1
|
Taheri E, Ehtesham H, Safdari R, Hormati A. A scientometric analysis and visualization of research on fatty liver diseases in Iran from 2003 to 2023. J Diabetes Metab Disord 2025; 24:103. [PMID: 40224531 PMCID: PMC11992304 DOI: 10.1007/s40200-025-01606-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/09/2025] [Indexed: 04/15/2025]
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide and is now a major global health threat. Due to the mechanistic links between these conditions, along with the rising prevalence of fatty liver disease, diabetes, and obesity, MAFLD (metabolic-associatedfatty liver diseases) was introduced as a new terminology and then updated to MASLD (Metabolic Dysfunction-associated Steatotic Liver Disease. These changes reflect a growing recognition of the importance of fatty liver and its associated health risks. Methods This scientometric study analyzed publications on "fatty liver diseases" (FLD) indexed in Scopus from 2003 to 2023 in Iran. Online Analysis Platforms and VOSviewer were used to assess publication trends and identify research hotspots. Results We retrieved 1,857 English articles on fatty liver diseases published between 2003 and 2023. The Journal of Hepatitis Monthly was notable for Iranian publications on fatty liver disease. Most publications were original articles and Dr. Sahebkar AH was the most prolific author. Iranian researchers primarily collaborated with scholars from the United States. The leading institution in terms of productivity was Tehran University of Medical Sciences. Conclusion This study reveals an increasing trend in both the number of Iranian publications and the citations of articles in the field of FLD. We believe this study can serve as a roadmap for future research and policy development on fatty liver diseases, which are a significant public health concern in Iran. Additionally, new strategies are needed to foster multi-disciplinary research and enhance international cooperation. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-025-01606-8.
Collapse
Affiliation(s)
- Ehsaneh Taheri
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamideh Ehtesham
- Department of Health Information Technology, Birjand University of Medical Sciences, Birjand, Iran
| | - Reza Safdari
- Department of Health Information Management, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Hormati
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Huang CL, Qi-En S, Cen XF, Ye T, Qu HS, Chen SJ, Liu D, Xia HG, Xu CF, Zhu JS. TJ0113 attenuates fibrosis in metabolic dysfunction-associated steatohepatitis by inducing mitophagy. Int Immunopharmacol 2025; 156:114678. [PMID: 40252468 DOI: 10.1016/j.intimp.2025.114678] [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: 02/24/2025] [Revised: 04/01/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) fibrosis is a liver disease accompanied by inflammatory cell infiltration. There is growing evidence that insufficient mitophagy can exacerbate inflammation and liver fibrosis (LF). TJ0113 is a novel mitophagy inducer. The study aimed to explore the role of TJ0113 in ameliorating fibrosis in MASH and its mechanisms. METHODS A high-fat diet (HFD)-induced MASH mice model and a transforming growth factor (TGF)-β1-induced LX-2 cells model were used, and then they were treated with TJ0113. Changes in hepatocyte damage were observed using electron microscopy. Expression of key molecules related to mitophagy, mitochondrial damage and inflammation in liver was detected by immunofluorescence staining (IF), immunohistochemistry (IHC) and western blotting (WB). RESULT TJ0113 induces mitophagy through parkin/PINK1 and ATG5 signaling pathways and reduces lipid accumulation, inflammation and fibrosis in the liver of MASH mice. TJ0113 attenuated hepatic injury and lowered serum ALT, AST, TC and TG levels. TJ0113 reduced pro-inflammatory factors (IL-1β, IL-6, TNF-α), TGF-β1/Smad pathway activation and typical fibrosis-related molecules (α-SMA, Collagen-1) expression. In addition, NF-κB/NLRP3 signaling pathway activation after MASH was significantly attenuated by enhanced Mitophagy. We found that TJ0113 was able to effectively and safely induce mitophagy in vitro and reduce TGF-β1/Smad signaling and downstream pro-fibrotic responses in TGF-β1-treated LX-2 cells. CONCLUSION TJ0113 enhances mitophagy to inhibit lipid accumulation, inflammation and fibrosis formation in MASH, and is a candidate for MASH treatment.
Collapse
Affiliation(s)
- Chun-Lian Huang
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Shen Qi-En
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Xu-Feng Cen
- Research Center of Clinical Pharmacy of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Ting Ye
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Hang-Shuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China.
| | - Si-Jia Chen
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Dong Liu
- Hangzhou PhecdaMed Co., Ltd., Third Floor, Building 2, No. 2626. Yuhangtang Road, Yuhang District, Hangzhou 310003, China.
| | - Hong-Guang Xia
- Research Center of Clinical Pharmacy of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Cheng-Fu Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Jian-Sheng Zhu
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China.
| |
Collapse
|
3
|
He Q, Li Y, Yu R, Lin M. Association of FGF21 with Metabolic and Cardiovascular Diseases: A Mendelian Randomization Analysis. Exp Clin Endocrinol Diabetes 2025. [PMID: 40245929 DOI: 10.1055/a-2549-6889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Studies have covered a possible relevance between fibroblast growth factor 21 (FGF21) and obesity-related metabolic complications and cardiovascular disease (CVD). Nevertheless, whether FGF21 is a causative factor in these diseases is not known. Using a bidirectional, two-sample Mendelian randomization (MR) approach, this study sought to establish a causal relationship between FGF21 and seven metabolic diseases and six CVDs. A large-scale meta-analysis dataset of genome-wide association studies (GWAS) was analyzed to generate summary-level statistics for FGF21. The diseases we studied included non-alcoholic fatty liver disease (NAFLD), obesity, type 2 diabetes (T2DM), hypertension, gestational diabetes (GDM), gestational hypertension (GHTN), pre-eclampsia or eclampsia (PE), atherosclerosis, cardiomyopathy (CMP), coronary heart disease (CHD), coronary atherosclerosis, heart failure (HF), myocardial infarction (MI) and the corresponding summary GAWS data were retrieved from the FinnGen Biobank and IEU Open GWAS Project database. The inverse variance-weighted (IVW) algorithm was the primary approach utilized for the MR analysis. The MR-Egger regression and MR-PRESSO tests were implemented to evaluate horizontal pleiotropy. The heterogeneity of instrumental variables was subsequently assessed utilizing Cochran's Q statistics.When diseases are used as exposures, MR analysis results of the IVW method indicated that NAFLD (Beta=- 0.047, 95% CI=- 0.08 to - 0.014; p=0.006), obesity (Beta=0.087, 95% CI=0.021-0.153; p=0.009), T2DM (Beta=0.071, 95% CI=0.037-0.106; p<0.001) correlated causally with FGF21. Nevertheless, FGF21 was not causally related to the remaining metabolic diseases and CVDs, according to the results of the MR analysis (p>0.05). It was demonstrated that the aforementioned results were robust and devoid of pleiotropy.Our study supports a causal association between NAFLD, obesity, and T2DM with FGF21. No substantial evidence exists to establish a causal relationship between FGF21 and other diseases. This study provides opportunities for the early prevention and innovative therapy of NAFLD, obesity, and T2DM.
Collapse
Affiliation(s)
- Qingwen He
- Affiliated Women's Hospital of Jiangnan University, Wuxi, China
| | - Yuguang Li
- Affiliated Women's Hospital of Jiangnan University, Wuxi, China
| | - Renqiang Yu
- Affiliated Women's Hospital of Jiangnan University, Wuxi, China
| | - Mengyuan Lin
- Affiliated Women's Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
4
|
Chen Q, Hu Q, Zhang F, Lu W, Yuan Z, Qiao F. Mechanistic evaluation of Jiu Wei Qing Zhi Gao in non-alcoholic fatty liver disease: insights from network Pharmacology and experimental validation. Hereditas 2025; 162:59. [PMID: 40221773 PMCID: PMC11992867 DOI: 10.1186/s41065-025-00427-2] [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: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
CONTEXT Jiu Wei Qing Zhi Gao (JWQZG), a traditional Chinese medicine (TCM) formulation, is widely utilized in China for managing non-alcoholic fatty liver disease (NAFLD). OBJECTIVE This study aimed to elucidate the therapeutic mechanisms of JWQZG in the management of NAFLD. MATERIALS AND METHODS Network pharmacology was employed to predict the potential mechanisms of JWQZG in NAFLD management. In vivo experiments were conducted using C57BL/6J mice fed a high-fat diet (HFD) for 16 weeks, followed by treatment with JWQZG at three dosages (1.85, 3.7, and 7.4 g/kg/day) or metformin (150 mg/kg/day) for 8 weeks. In vitro studies utilized HepG2 cells exposed to 0.5 mM palmitic acid (PA) for 24 h to establish an NAFLD model, followed by exposure to JWQZG-containing serum at three concentrations for an additional 24 h. Western blot analysis was used to analyze the expression levels of key signaling pathway components. RESULTS Results of network pharmacology analysis identified the insulin signaling pathway as a potential mediator of the protective effects of JWQZG in NAFLD. Treatment with JWQZG markedly reduced hepatic steatosis and improved insulin resistance. This was accompanied by enhanced expression of key components in the insulin signaling pathway, including insulin receptor substrate 1 (IRS1), phosphorylated PI3K (p-PI3K), phosphorylated AKT (p-AKT), and phosphorylated GSK3β (p-GSK3β), compared to the NAFLD model group. CONCLUSIONS These findings provide robust evidence supporting the therapeutic potential of JWQZG in NAFLD and its modulation of the insulin signaling pathway. Furthermore, the study offers valuable insights for the discovery of anti-NAFLD compounds derived from TCM formulations.
Collapse
Affiliation(s)
- Qinlei Chen
- Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210001, China
| | - Qianfeng Hu
- Nanjing University of Chinese Medicine, Nanjing, China, 210046
| | - Fan Zhang
- Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210001, China
| | - Weiting Lu
- Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210001, China
| | - Zheng Yuan
- Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210001, China.
| | - Fei Qiao
- Department of Infectious Diseases, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210001, China.
| |
Collapse
|
5
|
Chen A, Fu C, Chen H. Influence of triglycerides on the link between gamma-glutamyl transferase to high-density lipoprotein cholesterol ratio and nonalcoholic fatty liver disease in nonobese Chinese adults: A secondary cohort study. Medicine (Baltimore) 2025; 104:e42078. [PMID: 40228278 PMCID: PMC11999458 DOI: 10.1097/md.0000000000042078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents a chronic hepatic disorder marked by excessive lipid deposition within liver tissues. Previous studies have identified the gamma-glutamyl transferase to high-density lipoprotein cholesterol (GGT/HDL-c) ratio as a potential marker associated with NAFLD risk. However, the influence of triglyceride (TG) levels on this association has not been fully elucidated. This study was designed to assess whether TG modulates the relationship between GGT/HDL-c and the incidence of NAFLD, and to explore the possible interaction between TG and GGT/HDL-c in relation to NAFLD risk. This investigation was a secondary analysis based on data from a cohort study, encompassing 11,692 nonobese Chinese adults. Participants were stratified into 2 groups according to their triglyceride levels: hypertriglyceridemia (TG ≥ 1.7 mmol/L) and non-hypertriglyceridemia (TG < 1.7 mmol/L). Additionally, the GGT/HDL-c ratio was divided into quartiles for detailed analysis. The association between GGT/HDL-c, TG, and the occurrence of NAFLD was evaluated using Cox proportional hazards regression models. Furthermore, the potential modifying effect of TG on the GGT/HDL-c-NAFLD relationship was examined by categorizing participants into 8 subgroups based on TG status and quartiles of GGT/HDL-c ratio. During a mean follow-up duration of 29.24 months, 1926 individuals (16.47%) were newly diagnosed with NAFLD. Both elevated GGT/HDL-c ratios and higher TG concentrations were significantly correlated with an increased risk of developing NAFLD. TG levels influenced the strength of the association between GGT/HDL-c and NAFLD. Specifically, the association was stronger among individuals without hypertriglyceridemia (HR = 1.019, 95% CI = 1.015-1.023, P < .0001), and comparatively weaker in the hypertriglyceridemia group (HR = 1.012, 95% CI = 1.008-1.016, P < .0001). Moreover, a significant interaction effect was observed between TG and GGT/HDL-c, with the greatest risk identified in participants exhibiting both elevated GGT/HDL-c ratios and high TG levels (HR = 6.662, 95% CI = 5.237-8.474, P < .0001). Triglyceride levels appear to modify the relationship between GGT/HDL-c and NAFLD risk, with a notable interaction effect between these 2 factors. Among nonobese Chinese adults, simultaneous management aimed at reducing both GGT/HDL-c ratios and TG concentrations may contribute to lowering the risk of NAFLD onset.
Collapse
Affiliation(s)
- Aihua Chen
- Department of General Practice, First People’s Hospital of Changde City, Changde, Hunan, PR China
| | - Cishuang Fu
- Department of General Practice, First People’s Hospital of Changde City, Changde, Hunan, PR China
| | - Haiying Chen
- Department of General Practice, First People’s Hospital of Changde City, Changde, Hunan, PR China
| |
Collapse
|
6
|
Liu L, Deng Y, Yang L, Wang M, Lai Y. Comparison of efficacy and safety of pioglitazone and SGLT2 inhibitors in treating Asian patients in MASLD associated with type 2 diabetes: A meta-analysis. J Diabetes Complications 2025; 39:108998. [PMID: 40043473 DOI: 10.1016/j.jdiacomp.2025.108998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/09/2025] [Accepted: 03/02/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVE To comprehensively evaluate the therapeutic efficacy of pioglitazone and SGLT2 inhibitors (SGLT2i) in patients with MASLD and Type 2 Diabetes(T2DM). METHODS Electronic databases, including Web of Science, PubMed, the Cochrane Library, China National Knowledge Internet (CNKI), Wan-Fang Digital Database, and China Science and Technology Journal Database (VIP) were searched from inception to December 2024. Two reviewers independently assessed study eligibility, performed continuous data extraction, and independently evaluated bias risks. Liver ultrasonography, computed tomography (CT), and biochemical indices were utilized to determine the impact of treatment in both groups. Improvement in liver biomarkers and fibrosis as primary outcome indicators; Improvement in body composition, metabolic parameters, glucose parameters, and incidence of adverse effects as a secondary outcome indicator. For continuous variables, mean and standard deviation (SD) were extracted. RevMan 5.4 software was used to systematically analyze the literature, including heterogeneity testing, odds ratios (OR) calculation, and 95 % confidence intervals (CI) for each influencing factor. RESULTS Nine randomly controlled trials with 755 Asian participants were included. Our research showed that SGLT2i was more effective than pioglitazone in improving fibrosis-4 score (SMD 0.41 [95%CI 0.18,0.64] p = 0.005), visceral fat area (SMD 0.34 [95%CI 0.14,0.54] p = 0.0007), BMI (SMD 0.29 [95%CI 0.03,0.56] p = 0.03), and low-density lipoprotein levels (SMD 0.21 [95%CI 0.04,0.38] p = 0.01). In contrast, no statistically significant differences were observed in other outcomes. CONCLUSIONS Our study demonstrated that in patients with MASLD and T2DM, SGLT2i was more effective overall in improving liver fibrosis, blood lipids, liver fat, and body composition in the short term. These findings establish a theoretical basis for safe and rational drug use in clinical practice. Additionally, they may contribute to new insights into the pathogenesis of MASLD and type 2 diabetes and drug discovery and development efforts.
