1
|
Tsuboi A, Khanom H, Kawabata R, Matsui T, Murakami S, Ito T. Taurine ameliorates cellular senescence associated with an increased hydrogen sulfide and a decreased hepatokine, IGFBP-1, in CCl4-induced hepatotoxicity in mice. Redox Biol 2025; 83:103640. [PMID: 40286436 PMCID: PMC12059708 DOI: 10.1016/j.redox.2025.103640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
This study investigated the protective effects of taurine against cellular senescence and hepatokine secretion in a mouse model of carbon tetrachloride (CCl4)-induced chronic liver injury. Oral taurine administration by tap water containing 3 % taurine significantly attenuated liver damage, as evidenced by reduced serum AST, ALT level and hepatic lipid peroxidation. Importantly, hepatic taurine level is reduced in CCl4-induced injury model, while taurine administration recovered it. Moreover, taurine administration decreased the numbers of p21-positive senescent cells in liver tissue of CCl4-treated mice. Taurine increases hydrogen sulfide (H2S) in liver of normal mice, suggesting anti-oxidative role through H2S production by taurine. Furthermore, inhibition of CTH, which is an enzyme responsible for H2S production from cysteine, by propagylglycine attenuated malondialdehyde-lowering effect of taurine in liver of CCl4-treated mice. Moreover, we found taurine treatment lowers insulin-like growth factor binding protein-1 (IGFBP-1) in liver of normal mice. Importantly, while chronic CCl4 injection caused an induction of IGFBP-1, taurine administration blocked it. These findings suggest that taurine exerts its protective effects by attenuating cellular senescence, which is associated with enhancing H2S production and inhibiting IGFBP-1 expression. This study highlights the potential of taurine as a therapeutic strategy for mitigating chronic liver injury by producing H2S and targeting IGFBP1.
Collapse
Affiliation(s)
| | | | | | | | | | - Takashi Ito
- Corresponding author. Department of Biosciences and Biotechnology, Fukui Prefectural University. 4-1-1 Matsuokakenjojima, Eiheiji-cho, Yoshida-gun, Fukui, 910-1195, Japan.
| |
Collapse
|
2
|
Mayengbam S, Raman M, Parnell JA, Eksteen B, Lambert JE, Eller LK, Nicolucci AC, Aktary ML, Reimer RA. Effects of combined prebiotic fiber supplementation and weight loss counseling in adults with metabolic dysfunction-associated steatotic liver disease: a randomized controlled trial. Eur J Nutr 2025; 64:144. [PMID: 40172664 DOI: 10.1007/s00394-025-03660-7] [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: 10/08/2024] [Accepted: 03/22/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE Our aim was to examine the effects of combined prebiotic fiber supplementation and weight loss counseling on liver fat, body composition, subjective appetite, serum metabolomics, and intestinal microbiota in adults with MASLD. METHODS In a double blind, placebo-controlled trial, adult participants aged 18-70 years old with MASLD were randomized to receive prebiotic (oligofructose-enriched inulin, 16 g/day; n = 22) or isocaloric placebo (maltodextrin; n = 20) for 24 weeks alongside weight loss counseling from a registered dietitian. Primary outcomes were change in intrahepatic fat % (IHF%) and hepatic injury from baseline to 24 weeks. Secondary outcomes included body composition, subjective appetite, serum lipids and cytokines, fecal microbiota, and serum metabolomics. RESULTS At baseline, participants had IHF of 14.4 ± 8.4%. The change in IHF from baseline to 24 weeks did not differ between prebiotic and placebo. Prebiotic participants had a greater decrease (p = 0.029) in percent trunk fat compared to placebo. Compared to placebo, prebiotic significantly decreased desire to eat and hunger ratings over the course of the intervention. Fecal microbiota analysis showed a significant increase in Bifidobacterium abundance with prebiotic. A pathway analysis based on untargeted serum metabolomics revealed a downregulation of taurine and hypotaurine metabolism in the placebo group which was conserved in the prebiotic group. CONCLUSION Adding prebiotic fiber supplementation to weight loss counseling for adults with MASLD enhanced reductions in trunk fat and had a beneficial effect on subjective appetite compared to placebo. Improvements in fecal microbial profile and taurine metabolism revealed specific beneficial effects of prebiotics in the management of MASLD. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov/study/NCT02568605.
Collapse
Affiliation(s)
- Shyamchand Mayengbam
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Maitreyi Raman
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Jill A Parnell
- Department of Health and Physical Education, Mount Royal University, Calgary, AB, Canada
| | | | - Jennifer E Lambert
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Lindsay K Eller
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Alissa C Nicolucci
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Michelle L Aktary
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
3
|
Houttu V, Boulund U, Troelstra M, Csader S, Stols-Gonçalves D, Mak AL, Dijk AMV, Bouts J, Winkelmeijer M, Verdoes X, van den Berg-Faay S, Lek D, Ronteltap T, de Haan F, Jorstad H, Männistö V, Savonen K, Pentikäinen H, Hanhineva K, Babu AF, Panagiotou G, van Delden O, Verheij J, Doukas M, Nederveen A, Schwab U, Grefhorst A, Nieuwdorp M, Holleboom AG. Deep phenotyping of patients with MASLD upon high-intensity interval training. JHEP Rep 2025; 7:101289. [PMID: 40051412 PMCID: PMC11883402 DOI: 10.1016/j.jhepr.2024.101289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 03/09/2025] Open
Abstract
Background & Aims Exercise is a key component of lifestyle management in patients with metabolic dysfunction-associated steatotic liver disease (MASLD), but neither its therapeutic effect on the active stage of the disease, that is metabolic dysfunction-associated steatohepatitis (MASH) nor the mediating mechanisms have been characterized. Therefore, we performed multi-omic phenotyping of patients with MASLD-MASH on an exercise program. Methods Fifteen patients with MASLD conducted high-intensity interval training (HIIT) combined with home-based training for 12 weeks. MASLD was evaluated using histology, transient elastography, and multiparametric magnetic resonance imaging (MRI) before and after the intervention. Change in maximal oxygen consumption (VO2max) and MRI-determined liver fat were compared with a control group of patients with MASLD (n = 22). RNA sequencing was performed on liver, muscle, and fat biopsies of patients in the exercise group. Stool was analyzed by shotgun metagenomics and untargeted metabolomics was performed on plasma, urine, adipose, and stool. Results HIIT increased VO2max by 10.1% and improved mitochondrial metabolism in skeletal muscle, indicating improved cardiorespiratory fitness and adherence. VO2max increased significantly in the exercise group compared with controls. Histologically, no reduction in steatosis, MASH, or liver fibrosis was observed; however, transient elastography tended to improve. MRI-determined liver fat did not change in the exercise group compared with controls. HIIT induced changes in mRNA expression of genes related to beiging of adipose tissue and fibrogenesis in liver. In addition, specific gut microbial taxa and metabolites changed. Conclusions HIIT increased cardiorespiratory fitness and induced beneficial gene expression changes in muscle, adipose tissue, and liver, but without translation into histological improvement of MASLD. Longer exercise intervention trials are warranted to validate or refute current recommendations for exercise as a cornerstone treatment for MASLD-MASH. Impact and implications Despite exercise being considered as a key component of lifestyle management for steatotic liver disease, neither the clinical effects nor the mechanisms involved are completely understood. We show that a high-intensity interval training (HIIT) program in 15 patients with metabolic dysfunction-associated steatotic liver disease (MASLD) improved cardiorespiratory fitness, compared with 22 control patients with MASLD who did not participate in an exercise program, however, it did not improve MASLD. HIIT induced a positive effect on fat tissue and muscle metabolism which was accompanied with changes in certain gut bacteria and metabolites in blood and urine. These findings improve our understanding of the effects of exercise on the whole-body metabolism in relation to steatotic liver disease. As such, this study provides a basis for future exercise interventions in patients with MASLD, required to thoroughly test current guideline advice for exercise as a cornerstone treatment for MASLD of all stages. Clinical trial registry Dutch Trial Register (registration number NL7932).
