1
|
Bakrania A, Mo Y, Zheng G, Bhat M. RNA nanomedicine in liver diseases. Hepatology 2025; 81:1847-1877. [PMID: 37725757 PMCID: PMC12077345 DOI: 10.1097/hep.0000000000000606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023]
Abstract
The remarkable impact of RNA nanomedicine during the COVID-19 pandemic has demonstrated the expansive therapeutic potential of this field in diverse disease contexts. In recent years, RNA nanomedicine targeting the liver has been paradigm-shifting in the management of metabolic diseases such as hyperoxaluria and amyloidosis. RNA nanomedicine has significant potential in the management of liver diseases, where optimal management would benefit from targeted delivery, doses titrated to liver metabolism, and personalized therapy based on the specific site of interest. In this review, we discuss in-depth the different types of RNA and nanocarriers used for liver targeting along with their specific applications in metabolic dysfunction-associated steatotic liver disease, liver fibrosis, and liver cancers. We further highlight the strategies for cell-specific delivery and future perspectives in this field of research with the emergence of small activating RNA, circular RNA, and RNA base editing approaches.
Collapse
Affiliation(s)
- Anita Bakrania
- Department of Medicine, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Medicine, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yulin Mo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Mamatha Bhat
- Department of Medicine, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Medicine, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Division of Gastroenterology, University Health Network and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Firouzi-Farsani K, Dehghani-Samani M, Gerami R, Sadat Moosavi R, Gerami M, Mahdevar M. Exploring non-coding RNA expression profiles of AKR1B10P1, RP11-465B22.3, WASH8P, and NPM1P25 as a predictive model for hepatocellular carcinoma patient survival. Discov Oncol 2025; 16:771. [PMID: 40372570 PMCID: PMC12081793 DOI: 10.1007/s12672-025-02475-6] [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: 10/31/2024] [Accepted: 04/23/2025] [Indexed: 05/16/2025] Open
Abstract
The primary aim of the study was to analyze novel long non-coding RNAs (lncRNAs) in hepatocellular carcinoma (HCC) to assess their roles as potential oncogenes and tumor suppressors and to develop a survival prediction model based on their expression levels. Data from The Cancer Genome Atlas, GSE135631, and GSE214846, were utilized to evaluate changes in lncRNA expression in HCC and their associations with patient prognosis. A risk model was created based on lncRNA expression to predict patient mortality. The co-expression network was employed to identify associated pathways, and the results were subsequently validated using the RT-qPCR method. The findings indicated that 14 lncRNAs were down-regulated in HCC, and their increased expression was associated with a favorable prognosis. Additionally, eight lncRNAs were overexpressed and correlated with poorer patient outcomes. The multivariate Cox regression analysis demonstrated that overexpression of AKR1B10P1, RP11-465B22.3, WASH8P, and the downregulation of NPM1P25 could independently predict patient survival, irrespective of clinical variables. The risk score model based on these lncRNAs effectively stratified patients by their mortality risk. Furthermore, the co-expression network analysis revealed that the identified lncRNAs might be involved in various pathways, including fatty acid metabolism, mTOR signaling, glycolysis, angiogenesis, Wnt-β-catenin pathway, and DNA repair. RT-qPCR results validated the significant increase in the expression level of WASH8P in cancer specimens compared to normal tissues. Our results unveiled that changes in the expression levels of AKR1B10P1, RP11-465B22.3, WASH8P, and NPM1P25 were significantly and independently associated with prognosis. Moreover, the patient mortality risk model constructed using these four lncRNAs exhibited a robust capacity to accurately predict patients' survival rates.
Collapse
Affiliation(s)
| | - Mina Dehghani-Samani
- Department of Bioinformatics, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Razieh Gerami
- Independent Researcher, Graduated of PhD Pharmacology from Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Reihaneh Sadat Moosavi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Marzieh Gerami
- Department of Computer Engineering, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohammad Mahdevar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Genius Gene, Genetics and Biotechnology Company, Isfahan, Iran
| |
Collapse
|
3
|
Zwartsen A, Zeilmaker M, de Boer WJ, Rorije E, van der Voet H. Uncertainties in the Extrapolation of In Vitro Data in Human Risk Assessment: A Case Study of qIVIVE for Imazalil Using the Monte Carlo Risk Assessment Platform. Chem Res Toxicol 2025. [PMID: 40368340 DOI: 10.1021/acs.chemrestox.4c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
New approach methodologies (NAMs) are promising for refining, reducing, and replacing animal experiments for hazard characterization. Quantitative in vitro-in vivo extrapolation (qIVIVE) is essential to extrapolate an in vitro-based point of departure to an in vitro-based human equivalent dose and subsequently to an in vitro-based health-based guidance or threshold value. The use of NAMs for hazard characterization leads to the need for various new extrapolations and linked uncertainties that preferably are quantified. Currently, qIVIVE is often performed without addressing these uncertainties. A clear description and, if possible, quantification of extrapolations and uncertainties when using NAMs for risk assessment will aid the regulatory implementation of NAMs for risk assessment. A case study of a qIVIVE-based assessment on the risk of liver steatosis from dietary exposure to imazalil is reported, using a human cell line in vitro test method as an example of a NAM to replace animal experiments. We consider the uncertainties related to the extrapolations from in vitro to in vivo effects, from in vitro nominal concentrations to in vitro intracellular concentrations, from in vitro concentrations to external doses (reverse dosimetry), from in vitro exposure durations to in vivo exposure situations, and from the average human to a sensitive individual. The case study addresses these uncertainties in a mainly quantitative approach, using available data and the Monte Carlo Risk Assessment platform.
Collapse
Affiliation(s)
- Anne Zwartsen
- National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Marco Zeilmaker
- National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Waldo J de Boer
- Wageningen University & Research (WUR) Biometris, 6708 PB Wageningen, The Netherlands
| | - Emiel Rorije
- National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Hilko van der Voet
- Wageningen University & Research (WUR) Biometris, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
4
|
Kim YH, Kim JB, Bae JE, Park NY, Kim SH, Park D, So JH, Lee JM, Jeong K, Choi DK, Jo DS, Cho DH. ZLDI-8 facilitates pexophagy by ROS-mediated activation of TFEB and ATM in HeLa cells. Bioorg Med Chem Lett 2025; 120:130130. [PMID: 39923905 DOI: 10.1016/j.bmcl.2025.130130] [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: 12/16/2024] [Revised: 01/18/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Autophagy-mediated organelle quality control is vital for cellular homeostasis. However, the mechanisms underlying selective autophagy of peroxisomes, known as pexophagy, are less well understood than those of other organelles, such as mitochondria. In this study, we screened a phosphatase inhibitor library using a cell-based system and identified several potent pexophagy inducers, including ZLDI-8, a known inhibitor of lymphoid-specific tyrosine phosphatase. Notably, treatment with ZLDI-8 selectively induces the loss of peroxisomes without affecting other organelles, such as mitochondria, the endoplasmic reticulum, or the Golgi apparatus. The peroxisome loss induced by ZLDI-8 was significantly blocked in ATG5-knockout HeLa cells, confirming its dependence on autophagy. We further found that ZLDI-8 treatment increases both cellular and peroxisomal reactive oxygen species (ROS), which were effectively scavenged by N-acetylcysteine (NAC). The increase in peroxisomal ROS leads to the activation of ATM kinase and the dephosphorylation of TFEB. Moreover, ROS scavenging prevents all of these processes. Taken together, these findings demonstrate that ZLDI-8 induces pexophagy through a mechanism involving peroxisomal ROS-mediated activation of TFEB and ATM. This study provides valuable insights into the molecular mechanisms regulating selective peroxisome degradation and potential therapeutic strategies for targeting pexophagy.
Collapse
Affiliation(s)
- Yong Hwan Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu 41566 Republic of Korea
| | - Joon Bum Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu 41566 Republic of Korea; Organelle Institute, Kyungpook National University, Daegu 41566 Republic of Korea
| | - Ji-Eun Bae
- Organelle Institute, Kyungpook National University, Daegu 41566 Republic of Korea
| | - Na Yeon Park
- Organelle Institute, Kyungpook National University, Daegu 41566 Republic of Korea
| | - Seong Hyun Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu 41566 Republic of Korea
| | - Daeun Park
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu 41566 Republic of Korea
| | - Jun Hee So
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu 41566 Republic of Korea
| | - Jae Man Lee
- Organelle Institute, Kyungpook National University, Daegu 41566 Republic of Korea; Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 41944 Republic of Korea
| | - Kwiwan Jeong
- Bio Industry Department, Gyeonggido Business & Science Accelerator, Suwon 16229 Republic of Korea
| | - Dong Kyu Choi
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu 41566 Republic of Korea
| | - Doo Sin Jo
- ORGASIS Corp. 260, Changyong-daero, Yongtong-gu, Suwon 08826 Republic of Korea.
| | - Dong-Hyung Cho
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu 41566 Republic of Korea; Organelle Institute, Kyungpook National University, Daegu 41566 Republic of Korea; ORGASIS Corp. 260, Changyong-daero, Yongtong-gu, Suwon 08826 Republic of Korea.
| |
Collapse
|
5
|
Yu M, Chen S, Liu X, Dong H, Wang DC. The impact of vitamin D supplementation on glycemic control and lipid metabolism in polycystic ovary syndrome: a systematic review of randomized controlled trials. BMC Endocr Disord 2025; 25:110. [PMID: 40259331 PMCID: PMC12010551 DOI: 10.1186/s12902-025-01920-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/01/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a prevalent endocrine condition affecting both metabolic and reproductive health in women. The impact of vitamin D on metabolic regulation has attracted growing interest. The purpose of this study is to investigate the impact of vitamin D supplementation on key metabolic parameters-namely blood glucose, insulin, and lipid levels-in individuals with PCOS. METHODS A systematic review was conducted to identify relevant studies in PubMed, Embase, Cochrane Library, Web of Science, and ClinicalTrials.gov. The search focused on randomized controlled trials (RCTs) evaluating the impact of vitamin D supplementation in patients with PCOS. Meta-analysis was performed using RevMan 5.3 software, and study quality was evaluated with the Cochrane Risk of Bias Tool. In addition, outcome-related evidence was graded using the GRADE system, and TSA was performed to determine if the number of participants met the required threshold. RESULTS A total of 691 individuals with PCOS from 13 RCTs were evaluated. The meta-analysis indicated that the supplementation of vitamin D led to a notable reduction in the subsequent metabolic parameters: fasting blood glucose[MD=-2.91 mg/dL, 95% CI (-4.78, -1.04) mg/dL, P = 0.002], insulin levels[MD=-1.98 µIU/mL, 95% CI (-3.32, -0.64) µIU/mL, P = 0.004], triglycerides[MD=-11.01 mg/dL, 95% CI (-16.42, -5.61) mg/dL, P < 0.0001], total cholesterol [MD=-11.69 mg/dL, 95% CI (-15.56, -7.82) mg/dL, P < 0.00001], very low-density lipoprotein cholesterol (VLDL-cholesterol) [MD=-2.64 mg/dL, 95% CI (-4.50, -0.79) mg/dL, P = 0.005], and low-density lipoprotein cholesterol (LDL-cholesterol) [MD=-5.85 mg/dL, 95% CI (-10.28, -1.42) mg/dL, P = 0.010]. Nevertheless, the supplementation of vitamin D did not exert a significant impact on high - density lipoprotein cholesterol (HDL - cholesterol) [MD=-0.21 mg/dL, 95% CI (-0.81, 1.22) mg/dL, P = 0.69]. Begg's and Egger's tests suggested a minimal probability of publication bias, and the TSA confirmed that the optimal sample size for major outcomes had been reached, supporting the robustness of the meta-analysis results. CONCLUSION Vitamin D supplementation shows significant benefits in improving metabolic parameters in PCOS patients, particularly in reducing fasting blood glucose, insulin, and lipid levels, suggesting a potential role of vitamin D in PCOS management. The long-term outcomes and most effective dose of vitamin D warrant further investigation in future research. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Miao Yu
- Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, 643000, China
| | - Shuai Chen
- Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, 643000, China
| | - Xia Liu
- Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, 643000, China
| | - Hui Dong
- Department of General Surgery, Zigong Fourth People's Hospital, 19 Tanmulin Road, Zigong, Sichuan, 643000, P.R. China
| | - Deng-Chao Wang
- Department of General Surgery, Zigong Fourth People's Hospital, 19 Tanmulin Road, Zigong, Sichuan, 643000, P.R. China.
| |
Collapse
|
6
|
Joerg R, Itariu BK, Amor M, Bilban M, Langer F, Prager G, Joerg F, Stulnig TM. The effect of long-chain n-3 PUFA on liver transcriptome in human obesity. Prostaglandins Leukot Essent Fatty Acids 2025; 204:102663. [PMID: 39752839 DOI: 10.1016/j.plefa.2024.102663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND AIMS Obesity is associated with a higher risk of severe diseases such as atherosclerotic cardiovascular disease, type 2 diabetes mellitus (T2DM), and metabolic dysfunction-associated steatotic liver disease (MASLD). Polyunsaturated fatty acids, of the omega-3 family (n-3 PUFA), have been shown to reduce adipose tissue inflammation in obesity, as well as to have lipid-lowering effects and improve insulin sensitivity. However, direct effects on liver transcriptome in humans have not been described. Our aim was to understand the impact of n-3 PUFA on gene expression in obese human liver. APPROACH AND RESULTS Patients with obesity (BMI ≥ 40 kg/m2) were treated for eight weeks with 3.36 g n-3 PUFAs (1.84 g eicosapentaenoic acid (EPA) and 1.53 g docosahexaenoic acid (DHA)), or with 5 g of butter as a control (n = 15 per group) before undergoing bariatric surgery where liver biopsies were taken. Liver samples were used for mRNA microarray analyses and subsequently Gene Set Enrichment Analysis (GSEA) was performed. This bioinformatic approach led us to identify 80 significantly dysregulated pathways that were divided into 9 different clusters including insulin and lipid metabolism, and immunity. N-3 PUFA treatment significantly affected pathways related to immunity, metabolism, and inflammation. Specifically, it upregulated pathways involved in T-cell and B-cell functions and lipid metabolism, while downregulating glucagon signalling. These findings highlight the impact of n-3 PUFAs on key metabolic and immune processes in the liver of patients with obesity. CONCLUSION This study provides further insights into the impact on n-3 PUFA on human liver gene expression, particularly in pathways associated with immunity, lipid metabolism, and inflammation, setting basis for further clinical research. SUMMARY Obesity increases the risk of diseases like atherosclerotic- cardiovascular disease, type 2 diabetes mellitus and metabolic dysfunction-associated steatotic liver disease (MASLD). Omega-3 polyunsaturated fatty acids (n-3 PUFA) are known for their anti-inflammatory and metabolic benefits, but their direct impact on liver gene expression in people with obesity, remains unclear. In this study, patients with obesity (BMI ≥ 40 kg/m2) were administered either n-3 PUFAs or butter before bariatric surgery. Liver biopsies were analysed for gene expression via Gene Set Enrichment Analysis (GSEA). The results revealed 80 dysregulated pathways across 9 clusters, including those related to insulin and lipid metabolism, and immunity. This sheds light on how n-3 PUFAs influence gene expression in the liver of patients with obesity, setting the groundwork for further clinical exploration.
