1
|
Zhang L, Fang L, Zou J, Zhou D, Xie H, Chen A, Wu Q. Causal associations of metabolic dysfunction-associated steatotic liver disease with gestational hypertension and preeclampsia: a two-sample Mendelian randomization study. Hypertens Pregnancy 2025; 44:2441862. [PMID: 39704480 DOI: 10.1080/10641955.2024.2441862] [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: 06/26/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Hypertensive disorders of pregnancy (HDPs), which include gestational hypertension (GH) and preeclampsia (PE), are the primary causes of maternal morbidity and mortality worldwide. Recent studies have found a correlation between metabolic dysfunction-associated steatotic liver disease (MASLD) and HDPs, but the causality of this association remains to be identified. Therefore, this study aims to evaluate the causal relationship between MASLD and HDPs through Mendelian randomization (MR) analysis. METHODS The summary statistics from genome-wide association studies were employed to conduct a two-sample MR analysis. Five complementary MR methods, including inverse variance weighting (IVW), MR-Egger, weighted median, simple mode and weighted mode were performed to assess the causality of MASLD on GH and PE. Furthermore, we conducted various sensitivity analyses to ensure the stability and reliability of the results. RESULTS Genetically predicted MASLD significantly increased the risk of GH (IVW: OR = 1.138, 95% CI: 1.062-1.220, p < 0.001), while there was little evidence of a causal relationship between MASLD and PE (IVW: OR = 0.980, 95% CI: 0.910-1.056, p = 0.594). The sensitivity analyses indicated no presence of heterogeneity and horizontal pleiotropy. CONCLUSION This MR study provided evidence supporting the causal effect of MASLD on GH. Our findings underscore the significance of providing more intensive prenatal care and early intervention for pregnant women with MASLD to prevent potential adverse obstetric outcomes.
Collapse
Affiliation(s)
- Lu Zhang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Liang Fang
- Department of Gastroenterology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Jiahua Zou
- Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Dong Zhou
- Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Haonan Xie
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Aihua Chen
- Department of Gastroenterology, China Resources & WISCO General Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Qingming Wu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Wan L, Li L, Zhang X, Li B, Brao KJ, Harro JM, Shirtliff ME. Evaluating palm olein and palm stearin on hepatic lipids in C57BL/6 J mice under low and high fat intakes: A quantitative lipidomic approach. Food Chem 2025; 484:144326. [PMID: 40252450 DOI: 10.1016/j.foodchem.2025.144326] [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: 11/30/2024] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
This work aims to study the effects of dietary lipid content and composition on hepatic lipids of mice fed palm olein (PO)-based low-fat diet, PO-based high-fat diet, palm stearin (PS)-based low-fat diet, or PS-based high-fat diet for 8 weeks. Results showed that high PO and PS intake significantly elevated serum LDL-c and TCHO levels and hepatic TG content without altering the expression levels of genes involved in DNL. Overall, quantitative lipidomics revealed that accumulated lipids were polyunsaturated PE and saturated & monounsaturated SM/Cer_NS in response to high PO intake. While high PS intake led to increased levels of polyunsaturated PC and mono- & polyunsaturated TG. These enriched lipids may lead to disturbed hepatic PUFA metabolism. When lipid intake was constant, only under high fat intake did PS show minor effects on hepatic lipids. These results highlighted the role of lipid quantity instead of lipid quality on hepatic lipid profiles.
Collapse
Affiliation(s)
- Liting Wan
- School of Food Science and Technology, Guangdong Provincial Key Laboratory of Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou 510640, China; Department of Microbial Pathogenesis, School of Dentistry, University of Maryland - Baltimore, Baltimore, MD 21201, USA
| | - Lin Li
- School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan 523808, China
| | - Xia Zhang
- School of Food Science and Technology, Guangdong Provincial Key Laboratory of Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou 510640, China
| | - Bing Li
- School of Food Science and Technology, Guangdong Provincial Key Laboratory of Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou 510640, China.
| | - Kristen J Brao
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland - Baltimore, Baltimore, MD 21201, USA
| | - Janette M Harro
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland - Baltimore, Baltimore, MD 21201, USA.
| | - Mark E Shirtliff
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland - Baltimore, Baltimore, MD 21201, USA; Department of Microbiology and Immunology, School of Medicine, University of Maryland - Baltimore, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
Lee YH, Lee J, Jeong J, Park K, Baik B, Kwon Y, Kim K, Khim KW, Ji H, Lee JY, Kim K, Kim JW, Dao T, Kim M, Lee TY, Yang YR, Yoon H, Ryu D, Hwang S, Lee H, Nam D, Kim WK, Park NH, Yun H, Choi JH. Hepatic miR-93 promotes the pathogenesis of metabolic dysfunction-associated steatotic liver disease by suppressing SIRT1. Metabolism 2025; 169:156266. [PMID: 40228656 DOI: 10.1016/j.metabol.2025.156266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 04/05/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND AND AIMS The molecular mechanisms underlying metabolic dysfunction-associated steatotic liver disease (MASLD) remain largely unclear; however, emerging evidence suggests that microRNAs (miRNAs) play a critical role in modulating transcriptional regulation of target genes involved in MASLD. This study aims to elucidate the role of miR-93 in lipid metabolism and MASLD progression. METHODS We comprehensively analyzed miRNA expression profiles in liver tissues from patients with MASLD and diet-induced obese mice. miR-93 knockout (KO) mice were fed a high-fat-high-fructose (HFHFr) diet to assess the impact of miR-93 deficiency on MASLD. Transcriptome analysis was performed to elucidate the molecular mechanisms and role of miR-93 in MASLD. Additionally, we employed a high-throughput screening system to identify drugs capable of modulating miR-93 expression. RESULTS miR-93 was significantly upregulated in the livers of patients with MASLD and diet-induced obese mice. miR-93 KO mice exhibited reduced hepatic steatosis. Specifically, miR-93 deficiency upregulated genes involved in fatty acid oxidation and downregulated genes associated with cholesterol biosynthesis. Sirtuin 1 (SIRT1) was identified as a direct target of miR-93, and miR-93 KO enhanced SIRT1 expression and activated the LKB1-AMPK signaling pathway. Niacin treatment downregulated miR-93, ameliorating hepatic steatosis by enhancing SIRT1 activity. CONCLUSIONS These findings implicate miR-93 as a novel therapeutic target for MASLD. The study demonstrates the therapeutic potential of niacin in modulating the miR-93/SIRT1 axis, providing a new potential treatment for MASLD, a disease with limited current treatment options.
Collapse
Affiliation(s)
- Yo Han Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jinyoung Lee
- College of Pharmacy and Research Institute for Drug Development, Pusan National University (PNU), Busan 46241, Republic of Korea
| | - Joonho Jeong
- Division of Hepatology, Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital (UUH), Ulsan 44033, Republic of Korea
| | - Kieun Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Bukyung Baik
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Yuseong Kwon
- College of Pharmacy and Research Institute for Drug Development, Pusan National University (PNU), Busan 46241, Republic of Korea
| | - Kimyeong Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Keon Woo Khim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Haneul Ji
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Ji Young Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Kwangho Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Ji Won Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Tam Dao
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea; Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine (SKKU), Suwon 16419, Republic of Korea
| | - Misung Kim
- Department of Pathology, University of Ulsan College of Medicine, Ulsan University Hospital (UUH), Ulsan 44033, Republic of Korea
| | - Tae Young Lee
- Department of Radiology, University of Ulsan College of Medicine, Ulsan University Hospital (UUH), Ulsan 44033, Republic of Korea
| | - Yong Ryoul Yang
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Haejin Yoon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea; Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine (SKKU), Suwon 16419, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University (PNU), Busan 46241, Republic of Korea
| | - Haeseung Lee
- College of Pharmacy and Research Institute for Drug Development, Pusan National University (PNU), Busan 46241, Republic of Korea
| | - Dougu Nam
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Neung Hwa Park
- Division of Hepatology, Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital (UUH), Ulsan 44033, Republic of Korea.
| | - Hwayoung Yun
- College of Pharmacy and Research Institute for Drug Development, Pusan National University (PNU), Busan 46241, Republic of Korea.
| | - Jang Hyun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| |
Collapse
|
4
|
Ma X, Sun CY, Zhang Y, Li J, Zhao DS. The hepatoprotective effect of Lonicera japonica Flos on rats with high-fat diet-induced non-alcoholic fatty liver disease. Fitoterapia 2025; 183:106516. [PMID: 40188994 DOI: 10.1016/j.fitote.2025.106516] [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: 07/11/2024] [Revised: 03/18/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025]
Abstract
Lonicerae Japonica Flos (LJF) is an edible-medicinal herb, rich in phenolic acids, flavonoids, iridoids and other bioactive ingredients, that has anti-inflammatory, antioxidant, antilipemic, and hepatoprotective effects. However, its effect on non-alcoholic fatty liver disease (NAFLD) remains to be elucidated. The aim of this study was to determine the effect of LJF on liver injury in rats with high-fat diet (HFD)-induced NAFLD. The administration of LJF extract to rats with HFD-induced NAFLD significantly improved their body weight and daily food intake, liver tissue steatosis, lipid droplet vacuolization, and inflammatory cell infiltration. In addition, the LJF extract also improved to varying degrees the serum biochemical parameters, liver lipid content, levels of inflammatory factors, and oxidative stress markers. Among the treatment groups, the LJF high-dose group (LJF-H) showed the most significant improvement effect. Additionally, the correlation matrix heatmap visualization indicated that LJF may ameliorate NAFLD mainly by lowering liver lipid content and improving serum biochemical parameters.
Collapse
Affiliation(s)
- Xue Ma
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, No. 4655 Daxue Road, Jinan 250355, China
| | - Chun-Yong Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, No. 4655 Daxue Road, Jinan 250355, China
| | - Yan Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, No. 4655 Daxue Road, Jinan 250355, China
| | - Jia Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, No. 4655 Daxue Road, Jinan 250355, China.
| | - Dong-Sheng Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, No. 4655 Daxue Road, Jinan 250355, China.
| |
Collapse
|
5
|
Wang B, Yang Y, Yin Z, Yang W. The causal impact of body mass index on metabolic biomarkers and nonalcoholic fatty liver disease risk. Sci Rep 2025; 15:10314. [PMID: 40133380 PMCID: PMC11937590 DOI: 10.1038/s41598-024-84165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/20/2024] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a growing global health concern linked to obesity. METHODS This study employed a Mendelian randomization approach to explore the causal influence of BMI on metabolic biomarkers and the subsequent risk of NAFLD. We analyzed data from multiple sources, including 249 metabolic traits, to establish direct and mediating relationships among BMI, metabolic factors, and NAFLD risk. RESULTS Our findings revealed a significant positive correlation between BMI and NAFLD across various datasets. We identified 176 metabolites associated with BMI, of which 106 were also linked to NAFLD. Importantly, 86 metabolites were found to mediate the relationship between BMI and NAFLD risk. Specifically, elevated levels of branched-chain amino acids, triglycerides, and certain cholesterol esters were notably associated with increased NAFLD risk, whereas changes in free cholesterol and phospholipid levels also played critical roles. CONCLUSION This study highlights the complex interactions between BMI, metabolic biomarkers, and NAFLD risk. By elucidating these relationships, we highlight potential targets for interventions aimed at reducing NAFLD incidence in populations with elevated BMI, ultimately contributing to improved metabolic health.
Collapse
Affiliation(s)
- Bo Wang
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Yanjiang Yang
- Department of Rheumatology and Immunology, The People's Hospital of Qiandongnan Autonomous Prefecture, Kaili, 556000, Guizhou Province, China
| | - Zhaoqiang Yin
- Department of Minimally Invasive and Biliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China.
| | - Wenwen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
6
|
Xi J, Chen Y, Jie C, Law JCS, Fan Z, Lv G. Life's Crucial 9 and NAFLD from association to SHAP-interpreted machine learning predictions. Sci Rep 2025; 15:9384. [PMID: 40102489 PMCID: PMC11920226 DOI: 10.1038/s41598-025-92777-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease worldwide. Cardiovascular disease (CVD) and NAFLD share multiple common risk factors. Life's Crucial 9 (LC9), a novel indicator for comprehensive assessment of cardiovascular health (CVH), has not yet been studied in terms of its association with or predictive value for NAFLD. This study analyzed data from 10,197 participants in the National Health and Nutrition Examination Survey (NHANES) from 2007 to 2018. The association between LC9 and NAFLD was assessed using weighted logistic regression, while weighted Cox proportional hazards models were applied to evaluate the relationship between LC9 and all-cause mortality among NAFLD patients. Restricted cubic spline (RCS) analysis was conducted to explore dose-response relationships, and Kaplan-Meier survival curves were utilized to examine differences in survival outcomes. Machine learning (ML) approaches were employed to construct predictive models, with the optimal model further interpreted using SHapley Additive exPlanations (SHAP). An increase of 10 points in LC9 was negatively associated with the risk of NAFLD (model 3: OR = 0.39, 95% CI = 0.36 - 0.42, P < 0.001) and all-cause mortality in NAFLD patients (model 3: HR = 0.78, 95% CI = 0.67 - 0.91, P < 0.001). A non-linear relationship was observed between LC9 and NAFLD (P < 0.0001 for nonlinearity). Among the eight ML models, the Support Vector Machine (SVM) demonstrated the best predictive performance (AUC = 0.873). SHAP analysis indicated that LC9 was the most significant predictor in the model. LC9 demonstrated a nonlinear negative association with NAFLD and a linear negative association with all-cause mortality in NAFLD patients. Maintaining a higher LC9 score may reduce the risk of NAFLD and all-cause mortality among NAFLD patients. The predictive model developed using Support Vector Machine (SVM) exhibited strong clinical predictive value, with LC9 being the most critical factor in the model, facilitating self-risk assessment and targeted intervention.