Collapse
Affiliation(s)
- Veera Houttu
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ulrika Boulund
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marian Troelstra
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne Csader
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Daniela Stols-Gonçalves
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne Linde Mak
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Marieke van Dijk
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Julia Bouts
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maaike Winkelmeijer
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Xanthe Verdoes
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandra van den Berg-Faay
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Donne Lek
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Ted Ronteltap
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Ferdinand de Haan
- Polifysiek, Amsterdam University of Applied Science, Amsterdam, The Netherlands
| | - Harald Jorstad
- Department of Cardiology, Amsterdam Movement Sciences, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland, and Kuopio University Hospital, Kuopio, Finland
| | - Kai Savonen
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | | | - Kati Hanhineva
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Afekta Technologies Ltd., Kuopio, Finland
- Department of Life Technologies, Food Chemistry, and Food Development Unit, University of Turku, Turku, Finland
| | - Ambrin Farizah Babu
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Afekta Technologies Ltd., Kuopio, Finland
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research, and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
- Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Otto van Delden
- Department of Interventional Radiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Michial Doukas
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Aart Nederveen
- Department of Radiology, and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ursula Schwab
- School of Medicine, Institute of Public Health, and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Aldo Grefhorst
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan Georgius Holleboom
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Hu X, Xu T, Chen Y, Zhang Q, Tang L, Zheng L, Wang C, Wang P, Dong S, Wang R, Zhang S, Zhang Q, Xie HQ, Xu L, Zhao B. Comprehensive metabolic profiling of dioxin-like compounds exposure in laying hens: Implications for toxicity assessment. J Environ Sci (China) 2025; 148:107-115. [PMID: 39095149 DOI: 10.1016/j.jes.2024.01.010] [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: 08/07/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 08/04/2024]
Abstract
The evaluation of toxicity related to polychlorinated dibenzo-p-dioxins and furans (PCDD/Fs) and dioxin-like polychlorinated biphenyls (DL-PCBs) is crucial for a comprehensive risk assessment in real-world exposure scenarios. This study employed a controlled feeding experiment to investigate the metabolic effects of dioxin-like compounds (DLCs) on laying hens via feed exposure. Diets enriched with two concentrations (1.17 and 5.13 pg toxic equivalents (TEQ)/g dry weight (dw)) were administered over 14 days, followed by 28 days of clean feed. Metabolomics analyses of blood samples revealed significant metabolic variations between PCDD/Fs and DL-PCBs exposed groups and controls, reflecting the induced metabolic disruption. Distinct changes were observed in sphingosine, palmitoleic acid, linoleate, linolenic acid, taurocholic acid, indole acrylic acid, and dibutyl phthalate levels, implying possible connections between PCDD/Fs and DL-PCBs toxic effects and energy-neuronal imbalances, along with lipid accumulation and anomalous amino acid metabolism, impacting taurine metabolism. Moreover, we identified three differential endogenous metabolites-L-tryptophan, indole-3-acetaldehyde, and indole acrylic acid-as potential ligands for the aryl hydrocarbon receptor (AhR), suggesting their role in mediating PCDD/Fs and DL-PCBs toxicity. This comprehensive investigation provides novel insights into the metabolic alterations induced by PCDD/Fs and DL-PCBs in laying hens, thereby enhancing our ability to assess risks associated with their exposure in human populations.
Collapse
Affiliation(s)
- Xiaoxu Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tong Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yangsheng Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Lijuan Tang
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Liping Zheng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chu Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pu Wang
- Hubei Key Laboratory of Industrial Fume and Dust Pollution Control, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Shujun Dong
- Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Ruiguo Wang
- Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Su Zhang
- Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Qinghua Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
5
|
Svobodová G, Horní M, Velecká E, Boušová I. Metabolic dysfunction-associated steatotic liver disease-induced changes in the antioxidant system: a review. Arch Toxicol 2025; 99:1-22. [PMID: 39443317 PMCID: PMC11748479 DOI: 10.1007/s00204-024-03889-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a heterogeneous condition characterized by liver steatosis, inflammation, consequent fibrosis, and cirrhosis. Chronic impairment of lipid metabolism is closely related to oxidative stress, leading to cellular lipotoxicity, mitochondrial dysfunction, and endoplasmic reticulum stress. The detrimental effect of oxidative stress is usually accompanied by changes in antioxidant defense mechanisms, with the alterations in antioxidant enzymes expression/activities during MASLD development and progression reported in many clinical and experimental studies. This review will provide a comprehensive overview of the present research on MASLD-induced changes in the catalytic activity and expression of the main antioxidant enzymes (superoxide dismutases, catalase, glutathione peroxidases, glutathione S-transferases, glutathione reductase, NAD(P)H:quinone oxidoreductase) and in the level of non-enzymatic antioxidant glutathione. Furthermore, an overview of the therapeutic effects of vitamin E on antioxidant enzymes during the progression of MASLD will be presented. Generally, at the beginning of MASLD development, the expression/activity of antioxidant enzymes usually increases to protect organisms against the increased production of reactive oxygen species. However, in advanced stage of MASLD, the expression/activity of several antioxidants generally decreases due to damage to hepatic and extrahepatic cells, which further exacerbates the damage. Although the results obtained in patients, in various experimental animal or cell models have been inconsistent, taken together the importance of antioxidant enzymes in MASLD development and progression has been clearly shown.
Collapse
Affiliation(s)
- Gabriela Svobodová
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05, Hradec Králové, Czech Republic
| | - Martin Horní
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05, Hradec Králové, Czech Republic
| | - Eva Velecká
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05, Hradec Králové, Czech Republic
| | - Iva Boušová
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05, Hradec Králové, Czech Republic.
| |
Collapse
|
6
|
Li X, Zheng S, Li H, Liu J, Yang F, Zhao X, Liang Y. 16S rRNA Sequencing and Metabolomics to Analyze Correlation Between Fecal Flora and Metabolites of Squabs and Parent Pigeons. Animals (Basel) 2025; 15:74. [PMID: 39795017 PMCID: PMC11718954 DOI: 10.3390/ani15010074] [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: 11/07/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Intestinal microorganisms are essential for maintaining homeostasis, health, and development, playing a critical role in nutrient digestion, growth, and exercise performance in pigeons. In young pigeons, the gut microbiota is primarily acquired through pigeon milk, meaning the microbial composition of parent pigeons directly influences microbial colonization in squabs. However, research on the correlation between the gut microbial diversity of parent pigeons and their offspring remains scarce. This study investigates the fecal microbiota and metabolites of 10 pairs of parent pigeons and 20 squabs raised under a 2 + 2 system. Fecal samples were collected at 15 days of age, and differences in the microbiota and metabolites between the two groups were analyzed using 16S rRNA sequencing and LC-MS/MS. The results showed the following: (1) Squabs exhibited significantly lower α diversity, with a reduction in their Chao1 index and observed OTUs compared to the parent pigeons. (2) Firmicutes dominated the fecal microbiota in both groups, but parent pigeon feces showed a notably higher abundance of Proteobacteria. At the family level, 10 distinct families were identified, with 9 at the genus level and 4 at the species level. (3) A LEfSe analysis identified 16 significantly different bacterial species in the parent pigeons and 7 in the squabs. Functional gene abundance was highest in the metabolism, genetic information processing, and environmental information processing pathways. (4) An LC-MS/MS analysis in cationic mode identified 218 metabolites, with 139 upregulated and 79 downregulated in the squabs relative to the parents. These metabolites were primarily concentrated in five functional categories and enriched in 33 pathways, 2 of which showed significant differences. In conclusion, significant differences in both the α and β diversity of fecal microbiota were observed between squabs and parent pigeons, with similar bacterial species but marked differences in abundance. Metabolite analysis revealed greater richness in the parent pigeon feces. These findings suggest that future gut modulation using beneficial bacteria, such as probiotics, could potentially enhance host health based on microbial and metabolite compositions.
Collapse
Affiliation(s)
- Xiaobin Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830091, China; (X.L.); (S.Z.); (F.Y.); (X.Z.); (Y.L.)
| | - Shengchen Zheng
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830091, China; (X.L.); (S.Z.); (F.Y.); (X.Z.); (Y.L.)
| | - Haiying Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830091, China; (X.L.); (S.Z.); (F.Y.); (X.Z.); (Y.L.)
| | - Jiajia Liu
- Moyu Blue Sea Pigeon Industry Co., Ltd., Hetian 848101, China;
| | - Fan Yang
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830091, China; (X.L.); (S.Z.); (F.Y.); (X.Z.); (Y.L.)
| | - Xiaoyu Zhao
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830091, China; (X.L.); (S.Z.); (F.Y.); (X.Z.); (Y.L.)
| | - Yafei Liang
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830091, China; (X.L.); (S.Z.); (F.Y.); (X.Z.); (Y.L.)