Collapse
Affiliation(s)
- Lingyan Liu
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Yongkun Deng
- Department of Medical Protection Center, The 926th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kaiyuan 661600, Yunnan Province, China.
| | - Lijuan Yang
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Miaojiao Wang
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Yong Lai
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali 671000, Yunnan Province, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali 671000, Yunnan Province, China; College of Pharmacy Dali University, Dali 671000, Yunnan Province, China.
| |
Collapse
|
7
|
Lei X, Xu Z, Huang Y, Huang L, Lang J, Qu M, Liu D. Regulation of Mitochondrial Quality Control of Intestinal Stem Cells in Homeostasis and Diseases. Antioxid Redox Signal 2025; 42:494-511. [PMID: 39225500 DOI: 10.1089/ars.2023.0489] [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: 09/04/2024]
Abstract
Significance: Intestinal stem cells (ISCs) are crucial for the continuous renewal and regeneration of the small intestinal epithelium. ISC fate decisions are strictly controlled by metabolism. Mitochondria act as the central hubs of energetic metabolism and dynamically remodel their morphology to perform required metabolic functions. Mitochondrial dysfunction is closely associated with a variety of gastrointestinal diseases. Recent Advances: In recent years, several studies have reported that mitochondria are potential therapeutic targets for regulating ISC function to alleviate intestinal diseases. However, how mitochondrial quality control mediates ISCs under physiological conditions and protects against intestinal injury remains to be comprehensively reviewed. Critical Issues: In this review, we summarize the available studies about how mitochondrial metabolism, redox state, dynamics, autophagy, and proteostasis impact ISC proliferation, differentiation, and regeneration, respectively. Future Directions: We propose that remodeling the function of mitochondria in ISCs may be a promising potential future direction for the treatment of intestinal diseases. This review may provide new strategies for therapeutically targeting the mitochondria of ISCs in intestinal diseases. Antioxid. Redox Signal. 42, 494-511.
Collapse
Affiliation(s)
- Xudan Lei
- Department of Experimental Research, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenni Xu
- Department of Experimental Research, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yujun Huang
- Department of Experimental Research, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Lingxiao Huang
- Department of Experimental Research, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jinyi Lang
- Department of Experimental Research, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Mingyue Qu
- The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Dengqun Liu
- Department of Experimental Research, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
8
|
Hong X, Liu H, Sun H, Zhuang Y, Xiao M, Li S, Li Y, Jing M. Yunnan medicine Jiangzhi ointment alleviates hyperlipid-induced hepatocyte ferroptosis by activating AMPK and promoting autophagy. Cytotechnology 2025; 77:73. [PMID: 40062225 PMCID: PMC11883071 DOI: 10.1007/s10616-025-00737-3] [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: 12/04/2024] [Accepted: 02/27/2025] [Indexed: 03/21/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a serious public health problem worldwide. The purpose of this study was to investigate whether Yunnan medicine Jiangzhi ointment (YMJO) can relieve the progression of NAFLD and to elucidate the specific mechanism involved. A NAFLD model was established in high-fat diet (HFD)-induced SD rats and free fatty acid (FFA)-induced BRL 3A cells. The expression of autophagy-related proteins and ferroptosis-related proteins was detected using Western blotting. The histopathological features of the livers of NAFLD rats were evaluated using hematoxylin and eosin (HE) and Oil Red O staining. The results revealed that in a successfully established HFD-induced NAFLD rat model, YMJO alleviated the progression of NAFLD, promoted autophagy, and inhibited ferroptosis. This regulatory mechanism is related to the activation of the AMPK pathway. Further study of the molecular mechanism via cell experiments revealed that YMJO activated FFA-induced liver cell autophagy through the AMPK signaling pathway and inhibited ferroptosis, thus alleviating the development of NAFLD. This study revealed that YMJO promotes phosphorylation by activating the AMPK pathway, enhances autophagy, ameliorates ferroptosis induced by high fat, and alleviates the occurrence and development of NAFLD.
Collapse
Affiliation(s)
- Xin Hong
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Haijing Liu
- Department of Acupuncture and Massage, Yunnan University of Chinese Medicine, Kunming, 650500 China
| | - Hongli Sun
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Yan Zhuang
- Department of Pharmaceutical Preparation, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Meizhen Xiao
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Shaoping Li
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Yandong Li
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Ming Jing
- Department of Acupuncture and Massage, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| |
Collapse
|
9
|
Zhang F, Yue K, Sun S, Lu S, Jia G, Zha Y, Zhang S, Chou CJ, Liao C, Li X, Duan Y. Targeting Histone Deacetylase 11 with a Highly Selective Inhibitor for the Treatment of MASLD. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412903. [PMID: 39976110 PMCID: PMC12005767 DOI: 10.1002/advs.202412903] [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: 10/14/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents the most prevalent chronic liver disorder globally. Due to its intricate pathogenesis and the current lack of efficacious pharmacological interventions, there is a pressing need to discover novel therapeutic targets and agents for MASLD treatment. Herein, it is found that histone deacetylase 11 (HDAC11), a subtype of HDAC family, is markedly overexpressed in both in vitro and in vivo models of MASLD. Furthermore, the knockdown of HDAC11 is observed to mitigate lipid accumulation in hepatic cells. A highly selective HDAC11 inhibitor, B6, which exhibits favorable pharmacokinetic property and liver distribution, is further designed and synthesized. Integrating RNA-seq data with in vivo and in vitro experiments, B6 is found to inhibit de novo lipogenesis (DNL) and promote fatty acid oxidation, thus mitigating hepatic lipid accumulation and pathological symptoms in MASLD mice. Further omics analysis and experiments reveal that B6 enhances the phosphorylation of AMPKα1 at Thr172 through the inhibition of HDAC11, consequently modulating DNL and fatty acid oxidation in the liver. In summary, this study identifies HDAC11 as a potential therapeutic target in MASLD and reports the discovery of a highly selective HDAC11 inhibitor with favorable drug-like properties for the treatment of MASLD.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Kairui Yue
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Simin Sun
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Shengyuan Lu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Geng Jia
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Yang Zha
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - C. James Chou
- Department of Drug Discovery and Biomedical SciencesCollege of PharmacyMedical University of South CarolinaCharlestonSC29425USA
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Xiaoyang Li
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
- Marine Biomedical Research Institute of QingdaoQingdaoShandong266071China
| | - Yajun Duan
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| |
Collapse
|
10
|
Gomonova VP, Raikhelson KL. [Cardiometabolic and genetic factors in the progression of metabolic dysfunction-associated steatotic liver disease]. TERAPEVT ARKH 2025; 97:149-156. [PMID: 40237751 DOI: 10.26442/00403660.2025.02.203203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/19/2025] [Indexed: 04/18/2025]
Abstract
AIM To evaluate the contribution of cardiometabolicfactors and PNPLA3 I148M (rs738409 C>G) gene polymorphism to the development of compensated advanced chronic liver disease (cACLD) in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). MATERIALS AND METHODS 108 patients with MASLD were enrolled and formed the internal validation group; 30 patients with MASLD were selected for external validation. Anamnestic data, anthropometric and laboratory parameters and the presence of PNPLA3 gene polymorphism I148M (rs738409 C>G) were assessed. Steatosis was detected by assessing the controlled attenuation parameter. Liver elasticity was assessed by transient elastography. cACLD was detected when the liver stiffness was ≥8 kPa. RESULTS Statistically significant difference was observed in the internal validation group during comparison of the incidence of cACLD depending on the presence of arterial hypertension (odds ratio - OR 5.58; 95% confidence interval - CI 1.21-25.71), type 2 diabetes mellitus - T2DM (OR 4.58; 95% CI 1.59-13.21), obesity (OR 3.13; 95% CI 1.1-8.9), dyslipidemia (OR 6.12; 95% CI 1.33-28.19) and the mutant G allele of the PNPLA3 gene (OR 3.9; 95% CI 1.28-11.88). Patients with cACLD had significantly higher mean values of waist circumference (WC), alanine aminotransaminase, aspartate aminotransaminase, gamma-glutamyl transferase and triglycerides, non-invasive markers of steatosis and fibrosis. The compiled prognostic model demonstrated a direct relationship between the likelihood of developing cACLD and the presence of T2DM (adjusted odds ratio - AOR 3.28; 95% CI 0.62-17.33), dyslipidemia (AOR 5.89; 95% CI 1.21-28.67) and WC value (AOR 1.05; 95% CI 1.01-1.11). PNPLA3 I148M gene polymorphism did not significantly affect the development of late stages of the disease. External validation of the model showed its moderate diagnostic ability. CONCLUSION T2DM, dyslipidemia and WC values are the determining factors in the development of cACLD in patients with MASLD. The PNPLA3 I148M gene polymorphism has no leading importance for the development of the progressive course of MASLD in the studied cohort.
Collapse
|
11
|
El-Sehrawy AAMA, Rashid TA, Ullah MI, Uthirapathy S, Ganesan S, Singh A, Devi A, Joshi KK, Jasim AS, Kadhim AJ. Cutting edge: ferroptosis in metabolic dysfunction-associated steatotic liver disease (MASLD) pathogenesis and therapy. Funct Integr Genomics 2025; 25:71. [PMID: 40131513 DOI: 10.1007/s10142-025-01579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Ferroptosis denotes a distinct form of controlled cell death marked by substantial iron buildup and significant lipid peroxidation, playing a crucial role in several disease processes linked to cell death. Given the liver's essential functions in iron and lipid metabolism and its vulnerability to oxidative damage, more research has investigated the correlation between ferroptosis and numerous hepatic diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD). NAFLD has arisen as a worldwide public health concern due to elevated morbidity and high death rates. The pathogenesis of MASLD remains incompletely elucidated. Recent data suggests that ferroptosis is crucial in the pathophysiology of MASLD; nevertheless, the specific processes by which ferroptosis influences MASLD remain unclear. The present review summarizes the molecular processes of ferroptosis and its intricate regulatory networks, outlines the differing impacts of ferroptosis at different stages of MASLD, and examines possible approaches targeting ferroptosis for the therapy of MASLD, suggesting a novel approach for its management.
Collapse
Affiliation(s)
| | - Teeba Ammar Rashid
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq.
| | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Aljouf, Saudi Arabia
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, 248002, Uttarakhand, India
- Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| | - Ahmed Salman Jasim
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 5100, Babylon, Iraq
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| |
Collapse
|
12
|
Hernandez R, Garcia-Rodriguez NS, Arriaga MA, Perez R, Bala AA, Leandro AC, Diego VP, Almeida M, Parsons JG, Manusov EG, Galan JA. The hepatocellular model of fatty liver disease: from current imaging diagnostics to innovative proteomics technologies. Front Med (Lausanne) 2025; 12:1513598. [PMID: 40109726 PMCID: PMC11919916 DOI: 10.3389/fmed.2025.1513598] [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: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is a prevalent chronic liver condition characterized by lipid accumulation and inflammation, often progressing to severe liver damage. We aim to review the pathophysiology, diagnostics, and clinical care of MASLD, and review highlights of advances in proteomic technologies. Recent advances in proteomics technologies have improved the identification of novel biomarkers and therapeutic targets, offering insight into the molecular mechanisms underlying MASLD progression. We focus on the application of mass spectrometry-based proteomics including single cell proteomics, proteogenomics, extracellular vesicle (EV-omics), and exposomics for biomarker discovery, emphasizing the potential of blood-based panels for noninvasive diagnosis and personalized medicine. Future research directions are presented to develop targeted therapies and improve clinical outcomes for MASLD patients.
Collapse
Affiliation(s)
- Renee Hernandez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Natasha S Garcia-Rodriguez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Marco A Arriaga
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Ricardo Perez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Auwal A Bala
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Ana C Leandro
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Vince P Diego
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Marcio Almeida
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Jason G Parsons
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Eron G Manusov
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Jacob A Galan
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| |
Collapse
|
13
|
Piana K, Ziomber-Lisiak A, Ruszczycki B, Bugajski A, Szczerbowska-Boruchowska M. Effects of high-calorie diet-induced obesity on molecular structures of lipids and proteins - A multi-organ study using FTIR spectroscopy. Arch Biochem Biophys 2025; 765:110325. [PMID: 39894381 DOI: 10.1016/j.abb.2025.110325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/19/2025] [Accepted: 01/31/2025] [Indexed: 02/04/2025]
Abstract
In the presented study, we evaluated changes in the molecular structures of lipids and proteins in organs/tissues at the early stage of obesity induced by a high-calorie diet (HCD), using animal models. We examined several different molecular parameters and the organs most affected by obesity. Fourier transform infrared (FTIR) spectroscopy combined with Principal Component Analysis (PCA) and Receiver Operating Characteristic (ROC) analysis were used to evaluate molecular changes in tissues taken from HCD-induced obese Wistar rats and their lean counterparts. We observed that at the early stage of obesity, changes occurred mainly in lipid structures, primarily affecting white epididymal adipose tissue (WAT) and the liver (Lr). No changes in protein molecular structures were observed in any of the examined organs. PCA showed distinctly different organ/tissue compositions, in terms of molecular parameters, for both groups. In turn, ROC analysis indicated that fatty acid chain length (FACL), lipid unsaturation (L_Unsat), and carbonyl/lipid ratio (Carb/L) for WAT, and FACL and lipid/protein ratio (L/P) for Lr, were the molecular parameters, whose levels differentiated the most between both groups. We demonstrated that studies using FTIR spectroscopy combined with advanced data mining methods could deepen the current knowledge about obesity and the biochemical changes occurring in the organs affected by this disease. Thus, they can help in the future with better and faster diagnosis and prevention of obesity and its complications.
