1
|
Chen Y, Wang Y, Shen T, Wang N, Bai X, Li Q, Fang S, He Z, Sun C, Feng R. Serum metabolic signatures and MetalnFF diagnostic score for mild and moderate metabolic dysfunction-associated steatotic liver disease. J Pharm Biomed Anal 2025; 260:116772. [PMID: 40048991 DOI: 10.1016/j.jpba.2025.116772] [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: 11/30/2024] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 04/01/2025]
Abstract
To explore serum metabolic changes in metabolic dysfunction-associated steatotic liver disease (MASLD) with mild or moderate steatosis and develop a diagnostic index based on liver fat content to differentiate these stages. A total of 149 participants were enrolled from the Nutrition Health Atlas Project in 2019 (Stage 1, n = 92) and 2022 (Stage 2, n = 57). Serum levels of amino acids, free fatty acids (FFAs) and other organic acids were quantified using liquid or gas chromatography-mass spectrometry. The relationships between serum metabolites and magnetic resonance imaging proton density hepatic fat fraction were analyzed and a predictive model fitting fat fraction was constructed in Stage 1 and validated in Stage 2. Patients with moderate MASLD had significantly higher pyruvic acid, 2-ketoglutaric acid, malic acid, 2-hydroxyisocaproic acid and FFA(C14:0) than mild MASLD. Pathway analysis indicated that liver fat accumulation is associated with alterations in amino acid, FFA metabolism and tricarboxylic acid cycle (TCA). The MetalnFF score was developed to discriminate among three groups, achieving an area under the curve (AUC) of 0.956 (95 %CI:0.905, 1.00) for MASLD and 0.857 (95 %CI:0.745, 0.968) for moderate MASLD in Stage 1, and was further validated in Stage 2 with an AUC of 0.986 (95 %CI: 0.951, 1.00) and 0.759 (95 %CI:0.607, 0.921), respectively. In the early stages of MASLD, disrupted amino acid, FFAs metabolism and TCA cycle have occurred. As the disease progresses, metabolic disturbances in pyruvate metabolism become more severe. These findings enhance a deeper understanding of pathogenesis and propose MetalnFF score as a potential diagnostic tool.
Collapse
Affiliation(s)
- Yang Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang, China; Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China; NHC Specialty Laboratory Cooperation Unit of Food Safety Risk Assessment and Standard Development, Heilongjiang, China
| | - Yiran Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang, China; Institute of Cancer Prevention and Treatment, Harbin Medical University, Heilongjiang, China
| | - Tianjiao Shen
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, US
| | - Nan Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang, China
| | - Xiao Bai
- Haxi New Area Community Health Service Center, Nangang District, Heilongjiang, China
| | - Qiyang Li
- Imaging Center, Harbin Medical University Cancer Hospital, Heilongjiang, China
| | - Siyue Fang
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang, China
| | - Zhe He
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang, China; Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China; NHC Specialty Laboratory Cooperation Unit of Food Safety Risk Assessment and Standard Development, Heilongjiang, China.
| | - Rennan Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Heilongjiang, China; Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China; NHC Specialty Laboratory Cooperation Unit of Food Safety Risk Assessment and Standard Development, Heilongjiang, China.
| |
Collapse
|
2
|
Saeed BI, Uthirapathy S, Kubaev A, Ganesan S, Shankhyan A, Gupta S, Joshi KK, Kariem M, Jasim AS, Ahmed JK. Ferroptosis as a key player in the pathogenesis and intervention therapy in liver injury: focusing on drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04115-w. [PMID: 40244448 DOI: 10.1007/s00210-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Globally, drug-induced hepatotoxicity or drug-induced liver injury (DILI) is a serious clinical concern. Knowing the processes and patterns of cell death is essential for finding new therapeutic targets since there are not many alternatives to therapy for severe liver lesions. Excessive lipid peroxidation is a hallmark of ferroptosis, an iron-reliant non-apoptotic cell death linked to various liver pathologies. When iron is pathogenic, concomitant inflammation may exacerbate iron-mediated liver injury, and the hepatocyte necrosis that results is a key element in the fibrogenic response. The idea that dysregulated metabolic pathways and compromised iron homeostasis contribute to the development of liver injury by ferroptosis is being supported by new data. Various ferroptosis-linked genes and pathways have been linked to liver injury, although the molecular processes behind ferroptosis's pathogenicity are not well known. Here, we delve into the features of ferroptosis, the processes governing ferroptosis, and our current knowledge of iron metabolism. We also provide an overview of ferroptosis's involvement in the pathophysiology of liver injury, particularly DILI. Lastly, the therapeutic possibilities of ferroptosis targeting for liver injury management have been provided. Natural products, nanoparticles (NPs), mesenchymal stem cell (MSC), and their exosomes have attracted increasing attention among such therapeutics.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
3
|
Xiao M, Zhou N, Tian Z, Sun C. Endogenous metabolites in metabolic diseases: pathophysiological roles and therapeutic implications. J Nutr 2025:S0022-3166(25)00227-5. [PMID: 40250565 DOI: 10.1016/j.tjnut.2025.04.017] [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: 04/05/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Breakthroughs in metabolomics technology have revealed the direct regulatory role of metabolites in physiology and disease. Recent data have highlighted the bioactive metabolites involved in the etiology and prevention, and treatment of metabolic diseases such as obesity, nonalcoholic fatty liver disease (NAFLD), type 2 diabetes mellitus (T2DM), and atherosclerosis. Numerous studies reveal that endogenous metabolites biosynthesized by host organisms or gut microflora regulate metabolic responses and disorders. Lipids, amino acids, and bile acids (BAs), as endogenous metabolic modulators, regulate energy metabolism, insulin sensitivity, and immune response through multiple pathways, such as insulin signaling cascade, chemical modifications, and metabolite-macromolecule interactions. Furthermore, the gut microbial metabolites short-chain fatty acids (SCFAs), as signaling regulators have a variety of beneficial impacts in regulating energy metabolic homeostasis. In this review, we will summarize information about the roles of bioactive metabolites in the pathogenesis of many metabolic diseases. Furthermore, we discuss the potential value of metabolites in the promising preventive and therapeutic perspectives of human metabolic diseases.
Collapse
Affiliation(s)
- Mengjie Xiao
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, P. R. China 150081; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, 157 Baojian Road, Harbin, P. R. China 150081
| | - Ning Zhou
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, P. R. China 150081; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, 157 Baojian Road, Harbin, P. R. China 150081
| | - Zhen Tian
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, P. R. China 150081; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, 157 Baojian Road, Harbin, P. R. China 150081.
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, P. R. China 150081; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, 157 Baojian Road, Harbin, P. R. China 150081.
| |
Collapse
|
4
|
Menzel JP, Birrer FE, Stroka D, Masoodi M. Skylite: Skyline-Based Lipid Isomer Retention Time Evaluation for Lipidomics in Metabolic Dysfunction-Associated Steatohepatitis. Anal Chem 2025. [PMID: 40226872 DOI: 10.1021/acs.analchem.4c06503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent liver disorder worldwide and can progress to steatohepatitis. Elevated de novo lipogenesis (DNL) is a key contributor to hepatic steatosis. Fatty acid (FA) desaturation produces several unsaturated lipid isomers that are structurally very similar but have diverse biological functions. However, due to their structural similarity, many conventional mass spectrometry approaches cannot detect such metabolic alterations. Thus, we introduce the Skylite (Skyline-based lipid isomer retention time evaluation) workflow using conventional liquid chromatography-mass spectrometry (LC-MS) to identify important isomer features. Retention times of isomeric phosphatidylcholines are compared with the well-characterized human plasma reference standard NIST 1950. Retention time trends correlate well with fixed-charge derivatized FA in liquid chromatography and ozone-induced dissociation mass spectrometry data. The interpretation is supported by double bond diagnostic fragments in LC-MS/MS experiments of epoxidized hydrolyzed fatty acids. We investigate hepatic lipid profiles, focusing on esterified fatty acids in two mouse models of metabolic dysfunction-associated steatohepatitis (MASH). Out of 37 phosphatidylcholine sum compositions, the workflow identifies 123 lipid features. Importantly, CCl4-induced and melanocortin-4 receptor knockout mice on a western diet (WD) have significantly higher levels of mead acid, branched-chain fatty acid, and n-7 PUFA incorporated into phosphatidylcholines. While the MASH mouse liver tissues contain notable amounts of n-7 PUFA, no n-10 PUFA were detected, potentially indicating a unique desaturation pattern. The screening for altered lipid isomer profiles bridges the gap between high-throughput analyses and specialized structure-resolved techniques.
Collapse
Affiliation(s)
- Jan Philipp Menzel
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Fabienne E Birrer
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Deborah Stroka
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Mojgan Masoodi
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| |
Collapse
|
5
|
Gachon F, Bugianesi E, Castelnuovo G, Oster H, Pendergast JS, Montagnese S. Potential bidirectional communication between the liver and the central circadian clock in MASLD. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:15. [PMID: 40225783 PMCID: PMC11981938 DOI: 10.1038/s44324-025-00058-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/25/2025] [Indexed: 04/15/2025]
Abstract
Most aspects of physiology and behaviour fluctuate every 24 h in mammals. These circadian rhythms are orchestrated by an autonomous central clock located in the suprachiasmatic nuclei that coordinates the timing of cellular clocks in tissues throughout the body. The critical role of this circadian system is emphasized by increasing evidence associating disruption of circadian rhythms with diverse pathologies. Accordingly, mounting evidence suggests a bidirectional relationship where disruption of rhythms by circadian misalignment may contribute to liver diseases while liver diseases alter the central clock and circadian rhythms in other tissues. Therefore, liver pathophysiology may broadly impact the circadian system and may provide a mechanistic framework for understanding and targeting metabolic diseases and adjust metabolic setpoints.
Collapse
Affiliation(s)
- Frédéric Gachon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus, Denmark
| | | | | | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | | | - Sara Montagnese
- Department of Medicine, University of Padova, Padova, Italy
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
6
|
Yang Y, Jiao L, Huang Y, Shang H, Li E, Chang H, Cui H, Wan Y. Evaluation of FXR Activity in Pollutants Identified in Sewage Sludge and Subsequent in Vitro and in Vivo Characterization of Metabolic Effects of Triphenyl Phosphate. ENVIRONMENTAL HEALTH PERSPECTIVES 2025; 133:47005. [PMID: 40048564 PMCID: PMC12010937 DOI: 10.1289/ehp15435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common liver disease worldwide, and increasing evidence suggests that exposure to environmental pollutants is associated with the increased incidence of MASLD. The farnesoid X receptor (FXR) plays an important role in the development of MASLD by regulating bile acids (BAs) and lipid metabolism. However, whether FXR-active pollutants are the environmental drivers of MASLD remains unclear. OBJECTIVES This study aimed to determine whether FXR-active pollutants exist in the environment and evaluate their ability to trigger MASLD development in mice. METHODS An FXR protein affinity pull-down assay and nontargeted mass spectrometry (MS) analysis were used to identify environmental FXR ligands in sewage sludge. A homogeneous time-resolved fluorescence coactivator recruitment assay and cell-based dual-luciferase reporter assay were used to determine the FXR activities of the identified pollutants. Targeted analysis of BAs, MS imaging, lipidomic analysis, 16S rRNA sequencing, and quantitative polymerase chain reaction were conducted to assess the ability of FXR-active pollutants to induce metabolic disorders of BAs and lipids and to contribute to MASLD development in C57BL/6N mice. RESULTS We identified 19 compounds in the sewage sludge that had FXR-antagonistic activity, and triphenyl phosphate (TPHP) was the FXR antagonist with the highest efficacy. Mice exposed to either 10 or 50 mg / kg TPHP for 30 d had higher levels of conjugated primary BAs in enterohepatic circulation, and the BA pool showed FXR antagonistic activities. The exposed mice also had greater lipogenesis (more Oil Red O staining and high triglyceride levels) in liver. CONCLUSIONS Nineteen FXR-antagonistic pollutants were identified in sewage sludge. FXR inhibition by the strongest antagonist TPHP may have a role in promoting MASLD development in mice by inducing a positive feedback loop between the FXR and BAs. https://doi.org/10.1289/EHP15435.
Collapse
Affiliation(s)
- Yi Yang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Ling Jiao
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Yixuan Huang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Hailin Shang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Enrui Li
- College of Environmental Science and Engineering, Beijing Forestry University, Beijing, China
| | - Hong Chang
- College of Environmental Science and Engineering, Beijing Forestry University, Beijing, China
| | - Hongyang Cui
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Yi Wan
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| |
Collapse
|
7
|
Jiang J, Chen G, Zhang W, Qin S, Li M, Zhong S, Yang Y, Yang L, Shao M, Wang K, Li Q, Jiang C, Yang J, Wang F, Qiu S, Li X. Pseudonatural Flavonols as Novel Copper Ionophores for NAFLD Intervention via Synergistic Copper Delivery and Flavonoid Activity. J Med Chem 2025; 68:6450-6461. [PMID: 40080431 DOI: 10.1021/acs.jmedchem.4c02927] [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: 03/15/2025]
Abstract
Copper plays crucial roles in fundamental life processes, including proliferation, metabolism, and survival. Copper deficiency is associated with multiple diseases, such as nonalcoholic fatty liver disease (NAFLD) and Wilson's disease. Therapeutic programs targeting copper supply are prospectively employed for disease intervention. Herein, we developed biofriendly copper ionophores (HQFs) by constructing pseudonatural flavonols, which possess flavonoid bioactivity and enhanced copper transport properties. In cell models and mice, we found that HQF-mediated copper delivery synergistically, safely, and efficiently intervened in the development of fatty liver. Mechanistically, NAFLD remission involves fatty acid metabolism, anti-inflammatory processes, and pentose phosphate pathway (PPP) enhancement. Our work is the first to propose the utilization of synergistic copper loading and flavonoid activity for NAFLD intervention, which may inform the clinical management of liver disease.