- Moyu Blue Sea Pigeon Industry Co., Ltd., Hetian 848101, China;
| |
Collapse
|
7
|
Zhou Y, Bai F, Xiao R, Chen M, Sun Y, Ye J. Proteomics and Its Combined Analysis with Transcriptomics: Liver Fat-Lowering Effect of Taurine in High-Fat Fed Grouper ( Epinephelus coioides). Animals (Basel) 2024; 14:2039. [PMID: 39061501 PMCID: PMC11274106 DOI: 10.3390/ani14142039] [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: 06/17/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
In order to understand the intervention effect of taurine on liver fat deposition induced by high fat intake in the orange-spotted grouper (Epinephelus coioides), we performed proteomic analysis and association analysis with previously obtained transcriptomic data. Three isoproteic (47% crude protein) diets were designed to contain two levels of fat and were named as the 10% fat diet (10F), 15% fat diet (15F), and 15% fat with 1% taurine (15FT). The 10F diet was used as the control diet. After 8 weeks of feeding, the 15F diet exhibited comparable weight gain, feed conversion ratio, and hepatosomatic index as the 10F diet, but the former increased liver fat content vs. the latter. Feeding with the 15FT diet resulted in an improvement in weight gain and a reduction in feed conversion ratio, hepatosomatic index, and liver fat content compared with feeding the 15F diet. When comparing liver proteomic data between the 15F and 15FT groups, a total of 133 differentially expressed proteins (DEPs) were identified, of which 51 were upregulated DEPs and 82 were downregulated DEPs. Among these DEPs, cholesterol 27-hydroxylase, phosphatidate phosphatase LPIN, phosphatidylinositol phospholipase C, and 6-phosphofructo-2-kinase were further screened out and were involved in primary bile acid biosynthesis, glycerolipid metabolism, the phosphatidylinositol signaling system, and the AMPK signaling pathway as key DEPs in terms of alleviating liver fat deposition of taurine in high-fat fed fish. With the association analysis of transcriptomic and proteomic data through KEGG, three differentially expressed genes (atp1a, arf1_2, and plcd) and four DEPs (CYP27α1, LPIN, PLCD, and PTK2B) were co-enriched into five pathways related to fat metabolism including primary bile acid synthesis, bile secretion, glycerolipid metabolism, phospholipid D signaling, or/and phosphatidylinositol signaling. The results showed that dietary taurine intervention could trigger activation of bile acid biosynthesis and inhibition of triglyceride biosynthesis, thereby mediating the liver fat-lowering effects in high-fat fed orange-spotted grouper. The present study contributes some novel insight into the liver fat-lowering effects of dietary taurine in high-fat fed groupers.
Collapse
Affiliation(s)
| | | | | | | | | | - Jidan Ye
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College of Jimei University, Xiamen 361021, China; (Y.Z.); (F.B.); (R.X.); (M.C.); (Y.S.)
| |
Collapse
|
8
|
Ahmed K, Choi HN, Park JS, Kim YG, Bae MK, Yim JE. Taurine supplementation alters gene expression profiles in white adipose tissue of obese C57BL/6J mice: Inflammation and lipid synthesis perspectives. Heliyon 2024; 10:e23288. [PMID: 38192788 PMCID: PMC10771985 DOI: 10.1016/j.heliyon.2023.e23288] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024] Open
Abstract
This work aimed to identify the mechanisms by which taurine exerts its anti-obesity effects in the C57BL/6J ob/ob mice model and determine if taurine supplementation increases the amelioration of inflammation and lipogenesis linked genes in the adipose and liver tissues. Three groups of C57BL/6J mice were fed a standard chow diet for a period of 10 weeks the C57BL/6J normal group, the C57BL/6J ob/ob negative control group with no taurine intake and C57BL/6J ob/ob taurine group with taurine intake. Real time PCR was used to examine the gene expression profile in the experimental groups intrascapular brown adipose tissue (BAT), inguinal white adipose tissue (WAT) and liver. TNF-alpha, Ccl2, Adgre and illb genes that are associated with inflammation were found to have varying level of expression in the three tissues. In comparison to BAT and liver these genes were expressed at a much lower level in WAT, with enhanced serum adiponectin levels.
Collapse
Affiliation(s)
- Kainat Ahmed
- Interdisciplinary Program in Senior Human Ecology, Changwon National University, Changwon, Korea
| | - Ha-Neul Choi
- Department of Food and Nutrition, Changwon National University, Changwon, Korea
| | - Ji-sook Park
- Department of Food and Nutrition, Changwon National University, Changwon, Korea
| | - Yu-Gyeong Kim
- Department of Food and Nutrition, Changwon National University, Changwon, Korea
| | - Min Kyung Bae
- Interdisciplinary Program in Senior Human Ecology, Changwon National University, Changwon, Korea
- Department of Food and Nutrition, Changwon National University, Changwon, Korea
| | - Jung-Eun Yim
- Interdisciplinary Program in Senior Human Ecology, Changwon National University, Changwon, Korea
- Department of Food and Nutrition, Changwon National University, Changwon, Korea
| |
Collapse
|
9
|
Liu H, Niu T, Qiu G, Cui S, Zhang D. Taurine promotes insulin synthesis by enhancing Isl-1 expression through miR-7a/RAF1/ERK1/2 pathway. In Vitro Cell Dev Biol Anim 2024; 60:23-35. [PMID: 38117455 DOI: 10.1007/s11626-023-00835-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/04/2023] [Indexed: 12/21/2023]
Abstract
It has been well established that the circulating taurine affects the insulin synthesis in pancreatic islet β-cells, whereas miR-7a and LIM-homeodomain transcription factor Isl-1 are important intracellular factors regulating insulin transcription and synthesis. However, it still remains unknown whether taurine regulates insulin synthesis by affecting miR-7a and/or Isl-1 expressions in mouse pancreatic islet β-cells. The present study was thus proposed to identify the effects of taurine on the expressions of miR-7a and/or Isl-1 and their relations to insulin synthesis in mouse pancreatic islet β-cells by using miR-7a2 knockout (KO) and taurine transporter (TauT) KO mouse models and the related in vitro experiments. The results demonstrated that taurine supplement significantly decreased the pancreas miR-7a expression, but sharply upregulated the pancreas Isl-1 and insulin expressions, and serum insulin levels. However, the enhanced effects of taurine on Isl-1 expression and insulin synthesis were mitigated in the TauT KO and miR-7a2 KO mice. In addition, our results confirmed that taurine markedly increased pancreas RAF1 and ERK1/2 expressions. Collectively, the present study firstly demonstrates that taurine regulates insulin synthesis through TauT/miR-7a/RAF1/ERK1/2/Isl-1 signaling pathway, which are crucial for our understanding the mechanisms of taurine affecting insulin synthesis, and also potential for establishing the therapeutic strategies for diabetes and the diseases related to metabolism.
Collapse
Affiliation(s)
- Hui Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Tongjuan Niu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Guobin Qiu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Di Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
Yu Z, Cheng M, Luo S, Wei J, Song T, Gong Y, Zhou Z. Comparative Lipidomics and Metabolomics Reveal the Underlying Mechanisms of Taurine in the Alleviation of Nonalcoholic Fatty Liver Disease Using the Aged Laying Hen Model. Mol Nutr Food Res 2023; 67:e2200525. [PMID: 37909476 DOI: 10.1002/mnfr.202200525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 06/18/2023] [Indexed: 11/03/2023]
Abstract
SCOPE Aged laying hen is recently suggested as a more attractive animal model than rodent for studying nonalcoholic fatty liver disease (NAFLD) of humans. This study aims to reveal effects and metabolic regulation mechanisms of taurine alleviating NAFLD by using the aged laying hen model. METHODS AND RESULTS Liver histomorphology and biochemical indices show 0.02% taurine effectively alleviated fat deposition and liver damage. Comparative liver lipidomics and gene expressions analyses reveal taurine promoted lipolysis, fatty acids oxidation, lipids transport, and reduced oxidative stress in liver. Furthermore, comparative serum metabolomics screen six core metabolites negatively correlated with NAFLD, including linoleic acid, gamma-linolenic acid, pantothenate, L-methionine, 2-methylbutyroylcarnitine, L-carnitine; and two core metabolites positively correlated with NAFLD, including lysophosphatidylcholine (14:0/0:0) and lysophosphatidylcholine (16:0/0:0). Metabolic pathway analysis reveals taurine mainly regulated linoleic acid metabolism, cysteine and methionine metabolism, carnitine metabolism, pantothenic acid and coenzyme A biosynthesis metabolism, and glycerophospholipid metabolism to up-adjust levels of six negatively correlated metabolites and down-adjust two positively correlated metabolites for alleviating NAFLD of aged hens. CONCLUSION This study firstly reveals underlying metabolic mechanisms of taurine alleviating NAFLD using the aged hen model, thereby laying the foundation for taurine's application in the prevention of NAFLD in both human and poultry.