Collapse
Affiliation(s)
- Kaja Piana
- AGH University of Krakow, Faculty of Physics and Applied Computer Science, Al. A. Mickiewicza 30, 30-059, Krakow, Poland
| | - Agata Ziomber-Lisiak
- Chair of Pathophysiology, Faculty of Medicine, Jagiellonian University Medical College, ul. Czysta 18, 31-121, Krakow, Poland
| | - Blazej Ruszczycki
- AGH University of Krakow, Faculty of Physics and Applied Computer Science, Al. A. Mickiewicza 30, 30-059, Krakow, Poland
| | - Andrzej Bugajski
- Chair of Pathophysiology, Faculty of Medicine, Jagiellonian University Medical College, ul. Czysta 18, 31-121, Krakow, Poland
| | | |
Collapse
|
14
|
Zhang X, Chan DCL, Zhu J, Sin DZY, Peng Y, Wong MKL, Zhu W, Tsui Y, Haqq AM, Ting JY, Kozyrskyj A, Chan FKL, Ng SC, Tun HM. Early-life antibiotic exposure aggravates hepatic steatosis through enhanced endotoxemia and lipotoxic effects driven by gut Parabacteroides. MedComm (Beijing) 2025; 6:e70104. [PMID: 39968496 PMCID: PMC11832435 DOI: 10.1002/mco2.70104] [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: 01/22/2024] [Revised: 11/13/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025] Open
Abstract
Compelling evidence supports a link between early-life gut microbiota and the metabolic outcomes in later life. Using an early-life antibiotic exposure model in BALB/c mice, we investigated the life-course impact of prenatal and/or postnatal antibiotic exposures on the gut microbiome of offspring and the development of metabolic dysfunction-associated steatotic liver disease (MASLD). Compared to prenatal antibiotic exposure alone, postnatal antibiotic exposure more profoundly affected gut microbiota development and succession, which led to aggravated endotoxemia and metabolic dysfunctions. This was primarily resulted from the overblooming of gut Parabacteroides and hepatic accumulation of cytotoxic lysophosphatidyl cholines (LPCs), which acted in conjunction with LPS derived from Parabacteroides distasonis (LPS_PA) to induce cholesterol metabolic dysregulations, endoplasmic reticulum (ER) stress and apoptosis. Integrated serum metabolomics, hepatic lipidomics and transcriptomics revealed enhanced glycerophospholipid hydrolysis and LPC production in association with the upregulation of PLA2G10, the gene controlling the expression of the group X secretory Phospholipase A2s (sPLA2-X). Taken together, our results show microbial modulations on the systemic MASLD pathogenesis and hepatocellular lipotoxicity pathways following early-life antibiotic exposure, hence help inform refined clinical practices to avoid any prolonged maternal antibiotic administration in early life and potential gut microbiota-targeted intervention strategies.
Collapse
Affiliation(s)
- Xi Zhang
- Microbiota I‐Center (MagIC)Hong Kong SARChina
- Jockey Club School of Public Health and Primary CareFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
- Li Ka Shing Institute of Health SciencesFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
- HKU‐Pasteur Research Pole, School of Public HealthLKS Faculty of Medicine, The University of Hong KongHong Kong SARChina
| | - Darren Chak Lun Chan
- HKU‐Pasteur Research Pole, School of Public HealthLKS Faculty of Medicine, The University of Hong KongHong Kong SARChina
| | - Jie Zhu
- Microbiota I‐Center (MagIC)Hong Kong SARChina
| | - Daniel Zhen Ye Sin
- Jockey Club School of Public Health and Primary CareFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
- Li Ka Shing Institute of Health SciencesFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
| | - Ye Peng
- Microbiota I‐Center (MagIC)Hong Kong SARChina
- Jockey Club School of Public Health and Primary CareFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
- Li Ka Shing Institute of Health SciencesFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
| | | | - Wenyi Zhu
- Microbiota I‐Center (MagIC)Hong Kong SARChina
- Li Ka Shing Institute of Health SciencesFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
- Department of Medicine and TherapeuticsFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Yee Tsui
- HKU‐Pasteur Research Pole, School of Public HealthLKS Faculty of Medicine, The University of Hong KongHong Kong SARChina
| | - Andrea M. Haqq
- Department of PediatricsUniversity of AlbertaEdmontonCanada
| | - Joseph Y. Ting
- Department of PediatricsUniversity of AlbertaEdmontonCanada
| | | | - Francis Ka Leung Chan
- Microbiota I‐Center (MagIC)Hong Kong SARChina
- Centre for Gut Microbiota ResearchThe Chinese University of Hong KongHong Kong SARChina
| | - Siew Chien Ng
- Microbiota I‐Center (MagIC)Hong Kong SARChina
- Li Ka Shing Institute of Health SciencesFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
- Department of Medicine and TherapeuticsFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Hein Min Tun
- Microbiota I‐Center (MagIC)Hong Kong SARChina
- Jockey Club School of Public Health and Primary CareFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
- Li Ka Shing Institute of Health SciencesFaculty of Medicine, The Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
15
|
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
|
16
|
Mohsenpoor MA, Parastouei K, Taghdir M, Akbarzadeh M, Abbaszadeh S, Abyazi Heris MA, Mansouri Rad MR, Jafari MA, Borzooei R. The effect of chitosan supplementation on liver function, hepatic steatosis predictors, and metabolic indicators in adults with non-alcoholic fatty liver disease: a randomized, double-blinded, placebo-controlled, clinical trial. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:60. [PMID: 40025618 PMCID: PMC11872331 DOI: 10.1186/s41043-025-00797-3] [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: 08/06/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a disease with high complications. An increment in dietary fiber consumption is an approach to NAFLD management, and chitosan as dietary fiber can play a role in the management of NAFLD. Thus, the present study aimed to investigate the effect of chitosan supplementation on liver function, hepatic steatosis predictors, and metabolic indices in adults with NAFLD. METHODS Seventy-two adults with NAFLD were randomly assigned to consume either 1.5 g/day chitosan or placebo along with a low-calorie (- 500 kcal/day) diet for 8 weeks in a parallel, randomized, double-blinded, placebo-controlled, clinical trial. Participants were assessed for dietary intake, physical activity, and anthropometric indices. Blood samples were taken to measure fasting blood sugar (FBS), cholesterol, triglycerides, high- and low-density lipoprotein (HDL and LDL). Liver function indices including alanine aminotransferase (ALT), aspartate transaminase (AST), and gamma-glutamyltransferase (GGT) were evaluated using blood samples as the primary outcomes. Fatty liver index (FLI), hepatic steatosis index (HSI), and triglyceride-glucose index (TyG) were calculated as hepatic steatosis predictors' indices. RESULTS After 8 weeks of study, 66 participants finished the study. In comparison with placebo, chitosan supplementation reduced weight (P = 0.041), waist circumference (P = 0.049), AST (P = 0.040), ALT (P = 0.001), and GGT (P = 0.028). Although the reduction of FBS, triglycerides, cholesterol, LDL, FLI, HSI, and TyG, and increment in HDL was higher in the chitosan group, the results were not significant (P > 0.05). CONCLUSIONS Eight-week supplementation with 1.5 g/day chitosan along with a low-calorie diet could possibly reduce weight, waist circumference, AST, ALT, and GGT, and ameliorate NAFLD. Further investigations are recommended. Trial registration The trial was registered at IRCT.ir as IRCT20140502017522N4 (March 2023).
Collapse
Affiliation(s)
| | - Karim Parastouei
- Health Research Centre, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Taghdir
- Health Research Centre, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
- Department of Nutrition and Food Hygiene, Faculty of Health, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Marzieh Akbarzadeh
- Nutrition Research Center, Department of Community Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Abbaszadeh
- Health Research Centre, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Abyazi Heris
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | - Ronak Borzooei
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Zhang J, Wang H, Wang Q, Mo J, Fu L, Peng S. EEF1A2 identified as a hub gene associated with the severity of metabolic dysfunction-associated steatotic liver disease. Mamm Genome 2025; 36:93-105. [PMID: 39414652 DOI: 10.1007/s00335-024-10078-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent chronic liver disease that ranges from metabolic dysfunction-associated steatotic liver (MASL) to metabolic dysfunction-associated steatohepatitis (MASH), and may eventually progress to cirrhosis and hepatocellular carcinoma (HCC). The underlying mechanism of MASLD remains incompletely understood. This study aimed to identify key gene implicated in MASLD pathogenesis and validate its correlation with disease severity through an integration of bioinformatics and experimental approaches. Liver transcriptome data from MASLD patients were obtained from the Gene Expression Omnibus (GEO) database. A diet-induced MASLD mouse model was developed, and liver RNA-sequencing was performed. Liver specimens and clinical data from patients were collected for further analysis. A total of 120 differentially expressed genes (DEGs) were shared between datasets GSE89632 and GSE213621, with functional enrichment in inflammatory, metabolic, and cell cycle-related pathways. Protein-protein interaction (PPI) network analysis identified three modules associated with MASLD, with the cell cycle-related module being the most notable. EEF1A2 was identified as a novel hub gene and revealed to be elevated with MASLD progression through dataset analysis. EEF1A2 was confirmed to be highly expressed in the livers of both MASLD mouse models and patients. Moreover, the increased expression of EEF1A2 in MASH was positively correlated with higher serum alanine aminotransferase (ALT), alanine aminotransferase (AST), total cholesterol (TC), and body mass index (BMI). In conclusion, EEF1A2 is a novel hub gene significantly associated with MASLD severity and is a promising biomarker and therapeutic target for MASLD.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huiwen Wang
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qianbing Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Mo
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Fu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
18
|
El-Kurjieh A, Al-Arab R, Hachem QA, Ibrahim JN, Kobeissy PH. ACSS2 and metabolic diseases: from lipid metabolism to therapeutic target. Lipids Health Dis 2025; 24:74. [PMID: 40001058 PMCID: PMC11853604 DOI: 10.1186/s12944-025-02491-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
Elevated incidence of metabolic disorders has been reported worldwide in the recent decade, highlighting the need for developing efficient therapies. These diseases result from a complex interplay of various factors that contribute to disease progression, complications, and resistance to current treatment options. Acetyl-CoA Synthetase Short Chain Family Member 2 (ACSS2) is a nucleo-cytosolic enzyme with both lipogenic and metabolic regulatory roles. Studies on ACSS2 have shown that it is involved in pathways commonly dysregulated in metabolic disorders, leading to fat deposition and disrupted cellular signaling. Although multiple studies have suggested a role of ACSS2 in the metabolic rewiring during tumorigenesis, few studies have examined its involvement in the pathophysiology of metabolic diseases. Recent evidence indicates that ACSS2 may contribute to the pathogenesis of various metabolic disorders making its examination of great interest and potentially aiding in the development of new therapeutic strategies. The objective of this review is to summarize the current understanding of ACSS2's role in metabolic disorders and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Alaa El-Kurjieh
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Reem Al-Arab
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Qamar Abou Hachem
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - José-Noel Ibrahim
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon.
| | - Philippe Hussein Kobeissy
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon.
| |
Collapse
|
19
|
Ditzenberger GL, Lake JE, Kitch DW, Kantor A, Muthupillai R, Moser C, Belaunzaran-Zamudio PF, Brown TT, Corey K, Landay AL, Avihingsanon A, Sattler FR, Erlandson KM. Effects of Semaglutide on Muscle Structure and Function in the SLIM LIVER Study. Clin Infect Dis 2025; 80:389-396. [PMID: 39046173 PMCID: PMC11848261 DOI: 10.1093/cid/ciae384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Semaglutide is highly effective for decreasing weight. Concomitant loss of muscle mass often accompanies weight loss and may have consequences on muscle function. METHODS This is a secondary analysis from the SLIM LIVER (Advancing Clinical Therapeutics Globally for HIV/AIDS and Other Infections, ACTG A5371) study, a single-arm study of semaglutide in people with human immunodeficiency virus (HIV, PWH) with metabolic dysfunction-associated steatotic liver diseases (MASLD). Participants received subcutaneous semaglutide for 24 weeks (titrated to 1 mg/week by week 4). Psoas volume and fat fraction were assessed from liver magnetic resonance imaging, and physical function was assessed by 10-time chair rise test and 4 m gait speed. Mean change from baseline to week 24 was estimated with linear regression modeling. RESULTS Fifty-one PWH were enrolled (muscle measures n = 46). The mean age was 50 years (standard deviation, 11), body mass index was 35.5 kg/m2 (5.6), 43% were women, 33% Black, and 39% Hispanic/Latino. Psoas muscle volume decreased by 9.3% (95% confidence interval [CI]: -13.4 to -5.2; P < .001) over 24 weeks, but psoas muscle fat did not significantly change (-0.42%; 95% CI: -1.00 to .17; P = .16). Chair rise and gait speed showed nonsignificant improvements of 1.27 seconds (95% CI: -2.7 to .10) and 0.05 m/sec (95% CI: -.01 to .10), respectively (both P > .07). The prevalence of slow gait speed (<1 m/sec) decreased from 63% to 46% (P = .029). CONCLUSIONS In PWH receiving semaglutide for MASLD, despite decreased psoas muscle volume, there was no significant change in physical function, suggesting function was maintained despite significant loss of muscle. CLINICAL TRIALS REGISTRATION NCT04216589.
