51
|
Liu X, Ma S, Li J, Song M, Li Y, Qi Y, Liu F, Fang Z, Zheng R. Clinical Features and Plasma Metabolites Analysis in Obese Chinese Children With Nonalcoholic Fatty Liver Disease. J Endocr Soc 2025; 9:bvaf032. [PMID: 40104566 PMCID: PMC11914974 DOI: 10.1210/jendso/bvaf032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Indexed: 03/20/2025] Open
Abstract
Objective This study aimed to investigate the clinical characteristics and plasma metabolites of nonalcoholic fatty liver disease (NAFLD) in obese Chinese children and to develop machine learning-based NAFLD diagnostic models. Methods We recruited 222 obese children aged 4 to 17 years and divided them into an obese control group and an obese NAFLD group based on liver ultrasonography. Mass spectrometry metabolomic analysis was used to measure 106 metabolites in plasma. Binary logistic regression was used to identify NAFLD-related clinical variables. NAFLD-specific metabolites were illustrated via volcano plots, cluster heatmaps, and metabolic network diagrams. Additionally, we applied 8 machine learning methods to construct 3 diagnostic models based on clinical variables, metabolites, and clinical variables combined with metabolites. Results By evaluating clinical variables and plasma metabolites, we identified 16 clinical variables and 14 plasma metabolites closely associated with NAFLD. We discovered that the level of 18:0 to 22:6 phosphatidylethanolamines was positively correlated with the levels of total cholesterol, triglyceride-glucose index, and triglyceride to high-density lipoprotein cholesterol ratio, whereas the level of glycocholic acid was positively correlated with the levels of alanine aminotransferase, gamma-glutamyl transferase, insulin, and the homeostasis model assessment of insulin resistance. Additionally, we successfully developed 3 NAFLD diagnostic models that showed excellent diagnostic performance (areas under the receiver operating characteristic curves of 0.917, 0.954, and 0.957, respectively). Conclusions We identified 16 clinical variables and 14 plasma metabolites associated with NAFLD in obese Chinese children. Diagnostic models using these features showed excellent performance, indicating their potential for diagnosis.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300000, China
| | - Shifeng Ma
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300000, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin 300000, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300000, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300000, China
| | - Mingkun Song
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300000, China
| | - Yun Li
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300000, China
| | - Yingyi Qi
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300000, China
| | - Fei Liu
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300000, China
| | - Zhongze Fang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300000, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300000, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin 300000, China
| | - Rongxiu Zheng
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300000, China
| |
Collapse
|
52
|
Dell T, Mesropyan N, Layer Y, Tischler V, Weinhold L, Chang J, Jansen C, Schmidt B, Jürgens M, Isaak A, Kupczyk P, Pieper CC, Meyer C, Luetkens J, Kuetting D. Photon-counting CT-derived Quantification of Hepatic Fat Fraction: A Clinical Validation Study. Radiology 2025; 314:e241677. [PMID: 40100026 DOI: 10.1148/radiol.241677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Background Steatosis is a critical health problem, creating a growing need for opportunistic screening. Early detection may allow for effective treatment and prevention of further liver complications. Purpose To evaluate photon-counting CT (PCCT) fat quantification on contrast-enhanced scans and validate the results against fat quantification via histopathologic assessment, controlled attenuation parameter (CAP) from transient elastography, and MRI proton density fat fraction (PDFF). Materials and Methods In this prospective, observational clinical study, PCCT-derived fat fraction quantification was assessed in participants with known or suspected liver disease. Participants underwent PCCT between February 2022 and January 2024. Participants also underwent biopsy, US with CAP measurement, or MRI with a PDFF sequence for hepatic fat fraction quantification. Liver fat fraction was measured on virtual noncontrast PCCT images using spectral processing software with a three-material decomposition algorithm for fat, liver tissue, and iodine. Steatosis was graded for each modality. Correlation between PCCT-based steatosis grades and biopsy- and CAP-based grades was assessed with the Spearman correlation coefficient. Agreement between PCCT and MRI PDFF measurements was assessed with the intraclass correlation coefficient. Receiver operating characteristic curve analysis was conducted to determine the optimal PCCT fat fraction threshold for distinguishing between participants with and those without steatosis. Results The study included 178 participants, of whom 27 (mean age, 60.7 years ± 15.2 [SD]; 18 male participants) underwent liver biopsy, 26 (mean age, 60.0 years ± 18.3; 15 male participants) underwent CAP measurement, and 125 (mean age, 61.2 years ± 13.1; 70 male participants) underwent MRI PDFF measurement. There was excellent agreement between PCCT and MRI PDFF assessment of liver fat fraction (intraclass correlation coefficient, 0.91 [95% CI: 0.87, 0.94]). In stratified analysis, the intraclass correlation coefficient was 0.84 (95% CI: 0.63, 0.93) in participants with known fibrosis and 0.92 (95% CI: 0.88, 0.94) in participants without fibrosis. There was moderate correlation of PCCT-based steatosis grade with histologic (ρ = 0.65) and CAP-based (ρ = 0.45) steatosis grade. Based on the Youden index, the PCCT fat fraction threshold that best discriminated between participants with and those without steatosis was 4.8%, with a maximum achievable sensitivity of 81% (38 of 47) and a specificity of 71% (55 of 78). Conclusion PCCT in a standard clinical setting allowed for accurate estimation of liver fat fraction compared with MRI PDFF-based reference standard measurements. © RSNA, 2025 See also the editorial by Kartalis and Grigoriadis in this issue.
Collapse
Affiliation(s)
- Tatjana Dell
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Narine Mesropyan
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Yannik Layer
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Verena Tischler
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Leonie Weinhold
- Institute for Medical Biometry, Informatics, and Epidemiology, Rhenish Friedrich Wilhelm University of Bonn, Bonn, Germany
| | - Johannes Chang
- Department of Internal Medicine I, Center for Cirrhosis and Portal Hypertension Bonn, University Hospital Bonn, Bonn, Germany
| | - Christian Jansen
- Department of Internal Medicine I, Center for Cirrhosis and Portal Hypertension Bonn, University Hospital Bonn, Bonn, Germany
| | - Bernhard Schmidt
- Department of Computed Tomography, Siemens Healthcare, Forchheim, Germany
| | - Markus Jürgens
- Department of Computed Tomography, Siemens Healthcare, Forchheim, Germany
| | - Alexander Isaak
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Patrick Kupczyk
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Claus Christian Pieper
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Carsten Meyer
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Julian Luetkens
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Daniel Kuetting
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
53
|
Zeng R, Wang Y, Wen J, Cen Z, Wang T, Duan M, Huang X, Zhao Z, Zhang Z, Yang C, Chen S. Hypoxia-inducible factor-1α inhibitor promotes non-alcoholic steatohepatitis development and increases hepatocellular lipid accumulation via TSKU upregulation. Arch Biochem Biophys 2025; 765:110313. [PMID: 39832609 DOI: 10.1016/j.abb.2025.110313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/20/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Non-alcoholic steatohepatitis (NASH) is the progressive form of non-alcoholic fatty liver disease (NAFLD) which is the most common chronic liver disease worldwide. Hypoxia-inducible factor-1α (HIF1α) inhibitor is emerging as a promising therapeutic strategy for diseases. However, the role of HIF1α inhibitor in NASH is still unclear. A choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD) -induced NASH mouse model was established to identify the impacts of HIF1α inhibitor KC7F2 on the development of NASH. We found that KC7F2 treatment substantially aggravated lipid accumulation, inflammation, and fibrosis in the liver of NASH mice presumably via increasing Tsukushi (TSKU) expression in the liver. Mechanistically, KC7F2 up-regulated expression of TSKU in hepatocyte in vitro, which led to increased hepatocellular lipid accumulation and was reversed when TSKU was knockdown in hepatocyte. Our findings indicated that HIF1α inhibitor promotes the development of NASH presumably via increasing TSKU expression in the liver, suggesting that HIF1α attenuates NASH, and that we should assess the potential liver toxicity when use HIF1α inhibitor or medicines that can decrease the expression of HIF1α to therapy other diseases.
Collapse
Affiliation(s)
- Renli Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China; Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China.
| | - Yuxin Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| | - Jielu Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| | - Zhipeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| | - Meng Duan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510000, China.
| | - Xiuyi Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China.
| | - Zhengde Zhao
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China.
| | - Zhongyu Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| | - Chuan Yang
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China.
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
54
|
Mullin SM, Kelly AJ, Ní Chathail MB, Norris S, Shannon CE, Roche HM. Macronutrient Modulation in Metabolic Dysfunction-Associated Steatotic Liver Disease-the Molecular Role of Fatty Acids compared with Sugars in Human Metabolism and Disease Progression. Adv Nutr 2025; 16:100375. [PMID: 39842721 PMCID: PMC11849631 DOI: 10.1016/j.advnut.2025.100375] [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: 09/12/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a significant public health concern, with its progression to metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis leading to severe outcomes including cirrhosis, hepatocellular carcinoma, and liver failure. Whereas obesity and excess energy intake are well-established contributors to the development and progression of MASLD, the distinct role of specific macronutrients is less clear. This review examines the mechanistic pathways through which dietary fatty acids and sugars contribute to the development of hepatic inflammation and fibrosis, offering a nuanced understanding of their respective roles in MASLD progression. In terms of addressing potential therapeutic options, human intervention studies that investigate whether modifying the intake of dietary fats and carbohydrates affects MASLD progression are reviewed. By integrating this evidence, this review seeks to bridge the gap in the understanding between the mechanisms of macronutrient-driven MASLD progression and the effect of altering the intake of these nutrients in the clinical setting and presents a foundation for future research into targeted dietary strategies for the treatment of the disease.
Collapse
Affiliation(s)
- Sinéad M Mullin
- School of Public Health, Physiotherapy and Sport Science, and Institute of Food and Health, University College Dublin, Belfield, Dublin, Ireland; Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Aidan J Kelly
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Méabh B Ní Chathail
- School of Public Health, Physiotherapy and Sport Science, and Institute of Food and Health, University College Dublin, Belfield, Dublin, Ireland; Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Suzanne Norris
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Christopher E Shannon
- Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland; School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Helen M Roche
- School of Public Health, Physiotherapy and Sport Science, and Institute of Food and Health, University College Dublin, Belfield, Dublin, Ireland; Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland; Institute for Global Food Security, Queen's University Belfast, Northern Ireland.
| |
Collapse
|
55
|
He F, Du W, Liu Y, Ling Y, Xu M, Liu J, Song P, Fang Z, Yue Z, Duan J, Wang L. Exosome-equipped TNF antisense oligodeoxynucleotide or 2-deoxy-D-glucose ameliorated nonalcoholic steatohepatitis by modulating superoxide dismutase 1 in mice. Redox Biol 2025; 80:103488. [PMID: 39778469 PMCID: PMC11763583 DOI: 10.1016/j.redox.2025.103488] [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: 10/05/2024] [Revised: 12/09/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Inflammatory mediators tumor necrosis factor (TNF) and interleukin 1 beta (IL1β), primarily derived from hepatic macrophages in the liver, play a crucial role in the progression of nonalcoholic steatohepatitis (NASH). Meanwhile, intravenously injected exosomes are mainly distributed in the liver and predominantly taken up by hepatic macrophage. Herein, we aimed to evaluate the feasibility of targeted inhibition of TNF and IL1β expression in hepatic macrophages via exosomes as a potential therapeutic strategy for NASH. In this study, we demonstrated that antisense oligodeoxynucleotide targeting TNF (ASO-TNF) or 2-deoxy-d-glucose (2DG) effectively suppressed the expression of TNF and/or IL1β in macrophages. Exosomes loaded with ASO-TNF or 2DG were able to suppress the expression of TNF and/or IL1β in macrophages in vitro or in vivo. Furthermore, infusion of Exo/ASO-TNF or Exo/2DG significantly attenuated experimental steatohepatitis in choline deficient amino acid-defined (CDAA) or methionine and choline deficient (MCD) diet-fed mice. RNA-seq results showed that treatment with Exo/ASO-TNF or Exo/2DG significantly inhibited pro-inflammatory signaling pathways. Mechanistically, we observed that administration of Exo/ASO-TNF or Exo/2DG could attenuate NASH progression by up-regulating the expression of superoxide dismutase 1 (Sod1). Combined, our findings demonstrated that infusion of exosomes loaded with ASO-TNF or 2DG alleviated experimental steatohepatitis in murine models. Thus, infusion of exosomes loaded with anti-inflammatory agents holds promise as a potential therapeutic strategy for NASH treatment.
Collapse
Affiliation(s)
- Fei He
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Wei Du
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Yingying Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Yuwei Ling
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Ming Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Jingjing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Ping Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Zhiqiang Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Zhensheng Yue
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Juanli Duan
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| |
Collapse
|
56
|
Li L, Yang M, Tan L, Ni Y, Wu Y. Loss of DDB2 in type II diabetes mellitus induces dysregulated ubiquitination of KMT2A in lipid metabolism disorders. J Steroid Biochem Mol Biol 2025; 247:106673. [PMID: 39798620 DOI: 10.1016/j.jsbmb.2025.106673] [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: 07/18/2024] [Revised: 10/22/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
The disorders of glucose and lipid metabolism contribute to severe diseases, including cardiovascular disease, diabetes, and fatty liver. Here, we identified DNA damage-binding protein 2 (DDB2), an E3 ubiquitin ligase, as a pivotal regulator of lipid metabolism disorders in type II diabetes mellitus (T2DM). A mouse model of T2DM and primary mouse hepatocytes with steatosis were induced. DDB2 overexpression alone or in combination with lysine N-methyltransferase 2 A (KMT2A) overexpression vectors were delivered into db/db mice and in vitro hepatocytes. DDB2 was expressed poorly, while KMT2A was expressed highly in liver tissues and primary hepatocytes of db/db mice. DDB2 ameliorated glucose intolerance and insulin resistance, decreased liver/body weight ratio, downregulated expression of lipogenesis-associated proteins (SREBP1, FASN, and SCD1) and gluconeogenesis-related proteins (PEPCK and G6Pase) in liver tissues and cells, and decreased triglyceride and total cholesterol levels in steatotic hepatocytes. DDB2 reduced KMT2A expression through ubiquitination modification. Overexpression of KMT2A promoted insulin resistance, lipogenesis and lipid deposition, and glycogen accumulation in the presence of DDB2. Overall, our data demonstrate that DDB2 alleviates hepatic lipogenesis and lipid deposition via degradation of KMT2A, thereby repressing lipid metabolism disorders in T2DM.