Collapse
Affiliation(s)
- Rebeka Joerg
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Austria.
| | - Bianca K Itariu
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Austria; Metabolism Centre N12 Antonigasse, 1090 Vienna, Austria.
| | - Melina Amor
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Austria.
| | - Martin Bilban
- Department of Genomics, Medical University of Vienna, Austria.
| | - Felix Langer
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Austria.
| | - Gerhard Prager
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Austria.
| | - Florian Joerg
- Department of Computational Biological Chemistry, Faculty of Chemistry, University of Vienna, Austria.
| | - Thomas M Stulnig
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Austria; Department of Medicine III and Karl Landsteiner Institute for Metabolic Diseases and Nephrology, Clinic Hietzing, Vienna, Austria.
| |
Collapse
|
7
|
Wang X, Pan L, Niu D, Zhou J, Shen M, Zeng Z, Gong W, Yang E, Tang Y, Cheng G, Sun C. Jingfang Granules alleviates the lipid peroxidation induced ferroptosis in rheumatoid arthritis rats by regulating gut microbiota and metabolism of short chain fatty acids. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119160. [PMID: 39608616 DOI: 10.1016/j.jep.2024.119160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, bone and cartilage damage, musculoskeletal pain, swelling, and stiffness. Inflammation is one of the key factors that induce RA. Jingfang Granule (JFG) is a traditional Chinese medicine (TCM) with significant anti-inflammatory effects. Clinical studies have confirmed that JFG can be used to treat RA, but the mechanism is still vague. PURPOSE This study was designed to evaluate the protective function and the mechanism of JFG on rats with RA. STUDY DESIGN AND METHODS Complete Freud's Adjuvant (CFA) was used to establish a rat RA model, and JFG or Diclofenac Sodium (Dic) was orally administered. Foot swelling and hematoxylin eosin (H&E) staining were used to test the therapeutic effect of JFG on RA treatment, while ELISA kits were used to detect serum cytokines. Malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), catalase (CAT), and reactive oxygen species (ROS) were used to evaluate oxidative stress levels. The integration of label-free proteomics, fecal short chain fatty acid (SCFA) targeted metabolomics, peripheral blood SCFA, medium and long chain fatty acid targeted metabolomics, and 16S rDNA sequencing of gut microbiota were used to screen the mechanism. Western blot technology was used to validate the results of multiple omics studies. Serum D-Lactic acid, lipopolysaccharide specific IgA antibody (LPS IgA), diamine oxidase (DAO), and colon Claudin 5 and ZO-1 were used to evaluate the intestinal barrier. RESULTS The results confirmed that JFG effectively protected rats from RA injury, which was confirmed by improved foot swelling and synovial pathology. At the same time, JFG reduced the levels of TNF-α, IL-1β, and IL-6 in serum by inhibiting the NLRP3 inflammasome signaling pathway and TLR4/NF-κB signaling pathway in synovial tissue. Multiple omics studies indicated that JFG increased the abundance of gut microbiota and regulated the number of gut bacteria, thereby increased the levels of Acetic acid, Propionic acid, and Butyric acid in the gut and serum of RA rats, which activated AMPK to regulate fatty acid metabolism and fatty acid biosynthesis, thereby inhibited lipid oxidative stress induced ferroptosis to improve tissue damage caused by RA. Meanwhile, JFG improved the intestinal barrier by upregulating the expresses of Claudin 5 and ZO-1, which was confirmed by low concentrations of D-Lactic acid, LPS-SIgA and DAO in serum. CONCLUSIONS This study confirmed that JFG improved the disturbance of fatty acid metabolism by modulating gut microbiota and the production of fecal SCFAs to activate AMPK, and then inhibited ferroptosis caused by lipid oxidative stress in synovium tissue and prevented AR injury. This study proposes for the first time to investigate the mechanism of JFG treatment for RA from the perspective of the "Gut-joint" axis, and provides a promising approach for the treatment of RA.
Collapse
Affiliation(s)
- Xiuwen Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jidong Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Wenqiao Gong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Enhua Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Yunfeng Tang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Guoliang Cheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Chenghong Sun
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, China.
| |
Collapse
|
8
|
Sun J, Jin X, Li Y. OTUD7B inhibited hepatic injury from NAFLD by inhibiting K48-linked ubiquitination and degradation of β-catenin. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167555. [PMID: 39520879 DOI: 10.1016/j.bbadis.2024.167555] [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: 06/07/2024] [Revised: 09/27/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the prevalent liver disease. Ovarian tumor domain-containing 7B (OTUD7B) is a deubiquitinating enzyme and its role in NAFLD remains unclear. In high-fat diet (HFD)-induced NAFLD mouse model and palmitic acid (PA)-treated HepG2 cells, OTUD7B expression was decreased. Adenoviral overexpression of OTUD7B in mice resulted in reduced body weight and liver injury, with decreased serum aminotransferase (ALT) and aspartate aminotransferase (AST) levels. OTUD7B overexpression attenuated hepatic lipid deposition (serum TG, TC, LDL-C, HDLC, and FFA levels), which might be through the suppression of lipogenesis and β-oxidation-related genes. The contents of hepatic inflammatory factors (TNF-α, IL-6, and IL-1β) were decreased following OTUD7B overexpression in NAFLD mice. A mechanism study indicated that the protective effect of OTUD7B overexpression might be associated with β-catenin signal activation. OTUD7B overexpression promoted PA-induced β-catenin activity in TopFlash assay, and increased total β-catenin and c-myc levels in cells. The increase in β-catenin levels was contributed to the stabilization via inhibiting K48-linked ubiquitination and proteasomal degradation by OTUD7B. NR4A2 role in NASH has been proved. NR4A2 ChIP-seq and RNA-seq data excluded transcriptional regulation of NR4A2 to OTUD7B, and it was experimentally evidenced that NR4A2 bound to OTUD7B promoter region and positively transcriptionally regulate OTUD7B expression. In summary, OTUD7B overexpression ameliorated hepatic inflammation and steatosis in NAFLD. The possible mechanism of OTUD7B might be through the inhibition of β-catenin degradation by removing K48-linked ubiquitination, which might be regulated by NR4A2.
Collapse
Affiliation(s)
- Jing Sun
- Department of Gastroenterology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiuli Jin
- Department of Gastroenterology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yiling Li
- Department of Gastroenterology, the First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
9
|
Tang M, Wei X, Ma Y, Tan Y, Cao H, Yao S, Wang J, Yang H, Liu F, Peng Y, Fan N. USP13 ameliorates metabolic dysfunction-associated steatohepatitis through targeting PTEN. Life Sci 2025; 360:123264. [PMID: 39571890 DOI: 10.1016/j.lfs.2024.123264] [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: 09/22/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
OBJECTIVE The role of ubiquitin-specific protease 13 (USP13) in metabolic dysfunction-associated steatohepatitis (MASH) remains unclear. This study aimed to elucidate the role of USP13 in MASH progression. METHODS THLE-2 cells were subjected to palmitate acid (PA) to generate an in vitro model of lipid accumulation and inflammation. Two in vivo models of MASH were established by feeding mice with a high-fat, high-fructose and high-cholesterol (HFFC) diet for 16 weeks and a methionine/choline-deficient diet (MCD) for 4 weeks, respectively. Usp13 overexpression and knockout (KO) techniques were employed to investigate its role in MASH. RESULTS USP13 expression was significantly downregulated in the livers of MASH mice and in the in vitro model of lipid accumulation and inflammation. Hepatic overexpression of Usp13 markedly alleviated liver steatosis, inflammation and fibrosis, while knockout of Usp13 exacerbated the MASH phenotype. Mechanistically, USP13 directly bound to phosphatase and tensin homolog (PTEN) and deubiquitinated it, thereby elevating the PTEN expression and improving the MASH phenotype. Notably, Pten overexpression in Usp13 knockout mice reversed the exacerbation of MASH brought from Usp13 deficiency. CONCLUSIONS Our findings revealed that USP13 alleviates MASH by directly binding to and deubiquitinating PTEN. The USP13-PTEN axis may represent a promising molecular target for MASH treatment.
Collapse
Affiliation(s)
- Min Tang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Endocrinology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaohui Wei
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunqin Ma
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yijiong Tan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Cao
- Department of Endocrinology and Metabolism, Shanghai General Hospital of Nanjing Medical University, Shanghai, China; Department of Endocrinology, Songjiang District Central Hospital, Shanghai, China
| | - Shuangshuang Yao
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Yang
- Department of Endocrinology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fang Liu
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongde Peng
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Endocrinology and Metabolism, Shanghai General Hospital of Nanjing Medical University, Shanghai, China.
| | - Nengguang Fan
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Guo F, Zheng H, Cao H, Wang Y, Zhi Y, Liu H, Li B, Wu J, Zhang K, Gao Y. Bergenin inhibits hepatic fat deposition by activating the AMPK signaling pathway, thereby attenuating alcoholic liver disease. Int Immunopharmacol 2024; 142:113169. [PMID: 39298826 DOI: 10.1016/j.intimp.2024.113169] [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: 08/05/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Alcoholic liver disease (ALD) is a prevalent liver condition that arises from prolonged and excessive alcohol intake. Bergenin (BER) is an effective phytotherapeutic agent that exhibits pharmacological properties, including anti-inflammatory and anti-oxidative effects. To establish an in vivo model of ALD, C57BL/6 mice were continuously fed a high-fat diet (HFD) and administered alcohol gavage for 8 weeks, while concurrently administering BER and evaluated for therapeutic effects. After modeling, the therapeutic effects of BER were evaluated by observing histopathological changes and the detection of relevant biochemical indicators in mice. In addition, RNA sequencing of liver tissues was performed to analyze differentially expressed genes and to investigate the associated signaling pathways in order to elucidate the protective mechanisms of BER. These differentially expressed genes were mainly enriched in lipid metabolism pathways and the cytochrome P450 metabolism of exogenous substances. Subsequently, HepG2 was co-treated with sodium oleate (NaOA) and ethanol to establish an in vitro model, and the specific mechanism by which BER ameliorates ALD was further analyzed in depth. AMPK inhibitor, Compound C (CC), was demonstrated to significantly inhibit the regulation of lipid metabolism by BER in vitro. Finally, the differentially expressed genes selected were validated through qRT-PCR and Western blot analysis. Collectively, our findings revealed that BER effectively alleviated liver injury caused by alcohol and HFD in mice, significantly suppressing lipid deposition in ALD, enhancing alcohol metabolism, and mitigating oxidative stress.
Collapse
Affiliation(s)
- Fengyue Guo
- Key Laboratory of Pharmacology for Prevention and Treatment of High Incidence Diseases in Guangxi Higher Education Institutions, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Huaping Zheng
- Affiliated Hospital of Guilin Medical University , Guilin Medical University, Guilin 541001, Guangxi, China
| | - Houkang Cao
- Key Laboratory of Pharmacology for Prevention and Treatment of High Incidence Diseases in Guangxi Higher Education Institutions, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Yongwang Wang
- Affiliated Hospital of Guilin Medical University , Guilin Medical University, Guilin 541001, Guangxi, China
| | - Yueping Zhi
- Key Laboratory of Pharmacology for Prevention and Treatment of High Incidence Diseases in Guangxi Higher Education Institutions, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Haiping Liu
- Key Laboratory of Pharmacology for Prevention and Treatment of High Incidence Diseases in Guangxi Higher Education Institutions, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Bo Li
- Key Laboratory of Pharmacology for Prevention and Treatment of High Incidence Diseases in Guangxi Higher Education Institutions, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Jianzhao Wu
- Key Laboratory of Pharmacology for Prevention and Treatment of High Incidence Diseases in Guangxi Higher Education Institutions, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Kefeng Zhang
- Key Laboratory of Pharmacology for Prevention and Treatment of High Incidence Diseases in Guangxi Higher Education Institutions, Guilin Medical University, Guilin 541199, Guangxi, China.
| | - Ya Gao
- Key Laboratory of Pharmacology for Prevention and Treatment of High Incidence Diseases in Guangxi Higher Education Institutions, Guilin Medical University, Guilin 541199, Guangxi, China.
| |
Collapse
|
11
|
Kuo HN, LaRussa Z, Xu FM, Consitt LA, Liu M, Davidson WS, Puri V, Coschigano KT, Shi H, Lo CC. Attenuation of high-fat diet-induced weight gain by apolipoprotein A4. Obesity (Silver Spring) 2024; 32:2321-2333. [PMID: 39540259 PMCID: PMC11589529 DOI: 10.1002/oby.24155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Apolipoprotein A4 (APOA4) is synthesized by the small intestine in response to dietary lipids. Chronic exposure to a high-fat diet (HFD) desensitizes lipid-induced APOA4 production and attenuates brown adipose tissue (BAT) thermogenesis. We hypothesized that exogenous APOA4 could increase BAT thermogenesis and energy expenditure in HFD-fed mice, resulting in decreased obesity and improved glucose tolerance. METHODS BAT and inguinal white adipose tissue (IWAT) thermogenesis, body composition, energy intake and expenditure, and locomotor activity were measured using an infrared camera, immunoblots, quantitative magnetic resonance imaging, and a comprehensive lab animal monitoring system. An intraperitoneal glucose tolerance test and hepatic lipid accumulation and steatosis were assayed. RESULTS Mice receiving continuous infusion of APOA4 for the last 4 weeks of 10 weeks of HFD feeding gained no additional body weight and had reduced fat mass but enhanced BAT and IWAT thermogenesis and energy expenditure, despite unaltered food intake and locomotor activity. Additionally, APOA4 infusion elevated fatty acid β oxidation; decreased lipogenesis, lipid accumulation, and steatosis in liver; and improved glucose tolerance. CONCLUSIONS Maintenance of plasma APOA4 via exogenous APOA4 protein parallels elevation of BAT and IWAT thermogenesis, hepatic fatty acid β oxidation, and overall energy expenditure, with subsequent prevention of additional weight gain in HFD-fed obese mice.