Collapse
Affiliation(s)
- Zhengwang Yu
- Department of Animal Nutrition and Feed Science, College of Animal Sciences & Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Shanghai Yuanyao Agriculture and Animal Husbandry Technology Co., Ltd, Shanghai, 200000, China
| | - Manman Cheng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Sciences & Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shimei Luo
- Department of Animal Nutrition and Feed Science, College of Animal Sciences & Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jingjing Wei
- Department of Animal Nutrition and Feed Science, College of Animal Sciences & Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Tieping Song
- Yichang Tianyou Huamu Technology Co.,Ltd, Yichang, 443000, China
| | - Yanzhang Gong
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Sciences & Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhongxin Zhou
- Department of Animal Nutrition and Feed Science, College of Animal Sciences & Technology, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
11
|
Ramirez-Hincapie S, Birk B, Ternes P, Giri V, Zickgraf FM, Haake V, Herold M, Kamp H, Driemert P, Landsiedel R, Richling E, Funk-Weyer D, van Ravenzwaay B. Application of high throughput in vitro metabolomics for hepatotoxicity mode of action characterization and mechanistic-anchored point of departure derivation: a case study with nitrofurantoin. Arch Toxicol 2023; 97:2903-2917. [PMID: 37665362 PMCID: PMC10504224 DOI: 10.1007/s00204-023-03572-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
Omics techniques have been increasingly recognized as promising tools for Next Generation Risk Assessment. Targeted metabolomics offer the advantage of providing readily interpretable mechanistic information about perturbed biological pathways. In this study, a high-throughput LC-MS/MS-based broad targeted metabolomics system was applied to study nitrofurantoin metabolic dynamics over time and concentration and to provide a mechanistic-anchored approach for point of departure (PoD) derivation. Upon nitrofurantoin exposure at five concentrations (7.5 µM, 15 µM, 20 µM, 30 µM and 120 µM) and four time points (3, 6, 24 and 48 h), the intracellular metabolome of HepG2 cells was evaluated. In total, 256 uniquely identified metabolites were measured, annotated, and allocated in 13 different metabolite classes. Principal component analysis (PCA) and univariate statistical analysis showed clear metabolome-based time and concentration effects. Mechanistic information evidenced the differential activation of cellular pathways indicative of early adaptive and hepatotoxic response. At low concentrations, effects were seen mainly in the energy and lipid metabolism, in the mid concentration range, the activation of the antioxidant cellular response was evidenced by increased levels of glutathione (GSH) and metabolites from the de novo GSH synthesis pathway. At the highest concentrations, the depletion of GSH, together with alternations reflective of mitochondrial impairments, were indicative of a hepatotoxic response. Finally, a metabolomics-based PoD was derived by multivariate PCA using the whole set of measured metabolites. This approach allows using the entire dataset and derive PoD that can be mechanistically anchored to established key events. Our results show the suitability of high throughput targeted metabolomics to investigate mechanisms of hepatoxicity and derive point of departures that can be linked to existing adverse outcome pathways and contribute to the development of new ones.
Collapse
Affiliation(s)
| | - Barbara Birk
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Varun Giri
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | | | | | | | | | - Robert Landsiedel
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
- Pharmacy, Pharmacology and Toxicology, Free University of Berlin, Berlin, Germany
| | - Elke Richling
- Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, Kaiserslautern, Germany
| | | | | |
Collapse
|
12
|
Wang M, Jiang Y, Wang S, Fu L, Liang Z, Zhang Y, Huang X, Li X, Feng M, Long D. Yak milk protects against alcohol-induced liver injury in rats. Food Funct 2023; 14:9857-9871. [PMID: 37853817 DOI: 10.1039/d3fo03675h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
The protective effects of yak milk (YM) against chronic alcoholic liver injury in rats were investigated in this study. Histologic and biochemical analyses demonstrated that YM consumption ameliorates alcohol-induced liver injury by increasing the liver antioxidant enzyme activity and reducing inflammation. Furthermore, microbiome and metabolomic analyses exploring YM's impact on gut microbiota and metabolism found that YM administration regulates gut microbiota composition. Specifically, there was a decrease in the relative abundance of Helicobacter, Streptococcus, Peptococcus and Tyzzerella, along with an increase in Turisibacter and Intestinimonas. Moreover, Pearson analysis indicated positive correlations between Peptococcus and Tyzzerella with ALT and AST levels, while showing a negative correlation with ADH levels. Furthermore, differential metabolite analysis of fecal samples from the YM group identified significant increases in the taurine (2-Aminoethanesulfonic acid), hypotaurine (2-Aminoethanesulfonic Acid) and isethionic acid levels. Finally, KEGG topology analysis highlighted taurine and hypotaurine metabolism as the primary pathways influenced by YM intervention. Therefore, these findings collectively suggest that YM may protect alcohol-exposed rats against liver injury by modulating oxidative stress, inflammatory response, gut microbiota disorder, and metabolic regulation.
Collapse
Affiliation(s)
- Man Wang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Yanshi Jiang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Siying Wang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Lin Fu
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Zujin Liang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Ying Zhang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Xiaodan Huang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Xin Li
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Meiying Feng
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Danfeng Long
- School of Public Health, Lanzhou University, Lanzhou, China.
| |
Collapse
|
13
|
Espe M, Adam AC, Saito T, Skjærven KH. Methionine: An Indispensable Amino Acid in Cellular Metabolism and Health of Atlantic Salmon. AQUACULTURE NUTRITION 2023; 2023:5706177. [PMID: 37927379 PMCID: PMC10624553 DOI: 10.1155/2023/5706177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023]
Abstract
Methionine is an indispensable amino acid with an important role as the main methyl donor in cellular metabolism for both fish and mammals. Metabolization of methionine to the methyl donor S-adenosylmethionine (SAM) has consequence for polyamine, carnitine, phospholipid, and creatine synthesis as well as epigenetic modifications such as DNA- and histone tail methylation. Methionine can also be converted to cysteine and contributes as a precursor for taurine and glutathione synthesis. Moreover, methionine is the start codon for every protein being synthetized and thereby serves an important role in initiating translation. Modern salmon feed is dominated by plant ingredients containing less taurine, carnitine, and creatine than animal-based ingredients. This shift results in competition for SAM due to an increasing need to endogenously synthesize associated metabolites. The availability of methionine has profound implications for various metabolic pathways including allosteric regulation. This necessitates a higher nutritional need to meet the requirement as a methyl donor, surpassing the quantities for protein synthesis and growth. This comprehensive review provides an overview of the key metabolic pathways in which methionine plays a central role as methyl donor and unfolds the implications for methylation capacity, metabolism, and overall health particularly emphasizing the development of fatty liver, oxidation, and inflammation when methionine abundance is insufficient focusing on nutrition for Atlantic salmon (Salmo salar).
Collapse
Affiliation(s)
- M. Espe
- Institute of Marine Research, P.O. Box 5817 Nordnes, Bergen, Norway
| | - A. C. Adam
- Institute of Marine Research, P.O. Box 5817 Nordnes, Bergen, Norway
| | - T. Saito
- Institute of Marine Research, P.O. Box 5817 Nordnes, Bergen, Norway
| | - K. H. Skjærven
- Institute of Marine Research, P.O. Box 5817 Nordnes, Bergen, Norway
| |
Collapse
|
14
|
San J, Hu J, Pang H, Zuo W, Su N, Guo Z, Wu G, Yang J. Taurine Protects against the Fatty Liver Hemorrhagic Syndrome in Laying Hens through the Regulation of Mitochondrial Homeostasis. Int J Mol Sci 2023; 24:10360. [PMID: 37373507 DOI: 10.3390/ijms241210360] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a chronic liver disease caused by fat deposition in the liver of humans and mammals, while fatty liver hemorrhagic syndrome (FLHS) is a fatty liver disease in laying hens which can increase the mortality and cause severe economic losses to the laying industry. Increasing evidence has shown a close relationship between the occurrence of fatty liver disease and the disruption of mitochondrial homeostasis. Studies have proven that taurine can regulate hepatic fat metabolism, reduce hepatic fatty deposition, inhibit oxidative stress, and alleviate mitochondrial dysfunction. However, the mechanisms by which taurine regulates mitochondrial homeostasis in hepatocytes need to be further studied. In this study, we determined the effects and mechanisms of taurine on high-energy low-protein diet-induced FLHS in laying hens and in cultured hepatocytes in free fatty acid (FFA)-induced steatosis. The liver function, lipid metabolism, antioxidant capacity, mitochondrial function, mitochondrial dynamics, autophagy, and biosynthesis were detected. The results showed impaired liver structure and function, mitochondrial damage and dysfunction, lipid accumulation, and imbalance between mitochondrial fusion and fission, mitochondrial autophagy, and biosynthesis in both FLHS hens and steatosis hepatocytes. Taurine administration can significantly inhibit the occurrence of FLHS, protect mitochondria in hepatocytes from disease induced by lipid accumulation and FFA, up-regulate the expression levels of Mfn1, Mfn2, Opa1, LC3I, LC3II, PINK1, PGC-1α, Nrf1, Nrf2, and Tfam, and down-regulate the expression levels of Fis1, Drp1, and p62. In conclusion, taurine can protect laying hens from FLHS through the regulation of mitochondrial homeostasis, including the regulation of mitochondrial dynamics, autophagy, and biosynthesis.