Collapse
Affiliation(s)
- Lvqiu Li
- Department of Endocrinology, the Second People's Hospital of Kunming, Kunming, Yunnan 650203, PR China
| | - Maogeng Yang
- Department of Orthopaedic Trauma, Baoshan People's Hospital, Baoshan, Yunnan 678000, PR China
| | - Longqiao Tan
- Department of Endocrinology, the Second People's Hospital of Kunming, Kunming, Yunnan 650203, PR China
| | - Yanhong Ni
- Department of Endocrinology, the Second People's Hospital of Kunming, Kunming, Yunnan 650203, PR China
| | - Yang Wu
- Department of Endocrinology, the Second People's Hospital of Kunming, Kunming, Yunnan 650203, PR China.
| |
Collapse
|
57
|
Zhang X, Lau HCH, Yu J. Pharmacological treatment for metabolic dysfunction-associated steatotic liver disease and related disorders: Current and emerging therapeutic options. Pharmacol Rev 2025; 77:100018. [PMID: 40148030 DOI: 10.1016/j.pharmr.2024.100018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; formerly known as nonalcoholic fatty liver disease) is a chronic liver disease affecting over a billion individuals worldwide. MASLD can gradually develop into more severe liver pathologies, including metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and liver malignancy. Notably, although being a global health problem, there are very limited therapeutic options against MASLD and its related diseases. While a thyroid hormone receptor agonist (resmetirom) is recently approved for MASH treatment, other efforts to control these diseases remain unsatisfactory. Given the projected rise in MASLD and MASH incidence, it is urgent to develop novel and effective therapeutic strategies against these prevalent liver diseases. In this article, the pathogenic mechanisms of MASLD and MASH, including insulin resistance, dysregulated nuclear receptor signaling, and genetic risk factors (eg, patatin-like phospholipase domain-containing 3 and hydroxysteroid 17-β dehydrogenase-13), are introduced. Various therapeutic interventions against MASH are then explored, including approved medication (resmetirom), drugs that are currently in clinical trials (eg, glucagon-like peptide 1 receptor agonist, fibroblast growth factor 21 analog, and PPAR agonist), and those failed in previous trials (eg, obeticholic acid and stearoyl-CoA desaturase 1 antagonist). Moreover, given that the role of gut microbes in MASLD is increasingly acknowledged, alterations in the gut microbiota and microbial mechanisms in MASLD development are elucidated. Therapeutic approaches that target the gut microbiota (eg, dietary intervention and probiotics) against MASLD and related diseases are further explored. With better understanding of the multifaceted pathogenic mechanisms, the development of innovative therapeutics that target the root causes of MASLD and MASH is greatly facilitated. The possibility of alleviating MASH and achieving better patient outcomes is within reach. SIGNIFICANCE STATEMENT: Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, and it can progress to more severe pathologies, including steatohepatitis, cirrhosis, and liver cancer. Better understanding of the pathogenic mechanisms of these diseases has facilitated the development of innovative therapeutic strategies. Moreover, increasing evidence has illustrated the crucial role of gut microbiota in the pathogenesis of MASLD and related diseases. It may be clinically feasible to target gut microbes to alleviate MASLD in the future.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
58
|
Li M, Hu D, Zhang D, Xu G, Wu D, Gao C, Meng L, Feng X, Cheng W, Wang Z, Yang Y, Tang X. Encapsulation of oleanolic acid into cyclodextrin metal-organic frameworks by co-crystallization: Preparation, structure characterization and its effect on a zebrafish larva NAFLD model. Food Res Int 2025; 204:115936. [PMID: 39986782 DOI: 10.1016/j.foodres.2025.115936] [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: 10/23/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/24/2025]
Abstract
Oleanolic acid (OA) is a common pentacyclic triterpenoid and has been used to relieve metabolic-related diseases. However, its low water solubility severely limits its clinical application. In this study, OA was encapsulated by a cyclodextrin metal-organic framework material (CD-MOF) using the co-crystallization method to obtain the OA and CD-MOF inclusion complex (OA@CD-MOF). The OA@CD-MOF was characterized using scanning electron microscopy, Fourier-transform infrared spectroscopy, X-ray diffraction, and thermogravimetric analysis, which proved that OA was successfully loaded into the CD-MOF. Molecular docking data revealed that OA tends to occupy the cyclodextrin dimolecular cavity outside the CD-MOF. The formation of OA@CD-MOF significantly increased the apparent water solubility of OA to 269.27 ± 19.40 μg/mL. Further zebrafish experiments showed that the OA@CD-MOF exhibited better anti-steatosis, anti-lipid accumulation, and anti-oxidative stress activities than OA. Overall, this study presents a promising approach for delivering OA, enhancing its bioavailability in vivo and mitigating metabolic-related diseases such as non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Meng Li
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Dongxia Hu
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Dian Zhang
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Guangya Xu
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Di Wu
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Chengcheng Gao
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Linghan Meng
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Xiao Feng
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Weiwei Cheng
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Zhenjiong Wang
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China.
| | - Yuling Yang
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China
| | - Xiaozhi Tang
- College of Food Science & Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023 China.
| |
Collapse
|
59
|
Zhong Q, Zhou R, Huang YN, Huang RD, Li FR, Chen HW, Wei YF, Liu K, Cao BF, Liao KY, Xu ZY, Wang SA, Wu XB. Frailty and risk of metabolic dysfunction-associated steatotic liver disease and other chronic liver diseases. J Hepatol 2025; 82:427-437. [PMID: 39218228 DOI: 10.1016/j.jhep.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/22/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND & AIMS Frailty is associated with multiple morbidities. However, its effect on chronic liver diseases remains largely unexplored. This study evaluated the association of frailty with the risk of incident metabolic dysfunction-associated steatotic liver disease (MASLD), cirrhosis, liver cancer, and liver-related mortality. METHODS A total of 339,298 participants without prior liver diseases from the UK Biobank were included. Baseline frailty was assessed by physical frailty and the frailty index, categorizing participants as non-frail, prefrail, or frail. The primary outcome was MASLD, with secondary outcomes, including cirrhosis, liver cancer, and liver-related mortality, confirmed through hospital admission records and death registries. RESULTS During a median follow-up of 11.6 years, 4,667 MASLD, 1,636 cirrhosis, 257 liver cancer, and 646 liver-related mortality cases were identified. After multivariable adjustment, the risk of MASLD was found to be higher in participants with prefrailty (physical frailty: hazard ratio [HR] 1.66, 95% CI 1.40-1.97; frailty index: HR 2.01, 95% CI 1.67-2.42) and frailty (physical frailty: HR 3.32, 95% CI 2.54-4.34; frailty index: HR 4.54, 95% CI 3.65-5.66) than in those with non-frailty. Similar results were also observed for cirrhosis, liver cancer, and liver-related mortality. Additionally, the frail groups had a higher risk of MASLD, which was defined as MRI-derived liver proton density fat fraction >5%, than the non-frail group (physical frailty: odds ratio 1.64, 95% CI 1.32-2.04; frailty index: odds ratio 1.48, 95% CI 1.30-1.68). CONCLUSIONS Frailty was associated with an increased risk of chronic liver diseases. Public health strategies should target reducing chronic liver disease risk in frail individuals. IMPACT AND IMPLICATIONS While frailty is common and associated with a poor prognosis in people with MASLD (metabolic dysfunction-associated steatotic liver disease) and advanced chronic liver diseases, its impact on the subsequent risk of these outcomes remains largely unexplored. Our study showed that frailty was associated with increased risks of MASLD, cirrhosis, liver cancer, and liver-related mortality. This finding suggests that assessing frailty may help identify a high-risk population vulnerable to developing chronic liver diseases. Implementing strategies that target frailty could have major public health benefits for liver-related disease prevention.
Collapse
Affiliation(s)
- Qi Zhong
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Rui Zhou
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Yi-Ning Huang
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Rui-Dian Huang
- Public Health Division, Hospital of Zhongluotan Town, Baiyun District, Guangzhou, China
| | - Fu-Rong Li
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China; School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Hao-Wen Chen
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Yan-Fei Wei
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Kuan Liu
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Bi-Fei Cao
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Kai-Yue Liao
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Zheng-Yun Xu
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Shi-Ao Wang
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China
| | - Xian-Bo Wu
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, China.
| |
Collapse
|
60
|
Cui J, Zhao G, Xie W, Yang Y, Fu X, Meng H, Liu H, Tan M, Chen D, Rong C, Wang Y, Wang Y, Zhang LW. Exacerbated hepatotoxicity in in vivo and in vitro non-alcoholic fatty liver models by biomineralized copper sulfide nanoparticles. BIOMATERIALS ADVANCES 2025; 168:214117. [PMID: 39580989 DOI: 10.1016/j.bioadv.2024.214117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/19/2024] [Accepted: 11/10/2024] [Indexed: 11/26/2024]
Abstract
Copper sulfide nanoparticles (NPs) synthesized through biomineralization have significant commercial potential as photothermal agents, while the safety evaluation of these NPs, especially for patients with non-alcoholic fatty liver (NAFL), remains insufficient. To explore the differential hepatotoxicity of copper sulfide NPs in NAFL conditions, we synthesized large-sized (LNPs, 15.1 nm) and small-sized (SNPs, 3.5 nm) BSA@Cu2-xS NPs. A NAFL rat model fed with high fat diet (HFD) was successfully established for a 14-day subacute toxicity study by daily repeated administration of BSA@Cu2-xS NPs. Our findings from serum biochemistry and histopathological examinations revealed that copper sulfide at both sizes NPs induced more pronounced liver damage in NAFL rats than rats fed with normal diet. Transcriptome sequencing analysis showed that LNPs activated inflammation and DNA damage repair pathways in the livers of NAFL rats, while SNPs displayed minimal inflammation. A three-dimensional spheroid model of NAFL developed in our in-house cell spheroid culture honeycomb chips demonstrated that LNPs, but not SNPs, triggered a distinct release of inflammatory factors and increased reactive oxygen species through Kupffer cells. These results highlight that NAFL condition exacerbated the hepatotoxicity of BSA@Cu2-xS NPs, with SNPs emerging as safer photothermal agents compared to LNPs, suggesting superior potential for clinical applications.
Collapse
Affiliation(s)
- Jinbin Cui
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Gang Zhao
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wei Xie
- The College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, Zhejiang, China
| | - Yang Yang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xing Fu
- Suzhou Vivoid Biotechnology Co., Ltd, Suzhou 215124, China
| | - Hezhang Meng
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - He Liu
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Mengfei Tan
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Dandan Chen
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Chao Rong
- Department of Pathology, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Yangyun Wang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yong Wang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Leshuai W Zhang
- School of Radiation Medicine and Protection, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
61
|
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
|
62
|
Meadows V, Antonio JM, Ferraris RP, Gao N. Ruminococcus gnavus in the gut: driver, contributor, or innocent bystander in steatotic liver disease? FEBS J 2025; 292:1252-1264. [PMID: 39589934 PMCID: PMC11927045 DOI: 10.1111/febs.17327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/29/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
The human gut microbiome plays a crucial role in regulating intestinal and systemic health, impacting host immune response and metabolic function. Dysbiosis of the gut microbiome is linked to various diseases, including steatotic liver diseases. Metabolic dysfunction-associated steatotic liver disease (MASLD), a chronic liver disease characterized by excess hepatic lipid content and impaired metabolism, is the leading cause of liver disease worldwide. Among the gut microbes, Ruminococcus gnavus (R. gnavus) has garnered attention for its association with inflammatory and metabolic diseases. While R. gnavus abundance correlates to liver fat accumulation, further research is needed to identify a causal role or therapeutic intervention in steatotic liver disease. This review surveys our current understanding of R. gnavus in the development and progression of steatotic liver diseases, highlighting its potential mechanisms through metabolite secretion, and emphasizes the need for comprehensive microbiome analyses and longitudinal studies to better understand R. gnavus' impact on liver health. This knowledge could pave the way for targeted interventions aimed at modulating gut microbiota to treat and prevent MASLD and its comorbidities.
Collapse
Affiliation(s)
- Vik Meadows
- Department of Biological Sciences, School of Arts & Sciences, Rutgers University, Newark, NJ, USA
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Jayson M Antonio
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Ronaldo P Ferraris
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Nan Gao
- Department of Biological Sciences, School of Arts & Sciences, Rutgers University, Newark, NJ, USA
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
63
|
Lan Y, Song R, Feng D, He J. Bioinformatic analysis of molecular expression patterns during the development and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Sci Rep 2025; 15:7294. [PMID: 40025132 PMCID: PMC11873118 DOI: 10.1038/s41598-025-90744-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025] Open
Abstract
The global incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to rise, primarily driven by the escalating obesity epidemic worldwide. MASLD, a spectrum of liver disorders, can progress to more severe conditions, metabolic dysfunction-associated steatohepatitis (MASH), ultimately culminating in hepatocellular carcinoma (HCC). Given the complex nature of MASLD, there is an urgent need to develop robust risk prediction models and design specialized cancer screening initiatives tailored specifically for individuals with MASLD. This study aimed to identify genes exhibiting trending expression patterns that could serve as potential biomarkers or therapeutic targets. Our approach involved analyzing expression patterns across the five stages of MASLD development and progression. Notably, we introduced an innovative two-phase classification-MASLD occurrence and MASLD progression-instead of categorizing differentially expressed genes (DEGs) into multiple types. Leveraging LASSO regression models, we demonstrated their relatively strong capability to predict and distinguish both MASLD occurrence and progression. Furthermore, our analysis identified CYP7A1 and TNFRSF12A as significantly associated with the prognosis of MASLD progressing to HCC. These findings contribute to the understanding of gene expression dynamics in MASLD and may pave the way for the development of effective prognostic tools and targeted therapies in the realm of liver disease.
Collapse
Affiliation(s)
- Yuanfeng Lan
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Ran Song
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Duiping Feng
- Department of Interventional Radiology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Junqi He
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
64
|
Zuo J, Ren J, Yin B, Wang Z, Cui Q, Liu J, Huang D, Pei H, Wen R, Zhang Y, Ma Y. Effects of Sesamin in Animal Models of Obesity-Associated Diseases: A Systematic Review and Meta-Analysis. Nutr Rev 2025; 83:e838-e851. [PMID: 39074164 DOI: 10.1093/nutrit/nuae089] [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] [Indexed: 07/31/2024] Open
Abstract
CONTEXT As living standards have improved and lifestyles have undergone changes, metabolic diseases associated with obesity have become increasingly prevalent. It is well established that sesamin (Ses) (PubChem CID: 72307), the primary lignans in sesame seeds and sesame oil, possess antioxidant and anti-inflammatory effects. OBJECTIVE In this study, a systematic review and meta-analysis of the effects of Ses on animal models of obesity-related diseases was performed to assess their impact on relevant disease parameters. Importantly, this study sought to provide insights for the design of future human clinical studies utilizing Ses as a nutritional supplement or drug. DATA SOURCES This study conducted a comprehensive search in PubMed, Web of Science, Embase, Scopus, and the Cochrane Library, identifying English language articles published from inception to April 2023. DATA EXTRACTION The search incorporated keywords such as "sesamin," "obesity," "non-alcoholic fatty liver disease," "type 2 diabetes mellitus," and "metabolic syndrome." The meta-analysis included 17 articles on non-alcoholic fatty liver disease, type 2 diabetes, and metabolic syndrome. DATA ANALYSIS Overall, the pooled results demonstrated that Ses significantly reduced levels of total serum cholesterol (P = .010), total serum triglycerides (P = .003), alanine transaminase (P = .003), and blood glucose (P < .001), and increased high-density lipoprotein cholesterol levels (P = .012) in animal models of nonalcoholic fatty liver disease. In the type 2 diabetes model, Ses mitigated drug-induced weight loss (P < .001), high-fat-diet-induced weight gain (P < .001), and blood glucose levels (P = .001). In the metabolic syndrome model, Ses was associated with a significant reduction in body weight (P < .001), total serum cholesterol (P < .001), total serum triglycerides (P < .001), blood glucose (P < .001), and alanine transaminase levels (P = .039). CONCLUSION The meta-analysis results of this study suggest that Ses supplementation yields favorable effects in animal models of obesity-related diseases, including hypolipidemic, insulin-lowering, and hypoglycemic abilities, as well as organ protection from oxidative stress and reduced inflammation. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration No. CRD42023438502.