Collapse
Affiliation(s)
- Jianxin Xi
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, No.1 Xinmin Street, Chaoyang District, Changchun City, Jilin Province, China
| | - Yuguo Chen
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, No.1 Xinmin Street, Chaoyang District, Changchun City, Jilin Province, China
| | - Chen Jie
- Department of Radiology, The First Hospital of Jilin University, Jilin, China
| | - Jason Chi Shing Law
- Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Zhongqi Fan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, No.1 Xinmin Street, Chaoyang District, Changchun City, Jilin Province, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, No.1 Xinmin Street, Chaoyang District, Changchun City, Jilin Province, China.
| |
Collapse
|
7
|
Chen R, Petrazzini BO, Duffy Á, Rocheleau G, Jordan D, Bansal M, Do R. Trans-ancestral rare variant association study with machine learning-based phenotyping for metabolic dysfunction-associated steatotic liver disease. Genome Biol 2025; 26:50. [PMID: 40065360 PMCID: PMC11892324 DOI: 10.1186/s13059-025-03518-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified common variants associated with metabolic dysfunction-associated steatotic liver disease (MASLD). However, rare coding variant studies have been limited by phenotyping challenges and small sample sizes. We test associations of rare and ultra-rare coding variants with proton density fat fraction (PDFF) and MASLD case-control status in 736,010 participants of diverse ancestries from the UK Biobank, All of Us, and BioMe and performed a trans-ancestral meta-analysis. We then developed models to accurately predict PDFF and MASLD status in the UK Biobank and tested associations with these predicted phenotypes to increase statistical power. RESULTS The trans-ancestral meta-analysis with PDFF and MASLD case-control status identifies two single variants and two gene-level associations in APOB, CDH5, MYCBP2, and XAB2. Association testing with predicted phenotypes, which replicates more known genetic variants from GWAS than true phenotypes, identifies 16 single variants and 11 gene-level associations implicating 23 additional genes. Two variants were polymorphic only among African ancestry participants and several associations showed significant heterogeneity in ancestry and sex-stratified analyses. In total, we identified 27 genes, of which 3 are monogenic causes of steatosis (APOB, G6PC1, PPARG), 4 were previously associated with MASLD (APOB, APOC3, INSR, PPARG), and 23 had supporting clinical, experimental, and/or genetic evidence. CONCLUSIONS Our results suggest that trans-ancestral association analyses can identify ancestry-specific rare and ultra-rare coding variants in MASLD pathogenesis. Furthermore, we demonstrate the utility of machine learning in genetic investigations of difficult-to-phenotype diseases in trans-ancestral biobanks.
Collapse
Affiliation(s)
- Robert Chen
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ben Omega Petrazzini
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Áine Duffy
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Jordan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meena Bansal
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
8
|
Yang D, Dai H, Wang Y, Zhang J, Wei M, Shan M, Zhang X. Association between the non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio and metabolic dysfunction-associated steatotic liver disease. Front Nutr 2025; 12:1557751. [PMID: 40098738 PMCID: PMC11912010 DOI: 10.3389/fnut.2025.1557751] [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: 01/09/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease (MASLD) is one of the most widespread chronic liver diseases and a serious global public health problem. Further research to identify novel risk factors associated with MASLD is urgently needed. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) was identified as a novel lipid marker. The objective of this research was to assess the association between NHHR and MASLD in adults. Methods This cross-sectional study utilized data from the 2017-2020 National Health and Nutrition Examination Survey (NHANES). MASLD was diagnosed in accordance with controlled attenuation parameter scores and a combination of cardiometabolic risk factors. Multivariate logistic regression analyses, in conjunction with the restricted cubic spline method, were employed to investigate the association between NHHR and MASLD risk. Furthermore, subgroup and interaction analyses were conducted. Results This study included 5,269 individuals, with 2,031 individuals diagnosed with MASLD and 3,238 without MASLD. Logistic regression analyses revealed a significant positive correlation between NHHR and MASLD. After the confounding factors were adjusted, each unit rise in NHHR was correlated with a 39% higher probability of MASLD (OR = 1.39, 95% CI: 1.13-1.69). Subgroup and interaction analyses revealed that the positive correlation between NHHR and MASLD held steady regardless of age, gender, race, poverty-to-income ratio, education level, physical activity, body mass index, diabetes, hypertension, dyslipidemia and smoking status (P for interaction >0.05). In addition, a non-linear relationship with an S-shaped manner between NHHR and MASLD was found, with an inflection point at 1.59. Conclusion Our findings imply that an increasing trend in NHHR is associated with a greater risk of MASLD development. NHHR has the potential to function as an indicator for estimating the likelihood of developing MASLD.
Collapse
Affiliation(s)
- Dandan Yang
- Department of Gastroenterology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Hongsheng Dai
- Department of Gastroenterology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Yulu Wang
- Department of Hepatology, The Fourth Peoples' Hospital of Huai'an, Huai'an, China
| | - Jiayi Zhang
- Department of Gastroenterology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Min Wei
- Department of Gastroenterology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Ming Shan
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Xiaoqian Zhang
- Department of Gastroenterology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
9
|
Kim M, Zheng Z. Walking the VLDL tightrope in cardiometabolic diseases. Trends Endocrinol Metab 2025; 36:278-291. [PMID: 39191606 PMCID: PMC11861388 DOI: 10.1016/j.tem.2024.07.020] [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: 04/15/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024]
Abstract
Very-low-density lipoprotein (VLDL), a triglyceride-rich lipoprotein secreted by hepatocytes, is pivotal for supplying peripheral tissues with fatty acids for energy production. As if walking on a tightrope, perturbations in the balance of VLDL metabolism contribute to cardiometabolic dysfunction, promoting pathologies such as cardiovascular disease (CVD) or metabolic dysfunction-associated steatotic liver disease (MASLD). Despite the advent of lipid-lowering therapies, including statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, risks for cardiovascular events persist. With limitations to currently available CVD therapeutics and no US Food and Drug Administration (FDA)-approved treatment for MASLD, this review summarizes the current understanding of VLDL metabolism that sheds light on novel therapeutic avenues to pursue for cardiometabolic disorders.
Collapse
Affiliation(s)
- Mindy Kim
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, 53226, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, 53226, USA.
| | - Ze Zheng
- Department of Physiology, Medical College of Wisconsin, Milwaukee, 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226, USA; Thrombosis & Hemostasis Program, Versiti Blood Research Institute, Milwaukee, 53226, USA.
| |
Collapse
|
10
|
Li H, Hou Y, Xin W, Ding L, Yang Y, Zhang Y, Wu W, Wang Z, Ding W. The efficacy of sodium-glucose transporter 2 inhibitors in patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis. Pharmacol Res 2025; 213:107647. [PMID: 39929274 DOI: 10.1016/j.phrs.2025.107647] [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: 07/19/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
The efficacy of sodium-glucose transporter 2 (SGLT-2) inhibitors for nonalcoholic fatty liver disease (NAFLD) is unclear. Therefore, we conducted a systematic review and meta-analysis to evaluate SGLT-2 inhibitors efficacy for NAFLD treatment. We systematically searched major electronic databases (PubMed, Cochrane Library, Web of Science, Embase) from inception until 11/2023, identifying randomized controlled trials (RCTs) of SGLT-2 inhibitors treatment for patients with NAFLD. The mean differences (MD or SMD) and 95 % confidence intervals (CIs) were calculated via random-effects models. Eleven articles (n = 805 patients with NAFLD) were included in this study. Of these, 408 participants received SGLT-2 inhibitors, while 397 participants were in the control group. SGLT-2 inhibitors significantly reduced liver enzyme levels, including aspartate alanine aminotransferase (ALT) (MD [95 % CI]; -9.31 U/L [-13.41, -5.21], p < 0.00001), aspartate aminotransferase (AST) (MD [95 % CI]; -6.06 U/L [-10.98, -1.15], p = 0.02), and gamma-glutamyltransferase (GGT) (MD [95 % CI]; -11.72 U/L [-15.65, -7.80], p < 0.00001). SGLT-2 inhibitors intervention was also associated with significant reductions in body weight (MD [95 % CI]; -2.72 kg [-3.49, -1.95], p < 0.00001) and BMI (MD [95 % CI]; -1.11 kg/m2 [-1.39, -0.82], p < 0.00001) and improvements in glycaemic indices, triglyceride (TG) and high-density lipoprotein cholesterol (HDL-C). However, no significant changes in total cholesterol (TC) or low-density lipoprotein cholesterol (LDL-C) were observed. The meta-analysis revealed a beneficial effect of SGLT-2 inhibitors on liver functions and body weight, BMI, TG, HDL-C, and glucose homeostasis in patients with NAFLD, indicating that SGLT-2 inhibitors might be a clinical therapeutic strategy for these patients, especially individuals with concurrent type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Hongsheng Li
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China
| | - Yanli Hou
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China; Shandong Institute of Endocrine and Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China
| | - Wenyong Xin
- Department of Retirement Affairs, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China
| | - Lina Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China; Shandong Institute of Endocrine and Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China
| | - Ying Yang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China; Shandong Institute of Endocrine and Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China
| | - Yikun Zhang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China; Shandong Institute of Endocrine and Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China
| | - Wenqi Wu
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China
| | - Zhibin Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China; Shandong Institute of Endocrine and Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China.
| | - Wenyu Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877, Jingshi Rd, Jinan 250062, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China; Shandong Institute of Endocrine and Metabolic Diseases, 18877, Jingshi Rd, Jinan 250062, China.
| |
Collapse
|
11
|
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
|
12
|
Li B, Liu Y, Ma X, Guo X. The association between non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio and hepatic steatosis and liver fibrosis among US adults based on NHANES. Sci Rep 2025; 15:6527. [PMID: 39988726 PMCID: PMC11847945 DOI: 10.1038/s41598-025-90773-y] [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: 11/01/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
Recently, the non-high-density to high-density lipoprotein cholesterol ratio (NHHR) has gained growing attention as an indicator for predicting diseases associated with lipid metabolism. Hepatic steatosis and fibrosis are tightly associated lipid metabolism. Our study aims to analyze the correlations among NHHR, hepatic steatosis, and fibrosis. This study analysed data from 14,578 adults in the US National Health and Nutrition Examination Survey (2005-2018). The degree of hepatic steatosis was measured through the Fatty Liver Index (FLI), while liver fibrosis severity was evaluated with the Fibrosis-4 (FIB-4) index. Multivariate linear regression assessed the association between NHHR and the FLI and FIB-4 score. Smooth curve describing the relationship between NHHR and FLI or FIB-4. Additionally, a two-part linear regression model adopted in order to more accurately account for the nonlinear relationship, with threshold effects estimated through its two components. To confirm the robustness of the findings, interaction tests and subgroup analyses were conducted. The multivariate logistic regression analysis demonstrated a significantly positive correlation of lnNHHR with FLI across all three models. In Model 3, the association was (β = 11.14, 95%CI:10.38,11.90). Curve fitting indicated a nonlinear relationship. The positive correlation between lnNHHR and FLI persists across gender, BMI, and physical activity groups. Nevertheless, a notable negative correlation between lnNHHR and FIB-4 was observed in all three models. In Model 3, the relationship between lnNHHR and FIB-4 was as follows: (β = -0.20; 95% CI: -0.22, -0.17). Curve fitting revealed a V-shaped relationship, with threshold effect analysis identifying a breakpoint at 1.51. Above this threshold, the relationship was found to be statistically insignificant (p-value = 0.424). Receiver operating characteristic (ROC) curve analysis demonstrated that NHHR exhibited better predictive performance for MASLD compared to non-HDL-C, HDL-C, and LDL-C/HDL-C. The current study's findings suggest that elevated levels of NHHR correlate with a greater risk of hepatic steatosis among adults in the U.S. Our findings imply that NHHR may be a valuable tool in improving MASLD prevention strategies in the general population.
Collapse
Affiliation(s)
- Baoyu Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China
| | - Yuwei Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China
| | - Xiaorong Ma
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China.
| | - Xiaoyan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China.
| |
Collapse
|
13
|
Seagle HM, Akerele AT, DeCorte JA, Hellwege JN, Breeyear JH, Kim J, Levin M, Khodurksy S, Bress A, Lee K, Meiler J, Gill D, Lee JS, Heberer K, Miller DR, Reaven P, Chang KM, Lynch JA, Khankari NK, Shuey MM, Edwards TL, Vujkovic M. Genomics-Informed Drug Repurposing Strategy Identifies Novel Therapeutic Targets for Metabolic Dysfunction-Associated Steatotic Liver Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.18.25321035. [PMID: 40034783 PMCID: PMC11875238 DOI: 10.1101/2025.02.18.25321035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Identification of drug-repurposing targets with genetic and biological support is an economically and temporally efficient strategy for improving treatment of diseases. We employed a cross-disciplinary approach to identify potential treatments for metabolic dysfunction associated steatotic liver disease (MASLD) using humans as a model organism. We identified 212 putative causal genes associated with MASLD using data from a large multi-ancestry genetic association study, of which 158 (74.5%) are novel. From this set we identified 57 genes that encode for druggable protein targets, and where the effects of increasing genetically predicted gene expression on MASLD risk align with the function of that drug on the protein target. These potential targets were then evaluated for evidence of efficacy using Mendelian randomization, pathway analysis, and protein structural modeling. Using these approaches, we present compelling evidence to suggest activation of FADS1 by icosopent ethyl as well as S1PR2 by fingolimod could be promising therapeutic strategies for MASLD.