Collapse
Affiliation(s)
- Grace L Ditzenberger
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jordan E Lake
- Department of Internal Medicine, UTHealth, Houston, Texas, USA
| | - Douglas W Kitch
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Amy Kantor
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Raja Muthupillai
- School of Engineering Medicine, Texas A&M University, Houston, Texas, USA
| | - Carlee Moser
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Todd T Brown
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kathleen Corey
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alan L Landay
- Departments of Internal Medicine and Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Fred R Sattler
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Kristine M Erlandson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
20
|
Jin L, Cui Y, Zhang Q, Xiong Y, Zheng X, Ye J, Zhang A, Xiao Z, Zhuang Z, Liang G, Hu X, Luo W, Zhu W. The natural product-derived JM-9 alleviates high-fat diet-induced fatty liver in mice by targeting MD2. Int Immunopharmacol 2025; 148:114053. [PMID: 39827672 DOI: 10.1016/j.intimp.2025.114053] [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: 10/30/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), is gradually emerging as one of the most prevalent liver diseases worldwide. Previous research demonstrated the involvement of myeloid differentiation factor 2 (MD2), a co-receptor of TLR4, as a key mediator in MASLD pathogenesis. The current study identifies JM-9 as a novel MD2 inhibitor, and focuses on evaluating its potential therapeutic effects in mitigating MASLD progression. METHODS Drug affinity responsive target stability (DARTS) assay and surface plasmon resonance assay were utilized to evaluate the MD2-targeting specificity of JM-9. In vitro, hepatocytes and macrophages were stimulated with palmitic acid (PA) followed by JM-9 treatment. In vivo, a high-fat diet (HFD)-induced MASLD model was established and subjected to JM-9 administration during the last 2 months. RESULTS JM-9 directly bound the Phe76 residue of MD2 to disrupt the PA-induced MD2/TLR4 complex formation, thus further restoring AMPK phosphorylation and inhibiting NF-κB activation to reducing lipid accumulation and inflammation, respectively. In the HFD-mediated MASLD mouse model, JM-9 alleviated the binding between MD2 and TLR4 in the liver and counteracted hepatic TBK1 and p65 activation and AMPK suppression, thereby mitigating liver inflammation, steatosis, and fibrosis. CONCLUSION JM-9, as a novel MD2 inhibitor, holds potential as a viable treatment option for MASLD by playing a dual role in regulating both lipid metabolism and inflammation response.
Collapse
Affiliation(s)
- Leiming Jin
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Yaqian Cui
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Qianhui Zhang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Yongqiang Xiong
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; Department of Endocrinology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiangsheng Zheng
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; Department of Endocrinology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiaxi Ye
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Anqi Zhang
- Department of Anesthesiology and Operation Room, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhongxiang Xiao
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325600, China
| | - Zaishou Zhuang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China
| | - Guang Liang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Xiang Hu
- Department of Endocrinology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Wu Luo
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; Department of Endocrinology and Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| | - Weiwei Zhu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035 Zhejiang, China; The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325035 Zhejiang, China.
| |
Collapse
|
21
|
Hao X, Li D, Huang X, Wang T, Wu P, Shen L, Zhang K, Sun S. Remnant cholesterol, a potential risk factor of metabolic dysfunction-associated fatty liver disease. Nutr Metab (Lond) 2025; 22:13. [PMID: 39966919 PMCID: PMC11837628 DOI: 10.1186/s12986-025-00898-0] [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: 04/28/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVES This study aimed to explore the association between remnant cholesterol and metabolic dysfunction-associated fatty liver disease (MAFLD) in an adult population in the United States. METHODS Data were collected from the National Health and Nutrition Examination Survey database during 2017-2020. Weighted multivariable logistic regression analyses and receiver operating characteristic (ROC) curves were used to investigate the association between remnant cholesterol and the risk of MAFLD. Subgroup and interaction analyses were performed. To further investigate the possible non-linear relationship between remnant cholesterol and MAFLD, a restricted cubic spline was used. RESULTS Among the included 3633 participants, the prevalence rate of MAFLD was 34.56%. After full adjustment, higher remnant cholesterol was associated with the risk of MAFLD (odds ratio [OR], 1.04; 95% confidence interval [CI], 1.01-1.06; P = 0.02), and compared with the lowest quartile of remnant cholesterol, the highest quartile of remnant cholesterol was more likely to be associated with MAFLD (OR, 3.70; 95%CI, 2.37,5.76; P < 0.0001). A non-linear relationship between remnant cholesterol and MAFLD was found in the restricted cubic spline regression model, suggesting that the risk of MAFLD initially increased rapidly and then gradually slowed down. CONCLUSION Remnant cholesterol was identified as a potential risk factor for MAFLD, and a non-linear relationship between remnant cholesterol and the prevalence of MAFLD was detected. Large-scale, high-quality prospective studies are required to validate these findings.
Collapse
Affiliation(s)
- Xuanyu Hao
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Dongyang Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| | - Siyu Sun
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
22
|
Zhu C, Cheng Y, Yang L, Lyu Y, Li J, Zhao P, Zhu Y, Xin X, Yin L. Notch1 siRNA and AMD3100 Ameliorate Metabolic Dysfunction-Associated Steatotic Liver Disease. Biomedicines 2025; 13:486. [PMID: 40002899 PMCID: PMC11853639 DOI: 10.3390/biomedicines13020486] [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: 12/20/2024] [Revised: 01/21/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Background and Objectives: As a key mechanism of metabolic dysfunction-associated steatotic liver disease (MASLD) pathogenesis, inflammation triggered by chronic liver injury and immune cells with macrophages enables MASLD to progress to an advanced stage with irreversible processes such as fibrosis, cell necrosis, and cancer in the liver. The complexity of MASLD, including crosstalk between multiple organs and the liver, makes developing a new drug for MASLD challenging, especially in single-drug therapy. It was reported that upregulation of Notch1 is closely associated with the function of pro-inflammatory macrophages. To leverage this signaling pathway in treating MASLD, we developed a combination therapy. Materials and Methods: We chose Notch1 siRNA (siNotch1) to block the Notch pathway so that phenotypic regulation and functional recovery can be achieved in macrophages, combining with small molecule drug AMD3100. AMD3100 can cut off the migration of inflammatory cells to the liver to impede the development of inflammation and inhibit the CXCL12/CXCR4 biological axis in liver fibrosis to protect against the activation of HSCs. Then, we investigated the efficacy of the combination therapy on resolving inflammation and MASLD. Results: We demonstrated that in liver cells, siNotch1 combined with AMD3100 not only directly modulated macrophages by downregulating multiple pathways downstream of Notch, exerting anti-inflammatory, anti-migration, and switch of macrophage phenotype, but also modulated macrophage phenotypes through inhibiting NET release. The restored macrophages further regulate HSC and neutrophils. In in vivo pharmacodynamic studies, combination therapy exhibits a superior therapeutical effect over monotherapy in MASLD models. Conclusions: These results constitute an siRNA therapeutical approach combined with a small molecule drug against inflammation and liver injury in MASLD, offering a promising therapeutic intervention for MASLD.
Collapse
Affiliation(s)
- Chunli Zhu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
| | - Yiheng Cheng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
| | - Lei Yang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
| | - Yifu Lyu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
| | - Jingjing Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
| | - Pengbo Zhao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
| | - Ying Zhu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
| | - Xiaofei Xin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
| | - Lifang Yin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; (C.Z.); (Y.C.); (L.Y.); (Y.L.); (J.L.); (P.Z.); (Y.Z.)
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
23
|
Mei J, Xie Y, Huang P, Jin Y, Wang X, Chen Y. The effects of HAPA theory-based case management in patients with metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2025; 30:101790. [PMID: 39954736 DOI: 10.1016/j.aohep.2025.101790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 02/17/2025]
Abstract
INTRODUCTION AND OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common liver disease affecting people's health, with its incidence increasing year by year. This study aims to determine the effects of Health Action Process Approach (HAPA)-based health management on patients diagnosed with MASLD. PATIENTS AND METHODS 204 MASLD patients were selected from the hospital's physical examination center from January 1, 2023, through April 1, 2023. Patients are randomly assigned to two groups (n = 102 each) using an envelope-based. Individuals in the experimental group received case management based on the HAPA theory, while standard health management was employed for control group patients. All subjects were monitored over a 6-month period, comparing body mass index (BMI), waist-to-hip ratio (WHR), levels of liver function (AST, ALT), blood lipid levels (total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and health behaviours (as assessed with the HPLP-II health promotion lifestyle scale) before and after the intervention. RESULTS Post-intervention, the experimental group showed significant improvement in fatty liver (P < 0.01) and reductions in biochemical indices, BMI, and WHR levels (P < 0.05), and a corresponding improvement in HDL-C levels (P < 0.001), compared to the control group. Additionally, patients in the experimental group exhibited significantly better health behaviour scores related to stress management, exercise, diet, and health responsibility compared to controls (P < 0.01). CONCLUSIONS HAPA theory-based case management can improve blood lipid profiles, liver function, and health-related behaviours in MASLD patients, highlighting its potential as an effective management strategy for this condition.
Collapse
Affiliation(s)
- Jia Mei
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yuncai Xie
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Pingping Huang
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yudi Jin
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Xia Wang
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Ying Chen
- Health Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| |
Collapse
|
24
|
Li S, Yang H, Zhou W, Wang R, Li L, Zhang C, Zhang J, Liu Y, Huang Z, Li G. Selenium Nanoparticles Decorated With Stevioside Potentially Attenuate Fructose Palmitate Induced Lipid Accumulation in HepG2 Cells. Mediators Inflamm 2025; 2025:7942947. [PMID: 39981401 PMCID: PMC11842145 DOI: 10.1155/mi/7942947] [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: 05/22/2024] [Revised: 10/12/2024] [Accepted: 12/23/2024] [Indexed: 02/22/2025] Open
Abstract
The excessive accumulation of lipid droplets within hepatocytes stands as a hallmark characteristic of metabolic-associated fatty liver disease (MAFLD). Selenium (Se) nanoparticles (NPs) have garnered considerable attention for their notable bioavailability, minimal toxicity, and exceptional antioxidant properties. However, a critical limitation lies in the propensity of SeNPs to aggregate into the biologically inactive elemental Se, thereby constraining their utility. Here, we utilized Stevioside (SV), a natural sweetener, to modify SeNPs and obtained the SV-SeNPs with a size of about 187 ± 7 nm. We aimed to investigate the effect of SV-SeNPs on high fructose-palmitate (HFP) induced lipid accumulation in HepG2 cells. Noteworthy is the absence of overt cytotoxicity attributed to SV-SeNPs on normal HepG2 cells. Of significance, our findings delineate the profound inhibitory effects of SV-SeNPs on the expression of key genes implicated in de novo lipogenesis, such as fatty-acid synthase (FASN), acetyl-CoA-carboxylase 1 (ACC1), and stearoyl-CoA desaturase-1 (SCD1) within HFP-induced HepG2 cells. Furthermore, our investigation reveals that SV-SeNPs mediate a significant reduction in lipid accumulation by activating the PI3K/AKT/Nrf2 signaling cascades. Additionally, the antioxidative properties of SV-SeNPs are underscored by their ability to counteract oxidative stress via the upregulation of two pivotal antioxidant enzymes, superoxide dismutase (SOD) and glutathione peroxidase (GSHPx). In conclusion, our study unveils the potential beneficial effects of SV-SeNPs on the prevention and treatment of MAFLD by effectively suppressing lipid accumulation and ameliorating oxidative stress.
Collapse
Affiliation(s)
- Shuai Li
- Medical Department, Jingchu University of Technology, Jingmen, China
- Center for Clinical Epidemiology and Methodology (CCEM), The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Hui Yang
- Department of Health Management of the Guangdong Second Provincial General Hospital and Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, China
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Wenjun Zhou
- Medical Department, Jingchu University of Technology, Jingmen, China
| | - Ruoting Wang
- Center for Clinical Epidemiology and Methodology (CCEM), The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Likang Li
- Center for Clinical Epidemiology and Methodology (CCEM), The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Changfa Zhang
- Center for Clinical Epidemiology and Methodology (CCEM), The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Jingyi Zhang
- Center for Clinical Epidemiology and Methodology (CCEM), The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Yingxin Liu
- Center for Clinical Epidemiology and Methodology (CCEM), The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Zhi Huang
- Department of Health Management of the Guangdong Second Provincial General Hospital and Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, China
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Guowei Li
- Center for Clinical Epidemiology and Methodology (CCEM), The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, 1280 Main St West, Hamilton, Ontario, Canada
| |
Collapse
|
25
|
Wang Y, Wang P. Development and validation of a new diagnostic prediction model for NAFLD based on machine learning algorithms in NHANES 2017-2020.3. Hormones (Athens) 2025:10.1007/s42000-025-00634-6. [PMID: 39939537 DOI: 10.1007/s42000-025-00634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
AIMS Nonalcoholic fatty liver disease (NAFLD) is a multisystem disease that can trigger the metabolic syndrome. Early prevention and treatment of NAFLD is still a huge challenge for patients and clinicians. The aim of this study was to develop and validate machine learning (ML)-based predictive models. The model with optimal performance would be developed as a set of simple arithmetic tools for predicting the risk of NAFLD individually. METHODS Statistical analyses were performed in 2428 individuals extracted from the National Health and Nutrition Examination Survey (NHANES, cycle 2017-2020.3) database. Feature variables were selected by the least absolute shrinkage and selection operator (LASSO) regression. Seven ML algorithms, including logistic regression (LR), decision tree (DT), random forest (RF), extreme gradient boosting (XGB), K-nearest neighbor (KNN), light gradient boosting machine (LightGBM), and multilayer perceptron (MLP), were used to construct models based on the feature variables and evaluate their performance. The model with the best performance was transformed into a diagnostic predictive nomogram (DPN). The DPN was developed into an online calculator and an Excel algorithm tool. Receiver operating characteristic (ROC) curve, decision curve analysis (DCA), and subgroup analyses were used to compare and assess the predictive abilities of the DPN and six existing NAFLD predictive models, including the ZJU index, the hepatic steatosis index (HSI), the triglyceride-glucose index (TyG), the Framingham steatosis index (FSI), the fatty liver index (FLI), and the visceral adiposity index (VAI). RESULTS Among the 2428 participants, the prevalence of NAFLD was 47.45%. LASSO regression identified eight variables from 39 variables, including body mass index (BMI), waist circumference (WC), alanine aminotransferase (ALT), triglyceride (TG), diabetes, hypertension, uric acid (UA), and race. Among the models constructed by the seven algorithms mentioned above, the LR-based model performed the best, demonstrating outstanding performance in terms of area under the curve (AUC, 0.823), accuracy (0.754), precision (0.768), specificity (0.804), and positive predictive value (0.768). It was then transformed into the DPN, which was successfully developed as an online calculator and an Excel algorithm tool. The diagnostic accuracy (AUC 0.856, 95% confidence interval (CI) 0.839-0.874, and AUC 0.823, 95% CI 0.793-0.854, respectively) and net clinical benefit of DPN in the training and validation sets were superior to those of the ZJU, HSI, TyG, FSI, FLI, and VAI. The results were maintained in subgroup analyses. CONCLUSIONS The LR model based on ML was developed, exhibiting good performance. DPN can be used as an individualized tool for rapid detection of NAFLD.