Collapse
Affiliation(s)
- Jun Jiang
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, P. R. China
| | - Gang Chen
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, P. R. China
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, P. R. China
| | - Weiqin Zhang
- MetWare Biotechnology Co., Ltd., Wuhan 430075, Hubei, P. R. China
| | - Si Qin
- Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, Chongqing, P. R. China
| | - Man Li
- School of Pharmacy, Hubei University of Science and Technology; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, Hubei, P. R. China
| | - Shiyi Zhong
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, P. R. China
| | - Ying Yang
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, P. R. China
| | - Liuxin Yang
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, P. R. China
| | - Muqing Shao
- Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, Chongqing, P. R. China
| | - Kai Wang
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, P. R. China
| | - Qin Li
- Department of Endocrinology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, Chongqing, P. R. China
| | - Chen Jiang
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, P. R. China
| | - Jingfang Yang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, Beijing, P. R. China
| | - Fang Wang
- MetWare Biotechnology Co., Ltd., Wuhan 430075, Hubei, P. R. China
| | - Shuang Qiu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, Heilongjiang, P. R. China
| | - Xiang Li
- School of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, P. R. China
- National Local Joint Engineering Laboratory for Advanced Textile Processing and Clean Production, Wuhan Textile University, Wuhan 430073, Hubei, P. R. China
| |
Collapse
|
8
|
Zarrella S, Miranda MR, Covelli V, Restivo I, Novi S, Pepe G, Tesoriere L, Rodriquez M, Bertamino A, Campiglia P, Tecce MF, Vestuto V. Endoplasmic Reticulum Stress and Its Role in Metabolic Reprogramming of Cancer. Metabolites 2025; 15:221. [PMID: 40278350 DOI: 10.3390/metabo15040221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Endoplasmic reticulum (ER) stress occurs when ER homeostasis is disrupted, leading to the accumulation of misfolded or unfolded proteins. This condition activates the unfolded protein response (UPR), which aims to restore balance or trigger cell death if homeostasis cannot be achieved. In cancer, ER stress plays a key role due to the heightened metabolic demands of tumor cells. This review explores how metabolomics can provide insights into ER stress-related metabolic alterations and their implications for cancer therapy. Methods: A comprehensive literature review was conducted to analyze recent findings on ER stress, metabolomics, and cancer metabolism. Studies examining metabolic profiling of cancer cells under ER stress conditions were selected, with a focus on identifying potential biomarkers and therapeutic targets. Results: Metabolomic studies highlight significant shifts in lipid metabolism, protein synthesis, and oxidative stress management in response to ER stress. These metabolic alterations are crucial for tumor adaptation and survival. Additionally, targeting ER stress-related metabolic pathways has shown potential in preclinical models, suggesting new therapeutic strategies. Conclusions: Understanding the metabolic impact of ER stress in cancer provides valuable opportunities for drug development. Metabolomics-based approaches may help identify novel biomarkers and therapeutic targets, enhancing the effectiveness of antitumor therapies.
Collapse
Affiliation(s)
- Salvatore Zarrella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Verdiana Covelli
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy
| | - Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy
| | - Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy
| | - Manuela Rodriquez
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy
| |
Collapse
|
9
|
Bao Y, Shang X, Hu G, Wang J, Liu C, Lv Q, Che H, Han J, Shao T, Wang G. Stevia rebaudiana root polysaccharide modulates liver metabolism, bile acid, and gut microbiota improving HFD-induced NAFLD: Potential roles of ACSL1 and FADS2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156680. [PMID: 40220428 DOI: 10.1016/j.phymed.2025.156680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/27/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a prevalent metabolic disorder characterized by liver lipid accumulation and insulin resistance. However, effective therapeutic drugs for NAFLD are currently unavailable. Stevia rebaudiana root polysaccharides (SRRP) are inulin-type polysaccharides known for their hypoglycemic properties. Despite this, the effects of SRRP on improving NAFLD and the underlying mechanisms remain poorly understood. PURPOSE This study aims to evaluate the potential of SRRP in alleviating NAFLD and to explore its mechanisms of action. METHODS NAFLD was induced in male C57BL/6 J mice through high-fat diet (HFD) feeding, with subsequent SRRP administration over 8 weeks. Comprehensive assessments included serum biochemical profiling, hepatic histopathological examination, and proinflammatory enzyme activity quantification. Mechanistic investigations employed tripartite analytical approaches: gut microbiota analysis via 16S rRNA sequencing, hepatic metabolomic profiling and bile acid profiling, and validation of transport protein expression through Western blot (WB) techniques. RESULTS SRRP administration significantly alleviated NAFLD through reduced serum lipid concentrations, ameliorated inflammatory responses and oxidative stress, and decreased hepatic lipid deposition mechanistically, SRRP improved the structure of the gut microbiota by enhancing the proliferation of beneficial bacterial, including Lactobacillales and Bifidobacteriales, which subsequently elevated circulating Cholic acid (CA) and Chenodeoxycholic acid (CDCA), and improved hepatic lipid metabolites. Notably, KEGG from metabolomics indicated that the linoleic acid pathway might be associated with the improvement in hepatic lipid metabolite levels by SRRP. In Western blot analysis, SRRP significantly upregulated hepatic ACSL1 and FADS2 in NAFLD mice, demonstrating that the alleviation of NAFLD by SRRP may be achieved through the reduction of hepatic lipid accumulation. CONCLUSIONS SRRP exerts effects on improving NAFLD by modulating the gut microbiota, hepatic metabolites, bile acid levels, and the expression of ACSL1 and FADS2 proteins, providing more scientific evidence and support for the improvement of NAFLD by SRRP.
Collapse
Affiliation(s)
- Yulong Bao
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Xiaolong Shang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Guangdong Hu
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Jiapeng Wang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Chunyan Liu
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Qiuyue Lv
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China; Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wuhu 241002, China
| | - Hui Che
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China; Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wuhu 241002, China
| | - Jun Han
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wuhu 241002, China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Wannan Medical College, Wuhu, 241002, China; Anhui College of Traditional Chinese Medicine, Wuhu 241002, China.
| | - Taili Shao
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China; Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wuhu 241002, China.
| | - Guodong Wang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China; Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, Wuhu 241002, China.
| |
Collapse
|
10
|
Zhang J, Zhang Y, Qiu CX, Zeng W, Ruan Y, Gao Y, Ma W, Wu K, Zhang J, Cui J, Ye C, Liang J, Wang Z. Association of occupational noise exposure and shift work with non-alcoholic fatty liver disease: a cross-sectional study of male workers in the Chinese automobile manufacturing industry. BMJ Open 2025; 15:e085753. [PMID: 40074255 PMCID: PMC11904356 DOI: 10.1136/bmjopen-2024-085753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVE This study aimed to determine the relationship between occupational noise, shift work and non-alcoholic fatty liver disease (NAFLD) in male workers in the automobile manufacturing industry. DESIGN Cross-sectional study. SETTING This study was carried out at the Guangzhou Twelfth People's Hospital using data from April to September 2022. PARTICIPANTS A total of 4672 eligible participants were included in the study. PRIMARY AND SECONDARY OUTCOME MEASURES Diagnosis of NAFLD was made using ultrasound. Noise was detected according to the Measurement of Physical Factors in the Workplace-Part 8: Noise. Environmental noise intensity was assessed using an EDGE personal noise dosimeter manufactured by CASELLA (UK). The working status of workers was investigated by questionnaire. RESULTS The OR of NAFLD was 1.39 (1.03, 1.88) in the cumulative noise exposure (CNE)≥95 group compared with CNE<85 group. Improved risk of NAFLD in workers with shift work compared with those without shift work (OR=1.35, 95% CI: 1.09, 1.68). As stratified analyses showed, the ORs of NAFLD prevalence related to occupational noise and shift work exposure appear to be increased in young workers. When both shift work and noise exposure work are present simultaneously, the synergy index between them was 0.47 (95% CI: 0.25, 0.89). Combined effects analysis revealed that the OR of NAFLD was 2.02 (95% CI: 1.34, 2.99) in CNE≥95 and cumulative length of night shifts work>2920 hours. CONCLUSION Occupational noise exposure may be an independent risk factor for NAFLD. It may synergistically affect disease when combined with night shift work, particularly among younger workers. These findings underscore the importance for companies to prioritise the management and training of younger workers, along with targeted occupational health education initiatives, as crucial measures for reducing the incidence of NAFLD.
Collapse
Affiliation(s)
- Jinwei Zhang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Yuxia Zhang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Cong Xi Qiu
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Wenfeng Zeng
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Yanmei Ruan
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Yunxia Gao
- School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weiyu Ma
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Kangyong Wu
- School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingwen Zhang
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jiaxin Cui
- School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Cuiping Ye
- Department of Preventive Health, Guangzhou Red Cross Hospital, Guangzhou, Guangdong, China
| | - Jiabin Liang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Zhi Wang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Hernandez R, Garcia-Rodriguez NS, Arriaga MA, Perez R, Bala AA, Leandro AC, Diego VP, Almeida M, Parsons JG, Manusov EG, Galan JA. The hepatocellular model of fatty liver disease: from current imaging diagnostics to innovative proteomics technologies. Front Med (Lausanne) 2025; 12:1513598. [PMID: 40109726 PMCID: PMC11919916 DOI: 10.3389/fmed.2025.1513598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is a prevalent chronic liver condition characterized by lipid accumulation and inflammation, often progressing to severe liver damage. We aim to review the pathophysiology, diagnostics, and clinical care of MASLD, and review highlights of advances in proteomic technologies. Recent advances in proteomics technologies have improved the identification of novel biomarkers and therapeutic targets, offering insight into the molecular mechanisms underlying MASLD progression. We focus on the application of mass spectrometry-based proteomics including single cell proteomics, proteogenomics, extracellular vesicle (EV-omics), and exposomics for biomarker discovery, emphasizing the potential of blood-based panels for noninvasive diagnosis and personalized medicine. Future research directions are presented to develop targeted therapies and improve clinical outcomes for MASLD patients.
Collapse
Affiliation(s)
- Renee Hernandez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Natasha S Garcia-Rodriguez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Marco A Arriaga
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Ricardo Perez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Auwal A Bala
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Ana C Leandro
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Vince P Diego
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Marcio Almeida
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Jason G Parsons
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Eron G Manusov
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Jacob A Galan
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| |
Collapse
|
12
|
Bertran L, Capellades J, Abelló S, Richart C. Untargeted lipidomic analysis of metabolic dysfunction-associated steatohepatitis in women with morbid obesity. PLoS One 2025; 20:e0318557. [PMID: 40036208 DOI: 10.1371/journal.pone.0318557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/19/2025] [Indexed: 03/06/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatohepatitis (MASH) represents the severe condition of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Currently, there is a need to identify non-invasive biomarkers for an accurate diagnosis of MASH. Previously, omics studies identified alterations in lipid metabolites involved in MASLD. However, these studies require validation in other cohorts. In this sense, our aim was to perform lipidomics to identify the circulating lipid metabolite profile of MASH. We assessed a liquid chromatography coupled to a mass spectrometer-based untargeted lipidomic assay in serum samples of 216 women with morbid obesity that were stratified according to their hepatic diagnosis into Normal Liver (NL, n = 44), Simple Steatosis (SS, n = 66) and MASH (n = 106). First, we identified a profile of lipid metabolites that are increased in MASLD, composed of ceramides, triacylglycerols (TAG) and some phospholipids. Then, we identified that patients with SS have a characteristic profile of increased levels of ceramides, diacylglycerols DG (36:2) and DG (36:4), some TAG and a few phospholipids such as PC (32:1), PE (38:3), PE (40:6), PI (32:0) and PI (32:1). Later, in MASH patients, we found increased levels of ceramides, deoxycholic acid, a set of TAG, and some phospholipids such as PC, PE, PI and LPI; while we found decreased levels of the DG (36:0). Finally, we have reported a panel of lipid metabolites that might be used to differentiate patients with MASH from SS patients, made up of increased levels of 9-HODE some PC and PE, the LPI (16:0) and decreased levels of DG (36:0). To conclude, our investigation has suggested a lipid metabolite profile associated with MASLD and MASH. Specifically, a set of lipid metabolites seems to be discriminatory in MASH subjects compared to SS individuals. Thus, this panel of lipid metabolites could be used as a non-invasive diagnostic tool.
Collapse
Affiliation(s)
- Laia Bertran
- Department of Medicine and Surgery, Rovira i Virgili University, Tarragona, Spain
| | - Jordi Capellades
- Department of Electronic, Electric and Automatic Engineering, Higher Technical School of Engineering, Rovira i Virgili University, Tarragona, Spain
| | - Sonia Abelló
- Scientific and Technical Service, Rovira i Virgili University, Tarragona, Spain
| | - Cristóbal Richart
- Department of Medicine and Surgery, Rovira i Virgili University, Tarragona, Spain
| |
Collapse
|
13
|
Pan LX, Tian W, Huang ZH, Li JR, Su JY, Wang QY, Fan XH, Zhong JH. Identification of a liver fibrosis and disease progression-related transcriptome signature in non-alcoholic fatty liver disease. Int J Biochem Cell Biol 2025; 180:106751. [PMID: 39909111 DOI: 10.1016/j.biocel.2025.106751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD)-related liver fibrosis is closely associated with long-term outcomes of patients. This study aimed to establish a transcriptome signature to distinguish NAFLD patients with mild or advanced fibrosis and to monitor disease progression. Using least absolute shrinkage selection operator regression, we identified a signature of 11 hub genes by performing differential gene expression analysis in six bulk transcriptome profiles in the Gene Expression Omnibus database from liver fibrosis patients with different etiologies. Patients with NAFLD were classified using the 11-hub gene signature. Integrated analysis of signaling pathway enrichment, gene set enrichment, nearest template prediction, infiltration by hepatic stellate cells (HSCs) and pseudotime trajectories was performed on three bulk and one single-cell transcriptomes from NAFLD patients. Molecular features were compared between high-risk and low-risk groups, and associations were explored between hub gene signature expression and activation of HSCs. It was found that the high-risk group was characterized by advanced fibrosis stage, elevated risk for hepatocellular carcinoma, more significant infiltration by activated HSCs, as well as enrichment in signaling pathways related to fibrogenesis and NAFLD progression. Moreover, the 11-hub gene signature at the single-cell transcriptome level correlated with HSCs activation. In vitro experiments were conducted to evaluate the expression levels of hub genes, and IL6 was found to be up-regulated in activated LX-2 cells showing lipid accumulation. Our findings suggest that the 11-hub gene signature can help identify fibrosis stage in patients with NAFLD and detect disease progression. We also suggest that the role of IL6 in HSC activation deserves more investigation in the context of NAFLD.