Collapse
Affiliation(s)
- Jinshi Zuo
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Bowen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Ziyi Wang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qiqi Cui
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiarui Liu
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Dan Huang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Huanting Pei
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Rui Wen
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Yadong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| |
Collapse
|
65
|
Shi R, Chai K, Wang H, Zhou J, Yang S, Li J, Qiao C, Sheng X, Zhang X, Wu J. Clinical Assessment of Common Medications for Nonalcoholic Fatty Liver Disease: A Systematic Review and Bayesian Network Meta-Analysis. J Evid Based Med 2025; 18:e70002. [PMID: 39963857 PMCID: PMC11833758 DOI: 10.1111/jebm.70002] [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: 06/19/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
OBJECTIVE With a steadily rising prevalence, nonalcoholic fatty liver disease (NAFLD) was a leading global cause of liver-related health problems. In the clinical management of NAFLD, various western pharmaceuticals were widely utilized. This network meta-analysis aimed to evaluate the effectiveness of common western medications for NAFLD patients. METHODS We systematically reviewed and screened articles based on predesigned criterion about western medications for NAFLD, which were from Embase, Cochrane Library, PubMed, CNKI, WanFang, and China Science and Technology Journal Database until August 1, 2024. Eligible studies included randomized controlled trials of patients aged 18 or older with NAFLD, comparing Western medicines to placebos or other Western medicine treatments. The risk of bias assessment tool 2.0 from the Cochrane system was used to assess the quality of the included articles. A Bayesian network meta-analysis was conducted using WinBUGS 1.4.3 with a random-effects model and Markov Chain Monte Carlo methods. Treatment rankings were based on Surface Under the Cumulative Ranking Curve (SUCRA) values, and heterogeneity was assessed with I2 and Q statistics. The outcomes were analyzed in WinBUGS and visualized using Stata 14.0, generating network plots and cumulative probability rankings to compare treatment effects. The systematic review was registered in PROSPERO (CRD42024509176). RESULTS Based on 37 included articles involving 7673 patients, pioglitazone demonstrated the most significant effects in resolving nonalcoholic steatohepatitis without worsening fibrosis, increasing high-density lipoprotein cholesterol levels, and achieving a ≥ 2-point reduction in NAFLD activity scores (odds ratio [OR] = 0.09, 95% confidence interval [CI]: 0.01 to 0.81), with a SUCRA probability of 91.4%. Aldafermin showed remarkable effects in improving liver function markers, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), and γ-glutamyl transpeptidase, with cumulative probabilities of 90% for ALT and 69.8% for AST. Cluster analysis revealed that Resmetirom and Aldafermin were superior options for enhancing liver function, while pioglitazone emerged as the best treatment for the comprehensive improvement of NAFLD. CONCLUSIONS Pioglitazone outperformed other western medicines in terms of overall efficacy when treating NAFLD, but Aldafermin and Resmetirom showed superior improvement in liver function. This study provided a certain level of support for the use of specific clinical medications.
Collapse
Affiliation(s)
- Rui Shi
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Keyan Chai
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Haojia Wang
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Jiying Zhou
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Siyun Yang
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Jiaqi Li
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Chuanqi Qiao
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoguang Sheng
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Xiaomeng Zhang
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| | - Jiarui Wu
- Department of Clinical Chinese PharmacySchool of Chinese MateriaBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
66
|
Jiang Z, Yang L, Liu Q, Qiu M, Chen Y, Teng M, Zhang Y, Liu X, Zhao Z, Zheng Y, Andersen M, Qu W. Haloacetamides exacerbate non-alcoholic fatty liver disease induced by a high-fat diet in C57BL/6J mice. Toxicol Sci 2025; 204:57-69. [PMID: 39689017 DOI: 10.1093/toxsci/kfae160] [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] [Indexed: 12/19/2024] Open
Abstract
Obesity, a significant global health issue, heightens the risk of non-alcoholic fatty liver disease (NAFLD). Its interaction with environmental pollutants might exacerbate NAFLD's severity. Haloacetamides (HAcAms), a group of emerging nitrogenous disinfection byproducts (DBPs) and potent oxidative stressors, are found in chlorinated drinking water. Since oxidative stress is associated with HAcAms-DBP cytotoxicity and a key factor in NAFLD pathogenesis, we hypothesize that HAcAms-DBPs could exacerbate liver injury and NAFLD, particularly with high-fat diets. This study examined HAcAms-DBPs' impact on liver lipid metabolism in mice treated with 1 to 100 times the background drinking water level (13.05 µg/L) for up to 16 weeks of oral administration. Compared to a high-fat-only group, mice co-exposed to a high-fat diet and HAcAms-DBPs for 16 weeks had elevated serum alanine transaminase, aspartate transaminase, triglyceride, hepatic lipid aggregation, and inflammation response. Under high-fat conditions, background drinking water levels of HAcAms significantly upregulated liver Acetyl-CoA carboxylase 1, fatty acid synthase, peroxisome proliferator-activated receptor gamma (PPARγ), PPARγ coactivator-1α, glucose transporter 1 and 4 protein expression in C57BL/6J mice; 10 times background significantly increased expression of inflammatory marker tumor necrosis factor and liver fibrosis marker protein alpha-smooth muscle actin; 100 times further increased both liver damage and markers of early non-alcoholic steatohepatitis phenotypes like steatosis and lobular inflammation. HAcAms-DBPs plus high-fat conditions worsened liver damage. The possible health risks of NAFLD induced by HAcAms in obese individuals deserve further study.
Collapse
Affiliation(s)
- Zhiqiang Jiang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Lili Yang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Qinxin Liu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Meiyue Qiu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yu Chen
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Mengying Teng
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- Key Laboratory of the Public Health Safety, Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Fudan University, Shanghai 200032, China
| | - Xing Liu
- Key Laboratory of the Public Health Safety, Ministry of Education, Department of Epidemiology, School of Public Health, Fudan University, Shanghai 200032, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Melvin Andersen
- ScitoVation LLC, Research Triangle Park, NC 27713, United States
| | - Weidong Qu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| |
Collapse
|
67
|
Zhang X, Chen T, Li Z, Wan L, Zhou Z, Xu Y, Yan D, Zhao W, Chen H. NORAD exacerbates metabolic dysfunction-associated steatotic liver disease development via the miR-511-3p/Rock2 axis and inhibits ubiquitin-mediated degradation of ROCK2. Metabolism 2025; 164:156111. [PMID: 39710000 DOI: 10.1016/j.metabol.2024.156111] [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: 08/16/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND & AIMS Abnormal regulation of lncRNA is strongly linked to metabolic dysfunction-associated steatotic liver disease (MASLD). However, the precise molecular mechanisms remain unclear. This study explores the roles of noncoding RNA activated by DNA damage (NORAD)/miR-511-3p/Rho-associated protein kinase 2 (Rock2) axis and the NORAD/ROCK2 interaction in the development of MASLD. METHODS In vitro and in vivo models of MASLD were created using high-fat diet-fed mice and free fatty acid (FFA)-treated hepatocytes. To examine the relationships between NORAD, miR-511-3p, and ROCK2, we employed bioinformatics, luciferase assays, RNA immunoprecipitation, and biotinylated NORAD pull-down assays. MASLD progression was assessed based on food intake, energy expenditure, insulin resistance, hepatic steatosis, inflammation, white fat growth, and liver fibrosis. RESULTS NORAD and ROCK2 were upregulated, while miR-511-3p was downregulated in MASLD liver tissues and FFA-treated hepatocytes. Mechanistically, NORAD competitively interacted with miR-511-3p to modulate Rock2 mRNA expression, and directly stabilized ROCK2 protein by abrogating its ubiquitination degradation. Functionally, liver-specific knockdown of NORAD or overexpression of miR-511-3p significantly slowed MASLD progression. Overexpression of NORAD or ROCK2 partially reversed miR-511-3p-induced inhibition of MASLD. Additionally, ROCK2 knockdown attenuated NORAD-induced worsening of MASLD. Moreover, overexpressing NORAD or ROCK2 or interfering miR-511-3p influenced resmetirom treatment to suppress MASLD development. Finally, metabolic changes in liver driven by the NORAD/miR-511-3p/Rock2 axis and NORAD/ROCK2 interaction also influenced white adipose growth, pancreatic β-cell dedifferentiation, and liver fibrosis. CONCLUSIONS The NORAD/miR-511-3p/Rock2 axis and the NORAD/ROCK2 interaction play critical roles in MASLD progression, identifying potential therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Xu Zhang
- The Affiliated LiHuiLi Hospital of Ningbo University, Ningbo, China
| | | | - Zhenhan Li
- Department of Pathology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, China; School of Clinical Medicine, Wannan Medical College, Wuhu, China; The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lingfeng Wan
- Fatty liver disease center of integrated Chinese and Western medicine, Jiangsu Province Hospital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing, China
| | - Zhihang Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Xu
- School of Clinical Medicine, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China; School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Dong Yan
- Nanjing University of TCM, Nanjing, China; Jiangsu Province Hospital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing, China
| | - Wei Zhao
- School of Clinical Medicine, The First Affiliated Hospital, Chengdu Medical College, Chengdu, China; School of Laboratory Medicine, Chengdu Medical College, Chengdu, China.
| | - Hao Chen
- Department of Pathology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, China; Postdoctoral Research Station of Clinical Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
68
|
Hong J, Xu Z, Xu F, Wu H, Liu J, Qu L. Immune-related diagnostic indicators and targeted therapies for COPD combined with NASH were identified and verified via WGCNA and LASSO. Front Immunol 2025; 16:1514422. [PMID: 40093012 PMCID: PMC11906333 DOI: 10.3389/fimmu.2025.1514422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction The incidence of chronic obstructive pulmonary disease (COPD) and non-alcoholic fatty liver disease (NAFLD) has increased significantly in past decades, posing a significant public health burden. An increasing amount of research points to a connection between COPD and NAFLD. This study aimed to identify the key genes of these two diseases, construct a diagnostic model, and predict potential therapeutic agents based on critical genes. Methods NAFLD and COPD datasets were obtained from the GEO database, differential genes were identified by differential analysis and WGCNA, PPI networks were constructed and enriched for differential genes and COPD-associated secreted proteins, small molecule compounds were screened, and immune cell infiltration was assessed. Meanwhile, LASSO and RF further screened the essential genes, and finally, two key genes were obtained. Subsequently, the nomogram diagnostic model and lncRNA-miRNA-mRNA network were constructed based on these two core genes, subjected to drug prediction and GSEA enrichment analysis, and validated in an external cohort using qRT-PCR. Results KEGG enrichment analysis indicated that the NF-kappa B and TNF signaling pathways may be associated with COPD and NASH co-morbidities. Ten small-molecule drugs associated with COPD and NASH were identified through cMAP analysis, including ansoprazole and atovaquone. In addition, we further identified the hub genes S100A9 and MYH2 for NAFLD and COPD by machine learning methods. The immune infiltration indicated that these two core genes might be involved in the immunomodulatory process of NASH by regulating the function or recruitment of specific immune cell types. A nomogram diagnostic model was constructed based on these two core genes. The AUC value for S100A9 was 0.887, for MYH2 was 0.877, and for the nomogram was 0.889, demonstrating excellent diagnostic efficacy. Two hundred fifty-four potential drugs targeting S100A9 and 67 MYH2 were searched in the DGIdb database. Meanwhile, the lncRNA-miRNA-mRNA network was constructed by predicting target miRNAs of biomarkers and further predicting lncRNAs targeting miRNAs. qRT-PCR analysis revealed that S100A9 was upregulated in both COPD and NAFLD, consistent with bioinformatic predictions, while MYH2 showed increased expression in COPD but decreased expression in NAFLD, diverging from the predicted downregulation in both diseases. These findings suggest that S100A9 serves as a common inflammatory marker for both diseases, whereas MYH2 may be regulated by disease-specific mechanisms, highlighting its potential for distinguishing COPD from NAFLD. Conclusion The hub genes S100A9 and MYH2 in COPD and NASH were identified by various bioinformatics methods and a diagnostic model was constructed to improve the diagnostic efficiency. We also revealed some potential biological mechanisms of COPD and NASH and potential drugs for COPD-related NASH. Our findings provide potential new diagnostic and therapeutic options for COPD-associated NASH and may help reduce its prevalence.
Collapse
Affiliation(s)
- Jianwei Hong
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zikai Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Fangrui Xu
- Department of Medical Imaging, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Haifeng Wu
- Department of Emergency Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, China
| | - Jinxia Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Lishuai Qu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
69
|
Hu M, Huang H, Jia M, Xu M, Chen M, Wu J, Gu S, Liang H, Zhou H, Gong Y. A panel of miRNAs in the serum extracellular vesicles serve as novel diagnostic biomarkers for MASLD. Biomed J 2025:100838. [PMID: 40024368 DOI: 10.1016/j.bj.2025.100838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/01/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025] Open
Abstract
The increased prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) and its profound implications for global health have sparked extensive research endeavors aimed at developing potential diagnostic methods for this condition. Despite the achievements in defining various environmental factors and genetic predispositions linked to MASLD, diagnosis and clinical staging of the disease remain challenging. Recently, extracellular vesicles (EVs) have garnered considerable attention owing to their roles in metabolic dysfunctions and their potential as biomarkers for various conditions. This study aimed to investigate whether microRNAs (miRNAs) in serum EVs could be utilized for diagnosing and staging MASLD. We applied an innovative and efficient approach that involves capturing and analyzing extracellular vesicles using wheat germ agglutinin (WGA)-coupled magnetic beads, subsequently employing reverse transcription quantitative polymerase chain reaction (RT-qPCR) for analysis. MiR-574-3p, miR-542-3p, and miR-200a-3p in serum extracellular vesicles were significantly elevated in patients with MASLD, indicating their potential as diagnostic markers. This study has established a straightforward assay platform for isolating extracellular vesicles without the need for purification and for quantitatively detecting miR-574-3p, miR-542-3p, and miR-200a-3p in serum extracellular vesicles.