Collapse
Affiliation(s)
- Hannah M Seagle
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Joseph Maxwell Cleland Atlanta VA Medical Center, Atlanta, Georgia, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alexis T Akerele
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- School of Graduate Studies and Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
- Division of Quantitative Science, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Joseph A DeCorte
- Vanderbilt Medical Scientist Training Program, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jacklyn N Hellwege
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System (626), Nashville, Tennessee, United States of America
| | - Joseph H Breeyear
- Biostatistics and Computational Biology Branch, National Institute for Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Jeewoo Kim
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Quantitative Science, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Medical Scientist Training Program, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Michael Levin
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Samuel Khodurksy
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Adam Bress
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
- University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Kyung Lee
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
| | - Jens Meiler
- Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Jennifer S Lee
- Stanford University, Stanford, California, United States of America
- Palo Alto VA Medical Center, Palo Alto, California, United States of America
| | - Kent Heberer
- Palo Alto VA Medical Center, Palo Alto, California, United States of America
| | - Donald R Miller
- VA Center for Medication Safety, Department of Veterans Affairs, Chicago, Illinois, United States of America
- Center for Population Health, Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, United States of America
| | - Peter Reaven
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Kyong-Mi Chang
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz Philadelphia VA Medical Center, Philadelphia, Pennsylvania, United States of America
| | - Julie A Lynch
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
- University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Nikhil K Khankari
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Megan M Shuey
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System (626), Nashville, Tennessee, United States of America
| | - Marijana Vujkovic
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz Philadelphia VA Medical Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
14
|
Dakal TC, Xiao F, Bhusal CK, Sabapathy PC, Segal R, Chen J, Bai X. Lipids dysregulation in diseases: core concepts, targets and treatment strategies. Lipids Health Dis 2025; 24:61. [PMID: 39984909 PMCID: PMC11843775 DOI: 10.1186/s12944-024-02425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/30/2024] [Indexed: 02/23/2025] Open
Abstract
Lipid metabolism is a well-regulated process essential for maintaining cellular functions and energy homeostasis. Dysregulation of lipid metabolism is associated with various conditions, including cardiovascular diseases, neurodegenerative disorders, and metabolic syndromes. This review explores the mechanisms underlying lipid metabolism, emphasizing the roles of key lipid species such as triglycerides, phospholipids, sphingolipids, and sterols in cellular physiology and pathophysiology. It also examines the genetic and environmental factors contributing to lipid dysregulation and the challenges of diagnosing and managing lipid-related disorders. Recent advancements in lipid-lowering therapies, including PCSK9 inhibitors, ezetimibe, bempedoic acid, and olpasiran, provide promising treatment options. However, these advancements are accompanied by challenges related to cost, accessibility, and patient adherence. The review highlights the need for personalized medicine approaches to address the interplay between genetics and environmental factors in lipid metabolism. As lipidomics and advanced diagnostic tools continue to progress, a deeper understanding of lipid-related disorders could pave the way for more effective therapeutic strategies.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia, University, Udaipur, 313001, India
| | - Feng Xiao
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Chandra Kanta Bhusal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | | | - Rakesh Segal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Juan Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Xiaodong Bai
- Department of Plastic Surgery, Southern University of Science and Technology Hospital, Shenzhen, 518055, China.
| |
Collapse
|
15
|
Xu Z, Wang Y, Li X, Hou X, Yue S, Wang J, Ye S, Wu J. Interacting and joint effects of frailty and inflammation on cardiovascular disease risk and the mediating role of inflammation in middle-aged and elderly populations. BMC Cardiovasc Disord 2025; 25:118. [PMID: 39979798 PMCID: PMC11841180 DOI: 10.1186/s12872-025-04567-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Frailty and inflammation may increase the risk of cardiovascular disease (CVD), but their interacting and joint effects on CVDs remain unclear. To explore the interaction effects of frailty and inflammation on CVDs and the role of inflammation in the relationship between frailty and CVDs to provide better understanding of the underlying pathogenesis of CVD. METHODS A total of 220,608 initially CVD-free participants were recruited from the UK Biobank database and were categorized into non-frailty, pre-frailty, and frailty groups based on Fried's criteria. The participants were also grouped according to the low-grade inflammation (INFLA) score and its components: the neutrophil-lymphocyte ratio, C-reactive protein, white blood cell count, and platelet count. Cox proportional hazards models with hazard ratios (HRs) and 95% confidence intervals (CIs) were used to assess the effects of frailty phenotypes and inflammation on CVD risk. Mediation analysis was used to quantify the role of inflammation in the association between frailty and CVDs. The potential interactions between frailty and inflammation in terms of CVD risk were also evaluated using additive and multiplicative scales. RESULTS During a median follow-up of 13.3 years, 48,978 participants developed CVDs. After adjusting for various confounders, participants with pre-frailty and frailty had a higher risk of CVDs than those with non-frailty (HRs: 1.20 (95% CI: 1.18-1.23) and 1.80 (95% CI: 1.69-1.91), respectively). A higher risk of CVDs was observed among participants with moderate and high INFLA scores than those with low INFLA scores (HRs: 1.09 (95% CI: 1.07-1.12) and 1.27 (95% CI: 1.24-1.30), respectively). The INFLA score and its components had limited mediating effects in the association between frailty and CVDs. Significant interactions were observed between frailty phenotypes and INFLA scores on CVDs on the multiplicative scale but not on the additive scale. CONCLUSION Inflammation may amplify the harmful effect of frailty on the incidence of CVDs. Improving frailty alone might not substantially reduce the risk of CVDs, but effectively controlling inflammation might help to reduce the negative effects of frailty on cardiovascular health.
Collapse
Affiliation(s)
- Zihan Xu
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Yingbai Wang
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Xiaolin Li
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Xuefei Hou
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Suru Yue
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Jia Wang
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Shicai Ye
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| | - Jiayuan Wu
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
16
|
Wang X, Song Q, Zhang Q, Li X, Wang J, Gong J, Zhang Z, Tan N, Tsang SY, Wong WT, Ma D, Jiang L. The Prognostic Significance of the DBIL/HDLC Ratio in Patients With Dilated Cardiomyopathy. Cardiovasc Ther 2025; 2025:8835736. [PMID: 40170700 PMCID: PMC11961277 DOI: 10.1155/cdr/8835736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025] Open
Abstract
Background: In cardiovascular pathology, both direct bilirubin (DBIL) and high-density lipoprotein cholesterol (HDLC) have been associated with adverse clinical outcomes. However, the prognostic significance of these biomarkers in the context of dilated cardiomyopathy (DCM) remains unclear. To address this gap, this study conducted a retrospective analysis to evaluate the prognostic value of the DBIL/HDLC ratio in patients diagnosed with DCM. Methods and Results: A total of 986 consecutive DCM patients were retrospectively enrolled from January 2010 to December 2019 and divided into two groups based on the DBIL/HDLC ratio cut-off value: ≤ 4.45 (n = 483) and > 4.45 (n = 503). Patients with lower DBIL/HDLC (≤ 4.45) experienced lower in-hospital mortality, long-term mortality, and major adverse clinical events (MACEs) (0.8%, 32.9%, and 12.2%, respectively) compared to those with higher DBIL/HDLC (> 4.45) (6.4%, 59.1%, and 16.7%, respectively). Multivariate analysis identified DBIL/HDLC as an independent risk factor for long-term mortality (odds ratio: 1.026; 95% confidence interval (CI): 1.005-1.048; p = 0.016) and all-cause mortality over a median follow-up of 67 ± 1.8 months (hazard ratio: 1.011; 95% CI: 1.005-1.018; p < 0.001). The receiver operating characteristic curve showed good discrimination for long-term mortality (area under the curve (AUC): 0.675; 95% CI: 0.692-0.708; p < 0.001). The Kaplan-Meier survival analysis demonstrated a better prognosis for patients with DBIL/HDLC ≤ 4.45 (log-rank χ 2 = 40.356, p < 0.001). Furthermore, the impact of additional variables on the results was investigated by a subgroup analysis. Conclusion: The DBIL/HDLC ratio could serve as a simple and cost-effective tool for evaluating prognosis in DCM.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qiqi Song
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Qingqing Zhang
- Department of Anesthesiology, Guangdong Provincial People's Hospital Ganzhou Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Xinyi Li
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiaqi Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiantao Gong
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ziyi Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ning Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Suk-Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing-tak Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Dunliang Ma
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lei Jiang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
17
|
Zeng J, Lo CH. Editorial: Lipid metabolism dysregulation in obesity-related diseases and neurodegeneration. Front Endocrinol (Lausanne) 2025; 16:1564003. [PMID: 40007811 PMCID: PMC11850260 DOI: 10.3389/fendo.2025.1564003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, United States
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, United States
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, United States
- Department of Biology, Syracuse University, Syracuse, NY, United States
| |
Collapse
|
18
|
Wang W, Qin J, Bai S, Tian J, Zhou Y, Qin X, Gao X. Integrative transcriptomics and lipidomics unravels the amelioration effects of Radix Bupleuri on non-alcoholic fatty liver disease. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119005. [PMID: 39490432 DOI: 10.1016/j.jep.2024.119005] [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] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix Bupleuri (Bupleurum chinense DC.) is the most commonly used traditional Chinese medicine (TCM) for the treatment of liver diseases. While the effects of Radix Bupleuri (BR) on lipid-lowering and liver protection have been established, its role in the development of non-alcoholic fatty liver disease (NAFLD) induced by a high-fat diet remains unclear. AIM OF THE STUDY The objective of this study was to evaluate the alleviation effects of the active fraction of BR on NAFLD in vivo and to explore the underlying mechanisms through an analysis of liver transcriptome and lipidomics. MATERIALS AND METHODS The NAFLD model was established in SD rats by administering a high-fat diet (HFD) for 8 weeks. Subsequently, the NAFLD model rats were continuously gavaged with different polarity fractions of BR (25 g/kg/d) and melatonin (MT) (30 mg/kg/d) for an additional 6 weeks to assess therapeutic effects. The potential mechanism of the low polarity fraction of BR (LBR) in treating NAFLD was investigated through hepatic transcriptome analysis, non-targeted lipidomics, RT-qPCR, protein-protein interaction (PPI) network construction, molecular docking, and Western blotting, aiming to elucidate the underlying mechanisms by which LBR may ameliorate NAFLD. RESULTS These results demonstrated that LBR significantly alleviated the effects of HFD-induced NAFLD, as evidenced by reductions in body weight (BW), liver weight (LW), and epididymal fat weight (EFW) compared to model rats and other polarity fractions of BR. Furthermore, LBR notably down-regulated serum and liver levels of total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C), while up-regulating high-density lipoprotein cholesterol (HDL-C) in serum. Mechanistically, liver transcriptome analysis indicated that fatty acid metabolism may be a crucial pathway for the improvement of NAFLD following LBR treatment. Lipidomics data suggested that LBR can modulate the metabolic profile in NAFLD rats. Enrichment analysis revealed that glycerophospholipid and glycerolipid metabolism might be key pathways involved in the development of NAFLD. RT-qPCR analysis demonstrated that LBR could regulate the expression of lipid-related genes in these critical pathways. Additionally, Spearman correlation analysis showed a strong relationship between lipid metabolic biomarkers, pathological indices, and lipid-related genes. Moreover, protein-protein interaction (PPI) network and molecular docking analyses identified seven key targets with six ingredients of LBR exhibiting good binding capacity (<-5.0 kcal/mol). Finally, Western blotting analysis indicated that LBR up-regulates the expression levels of PPARα, CPT1, and FABP1 while down-regulating the expression levels of SREBF1 and SCD1, thereby improving metabolism and exerting a lipid-lowering effect. CONCLUSION In conclusion, the present research elucidated the lipid-lowering mechanisms of the active fractions of BR. Both BR and LBR presented themselves as promising candidates for the development of novel pharmacological agents targeting NAFLD. LBR effectively ameliorated lipid disturbances associated with HFD-induced NAFLD by modulating the metabolism of fatty acids, cholesterol, glycerolipid, and glycerophospholipids. Consequently, LBR held significant potential for development as an effective lipid-lowering therapeutic.
Collapse
Affiliation(s)
- Weiyu Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Jiaxin Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Shuaidong Bai
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Junsheng Tian
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Yuzhi Zhou
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China
| | - Xiaoxia Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| |
Collapse
|
19
|
Lau PP, Wei CY, Lin MR, Chou WH, Wan YJY, Chang WC. Genome-wide association study of the fatty liver index in the Taiwanese population reveals shared and population-specific genetic risk factors across ethnicities. Cell Biosci 2025; 15:19. [PMID: 39923089 PMCID: PMC11807309 DOI: 10.1186/s13578-025-01346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/08/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Although the incidence of fatty liver disease (FLD) is increasing worldwide, the genetic basis of this disease is not fully understood. This study uses the fatty liver index (FLI) to identify and compare genetic variants associated with FLD in Taiwanese and European populations. RESULTS In this study, a total of 145,356 Taiwan Biobank participants were included in the discovery analysis. Subjects with elevated FLI were found to have a significantly greater risk of developing FLD, as confirmed by imaging data (OR: 4.43; 95% CI: 3.88-5.06). Through genome-wide association studies (GWAS), we identified 6 variants previously associated with nonalcoholic fatty liver disease (NAFLD) and validated 50 shared risk variants located in ZPR1 and FTO between the Taiwanese and European populations. Conditional analysis of 423 significant variants from FLI-defined FLD further revealed 16 independent variants within 14 genes. Pathway analysis of GWAS significant genes revealed that lipid metabolism and the peroxisome proliferator-activated receptor (PPAR) signaling pathway are causes of hepatic fat accumulation. CONCLUSION This study identified six independent NAFLD-associated variants in GCKR, LPL, TRIB1AL, and FTO and emphasized ZPR1 and FTO as shared risk genes for FLI-defined FLD in both Taiwanese and European populations. These findings support the utility of the FLI for FLD prediction, provide new genetic insights, and reveal the common genetic pathways of FLD across two ethnic groups. This research offers a valuable framework for advancing personalized medicine and therapeutic strategies for FLD.
Collapse
Affiliation(s)
- Pei Pei Lau
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chun-Yu Wei
- Core Laboratory of Neoantigen Analysis for Personalized Cancer Vaccine, Office of R&D, Taipei Medical University, Taipei, Taiwan
| | - Min-Rou Lin
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wan-Hsuan Chou
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Jui Yvonne Wan
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- Core Laboratory of Neoantigen Analysis for Personalized Cancer Vaccine, Office of R&D, Taipei Medical University, Taipei, Taiwan.