Collapse
Affiliation(s)
- Li-Xin Pan
- Department of Microbiology, School of Preclinical Medicine, Center for Genomics and Personalized Medicine, Guangxi Medical University, Nanning 530021, China; Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Wei Tian
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China; School of Life Sciences, Guangxi Medical University, Nanning 530021, China
| | - Zhi-Hao Huang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Jian-Rong Li
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Jia-Yong Su
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Qiu-Yan Wang
- School of Life Sciences, Guangxi Medical University, Nanning 530021, China.
| | - Xiao-Hui Fan
- Department of Microbiology, School of Preclinical Medicine, Center for Genomics and Personalized Medicine, Guangxi Medical University, Nanning 530021, China.
| | - Jian-Hong Zhong
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning 530021, China; Key Laboratory of Early Prevention and Treatment for Regional High, Frequency Tumors (Guangxi Medical University), Ministry of Education, Nanning 530021, China; Guangxi Key Laboratory of Early Prevention and Treatment for Regional High, Frequency Tumors, Nanning 530021, China.
| |
Collapse
|
14
|
Zhou Y, Liu J. The role of lipoprotein sulfatides in MASLD fibrosis transition: A new frontier in hepatic immunomodulation. Metabol Open 2025; 25:100335. [PMID: 40176831 PMCID: PMC11963198 DOI: 10.1016/j.metop.2024.100335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 04/04/2025] Open
Affiliation(s)
- Yifan Zhou
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junli Liu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
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
|
16
|
Zhang S, Wu J, Wang L, Zhang C, Zhang Y, Feng Y. Exploring the hepatic-ophthalmic axis through immune modulation and cellular dynamics in diabetic retinopathy and non-alcoholic fatty liver disease. Hum Genomics 2025; 19:19. [PMID: 40011971 DOI: 10.1186/s40246-025-00730-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: 10/04/2024] [Accepted: 02/13/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Dysfunctions within the liver system are intricately linked to the progression of diabetic retinopathy (DR) and non-alcoholic fatty liver disease (NAFLD). This study leverages systematic analysis to elucidate the complex cross-talk and communication pathways among diverse cell populations implicated in the pathogenesis of DR and NAFLD. METHODS Single-cell RNA sequencing data for proliferative diabetic retinopathy (PDR) and NAFLD were retrieved from the Gene Expression Omnibus (GEO) database. Differential gene expression analysis was conducted and followed by pseudo-time analysis to delineate dynamic changes in core cells and differentially expressed genes (DEGs). CellChat was employed to predict intercellular communication and signaling pathways. Additionally, gene set enrichment and variation analyses (GSEA and GSVA) were performed to uncover key functional enrichments. RESULTS Our comparative analysis of the two datasets focused on T cells, macrophages and endothelial cells, revealing SYNE2 as a notable DEG. Notably, common genes including PYHIN1, SLC38A1, ETS1 (T cells), PPFIBP1, LIFR, HSPG2 (endothelial cells), and MSR1 (macrophages), emerged among the top 50 DEGs across these cell types. The CD45 signaling pathway was pivotal for T cells and macrophages, exerting profound effects on other cells in both PDR and NAFLD. Moreover, GSEA and GSVA underscored their involvement in cellular communication, immune modulation, energy metabolism, mitotic processes. CONCLUSION The comprehensive investigation of T cells, macrophages, endothelial cells, and the CD45 signaling pathway advances our understanding of the intricate biological processes underpinning DR and NAFLD. This research underscores the imperative of exploring immune-related cell interactions, shedding light on novel therapeutic avenues in these disease contexts.
Collapse
Affiliation(s)
- Shuyan Zhang
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jiajun Wu
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Leilei Wang
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yinjian Zhang
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
17
|
Dakal TC, Xiao F, Bhusal CK, Sabapathy PC, Segal R, Chen J, Bai X. Lipids dysregulation in diseases: core concepts, targets and treatment strategies. Lipids Health Dis 2025; 24:61. [PMID: 39984909 PMCID: PMC11843775 DOI: 10.1186/s12944-024-02425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/30/2024] [Indexed: 02/23/2025] Open
Abstract
Lipid metabolism is a well-regulated process essential for maintaining cellular functions and energy homeostasis. Dysregulation of lipid metabolism is associated with various conditions, including cardiovascular diseases, neurodegenerative disorders, and metabolic syndromes. This review explores the mechanisms underlying lipid metabolism, emphasizing the roles of key lipid species such as triglycerides, phospholipids, sphingolipids, and sterols in cellular physiology and pathophysiology. It also examines the genetic and environmental factors contributing to lipid dysregulation and the challenges of diagnosing and managing lipid-related disorders. Recent advancements in lipid-lowering therapies, including PCSK9 inhibitors, ezetimibe, bempedoic acid, and olpasiran, provide promising treatment options. However, these advancements are accompanied by challenges related to cost, accessibility, and patient adherence. The review highlights the need for personalized medicine approaches to address the interplay between genetics and environmental factors in lipid metabolism. As lipidomics and advanced diagnostic tools continue to progress, a deeper understanding of lipid-related disorders could pave the way for more effective therapeutic strategies.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia, University, Udaipur, 313001, India
| | - Feng Xiao
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Chandra Kanta Bhusal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | | | - Rakesh Segal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Juan Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Xiaodong Bai
- Department of Plastic Surgery, Southern University of Science and Technology Hospital, Shenzhen, 518055, China.
| |
Collapse
|
18
|
Bourganou MV, Chondrogianni ME, Kyrou I, Flessa CM, Chatzigeorgiou A, Oikonomou E, Lambadiari V, Randeva HS, Kassi E. Unraveling Metabolic Dysfunction-Associated Steatotic Liver Disease Through the Use of Omics Technologies. Int J Mol Sci 2025; 26:1589. [PMID: 40004054 PMCID: PMC11855544 DOI: 10.3390/ijms26041589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), now referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is the most prevalent liver disorder globally, linked to obesity, type 2 diabetes, and cardiovascular risk. Understanding its potential progression from simple steatosis to cirrhosis and hepatocellular carcinoma (HCC) is crucial for patient management and treatment strategies. The disease's complexity requires innovative approaches for early detection and personalized care. Omics technologies-such as genomics, transcriptomics, proteomics, metabolomics, and exposomics-are revolutionizing the study of MASLD. These high-throughput techniques allow for a deeper exploration of the molecular mechanisms driving disease progression. Genomics can identify genetic predispositions, whilst transcriptomics and proteomics reveal changes in gene expression and protein profiles during disease evolution. Metabolomics offers insights into the metabolic alterations associated with MASLD, while exposomics links environmental exposures to MASLD progression and pathology. By integrating data from various omics platforms, researchers can map out the intricate biochemical pathways involved in liver disease progression. This review discusses the roles of omics technologies in enhancing the understanding of disease progression and highlights potential diagnostic and therapeutic targets within the MASLD spectrum, emphasizing the need for non-invasive tools in disease staging and treatment development.
Collapse
Affiliation(s)
- Maria V. Bourganou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kyrou
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 IGB, UK
| | - Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vaia Lambadiari
- 2nd Department of Internal-Medicine, Diabetes Centre, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
19
|
Wang X, Cheng L, Liu A, Liu L, Gong L, Shen G. Metabolomics approach reveals key plasma biomarkers in multiple myeloma for diagnosis, staging, and prognosis. J Transl Med 2025; 23:163. [PMID: 39915820 PMCID: PMC11800462 DOI: 10.1186/s12967-024-05848-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/30/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Multiple myeloma (MM) is the most aggressive and prevalent primary malignant tumor within the blood system, and can be classified into grades RISS-I, II, and III. High-grade tumors are associated with decreased survival rates and increased recurrence rates. To better understand metabolic disorders and expand the potential targets for MM, we conducted large-scale untargeted metabolomics on plasma samples from MM patients and healthy controls (HC). METHODS Our study included 33 HC, 38 newly diagnosed MM patients (NDMM) categorized into three RISS grades (grade I: n = 5; grade II: n = 19; grade III: n = 8), and 92 MM patients post-targeted therapy with bortezomib-based regimens. Simultaneously, MM cell lines were employed for validation studies. Metabolites were analyzed and identified using ultra high liquid chromatography coupled with Q Orbitrap mass spectrometry (UPLC-HRMS), followed by verification through a self-built database. RESULTS Compared with HC participants, a total of 70 metabolites were identified as undergoing significant changes in NDMM. These metabolites were significantly enriched in citrate cycle, choline metabolism, glycerophospholipid metabolism, and sphingolipid metabolism, etc. Notably, a panel of circulating plasma metabolite biomarkers, including lactic acid and leucine, has emerged not only as diagnostic indicators but also as valuable tools for tumor surveillance, aiding in the assessment of disease stage and prognostic evaluation. Moreover, 14 differential metabolites were identified in both MM cell lines and MM patients. Among these, intracellular levels of lactate and leucine significantly decreased in vitro, aligning with the plasma results. CONCLUSION Our findings on key metabolites and metabolic pathways provide novel insights into the exploration of diagnostic and therapeutic targets for MM. A prospective study is essential to validate these discoveries for future MM patient care.
Collapse
Affiliation(s)
- Xiaoxue Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Longhao Cheng
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Capital Medical University, No. 2 YingHua Road, Beijing, 100029, China
| | - Aijun Liu
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Lihong Liu
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Capital Medical University, No. 2 YingHua Road, Beijing, 100029, China
| | - Lili Gong
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Capital Medical University, No. 2 YingHua Road, Beijing, 100029, China.
| | - Guolin Shen
- Institute of Chemicals Safety, Chinese Academy of Inspection and Quarantine, No. 11 Rong Hua Middle Road, Economic-Technological Development Area, Beijing, 100176, China.
| |
Collapse
|
20
|
Martín-Grau M, Casanova P, Moreno-Morcillo L, Morales JM, Marrachelli VG, Monleón D. Microbiota Co-Metabolism Alterations Precede Changes in the Host Metabolism in the Early Stages of Diet-Induced MASLD in Wistar Rats. Int J Mol Sci 2025; 26:1288. [PMID: 39941056 PMCID: PMC11818068 DOI: 10.3390/ijms26031288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD) affects around 30% of the global population. The sexual dimorphism and gut microbiota play an important role in the early development of MASLD. The main objective of this research was to investigate metabolic changes during the early subclinical MASLD progression, for identifying the sequence of events and evaluating the impact of sexual dimorphism and the microbiota on the initial stages of MASLD development. Male and female Wistar rats 18 weeks old were randomly divided into different groups and fed a chow diet or a 45% high-fat diet for 21 weeks. Every three weeks, samples of serum, urine, and faeces were collected and studied by metabolomics. Furthermore, the liver was analysed at the endpoint. In addition, the gut microbiota was analysed from faecal samples over time using 16S rRNA gene-targeted group-specific primers. Our results revealed that three weeks on an HFD reduced the bacterial diversity in the faecal microbiota of Wistar rats, accompanied by changes in the faecal and urine metabolome. The HFD-induced alterations in microbiota-related co-metabolites in the liver, blood, urine, and faeces indicate a significant role of host-microbiota co-metabolism changes in the early stages of MASLD. In this study, we provide a comprehensive longitudinal analysis, detailing the sequence of events in the early development of MASLD. Our findings suggest that alterations in the gut microbiota diversity and co-metabolism occur before changes in host metabolism in the early onset of liver steatosis, a subclinical phase of MASLD.
Collapse
Affiliation(s)
- María Martín-Grau
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Pilar Casanova
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Laura Moreno-Morcillo
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - José Manuel Morales
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Vannina G. Marrachelli
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
- Departament de Fisiologia, Universitat de València, 46010 Valencia, Spain
| | - Daniel Monleón
- Departament de Patologia, Universitat de València, 46010 Valencia, Spain; (P.C.); (L.M.-M.); (J.M.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| |
Collapse
|
21
|
Zhou Y, Wang M, Wang Z, Qiu J, Wang Y, Li J, Dong F, Huang X, Zhao J, Xu T. Polysaccharides from hawthorn fruit alleviate high-fat diet-induced NAFLD in mice by improving gut microbiota dysbiosis and hepatic metabolic disorder. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156458. [PMID: 39919328 DOI: 10.1016/j.phymed.2025.156458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/20/2025] [Accepted: 02/01/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Hawthorn fruit, renowned as both a functional food and herbal medicine with lipid-lowering effects, is abundant in polysaccharides. However, there is limited research on the effects and mechanisms of hawthorn fruit polysaccharides (HP) in addressing non-alcoholic fatty liver disease (NAFLD). PURPOSE This study aims to investigate the effects of HP on NAFLD both in vivo and in vitro, and to elucidate the underlying mechanisms by which HP exerts its anti-NAFLD activity. METHODS NAFLD mice induced by a high-fat diet were employed as the in vivo model, while oleate/palmitate-induced HepG2 cells served as the in vitro model. H&E and Oil Red O staining were employed to examine fat accumulation in hepatocytes. Serum aminotransferase (ALT), aspartate aminotransferase (AST), hepatic malondialdehyde (MDA), superoxide dismutase (SOD), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6) were measured using corresponding ELISA kits. Hepatic metabolomics analysis based on UHPLC-QTOF/MS was utilized to examine the role of HP in improving hepatic metabolic disorders. 16S rRNA sequencing was conducted to explore the effect of HP in alleviating gut microbiota dysbiosis. GC-MS was applied to detect short-chain fatty acids (SCFAs) to clarify the impact of HP in NAFLD mice. RESULTS HP significantly inhibited weight gain and hepatic fat accumulation in NAFLD mice. The reduction in serum ALT and AST levels indicated that HP mitigated liver function damage, while the decreased MDA levels and increased SOD activity suggested that HP alleviated hepatic oxidative stress. Furthermore, HP diminished the release of inflammatory cytokines such as IL-1β and IL-6 in the liver. HP significantly regulated metabolic pathways related to amino acids, lipids, and vitamins. Key metabolites such as l-tyrosine, urocanic acid, undecanedioic acid, oleamide, vitamin A, and vitamin B7 were restored to near-normal levels under the regulatory effects of HP. Gut microbiota dysbiosis in NAFLD mice was also ameliorated by HP, with genera such as unclassified_f__Lachnospiraceae and Dubosiella being notably affected. Correlation analysis indicated a significant correlation between the regulatory effects of HP on liver metabolism and gut microbiota. Additionally, HP showed no effect in vitro but increased acetic acid level in the gut of NAFLD mice. CONCLUSIONS These findings demonstrate that HP exhibits its anti-NAFLD effects, including alleviating lipid accumulation, liver dysfunction, oxidative stress, and inflammation. Mechanistically, HP primarily improves gut microbiota dysbiosis, thereby elevating intestinal SCFA levels and restoring hepatic metabolic disorders in NAFLD mice.