Collapse
Affiliation(s)
- Moran Hu
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hai Huang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China; Department of Emergency, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Meng Jia
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Min Xu
- Institute of Geriatric Medicine, Jiangsu Province Geriatric Hospital, Nanjing, Jiangsu 210009, China
| | - Malin Chen
- The First Clinical College of Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Junxiang Wu
- Department of Emergency, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Shouyong Gu
- Institute of Geriatric Medicine, Jiangsu Province Geriatric Hospital, Nanjing, Jiangsu 210009, China
| | - Hongwei Liang
- Department of Emergency, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| | - Hongwen Zhou
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Yingyun Gong
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
70
|
Yang D, Wuyunsiqin, YanNiu, Hashentuya, Tana, Anna, Ma M, Zhao W, Menggenduxi, Wang M. Traditional Mongolian Medicine Qiqirigan-8 alleviates non-alcoholic fatty liver disease via restoring gut microbiota and metabolism. Front Microbiol 2025; 16:1517082. [PMID: 40083784 PMCID: PMC11905161 DOI: 10.3389/fmicb.2025.1517082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/31/2025] [Indexed: 03/16/2025] Open
Abstract
Background Mongolian Medicine Qiqirigan-8 (MMQ-8) is a traditional Mongolian medicine formula used to treat fatty liver disease. However, the material basis and in vivo metabolic process of the therapeutic effect of MMQ-8 on non-alcoholic fatty liver disease (NAFLD) remain unclear. Methods The chemical composition of MMQ-8 was determined using Ultra-high-performance liquid chromatography-quadrupole Exactive Mass spectrometry analysis (UHPLC-QE-MS). C57BL/6J mice were fed a choline-deficient diet for 12 weeks to induce a NAFLD model. Hematoxylin and Eosin (H&E)-staining, combined with serum biochemical indexes, was used to observe liver appearance and characterize the pathological changes and functions of the liver. HE staining and Alcian Blue-Phosphoric Acid Schiff (AB-PAS) staining of the colon, along with ZO-1 immunofluorescence expression in the colon were used to reveal the effect of MMQ-8 on the disruption of the intestinal epithelial mucosal barrier in the NAFLD. The expression of intestinal tight junction genes was analyzed by qRT-PCR to observe the protective effect of MMQ-8 against intestinal epithelial mucosal barrier disruption. Fecal metagenomics and serum non-targeted metabolomics were used to reveal the effects of MMQ-8 on the gut microbiota and metabolism in mice with NAFLD. Finally, we emphasize the interaction between gut microbiota and metabolites through Spearman correlation coefficient analysis. Results Mongolian Medicine Qiqirigan-8 contains 17 active ingredients, which can reduce hepatic steatosis and lobular inflammation in mice with NAFLD, and have protective effects against liver injury. MMQ-8 reduced the infiltration of inflammatory cells in the colon epithelium of model mice while restoring the number of goblet cells. MMQ-8 significantly enhanced ZO-1 protein expression in the colon, as well as the mRNA expression of both ZO-1 and Occludin. Fecal metagenomics results showed that MMQ-8 reduced the Bacillota/Bacteroidota ratio in NAFLD mice. Increased the abundance of beneficial bacteria such as Porphyromonadaceae, Prevotella, and Bacteroidota. and suppressed the abundance of dysfunctional bacteria, such as Bacillota, Acetatifactor, and Erysipelotrichaceae. Furthermore, metabolomics studies revealed that MMQ-8 intervention significantly regulated the expression of metabolites related to glutathione metabolism, butyric acid metabolism, sphingolipid metabolism, and glycerophospholipid metabolism in NAFLD mice compared to the model group. These metabolic pathways play key roles in NAFLD. According to Spearman's correlation coefficient analysis, up-regulation of Porphyromonadaceae, Prevotella, and Bacteroidota after MMQ-8 intervention was negatively correlated with LPC levels in glycerophospholipid metabolic pathways, while positively correlated with PC levels. In contrast, the relationship between Bacillota and Acetatifactor, which were down-regulated after MMQ-8 intervention, was the opposite. In addition, the up-regulation of Porphyromonadaceae, Prevotella, and Bacteroidota after MMQ-8 intervention was positively correlated with fumaric acid, 2-oxoglutaric acid, adenosine, and L-glutathione levels, while those down-regulated after MMQ-8 intervention were positively correlated with the levels of Bacillota, Acetatifactor were negatively correlated with all the above metabolites. Thus, glutathione metabolism, butyric acid metabolism, sphingolipid metabolism, glycerophospholipid metabolism and gut microbial ecosystem are tightly intertwined in this process. Conclusion In summary, these findings indicate that MMQ-8 has a synergistic anti-NAFLD effect through its multi-component, multi-target, gut microbiota-modulating and multi metabolic pathway characteristics. The host's regulation of specific gut microbiota and involvement in multiple metabolic pathways may be one of the important mechanisms by which MMQ-8 exerts its therapeutic effects on NAFLD. It is worth noting that metabolic pathways such as glutathione metabolism, butyric acid metabolism, sphingolipid metabolism, glycerophospholipid metabolism, and the gut microbiota ecosystem are closely intertwined in this process.
Collapse
Affiliation(s)
- Dandan Yang
- School of Traditional Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- Key Laboratory of Quality Research and Pharmacodynamic Evaluation of Traditional Chinese Medicine and Mongolia Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Wuyunsiqin
- School of Traditional Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- Key Laboratory of Quality Research and Pharmacodynamic Evaluation of Traditional Chinese Medicine and Mongolia Medicine, Inner Mongolia Medical University, Hohhot, China
| | - YanNiu
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Hashentuya
- School of Traditional Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Tana
- School of Traditional Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Anna
- School of Traditional Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Mingxing Ma
- School of Traditional Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Wenhui Zhao
- School of Traditional Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Menggenduxi
- School of Traditional Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Minjie Wang
- Key Laboratory of Quality Research and Pharmacodynamic Evaluation of Traditional Chinese Medicine and Mongolia Medicine, Inner Mongolia Medical University, Hohhot, China
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
71
|
Zhang LH, Liu ST, Zhao Q, Liu XY, Liu T, Zhang Q, Liu MH, Zhao WX. Role of triggering receptor expressed on myeloid cells 2 in the pathogenesis of non-alcoholic fatty liver disease. World J Hepatol 2025; 17:102328. [PMID: 40027566 PMCID: PMC11866134 DOI: 10.4254/wjh.v17.i2.102328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/04/2025] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive disease. Without effective interventions, NAFLD can gradually develop to non-alcoholic steatohepatitis, fatty liver fibrosis, liver cirrhosis and even hepatocellular carcinoma. It is still to investigate the precise molecular mechanism behind the pathophysiology of NAFLD. Triggering receptor expressed on myeloid cells 2 (TREM2) can sense tissue injury and mediate immune remodeling, thereby inducing phagocytosis, lipid metabolism, and metabolic transfer, promoting cell survival and combating inflammatory activation. NAFLD might develop as a result of TREM2's regulatory role. We here briefly summarize the biological characteristics of TREM2 and its functions in the disease progression of NAFLD. Moreover, we propose to broaden the therapeutic strategy for NAFLD by targeting TREM2.
Collapse
Affiliation(s)
- Li-Hui Zhang
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Su-Tong Liu
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Qing Zhao
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Xiao-Yan Liu
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Tong Liu
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Qiang Zhang
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Ming-Hao Liu
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Wen-Xia Zhao
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Department of Spleen, Stomach, Liver and Gallbladder Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China.
| |
Collapse
|
72
|
Pu S, Zhao B, Jiang Y, Cui X. Hypothyroidism/subclinical hypothyroidism and metabolic dysfunction-associated steatotic liver disease: advances in mechanism and treatment. Lipids Health Dis 2025; 24:75. [PMID: 40016726 PMCID: PMC11866868 DOI: 10.1186/s12944-025-02474-0] [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: 10/13/2024] [Accepted: 02/09/2025] [Indexed: 03/01/2025] Open
Abstract
Hypothyroidism is a risk factor for metabolic dysfunction-associated steatotic liver disease (MASLD) but it is not clear whether subclinical hypothyroidism (SCH) increases the risk of MASLD and whether SCH patients with MASLD require treatment. In this study, we reviewed articles published in PubMed from 2013 to 2024 with SCH/hypothyroidism and MASLD as keywords. According to the studies retrieved, SCH increases the likelihood of developing MASLD. Thyroid hormones influence energy metabolism and storage in adipose tissues, as well as fatty acid and cholesterol metabolism and transport in the liver. L-T4 replacement therapy reduces the prevalence of MASLD, especially in patients with severe SCH or mild SCH with dyslipidemia. Recent studies showed that thyroid hormone analogues and thyroid hormone β receptor agonists obtained positive results in the treatment of MASLD in animal models and clinical trials, and Resmetirom has been approved by the US. Food and Drug Administration (FDA) under the name Rezdiffra for use in conjunction with dietary and exercise regimens for managing non-cirrhotic NASH in adults with moderate to advanced fibrosis.
Collapse
Affiliation(s)
- Sicheng Pu
- China Medical University, Shenyang, China
| | | | | | - Xuejiao Cui
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
73
|
Xu K, Corona-Avila I, Frutos MD, Nunez-Sanchez MA, Makhanasa D, Shah PV, Guzman G, Ramos-Molina B, Priyadarshini M, Khan MW. Hepatic HKDC1 Deletion Alleviates Western Diet-Induced MASH in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.26.625530. [PMID: 39651120 PMCID: PMC11623584 DOI: 10.1101/2024.11.26.625530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The global prevalence of Metabolic dysfunction-associated steatohepatitis (MASH) has been rising sharply, closely mirroring the increasing rates of obesity and metabolic syndrome. MASH exhibits a strong sexual dimorphism where females are affected with more severe forms after menopause. Hexokinase domain-containing protein 1 (HKDC1) has recently been recognized for its role in liver diseases, where its expression is minimal under normal conditions but significantly increases in response to metabolic stressors like obesity and liver injury. This selective upregulation suggests HKDC1s potential specialization in hepatic glucose and lipid dysregulation, linking it closely to the progression of MASLD and MASH. This study aims to clarify the role of HKDC1 in Western diet-induced MASH in female mice by examining its impact on hepatic glucose and lipid metabolism, offering insights into its potential as a therapeutic target and addressing the need for sex-specific research in liver disease. This study reveals that HKDC1 expression is elevated in obese women with MASH and correlates with liver pathology. In a mouse model, liver-specific HKDC1 knockout (HKDC1LKO) protected against Western diet-induced obesity, glucose intolerance, and MASH features, including steatosis, inflammation, and fibrosis. Transcriptomic analysis showed that HKDC1 deletion reduced pro-inflammatory and pro-fibrotic gene expression, while gut microbiome analysis indicated a shift toward MASH-protective bacteria. These findings suggest that HKDC1 may exacerbate MASH progression through its role in metabolic and inflammatory pathways, making it a potential therapeutic target.
Collapse
|
74
|
Zhang X, Ge Y, Ye M, Wang X, Tong Y, Liu C, Xu S, Zhao Z, You Q, Guo X, Jiang Z. A Keap1-recruiting BRD4 degrader offers a single-molecular polypharmacology approach for the treatment of metabolic dysfunction-associated steatohepatitis. Free Radic Biol Med 2025; 232:15-27. [PMID: 40023298 DOI: 10.1016/j.freeradbiomed.2025.02.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
The pathogenesis of metabolic dysfunction-associated steatohepatitis (MASH) involves multiple pathophysiological processes, including abnormal lipid metabolism, insulin resistance, oxidative stress, endoplasmic reticulum stress, inflammatory response, and fibrosis. These factors interact to form a complex network and the development of synergistic and pleiotropic drug modalities targeting multiple pathogenesis of MASH may have a better therapeutic effect. Herein, the bifunctional proteolytic targeting chimeras (PROTAC) technology was utilized for developing pleiotropic drugs for MASH treatment. We constructed a Keap1-recruiting degrader KB-3 which stabilizes the natural Keap1 target Nrf2 and degrades BRD4 synergistically, exhibiting combined therapeutic advantages against MASH-related pathologies. Experimental results confirmed that KB-3 could effectively alleviate MASH in mice by improving lipid metabolic disorder, enhancing the defense against oxidative stress, reducing inflammation, and delaying the progression of liver fibrosis. Such Keap1-recruiting degrader offering a single-molecular approach with polypharmacology effects may be an attractive strategy for the treatment of multifactorial disease.
Collapse
Affiliation(s)
- Xian Zhang
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuxin Ge
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Mengjie Ye
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiaolu Wang
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuanyuan Tong
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Chihong Liu
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Shicheng Xu
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ziquan Zhao
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaoke Guo
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zhengyu Jiang
- State Key Laboratory of Natural Medicines, and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
75
|
Zhu R, Xu C, Jiang S, Xia J, Wu B, Zhang S, Zhou J, Liu H, Li H, Lou J. Risk factor analysis and predictive model construction of lean MAFLD: a cross-sectional study of a health check-up population in China. Eur J Med Res 2025; 30:137. [PMID: 40001266 PMCID: PMC11863909 DOI: 10.1186/s40001-025-02373-1] [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: 09/05/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
AIM Cardiovascular disease morbidity and mortality rates are high in patients with metabolic dysfunction-associated fatty liver disease (MAFLD). The objective of this study was to analyze the risk factors and differences between lean MAFLD and overweight MAFLD, and establish and validate a nomogram model for predicting lean MAFLD. METHODS This retrospective cross-sectional study included 4363 participants who underwent annual health checkup at Yuyao from 2019 to 2022. The study population was stratified into three groups: non-MAFLD, lean MAFLD (defined as the presence of fatty liver changes as determined by ultrasound in individuals with a BMI < 25 kg/m2), and overweight MAFLD (BMI ≥ 25.0 kg/m2). Subsequent modeling analysis was conducted in a population that included healthy subjects with < 25 kg/m2 (n = 2104) and subjects with lean MAFLD (n = 849). The study population was randomly split (7:3 ratio) to a training vs. a validation cohort. Risk factors for lean MAFLD was identify by multivariate regression of the training cohort, and used to construct a nomogram to estimate the probability of lean MAFLD. Model performance was examined using the receiver operating characteristic (ROC) curve analysis and k-fold cross-validation (k = 5). Decision curve analysis (DCA) was applied to evaluate the clinical usefulness of the prediction model. RESULTS The multivariate regression analysis indicated that the triglycerides and glucose index (TyG) was the most significant risk factor for lean MAFLD (OR: 4.03, 95% CI 2.806-5.786). The restricted cubic spline curves (RCS) regression model demonstrated that the relationships between systolic pressure (SBP), alanine aminotransferase (ALT), serum urate (UA), total cholesterol (TCHO), triglyceride (TG), triglyceride glucose (TyG) index, high density lipoprotein cholesterol (HDLC), and MAFLD were nonlinear and the cutoff values for lean MAFLD and overweight MAFLD were different. The nomogram was constructed based on seven predictors: glycosylated hemoglobin A1c (HbA1c), serum ferritin (SF), ALT, UA, BMI, TyG index, and age. In the validation cohort, the area under the ROC curve was 0.866 (95% CI 0.842-0.891), with 83.8% sensitivity and 76.6% specificity at the optimal cutoff. The PPV and NPV was 63.3% and 90.8%, respectively. Furthermore, we used fivefold cross-validation and the average area under the ROC curve was 0.866 (Figure S3). The calibration curves for the model's predictions and the actual outcomes were in good agreement. The DCA findings demonstrated that the nomogram model was clinically useful throughout a broad threshold probability range. CONCLUSIONS Lean and overweight MAFLD exhibit distinct metabolic profiles. The nomogram model developed in this study is designed to assist clinicians in the early identification of high-risk individuals with lean MAFLD, including those with a normal BMI but at metabolic risk, as well as those with abnormal blood lipid, glucose, uric acid or transaminase levels. In addition, this model enhances screening efforts in communities and medical screening centers, ultimately ensuring more timely and effective medical services for patients.