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
20
|
Ren Y, Chen B, Zhang H, Xu S. A cohort study reveals shared and distinct serum metabolic biomarkers for major adverse cardiovascular events in middle-aged and older adults. GeroScience 2025:10.1007/s11357-025-01544-6. [PMID: 39904969 DOI: 10.1007/s11357-025-01544-6] [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: 10/07/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
We assessed the association of serum metabolites with the occurrence of major adverse cardiovascular events (MACE) in middle-aged and elderly individuals, explored the value of metabolomics in predicting MACE, and compared the distinctions in MACE risk-related metabolic biomarkers between middle-aged and elderly groups. Among the participants of the UK Biobank who underwent baseline assessment through nuclear magnetic resonance (NMR)-based metabolomic profiling of 168 serum metabolites and had complete covariates and clinical lipid parameters, we included those without a previous diagnosis of ischemic heart disease, cerebrovascular disease, heart failure, or cardiac arrest and not on lipid-lowering medications. Relevant covariates included sociodemographic characteristics, lifestyle factors, clinical information, and fasting time. Cox regression gave adjusted hazard ratios for metabolites, including the concentrations of various lipoprotein particles, compositional profiles of different lipoproteins, ketone bodies, amino acids, fatty acids, and additional low-molecular-weight metabolic biomarkers. The least absolute shrinkage and selection operator (LASSO) regression was applied to these metabolites to screen characteristic metabolic variables. Selected feature metabolic biomarkers were added to the established model for predicting MACE risk; risk differentiation (C-statistic) and reclassification (continuous net reclassification improvement [NRI], integrated differentiation index [IDI]) were evaluated. This study included 54,561 UK Biobank participants (34,797 middle-aged adults and 19,764 elderly adults) and was followed for a median of more than 12 years. Of these, there are 1799 middle-aged individuals and 2527 elderly individuals incident of MACE (ischemic heart disease, stroke, and cardiovascular deaths). After adjusting for relevant covariates, Cox regression yielded metabolic biomarkers associated with the occurrence of MACE in the population (false discovery rate controlled P < 0.05). In the elderly, the metabolites associated with increased MACE risk were notably diminished compared to the middle-aged; and the elderly group underscored the protective function of medium and small HDL and their constituents, docosahexaenoic acid, and glycine. The more comprehensive model, which additionally includes the feature metabolic biomarkers, demonstrated enhanced discriminatory power and predictive accuracy for MACE occurrence among middle-aged individuals, evidenced by improved C-statistics (from 0.711 [95% CI 0.699-0.722] to 0.723 [0.711-0.734]), a continuous NRI of 0.247 [0.207-0.315], and an absolute IDI of 0.005 [0.004-0.008]. Its evaluation value is superior to that in the elderly. Our study explored the association of circulating metabolites with MACE risk in middle-aged and elderly adults and made comparisons. Metabolomic insights have revealed biomarkers associated with new-onset MACE in different age populations, highlighting the value of protective metabolites in the elderly. This provides instrumental information to possibly implement precision medicine for preventing MACE.
Collapse
Affiliation(s)
- Yi Ren
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Department of Pathology and Pathophysiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Bo Chen
- Department of Endocrinology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
- Center for Clinical Evidence-Based and Translational Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Honggang Zhang
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing, China.
- Department of Pathology and Pathophysiology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Shaoyong Xu
- Department of Endocrinology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China.
- Center for Clinical Evidence-Based and Translational Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China.
| |
Collapse
|
21
|
Liu K, He H, Liu M, Hu YQ, Lu LW, Liu B, Chen JH. Evaluating the differential benefits of varying carbohydrate-restricted diets on lipid profiles and cardiovascular risks in dyslipidemia: a meta-analysis and systematic review. Food Funct 2025; 16:831-852. [PMID: 39807892 DOI: 10.1039/d4fo05125d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Background: carbohydrate-restricted diets (CRDs) have gained attention to address metabolic dysregulation commonly observed in dyslipidemia, a condition posing significant risks to cardiovascular health. However, the effectiveness of CRDs in improving cardiovascular health remains contentious. This meta-analysis comprehensively evaluated the long-term effects of CRDs on glucolipid metabolism and weight loss in individuals with dyslipidemia. Methods: extensive searches were conducted in PubMed, Web of Science, Scopus, the Cochrane Library, and EMBASE. Randomized controlled trials examining the effects of CRDs on glucolipid metabolism and weight loss in adults with dyslipidemia over a minimum of three weeks were included. This analysis compared the differential effects between moderate-low carbohydrate diets (MLCDs) and low carbohydrate diets (LCDs), including a targeted evaluation of animal-based CRDs and dyslipidemic individuals based on the BMI status, thereby addressing gaps in current knowledge. Results: Our findings indicated that CRDs significantly enhanced lipid profiles, including total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), and apolipoprotein B (ApoB), and contributed to weight management in individuals with dyslipidemia. MLCDs were more effective than LCDs in improving all lipid profiles except for TG, which was more effectively managed by LCDs. Animal-based CRDs did not significantly impact lipid profiles. Dyslipidemic individuals with overweight and obesity showed significant changes in TG and ApoB. A noteworthy negative correlation was also observed between TC, TG, and low-density lipoprotein cholesterol levels with higher dietary fiber intake, supporting the beneficial impact of fiber on cardiovascular health. Conclusions: These results for the first time highlighted the potential of adopting MLCDs, particularly those with sufficient fiber content, as a powerful strategy for reducing the risk of cardiovascular diseases in patients suffering from dyslipidemia.
Collapse
Affiliation(s)
- Kang Liu
- School of Medicine, Guangzhou Medical University, Guangzhou 511436, China
| | - Hui He
- School of Medicine, Guangzhou Medical University, Guangzhou 511436, China
| | - Min Liu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yu-Qi Hu
- School of Medicine, Guangzhou Medical University, Guangzhou 511436, China
| | - Louise Weiwei Lu
- School of Biological Sciences, Faculty of Science, the University of Auckland, Auckland, New Zealand
| | - Bin Liu
- Department of Traditional Chinese Medicine, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgangdong Road, Guangzhou 510260, China
| | - Jie-Hua Chen
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China.
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
22
|
Hirano T. Excess Triglycerides in Very Low-Density Lipoprotein (VLDL) Estimated from VLDL-Cholesterol could be a Useful Biomarker of Metabolic Dysfunction Associated Steatotic Liver Disease in Patients with Type 2 Diabetes. J Atheroscler Thromb 2025; 32:253-264. [PMID: 39231648 PMCID: PMC11802249 DOI: 10.5551/jat.65164] [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: 05/27/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
AIMS We report that small dense low-density lipoprotein cholesterol (sdLDL-C) levels are sensitive biomarkers of metabolic dysfunction-associated steatotic liver disease (MASLD). Since triglyceride (TG)-rich very low-density lipoprotein (VLDL) is a precursor of sdLDL and is overproduced by MASLD, the composition of VLDL may be more directly associated with MAFLD than sdLDL-C or plasma TG. To identify TG-rich VLDL, this author proposed "Excess TG" and examined its association with MASLD. METHODS Patients with type 2 diabetes (n=1295), excluding fasting hypertriglyceridemia (TG ≥ 400 mg/dL) and heavy drinkers were examined. Liver steatosis and visceral fat area (VFA) were evaluated using CT. VLDL-C was calculated as the total C minus direct LDL-C minus HDL-C. The average VLDL-TG level can be estimated using VLDL-C×5, according to the principle of the Friedewald equation for LDL-C. Thus, VLDL-TG was estimated as VLDL-C×5, and Excess TG was calculated as plasma TG minus VLDL-C×5. RESULTS Patients with MASLD were younger, more likely to be men and drinkers, and had higher VFA, TG, sdLDL-C, and excess TG, while VLDL-C was comparable. Excess TG was found to be the most sensitive lipid parameter for identifying MASLD, independent of sdLDL-C, TG, TG/VLDL-C, and VFA. The odds ratios for MASLD were 2.4-, 3.7-, and 3.9-fold higher for Excess TG ranges of 0-24, 25-49, and ≥ 50 mg/dL, respectively, relative to <0 mg, and a close relationship remained significant after adjustment for lipid- and adiposity-related parameters. CONCLUSIONS Excess TG in VLDL was strongly associated with MASLD beyond TG and sdLDL-C levels, which may reflect the presence of TG-rich VLDL.
Collapse
Affiliation(s)
- Tsutomu Hirano
- Diabetes Center, Ebina General Hospital, Kanagawa, Japan
| |
Collapse
|
23
|
Yu X, Cao Y, Li X, Liang Q, Dong X, Liang B. Association Between Arterial Stiffness Index and Age-Related Diseases: A Mendelian Randomization Study. Rejuvenation Res 2025; 28:9-16. [PMID: 39291781 PMCID: PMC11844224 DOI: 10.1089/rej.2024.0041] [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: 05/29/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024] Open
Abstract
Arterial stiffness is an emerging indicator of cardiovascular risk, but its causal relationship with a variety of age-related diseases is unclear. The objective is to assess the causal relationship between arterial stiffness index (ASI) and age-related diseases by Mendelian randomization (MR) analysis. We obtained instrumental variables associated with age-related diseases from genome-wide association studies (GWAS) of 484,598 European individuals, and data for ASI were obtained from the UK Biobank GWAS of 127,127 participants. We used the inverse variance-weighted as the primary analysis method. In addition, several sensitivity analyses including MR-Egger, weighted-median (WM), Mendelian randomization pleiotropy residual sum and outlier, and Cochran's Q test were performed to test the robustness of the results. Reverse MR analysis was also performed to assess reverse causal relationships between age-related diseases and ASI. We verified the causal relationship between eight age-related diseases and ASI, of which cardiovascular disease (β = 0.19), gallbladder disease (β = 0.85), liver, biliary, or pancreas problem (β = 1.02), hypertension (β = 0.19), joint disorder (β = 0.53), and esophageal disorder (β = 2.10) elevated ASI. In contrast, hyperthyroidism or thyrotoxicosis (β = -2.17) and bowel problems (β = -1.83) may reduce ASI. This MR analysis reveals causal relationships between ASI and several age-related diseases. ASI is expected to be a potential indicator of health conditions for older populations.
Collapse
Affiliation(s)
- Xiaojie Yu
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Yang Cao
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Xinyi Li
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Xiaodan Dong
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Bing Liang
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
24
|
Zhu Y, Zhang KX, Bu QY, Song SX, Chen Y, Zou H, You XY, Zhao GP. Ginsenosides From Panax ginseng Improves Hepatic Lipid Metabolism Disorders in HFD-Fed Rats by Regulating Gut Microbiota and Cholesterol Metabolism Signaling Pathways. Phytother Res 2025; 39:714-732. [PMID: 39660634 DOI: 10.1002/ptr.8402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/11/2024] [Accepted: 02/27/2024] [Indexed: 12/12/2024]
Abstract
A high-fat diet (HFD) is often associated with hepatic lipid metabolism disorders, leading to dysfunction in multiple body systems. Ginsenosides derived from Panax ginseng have been reported to possess potential effects in ameliorating lipid metabolism disorders; however, their underlying mechanisms remain insufficiently explored. This study aims to investigate the bioactivities of ginsenosides in combating lipid metabolism disorders and obesity, with a focus on their mechanisms involving the cholesterol metabolism signaling pathway and gut microbiota. Our results demonstrated that ginsenoside treatment significantly reduced overall body weight, body weight changes, liver weight, and eWAT weight, as well as alleviated hepatic steatosis and dyslipidemia in HFD-fed rats, without affecting food intake. These effects were dose-dependent. Furthermore, 16S rRNA sequencing revealed that ginsenosides significantly increased the relative abundance of Akkermansia muciniphila, Blautia, Eisenbergiella, Clostridium clusters XI, XVIII, and III, while decreasing the relative abundance of Clostridium subcluster XIVa and Dorea. In addition, ginsenoside treatment significantly regulated the expression of hepatic genes and proteins involved in the cholesterol metabolism signaling pathway (FXR, CYP7A1, CYP7B1, CYP27A1, ABCG5, ABCG8, Insig2, and Dhcr7), potentially inhibiting hepatic cholesterol biosynthesis while promoting cholesterol transport to HDL and its excretion via bile and feces. Notably, levels of 7-dehydrocholesterol (7-DHC) and 27-hydroxycholesterol (27-OHC) were reduced, while 5β,6β-epoxycholesterol (5,6β-epoxy) levels were elevated following ginsenoside treatment, indicating significant modulation of oxysterols by ginsenosides. Moreover, bile acid enterohepatic circulation was regulated through the enhancement of hepatic FXR-CYP7A1 signaling and intestinal FXR-FGF15 signaling in HFD-fed rats treated with ginsenosides, which was closely linked to gut microbiota composition. Collectively, our findings suggest that ginsenosides alleviate hepatic lipid metabolism disorders by modulating gut microbiota and the cholesterol metabolism signaling pathway in HFD-fed rats.
Collapse
Affiliation(s)
- Yue Zhu
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Kang-Xi Zhang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Qing-Yun Bu
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Haihe Laboratory of Synthetic Biology, Tianjin, China
| | - Shu-Xia Song
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Haihe Laboratory of Synthetic Biology, Tianjin, China
| | - Yue Chen
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Hong Zou
- CAS Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Xiao-Yan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Guo-Ping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
25
|
Pirahesh K, Zarrinnia A, Nikniaz L, Nikniaz Z. Association between sleep duration and risk of nonalcoholic fatty liver disease: A systematic review and meta-analysis. Prev Med Rep 2025; 50:102968. [PMID: 39897736 PMCID: PMC11783128 DOI: 10.1016/j.pmedr.2025.102968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Introduction Considering that both long and short sleep duration may have an association with nonalcoholic fatty liver disease (NAFLD), in this meta-analysis, we analyzed the dose-response association between sleep duration and NAFLD along with meta-analyses of the differences in mean sleep duration between NAFLD patients and healthy controls, and linear meta-analysis of the association between sleep duration and NAFLD. Methods PubMed (665 articles), Scopus (442 articles), and Web of Sciences (200 articles) were searched from inception until November 2023. Observational studies were included if they assess the association between sleep duration and NAFLD or compare the mean sleep duration between patients with NAFLD and healthy population. All studies done in humans without restriction on sex, age, and language were included. The methological quality of studies was assessed by Joanna Briggs Institute (JBI) tools. The meta-analysis was conducted using STATA. Results Thirty-one studies that included 836,117 participants were included in this systematic review. The results indicated no significant differences between NAFLD patients and healthy controls regarding mean sleep duration [Mean difference: -7.08, 95 % CI: -20.10, 5.94]. The subgroup meta-analysis did not show any significant differences between groups. The long versus short sleep duration meta-analysis showed a significant association between sleep duration and NAFLD (OR: 0.8 [95 % CI, 0.74-0.91]). The results of the dose-response meta-analysis do not suggest a linear or nonlinear relationship between sleep duration and NAFLD (p-value = 0.9). Conclusion The highest category of sleep duration was associated with a lower risk of NAFLD. However, no dose-response association was observed.