Collapse
Affiliation(s)
- Hsuan‐Chih N. Kuo
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
- Department of Biological SciencesOhio UniversityAthensOhioUSA
| | - Zachary LaRussa
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
- Department of Biological SciencesOhio UniversityAthensOhioUSA
| | | | - Leslie A. Consitt
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
| | - Min Liu
- Department of Pathology and Laboratory MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - W. Sean Davidson
- Department of Pathology and Laboratory MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Vishwajeet Puri
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
| | - Karen T. Coschigano
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
| | - Haifei Shi
- Department of BiologyMiami UniversityOxfordOhioUSA
| | - Chunmin C. Lo
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
- Department of Biological SciencesOhio UniversityAthensOhioUSA
| |
Collapse
|
12
|
Hong Y, Chen X, Yuan H, Huang Z, Tao S, Xie F, Xie W. Novel non-HDLc/HDLc ratio for predicting MASLD: a cross-sectional study in a Chinese health screening population. BMC Gastroenterol 2024; 24:439. [PMID: 39609681 PMCID: PMC11603918 DOI: 10.1186/s12876-024-03525-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Previous studies have shown a positive correlation between the non-high-density lipoprotein cholesterol/high-density lipoprotein cholesterol(non-HDLc/HDLc ratio) and metabolism-associated steatohepatitis (MASLD), suggesting it is a superior predictor of MASLD.The aim of this study is to assess the predictive value of the non-HDLc/HDLc ratio for MASLD. METHODS In this study, we retrospectively analyzed data from 4,498 adult patients at the Health Management Centre of Guangdong Provincial Hospital of Integrative Medicine, regardless of gender, and the diagnostic criteria for MASLD were derived from guidelines. We used univariate and multivariate logistic regression to verify the relationship between the non-HDLc/HDLc ratio and MASLD and assessed the stability of the results through interaction tests. ROC curve analysis was then employed to compare the diagnostic efficacy of several lipid indices, including the non-HDLc/HDLc ratio, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), triglycerides (TG), total cholesterol (TC), and the composite lipid profile in diagnosing MASLD. RESULTS These data suggested that non-HDLc/HDLc, LDL-C, TG, and TC were positively correlated with MASLD, while HDL-C was inversely correlated, even after adjusting for confounders such as gender, age, body mass index, ALT, AST, uric acid, blood glucose, and hypertension. Among them, non-HDLc/HDLc ratio(OR: 2.709, 95% CI: 2.316-3.169, p < 0.001), LDL-C (OR: 1.294, 95% CI: 1.125-1.489, p < 0.001), TG (OR: 2.854, 95% CI: 2.473-3.293, p < 0.001), TC (OR: 1.242, 95% CI: 1.122-1.374, p < 0.001), and HDL-C (OR: 0.074, 95% CI: 0.044-0.123, p < 0.001) were identified. The interaction results showed that gender did not affect lipid levels in MASLD (p > 0.05). ROC analysis confirmed the validity of the non-HDL cholesterol/HDL cholesterol ratio in predicting MASLD incidence, with an AUC of 0.801. Moreover, the composite lipid indices demonstrated greater predictive power for MASLD compared to individual indices (AUC: 0.857), and validation set results were consistent with those of the training set. CONCLUSION The present study revealed that the non-HDLc/HDLc ratio was positively correlated with the development of MASLD, and the ratio was also effective in predicting MASLD.
Collapse
Affiliation(s)
- Yan Hong
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong Province, China
| | - Xinrong Chen
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Hangtao Yuan
- Affiliated Dongfang Hospital of Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Fengtai District, Beijing, China
| | - Zixuan Huang
- Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Shaohong Tao
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong Province, China
| | - Fang Xie
- Department of Liver Disease, Jinling Hospital affiliated to Medical College of Nanjing University, Nanjing, Jiangsu Province, China
| | - Weining Xie
- Infectious Disease Department, Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, Guangdong Province, China.
| |
Collapse
|
13
|
Geng X, Zhuang M, Tian W, Shang H, Gong Z, Lv Y, Li J. Green Radish Polysaccharide Prevents Alcoholic Liver Injury by Interfering with Intestinal Bacteria and Short-Chain Fatty Acids in Mice. Foods 2024; 13:3733. [PMID: 39682806 DOI: 10.3390/foods13233733] [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: 09/24/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
This study aimed to ascertain the potential benefits of green radish polysaccharide (GRP) in treating alcoholic liver disease (ALD) in mice and explore its mechanism of action. Using biochemical analysis, high-throughput sequencing of gut microbiota, and gas chromatography-mass spectrometry to measure short-chain fatty acids (SCFAs) in feces, we found that GRP intervention significantly improved lipid metabolism and hepatic function in mice subjected to excessive alcohol intake. The GRP intervention reduced malondialdehyde levels by 66% and increased total superoxide dismutase levels by 22%, thereby mitigating alcohol-induced oxidative stress. Furthermore, GRP intervention in mice with alcohol consumption resulted in a reduction in tumor necrosis factor, interleukin 6, and lipopolysaccharide levels by 12%, 9%, and 25%, respectively, effectively attenuating alcoholic liver inflammation. 16S rRNA amplicon sequencing demonstrated that excessive alcohol consumption markedly altered the gut microbiota composition in mice. The GRP treatment resulted in a significant reduction in the number of beneficial bacteria (Lactobacillus and Lachnospiraceae_NK4A136_group) and an increase in the proportion of harmful bacteria (Muribaculaceae and Verrucomicrobiota). The metabolomic analyses of the SCFAs demonstrated an increase in the contents of SCFAs, acetic acid, propionic acid, and butyric acid, following GRP supplementation. Furthermore, the metabolic levels of cholinergic synapses and glycolysis/gluconeogenesis were found to be modulated. In conclusion, these findings suggest that GRP may attenuate alcohol-induced oxidative damage in the liver by modulating the gut microbiota and hepatic metabolic pathways. This may position GRP as a potential functional component for ALD prevention.
Collapse
Affiliation(s)
- Xiong Geng
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Miaomiao Zhuang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Weina Tian
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Huayan Shang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Ziyi Gong
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Yanfang Lv
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Jianrong Li
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| |
Collapse
|
14
|
Teka T, Wu J, Oduro PK, Li Z, Wang C, Chen H, Zhang L, Wang H, Wang L, Han L. Integrated multi-omics analyses combined with western blotting discovered that cis-TSG alleviated liver injury via modulating lipid metabolism. Front Pharmacol 2024; 15:1485035. [PMID: 39635428 PMCID: PMC11614611 DOI: 10.3389/fphar.2024.1485035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Background: Polygonum multiflorum shows dual hepatoprotective and hepatotoxic effects. The bioactive components responsible for these effects are unknown. This study investigates whether cis-2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside (cis-TSG), a stilbene glycoside, has hepatoprotective and/or hepatotoxic effects in a liver injury model. Methods: C57BL/6J mice were administered α-naphthylisothiocyanate (ANIT) to induce cholestasis, followed by treatment with cis-TSG. Hepatoprotective and hepatotoxic effects were assessed using serum biomarkers, liver histology, and metabolomic and lipidomic profiling. Transcriptomic analysis were conducted to explore gene expression changes associated with lipid and bile acid metabolism, inflammation, and oxidative stress. Results and Discussion: ANIT administration caused significant liver injury, evident from elevated alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and dysregulated lipid metabolism. cis-TSG treatment markedly reduced ALT and AST levels, normalized lipid profiles, and ameliorated liver damage, as seen histologically. Metabolomic and lipidomic analyses revealed that cis-TSG influenced key pathways, notably glycerophospholipid metabolism, sphingolipid metabolism, and bile acid biosynthesis. The treatment with cis-TSG increased monounsaturated and polyunsaturated fatty acids (MUFAs and PUFAs), enhancing peroxisome proliferator-activated receptor alpha (PPARα) activity. Transcriptomic data confirmed these findings, showing the downregulation of genes linked to lipid metabolism, inflammation, and oxidative stress in the cis-TSG-treated group. The findings suggest that cis-TSG has a hepatoprotective effect through modulation of lipid metabolism and PPARα activation.
Collapse
Affiliation(s)
- Tekleab Teka
- State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Pharmacy, Wollo University, Dessie, Ethiopia
| | - Jiang Wu
- Shenzhen Technology University, Shenzhen, China
| | - Patrick Kwabena Oduro
- Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, NY, United States
| | - Ze Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chenxi Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hao Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haitao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Liming Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Lifeng Han
- State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| |
Collapse
|
15
|
Heusinkveld HJ, Zwart EP, de Haan A, Braeuning A, Alarcan J, van der Ven LTM. The zebrafish embryo as a model for chemically-induced steatosis: A case study with three pesticides. Toxicology 2024; 508:153927. [PMID: 39151607 DOI: 10.1016/j.tox.2024.153927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
There is an increasing incidence and prevalence of fatty liver disease in the western world, with steatosis as the most prevalent variant. Known causes of steatosis include exposure to food-borne chemicals, and overconsumption of alcohol, carbohydrates and fat, and it is a well-known side effect of certain pharmaceuticals such as tetracycline, amiodarone and tamoxifen (drug-induced hepatic steatosis). Mechanistic knowledge on chemical-induced steatosis has greatly evolved and has been organized into adverse outcome pathways (AOPs) describing the chain of events from first molecular interaction of a substance with a biological system to the adverse outcome, intrahepatic lipid accumulation. In this study, three known steatosis-inducing pesticides (imazalil, clothianidin, and thiacloprid) were tested for their ability to induce hepatic triglyceride accumulation in the zebrafish (Danio rerio) embryo (ZFE) at 5 days post fertilization, both as single compounds and equipotent binary mixtures. The results indicate that the ZFE is very well applicable as a higher tier testing model to confirm effects in downstream key events in AOPs, that is, chemically-induced triglyceride accumulation in the whole organism and production of visible steatosis. Moreover, dose addition could be concluded for binary mixtures of substances with similar and with dissimilar modes of action.
Collapse
Affiliation(s)
- Harm J Heusinkveld
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands.
| | - Edwin P Zwart
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Angela de Haan
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, Berlin 10589, Germany
| | - Jimmy Alarcan
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, Berlin 10589, Germany
| | - Leo T M van der Ven
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| |
Collapse
|
16
|
Feng X, Wang M, Wen S, Hu L, Lan Y, Xu H. Lactiplantibacillus plantarum P101 Alleviated Alcohol-Induced Hepatic Lipid Accumulation in Mice via AMPK Signaling Pathway: Gut Microbiota and Metabolomics Analysis. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10373-6. [PMID: 39388022 DOI: 10.1007/s12602-024-10373-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Mitigating steatosis is essential for delaying the progression of alcoholic liver disease. The effect and mechanism of Lactiplantibacillus plantarum P101 (LP.P101) on alleviating alcohol-induced hepatic lipid accumulation were investigated in our study. The mouse model was constructed by a short-term (10-day)-plus-binge ethanol feeding and gavaged with 108 CFU/mL of LP.P101 daily. Lipid droplet in the liver was significantly reduced by LP.101 intervention on AMPK activation. However, when AMPK was inhibited by dorsomorphin, the levels of related indicators (ALT, TG, etc.) and the expression levels of AMPK and relevant genes in the liver converged to that of the alcohol-fed group. Compared with the alcohol-fed group, LP.P101 reduced the relative abundance of Firmicutes and increased that of Bacteroidetes. Parabacteroides merdae was negatively correlated with lipid accumulation, and unclassified Negativibacillus was negatively associated with AMPK activation. Importantly, LP.P101 modified the compositions of the serum metabolites. The potential biomarker stercobilinogen was positively correlated with AMPK activation and negatively associated with lipid accumulation. This work confirmed that LP.P101 attenuated alcohol-induced hepatic lipid accumulation in mice through AMPK activation, and the alterations in gut microbiota and metabolites may play a significant role on AMPK activation.
Collapse
Affiliation(s)
- Xiaoyan Feng
- State Key Laboratory of Food Science and Resource, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, P. R. China
| | - Mengqi Wang
- State Key Laboratory of Food Science and Resource, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, P. R. China
| | - Siyue Wen
- State Key Laboratory of Food Science and Resource, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, P. R. China
| | - Liehai Hu
- State Key Laboratory of Food Science and Resource, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, P. R. China
| | - Yuzhi Lan
- State Key Laboratory of Food Science and Resource, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, P. R. China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resource, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, P. R. China.
- International Institute of Food Innovation Co., Ltd., Nanchang University, Nanchang, 330200, P. R. China.
| |
Collapse
|
17
|
Giangregorio F, Mosconi E, Debellis MG, Provini S, Esposito C, Garolfi M, Oraka S, Kaloudi O, Mustafazade G, Marín-Baselga R, Tung-Chen Y. A Systematic Review of Metabolic Syndrome: Key Correlated Pathologies and Non-Invasive Diagnostic Approaches. J Clin Med 2024; 13:5880. [PMID: 39407941 PMCID: PMC11478146 DOI: 10.3390/jcm13195880] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background and Objectives: Metabolic syndrome (MetS) is a condition marked by a complex array of physiological, biochemical, and metabolic abnormalities, including central obesity, insulin resistance, high blood pressure, and dyslipidemia (characterized by elevated triglycerides and reduced levels of high-density lipoproteins). The pathogenesis develops from the accumulation of lipid droplets in the hepatocyte (steatosis). This accumulation, in genetically predisposed subjects and with other external stimuli (intestinal dysbiosis, high caloric diet, physical inactivity, stress), activates the production of pro-inflammatory molecules, alter autophagy, and turn on the activity of hepatic stellate cells (HSCs), provoking the low grade chronic inflammation and the fibrosis. This syndrome is associated with a significantly increased risk of developing type 2 diabetes mellitus (T2D), cardiovascular diseases (CVD), vascular, renal, pneumologic, rheumatological, sexual, cutaneous syndromes and overall mortality, with the risk rising five- to seven-fold for T2DM, three-fold for CVD, and one and a half-fold for all-cause mortality. The purpose of this narrative review is to examine metabolic syndrome as a "systemic disease" and its interaction with major internal medicine conditions such as CVD, diabetes, renal failure, and respiratory failure. It is essential for internal medicine practitioners to approach this widespread condition in a "holistic" rather than a fragmented manner, particularly in Western countries. Additionally, it is important to be aware of the non-invasive tools available for assessing this condition. Materials and Methods: We conducted an exhaustive search on PubMed up to July 2024, focusing on terms related to metabolic syndrome and other pathologies (heart, Lung (COPD, asthma, pulmonary hypertension, OSAS) and kidney failure, vascular, rheumatological (osteoarthritis, rheumatoid arthritis), endocrinological, sexual pathologies and neoplastic risks. The review was managed in accordance with the PRISMA statement. Finally, we selected 300 studies (233 papers for the first search strategy and 67 for the second one). Our review included studies that provided insights into metabolic syndrome and non-invasive techniques for evaluating liver fibrosis and steatosis. Studies that were not conducted on humans, were published in languages other than English, or did not assess changes related to heart failure were excluded. Results: The findings revealed a clear correlation between metabolic syndrome and all the pathologies above described, indicating that non-invasive assessments of hepatic fibrosis and steatosis could potentially serve as markers for the severity and progression of the diseases. Conclusions: Metabolic syndrome is a multisystem disorder that impacts organs beyond the liver and disrupts the functioning of various organs. Notably, it is linked to a higher incidence of cardiovascular diseases, independent of traditional cardiovascular risk factors. Non-invasive assessments of hepatic fibrosis and fibrosis allow clinicians to evaluate cardiovascular risk. Additionally, the ability to assess liver steatosis may open new diagnostic, therapeutic, and prognostic avenues for managing metabolic syndrome and its complications, particularly cardiovascular disease, which is the leading cause of death in these patients.