Collapse
Affiliation(s)
- Yazhi Wang
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Peng Wang
- The Department of Pharmacy, The 987th Hospital of Joint Logistics Support Force of People's Liberation Army, Baoji, Shaanxi, 721004, China.
| |
Collapse
|
26
|
Shariq A, Khan S, Usmani SUR. The Role of Dietary Protein in Mitigating the Risk of Nonalcoholic Fatty Liver Disease. Nutr Rev 2025:nuae229. [PMID: 39921657 DOI: 10.1093/nutrit/nuae229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent liver disease, mainly associated with excessive accumulation of fat in the liver. It has become a global health concern. The diagnosis of NAFLD is often done through liver biopsy; however, noninvasive methods have their own advantages. Dietary intervention, especially increased dietary protein alongside managing overall body weight, have been shown to be a promising strategy to lessen the impact of NAFLD. Dietary protein has been shown to reduce fat accumulation in the liver by increasing liver metabolism, eliciting satiety, improving insulin sensitivity, and enhancing muscle mass retention, collectively aiding in weight management. Both animal and plant proteins have benefits; however, plant proteins have demonstrated more metabolic advantages, while animal proteins have more downsides. Bridging the protein gap is critical, particularly in areas with limited availability to high-quality protein or in populations where dietary protein intake is inadequate. This commentary highlights the importance of obtaining sufficient protein from readily available and sustainable food sources. Furthermore, diets high in protein, like the Mediterranean diet, have proven to delay the advancement and likelihood of NAFLD. In conclusion, adequate dietary protein plays a crucial part in diminishing the risk of NAFLD, and efforts in public health should concentrate on addressing protein deficiency to decrease the growing burden of liver disease.
Collapse
Affiliation(s)
- Abia Shariq
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Sarosh Khan
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | | |
Collapse
|
27
|
Song H, Zhang M, Guo C, Guo X, Ma Y, Ma Y. Implication of protein post translational modifications in gastric cancer. Front Cell Dev Biol 2025; 13:1523958. [PMID: 39968176 PMCID: PMC11833226 DOI: 10.3389/fcell.2025.1523958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Gastric cancer (GC) is one of the most common and highly lethal malignant tumors worldwide, and its occurrence and development are regulated by multiple molecular mechanisms. Post-translational modifications (PTM) common forms include ubiquitylation, phosphorylation, acetylation and methylation. Emerging research has highlighted lactylation and glycosylation. The diverse realm of PTM and PTM crosstalk is linked to many critical signaling events involved in neoplastic transformation, carcinogenesis and metastasis. This review provides a comprehensive overview of the impact of PTM on the occurrence and progression of GC. Specifically, aberrant PTM have been shown to alter the proliferation, migration, and invasion capabilities of GC cells. Moreover, PTM are closely associated with resistance to chemotherapeutic agents in GC. Notably, this review also discusses the phenomenon of PTM crosstalk, highlighting the interactions among PTM and their roles in regulating signaling pathways and protein functions. Therefore, in-depth investigation into the mechanisms of PTM and the development of targeted therapeutic strategies hold promise for advancing early diagnosis, treatment, and prognostic evaluation of GC, offering novel insights and future research directions.
Collapse
Affiliation(s)
- Houji Song
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Mingze Zhang
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Chengwang Guo
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xi Guo
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yuqi Ma
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yuntao Ma
- The First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
28
|
Wang J, Wang Z, Yu Y, Cheng S, Wu J. Advances in research on metabolic dysfunction-associated steatotic liver disease. Life Sci 2025; 362:123362. [PMID: 39761743 DOI: 10.1016/j.lfs.2024.123362] [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: 09/14/2024] [Revised: 12/13/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
The global increase in obesity-related metabolic disorders has led to metabolic dysfunction-associated steatotic liver disease (MASLD) emerging as one of the most prevalent chronic liver disease worldwide. Despite growing concerns, the exact pathogenesis of MASLD remains unclear and no definitive treatments have been made available. Consequently, the need for comprehensive research on MASLD is more critical than ever. Gaining insight into the mechanisms of the disease can lay the groundwork for identifying new therapeutic targets and can facilitate the development of diagnostic tools that enable the early detection and intervention of MASLD. Research has discovered a multifactorial etiology for MASLD, suggesting that potential therapeutic strategies should be considered from a variety of perspectives. This review delves into the pathogenesis of MASLD, current diagnostic approaches, potential therapeutic targets, the status of clinical trials for emerging drugs, and the most promising treatment methods available today. With a focus on therapeutic targets, the aim is to offer fresh insights and guide for future research in the treatment of MASLD.
Collapse
Affiliation(s)
- Jiawang Wang
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Zhongyu Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yao Yu
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Si Cheng
- Beijing Tiantan Hospital, Capital Medical University, Beijing 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 10070, China.
| | - Jianping Wu
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China; Department of Pharmacology, Hubei University of Medicine, Shiyan 440070, China; Beijing Tiantan Hospital, Capital Medical University, Beijing 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 10070, China.
| |
Collapse
|
29
|
Ashraf A, Rather SA, Mehraj M. "Evaluation of Curcuma zedoaria Rosc. in the management of non-alcoholic fatty liver Disease: A Randomized, single blind, controlled trial". Arab J Gastroenterol 2025; 26:112-119. [PMID: 39880723 DOI: 10.1016/j.ajg.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/11/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND AND STUDY AIMS Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disorder, affecting 23% to 32% of the global population. This clinical study aimed to assess the efficacy of Curcuma zedoaria Rosc. compared to vitamin E in managing NAFLD. PATIENTS AND METHODS In this randomized, single-blind, standard-controlled study, 68 patients with grade 1 (mild) and grade 2 (moderate) NAFLD were randomly assigned to receive either Curcuma zedoaria Rosc. powder in capsule form (500 mg orally, twice a day) in the test group or vitamin E (400 mg orally, twice a day) in the control group for 60 days. Secondary endpoints included improvements in fatty liver grades, ultra-sonographic liver span, lipid profile, and liver function parameters after 60 days. Primary endpoints included improvements in dull ache intensity in the right hypochondrium (RHC), dyspepsia, anorexia, and severity of malaise assessed at days 0, 15, 30, 45, and 61. RESULTS Per protocol analysis was performed on 50 patients who completed the study. Both test and control groups showed significant improvement in dull ache severity in the RHC (p < 0.0001). The test group exhibited more favorable outcomes post-treatment (Chi-sq = 23.17, df = 2, p < 0.0001). Dyspepsia severity significantly improved in both groups post-treatment (p = 0.005 and p = 0.010, respectively), with the test group showing slightly better outcomes. Anorexia significantly improved in the test group (p = 0.016) from 72.00 % reporting absence post-treatment to 100.00 % absence, while the control group showed improvement without statistical significance (p = 0.102). Malaise severity significantly improved in the test group (p < 0.0001), with 84.00 % reporting absence post-treatment compared to 8.00 % in the control group, showing significant differences (p < 0.0001). Both groups exhibited a significant reduction in liver span post-treatment (p-value < 0.0001) without inter- group differences. Fatty liver grades improved significantly in both groups post-treatment (p < 0.0001), with no significant difference between groups (Chi-sq = 4, df = 2, p = 0.1353). There were no changes in liver function markers and lipid parameters in both groups, though the test drug demonstrated a slight reduction in serum triglyceride levels. No drug-related adverse events were observed during the trial. CONCLUSION The study revealed that Curcuma zedoaria Rosc. is effective in managing NAFLD, showing better outcomes than vitamin E in subjective parameters like dyspepsia, malaise, anorexia, and dull ache in RHC. With no observed drug-related adverse events, Curcuma zedoaria Rosc. could be a suitable alternative to conventional treatment modalities for NAFLD.
Collapse
Affiliation(s)
- Aaqib Ashraf
- Dept. of Moalajat,(Medicine) RRIUM, Srinagar, India
| | | | | |
Collapse
|
30
|
Miura Y, Voican C, Sakai Y, Nishikawa M, Leclerc E. A computational model of the crosstalk between hepatocyte fatty acid metabolism and oxidative stress highlights the key enzymes, metabolites, and detoxification pathways in the context of MASLD. Toxicol Appl Pharmacol 2025; 495:117185. [PMID: 39631537 DOI: 10.1016/j.taap.2024.117185] [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: 09/10/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; formerly known as NAFLD) is a common liver disease worldwide and carries the risk of progressing to severe liver conditions, such as fibrosis and liver cancer. In the context of MASLD, evaluating fat accumulation in the liver and the subsequent production of oxidative stress is essential to understand the disease propagation. However, clinical studies using human patients to investigate the fat accumulation and the onset of oxidative stress in MASLD face ethical and technical challenges, highlighting the importance of alternative methods. To understand the relationship between fatty acid metabolism, lipid accumulation, oxidative stress generation, and antioxidant mechanisms in hepatocytes, we proposed a new mathematical model. The importance of this model lies in its ability to track the time-dependent changes in oxidative stress and glutathione concentration in response to the input of fatty acids. Furthermore, the model allows for the evaluation of the effects of altering the activity of the key enzymes involved in those mechanisms. Our model is anticipated to provide new insights into MASLD therapy strategies by identifying key pathways and predicting the effects of drug-induced changes in enzyme activity.
Collapse
Affiliation(s)
- Yuki Miura
- Department of Chemical System Engineering, Graduate school of Engineering, the University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Cosmin Voican
- Department of Hepatogastroenterology and Nutrition, Antoine-Béclère University Hospital, AP-HP Paris-Saclay University, 92140 Clamart, France; INSERM U996, 91400 Orsay, France; Faculty of Medicine, Paris-Saclay University, 94270 Le Kremlin-Bicêtre, France
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate school of Engineering, the University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.; CNRS IRL 2820; Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, Graduate school of Engineering, the University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Eric Leclerc
- CNRS IRL 2820; Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan.
| |
Collapse
|
31
|
King L, Xia L, Chen J, Li W, Wang Q, Huang Y, Wang P, Liang X, Li Y, Chen L, Shan Z, Peng X, Liu L. Exposure to perchlorate and cardiovascular disease in China: A community-based cross-sectional study and benchmark dose estimation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125429. [PMID: 39617200 DOI: 10.1016/j.envpol.2024.125429] [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: 08/01/2024] [Revised: 10/22/2024] [Accepted: 11/28/2024] [Indexed: 12/08/2024]
Abstract
The association between exposure to perchlorate, which inhibits thyroidal iodine uptake, and cardiovascular disease (CVD) is unclear in China. Moreover, the point of departure (POD) for perchlorate based on observed adverse health effect in Chinese populations remains absent. A total of 2355 adults (mean age 50.4 years and 39.2% male) from four communities in Shenzhen were included in analyses. Spot urine specimens were collected to measure urinary perchlorate concentrations, which were applied to estimate daily intakes of perchlorate. Multivariable logistic regression model was applied to examine the association between perchlorate and CVD. The roles of cardiometabolic risk factors, including obesity, abdominal obesity, hypertension, diabetes, and hyperlipidemia, were evaluated with mediation analyses. We further employed Bayesian benchmark dose (BMD) modeling to derive the POD for risk assessment. Comparing extreme tertiles, subjects in the highest perchlorate tertile had a significantly elevated risk of prevalent CVD (OR: 2.16; 95% CI: 1.28, 3.65). Multivariable-adjusted ORs for hypertension, diabetes, and hyperlipidemia associated with per doubling in urinary perchlorate concentration were 1.11 (95% CI: 1.01, 1.21), 1.15 (95% CI: 1.02, 1.28), and 1.11 (95% CI: 1.01, 1.20), respectively. Hypertension, diabetes, and hyperlipidemia partially mediated the perchlorate-CVD association (mediated proportion ranged from 7.75% to 11.30%). Given a benchmark response of 5% and 10%, the model-averaged BMD lower bounds (BMDLs) of perchlorate exposure on CVD were 0.15 and 0.40 μg/kg-bw day, respectively. Our estimated POD for perchlorate was lower than those recommended by other groups. These findings call for stricter regulations on perchlorate contamination to promote cardiovascular health in China.
Collapse
Affiliation(s)
- Lei King
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Xia
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanyi Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Wang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Huang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Wang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoling Liang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonggang Li
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhilei Shan
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolin Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Non-communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China.
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
32
|
Zhang L, Zheng Y, Shao M, Chen A, Liu M, Sun W, Li T, Fang Y, Dong Y, Zhao S, Luo H, Feng J, Wang Q, Li L, Zheng Y. AlphaFold-based AI docking reveals AMPK/SIRT1-TFEB pathway modulation by traditional Chinese medicine in metabolic-associated fatty liver disease. Pharmacol Res 2025; 212:107617. [PMID: 39832686 DOI: 10.1016/j.phrs.2025.107617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a chronic, progressive disorder characterized by hepatic steatosis and excessive lipid accumulation. Its high global adult prevalence (approximately 50.7 %) is a significant concern worldwide. However, FDA-approved therapeutic drugs remains lacking. Qigui Jiangzhi Formula (QGJZF) shows promise in treating MAFLD by effectively decreasing lipid levels and improving hepatic steatosis, however its mechanisms remain unclear. This study investigated QGJZF's effects in high-fat diet-induced zebrafish and golden hamsters, and in palmitate (PA) and oleic acid (OA) - induced HepG2 cells, using the SymMap database to identify potential targets and pathways of QGJZF in MAFLD and AlphaFold algorithms to predict protein structures. In vivo, QGJZF significantly alleviated hepatic lipid deposition. Intriguingly, QGJZF decreased lipid droplets and its levels are negative correlated with the numbers of autolysosomes, indicating that QGJZF's mechanism of ameliorating liver lipid deposition may be related to the regulation of autophagy. QGJZF upregulated the expressions of phosphorylated -Adenosine 5'-monophosphate (AMP) - activated protein kinase (p-AMPK), Sirtuin deacetylase 1 (SIRT1) and Transcription factor EB (TFEB), accompanied by the changes in autophagy-related proteins. In vitro, QGJZF inhibited the lipid deposition in PA/OA-stimulated HepG2 cells, and its effect was blocked by an autophagy inhibitor Baf-A1, which was mediated through upregulation of TFEB and its mediated autophagy-lysosomal pathway. Moreover, cotreatment with AMPK inhibitor Compound C, the regulation of QGJZF on TFEB, SIRT1, autophagy-related protein levels, and lipid deposition were reversed. Network pharmacology identified the PRKAA2 (AMPK) and SIRT1 as key hub targets. Futher analysis of their structures using AlphaFold3 algorithms, yielded high-ranking scores of 0.97 and 0.93, respectively. Liquid chromatography-mass spectrometry combined with molecular docking expounded its five compounds in QGJZF binding to AMPK protein. These findings suggest that QGJZF as a therapeutic agent in augmenting autophagy-facilitated lipid clearance for the management of MAFLD via AMPK/SIRT1-TFEB axis.