Collapse
Affiliation(s)
- Ruya Zhu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Caicai Xu
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China
| | - Suwen Jiang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jianping Xia
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Boming Wu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Sijia Zhang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongliang Liu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China.
| | - Jianjun Lou
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China.
| |
Collapse
|
76
|
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
|
77
|
Pecoraro V, Nascimbeni F, Cuccorese M, Gabrielli F, Fasano T, Trenti T. Diagnostic Accuracy of Golgi Protein 73 (GP73) for Liver Fibrosis Staging in Metabolic Dysfunction-Associated Steatotic Liver Disease: A Scoping Review and Cohort Study. Diagnostics (Basel) 2025; 15:544. [PMID: 40075792 PMCID: PMC11898419 DOI: 10.3390/diagnostics15050544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Golgi protein 73 (GP73) is a transmembrane protein expressed by epithelial cells of the bile duct in the normal liver. High serum levels of GP73 have been detected in patients with acute or chronic liver diseases, MASLD, and its measurement has been suggested as a potential biomarker for liver fibrosis staging. We evaluated the utility of GP73 in the diagnosis of MASLD, MASH, and for liver fibrosis staging. Methods: We performed a literature scoping review to map the current evidence about the accuracy of GP73 in patients with MASLD. We searched in Medline and EMBASE for English studies reporting an AUC value of GP73 in diagnosing MASLD and MASH and evaluating GP73 for fibrosis staging. A narrative synthesis of the evidence was conducted. Moreover, we performed an observational study including 84 patients with MASLD, of which 60 were biopsy-confirmed MASH, and different liver fibrosis stages, and 15 healthy controls. Serum GP73 levels were determined using a chemiluminescent assay and reported as mean and standard deviation (SD). Sensitivity (SE), specificity (SP), the area under the receiver operating characteristic (AUROC) curve, and the optimal cut-off value were calculated. Data were considered statistically significant when p < 0.05. Results: Available studies evaluating GP73 in MASLD reported the ability to discriminate MASH from simple steatosis and distinguish patients at different fibrotic stages, but the evidence is still scarce. Our experimental study showed that the serum levels of GP73 were 30 ± 12 ng/mL in MASLD and 32 ± 12 ng/mL in MASH patients and were statistically higher than those of the control group (19 ± 30 ng/mL), increasing from liver fibrosis stage F0 to F4. GP73 levels were significantly higher in patients with significant and advanced fibrosis than controls and no significant fibrosis (p > 0.05). ROC analysis demonstrated that serum GP73 had a good diagnostic potential for MASLD (AUROC 0.85; SE 90%; SP 73%), MASH (AUROC 0.75; SE 82%; SP64%), and significant fibrosis (AUROC 0.7; SE 56%; SP 79%) and was better than other biomarkers for chronic liver diseases. Conclusions: Serum GP73 could support clinicians in the evaluation of patients with MASH and significant fibrosis.
Collapse
Affiliation(s)
- Valentina Pecoraro
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | | - Michela Cuccorese
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | | - Tommaso Fasano
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | |
Collapse
|
78
|
He Q, Liu X, Ding G, Wang Y, Luo X, Cao W, Xing W. The relationship between serum uric acid level and non-alcoholic fatty liver disease in northern China: a retrospective cohort study. BMC Public Health 2025; 25:718. [PMID: 39984884 PMCID: PMC11843771 DOI: 10.1186/s12889-025-21943-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease among adults. High uric acid (UA) increases the incidence of NAFLD in the general population. However, further exploration is warranted to determine the relationship between UA levels and NAFLD in various populations. We conducted a historical cohort study to investigate the causality between UA and NAFLD across different weight categories. METHODS A historical cohort was established from the Jidong community cohort. All participants were enrolled and followed up from July 1st, 2013 to August 1st, 2018. The study participants were retrospectively assigned to four groups according to their UA levels (Q1, 69-210 μmol/L; Q2, 211-255 μmol/L; Q3, 256-310 μmol/L; Q4, 311-593 μmol/L). The NAFLD incidence was investigated in each group. We used the UA level determined by an automatic analyzer. NAFLD was diagnosed with abdominal ultrasonography examination. Demographic information, lifestyle history, clinical anthropometric data, and blood samples of participants were collected. Univariate analysis and multivariable Cox regression were applied to analyze the relationship between UA and NAFLD by stratification of participants' body mass index (BMI) categories (underweight, normal weight, overweight, and obese). RESULTS Two thousand nine hundred eighty four participants were enrolled. 740 (24.8%) were assigned to UA Q1 group, 755 (25.3%) to UA Q2, 743 (24.9%) to UA Q3, and 746 (25.0%) to UA Q4. The global incidence of NAFLD was 26.0% (777/2984). The risk of NAFLD significantly increased with elevated UA levels in underweight and normal-weight participants (HR = 3.498, 95% CI: 2.413-5.072, P < 0.05). In multivariable analysis, UA showed a positive association with NAFLD, independent of other risk factors in underweight and normal-weight participants (UA Q2: 1.152 (0.761-1.743), UA Q3: 2.168 (1.489-3.157), UA Q4: 3.075 (2.103-4.196), P < 0.05). In the absence of other risk factors, high UA levels independently explained 17% of NAFLD risk in underweight and normal-weight participants. CONCLUSIONS High UA levels serve as an independent risk factor for NAFLD in underweight and normal-weight individuals, highlighting the necessity of early NAFLD screening through monitoring liver function and UA levels, and personalized treatment plans for NAFLD patients with higher UA levels, which may include uric acid-lowering therapy and lifestyle modifications. However, the relationship between UA levels and NAFLD in overweight and obese individuals remains inconclusive.
Collapse
Affiliation(s)
- Qian He
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xinyue Liu
- Tai'an City Center for Disease Control and Prevention, Taian, China
| | - Guoyong Ding
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yiying Wang
- Department of Medical, Rizhao Mental Health Center, Rizhao, China
| | - Xiaoting Luo
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenyuan Cao
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Weijia Xing
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Shandong First Medical University & Shandong Academy of Medical Sciences, The Second Affiliated Hospital, Taian, 271000, China.
| |
Collapse
|
79
|
Lv Y, Sun M, He Y, Zhang X, Min Y, Liu L, Yu W. Effects of induced molting on lipid accumulation in liver of aged laying hens. Poult Sci 2025; 104:104941. [PMID: 40020412 PMCID: PMC11910710 DOI: 10.1016/j.psj.2025.104941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025] Open
Abstract
As the age of laying increases, the metabolic capacity of the liver decreases, leading to excessive lipid accumulation, which seriously affects the laying performance of laying hens. Induced molting (IM) can rejuvenate the reproductive system of older laying hens, allowing them to enter a new laying cycle. However, it remains unclear whether induced molting can enhance lipid accumulation in the liver of aged laying hens and what the underlying mechanism might be. In this study, fasting-induced molting was performed on 70-week-old Hy-line brown laying hens, and the resulting metabolic changes were analyzed using non-targeted metabolomics. Serum lipid levels, liver oxidative stress, and inflammation were measured using kits, while autophagy and lipid metabolism-related factors were assessed through immunofluorescence and western blotting. The results showed that IM could promote hepatic lipid deposition in aged laying hens, reduce hepatic steatosis and injury, lower the blood lipid level, improve hepatic antioxidant capacity and increase egg production rate. During the fasting period, the hepatic autophagic system was activated in laying hens and the level of hepatic autophagy increased. Additionally, AMPK phosphorylation levels increased, while the expression of fatty acid synthesis genes SREBP-1C, ACC, and FASN decreased (P < 0.01). The expression of PPARα, PGC 1α and CPT1A, which are associated with fatty acid oxidation, was upregulated (P < 0.01). In conclusion, IM enhanced lipid metabolism, increased liver autophagy, and improved liver function in aged laying hens.
Collapse
Affiliation(s)
- Yibo Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Mengqing Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yefei He
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaohan Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yahong Min
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Institute of Traditional Chinese Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Lin Liu
- Guangdong Haida Group Co., Ltd. Research Institute, Guangzhou 510535, PR China
| | - Wenhui Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Institute of Traditional Chinese Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
80
|
Xia L, Yu SX, Bai YS, Liang X, Wu FG, Gao Y, Chen XL, Xiao ZX, Li M. Effect of surgery on overall survival and cancer-specific survival in patients with primary HCC: A study based on PSM in the SEER cohort. Medicine (Baltimore) 2025; 104:e41521. [PMID: 39993067 PMCID: PMC11857015 DOI: 10.1097/md.0000000000041521] [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: 06/13/2024] [Revised: 12/04/2024] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
This study was designed to assess the effects of surgery method on overall survival (OS) and cancer-specific survival (CSS) in patients with hepatocellular carcinoma (HCC). This is a retrospective study. Patients diagnosed with primary HCC (N = 10,174) were identified from the Surveillance, Epidemiology, and End Results Database from 2010 to 2017 and categorized into surgical (N = 4950) and nonsurgical (N = 5224) groups. The characteristics of patients were balanced by propensity score matching. Multivariate Cox analysis was used to explore independent prognostic factors for outcomes in both groups, and the Kaplan-Meier curve showed survival rates in each group. The surgical patients were subclassified by surgical method, whether local tumor destruction, wedge or segmental resection, lobotomy resection, or liver transplantation (LT). Finally, survival rates in the 2 groups were investigated by subgroup analysis. After propensity score matching, sex, grade, tumor node metastasis III/IV, surgery, chemotherapy, alpha-fetoprotein, number of regional lymph nodes, other race, and age > 70 were independent prognostic factors in the 2 groups. The OS (HR = 0.290, P < .001) and CSS (HR = 0.274, P < .001) rates of patients were higher in the surgical group than in the nonsurgical group. There was no obvious improvement in CSS in patients who received radiotherapy combined with surgery compared with patients who only received radiotherapy (HR = 0.813, P = .279). LT was consistently found to be the best of the 4 surgical methods. The OS of stage II patients undergoing LT was better than that of corresponding stage III patients, and lobectomy resection was the best choice for stage IV patients (HR = 0. 417, P = .023). In grade III patients, the median CSS time was longer than the OS time. The survival rate of patients treated with chemotherapy combined with LT was higher than that of patients who did not receive chemotherapy and only received LT. Patients with HCC who underwent surgery had better OS and CSS. Subgroup analysis showed that LT can improve the survival rate and median survival time of patients.
Collapse
Affiliation(s)
- Lin Xia
- School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shuai-Xin Yu
- School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yu-Shuai Bai
- School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiao Liang
- School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fu-Gui Wu
- School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yang Gao
- School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiu-Li Chen
- Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei, China
| | | | - Man Li
- School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
81
|
Wang Y, Chen Z, Wei B, Zhang R, Zhang X, Xu W. Metabolic dysfunction associated steatotic liver disease is associated with atrial fibrillation recurrence following cryoballoon ablation. Sci Rep 2025; 15:6287. [PMID: 39984596 PMCID: PMC11845672 DOI: 10.1038/s41598-025-90667-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/14/2025] [Indexed: 02/23/2025] Open
Abstract
Atrial fibrillation (AF) is a common arrhythmia often treated with cryoballoon ablation. The impact of Metabolic-associated fatty liver disease (MASLD), a condition newly defined by a fatty liver index ≥ 60, on AF recurrence post-ablation is unclear. We analyzed 303 patients undergoing cryoballoon ablation for AF. Cox proportional hazards models were used to assess the relationship between MASLD and AF recurrence. Paroxysmal atrial fibrillation was present in 61.1% of patients and 63% were male. Among the patients, 23.4% had MASLD. These patients exhibited larger left atrial diameter and left ventricular end-diastolic dimension. During a median follow-up of 14 months, AF recurrence was more frequent in MASLD patients (45.1% vs. 20.7%). MASLD independently predicted AF recurrence (HR, 2.24 [95% CI 1.35-3.74], P = 0.002), alongside persistent AF, longer AF duration, and larger left atrial diameter. MASLD consistently demonstrated a significant association with an increased risk of AF recurrence in both paroxysmal (HR, 2.38 [95% CI, 1.08-5.23], P = 0.031) and persistent AF (HR, 2.55 [95% CI, 1.23-5.26], P = 0.011). MASLD significantly increases the risk of AF recurrence after cryoballoon ablation, highlighting the importance of supporting targeted interventions of MASLD in the periprocedural management of AF.
Collapse
Affiliation(s)
- Yu Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321, Zhongshan Road, Nanjing, 210008, China
| | - Zheng Chen
- Department of Cardiology, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - Bingqian Wei
- Department of Cardiology, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - RuiXin Zhang
- Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinlin Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321, Zhongshan Road, Nanjing, 210008, China.
| | - Wei Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321, Zhongshan Road, Nanjing, 210008, China.
- Department of Cardiology, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
82
|
Jiang W, Zeng Q, Liu CH, Wang Y, Wang S, Chen E, Wang M, Zhou T, Bai L, Wu D, Tang H. Huc-MSCs-derived exosomes alleviate non-alcoholic steatohepatitis by regulating macrophages polarization through miR-24-3p/STING axis. Stem Cell Res Ther 2025; 16:74. [PMID: 39984996 PMCID: PMC11846240 DOI: 10.1186/s13287-025-04197-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND There's a scarcity of drugs effective against nonalcoholic steatohepatitis (NASH). Exosomes from Human umbilical cord mesenchymal stem cells (huc-MSCs) show potential in managing glycolipid metabolism and the immune response. Therefore, further investigations are required to explore their application in NASH and the underlying mechanisms. METHODS C57BL/6J mice were fed with a western diet for 12 weeks to induce NASH, and huc-MSCs exosomes (MSCs-exo) were administered during the feeding period. The effect of MSCs-exo was evaluated by monitoring changes in body weight, fat distribution, blood glucose, and insulin levels, and analyzing pathological alterations in liver tissue. Mechanism investigations were carried out using flow cytometry, immunofluorescence staining, and other experimental techniques. RESULTS MSCs-exo could reduce liver fat, inflammation, fibrosis, and improved metabolism to alleviate the progression of NASH. Besides, MSCs-exo could decrease macrophage accumulation in the liver, encouraging M2 over M1 macrophage polarization. Furthermore, our study found that MSCs-exo had a high expression of miR-24-3p, which may regulate macrophage polarization by targeting the interferon-stimulated genes (STING) gene in macrophages, with its overexpression amplifying MSCs-exo's NASH benefits. CONCLUSIONS These findings suggest that the therapeutic effect of MSCs-exo on NASH may be attributed to the regulation of macrophage M2 polarization through miR-24-3p targeting STING. This provides a scientific basis for future clinical application.
Collapse
Affiliation(s)
- Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yonghong Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Shisheng Wang
- Liver Surgery and Liver Transplant Center, Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Taoyou Zhou
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
83
|
Li Q, Xiang J. METTL3 promotes the progression of non-alcoholic fatty liver disease by mediating m6A methylation of FAS. Sci Rep 2025; 15:6162. [PMID: 39979577 PMCID: PMC11842791 DOI: 10.1038/s41598-025-90419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
N6-methyladenosine (m6A) is involved in the development of non-alcoholic fatty liver disease (NAFLD). Here, we aimed to investigate the effect of m6A methyltransferase METTL3 on liver damage in high-fat diet (HFD)-induced mouse model and hepatocyte damage treated with free fatty acid (FFA). Plasma lipid, lipogenesis, viability, and apoptosis were measured to assess injury. m6A methylation was evaluated using m6A dot blot, methylated RNA immunoprecipitation, dual-luciferase reporter assay, and RNA decay assay. The results indicated that METTL3 was highly expressed in the liver of HFD mice, which knockdown improved plasma lipid and reduced liver lipids. Additionally, silencing of METTL3 promoted cell viability, inhibited apoptosis, reduced lipid concentrations, and downregulated lipogenesis-related marker levels. Moreover, METTL3 promoted the m6A methylation of FAS and enhanced its stability. In conclusion, silencing of METTL3 attenuates the progression of NAFLD by FAS m6A methylation, suggesting that METTL3 may be a promising target for treating NAFLD.