Collapse
Affiliation(s)
- Francesco Giangregorio
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Emilio Mosconi
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Maria Grazia Debellis
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Stella Provini
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Ciro Esposito
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Matteo Garolfi
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Simona Oraka
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Olga Kaloudi
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Gunel Mustafazade
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Raquel Marín-Baselga
- Department of Internal Medicine, Hospital Universitario La Paz, Paseo Castellana 241, 28046 Madrid, Spain;
| | - Yale Tung-Chen
- Department of Internal Medicine, Hospital Universitario La Paz, Paseo Castellana 241, 28046 Madrid, Spain;
| |
Collapse
|
18
|
Jin Y, Kozan D, Young ED, Hensley MR, Shen MC, Wen J, Moll T, Anderson JL, Kozan H, Rawls JF, Farber SA. A high-cholesterol zebrafish diet promotes hypercholesterolemia and fasting-associated liver steatosis. J Lipid Res 2024; 65:100637. [PMID: 39218217 PMCID: PMC11913794 DOI: 10.1016/j.jlr.2024.100637] [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: 10/19/2023] [Revised: 07/22/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Zebrafish are an ideal model organism to study lipid metabolism and to elucidate the molecular underpinnings of human lipid-associated disorders. Unlike murine models, to which various standardized high lipid diets such as a high-cholesterol diet (HCD) are available, there has yet to be a uniformly adopted zebrafish HCD protocol. In this study, we have developed an improved HCD protocol and thoroughly tested its impact on zebrafish lipid deposition and lipoprotein regulation in a dose- and time-dependent manner. The diet stability, reproducibility, and fish palatability were also validated. Fish fed HCD developed hypercholesterolemia as indicated by significantly elevated ApoB-containing lipoproteins (ApoB-LPs) and increased plasma levels of cholesterol and cholesterol esters. Feeding of the HCD to larvae for 8 days produced hepatic steatosis that became more stable and sever after 1 day of fasting and was associated with an opaque liver phenotype (dark under transmitted light). Unlike larvae, adult fish fed HCD for 14 days followed by a 3-day fast did not develop a stable fatty liver phenotype, though the fish had higher ApoB-LP levels in plasma and an upregulated lipogenesis gene fasn in adipose tissue. In conclusion, our HCD zebrafish protocol represents an effective and reliable approach for studying the temporal characteristics of the physiological and biochemical responses to high levels of dietary cholesterol and provides insights into the mechanisms that may underlie fatty liver disease.
Collapse
Affiliation(s)
- Yang Jin
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Aas, Norway
| | - Darby Kozan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Eric D Young
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Division of Gastrointestinal and Liver Pathology, Department of Pathology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Monica R Hensley
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Meng-Chieh Shen
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Jia Wen
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Tabea Moll
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer L Anderson
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Hannah Kozan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Steven A Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
19
|
Tzirkel-Hancock N, Raz C, Sharabi L, Argov-Argaman N. The Stressogenic Impact of Bacterial Secretomes Is Modulated by the Size of the Milk Fat Globule Used as a Substrate. Foods 2024; 13:2429. [PMID: 39123620 PMCID: PMC11312077 DOI: 10.3390/foods13152429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Milk fat globules (MFGs) are produced by mammary epithelial cells (MECs) and originate from intracellular lipid droplets with a wide size distribution. In the mammary gland and milk, bacteria can thrive on MFGs. Herein, we aimed to investigate whether the response of MECs to the bacterial secretome is dependent on the MFG size used as a substrate for the bacteria, and whether the response differs between pathogenic and commensal bacteria. We used secretomes from both Bacillus subtilis and E. coli. Proinflammatory gene expression in MECs was elevated by the bacteria secretomes from both bacteria sources, while higher expression was found in cells exposed to the secretome of bacteria grown on large MFGs. The secretome of B. subtilis reduced lipid droplet size in MECs. When the secretome originated from E. coli, lipid droplet size in MEC cytoplasm was elevated with a stronger response to the secretome from bacteria grown on large compared with small MFGs. These results indicate that MEC response to bacterial output is modulated by bacteria type and the size of MFGs used by the bacteria, which can modulate the stress response of the milk-producing cells, their lipid output, and consequently milk quality.
Collapse
Affiliation(s)
| | | | | | - Nurit Argov-Argaman
- Department of Animal Science, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (N.T.-H.)
| |
Collapse
|
20
|
Léger T, Alilat S, Ferron PJ, Dec L, Bouceba T, Lanceleur R, Huet S, Devriendt-Renault Y, Parinet J, Clément B, Fessard V, Le Hégarat L. Chlordecone-induced hepatotoxicity and fibrosis are mediated by the proteasomal degradation of septins. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135177. [PMID: 39018595 DOI: 10.1016/j.jhazmat.2024.135177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Chlordecone (CLD) is a pesticide persisting in soils and contaminating food webs. CLD is sequestered in the liver and poorly metabolized into chlordecol (CLDOH). In vitro liver cell models were used to investigate the fate and mechanistic effects of CLD and CLDOH using multiomics. A 3D-cell model was used to investigate whether CLD and CLDOH can affect susceptibility to the metabolic dysfunction-associated steatotic liver disease (MASLD). Hepatocytes were more sensitive to CLD than CLDOH. CLDOH was intensively metabolized into a glucuronide conjugate, whereas CLD was sequestered. CLD but not CLDOH induced a depletion of Septin-2,- 7,- 9,- 10,- 11 due to proteasomal degradation. Septin binding with CLD and CLDOH was confirmed by surface plasmon resonance. CLD disrupted lipid droplet size and increased saturated long-chain dicarboxylic acid production by inhibiting stearoyl-CoA desaturase (SCD) abundance. Neither CLD nor CLDOH induced steatosis, but CLD induced fibrosis in the 3D model of MASLD. To conclude, CLD hepatoxicity is specifically driven by the degradation of septins. CLDOH, was too rapidly metabolized to induce septin degradation. We show that the conversion of CLD to CLDOH reduced hepatotoxicity and fibrosis in liver organoids. This suggests that protective strategies could be explored to reduce the hepatotoxicity of CLD.
Collapse
Affiliation(s)
- Thibaut Léger
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France.
| | - Sarah Alilat
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Pierre-Jean Ferron
- INSERM, University of Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer) UMR_A 1317, UMR_S 1241, Previtox Network, 35000 Rennes, France
| | - Léonie Dec
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Tahar Bouceba
- Sorbonne University, CNRS, Institut de Biologie Paris-Seine (IBPS), Protein Engineering Platform, Molecular Interaction Service, Paris, France
| | - Rachelle Lanceleur
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Sylvie Huet
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Yoann Devriendt-Renault
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Pesticides and Marine Biotoxins (PBM) unit, Maison-Alfort Laboratory, 94701 Maison-Alfort CEDEX, France
| | - Julien Parinet
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Pesticides and Marine Biotoxins (PBM) unit, Maison-Alfort Laboratory, 94701 Maison-Alfort CEDEX, France
| | - Bruno Clément
- INSERM, University of Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer) UMR_A 1317, UMR_S 1241, Previtox Network, 35000 Rennes, France
| | - Valérie Fessard
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Ludovic Le Hégarat
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| |
Collapse
|
21
|
Zhang Z, Yang Q, Jin M, Wang J, Chai Y, Zhang L, Jiang Z, Yu Q. Tamoxifen upregulates the peroxisomal β-oxidation enzyme Enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase ameliorating hepatic lipid accumulation in mice. Int J Biochem Cell Biol 2024; 172:106585. [PMID: 38734232 DOI: 10.1016/j.biocel.2024.106585] [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: 10/07/2023] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
Tamoxifen is an estrogen receptor modulator that has been reported to alleviate hepatic lipid accumulation in mice, but the mechanism is still unclear. Peroxisome fatty acid β-oxidation is the main metabolic pathway for the overload of long-chain fatty acids. As long-chain fatty acids are a cause of hepatic lipid accumulation, the activation of peroxisome fatty acid β-oxidation might be a novel therapeutic strategy for metabolic associated fatty liver disease. In this study, we investigated the mechanism of tamoxifen against hepatic lipid accumulation based on the activation of peroxisome fatty acid β-oxidation. Tamoxifen reduced liver long-chain fatty acids and relieved hepatic lipid accumulation in high fat diet mice without sex difference. In vitro, tamoxifen protected primary hepatocytes against palmitic acid-induced lipotoxicity. Mechanistically, the RNA-sequence of hepatocytes isolated from the liver revealed that peroxisome fatty acid β-oxidation was activated by tamoxifen. Protein and mRNA expression of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase were significantly increased in vivo and in vitro. Small interfering RNA enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase in primary hepatocytes abolished the therapeutic effects of tamoxifen in lipid accumulation. In conclusion, our results indicated that tamoxifen could relieve hepatic lipid accumulation in high fat diet mice based on the activation of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase-mediated peroxisome fatty acids β-oxidation.
Collapse
Affiliation(s)
- Ziling Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Qinqin Yang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Ming Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Jie Wang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Yuanyuan Chai
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
22
|
Agarwal M, Roth K, Yang Z, Sharma R, Maddipati K, Westrick J, Petriello MC. Loss of flavin-containing monooxygenase 3 modulates dioxin-like polychlorinated biphenyl 126-induced oxidative stress and hepatotoxicity. ENVIRONMENTAL RESEARCH 2024; 250:118492. [PMID: 38373550 PMCID: PMC11102846 DOI: 10.1016/j.envres.2024.118492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/21/2024]
Abstract
Dioxin-like pollutants (DLPs), such as polychlorinated biphenyl 126 (PCB 126), are synthetic chemicals classified as persistent organic pollutants. They accumulate in adipose tissue and have been linked to cardiometabolic disorders, including fatty liver disease. The toxicity of these compounds is associated with activation of the aryl hydrocarbon receptor (Ahr), leading to the induction of phase I metabolizing enzyme cytochrome P4501a1 (Cyp1a1) and the subsequent production of reactive oxygen species (ROS). Recent research has shown that DLPs can also induce the xenobiotic detoxification enzyme flavin-containing monooxygenase 3 (FMO3), which plays a role in metabolic homeostasis. We hypothesized whether genetic deletion of Fmo3 could protect mice, particularly in the liver, where Fmo3 is most inducible, against PCB 126 toxicity. To test this hypothesis, male C57BL/6 wild-type (WT) mice and Fmo3 knockout (Fmo3 KO) mice were exposed to PCB 126 or vehicle (safflower oil) during a 12-week study, at weeks 2 and 4. Various analyses were performed, including hepatic histology, RNA-sequencing, and quantitation of PCB 126 and F2-isoprostane concentrations. The results showed that PCB 126 exposure caused macro and microvesicular fat deposition in WT mice, but this macrovesicular fatty change was absent in Fmo3 KO mice. Moreover, at the pathway level, the hepatic oxidative stress response was significantly different between the two genotypes, with the induction of specific genes observed only in WT mice. Notably, the most abundant F2-isoprostane, 8-iso-15-keto PGE2, increased in WT mice in response to PCB 126 exposure. The study's findings also demonstrated that hepatic tissue concentrations of PCB 126 were higher in WT mice compared to Fmo3 KO mice. In summary, the absence of FMO3 in mice led to a distinctive response to dioxin-like pollutant exposure in the liver, likely due to alterations in lipid metabolism and storage, underscoring the complex interplay of genetic factors in the response to environmental toxins.
Collapse
Affiliation(s)
- Manisha Agarwal
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Katherine Roth
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Zhao Yang
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Rahul Sharma
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Krishnarao Maddipati
- Department of Pathology, Lipidomic Core Facility, Wayne State University, Detroit, MI, 48202, USA
| | - Judy Westrick
- Department of Chemistry, Lumigen Instrumentation Center, Wayne State University, Detroit, MI, 48202, USA
| | - Michael C Petriello
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
23
|
Liu Z, Zhu H, Zhao J, Yu L, Que S, Xu J, Geng L, Zhou L, Valenti L, Zheng S. Multi-omics analysis reveals a crosstalk between ferroptosis and peroxisomes on steatotic graft failure after liver transplantation. MedComm (Beijing) 2024; 5:e588. [PMID: 38868330 PMCID: PMC11167151 DOI: 10.1002/mco2.588] [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: 07/21/2023] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 06/14/2024] Open
Abstract
To identify the mechanism underlying macrosteatosis (MaS)-related graft failure (GF) in liver transplantation (LT) by multi-omics network analysis. The transcriptome and metabolome were assayed in graft and recipient plasma in discovery (n = 68) and validation (n = 89) cohorts. Differentially expressed molecules were identified by MaS and GF status. Transcriptional regulatory networks were generated to explore the mechanism for MaS-related inferior post-transplant prognosis. The differentially expressed molecules associated with MaS and GF were enriched in ferroptosis and peroxisome-related pathways. Core features of MaS-related GF were presented on decreased transferrin and impaired anti-oxidative capacity dependent upon dysregulation of transcription factors hepatocyte nuclear factor 4A (HNF4A) and hypoxia-inducible factor 1A (HIF1A). Furthermore, miR-362-3p and miR-299-5p inhibited transferrin and HIF1A expression, respectively. Lower M2 macrophages but higher memory CD4 T cells were observed in MaS-related GF cases. These results were validated in clinical specimens and cellular models. Systemic analysis of multi-omics data depicted a panorama of biological pathways deregulated in MaS-related GF. Transcriptional regulatory networks centered on transferrin and anti-oxidant responses were associated with poor MaS graft quality, qualifying as potential targets to improve prognosis of patients after LT.