Collapse
Affiliation(s)
- Jishuang San
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Jianmin Hu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Huiping Pang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Wenjun Zuo
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Na Su
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Zimeng Guo
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Gaofeng Wu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Jiancheng Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
15
|
Song Q, Guo JX, Ma YX, Ou T, Zhang J, Li HZ, Mi SQ, Zhang YZ, Oda H, Chen W. Taurine alleviated hepatic steatosis in oleic acid-treated-HepG2 cells and rats fed a high-fat diet. Heliyon 2023; 9:e16401. [PMID: 37274675 PMCID: PMC10238701 DOI: 10.1016/j.heliyon.2023.e16401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/21/2023] [Accepted: 05/16/2023] [Indexed: 06/06/2023] Open
Abstract
Taurine has been proven in many trials to alleviate the symptoms of metabolic associated fatty liver disease. Here its protective effect for hepatic steatosis and modulation of AMP-activated protein kinase and insulin signaling pathway were investigated. Steatotic HepG2 cell established with oleic acid (0.05 mmol/L), treated with taurine (5 mmol/L), dorsomorphin (10 μmol/L) for 24 h. Sprague Dawley rats were divided into regular and high-fat diet (HFD) groups, and their corresponding taurine (70 or 350 mg/kg BW/d) groups, fed for 8 weeks. In steatotic cell, taurine reduced the TG concentration and SREBP-1c, PPARγ, FAS, ACC, SCD1 protein levels, decreased phosphorylation of mTOR, IRS1 (Ser302), increased phosphorylation of AMPKα, LKB1, PI3K, Akt, ACC. While dorsomorphin eliminated taurine's TG-lowering effect. In HFD-fed rats, taurine reduced liver TG, serum TG, ALT, AST, IL-1β, IL-4, TNF-α. The effects of taurine on the main factors of fatty acid synthesis were mostly consistent with cell experiments, and the reduction of microRNAs (451, 33, 291b) was aligned with the improvement in LKB1 and AMPK expression in HFD rats. Taurine alleviated steatosis-induced inhibition of IRS1-PI3K-Akt pathway, but suppressed its positively regulated downstream factor mTOR. In parallel, taurine reduced steatosis by activating LKB1-AMPKα pathway via phosphorylation and no-phosphorylation manner, then inhibiting SREBP-1c directly or by suppressing mTOR phosphorylation.
Collapse
Affiliation(s)
- Qi Song
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya, 464-8601, Japan
| | - Jun xia Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| | - Yu xun Ma
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| | - Tong Ou
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| | - Jing Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| | - Hui zi Li
- Department of Nutrition, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Sheng quan Mi
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| | - Yan zhen Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| | - Hiroaki Oda
- Laboratory of Nutritional Biochemistry, Nagoya University, Nagoya, 464-8601, Japan
| | - Wen Chen
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| |
Collapse
|
16
|
Zhang D, Fan J, Liu H, Qiu G, Cui S. Testosterone enhances taurine synthesis by upregulating androgen receptor and cysteine sulfinic acid decarboxylase expressions in male mouse liver. Am J Physiol Gastrointest Liver Physiol 2023; 324:G295-G304. [PMID: 36749568 DOI: 10.1152/ajpgi.00076.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Taurine is an end-product of cysteine metabolism, whereas cysteine dioxygenase (CDO) and cysteine sulfinate decarboxylase (CSAD) are key enzymes regulating taurine synthesis. Sex steroids, including estrogens and androgens, are associated with liver physiopathological processes; however, we still do not know whether taurine and sex steroids interact in regulating liver physiology and hepatic diseases, and whether there are sex differences, although our recent study shows that the estrogen is involved in regulating taurine synthesis in mouse liver. The present study was thus proposed to identify whether 17-β-estradiol and testosterone (T) play their roles by regulating CDO and CSAD expression and taurine synthesis in male mouse liver. Our results demonstrated that testosterone did not have a significant influence on CDO expression but significantly enhanced CSAD, androgen receptor (AR) expressions, and taurine levels in mouse liver, cultured hepatocytes, and HepG2 cells, whereas these effects were abrogated by AR antagonist flutamide. Furthermore, our results showed that testosterone increased CSAD-promoter-luciferase activity through the direct interaction of the AR DNA binding domain with the CSAD promoter. These findings first demonstrate that testosterone acts as an important factor to regulate sulfur amino acid metabolism and taurine synthesis through AR/CSAD signaling pathway. In addition, the in vivo and in vitro experiments showed that 17-β-estradiol has no significant effects on liver CSAD expression and taurine synthesis in male mice and suggest that the effects of sex steroids on the taurine synthesis in mouse liver have sex differences. These results are crucial for understanding the physiological functions of taurine/androgen and their interacting mechanisms in the liver.NEW & NOTEWORTHY This study demonstrates that testosterone functions to enhance taurine synthesis by interacting with androgen receptor and binding to cysteine sulfinate decarboxylase (CSAD) promoter zone. Whereas estrogen has no significant effects either on liver CSAD expression or taurine synthesis in male mice and suggests that the effects of sex steroids on taurine synthesis in the liver have gender differences. These new findings are the potential for establishing effective protective and therapeutic strategies for liver diseases.
Collapse
Affiliation(s)
- Di Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| | - Jingjing Fan
- College of Biological and Agricultural Engineering, Weifang University, Weifang, People's Republic of China
| | - Hui Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| | - Guobin Qiu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
17
|
Ji X, Tang Z, Zhang F, Zhou F, Wu Y, Wu D. Dietary taurine supplementation counteracts deoxynivalenol-induced liver injury via alleviating oxidative stress, mitochondrial dysfunction, apoptosis, and inflammation in piglets. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114705. [PMID: 36863159 DOI: 10.1016/j.ecoenv.2023.114705] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
Deoxynivalenol (DON), as a widespread Fusarium mycotoxin in cereals, food products, and animal feed, is detrimental to both human and animal health. The liver is not only the primary organ responsible for DON metabolism but also the principal organ affected by DON toxicity. Taurine is well known to display various physiological and pharmacological functions due to its antioxidant and anti-inflammatory properties. However, the information regarding taurine supplementation counteracting DON-induced liver injury in piglets is still unclear. In our work, twenty-four weaned piglets were subjected to four groups for a 24-day period, including the BD group (a basal diet), the DON group (3 mg/kg DON-contaminated diet), the DON+LT group (3 mg/kg DON-contaminated diet + 0.3% taurine), and the DON+HT group (3 mg/kg DON-contaminated diet + 0.6% taurine). Our findings indicated that taurine supplementation improved growth performance and alleviated DON-induced liver injury, as evidenced by the reduced pathological and serum biochemical changes (ALT, AST, ALP, and LDH), especially in the group with the 0.3% taurine. Taurine could counteract hepatic oxidative stress in piglets exposed to DON, as it reduced ROS, 8-OHdG, and MDA concentrations and improved the activity of antioxidant enzymes. Concurrently, taurine was observed to upregulate the expression of key factors involved in mitochondrial function and the Nrf2 signaling pathway. Furthermore, taurine treatment effectively attenuated DON-induced hepatocyte apoptosis, as verified through the decreased proportion of TUNEL-positive cells and regulation of the mitochondria-mediated apoptosis pathway. Finally, the administration of taurine was able to reduce liver inflammation due to DON, by inactivating the NF-κB signaling pathway and declining the production of pro-inflammatory cytokines. In summary, our results implied that taurine effectively improved DON-induced liver injury. The underlying mechanism should be that taurine restored mitochondrial normal function and antagonized oxidative stress, thereby reducing apoptosis and inflammatory responses in the liver of weaned piglets.
Collapse
Affiliation(s)
- Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230001, China; Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou 233100, China
| | - Zhongqi Tang
- College of Animal Science, Anhui Science and Technology University, Chuzhou 233100, China
| | - Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou 233100, China; Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou 233100, China; Fengyang Xiaogang Minyi Land Shares Cooperatives, Chuzhou 233100, China
| | - Fen Zhou
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230001, China
| | - Yijing Wu
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230001, China
| | - Dong Wu
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230001, China.