Collapse
Affiliation(s)
- Qunhua Li
- Department of Gastroenterology, Affiliated Hospital of Chengdu University, 2nd N Section of 2nd Ring Rd, Chengdu, 610036, Sichuan, China
| | - Junying Xiang
- Department of Gastroenterology, Affiliated Hospital of Chengdu University, 2nd N Section of 2nd Ring Rd, Chengdu, 610036, Sichuan, China.
| |
Collapse
|
84
|
Mi K, Ye T, Zhu L, Pan CQ. Risk-stratified hepatocellular carcinoma surveillance in non-cirrhotic patients with MASLD. Gastroenterol Rep (Oxf) 2025; 13:goaf018. [PMID: 39980834 PMCID: PMC11842057 DOI: 10.1093/gastro/goaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 02/22/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly emerging as the leading global liver disorder and is poised to become the primary cause of hepatocellular carcinoma (HCC). Research indicates that nearly 50% of HCC cases in MASLD patients occur without cirrhosis, often presenting with more advanced and larger tumors. Despite this, current guidelines primarily focus on HCC screening in cirrhotic patients, with limited guidance for non-cirrhotic MASLD individuals. This narrative review seeks to identify key risk factors for HCC development, consolidate available screening methods, and propose a practical, risk-stratified algorithm for HCC surveillance in non-cirrhotic MASLD patients. We conducted a comprehensive review of studies published between 2017 and 2023 using PubMed, Embase, and CNKI, focusing on HCC risk factors and emerging screening strategies for non-cirrhotic MASLD cohorts. Key risk factors for HCC development in these patients include male sex, age over 65, hypertension, diabetes, mild alcohol consumption, smoking, dyslipidemia, elevated alanine aminotransferase levels, and a platelet count ≤ 150 × 109/L. Among the screening methods evaluated, circulating free DNA, alpha-fetoprotein (AFP) combined with protein induced by vitamin K absence or antagonist-II (PIVKA-II), and the GALAD score (incorporating Glypican-3, AFP, alpha-1-Antitrypsin, and des-gamma-carboxy prothrombin) demonstrated the highest performance. Based on these findings, we proposed a risk-stratified HCC surveillance algorithm that integrates GALAD and PIVKA-II into the existing sonography and AFP screening protocols. This review aims to provide clinicians with actionable recommendations for HCC screening in non-cirrhotic MASLD patients.
Collapse
Affiliation(s)
- Ke Mi
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Tingdan Ye
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Lin Zhu
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
| | - Calvin Q Pan
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P. R. China
- Division of Gastroenterology and Hepatology, Department of Medicine, NYU Langone Health, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
85
|
Wang L, Tang W, Sun N, Lv J, Hu J, Tao L, Zhang C, Wang H, Chen L, Xu DX, Zhang Y, Huang Y. Low-dose tire wear chemical 6PPD-Q exposure elicit fatty liver via promoting fatty acid biosynthesis in ICR mice. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137574. [PMID: 39986096 DOI: 10.1016/j.jhazmat.2025.137574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/13/2025] [Accepted: 02/09/2025] [Indexed: 02/24/2025]
Abstract
N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine-quinone (6PPD-Q) as a major metabolite of tire wear chemical 6PPD has been demonstrated to be an emerging burden of exposure in human populations, via contamination from drinking water, air particulate matter and food sources. Whilst increasing attention has been moved toward its adverse effect, the potential hepatotoxicity of 6PPD-Q in mammals at realistic dose remains unknown. Here, the toxic effects of 6PPD-Q at environmentally relevant dose on the liver of adult mice and its underlying mechanism were investigated through an integrative approach combining transcriptomic and lipidomic analyses. We found that 6PPD-Q exposure induced excessive lipid deposition following three weeks of exposure, ultimately contributing to the pathogenesis of fatty liver disease. Mechanistically, 6PPD-Q exposure caused a remarkable increase in the contents of fatty acids within the hepatic tissue of mice by enhancing their biosynthesis, thereby facilitating lipid deposition. In summary, this study provides a new understanding on the endocrine disrupting effects of 6PPD-Q on hepatic lipid metabolism and how it may contribute to elevated risk of fatty liver disease. Our findings call for a potential public health attention on the risk assessment of 6PPD-Q, particularly towards the risk of chronic metabolic diseases.
Collapse
Affiliation(s)
- Lili Wang
- Department of General Practice, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Weitian Tang
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Nan Sun
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Jia Lv
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Jiayue Hu
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Lin Tao
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Cheng Zhang
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Hua Wang
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Li Chen
- Department of General Practice, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - De-Xiang Xu
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Yihao Zhang
- School of Public Health, Anhui Medical University, Hefei 230032, China.
| | - Yichao Huang
- School of Public Health, Anhui Medical University, Hefei 230032, China; Clinical Research Center, Suzhou Hospital of Anhui Medical University, Suzhou 234099, China.
| |
Collapse
|
86
|
Zeng Y, Wu Q, Guo M, Teng F, Jiang C, Chen J, Tan X, Zeng C, Long Y, Law BYK, Xu Y. Gut microbiota-derived imidazole propionate: an emerging target for the prevention and treatment of cardiometabolic diseases. Front Endocrinol (Lausanne) 2025; 16:1409119. [PMID: 40034229 PMCID: PMC11872695 DOI: 10.3389/fendo.2025.1409119] [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: 03/29/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Despite significant advancements in prevention and treatment, cardiometabolic diseases continue to pose a high burden of incidence and mortality. The chronic progression of these diseases necessitates the identification of early and complementary therapeutic targets to elucidate and mitigate residual risks in patient care. The gut microbiota acts as a sentinel between internal and external environments, transmitting modified risks associated with these factors to the host. Imidazole propionate (ImP), a histidine metabolite originating from the gut microbiota, gained attention after being found to impair glucose tolerance and insulin signaling several years ago. Epidemiological studies over the past five years have demonstrated a robust correlation between ImP and an increased risk of onset of type 2 diabetes (T2D) and obesity, exacerbation of kidney traits in chronic kidney disease (CKD), progression of atherosclerotic plaques, and elevated mortality rates in heart failure (HF). These findings suggest that ImP may serve as a pivotal target for the prevention and treatment of cardiometabolic diseases. Mechanistic insights have uncovered associations between ImP and insulin resistance, impaired glucose metabolism, chronic inflammation, and intestinal barrier damage. This review provides a comprehensive summary of the current evidence regarding the association between ImP and cardiometabolic impairment, highlighting its potential in advancing personalized approaches to disease prevention and management, and exploring the intricate interplay of diet, gut microbiota, and ImP in cardiovascular metabolic impairment. Overall, this review offers valuable insights into the multifaceted roles of ImP in cardiometabolic diseases, identifies current knowledge gaps, and discusses future research directions.
Collapse
Affiliation(s)
- Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, The Afiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Man Guo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Fangyuan Teng
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxia Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jiao Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaozhen Tan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Long
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
87
|
Zhou L, Jiang L, An Y, Liu J, Wang G, Wang Y, Yang N. Association of Sensitivity to Thyroid Hormones and Non-Alcoholic Fatty Liver Disease and the Severity of Liver Fibrosis in Euthyroid Adults: A Retrospective Study. Diabetes Metab Syndr Obes 2025; 18:479-490. [PMID: 39990180 PMCID: PMC11844316 DOI: 10.2147/dmso.s499517] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/05/2025] [Indexed: 02/25/2025] Open
Abstract
Purpose The relationship between non-alcoholic fatty liver disease (NAFLD) and thyroid function is controversial. A mild acquired thyroid hormone resistance may exist and explain these contradictions. This study aims to investigate the associations of thyroid hormone sensitivity with NAFLD and the severity of liver fibrosis in euthyroid populations. Patients and Methods Twenty-nine thousand three hundred and eighty-six adult subjects were recruited from the medical examination center of the Beijing Chao-Yang Hospital. Free triiodothyronine (FT3)/free thyroxine (FT4), thyroid feedback quartile-based index for FT4 (TFQIFT4) and for FT3 (TFQIFT3), thyroid stimulating hormone index (TSHI), thyrotropin thyroxine resistance index (TT4RI), and thyrotropin triiodothyronine resistance index (TT3RI) were calculated. Logistic regression analysis was used to analyze the associations of thyroid hormone sensitivity indices with the risks of NAFLD and related metabolic disorders. The correlation between thyroid hormone sensitivity and the severity of liver fibrosis evaluated by noninvasive fibrosis indices was analyzed through ordinal logistic regression. Results Euthyroid adults with NAFLD had elevated levels of serum FT3 and FT4, reduced TSH, and impaired sensitivity to thyroid hormones. Compared with participants in the first quartile group, the risk of NAFLD was higher in the fourth quartile of TFQIFT3 (OR 1.25, 95% CI 1.13-1.39, P < 0.001) and FT3/FT4 (OR 1.45, 95% CI 1.32-1.60, P < 0.001) after adjusting for metabolic confounders. Among NAFLD patients, higher FT3/FT4 positively correlated with increased severity of liver fibrosis, with ORs per SD of 10.80 (95% CI 4.12-28.53, P < 0.001) for aminotransferase-to-platelet ratio index, 4.74 (95% CI 1.56-14.38, P = 0.006) for NAFLD fibrosis score and 3.21 (95% CI 1.02-10.08, P = 0.046) for fibrosis-4 index. Conclusion Impaired central sensitivity to FT3 and higher FT3/FT4 were associated with increased risks of NAFLD and related metabolic disorders. In patients with NAFLD, higher FT3/FT4 correlated with its progression to liver fibrosis. These findings might provide novel insight into the monitoring and evaluation of the risk of NAFLD and the severity of liver fibrosis.
Collapse
Affiliation(s)
- Liyuan Zhou
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Lanxuan Jiang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Yu An
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Ying Wang
- Medical Examination Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| | - Ning Yang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People’s Republic of China
| |
Collapse
|
88
|
Shi D, Tan Q, Zhang Y, Qi X, Xu X, Xu G, Bai R, Deng J, Chen M, Jiang T, Mei Y. Serum uric acid trajectories and risk of metabolic dysfunction-associated steatotic liver disease in China: a 2019-2021 cohort health survey. BMC Public Health 2025; 25:653. [PMID: 39962427 PMCID: PMC11834244 DOI: 10.1186/s12889-024-21214-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 12/26/2024] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE To investigate the associations between serum uric acid (UA) trajectories and the risk of metabolic dysfunction-associated steatotic liver disease (MASLD) in large cohort survey 2019-2021. METHODS This cohort health survey included 11,644 participants without MASLD before January 1, 2021. Among them, 5578 (47.90%) were men and 6066 (52.10%) were women. The group-based trajectory model method was applied to identify serum UA trajectories from January 1, 2019, to December 30, 2021. New-onset MASLD events in 2021 were treated as outcomes. A logistic regression model was used to assess associations between UA trajectories and incidence of MASLD. RESULTS Four distinct serum UA trajectories among both sexes were identified: "low-stable" trajectory 1 (n = 783 men; n = 1143 women), "moderate-moderate increasing" trajectory 2 (n = 2794 men; n = 3266 women), "moderate high-moderate increasing" trajectory 3 (n = 1660 men; n = 1464 women), and "high-increasing" trajectory 4 (n = 341 men; n = 193 women). During the 3-year follow-up period, 840 (15.06%) men and 408 (6.72%) women developed MASLD, respectively. Compared with the trajectory 1 group, the trajectory 4 group had the highest risk (odds ration [OR] 2.99 [95% confidence interval {CI} 1.70, 5.24] for men; OR 2.37 [95% CI 1.04, 5.33] for women), followed by the trajectory 3 (OR 2.23 [95% CI 1.52, 3.30] for men; OR 2.37 [95% CI 1.45, 3.92] for women) and trajectory 2 (OR 1.43 [95% CI 1.07, 1.94] for men; OR 1.37 [95% CI 0.93, 2.03] for women) groups. CONCLUSIONS High serum UA trajectories were independent risk factors for MASLD in both sexes, which is critical for informing prevention and treatment strategies in public health initiatives and clinical practice.
Collapse
Affiliation(s)
- Dan Shi
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Qilong Tan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
- School of Public Health, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yong Zhang
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Xiaoya Qi
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Xiaoyang Xu
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Guoqiong Xu
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Ruixue Bai
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Jing Deng
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Mengxue Chen
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China
| | - Tao Jiang
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China.
| | - Ying Mei
- Department of Health Management, The Second Hospital Affiliated to Chongqing Medical University, Chongqing, 400011, China.
| |
Collapse
|
89
|
Byrne CD, Armandi A, Pellegrinelli V, Vidal-Puig A, Bugianesi E. Μetabolic dysfunction-associated steatotic liver disease: a condition of heterogeneous metabolic risk factors, mechanisms and comorbidities requiring holistic treatment. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-025-01045-z. [PMID: 39962331 DOI: 10.1038/s41575-025-01045-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 03/09/2025]
Abstract
Μetabolic dysfunction-associated steatotic liver disease (MASLD) comprises a heterogeneous condition in the presence of steatotic liver. There can be a hierarchy of metabolic risk factors contributing to the severity of metabolic dysfunction and, thereby, the associated risk of both liver and extrahepatic outcomes, but the precise ranking and combination of metabolic syndrome (MetS) traits that convey the highest risk of major adverse liver outcomes and extrahepatic disease complications remains uncertain. Insulin resistance, low-grade inflammation, atherogenic dyslipidaemia and hypertension are key to the mechanisms of liver and extrahepatic complications. The liver is pivotal in MetS progression as it regulates lipoprotein metabolism and secretes substances that affect insulin sensitivity and inflammation. MASLD affects the kidneys, heart and the vascular system, contributing to hypertension and oxidative stress. To address the global health burden of MASLD, intensified by obesity and type 2 diabetes mellitus epidemics, a holistic, multidisciplinary approach is essential. This approach should focus on both liver disease management and cardiometabolic risk factors. This Review examines the link between metabolic dysfunction and liver dysfunction and extrahepatic disease outcomes, the diverse mechanisms in MASLD due to metabolic dysfunction, and a comprehensive, personalized management model for patients with MASLD.