Collapse
Affiliation(s)
- Zhengtao Liu
- Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang ProvinceShulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
- NHC Key Laboratory of Combined Multi‐Organ TransplantationKey Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMS, First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Organ TransplantationFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Shulan Hospital (Hangzhou)HangzhouChina
| | - Hai Zhu
- NHC Key Laboratory of Combined Multi‐Organ TransplantationKey Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMS, First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Organ TransplantationFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Junsheng Zhao
- Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang ProvinceShulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
| | - Lu Yu
- Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
- Shulan Hospital (Hangzhou)HangzhouChina
- School of MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | | | - Jun Xu
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Lei Geng
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Lin Zhou
- NHC Key Laboratory of Combined Multi‐Organ TransplantationKey Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMS, First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Organ TransplantationFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Luca Valenti
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
- Transfusion Medicine UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Biological Resource Center UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Shusen Zheng
- Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang ProvinceShulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
- NHC Key Laboratory of Combined Multi‐Organ TransplantationKey Laboratory of the Diagnosis and Treatment of Organ TransplantationCAMS, First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Organ TransplantationFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Shulan Hospital (Hangzhou)HangzhouChina
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
24
|
Shi Y, Zhong L, Liu Y, Xu S, Dai J, Zhang Y, Hu Y. Dietary sanguinarine supplementation recovers the decrease in muscle quality and nutrient composition induced by high-fat diets of grass carp ( Ctenopharyngodon idella). ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:208-219. [PMID: 38800733 PMCID: PMC11126770 DOI: 10.1016/j.aninu.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/13/2024] [Accepted: 04/01/2024] [Indexed: 05/29/2024]
Abstract
The intake of high-fat diets (HFD) has been shown to diminish the muscle quality of aquatic animals. Sanguinarine, as an excellent additive, exhibits the capability to reduce fat deposition and alleviate inflammation. However, its role in the muscle quality reduction caused by HFD remains unclear. An eight-week trial was conducted to investigate the impacts of dietary supplementation of sanguinarine at 1200 μg/kg (HFDS; crude fat = 10%) on the muscle quality of grass carp (Ctenopharyngodon idellus) in comparison to a basic diet (CON, crude fat = 5%). Each group had 3 replicates, with 40 fish per replicate. This experiment employed one-way ANOVA and Duncan's multiple comparisons of the means. The results showed that the HFD exhibited lower growth performance, reduced protein deposition, myofiber diameter, and muscle hardness, coupled with higher levels of fat deposition and inflammation when compared with the CON. However, HFDS improved growth performance (P < 0.05), fat metabolism (ppar-α ( P = 0.001), lpl (P < 0.001), atgl (P < 0.001), and cpt1 (P = 0.001) expression exhibited a significant elevation), protein deposition (the protein and mRNA levels of AKT (P = 0.004), PI3K (P = 0.027), TOR (P = 0.005), and P70S6K (P = 0.007) demonstrated a marked increase), myofiber diameter, muscle hardness, and the total content of eicosapentaenoic acid and docosahexaenoic acid. Furthermore, the HFDS reduced oxidative damage caused by fat deposition by significantly downregulating nf-κb (P < 0.001), il-1β (P < 0.001), il-6 (P < 0.001), il-8 (P = 0.003), and tnf-α (P < 0.001) expression and markedly upregulated nrf2 (P < 0.001), gpx4 (P < 0.001), cat (P < 0.001), sod (P < 0.001), and gr (P = 0.003) expression. The findings from this study suggest that sanguinarine has the potential to alleviate the adverse effects of HFD on growth and muscle quality, providing a theoretical foundation for its practical implementation.
Collapse
Affiliation(s)
- Yong Shi
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Research Center for Utilization of Characteristics of Aquatic Resources, Hunan Agricultural University, Changsha, 410128, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lei Zhong
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Research Center for Utilization of Characteristics of Aquatic Resources, Hunan Agricultural University, Changsha, 410128, China
| | - Yuanxiang Liu
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Research Center for Utilization of Characteristics of Aquatic Resources, Hunan Agricultural University, Changsha, 410128, China
| | - Shude Xu
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Research Center for Utilization of Characteristics of Aquatic Resources, Hunan Agricultural University, Changsha, 410128, China
| | - Jihong Dai
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Research Center for Utilization of Characteristics of Aquatic Resources, Hunan Agricultural University, Changsha, 410128, China
| | - Yaozhengtai Zhang
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Research Center for Utilization of Characteristics of Aquatic Resources, Hunan Agricultural University, Changsha, 410128, China
| | - Yi Hu
- Fisheries College, Hunan Agricultural University, Changsha, 410128, China
- Hunan Engineering Research Center for Utilization of Characteristics of Aquatic Resources, Hunan Agricultural University, Changsha, 410128, China
| |
Collapse
|
25
|
Gu W, Li T, Huang Y, Wang R, Zhang L, Chen R, Li R, Liu C. Metabolic Profile and Lipid Metabolism Phenotype in Mice with Conditional Deletion of Hepatic BMAL1. Int J Mol Sci 2024; 25:6070. [PMID: 38892255 PMCID: PMC11172555 DOI: 10.3390/ijms25116070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The disruption of circadian rhythms (CRs) has been linked to metabolic disorders, yet the role of hepatic BMAL1, a key circadian regulator, in the whole-body metabolism and the associated lipid metabolic phenotype in the liver remains unclear. Bmal1 floxed (Bmal1f/f) and hepatocyte-specific Bmal1 knockout (Bmal1hep-/-) C57BL/6J mice underwent a regular feeding regimen. Hepatic CR, lipid content, mitochondrial function, and systemic metabolism were assessed at zeitgeber time (ZT) 0 and ZT12. Relevant molecules were examined to elucidate the metabolic phenotype. Hepatocyte-specific knockout of Bmal1 disrupted the expression of rhythmic genes in the liver. Bmal1hep-/- mice exhibited decreased hepatic TG content at ZT0, primarily due to enhanced lipolysis, reduced lipogenesis, and diminished lipid uptake. The β-oxidation function of liver mitochondria decreased at both ZT0 and ZT12. Our findings on the metabolic profile and associated hepatic lipid metabolism in the absence of Bmal1 in hepatocytes provides new insights into metabolic syndromes from the perspective of liver CR disturbances.
Collapse
Affiliation(s)
- Weijia Gu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; (W.G.)
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Ting Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; (W.G.)
| | - Yuxin Huang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; (W.G.)
| | - Ruiqing Wang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; (W.G.)
| | - Lu Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; (W.G.)
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Rucheng Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; (W.G.)
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; (W.G.)
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; (W.G.)
- Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| |
Collapse
|
26
|
Geng X, Lin R, Hasegawa Y, Chao L, Shang H, Yang J, Tian W, Ma W, Zhuang M, Li J. Effects of Scallop Mantle Toxin on Intestinal Microflora and Intestinal Barrier Function in Mice. Toxins (Basel) 2024; 16:247. [PMID: 38922142 PMCID: PMC11209270 DOI: 10.3390/toxins16060247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
Previous studies have shown that feeding mice with food containing mantle tissue from Japanese scallops results in aggravated liver and kidney damage, ultimately resulting in mortality within weeks. The aim of this study is to evaluate the toxicity of scallop mantle in China's coastal areas and explore the impact of scallop mantle toxins (SMT) on intestinal barrier integrity and gut microbiota in mice. The Illumina MiSeq sequencing of V3-V4 hypervariable regions of 16S ribosomal RNA was employed to study the alterations in gut microbiota in the feces of SMT mice. The results showed that intestinal flora abundance and diversity in the SMT group were decreased. Compared with the control group, significant increases were observed in serum indexes related to liver, intestine, inflammation, and kidney functions among SMT-exposed mice. Accompanied by varying degrees of tissue damage observed within these organs, the beneficial bacteria of Muribaculaceae and Marinifilaceae significantly reduced, while the harmful bacteria of Enterobacteriaceae and Helicobacter were significantly increased. Taken together, this article elucidates the inflammation and glucose metabolism disorder caused by scallop mantle toxin in mice from the angle of gut microbiota and metabolism. SMT can destroy the equilibrium of intestinal flora and damage the intestinal mucosal barrier, which leads to glucose metabolism disorder and intestinal dysfunction and may ultimately bring about systemic toxicity.
Collapse
Affiliation(s)
- Xiong Geng
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China; (X.G.); (R.L.); (H.S.); (W.T.); (W.M.); (M.Z.)
| | - Ran Lin
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China; (X.G.); (R.L.); (H.S.); (W.T.); (W.M.); (M.Z.)
- College of Environmental Technology, Muroran Institute of Technology, Muroran 050-8585, Japan;
| | - Yasushi Hasegawa
- College of Environmental Technology, Muroran Institute of Technology, Muroran 050-8585, Japan;
| | - Luomeng Chao
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Huayan Shang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China; (X.G.); (R.L.); (H.S.); (W.T.); (W.M.); (M.Z.)
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Jingjing Yang
- Kerqin District Testing Institute for Food and Drug Control, Tongliao 028000, China;
| | - Weina Tian
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China; (X.G.); (R.L.); (H.S.); (W.T.); (W.M.); (M.Z.)
- Kerqin District Testing Institute for Food and Drug Control, Tongliao 028000, China;
| | - Wenting Ma
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China; (X.G.); (R.L.); (H.S.); (W.T.); (W.M.); (M.Z.)
| | - Miaomiao Zhuang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China; (X.G.); (R.L.); (H.S.); (W.T.); (W.M.); (M.Z.)
| | - Jianrong Li
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China; (X.G.); (R.L.); (H.S.); (W.T.); (W.M.); (M.Z.)
| |
Collapse
|
27
|
Yang YJ, Kim MJ, Yang JH, Heo JW, Kim HH, Kim WH, Kim GS, Lee HJ, Kim YW, Kim KY, Park KI. Liquid Chromatography/Tandem Mass Spectrometry Analysis of Sophora flavescens Aiton and Protective Effects against Alcohol-Induced Liver Injury and Oxidative Stress in Mice. Antioxidants (Basel) 2024; 13:541. [PMID: 38790646 PMCID: PMC11117756 DOI: 10.3390/antiox13050541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
In this study, we investigated the hepatoprotective effects of an ethanol extract of Sophora flavescens Aiton (ESF) on an alcohol-induced liver disease mouse model. Alcoholic liver disease (ALD) was caused by the administration of ethanol to male C57/BL6 mice who were given a Lieber-DeCarli liquid diet, including ethanol. The alcoholic fatty liver disease mice were orally administered ESF (100 and 200 mg/kg bw/day) or silymarin (50 mg/kg bw/day), which served as a positive control every day for 16 days. The findings suggest that ESF enhances hepatoprotective benefits by significantly decreasing serum levels of aspartate transaminase (AST) and alanine transaminase (ALT), markers for liver injury. Furthermore, ESF alleviated the accumulation of triglyceride (TG) and total cholesterol (TC), increased serum levels of superoxide dismutase (SOD) and glutathione (GSH), and improved serum alcohol dehydrogenase (ADH) activity in the alcoholic fatty liver disease mice model. Cells and organisms rely on the Kelch-like ECH-associated protein 1- Nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2) system as a critical defensive mechanism in response to oxidative stress. Therefore, Nrf2 plays an important role in ALD antioxidant responses, and its level is decreased by increased reactive oxidation stress (ROS) in the liver. ESF increased Nrf2, which was decreased in ethanol-damaged livers. Additionally, four polyphenol compounds were identified through a qualitative analysis of the ESF using LC-MS/MS. This study confirmed ESF's antioxidative and hangover-elimination effects and suggested the possibility of using Sophora flavescens Aiton (SF) to treat ALD.
Collapse
Affiliation(s)
- Ye Jin Yang
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Min Jung Kim
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Ju-Hye Yang
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Ji Woong Heo
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Hun Hwan Kim
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Woo H. Kim
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Gon Sup Kim
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Hu-Jang Lee
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Young Woo Kim
- School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea;
| | - Kwang Youn Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Kwang Il Park
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| |
Collapse
|
28
|
Shen Y, Zhao Y, Zhong C, Huang H, Yang Z, Wu M, Lu L, Yang R, Ke X. Primary study on the effects and mechanisms of separate and combined decoctions of Scutellaria baicalensis Georgi - Coptis chinensis Franch extracts in relieving acute alcoholic liver injury in rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117790. [PMID: 38253276 DOI: 10.1016/j.jep.2024.117790] [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: 11/10/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellaria baicalensis Georgi (SBG) and Coptis chinensis Franch (CCF) are traditional herbal medicine pairs used for clearing heat and eliminating dampness, stopping diarrhea, and detoxification. Traditionally, these two herbs are combined and decocted together, but the modern preparation procedures separate them to avoid the large amount of precipitation generated from co-decoction. Thus, a conflict lies between the traditional and modern extraction processes of Scutellaria baicalensis Georgi - Coptis chinensis Franch (SBG-CCF). AIM OF STUDY There is a conflict between traditional medical practices of SBG-CCF and the modern formulation industry. In this study, we investigated the differences in the effects and mechanisms of SBG-CCF extracted by decocting separately and combining decoctions, as well as the scientific effectiveness of traditional and modern treatment methods on both. Acute alcoholic liver injury (ALI) rats were used as the pathological model. MATERIALS AND METHODS SD rats were divided into 8 groups, including blank group, model group, low, medium, and high dose groups of SBG-CCF separated decoction, low, medium, and high dose groups of SBG-CCF combined decoction. Acute alcoholic liver injury model was induced in rats by gradually increasing the dose of alcohol through gavage everyday using white wine with an alcohol content 52%. Aspartate aminotransferase (AST), alanine aminotransferase (ALT), total cholesterol (TC), triglyceride (TG), lactate dehydrogenase (LDH), malondialdehyde (MDA), superoxide dismutase (SOD), and reduced glutathione (GSH) were used as indicators to assess the intervention effect of SBG-CCF. And the potential active ingredients of SBG-CCF and the targets related to ALI were screened using network pharmacology, and the prediction results of network pharmacology were verified by quantitative real-time fluorescence PCR (qRT-PCR). RESULTS SBG-CCF decoction alone and six combinations of decoctions have different degrees of improvement on alcoholic liver injury, with significant efficacy in the middle-dose group, and the combined decoction was superior to the individual decoction. SBG-CCF gavage can reduce the activity of AST, ALT, TC, TG, LDH, and MDA in the serum and liver of ALI rats, while increasing the levels of SOD and GSH. Network pharmacological analysis identified 39 active components, mainly flavonoids and alkaloids. Enrichment analysis suggested that SBG-CCF may treat ALI through the regulation of tumor necrosis factor (TNF), mitogen-activated protein kinase (MAPK), interleukin-17 (IL-17), apoptosis, and the Toll-like receptor signaling pathways. The key targets in the Disease-Signaling Pathway-Target Network were MAPK8, IKBKB, MAPK10, MAPK3, MAPK1, and AKT1. qRT-PCR results indicated that targets regulating inflammation and lipid metabolism are MAPK8, MAPK10, MAPK3, and AKT1. CONCLUSION SBG-CCF separately extracts and combines decoction can alleviate acute alcoholic liver injury, and the effect of combined decoction is more significant than separate decoction, implying that the precipitate produced by the combination of the two is also an active substance. The resistance mechanism of SBG-CCF ALI may be related to the modulation of lipid metabolism, inhibition of lipid peroxidation, and oxidative stress. SBG-CCF has the characteristics of multi-component, multi-pathway, and multi-target resistance to ALI.