Collapse
Affiliation(s)
- Kasra Pirahesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Zarrinnia
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Students` Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Nikniaz
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Nikniaz
- Tabriz Health Services Management Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Liu ZC, Fu HJ, Li NC, Deng FJ, Gan YK, Ye YJ, Huang BH, Liu C, Chen JH, Li XF. Systematic pharmacology and experimental validation to elucidate the inflammation-associated mechanism of Huanglian Wendan (HLWD) decoction in the treatment of MAFLD associated with atherosclerosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118841. [PMID: 39299361 DOI: 10.1016/j.jep.2024.118841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Metabolic-associated fatty liver disease (MAFLD) and atherosclerosis are very common disorders that frequently coexist. The therapeutic efficacy of Huanglian Wendan (HLWD) decoction, a traditional Chinese medicine (TCM) prescription, is satisfactory in treating MAFLD associated with atherosclerosis. However, the underlying mechanisms through which HLWD exerts its effects need to be elucidated. Given the complex composition of HLWD and its multiple therapeutic targets, pharmacological investigation is challenging. AIM OF THIS STUDY This study aimed to identify the effective compounds in HLWD and elucidate the mechanisms involved in its therapeutic effect on MAFLD associated with atherosclerosis. MATERIALS AND METHODS We used a systematic pharmacology method to identify effective compounds present in HLWD and determine the mechanism by which it affects MAFLD associated with atherosclerosis. The effective components of HLWD were identified through ultrahigh-performance liquid chromatography-q exactive-orbitrap high resolution mass spectrometry (UHPLC-Q-Orbitrap HRMS). Next, a comprehensive in silico method was used to predict potential related targets and disease targets for these compounds to establish corresponding pathways. The accuracy of our assumed systemic pharmacology results was determined by conducting follow-up experiments. RESULTS By conducting UHPLC-Q-Orbitrap HRMS combined with network analysis, we identified 18 potentially active components of HLWD and assessed the inflammatory regulatory mechanism by which it affects MAFLD associated with atherosclerosis on the basis of 52 key targets. We used a high-fat, high-cholesterol (HFHC)-induced mice model of MAFLD associated with atherosclerosis to confirm our results. We found that administering HLWD significantly improved the appearance of their liver and reduced their body weight, liver weight, blood lipids, hepatic damage, and hepatic pathology. HLWD also decreased atherosclerotic lesion areas, foam cells, and inflammatory cells in the aorta. HLWD showed anti-inflammatory effects, suppressed M1 polarization, and promoted M2 polarization in the liver and aorta. HLWD might also regulate peroxisome proliferator-activated receptor-γ (PPARγ)/nuclear factor kappa-B (NF-κB) signaling to influence macrophage polarization and inflammation. CONCLUSIONS Our results showed that HLWD protected against HFHC diet-induced MAFLD associated with atherosclerosis by regulating PPARγ/NF-κB signaling, thus adjusting macrophage polarization and inflammation. Additionally, pharmacochemistry research, network pharmacology analysis, and experimental verification can be combined to form a comprehensive model used in studies on TCM.
Collapse
Affiliation(s)
- Zhi-Chao Liu
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Huan-Jie Fu
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, PR China.
| | - Ning-Cen Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Fang-Jun Deng
- Department of Cardiovascular, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300150, PR China.
| | - Yong-Kang Gan
- Department of Vascular Surgery, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300150, PR China.
| | - Yu-Jia Ye
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Bing-Hui Huang
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Chang Liu
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Jin-Hong Chen
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong Province, 261053, PR China.
| | - Xiao-Feng Li
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, PR China.
| |
Collapse
|
27
|
Chen D, Shen Y, Huang F, Huang B, Xu S, Li L, Liu J, Li Z, Li X. Ethanol extract of Polygonatum cyrtonema Hua mitigates non-alcoholic steatohepatitis in mice. Front Pharmacol 2025; 15:1487738. [PMID: 39949396 PMCID: PMC11821971 DOI: 10.3389/fphar.2024.1487738] [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: 08/28/2024] [Accepted: 12/20/2024] [Indexed: 02/16/2025] Open
Abstract
Background Polygonum cyrtonema Hua is a kind of traditional Chinese botanic drug. Modern pharmacological research has confirmed that Polygonum cyrtonema Hua is able to alleviate nonalcoholic fatty liver disease, but the precise mechanism requires further investigation. This study investigated the protective effects and underlying mechanisms of Polygonatum cyrtonema ethanol extract (PCE) against Non-alcoholic steatohepatitis (NASH) in mice. Methods UHPLC-MS/MS was utilized to analyze the metabolites of PCE. The NASH mouse model was establishment in C57BL/6J mice via high-fat diet (HFD) feeding for 12 weeks, and from the 9th week, mice were gavaged with PCE (100, 300, and 900 mg/kg/day), simvastatin (4 mg/kg) or saline. One hand, liver injury was assessed by serum enzymes, biochemistry, and histopathology; On the other hand, RNA-seq, qPCR, and Western blot were employed to investigate the related molecular mechanisms. Results 211 metabolites were identified through UHPLC-MS/MS analysis. PCE ameliorated HFD induced liver injury and improved hepatocellular degeneration and steatosis in a dose-dependent way. PCE restored the expression of AMPK, SIRT1, SREBP1 and PPAR-α both in mRNA and protein levels. RNAseq identified unique gene expression profiles in response to high-fat diet (HFD) compared to the PCE treatments. HFD-induced DEGs were attenuated or abolished following PCE treatments. Ingenuity pathway analysis of RNA-seq data revealed key canonical pathways and upstream molecules regulated by PCE. Conclusion Our findings confirm the ability of PCE in alleviating NASH and underscores AMPK/SIRT1 pathway as a potential theraputic target for NASH treatment.
Collapse
Affiliation(s)
- Dongliang Chen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yue Shen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Department of Pharmacy, Bijie City Qixingguan District Hospital of Traditional Chinese Medicine, Bijie, Guizhou, China
| | - Fang Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Bo Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Shangfu Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lisheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jie Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Zheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Xia Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
28
|
Amirkhizi F, Taghizadeh M, Khalese-Ranjbar B, Hamedi-Shahraki S, Asghari S. The clinical value of serum sirtuin-1 concentration in the diagnosis of metabolic dysfunction-associated steatotic liver disease. BMC Gastroenterol 2025; 25:27. [PMID: 39844087 PMCID: PMC11753077 DOI: 10.1186/s12876-025-03613-8] [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: 08/28/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease and can affect individuals without producing any symptoms. We aimed to explore the value of serum sirtuin-1 (Sirt-1) in the diagnosis of MASLD. METHODS This case-control study analyzed data collected from 190 individuals aged 20 to 60 years. Anthropometric parameters, demographic information, and serum biochemical variables-including glycemic parameters, lipid profiles, liver enzymes, and Sirt-1 levels-were assessed. The correlation between serum Sirt-1 and biochemical variables was examined using Pearson's correlation coefficient. Receiver operating characteristic (ROC) curve analysis was employed to evaluate the diagnostic value of serum Sirt-1 in the context of MASLD. RESULTS Serum Sirt-1 levels was significantly lower in the MASLD group (p < 0.001) and was inversely correlated with serum insulin (r = -0.163, p = 0.025), HOMA-IR (r = -0.169, p = 0.020) and triglyceride (r = -0.190, p = 0.009) and positively correlated with serum levels of high-density lipoprotein cholesterol (HDL-C) (r = 0.214, p = 0.003). The area under the curve (AUC) of Sirt-1 to predict the presence of MASLD was 0.76 (p < 0.001, 95% CI: 0.69, 0.82) with a sensitivity of 78.9, specificity of 61.1, positive predictive value (PPV) of 67.0%, and negative predictive value (NPV) of 74.0%. The optimal cutoff, determined using Youden's index, was 23.75 ng/mL. This indicates that serum Sirt-1 levels below 23.75 ng/mL may be indicative of MASLD. CONCLUSIONS The present study demonstrated that serum Sirt-1 levels were significantly lower in patients with MASLD. Furthermore, these levels were correlated with various metabolic parameters, including insulin resistance and the serum lipid profile. A serum Sirt-1 level below the cutoff of 23.75 ng/mL exhibited a significant association with the presence of MASLD, suggesting its potential utility in identifying patients with this condition.
Collapse
Affiliation(s)
- Farshad Amirkhizi
- Department of Nutrition, School of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Mahdiyeh Taghizadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran
| | - Banafshe Khalese-Ranjbar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran
| | - Soudabeh Hamedi-Shahraki
- Department of Epidemiology and Biostatistics, School of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Somayyeh Asghari
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran.
| |
Collapse
|
29
|
Zhu N, Li Y, Lin Y, Cui X, Li X. Association between neutrophil-to-high-density lipoprotein cholesterol ratio and non-alcoholic fatty liver disease or metabolic dysfunction-associated steatotic liver disease: evidence from NHANES 2017-2020. Front Med (Lausanne) 2025; 11:1491858. [PMID: 39882525 PMCID: PMC11774988 DOI: 10.3389/fmed.2024.1491858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) or metabolic dysfunction-associated steatotic liver disease (MASLD) is closely associated with chronic inflammation and lipid metabolism disorders. The neutrophil-to-high-density lipoprotein cholesterol ratio (NHR) is an integrative marker reflecting inflammatory responses and lipid metabolism disorders and is associated with various diseases. This cross-sectional study aimed to determine the association between NHR and NAFLD, MASLD, and liver fibrosis. Methods Data for this study were obtained from the 2017-2020 National Health and Nutrition Examination Survey (NHANES), we employed weighted multiple regression and restricted cubic spline (RCS) analysis to assess the relationship between NHR and NAFLD, MASLD, and liver fibrosis. Additionally, we performed stratified analyses based on gender, age, body mass index, diabetes, hypertension, smoking status, and history of cardiovascular disease to evaluate the consistency of these associations across different subgroups. Results A total of 6,526 participants were included in the study. 2,839 (weighted 44.1%) participants were diagnosed with NAFLD and 2,813 (weighted 43.7%) participants were diagnosed with MASLD. After adjusting for confounders, NHR was positively associated with the risk of NAFLD/MASLD, and the correlation was particularly significant in the subgroups of females, those without hypertension, and those without diabetes (p < 0.05). By the NHR quartile, the risk of NAFLD/MASLD increased progressively with higher NHR levels (P for trend <0.001). In addition, RCS analysis showed a nonlinear association between NHR and NAFLD/MASLD and liver fibrosis (P-non-linear <0.05). Conclusion NHR may serve as a potential marker for NAFLD/MASLD and liver fibrosis, and lowering NHR levels could help reduce the incidence of these conditions.
Collapse
Affiliation(s)
- Na Zhu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yanyan Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yingying Lin
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing, China
| | - XinYu Cui
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xin Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing, China
| |
Collapse
|
30
|
Chen MQ, Wang A, Wan CX, Ruan BQ, Tong J, Shen JY. Prognostic value of atherogenic index of plasma in pulmonary hypertension. Front Med (Lausanne) 2025; 11:1490695. [PMID: 39871832 PMCID: PMC11769793 DOI: 10.3389/fmed.2024.1490695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/16/2024] [Indexed: 01/29/2025] Open
Abstract
Background The atherogenic index of plasma (AIP) is a brand-new lipid parameter that has been used to assess various cardiovascular events. This study aimed to investigate the prognostic value of AIP in patients with pulmonary hypertension (PH). Methods This retrospective study was conducted at Shanghai Jiao Tong University School of Medicine affiliated Renji Hospital, and included data from 125 PH patients treated during 2014-2018. The endpoint events of this study were clinical worsening outcomes. PH patients include those from group 1 and group 4. AIP was determined as the logarithm of the blood triglycerides ratio to high-density lipoprotein cholesterol. Results The 1-year, 3-year, and 5-year incidence rates of clinical worsening outcomes in PH patients in this study were 20.0, 44.8, and 54.4%, respectively. The median age of the PH patients was 38.00 years, with females accounting for 90.4%. After controlling for multivariable factors, the results of Cox regression analysis indicated that AIP was an independent predictor of adverse outcomes with a hazard ratio and 95% confident interval (CI) of 2.426 (1.021-5.763). The positive linear relationship of AIP was evaluated using restricted cubic spline analysis. Kaplan-Meier curves showed a significantly higher events rate in patients with AIP ≥ 0.144 compared to those with AIP < 0.144 (p = 0.002). Four potential prognostic variables, including AIP, were identified by LASSO regression to construct a nomogram. Compared to the model minus AIP, the AUC of the nomogram displayed a non-significant improvement (0.749 vs. 0.788, p = 0.298). In contrast, the results of net reclassification improvement (0.306, 95% CI: 0.039-0.459, p < 0.001) and integrated discrimination improvement (0.049, 95% CI: 0.006-0.097, p = 0.020) demonstrated significant enhancements in the predictive ability of the model when AIP was added to the clinical model. Conclusion AIP is an independent predictor of long-term clinical worsening in PH patients, and its inclusion in prognostic models could improve risk stratification and management.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie-Yan Shen
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Khalafi M, Rosenkranz SK, Ghasemi F, Kheradmand S, Habibi Maleki A, Korivi M, Tsao JP. Efficacy of intermittent fasting on improving liver function in individuals with metabolic disorders: a systematic review and meta-analysis. Nutr Metab (Lond) 2025; 22:1. [PMID: 39762987 PMCID: PMC11706068 DOI: 10.1186/s12986-024-00885-x] [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: 06/26/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Intermittent fasting (IF) can be an effective dietary therapy for weight loss and improving cardiometabolic health. However, there is scant evidence regarding the role of IF on indicators of liver function, particularly in adults with metabolic disorders. Therefore, we performed a systematic review and meta-analysis to investigate the effects of IF on liver function in adults with metabolic disorders. METHODS Three primary electronic databases including PubMed, Web of Science, and Scopus, were searched from inception to September 2024 to identify original studies that used IF interventions with or without control groups in adults with metabolic disorders. Inclusion criteria were (1) studies of human participants with metabolic diseases, (2) interventions that evaluated the effects of IF, (3) with or without a control group, and (4) measured liver fat, liver steatosis, liver fibrosis, or liver enzymes, including alanine aminotransferase (ALT) and aspartate aminotransferase (AST) as primary outcomes. Standardized mean differences (SMD) and 95% confidence intervals were calculated using random effects models. Heterogeneity was assessed using the Cochran's Q statistic and I-squared statistic (I2). Publication bias was assessed using the visual inspection of funnel plots and Egger's tests. The risk of bias was assessed using the PEDro scale and the NIH quality assessment tool. RESULTS A total 21 studies involving 1,226 participants with metabolic disorders were included in the meta-analysis. Overall, IF effectively decreased liver fat with a large effect size [SMD: -1.22 (95% CI: -1.63 to -0.80), p = 0.001], liver steatosis with a medium effect size [SMD: -0.73 (95% CI: -1.12 to -0.35), p = 0.001], ALT with a small effect size [SMD: -0.44 (95% CI: -0.58 to -0.30), p = 0.001], and AST with a small effect size [SMD: -0.30 (95% CI: -0.49 to -0.11), p = 0.001], but not liver fibrosis [SMD: -0.28 (95% CI: -0.59 to 0.02), p = 0.07]. Subgroup analyses showed that IF decreased liver fat and ALT significantly, independent of IF mode, participant age, health status, weight status, and intervention duration. IF significantly decreased liver fibrosis in those with obesity; and decreased AST following 5:2 diets, in middle-aged adults, adults with obesity, and regardless of health status or intervention duration. CONCLUSIONS IF seems to be an effective dietary therapy for improving liver function in adults with metabolic disorders, and many of liver function-related benefits occur regardless of IF mode, intervention duration, or participant health status. LIMITATIONS Significant heterogeneity, small numbers of studies and inclusion of non-randomized trials or single-group pre-post trials were the main limitation of our meta-analysis. Further randomized clinical trials are needed to elucidate the effects of IF on liver function in adults with metabolic disorders.