Collapse
Affiliation(s)
- Yuan Zhou
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mengyao Wang
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zichuan Wang
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junjie Qiu
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yichen Wang
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianbiao Li
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Fengqi Dong
- Department of Pharmacy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Xianzhe Huang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing 314400, China
| | - Jiahui Zhao
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Tengfei Xu
- Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
22
|
Shen D, Sha L, Yang L, Gu X. Identification of multiple complications as independent risk factors associated with 1-, 3-, and 5-year mortality in hepatitis B-associated cirrhosis patients. BMC Infect Dis 2025; 25:151. [PMID: 39891059 PMCID: PMC11786570 DOI: 10.1186/s12879-025-10566-6] [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: 10/02/2024] [Accepted: 01/28/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Hepatitis B-associated cirrhosis (HBC) is associated with severe complications and adverse clinical outcomes. This study aimed to develop and validate a predictive model for the occurrence of multiple complications (three or more) in patients with HBC and to explore the effects of multiple complications on HBC prognosis. METHODS In this retrospective cohort study, data from 121 HBC patients treated at Nanjing Second Hospital from February 2009 to November 2019 were analysed. The maximum follow-up period was 10.75 years, with a median of 5.75 years. Eight machine learning techniques were employed to construct predictive models, including C5.0, linear discriminant analysis (LDA), least absolute shrinkage and selection operator (LASSO), k-nearest neighbour (KNN), gradient boosting decision tree (GBDT), support vector machine (SVM), generalised linear model (GLM) and naive Bayes (NB), utilising variables such as medical history, demographics, clinical signs, and laboratory test results. Model performance was evaluated via receiver operating characteristic (ROC) curve analysis, residual analysis, calibration curve analysis, and decision curve analysis (DCA). The influence of multiple complications on HBC survival time was assessed via Kaplan‒Meier curve analysis. Furthermore, LASSO and univariable and multivariable Cox regression analyses were conducted to identify independent prognostic factors for overall survival (OS) in patients with HBC, followed by ROC, C-index, calibration curve, and DCA curve analyses of the constructed prognostic nomogram model. This study utilized bootstrap resampling for internal validation and employed the Medical Information Mart for Intensive Care IV (MIMIC-IV) database for external validation. RESULTS The GBDT model exhibited the highest area under the curve (AUC) and emerged as the optimal model for predicting the occurrence of multiple complications. The key predictive factors included posthospitalisation fever (PHF), body mass index (BMI), retinol binding protein (RBP), total bilirubin (TB) levels, and eosinophils (EOS). Kaplan-Meier analysis revealed that patients with multiple complications had significantly worse OS than those with fewer complications. Additionally, multivariable Cox regression analysis, informed by least absolute shrinkage and LASSO selection, identified hepatocellular carcinoma (HCC), multiple complications, and lactate dehydrogenase (LDH) levels as independent prognostic factors for OS. The prognostic model demonstrated 1-year, 3-year, and 5-year OS ROC AUCs of 0.802, 0.793, and 0.817, respectively. For the internal validation cohort, the corresponding AUC values were 0.797, 0.832, and 0.835. In contrast, the external validation cohort yielded a 1-year ROC AUC of 0.707. Calibration curves indicated good consistency of the model, and DCA demonstrated the model's clinical utility, showing high net benefits within certain threshold ranges. Compared with the univariable models, the multivariable ROC curves indicated higher AUC values for this prognostic model, and the model also possessed the best c-index. CONCLUSION The GBDT prediction model provides a reliable tool for the early identification of high-risk HBC patients prone to developing multiple complications. The concurrent occurrence of multiple complications is an independent prognostic factor for OS in patients with HBC. The constructed prognostic model demonstrated remarkable predictive performance and clinical applicability, indicating its crucial role in enhancing patient outcomes through timely and targeted interventions.
Collapse
Affiliation(s)
- Duo Shen
- Department of Gastroenterology, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Ling Sha
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated to Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Ling Yang
- Department of Central Laboratory, Jurong Hospital Affiliated to Jiangsu University, 66 Ersheng Road, Jurong, Zhenjiang, Jiangsu, 212400, China
| | - Xuefeng Gu
- Department of Central Laboratory, Jurong Hospital Affiliated to Jiangsu University, 66 Ersheng Road, Jurong, Zhenjiang, Jiangsu, 212400, China.
- Department of Infectious Diseases, Jurong Hospital Affiliated to Jiangsu University, 66 Ersheng Road, Jurong, Zhenjiang, Jiangsu, 212400, China.
| |
Collapse
|
23
|
Singh P, Singh R, Pasricha C, Kumari P. Navigating liver health with metabolomics: A comprehensive review. Clin Chim Acta 2025; 566:120038. [PMID: 39536895 DOI: 10.1016/j.cca.2024.120038] [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: 09/21/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the main cause of chronic liver disease worldwide, affecting one-fourth of the world's population. With more than half of the world's population, the Asia-Pacific region contributed 62.6 % of liver-related fatal incidents in 2015. Currently, liver imaging techniques such as computed tomography (CT), nuclear magnetic resonance (NMR) spectroscopy, and ultrasound are non-invasive imaging methods to diagnose the disease. A liver biopsy is the gold standard test for establishing the definite diagnosis of non-alcoholic steatohepatitis (NASH). However, there are still significant problems with sample variability and the procedure's invasiveness. Numerous studies have indicated various non-invasive biomarkers for both fibrosis and steatosis to counter the invasiveness of diagnostic procedures. Metabolomics could be a promising method for detecting early liver diseases, investigating pathophysiology, and developing drugs. Metabolomics, when utilized with other omics technologies, can result in a deeper understanding of biological systems. Metabolomics has emerged as a prominent research topic, offering extensive opportunities to investigate biomarkers for liver diseases that are both sensitive and specific. In this review, we have described the recent studies involving the use of a metabolomics approach in the diagnosis of liver diseases, which would be beneficial for the early detection and treatment of liver diseases.
Collapse
Affiliation(s)
- Preetpal Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Ravinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Chirag Pasricha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pratima Kumari
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
24
|
Zheng Z, Yang S, Dai W, Sun Y, Wang J, Zhang X, Zheng Y, Kong J. Role of plasma metabolome in mediating the effect of plasma lipidome on NAFLD: a Mendelian randomization study. Front Endocrinol (Lausanne) 2025; 15:1436827. [PMID: 39916753 PMCID: PMC11798786 DOI: 10.3389/fendo.2024.1436827] [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: 05/22/2024] [Accepted: 12/23/2024] [Indexed: 02/09/2025] Open
Abstract
Background This study explored the causal connection among the plasma lipidome, nonalcoholic fatty liver disease (NAFLD), and potential metabolome mediators through Mendelian randomization (MR). Methods We obtained summary statistics for 179 plasma lipidome traits (N = 7,174), 1,400 plasma metabolome traits (N = 8,299), and one NAFLD trait from publicly available genome-wide association studies. A two-sample MR analysis was conducted to infer causality. Additionally, multiple sensitivity analyses were conducted to assess the heterogeneity, horizontal pleiotropy, and robustness of the MR findings. MetaboAnalyst 6.0 was used for the pathway analysis of the identified lipids and metabolites. Furthermore, we used mediation analysis to assess whether the effect of plasma lipidome on NAFLD was mediated by plasma metabolome. Results The MR analysis predicted a genetically determined causal relationship between plasma lipidomes and NAFLD. No compelling proof was found that genetically predicted NAFLD influenced the risk of the five plasma lipidomes mentioned earlier. Based on established causal relationships between lipids and metabolites, we identified that eight metabolic pathways are closely associated with NAFLD. Our mediation analysis revealed six mediation relationships, indicating the causal pathway from plasma lipids to NAFLD mediated by five specific metabolites. No potential pleiotropy was found in the sensitivity analysis. Conclusions In summary, our study identified causal relationships between plasma lipidomes, plasma metabolomes, and NAFLD. Certainly, the impact of plasma lipidomes on NAFLD is not limited to plasma metabolomes, indicating a need to further investigate into other possible mediators. These identified factors may become new biomarkers of the NAFLD contributing to its prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Zhuyuan Zheng
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shaojie Yang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wanlin Dai
- Innovation Institute of China Medical University, Shenyang, Liaoning, China
| | - Yang Sun
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingnan Wang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaolin Zhang
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yiming Zheng
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Kong
- Biliary Surgery (2nd General) Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
25
|
Zhang X, Zheng MH, Liu D, Lin Y, Song SJ, Chu ESH, Liu D, Singh S, Berman M, Lau HCH, Gou H, Wong GLH, Zhang N, Yuan HY, Loomba R, Wong VWS, Yu J. A blood-based biomarker panel for non-invasive diagnosis of metabolic dysfunction-associated steatohepatitis. Cell Metab 2025; 37:59-68.e3. [PMID: 39500327 DOI: 10.1016/j.cmet.2024.10.008] [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: 02/24/2024] [Revised: 08/08/2024] [Accepted: 10/07/2024] [Indexed: 01/11/2025]
Abstract
The current diagnosis of metabolic dysfunction-associated steatotic liver disease (MASLD) and its severe form, metabolic dysfunction-associated steatohepatitis (MASH), is suboptimal. Here, we recruited 700 individuals, including 184 from Hong Kong as a discovery cohort and 516 from San Diego, Wenzhou, and Hong Kong as three validation cohorts. A panel of 3 parameters (C-X-C motif chemokine ligand 10 [CXCL10], cytokeratin 18 fragments M30 [CK-18], and adjusted body mass index [BMI]) was formulated (termed N3-MASH), which discriminated patients with MASLD from healthy controls with an area under the receiver operating characteristic (AUROC) of 0.954. Among patients with MASLD, N3-MASH could identify patients with MASH with an AUROC of 0.823, achieving 90.0% specificity, 62.9% sensitivity, and 88.6% positive predictive value. The diagnostic performance of N3-MASH was confirmed in three validation cohorts with AUROC of 0.802, 0.805, and 0.823, respectively. Additionally, N3-MASH identifies patients with MASH improvement with an AUROC of 0.857. In summary, we developed a robust blood-based panel for the non-invasive diagnosis of MASH, which might help clinicians reduce unnecessary liver biopsies.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dehua Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Yufeng Lin
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Sherlot Juan Song
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Eagle Siu-Hong Chu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Dabin Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Seema Singh
- MASLD Research Center, Division of Gastroenterology, University of California at San Diego, La Jolla, San Diego, CA, USA
| | - Michael Berman
- MASLD Research Center, Division of Gastroenterology, University of California at San Diego, La Jolla, San Diego, CA, USA
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Hongyan Gou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Grace Lai-Hung Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China
| | - Ni Zhang
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hai-Yang Yuan
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology, University of California at San Diego, La Jolla, San Diego, CA, USA.
| | - Vincent Wai-Sun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China.
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong SAR, China.
| |
Collapse
|
26
|
Zhang X, Zheng Y, Yang J, Yang Y, He Q, Xu M, Long F, Yang Y. Abnormal ac4C modification in metabolic dysfunction associated steatotic liver cells. Sci Rep 2025; 15:1013. [PMID: 39762452 PMCID: PMC11704021 DOI: 10.1038/s41598-024-84564-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
The pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) remains unclear due to the complexity of its etiology. The emerging field of the epitranscriptome has shown significant promise in advancing the understanding of disease pathogenesis and developing new therapeutic approaches. Recent research has demonstrated that N4-acetylcytosine (ac4C), an RNA modification within the epitranscriptome, is implicated in progression of various diseases. However, the role of ac4C modification in MASLD remains unexplored. Herein, we performed acRIP-ac4c-seq and RNA-seq analysis in free fatty acids-induced MASLD model cells, identifying 2128 differentially acetylated ac4C sites, with 1031 hyperacetylated and 1097 hypoacetylated peaks in MASLD model cells. Functional enrichments analysis showed that ac4C differentially modified genes were significantly involved in processes related to MASLD, such as nuclear transport and MAP kinase (MAPK) signaling pathway. We also identified 341 differentially expressed genes (DEGs), including 61 lncRNAs and 280 mRNAs, between control and MASLD model cells. Bioinformatics analysis showed that DEGs were significantly enriched in long-chain fatty acid biosynthetic process. Notably, 118 genes exhibited significant changes in both ac4C modification and expression levels in MASLD model cells. Among these proteins, JUN, caveolin-1 (CAV1), fatty acid synthase (FASN), and heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) were identified as core proteins through protein-protein interaction (PPI) network analysis using cytoscape software. Collectively, our findings establish a positive correlation between ac4C modification and the pathogenesis of MASLD and suggest that ac4C modification may serve as a therapeutic target for MASLD.