Collapse
Affiliation(s)
- Yingyan Shen
- Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial, Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu University of Traditional Chinese Medicine, Chendu, China
| | - Yixin Zhao
- Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial, Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu University of Traditional Chinese Medicine, Chendu, China
| | - Chunxue Zhong
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Hanmei Huang
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Zaiqing Yang
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Mingxing Wu
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Lingzhi Lu
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China
| | - Rongping Yang
- Chongqing Key Laboratory of Chinese Medicine New Drug Screening, Southwest University, Chongqing, China.
| | - Xiumei Ke
- College of Pharmacy, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
29
|
Bai Y, Morita K, Kokaji T, Hatano A, Ohno S, Egami R, Pan Y, Li D, Yugi K, Uematsu S, Inoue H, Inaba Y, Suzuki Y, Matsumoto M, Takahashi M, Izumi Y, Bamba T, Hirayama A, Soga T, Kuroda S. Trans-omic analysis reveals opposite metabolic dysregulation between feeding and fasting in liver associated with obesity. iScience 2024; 27:109121. [PMID: 38524370 PMCID: PMC10960062 DOI: 10.1016/j.isci.2024.109121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/03/2023] [Accepted: 01/31/2024] [Indexed: 03/26/2024] Open
Abstract
Dysregulation of liver metabolism associated with obesity during feeding and fasting leads to the breakdown of metabolic homeostasis. However, the underlying mechanism remains unknown. Here, we measured multi-omics data in the liver of wild-type and leptin-deficient obese (ob/ob) mice at ad libitum feeding and constructed a differential regulatory trans-omic network of metabolic reactions. We compared the trans-omic network at feeding with that at 16 h fasting constructed in our previous study. Intermediate metabolites in glycolytic and nucleotide metabolism decreased in ob/ob mice at feeding but increased at fasting. Allosteric regulation reversely shifted between feeding and fasting, generally showing activation at feeding while inhibition at fasting in ob/ob mice. Transcriptional regulation was similar between feeding and fasting, generally showing inhibiting transcription factor regulations and activating enzyme protein regulations in ob/ob mice. The opposite metabolic dysregulation between feeding and fasting characterizes breakdown of metabolic homeostasis associated with obesity.
Collapse
Affiliation(s)
- Yunfan Bai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keigo Morita
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Toshiya Kokaji
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Data Science Center, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara, Japan
| | - Atsushi Hatano
- Department of Omics and Systems Biology, Graduate School of Medical and Dental Sciences, Niigata University, 757 Ichibancho, Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Satoshi Ohno
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
- Department of AI Systems Medicine, M&D Data Science Center, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Riku Egami
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Yifei Pan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Dongzi Li
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Katsuyuki Yugi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Institute for Advanced Biosciences, Keio University, Fujisawa 252-8520, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Saori Uematsu
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Hiroshi Inoue
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8641, Japan
| | - Yuka Inaba
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8641, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Masaki Matsumoto
- Department of Omics and Systems Biology, Graduate School of Medical and Dental Sciences, Niigata University, 757 Ichibancho, Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Masatomo Takahashi
- Division of Metabolomics/Mass Spectrometry Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics/Mass Spectrometry Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- Division of Metabolomics/Mass Spectrometry Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan
| | - Shinya Kuroda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
30
|
Shao J, Lai C, Zheng Q, Luo Y, Li C, Zhang B, Sun Y, Liu S, Shi Y, Li J, Zhao Z, Guo L. Effects of dietary arsenic exposure on liver metabolism in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116147. [PMID: 38460405 DOI: 10.1016/j.ecoenv.2024.116147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/11/2024]
Abstract
Arsenic, a ubiquitous environmental toxicant with various forms and complex food matrix interactions, can reportedly exert differential effects on the liver compared to drinking water exposure. To examine its specific liver-related harms, we targeted the liver in C57BL/6 J mice (n=48, 8-week-old) fed with arsenic-contaminated food (30 mg/kg) for 60 days, mimicking the rice arsenic composition observed in real-world scenarios (iAsV: 7.3%, iAsIII: 72.7%, MMA: 1.0%, DMA: 19.0%). We then comprehensively evaluated liver histopathology, metabolic changes, and the potential role of the gut-liver axis using human hepatocellular carcinoma cells (HepG2) and microbiota/metabolite analyses. Rice arsenic exposure significantly altered hepatic lipid (fatty acids, glycerol lipids, phospholipids, sphingolipids) and metabolite (glutathione, thioneine, spermidine, inosine, indole-derivatives, etc.) profiles, disrupting 33 metabolic pathways (bile secretion, unsaturated fatty acid biosynthesis, glutathione metabolism, ferroptosis, etc.). Pathological examination revealed liver cell necrosis/apoptosis, further confirmed by ferroptosis induction in HepG2 cells. Gut microbiome analysis showed enrichment of pathogenic bacteria linked to liver diseases and depletion of beneficial strains. Fecal primary and secondary bile acids, short-chain fatty acids, and branched-chain amino acids were also elevated. Importantly, mediation analysis revealed significant correlations between gut microbiota, fecal metabolites, and liver metabolic alterations, suggesting fecal metabolites may mediate the impact of gut microbiota and liver metabolic disorders. Gut microbiota and its metabolites may play significant roles in arsenic-induced gut-liver injuries. Overall, our findings demonstrate that rice arsenic exposure triggers oxidative stress, disrupts liver metabolism, and induces ferroptosis.
Collapse
Affiliation(s)
- Junli Shao
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Chengze Lai
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Qiuyi Zheng
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Yu Luo
- Guangzhou Liwan District Center for Disease Control and Prevention, Guangzhou, Guangdong 510176, China
| | - Chengji Li
- Yunfu Disease Control and Prevention Center, Guangdong Province 527300, China
| | - Bin Zhang
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Yanqin Sun
- Department of Pathology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan 523808, China
| | - Shizhen Liu
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Yingying Shi
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Jinglin Li
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Zuguo Zhao
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Lianxian Guo
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
31
|
Ye X, He X, Hu Z, Zheng F, Huang X, Xie X, Chen F, Ou H, Qiu R. Metabolomic analysis identifies dysregulation of lipid metabolism in the immune clearance phase of chronic hepatitis B patients. J Pharm Biomed Anal 2024; 239:115900. [PMID: 38064772 DOI: 10.1016/j.jpba.2023.115900] [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: 09/28/2023] [Revised: 11/13/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
There is an accelerated progression of liver necroinflammation and fibrosis in the liver during the immune clearance (IC) phase of Chronic hepatitis B virus (HBV) infection, which are critical indicators of antiviral treatment for chronic hepatitis B (CHB) infection. This study applied serum metabolomics to identify the potential metabolite biomarkers for differential diagnosis between the CHB immune tolerance (IT) and Immune clearance (IC) phases. A liquid chromatography-mass spectrometry (LC-MS)-based approach was applied to evaluate and compared the serum metabolic profiles of 28 patients in IT phase and 33 patients in IC phase and appropriate statistical methods with MetaboAnalystR 2.0 R package to analyze those metabolites. The differential metabolites between IT and TC groups were classified and the top altered classification were lipids and lipid-like molecules and fatty acyls, clearly indicating that there were differences in the lipid metabolomic profile of HBV-infected patients with IT vs. IR phase. We identified the top 10 potential metabolite biomarkers for differential diagnosis between IT and IR. There were four lipid metabolites among them and the AUC of two of them, octadecadienoyl-sn-glycero-3-phosphocholine and 3-Cycloheptene-l-acetic acid, were 0.983 and 0.933. octadecadienoyl-sn-glycero-3-phosphocholine is Diacylglycerol (18:2n6/18:0) and 3-Cycloheptene-l-acetic acid is hydroxy fatty acids, both of which were associated with lipid metabolism. This study not only provides the potential metabolic biomarkers but also insight into the mechanism of CHB progression during IT clearance phase.
Collapse
Affiliation(s)
- Xiangyang Ye
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China; Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Xiongzhi He
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Zhenting Hu
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Fengfeng Zheng
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Xiaogang Huang
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Xuemei Xie
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Feihua Chen
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Hanbing Ou
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Rongxian Qiu
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, Fujian 351100, China.
| |
Collapse
|
32
|
Tullio SCMC, McCoy K, Chalcraft DR. Chronic toxicity and liver histopathology of mosquito fish (Gambusia holbrooki) exposed to natural and modified nanoclays. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168060. [PMID: 37918747 DOI: 10.1016/j.scitotenv.2023.168060] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/25/2023] [Accepted: 10/21/2023] [Indexed: 11/04/2023]
Abstract
Nanoclays are found in the air, water, and soil, and modified nanoclays are being developed and used in several consumer products. For example, modified nanoclays are used to remove pollutants from wastewater. Ironically, however, nanoclays are now considered emerging contaminants. Indeed, release of modified nanoclays in aquatic systems, even as remediating agents, could adversely affect associated wildlife. However, aquatic organisms have interacted with natural nanoclays for millennia, and it is unclear if modified nanoclays induce stronger effects than the nanoclays that occur naturally. The concentrations over which nanoclays occur and illicit negative effects are not well studied. This study investigated the dose response of a natural nanoclay (Na+montmorillonite) relative to two modified nanoclays (Cloisite®30B and Novaclay™) on survival, body condition, and liver pathomorphology of Gambusia holbrooki after 14 days of exposure. Although none of the nanoclays affected survival and body condition of G. holbrooki over 14 days, each nanoclay induced histopathological changes in liver tissues at very low concentrations (LOAEL: 0.01 mgL-1). The effects of nanoclays on hepatic cell circulatory (blood cell aggregation with increased number of Kupffer cells and hemosiderin deposits), regressive (hepatocyte vacuolization), and degenerative (cell death) changes of mosquito fish varied among nanoclay types. Novaclay™ at low concentrations caused circulatory changes on hepatic tissues of G. holbrooki, whereas both natural nanoclays and Cloisite®30B showed little effect on circulatory endpoints. In contrast, all of the nanoclays induced regressive and degenerative changes on liver tissues of mosquito fish across all concentrations tested. This study clearly reveals that natural and modified nanoclays have important health implications for fish and other aquatic organisms. Consequently, the widespread use of modified nanoclays in several applications and increased release of natural nanoclays through erosion or other processes needs to be evaluated in more detail especially in the context of their safety for aquatic systems.
Collapse
Affiliation(s)
- S C M C Tullio
- Department of Biology, East Carolina University, 1000 E 5th Street, N108 Howell Science Building, Greenville, NC 27834, USA.
| | - K McCoy
- Harbor Branch Oceanographic Institute, Florida Atlantic University, 5600 US1 North, Fort Pierce, FL 34946, USA
| | - D R Chalcraft
- Department of Biology, East Carolina University, 1000 E 5th Street, N108 Howell Science Building, Greenville, NC 27834, USA
| |
Collapse
|
33
|
Nayak N, Mukherjee T, Pattnaik A. Comprehensive Role of GDF15 in Inhibiting Adipogenesis and Hyperlipidemia, Enhancing Cardiovascular Health and Alleviating Inflammation in Metabolic Disorders. Curr Pharm Des 2024; 30:2387-2399. [PMID: 38934286 DOI: 10.2174/0113816128318741240611114448] [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: 03/23/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024]
Abstract
Growth Differentiation Factor 15 (GDF15) has emerged as a pivotal signaling molecule implicated in diverse physiological processes, spanning metabolic regulation, inflammation, and cardiovascular health. This study provides a comprehensive exploration of GDF15's multifaceted role, primarily focusing on its association with obesity-related complications and therapeutic potential. GDF15's involvement in energy homeostasis, specifically its regulation of body weight and appetite through hindbrain neuron activation and the GFRAL-RET signaling pathway, underscores its significance as an appetite-regulating hormone. GDF15's intricate modulation within adipose tissue dynamics in response to dietary changes and obesity, coupled with its influence on insulin sensitivity, highlights its critical role in metabolic health. The manuscript delves into the intricate crosstalk between GDF15 and pathways related to insulin sensitivity, macrophage polarization, and adipose tissue function, elucidating its potential as a therapeutic target for metabolic disorders associated with obesity. GDF15's association with chronic low-grade inflammation and its impact on cardiovascular health, particularly during hyperlipidemia and ischemic events, are explored. The intricate relationship between GDF15 and cardiovascular diseases, including its effects on endothelial function, cardiac hypertrophy, and heart failure, emphasizes its multifaceted nature in maintaining overall cardiovascular well-being. Challenges regarding the therapeutic application of GDF15, such as long-term safety concerns and ongoing clinical investigations, are discussed. Lastly, future research directions exploring GDF15's potential in addressing obesity-related complications and cardiovascular risks are proposed, highlighting its promising role as a therapeutic target in reshaping treatment strategies for obesity and associated health conditions.
Collapse
Affiliation(s)
- Nikita Nayak
- Department of Pharmaceutical Sciences and Technology, Division of Pharmacology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Tuhin Mukherjee
- Department of Pharmaceutical Sciences and Technology, Division of Pharmacology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Ashok Pattnaik
- Department of Pharmaceutical Sciences and Technology, Division of Pharmacology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| |
Collapse
|
34
|
Syed-Abdul MM. Lipid Metabolism in Metabolic-Associated Steatotic Liver Disease (MASLD). Metabolites 2023; 14:12. [PMID: 38248815 PMCID: PMC10818604 DOI: 10.3390/metabo14010012] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Metabolic-associated steatotic liver disease (MASLD) is a cluster of pathological conditions primarily developed due to the accumulation of ectopic fat in the hepatocytes. During the severe form of the disease, i.e., metabolic-associated steatohepatitis (MASH), accumulated lipids promote lipotoxicity, resulting in cellular inflammation, oxidative stress, and hepatocellular ballooning. If left untreated, the advanced form of the disease progresses to fibrosis of the tissue, resulting in irreversible hepatic cirrhosis or the development of hepatocellular carcinoma. Although numerous mechanisms have been identified as significant contributors to the development and advancement of MASLD, altered lipid metabolism continues to stand out as a major factor contributing to the disease. This paper briefly discusses the dysregulation in lipid metabolism during various stages of MASLD.