Collapse
Affiliation(s)
- Lulu Zhang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, China
| | - Yi Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingyan Shao
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Aiping Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Meiyi Liu
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Tianxing Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yini Fang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Dong
- Monitoning and Statistical Research Center, National Administration of Traditional Chinese Medicine, Beijing 100600, China
| | - Shipeng Zhao
- Graduate School of China Academy of Chinese Medical Sciences, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hui Luo
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China.
| | - Qi Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Lingru Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yanfei Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
33
|
Arai K, Ono Y, Hirai N, Sugiura Y, Kaneko K, Matsuda S, Iio K, Kajino K, Saitoh T, Wei FY, Katagiri H, Inoue A. Chemogenetic activation of hepatic G 12 signaling ameliorates hepatic steatosis and obesity. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167566. [PMID: 39542224 DOI: 10.1016/j.bbadis.2024.167566] [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: 05/03/2024] [Revised: 09/20/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE Hepatic steatosis, the early stage of nonalcoholic fatty liver disease (NAFLD), currently lacks targeted pharmacological treatments. G protein-coupled receptors (GPCRs) in hepatocytes differentially regulate lipid metabolism depending on their coupling profile of G protein subtypes. Unlike Gs, Gi, and Gq signaling, the role of G12 signaling in hepatic steatosis remains elusive. The objective of this study was to investigate the effect of G12 signaling on hepatic steatosis and obesity and its mechanisms. METHODS We generated mice expressing a G12-coupled designer GPCR in a liver-specific manner. We performed phenotypic analysis in the mice under the condition of fasting (acute hepatic steatosis model) or high-fat diet feeding (chronic hepatic steatosis model). RESULTS In acute and chronic hepatic steatosis models, chemogenetic activation of hepatic G12 signaling suppressed the progression of hepatic steatosis. The treatment led to an increased triglyceride secretion with little effect on mitochondrial respiratory activity, fatty acid oxidation, de novo lipogenesis, and fatty acid uptake. Furthermore, in a high-fat-diet-induced obesity model, activation of the G12-coupled designer GPCR exerted anti-obesity effects with increased whole-body energy expenditure and fat oxidation. Anti-FGF21 antibody treatment showed that the anti-obesity effects of the hepatic G12D activation relied in part on the hepatokine FGF21. CONCLUSIONS Our findings indicate that the activation of G12 signaling in the liver has the potential to prevent hepatic steatosis and obesity. This discovery provides a strong rationale for the development of drugs targeting G12-coupled GPCRs expressed in the liver.
Collapse
Affiliation(s)
- Kaito Arai
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Yuki Ono
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Natsumi Hirai
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Yuki Sugiura
- Center for Cancer Immunotherapy and Immunobiology (CCII), Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan; Human Biology Microbiome Quantum Research Center (WPI-Bio2Q), Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Shigeru Matsuda
- Department of Modomics Biology and Medicine, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Keita Iio
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8571, Japan
| | - Keita Kajino
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8571, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Fan-Yan Wei
- Department of Modomics Biology and Medicine, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
34
|
Jarmakiewicz-Czaja S, Sokal-Dembowska A, Filip R. Effects of Selected Food Additives on the Gut Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). MEDICINA (KAUNAS, LITHUANIA) 2025; 61:192. [PMID: 40005309 PMCID: PMC11857189 DOI: 10.3390/medicina61020192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025]
Abstract
The purpose of this article is to present selected food additives as disruptors of normal intestinal homeostasis with a potential impact on the development of metabolic dysfunction-associated steatotic liver disease (MASLD). A comprehensive literature search was conducted in three major electronic databases: PubMed, ScienceDirect, and Google Scholar. MASLD is a prevalent liver condition that is closely related to the global rise in obesity. Its pathogenesis is multifactorial, with genetic, environmental, and metabolic factors playing a key role. The "multiple-hit" hypothesis suggests that a Western-style diet, rich in ultra-processed foods, saturated fats, and food additives, combined with low physical activity, contributes to obesity, which promotes lipid accumulation in the liver. Recent studies underscore the role of impaired intestinal homeostasis in the development of MASLD. Food additives, including preservatives, emulsifiers, and sweeteners, affect gut health and liver function. Selected preservatives inhibit pathogenic microorganisms but disrupt the intestinal microbiota, leading to changes in intestinal permeability and liver dysfunction. Some emulsifiers and thickeners can cause inflammation and alter the gut microbiome, contributing to liver steatosis. Furthermore, the use of sweeteners such as sucralose and aspartame has been linked to changes in liver metabolism and intestinal microbial composition, which in turn promotes metabolic disorders.
Collapse
Affiliation(s)
- Sara Jarmakiewicz-Czaja
- Faculty of Health Sciences and Psychology, University of Rzeszow, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Aneta Sokal-Dembowska
- Faculty of Health Sciences and Psychology, University of Rzeszow, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Rafał Filip
- Gastroenterology Clinic, Center for Comprehensive Treatment of Inflammatory, Bowel Disease Regional Hospital No. 2 in Rzeszow, 35-301 Rzeszow, Poland
- Department of Internal Medicine, Faculty of Medicine, University of Rzeszow, 35-959 Rzeszow, Poland
| |
Collapse
|
35
|
Wan X, Ma J, Bai H, Hu X, Ma Y, Zhao M, Liu J, Duan Z. Drug Advances in NAFLD: Individual and Combination Treatment Strategies of Natural Products and Small-Synthetic-Molecule Drugs. Biomolecules 2025; 15:140. [PMID: 39858534 PMCID: PMC11764138 DOI: 10.3390/biom15010140] [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: 12/15/2024] [Revised: 01/07/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease and is closely associated with metabolic diseases such as obesity, type 2 diabetes mellitus (T2DM), and metabolic syndrome. However, effective treatment strategies for NAFLD are still lacking. In recent years, progress has been made in understanding the pathogenesis of NAFLD, identifying multiple therapeutic targets and providing new directions for drug development. This review summarizes the recent advances in the treatment of NAFLD, focusing on the mechanisms of action of natural products, small-synthetic-molecule drugs, and combination therapy strategies. This review aims to provide new insights and strategies in treating NAFLD.
Collapse
Affiliation(s)
- Xing Wan
- The First Affiliated Hospital of Dalian Medical University, Dalian 116012, China; (X.W.); (H.B.); (M.Z.)
- Institute of Integrated Traditional Chinese and Western Medicine, Dalian Medical University, Dalian 116051, China
| | - Jingyuan Ma
- The First Clinical Medical College, Liaoning University of Traditional Chinese Medicine, Shenyang 110033, China; (J.M.); (Y.M.)
| | - He Bai
- The First Affiliated Hospital of Dalian Medical University, Dalian 116012, China; (X.W.); (H.B.); (M.Z.)
| | - Xuyang Hu
- The Second Clinical Medical College, Liaoning University of Traditional Chinese Medicine, Shenyang 110033, China;
| | - Yanna Ma
- The First Clinical Medical College, Liaoning University of Traditional Chinese Medicine, Shenyang 110033, China; (J.M.); (Y.M.)
| | - Mingjian Zhao
- The First Affiliated Hospital of Dalian Medical University, Dalian 116012, China; (X.W.); (H.B.); (M.Z.)
| | - Jifeng Liu
- The First Affiliated Hospital of Dalian Medical University, Dalian 116012, China; (X.W.); (H.B.); (M.Z.)
| | - Zhijun Duan
- The First Affiliated Hospital of Dalian Medical University, Dalian 116012, China; (X.W.); (H.B.); (M.Z.)
| |
Collapse
|
36
|
Li Q, Sheng J, Baruscotti M, Liu Z, Wang Y, Zhao L. Identification of Senkyunolide I as a novel modulator of hepatic steatosis and PPARα signaling in zebrafish and hamster models. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118743. [PMID: 39209000 DOI: 10.1016/j.jep.2024.118743] [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: 07/01/2024] [Revised: 08/19/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Non-alcoholic fatty liver disease (NAFLD) is the leading cause of liver-related morbidity and mortality, with hepatic steatosis being the hallmark symptom. Salvia miltiorrhiza Bunge (Smil, Dan-Shen) and Ligusticum striatum DC (Lstr, Chuan-Xiong) are commonly used to treat cardiovascular diseases and have the potential to regulate lipid metabolism. However, whether Smil/Lstr combo can be used to treat NAFLD and the mechanisms underlying its lipid-regulating properties remain unclear. PURPOSE To assess the feasibility and reliability of a short-term high-fat diet (HFD) induced zebrafish model for evaluating hepatic steatosis phenotype and to investigate the liver lipid-lowering effects of Smil/Lstr, as well as its active components. METHODS The phenotypic alterations of liver and multiple other organ systems were examined in the HFD zebrafish model using fluorescence imaging and histochemistry. The liver-specific lipid-lowering effects of Smil/Lstr combo were evaluated endogenously. The active molecules and functional mechanisms were further explored in zebrafish, human hepatocytes, and hamster models. RESULTS In 5-day HFD zebrafish, significant lipid accumulation was detected in the blood vessels and the liver, as evidenced by increased staining with Oil Red O and fluorescent lipid probes. Hepatic hypertrophy was observed in the model, along with macrovesicular steatosis. Smil/Lstr combo administration effectively restored the lipid profile and alleviated hepatic hypertrophy in the HFD zebrafish. In oleic-acid stimulated hepatocytes, Smil/Lstr combo markedly reduced lipid accumulation and cell damage. Subsequently, based on zebrafish phenotypic screening, the natural phthalide senkyunolide I (SEI) was identified as a major molecule mediating the lipid-lowering activities of Smil/Lstr combo in the liver. Moreover, SEI upregulated the expression of the lipid metabolism regulator PPARα and downregulated fatty acid translocase CD36, while a PPARα antagonist sufficiently blocked the regulatory effect of SEI on hepatic steatosis. Finally, the roles of SEI on hepatic lipid accumulation and PPARα signaling were further verified in the hamster model. CONCLUSIONS We proposed a zebrafish-based screening strategy for modulators of hepatic steatosis and discovered the regulatory roles of Smil/Lstr combo and its component SEI on liver lipid accumulation and PPARα signaling, suggesting their potential value as novel candidates for NAFLD treatment.
Collapse
Affiliation(s)
- Qingquan Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jian Sheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Mirko Baruscotti
- Department of Biosciences, University of Milano, Milan, 1-20133, Italy
| | - Zhenjie Liu
- Department of Vascular Surgery, The Second Affiliated Hospital of Zhejiang University Medical School, Hangzhou, 310003, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310020, China
| | - Lu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Department of Vascular Surgery, The Second Affiliated Hospital of Zhejiang University Medical School, Hangzhou, 310003, China; State Key Laboratory of Chinese Medicine Modernization, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
37
|
Hu M, Yang J, Gao B, Wu Z, Wu Y, Hu D, Shen Q, Chen L. Prediction of MASLD using different screening indexes in Chinese type 2 diabetes mellitus. Diabetol Metab Syndr 2025; 17:10. [PMID: 39780236 PMCID: PMC11716454 DOI: 10.1186/s13098-024-01571-x] [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: 09/21/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Formerly known as non-alcoholic fatty liver disease (NAFLD), metabolic dysfunction-associated steatotic liver disease (MASLD) has now become the most widespread chronic liver disease worldwide. The primary goal of this study is to assess the ability of different indexes (including VAI, TyG, HOMA-IR, BMI, LAP, WHtR, TyG-BMI, TyG-WC, and TyG-WHtR) to predict MASLD in individuals diagnosed with type 2 diabetes mellitus (T2DM), particularly within the Chinese population. METHODS This cross-sectional study involved 1,742 patients with T2DM, recruited from the Metabolic Management Centers (MMC) at Suzhou Municipal Hospital. Abdominal ultrasonography was employed for MASLD diagnosis in patients with T2DM. The predictive accuracy of various screening indexes for MASLD in the Chinese T2DM population was evaluated using logistic regression and receiver operating characteristic (ROC) curve analyses. RESULTS Among the 1,742 participants, 996 were diagnosed with MASLD. After adjusting for potential confounding factors, positive associations with the risk of MASLD were found for all the nine indexes. The lipid accumulation product (LAP) exhibited the greatest predictive value for detecting MASLD, with an area under the curve (AUC) of 0.786(95%CI 0.764,0.807), followed by BMI(AUC = 0.785), VAI(AUC = 0.744), TyG(AUC = 0.720), WHtR(AUC = 0.710) and HOMA-IR(AUC = 0.676). The composite Indexes (TyG-BMI, TyG-WC, TyG-WHtR) also showed considerable predictive ability with AUCs of 0.765, 0.752 and 0.748, respectively. CONCLUSION Our results indicated that all nine indexes have favorable correlations with the risk of MASLD, and most of them have a good performance in predicting MASLD. According to our study, LAP was a reliable index for predicting MASLD among Chinese T2DM patients. The exploration of non-invasive screenings will provide significant support for the early detection and diagnosis of MASLD.