Collapse
Affiliation(s)
- Xiqian Zhang
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Yaxian Zheng
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Jing Yang
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Yan Yang
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Qin He
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Min Xu
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Women's and Children's Hospital, Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610041, China.
| | - Yujie Yang
- Department of Pharmacy, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, 610014, China.
| |
Collapse
|
27
|
Ahmadizar F, Younossi ZM. Exploring Biomarkers in Nonalcoholic Fatty Liver Disease Among Individuals With Type 2 Diabetes Mellitus. J Clin Gastroenterol 2025; 59:36-46. [PMID: 39352015 PMCID: PMC11630663 DOI: 10.1097/mcg.0000000000002079] [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: 03/27/2024] [Accepted: 09/02/2024] [Indexed: 10/03/2024]
Abstract
Integrating biomarkers into a comprehensive strategy is crucial for precise patient management, especially considering the significant healthcare costs associated with diseases. Current studies emphasize the urgent need for a paradigm shift in conceptualizing nonalcoholic fatty liver disease (NAFLD), now renamed metabolic dysfunction-associated steatotic liver disease (MASLD). Biomarkers are emerging as indispensable tools for accurate diagnosis, risk stratification, and monitoring disease progression. This review classifies biomarkers into conventional and novel categories, such as lipids, insulin resistance, hepatic function, and cutting-edge imaging/omics, and evaluates their potential to transform the approach to MASLD among individuals with type 2 diabetes mellitus (T2D). It focuses on the critical role of biomarkers in early MASLD detection, enhancing predictive accuracy, and discerning responses to interventions (pharmacological or lifestyle modifications). Amid this discussion, the complexities of the relationship between T2D and MASLD are explored, considering factors like age, gender, genetics, ethnicity, and socioeconomic background. Biomarkers enhance the effectiveness of interventions and support global initiatives to reduce the burden of MASLD, thereby improving public health outcomes. This review recognizes the promising potential of biomarkers for diagnostic precision while candidly addressing the challenges in implementing these advancements in clinical practice. The transformative role of biomarkers emerges as a central theme, promising to reshape our understanding of disease trajectories, prognosis, and the customization of personalized therapeutic strategies for improved patient outcomes. From a future perspective, identifying early-stage biomarkers, understanding environmental impact through exposomes, and applying a multiomics approach may reveal additional insight into MASLD development.
Collapse
Affiliation(s)
- Fariba Ahmadizar
- Data Science and Biostatistics Department, Julius Global Health, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Beatty Liver and Obesity Research Program Center for Liver Diseases, Inova Health System, Falls Church, VA
| | - Zobair M. Younossi
- The Global NASH Council, Center for Outcomes Research in Liver Disease, Washington, DC
| |
Collapse
|
28
|
Takeda H, Izumi Y, Bamba T. Quantitative Lipidomics of Biological Samples Using Supercritical Fluid Chromatography Mass Spectrometry. Methods Mol Biol 2025; 2891:131-152. [PMID: 39812980 DOI: 10.1007/978-1-0716-4334-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Lipidomics has attracted attention in the discovery of unknown biomolecules and for capturing the changes in metabolism caused by genetic and environmental factors in an unbiased manner. However, obtaining reliable lipidomics data, including structural diversity and quantification data, is still challenging. Supercritical fluid chromatography (SFC) is a suitable technique for separating lipid molecules with high throughput and separation efficiency. Here, we describe a quantitative lipidomics method using SFC coupled with mass spectrometry. This technique is suitable for characterizing the structural diversity of lipids (e.g., phospholipids, sphingolipids, glycolipids, and glycerolipids) with high quantitative accuracy to understand their biological functions.
Collapse
Affiliation(s)
- Hiroaki Takeda
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
- RIKEN Center for Brain Science, Saitama, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
29
|
Fan YQ, Wang H, Wang PP, Shi ZY, Wang Y, Xu J. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio as a predictive indicator of CKD risk in NAFLD patients: NHANES 2017-2020. Front Nutr 2024; 11:1501494. [PMID: 39777076 PMCID: PMC11703712 DOI: 10.3389/fnut.2024.1501494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD) are both closely related to dyslipidemia. However, the relationship between dyslipidemia in patients with NAFLD and CKD is not yet clear. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) is an innovative and comprehensive lipid index. The purpose of this study was to investigate the correlation between NHHR and CKD risk in NAFLD patients with or without fibrosis. Methods This study used data from the National Health and Nutrition Examination Survey (NHANES) from 2017 to 2020 for analysis, including a total of 4,041 subjects diagnosed with NAFLD. Among the NAFLD subjects, 3,315 individuals without liver fibrosis and 726 individuals with fibrosis. Weighted multivariate linear regression, weighted logistic regression, restricted cubic spline (RCS) curves, and subgroup analysis were used to evaluate the correlation between NHHR and CKD in patients with NAFLD. Results Our findings indicate that in NAFLD subjects without liver fibrosis, the highest tertile of NHHR, as compared to the lowest tertile, was inversely related to glomerular filtration rate (eGFR) (β: -2.14, 95% CI: -3.97, -0.32, p < 0.05) and positively related to CKD (OR: 1.67, 95% CI: 1.12, 2.49, p < 0.05). No significant associations were observed between NHHR and eGFR, urinary albumin to creatinine ratio (ACR) in NAFLD subjects with liver fibrosis. The RCS revealed a linear relationship between NHHR and ACR, CKD in NAFLD subjects without liver fibrosis, while a U-shaped relationship was observed between NHHR and ACR, CKD in NAFLD subjects with liver fibrosis. Conclusion In patients with non-fibrotic NAFLD, a significantly elevated NHHR is closely associated with an increased risk of CKD and shows a linear relationship with CKD. In patients with fibrotic NAFLD, NHHR shows a U-shaped relationship with CKD. LD, Our findings underscore the practical utility of NHHR as a biomarker for early risk stratification of CKD in patients with NAFLD.
Collapse
Affiliation(s)
- Yong-Qiang Fan
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hao Wang
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Pei-Pei Wang
- Department of Respiratory, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhi-Yong Shi
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan Wang
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jun Xu
- Liver Transplantation Center, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
30
|
Tian P, Zhao L, Zhang G, Chen S, Zhang W, Ou M, Sun Y, Chen Y. A global lipid map of severe fever with thrombocytopenia syndrome virus infection reveals glycerophospholipids as novel prognosis biomarkers. mBio 2024; 15:e0262824. [PMID: 39535228 PMCID: PMC11633121 DOI: 10.1128/mbio.02628-24] [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: 08/26/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a rapidly progressing infectious disease caused by a novel bunyavirus characterized by high fever, thrombocytopenia, and multiple organ damage. While lipids play an important role in viral infections, the specific alterations in lipid metabolism during SFTSV infection remain unclear. This study aimed to elucidate the global lipid metabolic profiles of SFTS patients with mild, severe, and fatal outcomes. A total of 60 SFTS patients, consisting of 30 mild, 15 severe and 15 fatal patients, and 30 healthy controls, were enrolled for the investigation of global lipidomics in serum using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Our findings revealed global alterations in the lipid signature induced by SFTSV infection and further confirmed that the glycerophospholipid metabolism pathway was profoundly affected during the progression of mild, severe, and fatal outcomes in SFTS patients. Importantly, LysoPC (20:0) and LysoPC (P-16:0) are strongly correlated with the clinical parameters of SFTSV infection. Furthermore, we demonstrated the substantial prognostic value of LysoPC (20:0) and LysoPC (P-16:0) by receiver operating characteristic (ROC) curve analysis, providing evidence for their remarkable value as prognostic biomarkers for predicting SFTS clinical outcomes. In particular, LysoPC (20:0) and LysoPC (P-16:0), along with APTT, yielded superior prognostic performance for fatal SFTS [area under the curve (AUC) = 98.4%], outperforming routine clinical parameters. Collectively, our findings revealed the lipidomic landscape after SFTSV infection, which offers new insights into the mechanisms of SFTS disease progression and suggests that targeting lipid metabolism may serve as a potential therapeutic strategy. IMPORTANCE This study systematically investigated the lipid landscape profile of SFTS-infected patients with different clinical outcomes. Our results revealed a global alteration in the lipid signature, particularly the glycerophospholipid metabolic pathway, induced by SFTSV infection. Notably, LysoPC (20:0) and LysoPC (P-16:0) presented remarkable prognostic value as novel biomarkers for SFTSV infection and may contribute to the prognosis of SFTS progression and appropriate interventions.
Collapse
Affiliation(s)
- Panpan Tian
- Department of Laboratory Medicine, Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Liwei Zhao
- Department of Laboratory Medicine, Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Guiting Zhang
- Department of Laboratory Medicine, Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shixing Chen
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Wanying Zhang
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mingrong Ou
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yidan Sun
- Department of Laboratory Medicine, Nanjing Pukou People’s Hospital, Nanjing, Jiangsu, China
| | - Yuxin Chen
- Department of Laboratory Medicine, Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
31
|
Mouskeftara T, Kalopitas G, Liapikos T, Arvanitakis K, Theocharidou E, Germanidis G, Gika H. A Comprehensive Analysis of Liver Lipidomics Signature in Adults with Metabolic Dysfunction-Associated Steatohepatitis-A Pilot Study. Int J Mol Sci 2024; 25:13067. [PMID: 39684777 DOI: 10.3390/ijms252313067] [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: 10/20/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is the most common chronic liver disorder in Western countries, encompassing a range of conditions from steatosis to Metabolic Dysfunction-Associated Steatohepatitis (MASH), which can potentially progress to cirrhosis. Lipidomics approaches have revealed significant alterations in the hepatic lipidome associated with both steatosis and steatohepatitis, with these changes correlating with disease manifestation. While the transition from steatosis to MASH remains poorly understood, recent research indicates that both the quantity and quality of deposited lipids play a pivotal role in MASLD progression. In our study, we utilized untargeted and targeted analyses to identify intact lipids and fatty acids in liver biopsies from healthy controls and MASLD patients, categorized based on their histological findings. In total, 447 lipid species were identified, with 215 subjected to further statistical analysis. Univariate and multivariate analyses revealed alterations in triglyceride species and fatty acids, including FA 16:0, FA 16:1, FA 18:3 n6, the sum of MUFA, and the Δ9-desaturase activity ratio. This research provides insights into the connection between dysregulated lipid metabolism in the progression of MASLD, supporting previous findings. Further studies on lipid metabolism could improve risk assessment methods, particularly given the current limited understanding of the transition from steatosis to MASH.
Collapse
Affiliation(s)
- Thomai Mouskeftara
- Laboratory of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Georgios Kalopitas
- Division of Gastroenterology and Hepatology, 1st Department of Internal Medicine, AHEPA University Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
- Laboratory of Hygiene, Social and Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Theodoros Liapikos
- Laboratory of Analytical Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, 1st Department of Internal Medicine, AHEPA University Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Eleni Theocharidou
- 2nd Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, 1st Department of Internal Medicine, AHEPA University Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
- Laboratory of Hygiene, Social and Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Helen Gika
- Laboratory of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Biomic AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi Rd., 57001 Thessaloniki, Greece
| |
Collapse
|
32
|
Iliou A, Argyropoulou OD, Palamidas DA, Karagiannakou M, Benaki D, Tsezou KI, Vlachoyiannopoulos PG, Mikros E, Tzioufas AG. NMR-based metabolomics in giant cell arteritis and polymyalgia rheumatica sequential sera differentiates active and inactive disease. Rheumatology (Oxford) 2024; 63:3360-3369. [PMID: 37935429 DOI: 10.1093/rheumatology/kead590] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 09/28/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
OBJECTIVES GCA is an inflammatory disease following a chronic, relapsing course. The metabolic alterations related to the intense inflammatory process during the active phase and the rapid impact of steroid treatment remain unknown. This study aims to investigate the serum metabolome in active and inactive disease states. METHODS A total of 110 serum samples from 50 patients (33 GCA and 17 PMR) at three time points-0 (V1: active disease), 1 and 6 months (V2 and V3: remission)-of treatment with glucocorticoids (GCs) were subjected to NMR-based metabolomic analysis. Multi- and univariate statistical analyses were utilized to unveil metabolome alterations following treatment. RESULTS Distinct metabolic profiles were identified between activity and remission, independent of disease type. N-acetylglycoproteins and cholines of bound phospholipids emerged as predictive markers of disease activity. Altered levels of 4 of the 21 small molecules were also observed, including increased levels of phenylalanine and decreased glutamine, alanine and creatinine in active disease. Metabolic fingerprinting discriminated GCA from PMR in remission. GCA and PMR patients exhibited characteristic lipid alterations as a response and/or adverse effect of GC treatment. Correlation analysis showed that several identified biomarkers were further associated with acute phase reactants, CRP and ESR. CONCLUSION The NMR profile of serum metabolome could identify and propose sensitive biomarkers of inflammation. Metabolome alterations, following GC treatment, could provide predictors for future steroid-induced side effects.
Collapse
Affiliation(s)
- Aikaterini Iliou
- Department of Pharmaceutical Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Ourania D Argyropoulou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris-Anastasios Palamidas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Research Institute for Systemic Autoimmune Rheumatic Diseases (RISARD), Athens, Greece
| | - Marianna Karagiannakou
- Department of Pharmaceutical Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra Benaki
- Department of Pharmaceutical Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantina-Ismini Tsezou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Pharmagnose S.A., Inofyta, Greece
| | - Panayiotis G Vlachoyiannopoulos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Research Institute for Systemic Autoimmune Rheumatic Diseases (RISARD), Athens, Greece
| | - Emmanuel Mikros
- Department of Pharmaceutical Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- Pharmagnose S.A., Inofyta, Greece
- Pharma - Informatics Unit, Athena Research and Innovation Center in Information Communication & Knowledge Technologies, Marousi, Greece
| | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Research Institute for Systemic Autoimmune Rheumatic Diseases (RISARD), Athens, Greece
- Center of Stratified Medicine in Autoimmune and Rheumatic Diseases, Biomedical Research Foundation Academy of Athens, Athens, Greece
| |
Collapse
|
33
|
Carli F, Della Pepa G, Sabatini S, Vidal Puig A, Gastaldelli A. Lipid metabolism in MASLD and MASH: From mechanism to the clinic. JHEP Rep 2024; 6:101185. [PMID: 39583092 PMCID: PMC11582433 DOI: 10.1016/j.jhepr.2024.101185] [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: 04/23/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 11/26/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH) is recognised as a metabolic disease characterised by excess intrahepatic lipid accumulation due to lipid overflow and synthesis, alongside impaired oxidation and/or export of these lipids. But where do these lipids come from? The main pathways related to hepatic lipid accumulation are de novo lipogenesis and excess fatty acid transport to the liver (due to increased lipolysis, adipose tissue insulin resistance, as well as excess dietary fatty acid intake, in particular of saturated fatty acids). Not only triglycerides but also other lipids are secreted by the liver and are associated with a worse histological profile in MASH, as shown by lipidomics. Herein, we review the role of lipid metabolism in MASLD/MASH and discuss the impact of weight loss (diet, bariatric surgery, GLP-1RAs) or other pharmacological treatments (PPAR or THRβ agonists) on hepatic lipid metabolism, lipidomics, and the resolution of MASH.