| |
Collapse
|
18
|
Zhong W, Li Y, Zhong H, Cheng Y, Chen Q, Zhao X, Liu Z, Li R, Zhang R. Exploring the mechanism of anti-chronic heart failure effect of qiweiqiangxin І granules based on metabolomics. Front Pharmacol 2023; 14:1111007. [PMID: 36860302 PMCID: PMC9968974 DOI: 10.3389/fphar.2023.1111007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Background: Qiweiqiangxin І granules (QWQX І) is a traditional Chinese medicine preparation based on the basic theory of traditional Chinese medicine, which produces a good curative effect in treating chronic heart failure (CHF). However, its pharmacological effect and potential mechanism for CHF remain unknown. Aim of the study: The purpose of this study is to clarify the efficacy of QWQX І and its possible mechanisms. Materials and methods: A total of 66 patients with CHF were recruited and randomly assigned to the control or QWQX І groups. The primary endpoint was the effect of left ventricular ejection fraction (LVEF) after 4 weeks of treatment. The LAD artery of rats was occluded to establish the model of CHF. Echocardiography, HE and Masson staining were performed to evaluate the pharmacological effect of QWQX І against CHF. Ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF/MS) untargeted metabolomics was to screen endogenous metabolites in rat plasma and heart and elucidate the mechanism of QWQX І against CHF. Results: In the clinical study, a total of 63 heart failure patients completed the 4-week follow-up, including 32 in the control group and 31 in QWQX І group. After 4 weeks of treatment, LVEF was significantly improved in QWQX І group compared with the control group. In addition, the patients in QWQX І group had better quality of life than the control group. In animal studies, QWQX І significantly improved cardiac function, decreased B-type natriuretic peptide (BNP) levels, reduced inflammatory cell infiltration, and inhibited collagen fibril rate. Untargeted metabolomic analysis revealed that 23 and 34 differential metabolites were screened in the plasma and heart of chronic heart failure rats, respectively. 17 and 32 differential metabolites appeared in plasma and heart tissue after QWQX І treatment, which were enriched to taurine and hypotaurine metabolism, glycerophospholipid metabolism and linolenic acid metabolism by KEGG analysis. LysoPC (16:1 (9Z)) is a common differential metabolite in plasma and heart, which is produced by lipoprotein-associated phospholipase A2 (Lp-PLA2), hydrolyzes oxidized linoleic acid to produce pro-inflammatory substances. QWQX І regulates the level of LysoPC (16:1 (9Z)) and Lp-PLA2 to normal. Conclusion: QWQX І combined with western medicine can improve the cardiac function of patients with CHF. QWQX І can effectively improve the cardiac function of LAD-induced CHF rats through regulating glycerophospholipid metabolism and linolenic acid metabolism-mediated inflammatory response. Thus, QWQX I might provide a potential strategy for CHF therapy.
Collapse
Affiliation(s)
- Wanru Zhong
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, School of Pharmaceutical Sciences, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yihua Li
- The first clinical medical college, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haixiang Zhong
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, School of Pharmaceutical Sciences, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanyuan Cheng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, School of Pharmaceutical Sciences, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Chen
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, School of Pharmaceutical Sciences, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China,Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xinjun Zhao
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhongqiu Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, School of Pharmaceutical Sciences, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Zhongqiu Liu, ; Rong Li, ; Rong Zhang,
| | - Rong Li
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China,*Correspondence: Zhongqiu Liu, ; Rong Li, ; Rong Zhang,
| | - Rong Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, School of Pharmaceutical Sciences, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Zhongqiu Liu, ; Rong Li, ; Rong Zhang,
| |
Collapse
|
19
|
Zhang D, Wang Z, Luo X, Guo H, Qiu G, Gong Y, Gao H, Cui S. Cysteine dioxygenase and taurine are essential for embryo implantation by involving in E 2-ERα and P 4-PR signaling in mouse. J Anim Sci Biotechnol 2023; 14:6. [PMID: 36604722 PMCID: PMC9814424 DOI: 10.1186/s40104-022-00804-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/20/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Taurine performs multiple physiological functions, and the maintenance of taurine level for most mammals relies on active uptake from diet and endogenous taurine synthesis through its synthesis enzymes, including cysteine dioxygenase (CDO). In addition, uterus tissue and uterus fluid are rich in taurine, and taurine synthesis is regulated by estrogen (E2) and progesterone (P4), the key hormones priming embryo-uterine crosstalk during embryo implantation, but the functional interactions and mechanisms among which are largely unknown. The present study was thus proposed to identify the effects of CDO and taurine on embryo implantation and related mechanisms by using Cdo knockout (KO) and ovariectomy (OVX) mouse models. RESULTS The uterine CDO expression was assayed from the first day of plugging (d 1) to d 8 and the results showed that CDO expression level increased from d 1 to d 4, followed by a significant decline on d 5 and persisted to d 8, which was highly correlated with serum and uterine taurine levels, and serum P4 concentration. Next, Cdo KO mouse was established by CRISPER/Cas9. It was showed that Cdo deletion sharply decreased the taurine levels both in serum and uterus tissue, causing implantation defects and severe subfertility. However, the implantation defects in Cdo KO mice were partly rescued by the taurine supplementation. In addition, Cdo deletion led to a sharp decrease in the expressions of P4 receptor (PR) and its responsive genes Ihh, Hoxa10 and Hand2. Although the expression of uterine estrogen receptor (ERα) had no significant change, the levels of ERα induced genes (Muc1, Ltf) during the implantation window were upregulated after Cdo deletion. These accompanied by the suppression of stroma cell proliferation. Meanwhile, E2 inhibited CDO expression through ERα and P4 upregulated CDO expression through PR. CONCLUSION The present study firstly demonstrates that taurine and CDO play prominent roles in uterine receptivity and embryo implantation by involving in E2-ERα and P4-PR signaling. These are crucial for our understanding the mechanism of embryo implantation, and infer that taurine is a potential agent for improving reproductive efficiency of livestock industry and reproductive medicine.
Collapse
Affiliation(s)
- Di Zhang
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009 People’s Republic of China ,grid.268415.cJiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Zhijuan Wang
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, 100193 Beijing, People’s Republic of China
| | - Xuan Luo
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, 100193 Beijing, People’s Republic of China
| | - Hongzhou Guo
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009 People’s Republic of China ,grid.268415.cJiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Guobin Qiu
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009 People’s Republic of China ,grid.268415.cJiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Yuneng Gong
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009 People’s Republic of China ,grid.268415.cJiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Hongxu Gao
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009 People’s Republic of China ,grid.268415.cJiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Sheng Cui
- grid.268415.cCollege of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009 People’s Republic of China ,grid.268415.cJiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009 People’s Republic of China ,grid.268415.cInstitute of Reproduction and Metabolism, Yangzhou University, 225009 Jiangsu, People’s Republic of China
| |
Collapse
|
20
|
Mokhtari E, Ahmadirad H, Teymoori F, Mohammadebrahim A, Bahrololomi SS, Mirmiran P. The association between dietary amino acids and the risk of nonalcoholic fatty liver disease among Tehranian adults: a case-control study. BMC Nutr 2022; 8:155. [PMID: 36575550 PMCID: PMC9793580 DOI: 10.1186/s40795-022-00656-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Amino acids (AAs) are important bioactive components in the diet that can be involved in various underlying biological processes that contribute to the development of nonalcoholic fatty liver disease (NAFLD). The present study investigates the association between dietary intake of amino acids and NAFLD in Iranian adults. METHODS This study was conducted among 225 newly diagnosed cases of NAFLD and 450 controls. A valid and reliable 168-item semiquantitative food frequency questionnaire (FFQ) was used to collect participants' dietary intakes. Multivariable logistic regression models were used to assess the association between tertiles of branched-chain amino acids (BCAAs), aromatic amino acids (AAAs), and sulfuric amino acids (SAAs) intake with the odds of NAFLD among the study participants. RESULTS The mean ± standard deviation of age and BMI of participants (53% male) were 38.1 ± 8.8 years and 26.8 ± 4.3 kg/m2, respectively. In the final models, the OR and 95% CI of NAFLD among participants in the highest tertiles of BCAAs, AAAs, and SAAs intake compared with those in the lowest tertiles were (OR = 2.82; 95% CI: 1.50-5.30), (OR = 2.82; 95% CI: 1.50-5.30), (OR = 2.86; 95% CI: 1.49-5.48), respectively. CONCLUSION Our study indicated a direct association between the intake of AAs groups, including BCAAs, AAAs, SAAs, and the odds of NAFLD. We suggest that other researchers examine the association between AAs groups and NAFLD in large cohort studies.