Collapse
Affiliation(s)
- Mousa Khalafi
- Department of Physical Education and Sport Sciences, Faculty of Humanities, University of Kashan, Kashan, Iran
| | - Sara K Rosenkranz
- Department of Kinesiology and Nutrition Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Faeghe Ghasemi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Guilan, Iran
| | - Shokoufeh Kheradmand
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Mazandaran, Babolsar, Iran
| | - Aref Habibi Maleki
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mallikarjuna Korivi
- Institute of Human Movement and Sports Engineering, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China.
| | - Jung-Piao Tsao
- Department of Sports Medicine, China Medical University, Taichung City, Taiwan.
| |
Collapse
|
32
|
Xuan Y, He F, Liu Q, Dai D, Wu D, Shi Y, Yao Q. Elevated GGT to HDL ratio as a marker for the risk of NAFLD and liver fibrosis. Sci Rep 2025; 15:10. [PMID: 39748013 PMCID: PMC11695851 DOI: 10.1038/s41598-024-84649-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025] Open
Abstract
This study investigated the association between NAFLD and liver fibrosis and the ratio of gamma-glutamyl transferase to high-density lipoprotein cholesterol (GGT/HDL-C). In this cross-sectional study, we included 4764 subjects who participated in the National Health and Nutrition Examination Survey (NHANES) during 2017-2018. Adjusted multivariate logistic regression analysis was utilized to evaluate the relationships between GGT/HDL-C levels and NAFLD, fatty liver degree, and liver fibrosis. The non-linear link between NAFLD and the GGT/HDL-C ratio was examined using generalized additive models. There was a non-linear association between GGT/HDL-C and the risk of NAFLD, and all regression models demonstrated a strong relationship between GGT/HDL-C levels and the risk of liver fibrosis, the degree of hepatic steatosis, and the prevalence of NAFLD. Subgroup analyses revealed a significant correlation between the risk of NAFLD and the GGT/HDL-C ratio among Mexican Americans and young people in the 20-40 age range. The receiver operating characteristic (ROC) study showed that GGT/HDL-C was a more accurate predictor of NAFLD than GGT or HDL-C alone. In the U.S. population, an increased risk of NAFLD, the severity of hepatic steatosis, and the risk of liver fibrosis are independently correlated with an elevated GGT/HDL-C ratio.
Collapse
Affiliation(s)
- Yanyan Xuan
- Department of Hospital Infection, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Department of Hepatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Department of Geriatrics Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Fangfang He
- Department of Hospital Infection, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qing Liu
- Department of Hepatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Dandan Dai
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Dingting Wu
- Department of Clinical Nutrition, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanmei Shi
- Department of Hepatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.
| | - Qi Yao
- Department of Geriatrics Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
33
|
Li Z, Li J, He S, Chen J, Deng C, Duan J. Ellagic Acid Modulates Necroptosis, Autophagy, Inflammations, and Stress to Ameliorate Nonalcoholic Liver Fatty Disease in a Rat Model. Food Sci Nutr 2025; 13:e4694. [PMID: 39830906 PMCID: PMC11742184 DOI: 10.1002/fsn3.4694] [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: 08/28/2024] [Revised: 10/25/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered one of the most common metabolic disorders worldwide. Although the pathoetiology of NAFLD is not fully elucidated, recent evidence suggests the involvement of stress, inflammation, and programmed death in the onset and progression of the disease. This investigation aimed to evaluate the effects of ellagic acid (EA), a known herbal antioxidant, on a high-fat diet (HFD)-induced animal model of NAFLD by evaluating the status of lipid profile, necroptosis (RIPK1, RIPK3, and MLKL), autophagy (LC3, ATG5, and BECN1), inflammation (TNF-α, IL-6, IL-4, and IL-10), and stress (SOD, CAT, GR, GPx, and MDA). In this regard, rats were randomly divided into 6 groups as follows: normal diet controls, HFD (supplemented with high caloric diet model), EA low dose (HFD and 10 mg/kg/day EA), EA middle dose (HFD and 25 mg/kg/day EA), EA high dose (HFD and 50 mg/kg/day EA), and Rosiglitazone (HFD and 10 mg/kg/day Rosi). After the treatment, the levels of markers related to necroptosis and autophagy in the liver tissue as well as the lipid profiles, inflammation, and oxidative stress status were analyzed. It was shown that the dose of EA was able to improve the weight gain and lipid profile when compared to NAFLD animals (p-value < 0.001). Moreover, EA increased the level of LC3 and ATG5 while decreasing BECN 1, RIPK1, RIPK3, and MLKL compared to the HFD-induced NAFLD rats (p-value < 0.05). TNF-α and IL-6 were decreased after EA administration, whereas IL-4 and IL-10 levels were increased (p-value < 0.001). Furthermore, the increase in the activity of SOD, CAT, GR, and GPx along with the decrease in MDA levels indicated the suppression of oxidative stress by EA treatment compared to the NAFLD rats (p-value < 0.0001). The current findings may suggest that EA improves NAFLD via modulation of necroptosis, autophagy, inflammation, and stress.
Collapse
Affiliation(s)
- Zhuoheng Li
- Gastroenterology DepartmentKunming Children's HospitalKunmingChina
| | - Juan Li
- Gastroenterology DepartmentKunming Children's HospitalKunmingChina
| | - Shuli He
- Gastroenterology DepartmentKunming Children's HospitalKunmingChina
| | - Jun Chen
- Gastroenterology DepartmentKunming Children's HospitalKunmingChina
| | - Chengjun Deng
- Gastroenterology DepartmentKunming Children's HospitalKunmingChina
| | - Jintao Duan
- Gastroenterology DepartmentKunming Children's HospitalKunmingChina
| |
Collapse
|
34
|
Flam E, Haas JT, Staels B. Liver metabolism in human MASLD: A review of recent advancements using human tissue metabolomics. Atherosclerosis 2025; 400:119054. [PMID: 39586140 DOI: 10.1016/j.atherosclerosis.2024.119054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/27/2024]
Abstract
Global incidence of Metabolic dysfunction-Associated Steatotic Liver Disease (MASLD) is on the rise while treatments remain elusive. MASLD is a disease of dysregulated systemic and hepatic metabolism. Current understanding of disease pathophysiology as it relates to metabolome changes largely comes from studies on animal models and human plasma. However, human tissue data are crucial for transitioning from mechanisms to clinical therapies. The close relationship between MASLD and comorbidities like obesity, type 2 diabetes and dyslipidemia make it difficult to determine the contribution from liver disease itself. Here, we review recent metabolomics studies in liver tissue from human MASLD patients, which have predominately focused on lipid metabolism, but also include bile acid, tricarboxylic acid (TCA) cycle, and branched chain amino acid (BCAA) metabolism. Several clinical trials are underway to target various of these lipid-related pathways in MASLD. Although only the β-selective thyroid hormone receptor agonist resmetirom has so far been approved for use, many metabolism-targeting pharmaceuticals show promising results for halting disease progression, if not promoting outright reversal. Ultimately, the scarcity of human tissue data and the variability of confounding factors, like obesity, within and between cohorts are impediments to the pathophysiological understanding required for efficient development of metabolic treatments.
Collapse
Affiliation(s)
- Emily Flam
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Joel T Haas
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
35
|
Kwon Y, Gottmann P, Wang S, Tissink J, Motzler K, Sekar R, Albrecht W, Cadenas C, Hengstler JG, Schürmann A, Zeigerer A. Induction of steatosis in primary human hepatocytes recapitulates key pathophysiological aspects of metabolic dysfunction-associated steatotic liver disease. J Hepatol 2025; 82:18-27. [PMID: 38977136 DOI: 10.1016/j.jhep.2024.06.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 06/15/2024] [Accepted: 06/29/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common cause of chronic liver disease. Owing to limited available treatment options, novel pre-clinical models for target selection and drug validation are warranted. We have established and extensively characterized a primary human steatotic hepatocyte in vitro model system that could guide the development of treatment strategies for MASLD. METHODS Cryopreserved primary human hepatocytes from five donors varying in sex and ethnicity were cultured with free fatty acids in a 3D collagen sandwich for 7 days and the development of MASLD was followed by assessing classical hepatocellular functions. As proof of concept, the effects of the drug firsocostat (GS-0976) on in vitro MASLD phenotypes were evaluated. RESULTS Incubation with free fatty acids induced steatosis, insulin resistance, mitochondrial dysfunction, inflammation, and alterations in prominent human gene signatures similar to patients with MASLD, indicating the recapitulation of human MASLD in this system. The application of firsocostat rescued clinically observed fatty liver disease pathologies, highlighting the ability of the in vitro system to test the efficacy and potentially characterize the mode of action of drug candidates. CONCLUSIONS Altogether, our human MASLD in vitro model system could guide the development and validation of novel targets and drugs for the treatment of MASLD. IMPACT AND IMPLICATIONS Due to low drug efficacy and high toxicity, clinical treatment options for metabolic dysfunction-associated steatotic liver disease (MASLD) are currently limited. To facilitate earlier stop-go decisions in drug development, we have established a primary human steatotic hepatocyte in vitro model. As the model recapitulates clinically relevant MASLD characteristics at high phenotypic resolution, it can serve as a pre-screening platform and guide target identification and validation in MASLD therapy.
Collapse
Affiliation(s)
- Yun Kwon
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Pascal Gottmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany; German Institute of Human Nutrition (DIfE), Department of Experimental Diabetology, Nuthetal, Germany
| | - Surui Wang
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Joel Tissink
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karsten Motzler
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Revathi Sekar
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Department of Toxicology, Dortmund, Germany
| | - Cristina Cadenas
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Department of Toxicology, Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Department of Toxicology, Dortmund, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany; German Institute of Human Nutrition (DIfE), Department of Experimental Diabetology, Nuthetal, Germany
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
36
|
Wang P, Fan Y, Gao H, Wang B. Body roundness index as a predictor of all-cause and cardiovascular mortality in patients with diabetes and prediabetes. Diabetes Res Clin Pract 2025; 219:111958. [PMID: 39675484 DOI: 10.1016/j.diabres.2024.111958] [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: 10/01/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND There are limited population-based studies examining the correlation between body roundness index (BRI) and mortality in diabetes and prediabetes patients. METHOD Our final analysis encompassed 15,848 patients with diabetes and prediabetes sourced from the National Health and Nutrition Examination Survey(NHANES) spanning from 2003 to 2018. Cox proportional hazards model and restricted cubic splines (RCS) were utilized to assess the correlation between BRI and both all-cause mortality and cardiovascular mortality. RESULTS During an average follow-up period of 92.9 months, 2655 participants (12.73 %) died, including 730 (3.44 %) from cardiovascular diseases. RCS demonstrated a U-shaped nonlinear association between BRI with all-cause mortality and cardiovascular mortality, with threshold values of 5.54 and 5.21, respectively. When BRI was below the threshold, a negative correlation was observed between BRI and all-cause mortality (HR 0.87, 95 % CI 0.81-0.93).The correlation with cardiovascular mortality is not significant. Conversely, when BRI was above the threshold, a positive correlation was observed between BRI with all-cause mortality (HR 1.10, 95 % CI 1.06-1.14) and cardiovascular mortality (HR 1.13, 95 % CI 1.07-1.20). CONCLUSION Our research indicates that among US adults with diabetes or prediabetes, BRI exhibits a U-shaped relationship with all-cause and cardiovascular mortality, with threshold values of 5.54 and 5.21, respectively.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Respiratory, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yongqiang Fan
- Department of General Surgery, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Haoyue Gao
- Department of Respiratory, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Bei Wang
- Department of Respiratory, The Second Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
37
|
Liu A, Sun Y, Qi X, Zhou Y, Zhou J, Li Z, Wu X, Zou Z, Lv X, Li H, Li Y. Association between the ratio of serum uric acid to high-density lipoprotein cholesterol and liver fat content: evidence from a Chinese health examination dataset. Sci Rep 2024; 14:31397. [PMID: 39733068 PMCID: PMC11682173 DOI: 10.1038/s41598-024-83013-2] [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: 08/15/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
Despite numerous studies investigating the correlation between the serum uric acid and high-density lipoprotein cholesterol ratio (UHR) and fatty liver disease, the evidence for the dose-response relationship between UHR and liver fat content (LFC) remains uncertain. This study employs quantitative computed tomography (CT) to quantify LFC and aims to investigate the correlation and dose-response relationship between UHR levels and LFC in Chinese adults. Based on the health check-up data from 2021 at Henan Provincial People's Hospital, China, the objective of this cross-sectional study was to investigate the association between UHR levels and LFC among individuals of different genders. The analytical approach encompassed one-way ANOVA, multiple regression analysis, subgroup analysis, smooth curve fitting, and the evaluation of threshold and saturation effects. Upon adjusting for potential influencing factors, the multiple regression analysis indicated a positive correlation between UHR and LFC in both male and female subjects. This positive correlation was more significant in the highest UHR quartile (Male Q4 in model II: β = 2.119, 95% CI: 1.353-2.886, P < 0.05; Female Q4 in model II: β = 1.312, 95% CI: 0.499-2.124, P < 0.05). Subgroup and threshold saturation effect analyses demonstrated a positive correlation between UHR and LFC in the male population, independent of age, although the linear correlation trend was influenced by different body mass index (BMI) groups. In the female population, age also affected the association between UHR and LFC, with a negative association observed when age ≥ 45 years and UHR > 30.63. A positive association exists between UHR levels and LFC in both genders among Chinese adults, albeit exhibiting variations across different age and BMI groups. Consequently, early monitoring of UHR levels may be crucial for the early detection and intervention in high-risk groups exhibiting increased LFC.