Collapse
Affiliation(s)
- Mengmeng Hu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 126 Daoqian Street, Suzhou, 215000, China
| | - Jingyu Yang
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 126 Daoqian Street, Suzhou, 215000, China
| | - Beibei Gao
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 126 Daoqian Street, Suzhou, 215000, China
| | - Zhoulu Wu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 126 Daoqian Street, Suzhou, 215000, China
| | - Ying Wu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 126 Daoqian Street, Suzhou, 215000, China
| | - Dandan Hu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 126 Daoqian Street, Suzhou, 215000, China
| | - Qiong Shen
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 126 Daoqian Street, Suzhou, 215000, China
| | - Lei Chen
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 126 Daoqian Street, Suzhou, 215000, China.
| |
Collapse
|
38
|
Su S, Liu X, Zhu M, Liu W, Liu J, Yuan Y, Fu F, Rao Z, Liu J, Lu Y, Chen Y. Trehalose Ameliorates Nonalcoholic Fatty Liver Disease by Regulating IRE1α-TFEB Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:521-540. [PMID: 39680632 DOI: 10.1021/acs.jafc.4c08669] [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: 12/18/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), characterized by hepatic lipid deposition, is one of the most prevalent chronic metabolic disorders globally, and its pharmaceutical treatments are still limited. Excessive lipid accumulation triggers endoplasmic reticulum (ER) stress and autophagy flux dysfunction, which are important mechanisms for NAFLD. Trehalose (Tre), a natural disaccharide, has been identified to reduce hepatic steatosis and glucose intolerance. However, its underlying mechanisms for NAFLD remain unclear. In this study, a high-fat-diet (HFD)-induced mouse NAFLD model and a saturated fatty acid palmitic acid (PA)-stimulated cell model were constructed. The results indicated that Tre supplementation ameliorated hepatocyte lipid deposition in vitro, as well as hepatic steatosis and hyperlipidemia in vivo. Mechanistically, Tre alleviated both autophagy flux dysfunction and endoplasmic reticulum (ER) stress. Under the stimulation of HFD or PA, Tre remarkably increased the expression and nucleic translocation of the lysosomal master protein transcription factor EB (TFEB), while decreasing the accumulation of p62 and also decreasing the ER stress markers (inositol-requiring enzyme 1 (IRE1α), XBP-1, CHOP, and BIP). Similar results were observed in an ER stressor tunicamycin (TM)-induced in vivo and in vitro models. In addition, the transcriptomic analysis of NAFLD patients revealed significant differences in ER stress-related and autophagy-related biomarkers, including TFEB, ATG7, IRE1α, and CHOP. Molecular docking results demonstrated a strong affinity between Tre and both IRE1α and TFEB. Overall, Tre protected hepatocytes from lipotoxicity-related ER stress and autophagy dysfunction, and its regulatory effect on the IRE1α-TFEB signaling pathway may be a critical mechanism. These findings suggest that Tre, as a bioactive substance with significant medicinal potential, holds considerable promise for drug development and clinical application in treating NAFLD.
Collapse
Affiliation(s)
- Shan Su
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, P. R. China
| | - Xiaohong Liu
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Min Zhu
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Wen Liu
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Jingyi Liu
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Yujia Yuan
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Fudong Fu
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, P. R. China
| | - Zhiyong Rao
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Jingping Liu
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Yanrong Lu
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Younan Chen
- Department of Clinical Nutrition and National Health Commission (NHC) Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, P. R. China
| |
Collapse
|
39
|
Zhao H, Zhou J, Yuan L, Sun Z, Liu Y, Zhao X, Ye F. Exploring the alleviating effects of Bifidobacterium metabolite lactic acid on non-alcoholic steatohepatitis through the gut-liver axis. Front Microbiol 2025; 15:1518150. [PMID: 39850131 PMCID: PMC11756523 DOI: 10.3389/fmicb.2024.1518150] [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: 10/29/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Objective This study investigates the protective effects of lactic acid, a metabolite of Bifidobacterium, on non-alcoholic fatty liver disease (NAFLD) induced by a high-sugar, high-fat diet (HFD) in mice, in the context of the gut-liver axis. Methods A NAFLD mouse model was established using a HFD, and different intervention groups were set up to study the protective effects of Bifidobacterium and its metabolite lactic acid. The groups included a control group, NAFLD group, Bifidobacterium treatment group, Glyceraldehyde-3-P (G-3P) co-treatment group, and NOD-like receptor family pyrin domain containing 3 (NLRP3) overexpression group. The evaluation of liver function and lipid metabolism was conducted using the liver-to-body weight ratio, histological staining, and biochemical assays. Enzyme-linked immunosorbent assay (ELISA) was performed to measure inflammatory cytokines, and western blotting was used to analyze the expression of NLRP3 inflammasome and autophagy-related molecules. In vitro, an NAFLD cell model was established using oleic acid, with cells treated with lactic acid and NLRP3 overexpression to assess lipid droplet accumulation and inflammation. Results In vivo findings indicated that, in comparison to CBX group (Control group without antibiotic treatment), NAFLD/CBX group (NAFLD group without antibiotic administration) and NAFLD/ABX group (NAFLD group with antibiotic administration) exhibited increased liver-to-body weight ratio, higher lipid droplet accumulation, aggravated liver histopathological damage, and elevated levels of AST (Aspartate Aminotransferase), ALT (Alanine Aminotransferase), TC (Total Cholesterol), TG (Triglycerides), LDL-C (Low-Density Lipoprotein Cholesterol), IL-6 (Interleukin-6), TNF-α (Tumor Necrosis Factor-alpha), IL-1β (Interleukin-1 beta), and NLRP3-related molecules, while HDL-C (High-Density Lipoprotein Cholesterol) levels significantly decreased. Intervention with Bifidobacterium significantly reversed these adverse changes. Further addition of G-3P led to more pronounced improvement in NAFLD symptoms, while overexpression of NLRP3 weakened the protective effects of Bifidobacterium. In vitro results indicated that Ole group exhibited heightened lipid droplet accumulation and expression of NLRP3 inflammasome-related molecules relative to the control group. Treatment with lactic acid effectively reversed these changes; however, the protective effect of lactic acid was significantly weakened with NLRP3 overexpression. Conclusion Lactic acid can alleviate lipid metabolism disorders in NAFLD induced by diet through the inhibition of inflammation mediated by the NLRP3 inflammasome and the regulation of the autophagy process.
Collapse
Affiliation(s)
- Hongmei Zhao
- Department of Infectious Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Gastroenterology and Nutrition, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha, China
| | - Juan Zhou
- Department of Gastroenterology and Nutrition, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha, China
| | - Lingzhi Yuan
- Department of Gastroenterology and Nutrition, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha, China
| | - Zhiyi Sun
- University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yi Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyu Zhao
- Department of Pediatrics, Changsha County Maternal and Child Health Hospital, Changsha, China
| | - Feng Ye
- Department of Infectious Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
40
|
Zhang X, Zheng Y, Yang J, Yang Y, He Q, Xu M, Long F, Yang Y. Abnormal ac4C modification in metabolic dysfunction associated steatotic liver cells. Sci Rep 2025; 15:1013. [PMID: 39762452 PMCID: PMC11704021 DOI: 10.1038/s41598-024-84564-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
The pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) remains unclear due to the complexity of its etiology. The emerging field of the epitranscriptome has shown significant promise in advancing the understanding of disease pathogenesis and developing new therapeutic approaches. Recent research has demonstrated that N4-acetylcytosine (ac4C), an RNA modification within the epitranscriptome, is implicated in progression of various diseases. However, the role of ac4C modification in MASLD remains unexplored. Herein, we performed acRIP-ac4c-seq and RNA-seq analysis in free fatty acids-induced MASLD model cells, identifying 2128 differentially acetylated ac4C sites, with 1031 hyperacetylated and 1097 hypoacetylated peaks in MASLD model cells. Functional enrichments analysis showed that ac4C differentially modified genes were significantly involved in processes related to MASLD, such as nuclear transport and MAP kinase (MAPK) signaling pathway. We also identified 341 differentially expressed genes (DEGs), including 61 lncRNAs and 280 mRNAs, between control and MASLD model cells. Bioinformatics analysis showed that DEGs were significantly enriched in long-chain fatty acid biosynthetic process. Notably, 118 genes exhibited significant changes in both ac4C modification and expression levels in MASLD model cells. Among these proteins, JUN, caveolin-1 (CAV1), fatty acid synthase (FASN), and heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) were identified as core proteins through protein-protein interaction (PPI) network analysis using cytoscape software. Collectively, our findings establish a positive correlation between ac4C modification and the pathogenesis of MASLD and suggest that ac4C modification may serve as a therapeutic target for MASLD.
Collapse
Affiliation(s)
- Xiqian Zhang
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Yaxian Zheng
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Jing Yang
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Yan Yang
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Qin He
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Min Xu
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610041, China.
| | - Yujie Yang
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China.
| |
Collapse
|
41
|
Iturbe-Rey S, Maccali C, Arrese M, Aspichueta P, Oliveira CP, Castro RE, Lapitz A, Izquierdo-Sanchez L, Bujanda L, Perugorria MJ, Banales JM, Rodrigues PM. Lipotoxicity-driven metabolic dysfunction-associated steatotic liver disease (MASLD). Atherosclerosis 2025; 400:119053. [PMID: 39581063 DOI: 10.1016/j.atherosclerosis.2024.119053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/19/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a spectrum of liver lesions, ranging from simple steatosis to metabolic dysfunction-associated steatohepatitis (MASH), that may further progress to cirrhosis. MASLD is estimated to affect more than one third of the general population and it represents a risk factor for end-stage liver failure and liver cancer, substantially contributing to liver-related morbidity and mortality. Although the pathogenesis of MASLD is incompletely understood, it is known to consist of a multifactorial process influenced by extrinsic and intrinsic factors such as metabolic, environmental and demographic features, gut microbiota and genetics. Dysregulation of both extracellular and intracellular lipid composition is known to promote the generation of toxic lipid species, thereby triggering lipotoxicity and cellular stress. These events ultimately lead to the activation of distinct cell death pathways, resulting in inflammation, fibrogenesis and, eventually, carcinogenesis. In this manuscript, we provide a comprehensive review of the role of lipotoxicity during MASLD pathogenesis, discussing the most relevant lipid species and related molecular mechanisms, summarizing the cell type-specific effects and highlighting the most promising putative therapeutic strategies for modulating lipotoxicity and lipid metabolism in MASLD.
Collapse
Affiliation(s)
- Santiago Iturbe-Rey
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Claudia Maccali
- Clinical and Experimental Gastroenterology Laboratory LIM-07, Department of Gastroenterology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marco Arrese
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, 8330077, Chile
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain; Biobizkaia Health Research Institute, Cruces University Hospital, 48903, Barakaldo, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain
| | - Claudia P Oliveira
- Clinical and Experimental Gastroenterology Laboratory LIM-07, Department of Gastroenterology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Division of Clinical Gastroenterology and Hepatology, Hospital das Clínicas de São Paulo, HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Rui E Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ainhoa Lapitz
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; Department of Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| | - Pedro M Rodrigues
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
42
|
Khare T, Liu K, Chilambe LO, Khare S. NAFLD and NAFLD Related HCC: Emerging Treatments and Clinical Trials. Int J Mol Sci 2025; 26:306. [PMID: 39796162 PMCID: PMC11720452 DOI: 10.3390/ijms26010306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently renamed metabolic-associated fatty liver disease (MAFLD), is the most prevalent liver disease worldwide. It is associated with an increased risk of developing hepatocellular carcinoma (HCC) in the background of cirrhosis or without cirrhosis. The prevalence of NAFLD-related HCC is increasing all over the globe, and HCC surveillance in NAFLD cases is not that common. In the present review, we attempt to summarize promising treatments and clinical trials focused on NAFLD, nonalcoholic steatohepatitis (NASH), and HCC in the past five to seven years. We categorized the trials based on the type of intervention. Most of the trials are still running, with only a few completed and with conclusive results. In clinical trial NCT03942822, 25 mg/day of milled chia seeds improved NAFLD condition. Completed trial NCT03524365 concluded that Rouxen-Y gastric bypass (RYGB) or sleeve gastrectomy (SG) results in histological resolution of NASH without worsening of fibrosis, while NCT04677101 validated sensitivity/accuracy of blood biomarkers in predicting NASH and fibrosis stage. Moreover, trials with empagliflozin (NCT05694923), curcuvail (NCT06256926), and obeticholic acid (NCT03439254) were completed but did not provide conclusive results. However, trial NCT03900429 reported effective improvement in fibrosis by at least one stage, without worsening of NAFLD activity score (NAS), as well as improvement in lipid profile of the NASH patients by 80 or 100 mg MGL-3196 (resmetirom). Funded by Madrigal Pharmaceuticals, Rezdiffra (resmetirom), used in the clinical trial NCT03900429, is the first FDA-approved drug for the treatment of NAFLD/NASH.
Collapse
Affiliation(s)
- Tripti Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Harry S Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Karina Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA;
| | | | - Sharad Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Harry S Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| |
Collapse
|
43
|
Ling L, Li R, Xu M, Zhou J, Hu M, Zhang X, Zhang XJ. Species differences of fatty liver diseases: comparisons between human and feline. Am J Physiol Endocrinol Metab 2025; 328:E46-E61. [PMID: 39636211 DOI: 10.1152/ajpendo.00014.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most widespread chronic liver disease that poses significant threats to public health due to changes in dietary habits and lifestyle patterns. The transition from simple steatosis to nonalcoholic steatohepatitis (NASH) markedly increases the risk of developing cirrhosis, hepatocellular carcinoma, and liver failure in patients. However, there is only one Food and Drug Administration-approved therapeutic drug in the world, and the clinical demand is huge. There is significant clinical heterogeneity among patients with NAFLD, and it is challenging to fully understand human NAFLD using only a single animal model. Interestingly, felines, like humans, are particularly prone to spontaneous fatty liver disease. This review summarized and compared the etiology, clinical features, pathological characteristics, and molecular pathogenesis between human fatty liver and feline hepatic lipidosis (FHL). We analyzed the key similarities and differences between those two species, aiming to provide theoretical foundations for developing effective strategies for the treatment of NAFLD in clinics.