Collapse
Affiliation(s)
- Ebrahim Mokhtari
- grid.411600.2Student Research Committee, Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran ,grid.411600.2Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985717413, Tehran, Iran
| | - Hamid Ahmadirad
- grid.411600.2Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985717413, Tehran, Iran
| | - Farshad Teymoori
- grid.411600.2Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985717413, Tehran, Iran ,grid.411746.10000 0004 4911 7066Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Mohammadebrahim
- grid.411746.10000 0004 4911 7066Department of Nutrition, health and treatment center of shahriyar, Iran University of Medical Sciences, Tehran, Iran
| | - Samaneh Sadat Bahrololomi
- grid.411746.10000 0004 4911 7066Department of Nutrition, health and treatment center of shahriyar, Iran University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- grid.411600.2Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985717413, Tehran, Iran
| |
Collapse
|
21
|
CSAD Ameliorates Lipid Accumulation in High-Fat Diet-Fed Mice. Int J Mol Sci 2022; 23:ijms232415931. [PMID: 36555571 PMCID: PMC9783087 DOI: 10.3390/ijms232415931] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic metabolic disease manifested in hepatic steatosis, inflammation, fibrosis, etc., which affects over one-quarter of the population around the world. Since no effective therapeutic drugs are available to cope with this widespread epidemic, the functional research of genes with altered expression during NAFLD helps understand the pathogenesis of this disease and the development of new potential therapeutic targets for drugs. In the current work, we discovered via the analysis of the Gene Expression Omnibus (GEO) dataset that cysteine sulfinic acid decarboxylase (CSAD) decreased significantly in NAFLD patients, which was also confirmed in multiple NAFLD mouse models (HFD-fed C57BL/6J, db/db and HFHFrHC-fed C57BL/6J mice). Next, CSAD's function in the progression of NAFLD was explored using AAV-mediated liver-directed gene overexpression in an HFD-fed mouse model, where the overexpression of CSAD in the liver could alleviate NAFLD-associated pathologies, including body weight, liver/body weight ratio, hepatic triglyceride and total cholesterol, and the degree of steatosis. Mechanically, we found that the overexpression of CSAD could increase the expression of some genes related to fatty acid β-oxidation (Acad1, Ppara, and Acox1). Furthermore, we also detected that CSAD could improve mitochondrial injury in vitro and in vivo. Finally, we proposed that the effect of CSAD on lipid accumulation might be independent of the taurine pathway. In conclusion, we demonstrated that CSAD is involved in the development of NAFLD as a protective factor, which suggested that CSAD has the potential to become a new target for drug discovery in NAFLD.
Collapse
|
22
|
Zhang Z, Chen D, Yu J, Su X, Li L. Metabolic perturbations in human hepatocytes induced by bis (2-ethylhexyl)-2,3,4,5-tetrabromophthalate exposure: Insights from high-coverage quantitative metabolomics. Anal Biochem 2022; 657:114887. [PMID: 36150471 DOI: 10.1016/j.ab.2022.114887] [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: 05/06/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022]
Abstract
Bis (2-ethylhexyl)-2,3,4,5-tetrabromophthalate (TBPH) is an extensively used novel brominated flame retardant that is present ubiquitously in the environment and in biota. However, there is inadequate data on its potential hepatotoxicity to humans. In this study, high-coverage quantitative metabolomics based on 12C-/13C-dansylation labeling LC-MS was performed for the first time to assess the metabolic perturbations and underlying mechanisms of TBPH on human hepatocytes. HepG2 cells were exposed to TBPH at dosages of 0.1,1,10 μM for 24 or 72 h. Overall, 1887 and 1364 amine/phenol-containing metabolites were relatively quantified in cells and culture supernatant. Our results revealed that exposure to 0.1 μM TBPH showed little adverse effects, whereas exposure to 10 μM TBPH for 24 h enhanced intracellular protein catabolism and disrupted energy and lipid homeostasis-related pathways such as histidine metabolism, pantothenate and CoA biosynthesis, alanine, aspartate and glutamate metabolism. Nevertheless, most of these perturbations returned to the same levels as controls after 72 h of exposure. Additionally, prolonged TBPH exposure increased oxidative stress, as reflected by marked disturbances in taurine metabolism. This study sensitively revealed the dysregulations of intracellular and extracellular metabolome induced by TBPH, providing a comprehensive understanding of metabolic responses of cells to novel brominated flame retardants.
Collapse
Affiliation(s)
- Zhehua Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Deying Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiong Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiaoling Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
23
|
Guo Q, Zhang L, Yin Y, Gong S, Yang Y, Chen S, Han M, Duan Y. Taurine Attenuates Oxidized Fish Oil-Induced Oxidative Stress and Lipid Metabolism Disorder in Mice. Antioxidants (Basel) 2022; 11:antiox11071391. [PMID: 35883883 PMCID: PMC9311513 DOI: 10.3390/antiox11071391] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
The objective of this study was to determine the effect of dietary taurine on lipid metabolism and liver injury in mice fed a diet high in oxidized fish oil. The ICR mice (six weeks old) were randomly assigned to six groups and fed different diets for 10 weeks: control (CON), normal plus 15% fresh fish oil diet (FFO), normal plus 15% oxidized fish oil diet (OFO), or OFO plus 0.6% (TAU1), 0.9% (TAU2) or 1.2% (TAU3) taurine. Compared to the CON group, OFO mice showed increased liver index, aspartate aminotransferase (AST) and malondialdehyde (MDA) levels in serum (p < 0.05). In addition, OFO mice had increased cholesterol (CHOL)/high-density lipoprotein cholesterol (HDL-C) and decreased HDL-C/low-density lipoprotein cholesterol (LDL-C) and n-6/n-3 polyunsaturated fatty acid (PUFA) ratio in serum (p < 0.05) compared with CON mice. Notably, dietary taurine ameliorated the liver index and AST and MDA levels in serum and liver in a more dose-dependent manner than OFO mice. In addition, compared to OFO mice, decreased levels of CHOL and ratio of CHOL/HDL-C and n-6 PUFA/n-3 PUFA in serum were found in TAU3-fed mice. Supplementation with TAU2 and TAU3 increased the relative mRNA expression levels of peroxisome proliferator-activated receptor α, adipose triglyceride lipase, lipoprotein lipase, hormone-sensitive lipase and carnitine palmitoyl transferase 1 in liver compared with the OFO group (p < 0.05). Moreover, impaired autophagy flux was detected in mice fed with the OFO diet, and this was prevented by taurine. These findings suggested that dietary taurine might provide a potential therapeutic choice against oxidative stress and lipid metabolism disorder.
Collapse
Affiliation(s)
- Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Q.G.); (L.Z.); (Y.Y.); (S.G.); (Y.Y.); (S.C.); (M.H.)
| | - Lingyu Zhang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Q.G.); (L.Z.); (Y.Y.); (S.G.); (Y.Y.); (S.C.); (M.H.)
- National Engineering Laboratory for Rice and By-Product Deep Processing, Central South University of Forestry and Technology, Changsha 410004, China
| | - Yunju Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Q.G.); (L.Z.); (Y.Y.); (S.G.); (Y.Y.); (S.C.); (M.H.)
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Saiming Gong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Q.G.); (L.Z.); (Y.Y.); (S.G.); (Y.Y.); (S.C.); (M.H.)
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yuhuan Yang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Q.G.); (L.Z.); (Y.Y.); (S.G.); (Y.Y.); (S.C.); (M.H.)
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Sisi Chen
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Q.G.); (L.Z.); (Y.Y.); (S.G.); (Y.Y.); (S.C.); (M.H.)
- College of Advanced Agricultural Science, University of Chinese Academy of Sciences, Beijing 100039, China
| | - Mengmeng Han
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Q.G.); (L.Z.); (Y.Y.); (S.G.); (Y.Y.); (S.C.); (M.H.)
- College of Advanced Agricultural Science, University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (Q.G.); (L.Z.); (Y.Y.); (S.G.); (Y.Y.); (S.C.); (M.H.)
- Correspondence: ; Tel.: +86-0731-84619767
| |
Collapse
|
24
|
Feng X, Hu W, Hong Y, Ruan L, Hu Y, Liu D. Taurine Ameliorates Iron Overload-Induced Hepatocyte Injury via the Bcl-2/VDAC1-Mediated Mitochondrial Apoptosis Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4135752. [PMID: 35879990 PMCID: PMC9308541 DOI: 10.1155/2022/4135752] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/20/2022] [Accepted: 05/31/2022] [Indexed: 12/29/2022]
Abstract
Iron overload can induce reactive oxygen species (ROS) burst and liver damage. Taurine can reduce ROS production and ameliorate liver injury caused by iron overload; however, the underlying molecular mechanism remains elusive. Herein, L02 cells treated with 120 μM iron dextran for 48 h showed marked oxidative stress damage and significantly increased apoptosis. Taurine protected hepatocytes by stabilizing mitochondrial membranes and resisting oxidative stress damage caused by iron overload. However, transfection with siRNA Bcl-2 virus abrogated the observed protective effects. Following treatment with taurine, B cell lymphoma-2 (Bcl-2) could inhibit the opening of the mitochondrial permeability transition pore (mPTP), subsequently stabilizing the mitochondrial membrane potential by interacting with voltage-dependent anion channel 1 (VDAC1) of mPTP. The present study is the first to clarify the mechanism underlying taurine-afforded hepatocyte protection against iron overload-induced oxidative stress via Bcl-2-mediated inhibition of mPTP opening and the antiapoptotic pathway.