Collapse
Affiliation(s)
- Ao Liu
- Department of Medical Imaging, People's Hospital of Zhengzhou University, #7 Wei Wu Road, Zhengzhou, 450003, China
| | - Yongbing Sun
- Department of Medical Imaging, People's Hospital of Zhengzhou University, #7 Wei Wu Road, Zhengzhou, 450003, China
| | - Xin Qi
- Department of Medical Imaging, Henan Provincial People's Hospital, Xinxiang Medical College, Zhengzhou, 450003, China
| | - Yang Zhou
- Department of Medical Imaging, People's Hospital of Zhengzhou University, #7 Wei Wu Road, Zhengzhou, 450003, China
| | - Jing Zhou
- Department of Health Management, Chronic Health Management Laboratory, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Zhonglin Li
- Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Xiaoling Wu
- Department of Nuclear Medicine, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Zhi Zou
- Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Xue Lv
- Department of Health Management, Chronic Health Management Laboratory, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Hao Li
- Department of Health Management, Fuwai Central China Cardiovascular Hospital, #1 Fuwai Avenue, Zhengzhou, 451464, China
| | - Yongli Li
- Department of Health Management, Chronic Health Management Laboratory, Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
38
|
Wang W, Huang L, Qiu XP, Tu M, Guo XL. Monocytes to Apolipoprotein A1 ratio is associated with metabolic dysfunction-associated fatty liver disease in type 2 diabetes mellitus. Sci Rep 2024; 14:31396. [PMID: 39733102 PMCID: PMC11682227 DOI: 10.1038/s41598-024-82994-4] [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: 04/14/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
The monocyte-to-Apolipoprotein A1 ratio (MAR) emerges as a potentially valuable inflammatory biomarker indicative of metabolic dysfunction-associated fatty liver disease (MASLD). Accordingly, this investigation primarily aims to assess the correlation between MAR and MASLD risk. A cohort comprising 957 individuals diagnosed with type 2 diabetes mellitus (T2DM) participated in this study. The relationship between MAR and MASLD was analyzed through binomial logistic regression analysis and restricted cubic splines (RCS). Furthermore, a comparative assessment of MAR and monocyte to high-density lipoprotein ratio (MHR) in identifying MASLD efficacy was conducted using receiver operating characteristic curve analysis. Remarkably, even after adjusting for metabolic parameters and hepatic functional markers, MAR stood out as an independent predictor for MASLD (OR 1.58, 95% CI 1.36-1.84; P < 0.001) and displayed a nonlinear positive association with MASLD risk according to RCS analysis (P for nonlinearity and overall < 0.001). Notably, MAR exhibited superior diagnostic accuracy for identifying MASLD compared to MHR (AUC: 0.772 vs 0.722, P < 0.001). In summary, MAR emerges as a promising inflammatory indicator for MASLD, demonstrating potential as a valuable screening tool to bolster the management of MASLD within the T2DM population.
Collapse
Affiliation(s)
- Wei Wang
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, Fujian, China
| | - Lian Huang
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, Fujian, China
| | - Xiu Ping Qiu
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, Fujian, China
| | - Mei Tu
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, Fujian, China
| | - Xiu Li Guo
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 364000, Fujian, China.
| |
Collapse
|
39
|
Fan YQ, Wang H, Wang PP, Shi ZY, Wang Y, Xu J. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio as a predictive indicator of CKD risk in NAFLD patients: NHANES 2017-2020. Front Nutr 2024; 11:1501494. [PMID: 39777076 PMCID: PMC11703712 DOI: 10.3389/fnut.2024.1501494] [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: 09/25/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD) are both closely related to dyslipidemia. However, the relationship between dyslipidemia in patients with NAFLD and CKD is not yet clear. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) is an innovative and comprehensive lipid index. The purpose of this study was to investigate the correlation between NHHR and CKD risk in NAFLD patients with or without fibrosis. Methods This study used data from the National Health and Nutrition Examination Survey (NHANES) from 2017 to 2020 for analysis, including a total of 4,041 subjects diagnosed with NAFLD. Among the NAFLD subjects, 3,315 individuals without liver fibrosis and 726 individuals with fibrosis. Weighted multivariate linear regression, weighted logistic regression, restricted cubic spline (RCS) curves, and subgroup analysis were used to evaluate the correlation between NHHR and CKD in patients with NAFLD. Results Our findings indicate that in NAFLD subjects without liver fibrosis, the highest tertile of NHHR, as compared to the lowest tertile, was inversely related to glomerular filtration rate (eGFR) (β: -2.14, 95% CI: -3.97, -0.32, p < 0.05) and positively related to CKD (OR: 1.67, 95% CI: 1.12, 2.49, p < 0.05). No significant associations were observed between NHHR and eGFR, urinary albumin to creatinine ratio (ACR) in NAFLD subjects with liver fibrosis. The RCS revealed a linear relationship between NHHR and ACR, CKD in NAFLD subjects without liver fibrosis, while a U-shaped relationship was observed between NHHR and ACR, CKD in NAFLD subjects with liver fibrosis. Conclusion In patients with non-fibrotic NAFLD, a significantly elevated NHHR is closely associated with an increased risk of CKD and shows a linear relationship with CKD. In patients with fibrotic NAFLD, NHHR shows a U-shaped relationship with CKD. LD, Our findings underscore the practical utility of NHHR as a biomarker for early risk stratification of CKD in patients with NAFLD.
Collapse
Affiliation(s)
- Yong-Qiang Fan
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hao Wang
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Pei-Pei Wang
- Department of Respiratory, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhi-Yong Shi
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan Wang
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jun Xu
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
40
|
Ni H, Wu H, Wang J, Chan BKW, Chen K, Chan EWC, Li F, Chen S. Lincomycin as a growth-promoting antibiotic induces metabolic and immune dysregulation in animals. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177780. [PMID: 39612713 DOI: 10.1016/j.scitotenv.2024.177780] [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: 10/04/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Since animal growth promoters (AGPs) are used in large quantities and commonly released to the environment from animal farms, it is necessary to determine whether such agents should be regarded as an environmental toxin that poses a threat to the ecosystem and health risk to wildlife. In this study, a multi-omics approach was employed to explore the effects of a representative AGP, lincomycin, on key metabolic and physiological functions of animals, using a mouse model. The results indicated that exposure to lincomycin resulted in a significant increase in growth rate of mice (50.11 %) over an 8 weeks period, during which significant decrease (61.94 %) and increase (68.64 %) in the relative abundance of Firmicutes and Escherichia coli, respectively, was observed in the gut microbiota, indicating that the gut microbiota structure has been altered. Moreover, the mice exhibited altered lipid profiles and liver damage suggestive of early-stage non-alcoholic fatty liver disease (NAFLD). Disruptions in blood glucose and insulin levels associated with type 2 diabetes mellitus (T2DM) were also observed. Furthermore, lincomycin was found to cause suppression in inflammatory responses, as evidenced by the downregulation of related genes and elevated inflammatory mediators, potentially resulting in increased susceptibility to microbial infection. Our findings underscore the detrimental effects of lincomycin on animal health and highlight the necessity for comprehensive toxicological assessments of lincomycin and other AGPs before their environmental release.
Collapse
Affiliation(s)
- Hongyuhang Ni
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong; Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Haoze Wu
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Jing Wang
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Bill Kwan-Wai Chan
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Kaichao Chen
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Edward Wai-Chi Chan
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Fuyong Li
- Department of Animal Science and Technology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sheng Chen
- Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong; Shenzhen Key Lab for Food Biological Safety Control, The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China.
| |
Collapse
|
41
|
Long C, Wang X, Wang D, Chen Y, Zhang B. Deciphering the impact of heavy metal mixed exposure on lipid metabolism using three statistical models. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2024; 47:20. [PMID: 39692884 DOI: 10.1007/s10653-024-02328-1] [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: 04/28/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Lipid metabolism disorders pose a significant threat to human health. However, the relationship between heavy metal mixed exposure and lipid metabolism remains poorly understood. This study recruited 1717 residents living near a chromium factory in northeast China. The concentrations of blood Cr, Mn, Cd, Pb, V, and serum CHOL, TG, LDL and HDL levels were measured. Generalized linear model (GLM), quantile g-computation (Qg-comp), and Bayesian kernel machine regression (BKMR) were simultaneously employed to investigate the associations between heavy metal mixed exposure and lipid markers levels. GLM analysis revealed significant associations between blood Cr concentration and HDL (β = -0.07; 95%CI: -0.09, -0.05), LDL (β = -0.06; 95%CI: -0.11, -0.02), and CHOL (β = 0.07; 95%CI: 0.01, 0.12) levels. V concentration was positively associated with HDL (β = 0.12; 95%CI: 0.06, 0.18) and LDL (β = 0.17; 95%CI: 0.04, 0.30) levels. Qg-comp analysis indicated a negative association between heavy metal mixed exposure and HDL (β = -0.040; 95%CI: -0.073, -0.006) level. BKMR model further confirmed the negative relationship between heavy metal mixed exposure and HDL, with the interaction between blood Cr (> 1.05 μg/L) and blood V (> 5.16 μg/L) contributing to decreased HDL levels. Our findings suggested that heavy metal mixed exposure had impacts on HDL and CHOL levels, and the Cr and V may mutually play a predominant role in the observed abnormal HDL levels.
Collapse
Affiliation(s)
- Changmao Long
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Xiangjun Wang
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Dongsheng Wang
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yuqing Chen
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Baojun Zhang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
42
|
Tavaglione F, Marafioti G, Romeo S, Jamialahmadi O. Machine Learning Reveals the Contribution of Lipoproteins to Liver Triglyceride Content and Inflammation. J Clin Endocrinol Metab 2024; 110:218-227. [PMID: 38833012 PMCID: PMC11651681 DOI: 10.1210/clinem/dgae371] [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: 01/11/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
CONTEXT Metabolic dysfunction-associated steatotic liver disease (MASLD) is currently the most common chronic liver disease worldwide and is strongly associated with metabolic comorbidities, including dyslipidemia. OBJECTIVE Herein, we aim to estimate the prevalence of MASLD and metabolic dysfunction-associated steatohepatitis (MASH) in Europeans with isolated hypercholesterolemia and isolated hypertriglyceridemia in the UK Biobank and to estimate the independent contribution of lipoproteins to liver triglyceride content. METHODS We selected 218 732 Europeans from the UK Biobank without chronic viral hepatitis and other causes of liver disease, of whom 14 937 with liver magnetic resonance imaging data available. Next, to examine the relationships between traits in predicting liver triglyceride content, we compared the predictive performance of several machine learning methods and selected the best performing algorithms based on the minimum cross-validated mean squared error (MSE). RESULTS There was an approximately 3-fold and 4-fold enrichment of MASLD and MASH in individuals with isolated hypertriglyceridemia (P = 1.23 × 10-41 and P = 1.29 × 10-10, respectively), whereas individuals with isolated hypercholesterolemia had a marginal higher rate of MASLD and no difference in MASH rate compared with the control group (P = .019 and P = .97, respectively). Among machine learning methods, the feed-forward neural network had the best cross-validation MSE on the validation set. Circulating triglycerides, after body mass index, were the second strongest independent predictor of liver proton density fat fraction with the largest absolute mean Shapley additive explanation value. CONCLUSION Isolated hypertriglyceridemia is the second strongest, after obesity, independent predictor of MASLD/MASH. Individuals with hypertriglyceridemia, but not with hypercholesterolemia, should be screened for liver disease.