Collapse
Affiliation(s)
- Like Ling
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Ruilin Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Mengqiong Xu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Junjie Zhou
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Manli Hu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Xin Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Xiao-Jing Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
44
|
Soares De Oliveira L, Ritter MJ. Thyroid hormone and the Liver. Hepatol Commun 2025; 9:e0596. [PMID: 39699315 PMCID: PMC11661762 DOI: 10.1097/hc9.0000000000000596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/20/2024] Open
Abstract
It is known that thyroid hormone can regulate hepatic metabolic pathways including cholesterol, de novo lipogenesis, fatty acid oxidation, lipophagy, and carbohydrate metabolism. Thyroid hormone action is mediated by the thyroid hormone receptor (THR) isoforms and their coregulators, and THRβ is the main isoform expressed in the liver. Dysregulation of thyroid hormone levels, as seen in hypothyroidism, has been associated with dyslipidemia and metabolic dysfunction-associated fatty liver disease. Given the beneficial effects of thyroid hormone in liver metabolism and the advances illuminating the use of thyroid hormone analogs such as resmetirom as therapeutic agents in the treatment of metabolic dysfunction-associated fatty liver disease, this review aims to further explore the relationship between TH, the liver, and metabolic dysfunction-associated fatty liver disease. Herein, we summarize the current clinical therapies and highlight future areas of research.
Collapse
|
45
|
Tang L, Zhu J, Zhuge S, Yu J, Jiang G. Perfluorooctane sulfonate induces hepatotoxicity through promoting inflammation, cell death and autophagy in a rat model. J Toxicol Sci 2025; 50:45-55. [PMID: 39894534 DOI: 10.2131/jts.50.45] [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: 02/04/2025]
Abstract
Perfluorooctane sulfonate (PFOS) is reported to cause hepatotoxicity in animals and humans. However, the underlying mechanism by which it affects organelle toxicity in the liver are not well elucidated yet. This study aimed to investigate the mechanisms underlying PFOS-induced hepatic toxicity, focusing on inflammation, cell death, and autophagy. We established a PFOS-exposed Sprague-Dawley (SD) rat liver injury model by intraperitoneal injection of PFOS (1 mg/kg and 10 mg/kg body weight) every alternate day for 15 days. Our findings indicated that PFOS increased liver weight, caused lipid disorder and hepatic steatosis in rats. Meanwhile, PFOS disrupted the structure of mitochondria, increased accumulation of reactive oxygen species (ROS), repressed superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) levels, and elevated malondialdehyde (MDA) and nitric oxide synthase (NOS) amounts. We found PFOS induced inflammation as evidenced by activation of NOD-like receptor protein 3 (NLRP3), Cleaved cysteine-aspartic acid protease (caspase)1, tumor necrosis factor (TNF)α and interleukin (IL)-1β levels. Moreover, PFOS exposure significantly decreased B-cell lymphoma2 (Bcl2)/Bcl2 associated X (Bax) ratio and increased the protein expression of Cleaved caspase-3. Compared with the control group, PFOS upregulated the protein expression of necroptotic markers and autophagy-related proteins. In conclusion, PFOS induced inflammation, cell death, and autophagy through oxidative stress by ROS overload, thereby providing a mechanistic explanation for PFOS-induced hepatotoxicity.
Collapse
Affiliation(s)
- Leilei Tang
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, China
| | - Jianjun Zhu
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, China
| | - Sheng Zhuge
- Department of Surgery, The First People's Hospital of Yuhang District
| | - Jiawen Yu
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, China
| | - Guojun Jiang
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, China
| |
Collapse
|
46
|
Asikaer A, Sun C, Shen Y. Thiostrepton: multifaceted biological activities and its applications in treatment of inflammatory diseases. Inflammopharmacology 2025; 33:183-194. [PMID: 39487942 DOI: 10.1007/s10787-024-01587-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
Thiostrepton (TST) is a naturally occurring oligopeptide antibiotic with a demonstrated capacity to antagonize a broad spectrum of Gram-positive bacteria. It has been utilized as a safe antimicrobial agent in veterinary medicine. Despite its therapeutic potential, the clinical application of TST has been constrained by its poor solubility and bioavailability. However, an increasing number of studies indicate that TST possesses diverse pharmacological activities, including its significant role in microbe resistance and cancer countering. Notably, recent studies have pointed out that TST also possesses anti-inflammatory potential. It has exhibited promising therapeutic efficacy across various in vivo and in vitro disease models, including non-alcoholic fatty liver disease, inflammatory bowel disease, sepsis, psoriasis-like inflammation, and periodontitis. In this review, we describe the various pharmacological activities of TST, particularly its anti-inflammatory activity demonstrated in a variety of inflammatory diseases and the underlying mechanisms. These effects highlight the potential of TST as an anti-inflammatory agent for the treatment of inflammation diseases and for enhancing cellular therapies.
Collapse
Affiliation(s)
- Aiminuer Asikaer
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, PR, China
| | - Cai Sun
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, PR, China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, PR, China.
| |
Collapse
|
47
|
Schütz F, Longo L, Keingeski MB, Filippi-Chiela E, Uribe-Cruz C, Álvares-da-Silva MR. Lipophagy and epigenetic alterations are related to metabolic dysfunction-associated steatotic liver disease progression in an experimental model. World J Hepatol 2024; 16:1468-1479. [DOI: 10.4254/wjh.v16.i12.1468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Genetic and epigenetic alterations are related to metabolic dysfunction-associated steatotic liver disease (MASLD) pathogenesis.
AIM To evaluate micro (mi)RNAs and lipophagy markers in an experimental model of metabolic dysfunction-associated steatohepatitis (MASH).
METHODS Adult male Sprague Dawley rats were randomized into two groups: Control group (n = 10) fed a standard diet; and intervention group (n = 10) fed a high-fat-choline-deficient diet for 16 weeks. Molecular evaluation of lipophagy markers in liver tissue [sirtuin-1, p62/sequestosome-1, transcription factor-EB, perilipin-2 (Plin2), Plin3, Plin5, lysosome-associated membrane proteins-2, rubicon, and Cd36], and serum miRNAs were performed.
RESULTS Animals in the intervention group developed MASH and showed a significant decrease in sirtuin-1 (P = 0.020) and p62/sequestosome-1 (P < 0.001); the opposite was reported for transcription factor-EB (P = 0.020), Plin2 (P = 0.003), Plin3 (P = 0.031), and Plin5 (P = 0.005) compared to the control group. There was no significant difference between groups for lysosome-associated membrane proteins-2 (P = 0.715), rubicon (P = 0.166), and Cd36 (P = 0.312). The intervention group showed a significant increase in miR-34a (P = 0.005) and miR-21 (P = 0.043) compared to the control. There was no significant difference between groups for miR-375 (P = 0.905), miR-26b (P = 0.698), and miR-155 (P = 0.688).
CONCLUSION Animals with MASH presented expression changes in markers related to lysosomal stress and autophagy as well as in miRNAs related to inflammation and fibrogenesis, processes that promote MASLD progression.
Collapse
Affiliation(s)
- Felipe Schütz
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
| | - Larisse Longo
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
| | - Melina Belén Keingeski
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
| | - Eduardo Filippi-Chiela
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Rio Grande do Sul, Brazil
- Department of Morphological Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Rio Grande do Sul, Brazil
| | - Carolina Uribe-Cruz
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
- Facultad de Ciencias de la Salud, Universidad Católica de las Misiones, Posadas 3300, Misiones, Argentina
| | - Mário Reis Álvares-da-Silva
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-007, Rio Grande do Sul, Brazil
- Conselho Nacional de Desenvolvimento Científico e Tecnológico, Brasilia 71.605-001, Distrito Federal, Brazil
| |
Collapse
|
48
|
Ramoni D, Liberale L, Montecucco F. Inflammatory biomarkers as cost-effective predictive tools in metabolic dysfunction-associated fatty liver disease. World J Gastroenterol 2024; 30:5086-5091. [PMID: 39713167 PMCID: PMC11612858 DOI: 10.3748/wjg.v30.i47.5086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/01/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Qu and Li emphasize a fundamental aspect of metabolic dysfunction-associated fatty liver disease in their manuscript, focusing on the critical need for non-invasive diagnostic tools to improve risk stratification and predict the progression to severe liver complications. Affecting approximately 25% of the global population, metabolic dysfunction-associated fatty liver disease is the most common chronic liver condition, with higher prevalence among those with obesity. This letter stresses the importance of early diagnosis and intervention, especially given the rising incidence of obesity and metabolic syndrome. Research advancements provide insight into the potential of biomarkers (particularly inflammation-related) as predictive tools for disease progression and treatment response. This overview addresses pleiotropic biomarkers linked to chronic inflammation and cardiometabolic disorders, which may aid in risk stratification and treatment efficacy monitoring. Despite progress, significant knowledge gaps remain in the clinical application of these biomarkers, necessitating further research to establish standardized protocols and validate their utility in clinical practice. Understanding the complex interactions among these factors opens new avenues to enhance risk assessment, leading to better patient outcomes and addressing the public health burden of this worldwide condition.
Collapse
Affiliation(s)
- Davide Ramoni
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- Department of Internal Medicine, First Clinic of Internal Medicine, IRCCS Ospedale Policlinico San Martino, Italian Cardiovascular Network, Genoa 16132, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- Department of Internal Medicine, First Clinic of Internal Medicine, IRCCS Ospedale Policlinico San Martino, Italian Cardiovascular Network, Genoa 16132, Italy
| |
Collapse
|
49
|
Jiang S, Fan X, Hua J, Liu S, Feng Y, Shao D, Shen Y, Wang Z, Yan X, Wang J. Integrated metabolomics and network pharmacology analysis to reveal the protective effect of Complanatoside A on nonalcoholic fatty liver disease. Eur J Pharmacol 2024; 985:177074. [PMID: 39481627 DOI: 10.1016/j.ejphar.2024.177074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
INTRODUCTION The rising prevalence and severe consequences of nonalcoholic fatty liver disease (NAFLD) have driven the quest for preventive medications. Complanatoside A (CA) is the marked flavonoid of Astragali complanati semen, a traditional Chinese herb that acts on the liver meridian and is widely used to treat liver problems. CA has been proven to have considerable lipid-lowering and liver-protective effects in vitro. However, the efficacy of CA in preventing NAFLD has yet to be shown in vivo. METHODS First, the effectiveness of CA against NAFLD was assessed using a high-fat diet (HFD) mouse model. Second, the CA protective mechanism against NAFLD was investigated using a combined metabolomics and network pharmacology strategy. Differential metabolites were identified by metabolomics-based analyses, and metabolic pathway analysis was accomplished by MetaboAnalyst. Potential therapeutic targets were obtained through network pharmacology. Finally, key targets were identified via compound-target networks and validated by molecular docking and western blotting. RESULTS CA prevented NAFLD mainly by reducing liver lipid accumulation in HFD mice. Metabolomics identified 22 potential biomarkers for CA treatment of NAFLD, primarily involving glycerophospholipid and arachidonic acid metabolism. Fifty-one potential targets were determined by network pharmacology. Co-analysis revealed that albumin, peroxisome proliferator-activated receptor-alpha, retinoid X receptor alpha, interleukin-6, and tumor necrosis factor alpha were key targets. CONCLUSION This experiment revealed that CA has a preventive effect on NAFLD, primarily by regulating the peroxisome proliferator-activated receptor-alpha/retinoid X receptor alpha pathway. Furthermore, it provides evidence supporting the potential use of CA in the long-term prevention of NAFLD.
Collapse
Affiliation(s)
- Sijia Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Xiaoxu Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Jian Hua
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Shuangqiao Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Yingtong Feng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Danyue Shao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Yiwei Shen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Zhen Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Xuehua Yan
- Institute of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China.
| | - Jingxia Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 102488, Beijing, China.
| |
Collapse
|
50
|
Li F, Xie R, Li T, Ren S. Erhong Jiangzhi Decoction Inhibits Lipid Accumulation and Alleviates Nonalcoholic Fatty Liver Disease with Nrf2 Restoration Under Obesity. J Inflamm Res 2024; 17:10929-10942. [PMID: 39677297 PMCID: PMC11646429 DOI: 10.2147/jir.s491484] [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: 09/18/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) refers to the liver pathological changes caused by excessive fat accumulation in hepatocytes owing to various reasons, which has become an emerging health challenge. Erhong Jiangzhi Decoction (EHJD) is a traditional Chinese medicine decoction. This study aims to investigate the therapeutic effect of EHJD on NAFLD. Methods NAFLD model was constructed by high-fat diet (HFD)-induced mice and oleic acid-induced HepG2 cells. Mice were intragastrically administered with EHJD and HepG2 cells were treated with EHJD drug-containing serum. The effects of EHJD on NAFLD were explored in vivo and in vitro. Histological assessment was performed by hematoxylin-eosin and oil red O staining. ELISA was exploited to detect the expression of lipid accumulation, liver function, inflammation, and oxidative stress related indicators. The expression of Nrf2/HO-1 pathway was detected by qRT-PCR and Western blot. Results In HFD-induced NAFLD mice, the body weight was increased, and liver/weight, inguinal fat/weight, and epididymal fat/weight were higher, while EHJD reduced them. Staining results exhibited that EHJD decreased inflammatory cell infiltration and oil red lipid droplets in HFD-induced mice. In addition, EHJD treatment suppressed TC, TG, ALT and AST levels; TNF-α, IL-1β, IL-6 and MDA levels were inhibited by EHJD, while GSH-Px, CAT and T-AOC levels were increased in NAFLD through the in vivo and in vitro experiments. The suppression of Nrf2 weakened the inhibitory effect of EHJD on lipid metabolism, liver injury, inflammation and oxidative stress. Conclusion EHJD had a protective effect on NAFLD by alleviating lipid accumulation and liver injury, inhibiting inflammation, and oxidative stress, which was achieved by the restoration of Nrf2.
Collapse
Affiliation(s)
- Fang Li
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, People’s Republic of China
| | - Rong Xie
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, People’s Republic of China
| | - Tianlun Li
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, People’s Republic of China
| | - Shouzhong Ren
- College of Pharmacy, Hainan Medical University, HaiKou City, Hainan Province, People’s Republic of China
| |
Collapse
|