Collapse
Affiliation(s)
- Fabrizia Carli
- Cardiometabolic Risk Laboratory, Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy
| | - Giuseppe Della Pepa
- Cardiometabolic Risk Laboratory, Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy
| | - Silvia Sabatini
- Cardiometabolic Risk Laboratory, Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy
| | - Antonio Vidal Puig
- Metabolic Research Laboratories, Medical Research Council Institute of Metabolic Science University of Cambridge, Cambridge CB2 0QQ UK
- Centro de Investigacion Principe Felipe Valencia 46012 Spain
- Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, China
| | - Amalia Gastaldelli
- Cardiometabolic Risk Laboratory, Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy
| |
Collapse
|
34
|
Kim J, Kim MY, Han Y, Lee GY, Kim DH, Heo YJ, Park M. Development of an electrochemical biosensor for non-invasive cholesterol monitoring via microneedle-based interstitial fluid extraction. Talanta 2024; 280:126771. [PMID: 39191110 DOI: 10.1016/j.talanta.2024.126771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/09/2024] [Accepted: 08/24/2024] [Indexed: 08/29/2024]
Abstract
In this study, we present the development of an innovative electrochemical biosensor integrated into a microneedle-based system for non-invasive and sensitive quantification of cholesterol levels in interstitial fluid (ISF). The biosensor employs a graphene-based electrode with a polyelectrolyte interlayer to immobilize cholesterol oxidase (ChOx), enabling selective cholesterol detection. Graphene oxide is electrochemically reduced to form a conductive layer, and PANI is chosen as the optimal polyelectrolyte for ChOx immobilization. The biosensor's performance is thoroughly evaluated, demonstrating excellent sensitivity, stability, and selectivity. Furthermore, the biosensor is successfully applied to skin-mimicking agarose gel and porcine skin, showcasing its potential for real-world interstitial fluid extraction and cholesterol monitoring. The integrated microneedle-based system offers a promising approach for non-invasive monitoring of cholesterol levels, with implications for personalized healthcare diagnostics.
Collapse
Affiliation(s)
- JeeYoung Kim
- Major in Materials Science and Engineering, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea; Integrative Materials Research Institute, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea; Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea
| | - Mi Yeon Kim
- Major in Materials Science and Engineering, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea; Integrative Materials Research Institute, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea; Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea
| | - Yuna Han
- Department of Mechanical Engineering, Kyung Hee University, 1732 Deogyeong-daero, Yongin-si, Gyeonggi-do, 17104, Republic of Korea; Integrated Education Institute for Frontier Science & Technology (BK21 Four), Kyung Hee University, 1732 Deogyeong-daero, Yongin-si, Gyeonggi-do, 17104, Republic of Korea
| | - Ga Yeong Lee
- Department of Mechanical Engineering, Kyung Hee University, 1732 Deogyeong-daero, Yongin-si, Gyeonggi-do, 17104, Republic of Korea; Integrated Education Institute for Frontier Science & Technology (BK21 Four), Kyung Hee University, 1732 Deogyeong-daero, Yongin-si, Gyeonggi-do, 17104, Republic of Korea
| | - Da Hyeon Kim
- Major in Materials Science and Engineering, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea; Integrative Materials Research Institute, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea; Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea
| | - Yun Jung Heo
- Department of Mechanical Engineering, Kyung Hee University, 1732 Deogyeong-daero, Yongin-si, Gyeonggi-do, 17104, Republic of Korea; Integrated Education Institute for Frontier Science & Technology (BK21 Four), Kyung Hee University, 1732 Deogyeong-daero, Yongin-si, Gyeonggi-do, 17104, Republic of Korea.
| | - Min Park
- Major in Materials Science and Engineering, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea; Integrative Materials Research Institute, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea; Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, 1 Hallymdaehak-gil, Chuncheon-si, Gangwon-do, 24252, Republic of Korea.
| |
Collapse
|
35
|
Mendoza YP, Tsouka S, Semmler G, Seubnooch P, Freiburghaus K, Mandorfer M, Bosch J, Masoodi M, Berzigotti A. Metabolic phenotyping of patients with advanced chronic liver disease for better characterization of cirrhosis regression. J Hepatol 2024; 81:983-994. [PMID: 38944391 DOI: 10.1016/j.jhep.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND & AIMS Regression of cirrhosis has been observed in patients with viral and non-viral etiologies of liver disease in whom the underlying cause of liver injury was effectively suppressed. However, the understanding of the factors contributing to reversibility of fibrosis and cirrhosis is limited. Our aims were to assess clinical factors, perform genotyping of known variants, and comprehensive metabolic phenotyping to characterize the regression of fibrosis in patients with compensated advanced chronic liver disease (cACLD). METHODS In a case-control pilot study of 81 patients with cACLD, we compared individuals exhibiting histological or clinical evidence of cACLD regression ("regressors"; n = 44) with those showing no improvement ("non-regressors"; n = 37) after a minimum of 24 months of successful treatment of the cause of liver disease. Data were validated using an external validation cohort (n = 30). RESULTS Regardless of the cause of cACLD, the presence of obesity (odds ratio [OR] 0.267 95% CI 0.072-0.882; p = 0.049), high liver stiffness (OR 0.960, 95% CI 0.925-0.995; p = 0.032), and carriage of GCKR variant rs1260326 (OR 0.148, 95% CI 0.030-0.773; p = 0.019) are associated with a reduced likelihood of fibrosis regression in a subgroup of 60 patients with ACLD genotyped for known genetic variants. Using liver tissue transcriptomics, we identified metabolic pathways differentiating regressors from non-regressors, with top pathways associated with lipid metabolism - especially fatty acids, bile acids, phospholipids, triacylglycerides (biosynthesis), and the carnitine shuttle. In the entire discovery cohort, we further measured metabolites within the defined pathways, which led to the identification of 33 circulating markers differentiating regressors from non-regressors after etiological therapy. The validation cohort confirmed 14 of the differentially expressed markers. CONCLUSIONS We identified and validated a group of lipid biomarkers associated with regression of fibrosis that could be used as non-invasive biomarkers for detecting regression of fibrosis in cACLD. IMPACT AND IMPLICATIONS Regression of cirrhosis/advanced chronic liver disease (ACLD) after removal of the underlying cause of liver injury has been observed in human cirrhosis. However, detailed characterization of ACLD regression remains an unmet need. In this study, we provide a comprehensive phenotyping of individuals likely to experience ACLD regression. While obesity, carriage of GCKR variant rs1260326 and high liver stiffness were associated with lower likelihood of regression of ACLD, a signature of circulating lipid metabolites enabled differentiation of regressors from non-regressors after effective etiologic therapy. The lipid signature we discovered and externally validated could be used as non-invasive biomarker to detect regression of fibrosis in patients with compensated ACLD.
Collapse
Affiliation(s)
- Yuly P Mendoza
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland; Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland; Graduate School for Health Sciences (GHS), University of Bern, Switzerland
| | - Sofia Tsouka
- Institute of Clinical Chemistry, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Georg Semmler
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Patcharamon Seubnooch
- Institute of Clinical Chemistry, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Katrin Freiburghaus
- Institute of Clinical Chemistry, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Mattias Mandorfer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Jaime Bosch
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland; Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
| | - Mojgan Masoodi
- Institute of Clinical Chemistry, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland; Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland.
| |
Collapse
|
36
|
Mak LY, Liu K, Chirapongsathorn S, Yew KC, Tamaki N, Rajaram RB, Panlilio MT, Lui R, Lee HW, Lai JCT, Kulkarni AV, Premkumar M, Lesmana CRA, Hsu YC, Huang DQ. Liver diseases and hepatocellular carcinoma in the Asia-Pacific region: burden, trends, challenges and future directions. Nat Rev Gastroenterol Hepatol 2024; 21:834-851. [PMID: 39147893 DOI: 10.1038/s41575-024-00967-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/17/2024]
Abstract
Globally, nearly half of deaths from cirrhosis and chronic liver diseases (CLD) and three-quarters of deaths from hepatocellular carcinoma (HCC) occur in the Asia-Pacific region. Chronic hepatitis B is responsible for the vast majority of liver-related deaths in the region. Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common form of CLD, affecting an estimated 30% of the adult population. Compared with people of European descent, people from the Asia-Pacific region carry more genetic variants associated with MASLD and its progression. Alcohol is a fast-growing cause of CLD and HCC in Asia as a result of the rising per-capita consumption of alcohol. Drug-induced liver injury is under-recognized and probably has a high prevalence in this region. The epidemiological and outcome data of acute-on-chronic liver failure are heterogeneous, and non-unified definitions across regions contribute to this heterogeneity. CLDs are severely underdiagnosed, and effective treatments and vaccinations are underutilized. In this Review, we highlight trends in the burden of CLD and HCC in the Asia-Pacific region and discuss the rapidly changing aetiologies of liver disease. We examine the multiple gaps in the care cascade and propose mitigating strategies and future directions.
Collapse
Affiliation(s)
- Lung-Yi Mak
- The University of Hong Kong, Hong Kong, China
| | - Ken Liu
- The University of Sydney, Sydney, Australia
| | | | | | | | | | | | - Rashid Lui
- The Chinese University of Hong Kong, Hong Kong, China
| | - Hye Won Lee
- Yonsei University College of Medicine, Seoul, Korea
| | | | - Anand V Kulkarni
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | - Madhumita Premkumar
- Department of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Yao Chun Hsu
- Department of Medical Research, E-Da Hospital, Kaohsiung, Taiwan; School of Medicine and Graduate Institute of Medicine, I-Shou University, Kaohsiung, Taiwan
- School of Medicine and Graduate Institute of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Daniel Q Huang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore, Singapore.
| |
Collapse
|
37
|
Song Q, Pan J, Pan M, Zheng C, Fan W, Zhen J, Pi D, Liang Z, Shen H, Li Y, Yang Q, Zhang Y. Exploring the relationship between air pollution, non-alcoholic fatty liver disease, and liver function indicators: a two-sample Mendelian randomization analysis study. Front Endocrinol (Lausanne) 2024; 15:1396032. [PMID: 39678198 PMCID: PMC11637881 DOI: 10.3389/fendo.2024.1396032] [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: 03/05/2024] [Accepted: 11/06/2024] [Indexed: 12/17/2024] Open
Abstract
Background and aims Non-alcoholic fatty liver disease (NAFLD) is a common metabolic disorder worldwide, with an increasing incidence in recent years. While previous studies have suggested an association between the air pollutant PM2.5 and NAFLD, there is still considerable debate regarding the existence of a clear causal relationship between air pollution and NAFLD. This study aims to employ Mendelian randomization methods to evaluate the causal relationship between major air pollutants and NAFLD. Method We conducted Mendelian randomization analyses on a large-scale publicly available genome-wide association study (GWAS) dataset of European populations to dissect the association between air pollutants, NAFLD, and liver function indicators. We used five different analysis methods, including Inverse-variance weighted (IVW), Weighted median, MR-Egger, Simple mode, and Weighted mode, to analyze the data. We also tested for pleiotropy, heterogeneity, and sensitivity of the results. Results This study utilized four common exposures related to air pollution and four outcomes related to NAFLD. The results regarding the association between air pollutants and NAFLD (PM2.5: P=0.808, 95% CI=0.37-3.56; PM10: P=0.238, 95% CI=0.33-1.31; nitrogen dioxide: P=0.629, 95% CI=0.40-4.61; nitrogen oxides: P=0.123, 95% CI=0.13-1.28) indicated no statistically significant correlation between them. However, notably, there was a causal relationship between PM10 and serum albumin (ALB) levels (P=0.019, 95% CI=1.02-1.27). Conclusion This MR study found no evidence of a causal relationship between air pollution and NAFLD in European populations. However, a statistically significant association was observed between PM10 and ALB levels, suggesting that the air pollutant PM10 may impact the liver's ability to synthesize proteins.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Qinhe Yang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yupei Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
38
|
Beyoğlu D, Popov YV, Idle JR. Metabolomic Hallmarks of Obesity and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2024; 25:12809. [PMID: 39684520 DOI: 10.3390/ijms252312809] [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: 10/24/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
From a detailed review of 90 experimental and clinical metabolomic investigations of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD), we have developed metabolomic hallmarks for both obesity and MASLD. Obesity studies were conducted in mice, rats, and humans, with consensus biomarker groups in plasma/serum being essential and nonessential amino acids, energy metabolites, gut microbiota metabolites, acylcarnitines and lysophosphatidylcholines (LPC), which formed the basis of the six metabolomic hallmarks of obesity. Additionally, mice and rats shared elevated cholesterol, humans and rats shared elevated fatty acids, and humans and mice shared elevated VLDL/LDL, bile acids and phosphatidylcholines (PC). MASLD metabolomic studies had been performed in mice, rats, hamsters, cows, geese, blunt snout breams, zebrafish, and humans, with the biomarker groups in agreement between experimental and clinical investigations being energy metabolites, essential and nonessential amino acids, fatty acids, and bile acids, which lay the foundation of the five metabolomic hallmarks of MASLD. Furthermore, the experimental group had higher LPC/PC and cholesteryl esters, and the clinical group had elevated acylcarnitines, lysophosphatidylethanolamines/phosphatidylethanolamines (LPE/PE), triglycerides/diglycerides, and gut microbiota metabolites. These metabolomic hallmarks aid in the understanding of the metabolic role played by obesity in MASLD development, inform mechanistic studies into underlying disease pathogenesis, and are critical for new metabolite-inspired therapies.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| | - Yury V Popov
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jeffrey R Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| |
Collapse
|
39
|
Fan JG, Xu XY, Yang RX, Nan YM, Wei L, Jia JD, Zhuang H, Shi JP, Li XY, Sun C, Li J, Wong VWS, Duan ZP. Guideline for the Prevention and Treatment of Metabolic Dysfunction-associated Fatty Liver Disease (Version 2024). J Clin Transl Hepatol 2024; 12:955-974. [PMID: 39544247 PMCID: PMC11557364 DOI: 10.14218/jcth.2024.00311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024] Open
Abstract
With the rising epidemic of obesity, metabolic syndrome, and type 2 diabetes mellitus in China, metabolic dysfunction-associated non-alcoholic fatty liver disease has become the most prevalent chronic liver disease. This condition frequently occurs in Chinese patients with alcoholic liver disease and chronic hepatitis B. To address the impending public health crisis of non-alcoholic fatty liver disease and its underlying metabolic issues, the Chinese Society of Hepatology and the Chinese Medical Association convened a panel of clinical experts to revise and update the "Guideline of prevention and treatment of non-alcoholic fatty liver disease (2018, China)". The new edition, titled "Guideline for the prevention and treatment of metabolic dysfunction-associated fatty liver disease (Version 2024)", offers comprehensive recommendations on key clinical issues, including screening and monitoring, diagnosis and evaluation, treatment, and follow-up for metabolic dysfunction-associated fatty liver disease and metabolic dysfunction-associated steatotic liver disease. Metabolic dysfunction-associated fatty liver disease is now the preferred English term and is used interchangeably with metabolic dysfunction-associated steatotic liver disease. Additionally, the guideline emphasizes the importance of multidisciplinary collaboration among hepatologists and other specialists to manage cardiometabolic disorders and liver disease effectively.