Collapse
Affiliation(s)
- Majid Mufaqam Syed-Abdul
- Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
35
|
Craciunescu O, Seciu-Grama AM, Mihai E, Utoiu E, Negreanu-Pirjol T, Lupu CE, Artem V, Ranca A, Negreanu-Pirjol BS. The Chemical Profile, Antioxidant, and Anti-Lipid Droplet Activity of Fluid Extracts from Romanian Cultivars of Haskap Berries, Bitter Cherries, and Red Grape Pomace for the Management of Liver Steatosis. Int J Mol Sci 2023; 24:16849. [PMID: 38069172 PMCID: PMC10706173 DOI: 10.3390/ijms242316849] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
This study aimed to investigate, for the first time, the chemical composition and antioxidant activity of fluid extracts obtained from three Romanian cultivars of haskap berries (Lonicera caerulea L.) var. Loni, bitter cherries (Prunus avium var. sylvestris Ser.) var. Silva, and pomace from red grapes (Vitis vinifera L.) var. Mamaia, and their capacity to modulate in vitro steatosis, in view of developing novel anti-obesity products. Total phenolic, flavonoid, anthocyanin, and ascorbic acid content of fluid extracts was spectrophotometrically assessed and their free radical scavenging capacity was evaluated using Trolox Equivalent Antioxidant Capacity (TEAC) and free 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical inhibition assays. The Pearson coefficients showed a moderate correlation between the antioxidant activity of fluid extracts and their phenolic content, but a strong correlation between anthocyanin and ascorbic acid content. HPLC analysis identified and quantified the main phenolic compounds of chlorogenic and syringic acid, catechin, and glycosylated kaempferol, apigenin, and quercetin, in variable proportions. An in vitro experimental model of steatosis was developed in HepG2 hepatocytes treated with a mixture of free fatty acids. Cell culture analyses showed that cytocompatible concentrations of fluid extracts could significantly reduce the lipid accumulation and inhibit the reactive oxygen species, malondialdehyde, and nitric oxide secretion in stressed hepatocytes. In conclusion, these results put an emphasis on the chemical compounds' high antioxidant and liver protection capacity of unstudied fluid extracts obtained from Romanian cultivars of bitter cherries var. Silva and pomace of red grapes var. Mamaia, similar to the fluid extract of haskap berries var. Loni, in particular, the positive modulation of fat deposition next to oxidative stress and the lipid peroxidation process triggered by fatty acids in HepG2 hepatocytes. Consequently, this study indicated that these fluid extracts could be further exploited as hepatoprotective agents in liver steatosis, which provides a basis for the further development of novel extract mixtures with synergistic activity as anti-obesity products.
Collapse
Affiliation(s)
- Oana Craciunescu
- National Institute of R&D for Biological Sciences, 060031 Bucharest, Romania; (A.-M.S.-G.); (E.M.); (E.U.)
| | - Ana-Maria Seciu-Grama
- National Institute of R&D for Biological Sciences, 060031 Bucharest, Romania; (A.-M.S.-G.); (E.M.); (E.U.)
| | - Elena Mihai
- National Institute of R&D for Biological Sciences, 060031 Bucharest, Romania; (A.-M.S.-G.); (E.M.); (E.U.)
| | - Elena Utoiu
- National Institute of R&D for Biological Sciences, 060031 Bucharest, Romania; (A.-M.S.-G.); (E.M.); (E.U.)
| | - Ticuta Negreanu-Pirjol
- Faculty of Pharmacy, University Ovidius of Constanta, 900470 Constanta, Romania; (C.E.L.); (B.-S.N.-P.)
- Academy of Romanian Scientists, Ilfov Street, No. 3, 050044 Bucharest, Romania
| | - Carmen Elena Lupu
- Faculty of Pharmacy, University Ovidius of Constanta, 900470 Constanta, Romania; (C.E.L.); (B.-S.N.-P.)
| | - Victoria Artem
- Research-Development Station for Viticulture and Winemaking of Murfatlar, 905100 Murfatlar, Romania; (V.A.); (A.R.)
| | - Aurora Ranca
- Research-Development Station for Viticulture and Winemaking of Murfatlar, 905100 Murfatlar, Romania; (V.A.); (A.R.)
| | | |
Collapse
|
36
|
Gowda D, Shekhar C, B. Gowda SG, Chen Y, Hui SP. Crosstalk between Lipids and Non-Alcoholic Fatty Liver Disease. LIVERS 2023; 3:687-708. [DOI: 10.3390/livers3040045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), a complex liver disorder that can result in non-alcoholic steatohepatitis, cirrhosis, and liver cancer, is the accumulation of fat in the liver seen in people due to metabolic dysfunction. The pathophysiology of NAFLD is influenced by several variables, such as metabolic dysregulation, oxidative stress, inflammation, and genetic susceptibility. This illness seriously threatens global health because of its link to obesity, insulin resistance, type 2 diabetes, and other metabolic disorders. In recent years, lipid–NAFLD crosstalk has drawn a lot of interest. Through numerous methods, lipids have been connected to the onset and advancement of the illness. The connection between lipids and NAFLD is the main topic of the current review, along with the various therapeutic targets and currently available drugs. The importance of hepatic lipid metabolism in the progression of NAFLD is summarized with the latest results in the field.
Collapse
Affiliation(s)
- Divyavani Gowda
- Faculty of Health Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Chandra Shekhar
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Siddabasave Gowda B. Gowda
- Faculty of Health Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Graduate School of Global Food Resources, Hokkaido University, Sapporo 060-0812, Japan
| | - Yifan Chen
- Faculty of Health Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo 060-0812, Japan
| |
Collapse
|
37
|
Sturgeon JP, Njunge JM, Bourke CD, Gonzales GB, Robertson RC, Bwakura-Dangarembizi M, Berkley JA, Kelly P, Prendergast AJ. Inflammation: the driver of poor outcomes among children with severe acute malnutrition? Nutr Rev 2023; 81:1636-1652. [PMID: 36977352 PMCID: PMC10639108 DOI: 10.1093/nutrit/nuad030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Severe acute malnutrition (SAM) is the most life-threatening form of undernutrition and underlies at least 10% of all deaths among children younger than 5 years in low-income countries. SAM is a complex, multisystem disease, with physiological perturbations observed in conjunction with the loss of lean mass, including structural and functional changes in many organ systems. Despite the high mortality burden, predominantly due to infections, the underlying pathogenic pathways remain poorly understood. Intestinal and systemic inflammation is heightened in children with SAM. Chronic inflammation and its consequent immunomodulation may explain the increased morbidity and mortality from infections in children with SAM, both during hospitalization and in the longer term after discharge. Recognition of the role of inflammation in SAM is critical in considering new therapeutic targets in this disease, which has not seen a transformational approach to treatment for several decades. This review highlights the central role of inflammation in the wide-ranging pathophysiology of SAM, as well as identifying potential interventions that have biological plausibility based on evidence from other inflammatory syndromes.
Collapse
Affiliation(s)
- Jonathan P Sturgeon
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - James M Njunge
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Claire D Bourke
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - Gerard Bryan Gonzales
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
| | - Ruairi C Robertson
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | | | - James A Berkley
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Paul Kelly
- is with the Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
38
|
Robles-Matos N, Radaelli E, Simmons RA, Bartolomei MS. Preconception and developmental DEHP exposure alter liver metabolism in a sex-dependent manner in adult mouse offspring. Toxicology 2023; 499:153640. [PMID: 37806616 PMCID: PMC10842112 DOI: 10.1016/j.tox.2023.153640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/30/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
Environmental exposure to endocrine disrupting chemicals (EDCs) during critical periods of development is associated with an increased risk of metabolic diseases, including hepatic steatosis and obesity. Di-2-ethylhexyl-phthalate (DEHP) is an EDC strongly associated with these metabolic abnormalities. DEHP developmental windows of susceptibility are unknown yet have important public health implications. The purpose of this study was to identify these windows of susceptibility and determine whether developmental DEHP exposure alters hepatic metabolism later in life. Dams were exposed to control or feed containing human exposure relevant doses of DEHP (50 μg/kg BW/d) and high dose DEHP (10 mg/kg BW/d) from preconception until weaning or only exposed to DEHP during preconception. Post-weaning, all offspring were fed a control diet throughout adulthood. Using the Metabolon Untargeted Metabolomics platform, we identified 148 significant metabolites in female adult livers that were altered by preconception-gestation-lactation DEHP exposure. We found a significant increase in the levels of acylcarnitines, diacylglycerols, sphingolipids, glutathione, purines, and pyrimidines in DEHP-exposed female livers compared to controls. These changes in fatty acid oxidation and oxidative stress-related metabolites were correlated with hepatic changes including microvesicular steatosis, hepatocyte swelling, inflammation. In contrast to females, we observed fewer metabolic alterations in male offspring, which were uniquely found in preconception-only low dose DEHP exposure group. Although we found that preconception-gestational-lactation exposure causes the most liver pathology, we surprisingly found preconception exposure linked to an abnormal liver metabolome. We also found that two doses exhibited non-monotonic DEHP-induced changes in the liver. Collectively, these findings suggest that metabolic changes in the adult liver of offspring exposed periconceptionally to DHEP depends on the timing of exposure, dose, and sex.
Collapse
Affiliation(s)
- Nicole Robles-Matos
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Enrico Radaelli
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca A Simmons
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Marisa S Bartolomei
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Zhou T, Song G, Tian D, Liu Q, Shen J, Yang X, Zhao P. Nuciferine relieves type 2 diabetes mellitus via enhancing GLUT4 expression and translocation. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
40
|
Zhang Z, Lu W, Liu P, Li M, Ge X, Yu B, Wu Z, Liu G, Ding N, Cui B, Chen X. Microbial modifications with Lycium barbarum L. oligosaccharides decrease hepatic fibrosis and mitochondrial abnormalities in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155068. [PMID: 37690228 DOI: 10.1016/j.phymed.2023.155068] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/03/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Lycium barbarum L. is a typical Chinese herbal and edible plant and are now consumed globally. Low molecular weight L. barbarum L. oligosaccharides (LBO) exhibit better antioxidant activity and gastrointestinal digestibility in vitro than high molecular weight polysaccharides. However, the LBO on the treatment of liver disease is not studied. PURPOSE Modification of the gut microbial ecosystem by LBO is a promising treatment for liver fibrosis. STUDY DESIGN AND METHODS Herein, LBO were prepared and characterized. CCl4-treated mice were orally gavaged with LBO and the effects on hepatic fibrosis and mitochondrial abnormalities were evaluated according to relevant indicators (gut microbiota, faecal metabolites, and physiological and biochemical indices). RESULTS The results revealed that LBO, a potential prebiotic source, is a pyranose cyclic oligosaccharide possessing α-glycosidic and β-glycosidic bonds. Moreover, LBO supplementation restored the configuration of the bacterial community, enhanced the proliferation of beneficial species in the gastrointestinal tract (e.g., Bacillus, Tyzzerella, Fournierella and Coriobacteriaceae UCG-002), improved microbial metabolic alterations (i.e., carbohydrate metabolism, vitamin metabolism and entero-hepatic circulation), and increased antioxidants, including doxepin, in mice. Finally, LBO administration reduced serum inflammatory cytokine and hepatic hydroxyproline levels, improved intestinal and hepatic mitochondrial functions, and ameliorated mouse liver fibrosis. CONCLUSION These findings indicate that LBO can be utilized as a prebiotic and has a remarkable ability to mitigate liver fibrosis.
Collapse
Affiliation(s)
- Zheng Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Wenjia Lu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Pengfei Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Mengjie Li
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Xinyi Ge
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Bin Yu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Zhengzong Wu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Guimei Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Nannan Ding
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Bo Cui
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| | - Xiao Chen
- College of Health Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250353, China.
| |
Collapse
|
41
|
Reimers N, Do Q, Zhang R, Guo A, Ostrander R, Shoji A, Vuong C, Xu L. Tracking the Metabolic Fate of Exogenous Arachidonic Acid in Ferroptosis Using Dual-Isotope Labeling Lipidomics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:2016-2024. [PMID: 37523294 PMCID: PMC10487598 DOI: 10.1021/jasms.3c00181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 08/02/2023]
Abstract
Lipid metabolism is implicated in a variety of diseases, including cancer, cell death, and inflammation, but lipidomics has proven to be challenging due to the vast structural diversity over a narrow range of mass and polarity of lipids. Isotope labeling is often used in metabolomics studies to follow the metabolism of exogenously added labeled compounds because they can be differentiated from endogenous compounds by the mass shift associated with the label. The application of isotope labeling to lipidomics has also been explored as a method to track the metabolism of lipids in various disease states. However, it can be difficult to differentiate a single isotopically labeled lipid from the rest of the lipidome due to the variety of endogenous lipids present over the same mass range. Here we report the development of a dual-isotope deuterium labeling method to track the metabolic fate of exogenous polyunsaturated fatty acids, e.g., arachidonic acid, in the context of ferroptosis using hydrophilic interaction-ion mobility-mass spectrometry (HILIC-IM-MS). Ferroptosis is a type of cell death that is dependent on lipid peroxidation. The use of two isotope labels rather than one enables the identification of labeled species by a signature doublet peak in the resulting mass spectra. A Python-based software, D-Tracer, was developed to efficiently extract metabolites with dual-isotope labels. The labeled species were then identified with LiPydomics based on their retention times, collision cross section, and m/z values. Changes in exogenous AA incorporation in the absence and presence of a ferroptosis inducer were elucidated.
Collapse
Affiliation(s)
- Noelle Reimers
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Quynh Do
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Rutan Zhang
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Angela Guo
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Ryan Ostrander
- Department
of Mechanical Engineering, University of
Washington, Seattle Washington 98195, United States
| | - Alyson Shoji
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Chau Vuong
- Department
of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Libin Xu
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
42
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic fatty liver disease worldwide, particularly in obese and type 2 diabetic individuals. Currently, there are no therapies for NAFLD that have been approved by the US Food and Drug Administration. Herein, we examine the rationale for using ω3 polyunsaturated fatty acids (PUFAs) in NAFLD therapy. This focus is based on the finding that NAFLD severity is associated with a reduction of hepatic C20-22 ω3 PUFAs. Because C20-22 ω3 PUFAs are pleiotropic regulators of cell function, loss of C20-22 ω3 PUFAs has the potential to significantly impact hepatic function. We describe NAFLD prevalence and pathophysiology as well as current NAFLD therapies. We also present evidence from clinical and preclinical studies that evaluated the capacity of C20-22 ω3 PUFAs to treat NAFLD. Given the clinical and preclinical evidence, dietary C20-22 ω3 PUFA supplementation has the potential to decrease human NAFLD severity by reducing hepatosteatosis and liver injury.