Collapse
Affiliation(s)
- Federica Tavaglione
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Giuseppe Marafioti
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Stefano Romeo
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, 41345 Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
- Department of Medicine, Karolinska Institutet, Huddinge, 14152 Stockholm, Sweden
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, 41345 Gothenburg, Sweden
| |
Collapse
|
43
|
Wang W, Li X, Lv D, Wu X, Xie F, Xie W, Wang J, Zhao Z. Analyzing lipid profiles and dyslipidemia prevalence in hypertensive patients: a cross-sectional study from primary community health institutions. Front Med (Lausanne) 2024; 11:1425414. [PMID: 39741514 PMCID: PMC11685003 DOI: 10.3389/fmed.2024.1425414] [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: 05/14/2024] [Accepted: 10/15/2024] [Indexed: 01/03/2025] Open
Abstract
Background A significant proportion of hypertensive patients also suffer from comorbid dyslipidemia, which critically influences their treatment outcomes and overall prognosis. Given its implications, the lipid profiles of hypertensive individuals warrant increased attention for more effective clinical management. Methods We analyzed data from 92,443 hypertensive patients registered at primary community health institutions in 2021. Employing a cross-sectional study design, we assessed the distribution of lipid levels and the prevalence of various dyslipidemia subtypes. Stepwise forward logistic regression was used to identify factors associated with dyslipidemia, adjusting for gender, age, body size, and other relevant characteristics. Results According to the 2023 Chinese Guidelines for the Management of Lipids, the overall prevalence of dyslipidemia was 37.5%. Subtype analysis revealed prevalence of high total cholesterol (TC) at 11.2%, high triglycerides (TG) at 16.0%, low high-density lipoprotein cholesterol (HDL-C) at 16.0%, and high low-density lipoprotein cholesterol (LDL-C) at 10.2%. TG abnormalities were more common among males (16.8%), whereas TC abnormalities predominated in females (14.4%). Notably, hypertensive patients with diabetes had higher levels of TG compared to non-diabetics (p = 0.009). Those with stroke and liver disease comorbidities exhibited lower TG levels than their counterparts (p = 0.018 and p < 0.001, respectively). Additionally, HDL-C levels were significantly lower in hypertensives with diabetes, coronary artery disease, and central obesity (p < 0.001, p = 0.026, p < 0.001, respectively). Regression analysis indicated that dyslipidemia prevalence correlates significantly with gender, age, diabetes, coronary heart disease, stroke, family history of hypertension, body mass index (BMI), central obesity, frequency of physical activity, smoking status, regular alcohol consumption, and abdominal ultrasound findings. Conclusion Our study underscores the necessity of rigorous lipid monitoring and analysis of dyslipidemia-influencing factors for the development of effective health management strategies within the community. There is a critical need to examine lipid profiles comprehensively and implement targeted therapeutic interventions aimed at managing hyperlipidemia, a modifiable risk factor for cardiovascular disease.
Collapse
Affiliation(s)
- Wenxin Wang
- School of Public Health/Institute of Local Government Development, Shantou University, Shan-Tou, China
| | - Xinmin Li
- School of Public Health, Shantou University, Shan-Tou, China
| | - Deliang Lv
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Xiaobing Wu
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Fengzhu Xie
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Wei Xie
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Jinxiao Wang
- School of Public Health, Shantou University, Shan-Tou, China
| | - Zhiguang Zhao
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| |
Collapse
|
44
|
Tan EY, Muthiah MD, Sanyal AJ. Metabolomics at the cutting edge of risk prediction of MASLD. Cell Rep Med 2024; 5:101853. [PMID: 39657668 PMCID: PMC11722125 DOI: 10.1016/j.xcrm.2024.101853] [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: 07/25/2024] [Revised: 10/12/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major public health threat globally. Management of patients afflicted with MASLD and research in this domain are limited by the lack of robust well-established non-invasive biomarkers for diagnosis, prognostication, and monitoring. The circulating metabolome reflects both the systemic metabo-inflammatory milieu and changes in the liver in affected individuals. In this review we summarize the available literature on changes in the different components of the metabolome in MASLD with a focus on changes that are linked to the presence of underlying steatohepatitis, severity of disease activity, and fibrosis stage. We further summarize the existing literature around biomarker panels that are derived from interrogation of the metabolome. Their relevance to disease biology and utility in practice are also discussed. We further highlight potential direction for future studies particularly to ensure they are fit for purpose and suitable for widespread use.
Collapse
Affiliation(s)
- En Ying Tan
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore.
| | - Mark D Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
45
|
Wu Q, Peng Y, Gong C. Role of waist circumference, body mass index and high-sensitivity C-reactive protein in pediatric steatotic liver disease: A cross-sectional study. Ann Hepatol 2024; 30:101759. [PMID: 39638037 DOI: 10.1016/j.aohep.2024.101759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 09/23/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION AND OBJECTIVES Relationships and interactions among waist circumference (WC), body mass index (BMI) and high-sensitivity C-reactive protein (hs-CRP) with steatotic liver disease (SLD) in children have rarely been studied as a whole. We aimed to investigate the association among WC, BMI and hs-CRP with SLD and its related metabolic indictors. MATERIALS AND METHODS A total of 10,776 children aged 10-15 years were screened in our study. Anthropometric data, biochemical parameters and ultrasound assessments were collected. Metabolic indictors between children with and without SLD were compared. The correlation of waist circumference Z score (ZWC), body mass index Z score (ZBMI) and hs-CRP with SLD and its related metabolic indictors, and the interactive effect between ZWC with hs-CRP and ZBMI with hs-CRP upon SLD, respectively, was tested. RESULTS A total of 543 children with normal BMI (n = 287) and high BMI (n = 256) were examined. Hs-CRP, ZWC and ZBMI were all found to significantly correlate with SLD and its related metabolic indexes. The interaction effect analysis showed that ZWC and male was independent risk factor of SLD with OR (95 %CI) of 23.431 (7.253, 75.697) and 7.927 (2.766,22.713), respectively, whereas the same effect wasn't found in ZBMI. The cut-off value of ZWC for the prediction of SLD was 1.494 and 1.541 in boys and girls, respectively. CONCLUSIONS Increased WC, BMI and hs-CRP exerts adverse effect in pediatric SLD and its related metabolic indictors. WC and male gender could be independent risk factors for SLD, and WC was a powerful index for the prediction of SLD in children aged 10-15 years.
Collapse
Affiliation(s)
- Qiaoling Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Yongmei Peng
- Children's Hospital of Fudan University, Shanghai, China; Shanghai Center for Women and Children's Health, Shanghai, China.
| | - Chundan Gong
- Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
46
|
Xiao Y, Xu S, Hu W, Huang J, Jiang D, Na R, Yin Z, Zhang J, Chen H. Metabolic dysfunction-associated steatotic liver disease and risk of four intrahepatic and extrahepatic diseases. Ann Hepatol 2024; 30:101750. [PMID: 39638041 DOI: 10.1016/j.aohep.2024.101750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/15/2024] [Accepted: 09/30/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION AND OBJECTIVES Recently, Delphi consensus proposed an overarching term steatotic liver disease (SLD), with various subcategories such as MASLD, MetALD and ALD. Our aim was to investigate the association between MASLD/MetALD/ALD and four intrahepatic and extrahepatic diseases (liver diseases, renal diseases, cardiovascular diseases, and cancers) in the UK Biobank cohort. PATIENTS AND METHODS By defining hepatic steatosis as image-derived phenotype (IDP)-PDFF >5.21%, we used data from the UK Biobank to diagnose MASLD/ MetALD/ALD. The odd ratio (OR) and the hazard ratio (HR) were calculated using the logistic regression modals and Cox regression models, respectively. RESULTS Among 39,230 eligible individuals, 6,865 MASLD subjects, 2,379 MetALD subjects and 884 ALD subjects were diagnosed. The last follow-up time was October 13, 2023. Consistent with the logistic analyses, MASLD/MetALD/ALD were significantly associated with a higher risk of liver diseases (HR=3.04 [95%CI:2.60-3.56], HR = 2.69 [95% CI: 2.12-3.42] and HR =3.99 [95%CI:2.92-5.45], respectively). Subjects with MASLD also had an increased higher risk of renal diseases (HR = 1.40 [95%CI:1.20-1.64]) and subjects with ALD had an increased higher risk of cancers (HR = 1.36 [95%CI:1.15-1.60]). CONCLUSION It is the first study to report the association between MASLD, MetALD, ALD and common intrahepatic and extrahepatic diseases based on magnetic resonance imaging data-PDFF. We found that MASLD, MetALD and ALD were risk factors for liver diseases. Meanwhile, MASLD was also a risk factor for renal diseases and ALD was a risk factor for cancers.
Collapse
Affiliation(s)
- Yiyuan Xiao
- School of Public Health (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China.
| | - Sihua Xu
- School of Public Health (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China.
| | - Wenyan Hu
- School of Public Health (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China.
| | - Jiapeng Huang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 in Nanjing North Street, Heping Distinct, Shenyang, Liaoning Province, China.
| | - Deke Jiang
- State Key Laboratory of Organ Failure Research, Guangdong Key Laboratory of Viral Hepatitis Research, Institutes of Liver Diseases Research of Guangdong Province, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, China.
| | - Rong Na
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam Hong Kong SAR, Hong Kong, China (Hong Kong).
| | - Zhaoqing Yin
- Department of Pediatric, Dehong People Hospital, 13 Yonghan Street, Mangshi, Dehong Dai and Jingpo Autonomous Prefecture, Yunnan, China.
| | - Jingjing Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China.
| | - Haitao Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, China.
| |
Collapse
|
47
|
Xiang H, Lyu Q, Chen S, Ouyang J, Xiao D, Liu Q, Long H, Zheng X, Yang X, Lu H. PACS2/CPT1A/DHODH signaling promotes cardiomyocyte ferroptosis in diabetic cardiomyopathy. Cardiovasc Diabetol 2024; 23:432. [PMID: 39633391 PMCID: PMC11619700 DOI: 10.1186/s12933-024-02514-6] [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: 08/09/2024] [Accepted: 11/17/2024] [Indexed: 12/07/2024] Open
Abstract
OBJECTIVES The pathophysiology of diabetic cardiomyopathy (DCM) is a phenomenon of great interest, but its clinical problems have not yet been effectively addressed. Recently, the mechanism of ferroptosis in the pathophysiology of various diseases, including DCM, has attracted widespread attention. Here, we explored the role of PACS2 in ferroptosis in DCM through its downregulation of PACS2 expression. METHODS AND RESULTS Cardiomyocytes were treated with high glucose and palmitic acid (HGPA), and the detection of cardiomyocyte iron ions, lipid peroxides, and reactive oxygen species (ROS) revealed clear ferroptosis during these treatments. Silencing PACS2 downregulated CPT1A expression and upregulated DHODH expression significantly, reversing HGPA-induced ferroptosis. Further silencing of PACS2 with a CPT1A agonist exacerbated cardiomyocyte ferroptosis while promoting mitochondrial damage in cardiomyocytes. Using a mouse model of type 2 diabetes induced by streptozotocin (STZ) and a high-fat diet (HFD), we found that PACS2 deletion reversed these treatment-induced increases in cellular iron ions, impaired cardiac function, mitochondrial damage and ferroptosis in cardiac muscle tissues. CONCLUSIONS The PACS2/CPT1A/DHODH signalling pathway may be involved in ferroptosis in DCM by regulating cardiomyocyte mitochondrial function.
Collapse
MESH Headings
- Animals
- Ferroptosis/drug effects
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/genetics
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Signal Transduction
- Carnitine O-Palmitoyltransferase/metabolism
- Carnitine O-Palmitoyltransferase/genetics
- Mice, Inbred C57BL
- Diabetes Mellitus, Experimental/enzymology
- Male
- Mice, Knockout
- Mitochondria, Heart/pathology
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/drug effects
- Reactive Oxygen Species/metabolism
- Palmitic Acid/pharmacology
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Mice
- Diet, High-Fat
Collapse
Affiliation(s)
- Hong Xiang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qi Lyu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, China
| | - Jie Ouyang
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Di Xiao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Quanjun Liu
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - HaiJiao Long
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xinru Zheng
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan, China.
| | - Hongwei Lu
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.
- Department of Cardiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
48
|
Kim HK, Kim DY, Kang S, Kim H, Kim JM, Go GW. Lean metabolic dysfunction-associated steatotic disease is reversed by betulinic acid, a therapeutic triterpene from birch bark. FOOD BIOSCI 2024; 62:104376. [DOI: 10.1016/j.fbio.2024.104376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
49
|
Zhang Y, Han S, Li T, Zhu L, Wei F. Bisphenol A induces non-alcoholic fatty liver disease by promoting the O-GlcNAcylation of NLRP3. Arch Physiol Biochem 2024; 130:814-822. [PMID: 38038745 DOI: 10.1080/13813455.2023.2288533] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 12/02/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease. The mechanism by which bisphenol A (BPA) promots NAFLD remains unclear. Palmitic acid (PA) and lipopolysaccharide (LPS) were used to simulate NAFLD in HepG2 cells in vitro. Total cholesterol (TC), triglyceride (TG) content, and lipid accumulation were measured to evaluate lipid metabolism. The caspase-1-stained cells and NLRP3 inflammasome-associated proteins were evaluated for pyroptosis. Western blot analysis was used to detect protein levels and co-immunoprecipitation (Co-IP) was used to detect the association between the proteins. Cycloheximide (CHX) treatment combined with western blot was performed to access protein stability. This data have shown that BPA induces lipid metabolism dysfunction and pyroptosis by upregulating O-GlcNAc transferase (OGT) level. NLRP3 directly interacts with OGT, and elevated OGT enhanced the stability of NLRP3 protein. BPA promoted OGT-mediated O-GlcNAcylation to stabilised NLRP3, thus accelerating NAFLD progress in vitro. Our study reveals that BPA, as an environmental factor, may be involved in the promotion of NAFLD, and that targeting NLRP3 and OGT may inhibit BPA's induction of NAFLD.
Collapse
Affiliation(s)
- Yonghong Zhang
- Department of Endocrinology, First Affiliated Hospital of Baotou Medical Collage, Inner Mongolia University of Science and Technology, Baotou, PR China
| | - Shujuan Han
- Baotou Medical Collage, Inner Mongolia University of Science and Technology, Baotou, PR China
| | - Tian Li
- Baotou Medical Collage, Inner Mongolia University of Science and Technology, Baotou, PR China
| | - Li Zhu
- Department of Endocrinology, First Affiliated Hospital of Baotou Medical Collage, Inner Mongolia University of Science and Technology, Baotou, PR China
| | - Feng Wei
- Department of Endocrinology, First Affiliated Hospital of Baotou Medical Collage, Inner Mongolia University of Science and Technology, Baotou, PR China
| |
Collapse
|
50
|
Yadav P, Quadri K, Kadian R, Waziri A, Agrawal P, Alam MS. New approaches to the treatment of metabolic dysfunction-associated steatotic liver with natural products. ILIVER 2024; 3:100131. [DOI: 10.1016/j.iliver.2024.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|