Collapse
Affiliation(s)
- Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Yuan Xu
- Department of Infectious Diseases, Peking University First Hospital, Beijing, China
| | - Rui-Xu Yang
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue-Min Nan
- Department of Traditional and Western Medical Hepatology, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
| | - Lai Wei
- Hepatopancreatobiliary Centre, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Ji-Dong Jia
- Liver Research Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hui Zhuang
- Department of Microbiology and Centre for Infectious Diseases, Peking University Health Science Centre, Beijing, China
| | - Jun-Ping Shi
- Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiao-Ying Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chao Sun
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Li
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Vincent Wai-Sun Wong
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhong-Ping Duan
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chinese Society of Hepatology, Chinese Medical Association
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Infectious Diseases, Peking University First Hospital, Beijing, China
- Department of Traditional and Western Medical Hepatology, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Hepatopancreatobiliary Centre, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Liver Research Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Microbiology and Centre for Infectious Diseases, Peking University Health Science Centre, Beijing, China
- Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Lin AZ, Fu X, Jiang Q, Zhou X, Hwang SH, Yin HH, Ni KD, Pan QJ, He X, Zhang LT, Meng YW, Liu YN, Hammock BD, Liu JY. Metabolomics reveals soluble epoxide hydrolase as a therapeutic target for high-sucrose diet-mediated gut barrier dysfunction. Proc Natl Acad Sci U S A 2024; 121:e2409841121. [PMID: 39556751 PMCID: PMC11621843 DOI: 10.1073/pnas.2409841121] [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: 06/05/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Highsucrose diet (HSD) was reported as a causative factor for multiorgan injuries. The underlying mechanisms and therapeutic strategies remain largely uncharted. In the present study, by using a metabolomics approach, we identified the soluble epoxide hydrolase (sEH) as a therapeutic target for HSD-mediated gut barrier dysfunction. Specifically, 16-week feeding on an HSD caused gut barrier dysfunction, such as colon inflammation and tight junction impairment in a murine model. A metabolomics analysis of mouse colon tissue showed a decrease in the 5(6)-epoxyeicosatrienoic acid [5(6)-EET] level and an increase in soluble epoxide hydrolase, which is related to HSD-mediated injuries to the gut barrier. The mice treated with a chemical inhibitor of sEH and the mice with genetic intervention by intestinal-specific knockout of the sEH gene significantly attenuated HSD-caused intestinal injuries by reducing HSD-mediated colon inflammation and improving the impaired tight junction caused by an HSD. Further, in vitro studies showed that treatment with 5(6)-EET, but not its hydrolytic product 5,6-dihydroxyeicosatrienoic acid (5,6-DiHET), significantly ablated high sucrose-caused intestinal epithelial inflammation and impaired tight junction. Additionally, 5(6)-EET is anti-inflammatory and improves gut epithelial tight junction while 5,6-DiHET cannot do so. This study presents an underlying mechanism of and a therapeutic strategy for the gut barrier dysfunction caused by an HSD.
Collapse
Affiliation(s)
- Ai-Zhi Lin
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Xian Fu
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Qing Jiang
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Xue Zhou
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Sung Hee Hwang
- Department of Entomology and Nematology, University of California, Davis, CA95616
- Comprehensive Cancer Center, University of California, Davis, CA95616
| | - Hou-Hua Yin
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Kai-Di Ni
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Qing-Jin Pan
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Xin He
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Ling-Tong Zhang
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Yi-Wen Meng
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Ya-Nan Liu
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology, University of California, Davis, CA95616
- Comprehensive Cancer Center, University of California, Davis, CA95616
| | - Jun-Yan Liu
- Department of Anesthesia of the Second Affiliated Hospital and CNTTI of College of Pharmacy, Chongqing Medical University, Chongqing400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing400016, China
| |
Collapse
|
41
|
Li Z, Zhu X, Li C, Tang R, Zou Y, Liu S. Integrated serum metabolomics, 16S rRNA sequencing and bile acid profiling to reveal the potential mechanism of gentiopicroside against nonalcoholic steatohepatitis in lean mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118526. [PMID: 38972531 DOI: 10.1016/j.jep.2024.118526] [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: 04/15/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lean nonalcoholic steatohepatitis (NASH) poses a serious threat to public health worldwide. Herbs of the genus Gentiana have been used for centuries to treat hepatic disease or have been consumed for hepatic protection efficiency. Gentiopicroside (GPS), the main bioactive component of Gentiana herbs, has been shown to be beneficial for protecting the liver, improving intestinal disorders, modulating bile acid profiles, ameliorating alcoholic hepatosteatosis, and so on. It is plausible to speculate that GPS may hold potential as a therapeutic strategy for lean NASH. However, no related studies have been conducted thus far. AIM OF THE STUDY The present work aimed to investigate the benefit of GPS on NASH in a lean mouse model. MATERIALS AND METHODS NASH in a lean mouse model was successfully established via a published method. GPS of 50 and 100 mg/kg were orally administered to verify the effect. Untargeted metabolomics, 16S rDNA sequencing and bile acid (BA) profiling, as well as qPCR and Western blotting analysis were employed to investigate the mechanism underlying the alleviating effect. RESULTS GPS significantly reduced the increase in serum biochemicals and liver index, and attenuated the accumulation of fat in the livers of lean mice with NASH. Forty-two potential biomarkers were identified by metabolomics analysis, leading to abnormal metabolic pathways of primary bile acid biosynthesis and fatty acid biosynthesis, which were subsequently rebalanced by GPS. A decreased Firmicutes/Bacteroidetes (F/B) ratio and disturbed BA related GM profiles were revealed in lean mice with NASH but were partially recovered by GPS. Furthermore, serum profiling of 23 BAs confirmed that serum BA levels were elevated in the lean model but downregulated by GPS treatment. Pearson correlation analysis validated associations between BA profiles, serum biochemical indices and related GM. qPCR and Western blotting analysis further elucidated the regulation of genes associated with liver lipid synthesis and bile acid metabolism. CONCLUSIONS GPS may ameliorate steatosis in lean mice with NASH, regulating the metabolomic profile, BA metabolism, fatty acid biosynthesis, and BA-related GM. All these factors may contribute to its beneficial effect.
Collapse
Affiliation(s)
- Zeyun Li
- Department of pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Xueya Zhu
- Department of pharmacy, Zhumadian Central Hospital, Zhumadian, 463000, Henan, China.
| | - Chenhao Li
- Department of pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Ruiting Tang
- Department of pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yuanyuan Zou
- Yichun University, Yichun, 336000, Jiangxi, China.
| | - Shuaibing Liu
- Department of pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
42
|
Wu D, Wu H, Yu P, Liu H, Liu M, Wang J, Wang X, Xu F. Unraveling the molecular mechanism of aqueous extract of Sargentodoxa cuneata against ulcerative colitis from serum metabolomics and bioinformatics perspectives. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1249:124372. [PMID: 39561468 DOI: 10.1016/j.jchromb.2024.124372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 10/19/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024]
Abstract
Symptoms of ulcerative colitis (UC) are like "intestinal carbuncle" in Chinese medicine. The aqueous extract of Sargentodoxa cuneata (AESc) has good therapeutic effects on UC, but the underlying mechanism needs to be further elucidated. The mechanism of AESc against UC was studied based on metabolomics and bioinformatics in mice with UC. Dextran sodium sulfate was applied to induce a mouse model of UC. After the intervention of AESc, the general condition of the animals was recorded, and efficacy-related indicators were measured. Information on serum metabolites was determined. Multivariate analysis combined with bioinformatics methods were used to identify the differential metabolites. Furthermore, "metabolite-target-disease" network was obtained, and differential metabolites of UC were screened, and further analysis of the metabolites were performed. Molecular docking validation was also carried out. AESc improved general conditions such as blood in stool, hair of animals, and weight loss, reduced disease activity index scores and shortening of colon length in mice with UC. A total of 3445 serum metabolites were obtained, and 64 differentiated metabolites of AESc against UC were screened. Enrichment analysis showed that arachidonic acid metabolism, bile secretion, drug metabolism-other enzymes, and tyrosine metabolism were associated with AESc in the treatment of UC. In addition, based on "metabolite-target-disease" network, the serum metabolites cholylleucine, 9,10,13-TriHOME, birabresib, anthramycin methyl ether, trans-hexadec-2-enoyl carnitine, and lucidumol A were found to have the therapeutic potential for UC. Further, 14 core targets were obtained, and lipids and atherosclerosis, rheumatoid arthritis and multiple immune-inflammatory pathways were associated with AESc for the treatment of UC. AESc corrects serum metabolic disturbances in UC mice, and multiple serum metabolites have therapeutic potential for UC. AESc may treat UC by regulating biological processes such as lipid metabolism, amino acid metabolism, thereby restoring normal physiological function of the intestine.
Collapse
Affiliation(s)
- Dengli Wu
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Hongmei Wu
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Piao Yu
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Hongyun Liu
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Mei Liu
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Junyi Wang
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xiangpei Wang
- School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, China.
| | - Feng Xu
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
43
|
Ding J, Liu H, Zhang X, Zhao N, Peng Y, Shi J, Chen J, Chi X, Li L, Zhang M, Liu WY, Zhang L, Ouyang J, Yuan Q, Liao M, Tan Y, Li M, Xu Z, Tang W, Xie C, Li Y, Pan Q, Xu Y, Cai SY, Byrne CD, Targher G, Ouyang X, Zhang L, Jiang Z, Zheng MH, Sun F, Chai J. Integrative multiomic analysis identifies distinct molecular subtypes of NAFLD in a Chinese population. Sci Transl Med 2024; 16:eadh9940. [PMID: 39504356 DOI: 10.1126/scitranslmed.adh9940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/07/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become a common health care burden worldwide. The high heterogeneity of NAFLD remains elusive and impairs outcomes of clinical diagnosis and pharmacotherapy. Several NAFLD classifications have been proposed on the basis of clinical, genetic, alcoholic, or serum metabolic analyses. Yet, accurately predicting the progression of NAFLD to cirrhosis or hepatocellular carcinoma (HCC) in patients remains a challenge. Here, on the basis of a Chinese cohort of patients, we classified NAFLD into three distinct molecular subtypes (NAFLD-mSI, NAFLD-mSII, and NAFLD-mSIII) using integrative multiomics including whole-genome sequencing (WGS), proteomics, phosphoproteomics, lipidomics, and metabolomics across a broad range of liver, blood, and urine specimens. We found that NAFLD-mSI had higher expression of CYP1A2 and CYP3A4, which alleviate hepatic steatosis through mediating free fatty acid/bile acid-mTOR-FXR/PPARα signaling. NAFLD-mSII displayed an elevated risk of liver cirrhosis along with increased hepatic infiltration of M1 and M2 macrophages because of lipid-triggered hepatic CCL2 and CRP production. NAFLD-mSIII exhibited a potential risk for HCC development by increased transcription of CEBPB- and ERCC3-regulated oncogenes because of activation of the EGF-EGFR/CHKA/PI3K-PDK1-AKT cascade. Next, we validated the existence of these three NAFLD molecular subtypes in an external cohort comprising 92 patients with NAFLD across three different Chinese hospitals. These findings may aid in understanding the molecular features underlying NAFLD heterogeneity, thereby facilitating clinical diagnosis and treatment strategies with the aim of preventing the development of liver cirrhosis and HCC.