Collapse
Affiliation(s)
- Melinda H Spooner
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA;
| | - Donald B Jump
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA;
| |
Collapse
|
43
|
Bawden SJ, Hoad C, Kaye P, Stephenson M, Dolman G, James MW, Wilkes E, Austin A, Guha IN, Francis S, Gowland P, Aithal GP. Comparing magnetic resonance liver fat fraction measurements with histology in fibrosis: the difference between proton density fat fraction and tissue mass fat fraction. MAGMA (NEW YORK, N.Y.) 2023; 36:553-563. [PMID: 36538248 PMCID: PMC10468948 DOI: 10.1007/s10334-022-01052-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
OBJECTIVE Magnetic resonance spectroscopy (MRS) provides a powerful method of measuring fat fraction. However, previous studies have shown that MRS results give lower values compared with visual estimates from biopsies in fibrotic livers. This study investigated these discrepancies and considered whether a tissue water content correction, as assessed by MRI relaxometry, could provide better agreement. MATERIALS AND METHODS 110 patients were scanned in a 1.5 T Philips scanner and biopsies were obtained. Multiple echo MRS (30 × 30 × 30 mm volume) was used to determine Proton Density Fat Fraction (PDFF). Biopsies were assessed by visual assessment for fibrosis and steatosis grading. Digital image analysis (DIA) was also used to quantify fat fraction within tissue samples. T1 relaxation times were then used to estimate tissue water content to correct PDFF for confounding factors. RESULTS PDFF values across the four visually assessed steatosis grades were significantly less in the higher fibrosis group (F3-F4) compared to the lower fibrosis group (F0-F2). The slope of the linear regression of PDFF vs DIA fat fraction was ~ 1 in the low fibrosis group and 0.77 in the high fibrosis group. Correcting for water content based on T1 increased the gradient but it did not reach unity. DISCUSSION In fibrotic livers, PDFF underestimated fat fraction compared to DIA methods. Values were improved by applying a water content correction, but fat fractions were still underestimated.
Collapse
Affiliation(s)
- Stephen James Bawden
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2RD, UK.
- Sir Peter Mansfield Imaging Centre, SPMIC, University Park, Physics and Astronomy, University of Nottingham, Nottingham, UK.
| | - Caroline Hoad
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2RD, UK
| | - Philip Kaye
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2RD, UK
- Department of Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Mary Stephenson
- Clinical Imaging Research Centre (CIRC), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Grace Dolman
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2RD, UK
| | - Martin W James
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2RD, UK
| | - Emilie Wilkes
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2RD, UK
| | | | - Indra Neil Guha
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2RD, UK
| | - Susan Francis
- Sir Peter Mansfield Imaging Centre, SPMIC, University Park, Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre, SPMIC, University Park, Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Guruprasad P Aithal
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
44
|
Chatzi L, Baumert BO. Invited Perspective: PFAS and Dyslipidemia-The Perimenopausal Period as a Critical Time Window. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:81302. [PMID: 37552132 PMCID: PMC10408593 DOI: 10.1289/ehp13195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/06/2023] [Accepted: 07/06/2023] [Indexed: 08/09/2023]
Affiliation(s)
- Lida Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Brittney O. Baumert
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
45
|
Ikeda T, Ishikawa T, Ninagawa S, Okada T, Ono M, Mori K. Proteomic analysis of fatty liver induced by starvation of medaka fish larvae. Cell Struct Funct 2023; 48:123-133. [PMID: 37380437 PMCID: PMC10915113 DOI: 10.1247/csf.23014] [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: 03/05/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023] Open
Abstract
When medaka fish (Oryzias latipes) larvae are grown in the absence of exogenous nutrition, the liver becomes dark and positive to Oil Red O staining from 7 days post-hatch (dph). We determined the mechanism of this starvation-induced development of fatty liver by proteomic analysis using livers obtained from larvae grown in the presence or absence of 2% glucose at 5 dph. Results showed that changes in the expression levels of enzymes involved in glycolysis or the tricarboxylic acid cycle were modest, whereas the expression levels of enzymes involved in amino acid catabolism or β-oxidation of fatty acids were significantly elevated, suggesting that they become major energy sources under starvation conditions. Expression levels of enzymes for the uptake and β-oxidation of fatty acids as well as synthesis of triacylglycerol were elevated, whereas those for the synthesis of cholesterol as well as export of cholesterol and triacylglycerol were decreased under starvation conditions, which explains the accumulation of triacylglycerol in the liver. Our results provide the basis for future research to understand how gene malfunction(s) affects the development of fatty liver, which can lead to nonalcoholic steatohepatitis and then to liver cirrhosis.Key words: amino acid catabolism, β-oxidation, triacylglycerol, cholesterol, export.
Collapse
Affiliation(s)
- Tomoyo Ikeda
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Tokiro Ishikawa
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Satoshi Ninagawa
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Tetsuya Okada
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Masaya Ono
- National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Kazutoshi Mori
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
46
|
Jia L, Zhang L, Liu M, Ji H, Wen Z, Wang C. Mitochondrial Control for Healthy and Autoimmune T Cells. Cells 2023; 12:1800. [PMID: 37443834 PMCID: PMC10340733 DOI: 10.3390/cells12131800] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
T cells are critical players in adaptive immunity, driving the tissue injury and organ damage of patients with autoimmune diseases. Consequently, investigations on T cell activation, differentiation, and function are valuable in uncovering the disease pathogenesis, thus exploring promising therapeutics for autoimmune diseases. In recent decades, accumulating studies have pinpointed immunometabolism as the fundamental determinant in controlling T cell fate. Specifically, mitochondria, as a hub of intracellular metabolism, connect glucose, lipid, and amino acid metabolic pathways. Herein, we summarize metabolic adaptations of mitochondrial oxidative phosphorylation and the relevant glucose, lipid, and amino acid metabolism during T cell activation, differentiation, and function. Further, we focused on current updates of the molecular bases for metabolic reprogramming in autoimmune T cells and advances in exploring metabolic-targeted therapeutics against autoimmune diseases. This might facilitate the in-depth understanding of autoimmune pathogeneses and the clinical management of autoimmune diseases.
Collapse
Affiliation(s)
- Li Jia
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Mengdi Liu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Huiyan Ji
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Chunhong Wang
- Cyrus Tang Hematology Center, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| |
Collapse
|
47
|
Kim M, Kim SH, Choi JY, Park YJ. Investigating fatty liver disease-associated adverse outcome pathways of perfluorooctane sulfonate using a systems toxicology approach. Food Chem Toxicol 2023; 176:113781. [PMID: 37059384 DOI: 10.1016/j.fct.2023.113781] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
Adverse outcome pathway (AOP) frameworks help elucidate toxic mechanisms and support chemical regulation. AOPs link a molecular initiating event (MIE), key events (KEs), and an adverse outcome by key event relationships (KERs), which assess the biological plausibility, essentiality, and empirical evidence involved. Perfluorooctane sulfonate (PFOS), a hazardous poly-fluoroalkyl substance, demonstrates hepatotoxicity in rodents. PFOS may induce fatty liver disease (FLD) in humans; however, the underlying mechanism remains unclear. In this study, we evaluated the toxic mechanisms of PFOS-associated FLD by developing an AOP using publicly available data. We identified MIE and KEs by performing GO enrichment analysis on PFOS- and FLD-associated target genes collected from public databases. The MIEs and KEs were then prioritized by PFOS-gene-phenotype-FLD networks, AOP-helpFinder, and KEGG pathway analyses. Following a comprehensive literature review, an AOP was then developed. Finally, six KEs for the AOP of FLD were identified. This AOP indicated that toxicological processes initiated by SIRT1 inhibition led to SREBP-1c activation, de novo fatty acid synthesis, and fatty acid and triglyceride accumulation, culminating in liver steatosis. Our study provides insights into the toxic mechanism of PFOS-induced FLD and suggests approaches to assessing the risk of toxic chemicals.
Collapse
Affiliation(s)
- Moosoo Kim
- College of Pharmacy, Kyungsung University, Busan, 48434, Republic of Korea
| | - Sang Heon Kim
- College of Pharmacy, Kyungsung University, Busan, 48434, Republic of Korea
| | - Jun Yeong Choi
- College of Pharmacy, Kyungsung University, Busan, 48434, Republic of Korea
| | - Yong Joo Park
- College of Pharmacy, Kyungsung University, Busan, 48434, Republic of Korea.
| |
Collapse
|
48
|
Weiss JM, Palmieri EM, Gonzalez-Cotto M, Bettencourt IA, Megill EL, Snyder NW, McVicar DW. Itaconic acid underpins hepatocyte lipid metabolism in non-alcoholic fatty liver disease in male mice. Nat Metab 2023; 5:981-995. [PMID: 37308721 PMCID: PMC10290955 DOI: 10.1038/s42255-023-00801-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 04/06/2023] [Indexed: 06/14/2023]
Abstract
Itaconate, the product of the decarboxylation of cis-aconitate, regulates numerous biological processes. We and others have revealed itaconate as a regulator of fatty acid β-oxidation, generation of mitochondrial reactive oxygen species and the metabolic interplay between resident macrophages and tumors. In the present study, we show that itaconic acid is upregulated in human non-alcoholic steatohepatitis and a mouse model of non-alcoholic fatty liver disease. Male mice deficient in the gene responsible for itaconate production (immunoresponsive gene (Irg)-1) have exacerbated lipid accumulation in the liver, glucose and insulin intolerance and mesenteric fat deposition. Treatment of mice with the itaconate derivative, 4-octyl itaconate, reverses dyslipidemia associated with high-fat diet feeding. Mechanistically, itaconate treatment of primary hepatocytes reduces lipid accumulation and increases their oxidative phosphorylation in a manner dependent upon fatty acid oxidation. We propose a model whereby macrophage-derived itaconate acts in trans upon hepatocytes to modulate the liver's ability to metabolize fatty acids.
Collapse
Affiliation(s)
- Jonathan M Weiss
- Cancer Innovation Laboratory, Center for Cancer Research, NCI Frederick, Frederick, MD, USA
| | - Erika M Palmieri
- Cancer Innovation Laboratory, Center for Cancer Research, NCI Frederick, Frederick, MD, USA
| | - Marieli Gonzalez-Cotto
- Cancer Innovation Laboratory, Center for Cancer Research, NCI Frederick, Frederick, MD, USA
| | - Ian A Bettencourt
- Cancer Innovation Laboratory, Center for Cancer Research, NCI Frederick, Frederick, MD, USA
| | - Emily L Megill
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Nathaniel W Snyder
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Daniel W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, NCI Frederick, Frederick, MD, USA.
| |
Collapse
|
49
|
Kuo HCN, LaRussa Z, Xu FM, West K, Consitt L, Davidson WS, Liu M, Coschigano KT, Shi H, Lo CC. Apolipoprotein A4 Elevates Sympathetic Activity and Thermogenesis in Male Mice. Nutrients 2023; 15:2486. [PMID: 37299447 PMCID: PMC10255745 DOI: 10.3390/nu15112486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Long-chain fatty acids induce apolipoprotein A4 (APOA4) production in the small intestine and activate brown adipose tissue (BAT) thermogenesis. The increase in BAT thermogenesis enhances triglyceride clearance and insulin sensitivity. Acute administration of recombinant APOA4 protein elevates BAT thermogenesis in chow-fed mice. However, the physiological role of continuous infusion of recombinant APOA4 protein in regulating sympathetic activity, thermogenesis, and lipid and glucose metabolism in low-fat-diet (LFD)-fed mice remained elusive. The hypothesis of this study was that continuous infusion of mouse APOA4 protein would increase sympathetic activity and thermogenesis in BAT and subcutaneous inguinal white adipose tissue (IWAT), attenuate plasma lipid levels, and improve glucose tolerance. To test this hypothesis, sympathetic activity, BAT temperature, energy expenditure, body weight, fat mass, caloric intake, glucose tolerance, and levels of BAT and IWAT thermogenic and lipolytic proteins, plasma lipids, and markers of fatty acid oxidation in the liver in mice with APOA4 or saline treatment were measured. Plasma APOA4 levels were elevated, BAT temperature and thermogenesis were upregulated, and plasma triglyceride (TG) levels were reduced, while body weight, fat mass, caloric intake, energy expenditure, and plasma cholesterol and leptin levels were comparable between APOA4- and saline-treated mice. Additionally, APOA4 infusion stimulated sympathetic activity in BAT and liver but not in IWAT. APOA4-treated mice had greater fatty acid oxidation but less TG content in the liver than saline-treated mice had. Plasma insulin in APOA4-treated mice was lower than that in saline-treated mice after a glucose challenge. In conclusion, continuous infusion of mouse APOA4 protein stimulated sympathetic activity in BAT and the liver, elevated BAT thermogenesis and hepatic fatty acid oxidation, and consequently attenuated levels of plasma and hepatic TG and plasma insulin without altering caloric intake, body weight gain and fat mass.
Collapse
Affiliation(s)
- Hsuan-Chih N. Kuo
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Zachary LaRussa
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Flora Mengyang Xu
- Department of Biology, Miami University, Oxford, OH 45056, USA; (F.M.X.); (H.S.)
| | - Kathryn West
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
| | - Leslie Consitt
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
| | - William Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, USA; (W.S.D.); (M.L.)
| | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, USA; (W.S.D.); (M.L.)
| | - Karen T. Coschigano
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA; (F.M.X.); (H.S.)
| | - Chunmin C. Lo
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine and Diabetes Institute, Ohio University, Athens, OH 45701, USA; (H.-C.N.K.); (Z.L.); (K.W.); (L.C.); (K.T.C.)
| |
Collapse
|
50
|
Zhang J, Wang S, Wang J, Liu W, Gong H, Zhang Z, Lyu B, Yu H. Insoluble Dietary Fiber from Soybean Residue (Okara) Exerts Anti-Obesity Effects by Promoting Hepatic Mitochondrial Fatty Acid Oxidation. Foods 2023; 12:foods12102081. [PMID: 37238899 DOI: 10.3390/foods12102081] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/14/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Numerous investigations have shown that insoluble dietary fiber (IDF) has a potentially positive effect on obesity due to a high-fat diet (HFD). Our previous findings based on proteomic data revealed that high-purity IDF from soybean residue (okara) (HPSIDF) prevented obesity by regulating hepatic fatty acid synthesis and degradation pathways, while its intervention mechanism is uncharted. Consequently, the goal of this work is to find out the potential regulatory mechanisms of HPSIDF on hepatic fatty acid oxidation by determining changes in fatty acid oxidation-related enzymes in mitochondria and peroxisomes, the production of oxidation intermediates and final products, the composition and content of fatty acids, and the expression levels of fatty acid oxidation-related proteins in mice fed with HFD. We found that supplementation with HPSIDF significantly ameliorated body weight gain, fat accumulation, dyslipidemia, and hepatic steatosis caused by HFD. Importantly, HPSIDF intervention promotes medium- and long-chain fatty acid oxidation in hepatic mitochondria by improving the contents of acyl-coenzyme A oxidase 1 (ACOX1), malonyl coenzyme A (Malonyl CoA), acetyl coenzyme A synthase (ACS), acetyl coenzyme A carboxylase (ACC), and carnitine palmitoyl transferase-1 (CPT-1). Moreover, HPSIDF effectively regulated the expression levels of proteins involved with hepatic fatty acid β-oxidation. Our study indicated that HPSIDF treatment prevents obesity by promoting hepatic mitochondrial fatty acid oxidation.
Collapse
Affiliation(s)
- Jiarui Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Sainan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Junyao Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Wenhao Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Hao Gong
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Zhao Zhang
- Sinoglory Health Food Co., Ltd., Liaocheng 252000, China
| | - Bo Lyu
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Hansong Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|