Collapse
Affiliation(s)
- Christopher D Byrne
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton, UK
| | - Angelo Armandi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vanessa Pellegrinelli
- Institute of Metabolic Science, MRC MDU Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Antonio Vidal-Puig
- Institute of Metabolic Science, MRC MDU Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
90
|
Lajeunesse-Trempe F, Dugas S, Maltais-Payette I, Tremblay ÈJ, Piché ME, K. Dimitriadis G, Lafortune A, Marceau S, Biertho L, Tchernof A. Anthropometric Indices and Metabolic Dysfunction-Associated Fatty Liver Disease in Males and Females Living With Severe Obesity. Can J Gastroenterol Hepatol 2025; 2025:5545227. [PMID: 39989658 PMCID: PMC11847611 DOI: 10.1155/cjgh/5545227] [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: 10/23/2023] [Revised: 05/28/2024] [Accepted: 11/07/2024] [Indexed: 02/25/2025] Open
Abstract
Introduction: Metabolic dysfunction-associated fatty liver disease (MAFLD) is highly prevalent among people living with severe obesity (body mass index [BMI] ≥ 35 kg/m2). However, it remains unknown how sex and adipose tissue distribution are related to MAFLD onset and progression into metabolic dysfunction-associated steatohepatitis (MASH) or advanced stages of fibrosis. Methodology: We retrospectively studied patients with severe obesity who were eligible for bariatric surgery. Demographic characteristics, biomarkers, and cardiometabolic comorbidities were reported. Anthropometric indices such as BMI, waist circumference (WC), waist-to-hip ratio (WHR), waist-to-height ratio (WHtR), neck circumference (NC), lipid accumulation product (LAP), visceral adiposity index (VAI), body adiposity index (BAI), abdominal volume index (AVI), and body roundness index (BRI) were measured or calculated. MAFLD, MASH, and stages of fibrosis (F1-F4) were established from perioperative liver biopsies. Standardized univariate and multivariate logistic regression analyses were used to examine the association between demographic variables, anthropometric indices, cardiometabolic conditions, and the risk of MASH or severe fibrosis (F2-F4). Results: A total of 2091 participants with severe obesity were included in the analyses; BMI 47.9 ± 7.3 kg/m2, age 46.2 ± 11.2 years, and 68.4% females. Overall, MAFLD prevalence was 79.5%, with 44.5% having MASH and 24.4% having severe fibrosis (Stage 2 or higher). No anthropometric indices of adiposity were associated with MASH or fibrosis severity. In this population, female sex was a risk factor for severe fibrosis (OR: 1.27, 95% CI 1.01-1.59, p < 0.05). Conclusions: MAFLD and MASH are highly prevalent in individuals living with severe obesity, but no anthropometric indices or laboratory tests are good predictors of MAFLD or MASH in this population. When MAFLD is diagnosed, our results suggest that females with severe obesity might be at higher risk of advanced stages of fibrosis.
Collapse
Affiliation(s)
- Fannie Lajeunesse-Trempe
- Department of Specialized Medicine, Internal Medicine, Quebec Heart and Lung Institute, Laval University, Quebec City, Quebec, Canada
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
- Faculty of Life Sciences and Medicine, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | - Selena Dugas
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
| | - Ina Maltais-Payette
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
| | - Ève-Julie Tremblay
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
| | - Marie-Eve Piché
- Department of Medicine, Laval University, Quebec City, Quebec, Canada
| | - Georgios K. Dimitriadis
- Faculty of Life Sciences and Medicine, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
- Department of Endocrinology, King's College Hospital NHS Foundation Trust, London, UK
| | - Annie Lafortune
- Department of Surgery, Laval University, Quebec City, Quebec, Canada
| | - Simon Marceau
- Department of Surgery, Laval University, Quebec City, Quebec, Canada
| | - Laurent Biertho
- Department of Surgery, Laval University, Quebec City, Quebec, Canada
| | - André Tchernof
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
91
|
Tang SS, Zhao XF, An XD, Sun WJ, Kang XM, Sun YT, Jiang LL, Gao Q, Li ZH, Ji HY, Lian FM. Classification and identification of risk factors for type 2 diabetes. World J Diabetes 2025; 16:100371. [PMID: 39959280 PMCID: PMC11718467 DOI: 10.4239/wjd.v16.i2.100371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/24/2024] [Accepted: 11/26/2024] [Indexed: 12/30/2024] Open
Abstract
The risk factors for type 2 diabetes mellitus (T2DM) have been increasingly researched, but the lack of systematic identification and categorization makes it difficult for clinicians to quickly and accurately access and understand all the risk factors, which are categorized in this paper into five categories: Social determinants, lifestyle, checkable/testable risk factors, history of illness and medication, and other factors, which are discussed in a narrative review. Meanwhile, this paper points out the problems of the current research, helps to improve the systematic categorisation and practicality of T2DM risk factors, and provides a professional research basis for clinical practice and industry decision-making.
Collapse
Affiliation(s)
- Shan-Shan Tang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, China
| | - Xue-Fei Zhao
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xue-Dong An
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Wen-Jie Sun
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xiao-Min Kang
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Yu-Ting Sun
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Lin-Lin Jiang
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Qing Gao
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Ze-Hua Li
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Hang-Yu Ji
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Feng-Mei Lian
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| |
Collapse
|
92
|
Ko CJ, Lin HY, Hsieh PM, Wang WL, Chen SY, Chou LW, Chen YS, Huang YW, Ho WC, Lin CW. Association of concomitant MASLD and hepatitis B virus with clinical prognosis in hepatocellular carcinoma after curative resection. Am J Cancer Res 2025; 15:737-748. [PMID: 40084366 PMCID: PMC11897618 DOI: 10.62347/ksln5850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
The term "metabolic dysfunction-associated steatotic liver disease" (MASLD) was introduced to replace the term "nonalcoholic fatty liver disease". The prevalence of MASLD is increasing worldwide. The prevalence of concomitant MASLD and hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) is also increasing. This study explored the effect of the coexistence of MASLD and HBV on clinicopathological features and long-term clinical prognoses in patients with MASLD-related and/or HBV-related HCC after curative hepatectomy. The study retrospectively collected the data of 653 patients with HCC who had undergone curative hepatectomy between 2011 and 2022. We assessed the association of histologically confirmed MASLD with HCC recurrence and mortality. Of 653 patients, 320 (49.0%), 103 (15.8%), and 230 (35.2%) had concomitant MASLD and HBV, MASLD only, and HBV only, respectively. The median follow-up period was 5.1 years. Patients with concomitant MASLD and HBV were at a significantly increased risk of HCC recurrence (P = 0.013 and P = 0.041) and mortality (P = 0.044 and P = 0.026) than those with MASLD or HBV alone. In multivariable analyses, concomitant MASLD and HBV, male sex, body mass index < 23, absence of antiviral therapy, and tumor size ≥ 5 cm were significantly associated with increased HCC recurrence. Concomitant MASLD and HBV, male sex, type 2 diabetes mellitus, serum aspartate aminotransferase ≥ 40 U/L, tumor size ≥ 5 cm, tumor cell differentiation II-III, microvascular invasion, lymph node invasion, and tumor recurrence were significantly associated with increased mortality. In conclusion, patients with concomitant MASLD and HBV are at a significantly greater risk of HCC recurrence and mortality after curative hepatectomy than those with MASLD or HBV alone.
Collapse
Affiliation(s)
- Chih-Jan Ko
- Department of Public Health, China Medical UniversityTaichung, Taiwan
- Department of General Surgery, China Medical University Hsinchu HospitalHsinchu, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, and Research Center for Traditional Chinese Medicine, China Medical UniversityTaichung, Taiwan
| | - Hung-Yu Lin
- Department of Surgery, E-Da Cancer Hospital, I-Shou UniversityKaohsiung, Taiwan
- Department of Surgery, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
| | - Pei-Min Hsieh
- Department of Surgery, E-Da Dachang Hospital, I-Shou UniversityKaohsiung, Taiwan
- Division of Occupational Medicine, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
| | - Wen-Lung Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou UniversityKaohsiung, Taiwan
| | - Szu-Ying Chen
- School of Medicine, College of Medicine, I-Shou UniversityKaohsiung, Taiwan
- Division of Surgical Intensive Care, Department of Critical Care Medicine, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
- Department of Nursing, Fooyin UniversityKaohsiung, Taiwan
| | - Li-Wei Chou
- Department of Physical Medicine and Rehabilitation, China Medical University HospitalTaichung, Taiwan
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical UniversityTaichung, Taiwan
- Department of Physical Medicine and Rehabilitation, Asia University Hospital, Asia UniversityTaichung, Taiwan
| | - Yaw-Sen Chen
- Department of Surgery, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
| | - Yu-Wei Huang
- School of Medicine, College of Medicine, I-Shou UniversityKaohsiung, Taiwan
- Department of Nursing, Fooyin UniversityKaohsiung, Taiwan
- Department of Anesthesiology, Emergency and Critical Care Center, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
| | - Wen-Chao Ho
- Department of Public Health, China Medical UniversityTaichung, Taiwan
| | - Chih-Wen Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou UniversityKaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou UniversityKaohsiung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, and Research Center for Traditional Chinese Medicine, China Medical UniversityTaichung, Taiwan
| |
Collapse
|
93
|
Nie Q, Jiang Y, Li M, Liang Q, Mo X, Qiu T, Jiang Q, Huang K, Xie Y, Chen Y, Ma X, Li J, Jiang K. Global burden and international disparities in NASH-associated liver Cancer: mortality trends (1990-2021) and future projections to 2045. Front Public Health 2025; 13:1527328. [PMID: 40027504 PMCID: PMC11868124 DOI: 10.3389/fpubh.2025.1527328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Background NASH-associated liver cancer (NALC) is a significant contributor to global cancer mortality, closely linked to the increasing prevalence of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). This study comprehensively examines the global burden of NALC from 1990 to 2021. Methods This study used data from the Global Burden of Disease (GBD) 2021 database to analyze NALC death and age-standardized death rates (ASDR) globally and regionally from 1990 to 2021. We applied Joinpoint regression analysis to assess temporal trends, calculating the annual percent change (APC) and average annual percent change (AAPC). Decomposition analysis was performed to break down mortality changes into contributions from population aging, growth, and epidemiological changes. A frontier analysis was used to evaluate the relationship between NALC burden and sociodemographic development using the Socio-Demographic Index (SDI). Prediction analysis of NALC deaths and ASDR from 2021 to 2045 were estimated using the Nordpred model. Results From 1990 to 2021, the global burden of NALC deaths increased significantly, with the ASDR rising from 0.38 per 100,000 in 1990 to 0.48 per 100,000 in 2021. Age-specific data in 2021 revealed that NALC deaths peaked in the 65-69 age group for men and 70-74 age group for women. Decomposition analysis indicated that population growth was the most significant contributor to the global NALC death toll, followed by population aging and epidemiological changes. Frontier analysis showed that countries like Mongolia and Gambia were farthest from the disease burden frontier, while Morocco and Ukraine were closest. Prediction analysis suggest a significant increase in NALC deaths by 2045 compared to 2021, with a larger rise in deaths among women. Conclusion Through this study, a data-driven approach is provided to reduce the global disease burden of NALC. Essential data support for public health prevention strategies is offered, helping guide the development of targeted government interventions. Trends across global regions, countries, age groups, and genders have been analyzed, providing valuable insights for the formulation of evidence-based policies aimed at mitigating the impact of NALC worldwide.
Collapse
Affiliation(s)
- Qilong Nie
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yongwen Jiang
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Mingyang Li
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Qiuyan Liang
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Xiaoai Mo
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Tengyu Qiu
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Qunfang Jiang
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Kaizhou Huang
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Youqing Xie
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Ying Chen
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Xiaojun Ma
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Jianhong Li
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Kaiping Jiang
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| |
Collapse
|
94
|
Wang Y, Wang P. Development and validation of a new diagnostic prediction model for NAFLD based on machine learning algorithms in NHANES 2017-2020.3. Hormones (Athens) 2025:10.1007/s42000-025-00634-6. [PMID: 39939537 DOI: 10.1007/s42000-025-00634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
AIMS Nonalcoholic fatty liver disease (NAFLD) is a multisystem disease that can trigger the metabolic syndrome. Early prevention and treatment of NAFLD is still a huge challenge for patients and clinicians. The aim of this study was to develop and validate machine learning (ML)-based predictive models. The model with optimal performance would be developed as a set of simple arithmetic tools for predicting the risk of NAFLD individually. METHODS Statistical analyses were performed in 2428 individuals extracted from the National Health and Nutrition Examination Survey (NHANES, cycle 2017-2020.3) database. Feature variables were selected by the least absolute shrinkage and selection operator (LASSO) regression. Seven ML algorithms, including logistic regression (LR), decision tree (DT), random forest (RF), extreme gradient boosting (XGB), K-nearest neighbor (KNN), light gradient boosting machine (LightGBM), and multilayer perceptron (MLP), were used to construct models based on the feature variables and evaluate their performance. The model with the best performance was transformed into a diagnostic predictive nomogram (DPN). The DPN was developed into an online calculator and an Excel algorithm tool. Receiver operating characteristic (ROC) curve, decision curve analysis (DCA), and subgroup analyses were used to compare and assess the predictive abilities of the DPN and six existing NAFLD predictive models, including the ZJU index, the hepatic steatosis index (HSI), the triglyceride-glucose index (TyG), the Framingham steatosis index (FSI), the fatty liver index (FLI), and the visceral adiposity index (VAI). RESULTS Among the 2428 participants, the prevalence of NAFLD was 47.45%. LASSO regression identified eight variables from 39 variables, including body mass index (BMI), waist circumference (WC), alanine aminotransferase (ALT), triglyceride (TG), diabetes, hypertension, uric acid (UA), and race. Among the models constructed by the seven algorithms mentioned above, the LR-based model performed the best, demonstrating outstanding performance in terms of area under the curve (AUC, 0.823), accuracy (0.754), precision (0.768), specificity (0.804), and positive predictive value (0.768). It was then transformed into the DPN, which was successfully developed as an online calculator and an Excel algorithm tool. The diagnostic accuracy (AUC 0.856, 95% confidence interval (CI) 0.839-0.874, and AUC 0.823, 95% CI 0.793-0.854, respectively) and net clinical benefit of DPN in the training and validation sets were superior to those of the ZJU, HSI, TyG, FSI, FLI, and VAI. The results were maintained in subgroup analyses. CONCLUSIONS The LR model based on ML was developed, exhibiting good performance. DPN can be used as an individualized tool for rapid detection of NAFLD.
Collapse
Affiliation(s)
- Yazhi Wang
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Peng Wang
- The Department of Pharmacy, The 987th Hospital of Joint Logistics Support Force of People's Liberation Army, Baoji, Shaanxi, 721004, China.
| |
Collapse
|
95
|
Liu B, Shao T, Xiao D, Yang S, Lin W, Sun L, Zhang W, Luo M, Zhao J, Yang L, Bai S, Deng D, Wang C, Wang S, Zhang R, Liu Z, An L. Aqueous extract of Cornus officinalis alleviate NAFLD via protecting hepatocytes proliferation through regulation of the tricarboxylic acid cycle. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119330. [PMID: 39778783 DOI: 10.1016/j.jep.2025.119330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/02/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cornus officinalis (CO) has been widely used as Chinese herbal medicine and has a good clinical efficacy in liver disease. In particular, it has a significant therapeutic effect on metabolic liver disease. However, systematic pharmacological studies on its hepatoprotective effect on non-alcoholic fatty liver disease (NAFLD) are lacking. AIM OF THE STUDY We investigated the impact of Cornus officinalis extract (COE) on two mouse models of NAFLD, screened the potential mechanisms of action by using metabolomics assays, and explored the protective effects on hepatocyte proliferation by regulating glutamate metabolism and tricarboxylic acid (TCA) cycle. METHODS The main components of COE were identified by high performance liquid chromatograph (HPLC). Male C57BL/6J mice were subjected to construct carbon tetrachloride (CCl4) or methionine choline deficient (MCD) induced NAFLD mice. Liver function and lipid biochemical indicators were detected using commercial assay kits. Masson staining, Western blot, and immunohistochemistry analyses were used for assessing hepatic injury and fibrosis. LC-MS non-targeted analysis was performed using the 1290 Ultra-High Performance Liquid Chromatograph System and the 6540 Q-TOF Mass Spectrometry. Palmitic acid (PA) induced L-02 cell model was established. The mediators in glutamate metabolism and TCA cycle were assessed by assay kits. RESULTS In vivo experiments validated that COE significantly improved liver function in NAFLD mice, reduced lipid accumulation, and alleviated pathological damage and liver fibrosis. The non-targeted metabolomics analysis combined with Ingenuity Pathway Analysis (IPA) located glutamate metabolism and the downstream TCA cycle as potential mechanisms of COE, which was further confirmed in NAFLD model mice and PA-induced L-02 cells. Finally, we confirmed that COE could promote mitochondrial energy supply by remodeling the homeostasis of the TCA cycle, thereby enhancing hepatocyte proliferation. CONCLUSIONS This study demonstrated that COE could significantly improve CCl4 or MCD-induced NAFLD by promoting hepatocyte proliferation. These results highlighted the potential of COE as leads for the development of innovative treatments for NAFLD.