Collapse
Affiliation(s)
- Jingjing Ding
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Huaizheng Liu
- Department of Emergency, the Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xiaoxun Zhang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Nan Zhao
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Peng
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Junping Shi
- Department of Infectious Diseases and Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
| | - Jinjun Chen
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoling Chi
- Department of Hepatology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ling Li
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mengni Zhang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wen-Yue Liu
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Liangjun Zhang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiafeng Ouyang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qian Yuan
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Department of Pharmacy, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Min Liao
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ya Tan
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mingqiao Li
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ziqian Xu
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wan Tang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chuanming Xie
- Institute of Hepatobiliary Surgery, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi Li
- Department of Clinical Laboratory, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Qiong Pan
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Xu
- School of Clinical Medicine and the First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Shi-Ying Cai
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella 37024, Italy
| | - Xinshou Ouyang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Liqun Zhang
- Department of Clinical Laboratory, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Zhongyong Jiang
- Department of Medical Laboratory, Cheng du Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu 610213, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou 325000, China
| | - Fengjun Sun
- Department of Pharmacy, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jin Chai
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
44
|
Hoppmann A, Arriola Apelo SI. Fast and reliable method for analysis of derivatized plasma amino acids by liquid chromatography-single quadrupole-mass spectrometry. JDS COMMUNICATIONS 2024; 5:745-750. [PMID: 39650039 PMCID: PMC11624321 DOI: 10.3168/jdsc.2024-0546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/12/2024] [Indexed: 12/11/2024]
Abstract
The pool of free, genetically encoded AA in plasma plays an essential role not only as substrate for every protein synthesized in the body but also as signaling molecules that regulate a wide range of physiological processes. Here we present a method for the analysis of 19 of the 20 encoded AA (except Cys) in dairy cow plasma. Isolated plasma or standards for the 19 AA were gravimetrically mixed with an internal standard mix consisting of 13C isotopes of each AA. Plasma proteins were precipitated on acetonitrile and supernatants transferred to glass vials. For precolumn derivatization, plasma supernatants were buffered with sodium borate (pH 9.5-10), and AA were derivatized with 9-fluorenylmethoxycarbonyl (Fmoc) chloride. Analytes were isolated by solid-phase extraction using a strong-anionic ion exchange column and dry eluates were reconstituted in mobile phase consisting of 70% water solution of ammonium formate and 30% acetonitrile. Amino acid derivatives were separated by reverse-phase liquid chromatography over 17.5 min with a C18 column in which acetonitrile increased to 80% over the first 11 min of the method, before returning to initial levels. Electrospray ionization on negative mode was used for most AA, except Arg and Pro, for which positive mode yielded superior results. Single or double (only Lys) derivatives were measured by single quadrupole-mass spectrometry. We hypothesized that precolumn Fmoc derivatization would yield optimal resolution for quantitative analysis of the 19 targeted AA and their respective 13C internal standards, with limits of quantitation beyond physiological ranges. All 19 AA were detected with minimal background noise. An 11-point standard curve was developed for each AA. Limits of quantitation were beyond concentrations observed in plasma samples of lactating dairy cows, except for Gly where upper curve points had to be removed to maintain linearity, limiting quantitation to the upper range of physiological concentration. After removing the 4 highest concentrations from the Gly standard curve, coefficients of determination were greater than 0.999 for all of the AA. Recovery of spiked AA from plasma samples ranged from 89.9% for Phe to 100.3% for Trp. Instrument repeatability averaged 0.91 and ranged from 0.33 for Val to 2.29 for Arg. Meanwhile, sample preparation method repeatability averaged 2.02 and ranged from 1.14 for Tyr to 3.34 for Arg. Although robust methods have been developed, they depend on either availability of sophisticated instruments, mostly limited to core facilities (i.e., tandem MS methods), long and expensive chromatography without specific internal standards for each AA (i.e., HPLC-ultraviolet and HPLC-fluorescence detector), or unstable derivatization (GC-MS). Here we describe a method with high throughput, stable derivatization, high precision and recovery, and potentially more affordable than most existing methods. This method could help dairy nutritionists to consider plasma AA information for diet formulation strategies, potentially reducing feeding costs and N emissions.
Collapse
Affiliation(s)
- August Hoppmann
- Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI 53706
| | | |
Collapse
|
45
|
Geng C, Chen F, Sun H, Lin H, Qian Y, Zhang J, Xia Q. Serum Arginine Level for Predicting Early Allograft Dysfunction in Liver Transplantation Recipients by Targeted Metabolomics Analysis: A Prospective, Single-Center Cohort Study. Adv Biol (Weinh) 2024; 8:e2400128. [PMID: 39164220 DOI: 10.1002/adbi.202400128] [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: 03/06/2024] [Revised: 07/09/2024] [Indexed: 08/22/2024]
Abstract
Early allograft dysfunction (EAD) is a frequent phenomenon, leading to increased graft loss and higher mortality after liver transplantation (LT). Despite significant efforts for early diagnosis of EAD, there is no existing approach that can predict EAD on the first post-operative day. The aim is to define a metabolite-based biomarker on the first day after LT complicated with EAD. Ten patients diagnosed with EAD and 26 non-EAD are recruited for the study. A HPLC-MS/MS is used to determine 14 amino acids and 15 bile acids serum concentration. Comparative analyses are conducted between EAD and non-EAD groups. Arginine is identified as the most significant metabolite distinguishing the EAD and non-EAD groups, and therefore, is identified as a potential biomarker of EAD. The optimal cut-off value for arginine is 52.09 µmol L-1, with an AUROC of 0.804 (95% confidence interval: 0.638-0.917, p < 0.001), yielding a sensitivity of 100%, specificity of 53.8%, and Youden index of 0.54, NPVof 100%, and PPV of 45.45%. In summary, the study indicated that targeted metabolomics analysis would be a promising strategy for discovering novel biomarkers to predict EAD. The identified arginine may be helpful in developing an objective diagnostic method for EAD.
Collapse
Affiliation(s)
- Chunmei Geng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, P. R. China
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, P. R. China
| | - Fang Chen
- Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, P. R. China
| | - Hanyong Sun
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, P. R. China
| | - Houwen Lin
- Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, P. R. China
| | - Yongbing Qian
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, P. R. China
| | - Jianjun Zhang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, P. R. China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, P. R. China
| |
Collapse
|
46
|
Tang J, Tang T, Yu Q, Tong X, Liu C, Chen C, Hu S, Zhang S, You Y, Wang C, Li J, Wen W, Chen J, Zhang X, Wang M, Tong F. Association Between Remnant Cholesterol and Nonalcoholic Fatty Liver Disease: A Systemic Review and Meta-Analysis. Metab Syndr Relat Disord 2024; 22:637-645. [PMID: 39158352 DOI: 10.1089/met.2024.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
Lipid disorders are related to the risk of nonalcoholic fatty liver disease (NAFLD). Remnant cholesterol (RC), a nonclassical and once-neglected risk factor for NAFLD, has recently received new attention. In this study, we assessed the relationship between the RC levels and NAFLD risk. We searched across PubMed, Web of Science, Embase, Cochrane Library, and China National Knowledge Infrastructure, with no restrictions on publication languages. Retrospective cohort studies and cross-sectional studies were enrolled from the inception of the databases until August 6, 2023. A random-effect model was applied to construct the mean difference, and a 95% confidence interval was applied to assess the relationship between the RC levels and NAFLD risk. We used two methods to estimate RC levels: Calculated-1 subtracts low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol from total cholesterol; Calculated-2 uses the Friedewald formula for LDL-C when triglycerides are <4.0 mmol/L, otherwise directly measured. A total of 265 published studies were selected through preliminary retrieval. Of these, six studies met the inclusion requirements and were enrolled in the meta-analysis. The RC level in the NAFLD group was significantly higher than that in the non-NAFLD group (mean difference: 0.18, 95% confidence interval: 0.10-0.26, P < 0.00001). We conducted subgroup analyses of computational methods and geographic regions. Notably, in the subgroup analysis of Calculation Method 2, the NAFLD group had significantly higher RC levels than the non-NAFLD group. On the other hand, in Calculation Method 1, the difference between the two groups was insignificant. In both the Asian and non-Asian populations, the RC levels were significantly higher in the NAFLD group than in the non-NAFLD group. The association of RC with an increased NAFLD risk was not dependent on the triglyceride. This meta-analysis suggests that elevated RC levels are associated with an increased risk of NAFLD. In addition to the conventional risk factors for fatty liver, clinicians should be concerned about the RC levels in the clinic.
Collapse
Affiliation(s)
- Jiake Tang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Ting Tang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Qingwen Yu
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Xuhan Tong
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Chun Liu
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Chen Chen
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Siqi Hu
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Shenghui Zhang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Yao You
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Chunyi Wang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Jie Li
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Wen Wen
- Department of Cardiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Zhejiang, China
| | - Juan Chen
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Xingwei Zhang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Mingwei Wang
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
- Hangzhou Lin'an Fourth People's Hospital, Hangzhou, China
| | - Fan Tong
- Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Institute of Cardiovascular Diseases, Zhejiang Key Laboratory of Medical Epigenetics, Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
47
|
Zhou W, Du Z. Oleuropein mitigates non-alcoholic fatty liver disease (NAFLD) and modulates liver metabolites in high-fat diet-induced obese mice via activating PPARα. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:8634-8645. [PMID: 38952322 DOI: 10.1002/jsfa.13691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND This study aimed to elucidate the mechanism of oleuropein (OLE) ameliorates non-alcoholic fatty liver disease (NAFLD) and its underlying mechanisms. RESULTS Male C57BL/6J mice were fed either a low-fat diet (LFD), a high-fat diet (HFD), or a HFD supplemented with 0.03% (w/w) OLE for 16 weeks. OLE supplementation decreased body weight and liver weight, improved serum lipid profiles, and ameliorated HFD-induced hepatic dysfunction. Liver metabolomics analysis revealed that OLE increased the levels of nicotinamide, tauroursodeoxycholic acid, taurine, and docosahexaenoic acid, which were beneficial for lipid homeostasis and inflammation regulation. OLE exerted its protective effects by activating peroxisome proliferator-activated receptor alpha (PPARα), a key transcription factor that regulates fibroblast growth factor 21 (FGF21) expression and modulates lipid oxidation, lipogenesis and inflammation pathways. Importantly, OLE supplementation did not significantly affect body weight or liver weight in PPARα knockout (PPARα KO) mice, indicating that PPARα is essential for OLE-mediated NAFLD prevention. CONCLUSION Our results suggest that OLE alleviates NAFLD in mice by activating PPARα and modulating liver metabolites. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wei Zhou
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zheng Du
- The First People's Hospital of Lianyungang Public Health Department, Lianyungang, China
| |
Collapse
|
48
|
Taranto D, Kloosterman DJ, Akkari L. Macrophages and T cells in metabolic disorder-associated cancers. Nat Rev Cancer 2024; 24:744-767. [PMID: 39354070 DOI: 10.1038/s41568-024-00743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 10/03/2024]
Abstract
Cancer and metabolic disorders have emerged as major global health challenges, reaching epidemic levels in recent decades. Often viewed as separate issues, metabolic disorders are shown by mounting evidence to heighten cancer risk and incidence. The intricacies underlying this connection are still being unraveled and encompass a complex interplay between metabolites, cancer cells and immune cells within the tumour microenvironment (TME). Here, we outline the interplay between metabolic and immune cell dysfunction in the context of three highly prevalent metabolic disorders, namely obesity; two associated liver diseases, metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH); and type 2 diabetes. We focus primarily on macrophages and T cells, the critical roles of which in dictating inflammatory response and immune surveillance in metabolic disorder-associated cancers are widely reported. Moreover, considering the ever-increasing number of patients prescribed with metabolism disorder-altering drugs and diets in recent years, we discuss how these therapies modulate systemic and local immune phenotypes, consequently impacting cancer malignancy. Collectively, unraveling the determinants of metabolic disorder-associated immune landscape and their role in fuelling cancer malignancy will provide a framework essential to therapeutically address these highly prevalent diseases.
Collapse
Affiliation(s)
- Daniel Taranto
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan J Kloosterman
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leila Akkari
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
49
|
Lei L, Li J, Liu Z, Zhang D, Liu Z, Wang Q, Gao Y, Mo B, Li J. Identification of diagnostic markers pyrodeath-related genes in non-alcoholic fatty liver disease based on machine learning and experiment validation. Sci Rep 2024; 14:25541. [PMID: 39462099 PMCID: PMC11513955 DOI: 10.1038/s41598-024-77409-3] [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: 08/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) poses a global health challenge. While pyroptosis is implicated in various diseases, its specific involvement in NAFLD remains unclear. Thus, our study aims to elucidate the role and mechanisms of pyroptosis in NAFLD. Utilizing data from the Gene Expression Omnibus (GEO) database, we analyzed the expression levels of pyroptosis-related genes (PRGs) in NAFLD and normal tissues using the R data package. We investigated protein interactions, correlations, and functional enrichment of these genes. Key genes were identified employing multiple machine learning techniques. Immunoinfiltration analyses were conducted to discern differences in immune cell populations between NAFLD patients and controls. Key gene expression was validated using a cell model. Analysis of GEO datasets, comprising 206 NAFLD samples and 10 controls, revealed two key PRGs (TIRAP, and GSDMD). Combining these genes yielded an area under the curve (AUC) of 0.996 for diagnosing NAFLD. In an external dataset, the AUC for the two key genes was 0.825. Nomogram, decision curve, and calibration curve analyses further validated their diagnostic efficacy. These genes were implicated in multiple pathways associated with NAFLD progression. Immunoinfiltration analysis showed significantly lower numbers of various immune cell types in NAFLD patient samples compared to controls. Single sample gene set enrichment analysis (ssGSEA) was employed to assess the immune microenvironment. Finally, the expression of the two key genes was validated in cell NAFLD model using qRT-PCR. We developed a prognostic model for NAFLD based on two PRGs, demonstrating robust predictive efficacy. Our findings enhance the understanding of pyroptosis in NAFLD and suggest potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Liping Lei
- Department of Geriatric Medicine, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Jixue Li
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zirui Liu
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Dongdong Zhang
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zihan Liu
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Qing Wang
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Yi Gao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Biwen Mo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541002, Guangxi, China.
| | - Jiangfa Li
- Division of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, Guangxi, China.
| |
Collapse
|
50
|
Guo Z, Yao Z, Huang B, Wu D, Li Y, Chen X, Lu Y, Wang L, Lv W. MAFLD-related hepatocellular carcinoma: Exploring the potent combination of immunotherapy and molecular targeted therapy. Int Immunopharmacol 2024; 140:112821. [PMID: 39088919 DOI: 10.1016/j.intimp.2024.112821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
Hepatocellular carcinoma (HCC) is a common cause of cancer-related mortality and morbidity globally, and with the prevalence of metabolic-related diseases, the incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) related hepatocellular carcinoma (MAFLD-HCC) continues to rise with the limited efficacy of conventional treatments, which has created a major challenge for HCC surveillance. Immune checkpoint inhibitors (ICIs) and molecularly targeted drugs offer new hope for advanced MAFLD-HCC, but the evidence for the use of both types of therapy in this type of tumour is still insufficient. Theoretically, the combination of immunotherapy, which awakens the body's anti-tumour immunity, and targeted therapies, which directly block key molecular events driving malignant progression in HCC, is expected to produce synergistic effects. In this review, we will discuss the progress of immunotherapy and molecular targeted therapy in MAFLD-HCC and look forward to the opportunities and challenges of the combination therapy.
Collapse
Affiliation(s)
- Ziwei Guo
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ziang Yao
- Department of Traditional Chinese Medicine, Peking University People 's Hospital, Beijing 100044, China
| | - Bohao Huang
- Beijing University of Chinese Medicine, Beijing 100105, China
| | - Dongjie Wu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yanbo Li
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiaohan Chen
- Department of Hematology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yanping Lu
- Department of Hepatology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518100, China.
| | - Li Wang
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Wenliang Lv
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|