Collapse
Affiliation(s)
- Binjie Liu
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ting Shao
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dandan Xiao
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Shujie Yang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Weijie Lin
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lizhu Sun
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weiqin Zhang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Meiqing Luo
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jinlan Zhao
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lei Yang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shasha Bai
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Di Deng
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Caiyan Wang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| | - Shaogui Wang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Rong Zhang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China.
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China.
| | - Lin An
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China.
| |
Collapse
|
96
|
Zhang N, Li J, Xie X, Hu Y, Chen H, Zhang Y, Liu Y, Zhu X, Xu H, Wang Z, Baima K, Zhang X, Qin Z, Yu Z, Xiao X, Zhao X. Changes in drinking levels and metabolic dysfunction-associated steatotic liver disease: a longitudinal study from the China multi-ethnic cohort study. BMC Public Health 2025; 25:556. [PMID: 39934719 DOI: 10.1186/s12889-025-21752-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Little is known about the associations of changes in drinking levels with the newly defined metabolic dysfunction-associated steatotic liver disease (MASLD). We therefore sought to estimate the associations between changes in drinking levels and MASLD in less developed regions of China. METHODS This longitudinal study included 8727 participants from the China Multi-Ethnic Cohort (CMEC) in less developed regions, all participating in baseline and a follow-up survey. MASLD was defined as hepatic steatosis, along with the presence of at least one of five cardiometabolic risks, in addition to limiting excessive alcohol consumption. We applied the parametric g-formula to evaluate the association between changes in drinking levels and MASLD. We further estimated the association between changes in drinking levels and fibrosis scores (AST-to-platelet ratio and fibrosis-4 index) in patients with MASLD. RESULTS Compared with sustained non-drinking, sustained modest drinking was associated with a higher risk of MASLD (Mean Ratio (MR): 1.127 [95% CI: 1.040-1.242]). Compared to sustained non-drinking, the MR for those transitioning from non-drinking to modest drinking was 1.065 [95% CI: 0.983-1.169], while the MR for those changing from modest drinking to non-drinking was 1.059 [95% CI: 0.965, 1.173]. Non-invasive fibrosis scores tended to increase with modest drinking compared to sustained non-drinking. CONCLUSION In the less developed regions of China, sustained moderate drinking was associated with the risk of MASLD compared with sustained non-drinking. Increased drinking showed a trend towards a higher risk of MASLD. This study can inform drinking policies related to MASLD and liver fibrosis in less developed regions.
Collapse
Affiliation(s)
- Ning Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jingzhong Li
- Tibet Center for Disease Control and Prevention, Lhasa, China
| | - Xiaofen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yifan Hu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Hongxiang Chen
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuan Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yujie Liu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xingren Zhu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Hao Xu
- State Key Laboratory of Oral Diseases, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Sichuan University, Chengdu, China
| | - Zhenghong Wang
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing, China
| | - Kangzhuo Baima
- High Altitude Health Science Research Center of Tibet University, Lhasa, Tibet, China
| | - Xuehui Zhang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Zixiu Qin
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Zhimiao Yu
- Chengdu Center for Disease Control and Prevention, Chengdu, China
| | - Xiong Xiao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xing Zhao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
97
|
Li P, Zhang R, Hu P, Wang T, Wang J. Cepharanthine relieves nonalcoholic steatohepatitis through inhibiting STAT1/CXCL10 axis-mediated lipogenesis and inflammatory responses. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119358. [PMID: 39805479 DOI: 10.1016/j.jep.2025.119358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/24/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Stephania rotunda Lour., a medicinal herb, has been utilized in both Traditional Chinese Medicine (TCM) and Traditional Indian Medicine to treat conditions such as fever, dysentery, and inflammation. Cepharanthine (CEP), a primary active ingredient of Stephania rotunda Lour., has demonstrated a range of pharmacological activities, including anti-oxidative, anti-inflammatory, anti-cancer, anti-viral and anti-parasitic properties. However, the effects and underlying mechanisms of CEP on improving nonalcoholic steatohepatitis (NASH) remain unclear. AIM OF THE STUDY This study aimed to investigate the effects of CEP on mitigating diet-induced NASH and explore its underlying mechanisms. MATERIALS AND METHODS A High-Fat Diet (HFD) and the high levels of free fatty acids (FFA) were used to establish in vivo and in vitro NASH models to evaluate the intervention effect of CEP. Subsequently, RNA-sequencing, western blotting, quantitative real-time PCR (qRT-PCR) and siRNA transfection were employed to investigate its underlying mechanisms. RESULTS Our findings indicated that CEP significantly reduced lipogenesis and inflammatory responses in both HFD-fed rats and FFA-induced hepatic cells (including HepG2, L02 and AML12 cell lines), as is evidenced by the reduction of triglyceride (TG), lipid accumulation, and the release of inflammatory cytokines such as TNF-α, IL-6 and IL-1β. Mechanistically, CEP significantly inhibits CXC motif chemokine ligand 10 (CXCL10) expression both in vivo and in vitro. It also regulates sterol regulatory element binding protein-1c (SREBP1c)-induced lipogenic gene expression and CXCL10-mediated nuclear factor kappa B (NFκB) activation. Notably, knockdown of CXCL10 mimics the ability of CEP to reduce lipid accumulation and inflammatory responses, which is also observed following the blockade of signal transducer and activator of transcription 1 (STAT1) in HepG2 cells. CONCLUSION CEP alleviates NASH by inhibiting lipogenesis and inflammatory responses in a STAT1/CXCL10 axis-dependent manner.
Collapse
Affiliation(s)
- Pan Li
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Ruoyu Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, 999077, PR China
| | - Pingping Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing, 400016, PR China
| | - Tingting Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
98
|
van Eekeren LE, Vadaq N, Blaauw MJT, Groenendijk AL, Vos WAJW, Nelwan EJ, Verbon A, Stalenhoef JE, Berrevoets MAH, van Lunzen J, Netea MG, Weijers G, Riksen NP, Rutten JHW, de Mast Q, Tjwa ETTL, Joosten LAB, van der Ven AJAM. Distinct metabolic perturbations link liver steatosis and incident CVD in lean but not obese PWH. BMC Med 2025; 23:78. [PMID: 39934780 DOI: 10.1186/s12916-025-03914-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a key risk factor for cardiovascular disease (CVD), potentially driven by shared metabolic mechanisms. Metabolic perturbations associated with MASLD and CVD remain underexplored in people with HIV (PWH). METHODS We used data from the longitudinal multicenter 2000HIV study comprising 1895 virally suppressed PWH, out of which 970 had available liver and carotid artery measurements. Transient elastography with controlled attenuation parameter (CAP) was performed for the assessment of liver steatosis (CAP > 263 dB/m) and fibrosis (LSM ≥ 7.0). Historic and future incident CVD within 2-year follow-up, defined as myocardial infarction, stroke, peripheral arterial disease, and angina pectoris, were extracted from the medical files, while atherosclerotic plaque(s) in the carotid arteries were assessed using ultrasonography. Metabolic perturbations were analyzed using mass spectrometry-based untargeted metabolomics (n = 500 metabolites) and nuclear magnetic resonance spectroscopy for targeted lipids and other metabolites (n = 246 metabolites). RESULTS PWH with liver steatosis were more likely to have arterial plaques (47% vs. 36%; P value = 0.003) and CVD history (11% vs. 6.8%; P value = 0.021) than PWH without liver steatosis. These associations were only significant in lean PWH, in contrast to those with BMI ≥ 25 kg/m2. Metabolic pathways associated with liver steatosis and fibrosis primarily involved lipid and amino acid metabolism, and they were validated by targeted lipoproteomic measurements. Interestingly, metabolomic pathways and lipoproteomic signatures associated with MASLD were mostly distinct from those associated with CVD parameters. However, several metabolic pathways were shared, especially in lean PWH. These include arachidonic acid metabolism and formation of prostaglandin, purine metabolism, cholecalciferol metabolism, and glycine, serine, alanine, and threonine metabolism. CONCLUSION Metabolic disturbances linked to liver steatosis and CVD diverge across BMI categories in PWH. Lean PWH, unlike their overweight/obese counterparts, show common metabolic perturbations between MASLD and CVD, particularly involving arachidonic acid metabolism. This suggests that lean PWH with liver steatosis may face a heightened risk of CVD due to shared metabolic pathways, potentially opening avenues for targeted interventions, such as aspirin therapy, to mitigate this risk.
Collapse
Affiliation(s)
- Louise E van Eekeren
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands.
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands.
| | - Nadira Vadaq
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Division of Tropical Medicine and Infectious Disease, Department of Internal Medicine, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Marc J T Blaauw
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Department of Internal Medicine, Elisabeth-Tweesteden Hospital, Tilburg, the Netherlands
| | - Albert L Groenendijk
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Erni J Nelwan
- Division of Tropical Medicine and Infectious Disease, Department of Internal Medicine, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Annelies Verbon
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Marvin A H Berrevoets
- Department of Internal Medicine, Elisabeth-Tweesteden Hospital, Tilburg, the Netherlands
| | - Jan van Lunzen
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Department of Metabolism and Immunology, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Gert Weijers
- Medical UltraSound Imaging Center (MUSIC), Division of Medical Imaging, Radboudumc, Nijmegen, the Netherlands
| | - Niels P Riksen
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| | - Eric T T L Tjwa
- Department of Gastroenterology and Hepatology, Radboudumc, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca-Napoca, Romania
| | - André J A M van der Ven
- Department of Internal Medicine, Radboudumc, Nijmegen, 6500 HB, the Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, 6500 HB, the Netherlands
| |
Collapse
|
99
|
Lei Y, Tao S, Yang Y, Xie F, Xie W. Association between prognostic nutritional index and all-cause mortality and cardiovascular disease mortality in American adults with non-alcoholic fatty liver disease. Front Nutr 2025; 12:1526801. [PMID: 39996009 PMCID: PMC11847695 DOI: 10.3389/fnut.2025.1526801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Background The current research was to investigate the relationship between prognostic nutritional index (PNI) and mortality, with a focus on all-cause and cardiovascular disease (CVD) mortality, for those with non-alcoholic fatty liver disease (NAFLD). Methods Data from 20,142 patients who participated in the National Health and Nutrition Examination Survey (NHANES), which was carried out between 2005 and 2014, were included in this research. To examine the relationship between PNI and both all-cause and cardiovascular mortality, we employed weighted Cox regression models with multiple variables. Kaplan-Meier survival curves were utilized to visualize the survival distribution across different levels of PNI. The non-linear association between PNI and mortality was addressed through penalized spline smoothing. Subgroup analyses were conducted to examine the potential influence of relevant clinical variables on the relationship between PNI and mortality. The precision of PNI in forecasting the outcome of survival was assessed as well using time-dependent receiver operating characteristic curve (ROC) analysis. Results Kaplan-Meier analysis linked higher PNI to significantly reduced all-cause and CVD mortality. Multivariable Cox models demonstrated that increasing PNI consistently lowered mortality risks. With a threshold value of 50.5, the link between PNI and mortality showed a non-linear pattern after adjusting for confounding factors. Subgroup analyses confirmed robust associations, particularly in race, education, BMI, and fibrosis. Time-dependent ROC analysis highlighted the strong predictive performance of PNI across various time points. Conclusion PNI played a significant role as an effective predictor of prognosis in individuals diagnosed with NAFLD.
Collapse
Affiliation(s)
- Yuqing Lei
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Shaohong Tao
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yubo Yang
- Nanhai Hospital of Traditional Chinese Medicine, Jinan University, Foshan, Guangdong, China
| | - Fang Xie
- Department of Liver Disease, Jinling Hospital Affiliated to Medical College of Nanjing University, Nanjing, Jiangsu, China
| | - Weining Xie
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, Guangdong, China
| |
Collapse
|
100
|
Liang M, Xiao X, Chen M, Guo Y, Han W, Min Y, Jiang X, Yu W. Artemisia capillaris Thunb. Water extract alleviates metabolic dysfunction-associated Steatotic liver disease Disease by inhibiting miR-34a-5p to activate Sirt1-mediated hepatic lipid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119030. [PMID: 39515682 DOI: 10.1016/j.jep.2024.119030] [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: 08/03/2024] [Revised: 10/14/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia capillaris Thunb. (ACT) is a plant in the Asteraceae family. Its traditional effects are to clear away dampness and heat, promote gallbladder and reduce jaundice. Traditional Chinese medicine believes that MASLD is a damp-heat syndrome. The group's previous study showed that Artemisia capillaris Thunb. Water Extract (ACTE) has an improved effect on MASLD. AIM OF THE STUDY AND METHODS In order to further understand its mechanism of action, this study established a mouse MASLD model and a HepG2 cell lipid droplet model, combined small RNA sequencing and miRNA transfection experiments, to explore the mechanism of ACTE to improve MASLD by modulating miRNA-targeted mRNA. Non-targeted metabolomics method was used to detect and analyze ACTE. RESULTS This study screened miR-34a-5p and confirmed its target mRNA-Sirtuin 1 (Sirt1). MASLD induced high expression of miR-34a-5p and low expression of Sirt1, and ACE reversed these changes. When overexpressing miR-34a-5p or knocking down Sirt1, the effect of ACE in reducing PO (palmitic acid and oleic acid complex)-induced lipid accumulation in HepG2 cells was attenuated. ACTE reduces the expression of FASN, SCD1, ACC, and SREBP-1c, promotes the expression of CPT-1 and HSL, thereby reducing lipid accumulation. CONCLUSIONS ACTE activates Sirt1 by inhibiting the expression of miR-34a-5p, thereby reducing liver lipid accumulation and improving HFD-induced MASLD. These findings highlight the potential of ACTE in reducing weight, controlling obesity, and improving lipid metabolism disorders.
Collapse
Affiliation(s)
- Meng Liang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Xiao Xiao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Miao Chen
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Yi Guo
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Weiting Han
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Yahong Min
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Provincial Key Laboratory of Animal Pathogenesis and Comparative Medicine, Harbin, 150030, China.
| |
Collapse
|