Collapse
Affiliation(s)
- Xiaoyu Feng
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Wenfeng Hu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
- Department of Pharmacy, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang 332900, China
| | - Yujiao Hong
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Linlin Ruan
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Yueben Hu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Dan Liu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| |
Collapse
|
25
|
Taurine Ameliorates Streptozotocin-Induced Diabetes by Modulating Hepatic Glucose Metabolism and Oxidative Stress in Mice. Metabolites 2022; 12:metabo12060524. [PMID: 35736457 PMCID: PMC9228042 DOI: 10.3390/metabo12060524] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 12/30/2022] Open
Abstract
Taurine is a sulfated amino acid derivative that plays an important role in maintaining the cell function of the living body. Although taurine has been shown to ameliorate diabetes, its mechanism of action has not yet been fully elucidated. The present study investigated the effects of taurine on diabetes focusing on glucose metabolism and oxidative stress. Type 1 diabetes was induced by the administration of streptozotocin (STZ) to male C57BL/6J mice. Taurine was dissolved in drinking water at 3% (w/v) and allowed to be freely ingested by diabetic mice. The weight and blood glucose levels were measured weekly. After nine weeks, mice were sacrificed and their serum, liver, and kidney were removed and used for biochemical and histological analyses. A microarray analysis was also performed in normal mice. Taurine alleviated STZ-induced hyperglycemia and hyperketonemia, accompanied by the suppression of the decrease in hepatic glycogen and upregulation of the mRNA expression of hepatic glucose transporter GLUT-2. Furthermore, STZ-induced elevation of oxidative stress in the liver and kidney was suppressed by taurine treatment. These results showed that taurine ameliorated diabetes and diabetic complications by improving hepatic glucose metabolism and reducing oxidative stress.
Collapse
|
26
|
Zheng H, Ji H, Fan K, Xu H, Huang Y, Zheng Y, Xu Q, Li C, Zhao L, Li Y, Gao H. Targeting Gut Microbiota and Host Metabolism with Dendrobium officinale Dietary Fiber to Prevent Obesity and Improve Glucose Homeostasis in Diet-Induced Obese Mice. Mol Nutr Food Res 2022; 66:e2100772. [PMID: 35225418 DOI: 10.1002/mnfr.202100772] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/29/2021] [Indexed: 12/26/2022]
Abstract
SCOPE Obesity is becoming a major public health problem due to excess dietary fat intake. Dendrobium officinale (D. officinale) is a medicine food homology plant and exerts multiple health-promoting effects. However, its antiobesity effects and the potential mechanisms remain unclear. METHODS AND RESULTS High-fat diet (HFD)-fed mice are administered D. officinale dietary fiber (DODF) daily by gavage for 11 weeks. The results show that treatment with DODF alleviates obesity, liver steatosis, inflammation, and oxidant stress in HFD-induced obese mice. Improved glucose homeostasis in obese mice after DODF treatment is achieved by enhancing insulin pathway and hepatic glycogen synthesis. DODF restructures the gut microbiota in obese mice by decreasing the relative abundance of Bilophila and increasing the relative abundances of Akkermansia, Bifidobacterium, and Muribaculum. Also, DODF reshapes the metabolic phenotype of obese mice as indicated by up-regulating energy metabolism, increasing acetate and taurine, and reducing serum low density/very low density lipoproteins (LDL/VLDL). These beneficial effects are partly transferred by FMT, implying the gut microbiota as a target for the protective effect of DODF on obesity-related symptoms. CONCLUSION The results suggest that DODF can be used as a novel prebiotics to maintain the gut microbial homeostasis and improve metabolic health, preventing obesity and related metabolic syndrome.
Collapse
Affiliation(s)
- Hong Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.,Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325400, China
| | - Hui Ji
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Kai Fan
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hangying Xu
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yinli Huang
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325400, China
| | - Yafei Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qingqing Xu
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chen Li
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Liangcai Zhao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuping Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Hongchang Gao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
27
|
Higuchi K, Sugiyama K, Tomabechi R, Kishimoto H, Inoue K. Mammalian monocarboxylate transporter 7 (MCT7/Slc16a6) is a novel facilitative taurine transporter. J Biol Chem 2022; 298:101800. [PMID: 35257743 PMCID: PMC8980330 DOI: 10.1016/j.jbc.2022.101800] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 12/02/2022] Open
Abstract
Monocarboxylate transporter 7 (MCT7) is an orphan transporter expressed in the liver, brain, and in several types of cancer cells. It has also been reported to be a survival factor in melanoma and breast cancers. However, this survival mechanism is not yet fully understood due to MCT7’s unidentified substrate(s). Therefore, here we sought to identify MCT7 substrate(s) and characterize the transport mechanisms by analyzing amino acid transport in HEK293T cells and polarized Caco-2 cells. Analysis of amino acids revealed significant rapid reduction in taurine from cells transfected with enhanced green fluorescent protein-tagged MCT7. We found that taurine uptake and efflux by MCT7 was pH-independent and that the uptake was not saturated in the presence of taurine excess of 200 mM. Furthermore, we found that monocarboxylates and acidic amino acids inhibited MCT7-mediated taurine uptake. These results imply that MCT7 may be a low-affinity facilitative taurine transporter. We also found that MCT7 was localized at the basolateral membrane in polarized Caco-2 cells and that the induction of MCT7 expression in polarized Caco-2 cells enhanced taurine permeation. Finally, we demonstrated that interactions of MCT7 with ancillary proteins basigin/CD147 and embigin/GP70 enhanced MCT7-mediated taurine transport. In summary, these findings reveal that taurine is a novel substrate of MCT7 and that MCT7-mediated taurine transport might contribute to the efflux of taurine from cells.
Collapse
Affiliation(s)
- Kei Higuchi
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Koki Sugiyama
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Ryuto Tomabechi
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Hisanao Kishimoto
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan.
| |
Collapse
|
28
|
Satsu H, Fukumura M, Watari K. Regulation of CXCR4 Expression by Taurine in Macrophage-Like Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:41-49. [DOI: 10.1007/978-3-030-93337-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Li Y, Zhao D, Qian M, Liu J, Pan C, Zhang X, Duan X, Zhang Y, Jia W, Wang L. Amlodipine, an anti-hypertensive drug, alleviates non-alcoholic fatty liver disease by modulating gut microbiota. Br J Pharmacol 2021; 179:2054-2077. [PMID: 34862599 DOI: 10.1111/bph.15768] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/08/2021] [Accepted: 11/21/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Non-alcoholic fatty liver disease (NAFLD) represents a severe public health problem. It often coexists with hypertension in the context of metabolic syndrome. Here, we investigated the effects of amlodipine on non-alcoholic fatty liver disease combined with hypertension and the underlying mechanism. EXPERIMENTAL APPROACH mice were fed with high-fat diet and 0.05% N-Nitro-L-arginine methylester sterile water to induce NAFLD with hypertension. Gut microbiota composition and function were assessed by 16S ribosomal DNA and metagenomic sequencing. Untargeted metabolome profiles were applied to identify differential metabolites in mice cecum. KEY RESULTS Amlodipine besylate (AB) and amlodipine aspartate (AA) significantly decreased liver injury, hepatic steatosis and improved lipid metabolism with a concomitant reduction in the expression of lipogenic genes in mice with NAFLD and hypertension. Mechanistically, AA and AB have potential in restoring intestinal barrier integrity and improving antimicrobial defense along with the elevated abundances of Akkermansia, Bacteroides and Lactobacillus. Noteworthily, the gut microbiota in AB and AA-treated mice had higher abundance of functional genes involved in taurine and hypotaurine metabolism. Consistently, the strengthened taurine and hypotaurine metabolism was confirmed by the untargeted metabolome analysis. Based on the correlation and causal analysis, the altered gut microbiota composition and the enhancement of taurine and hypotaurine metabolism may synergistically decreased ALT, liver triglycerides, lipogenic genes and plasma cholesterol in HFD-fed hypertensive mice. CONCLUSION AND IMPLICATIONS Collectively, AA and AB exert multi-factorial improvements in NAFLD and hypertension by modulating gut microbiota, and may serve as a promising therapeutic agent for treating these diseases.
Collapse
Affiliation(s)
- Yang Li
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Danyang Zhao
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Minyi Qian
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Chuyue Pan
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xinxin Zhang
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xubin Duan
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yufei Zhang
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wenxin Jia
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lirui Wang
- Institute of Modern Biology, Nanjing University, Nanjing, China
| |
Collapse
|
30
|
Yao Z, Liang G, Lv ZL, Lan LC, Zhu FL, Tang Q, Huang L, Chen XQ, Yang MX, Shan QW. Taurine Reduces Liver Damage in Non-Alcoholic Fatty Liver Disease Model in Rats by Down-Regulating IL-9 and Tumor Growth Factor TGF-β. Bull Exp Biol Med 2021; 171:638-643. [PMID: 34617180 DOI: 10.1007/s10517-021-05285-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Indexed: 02/08/2023]
Abstract
The study employed a rat model to examine the effects of taurine (Tau) on prevention and therapy of non-alcoholic fatty liver disease (NAFLD). In model rats maintained on a high-fat diet (HFD), the serum levels of ALT, AST, triglycerides, cholesterol, and LDL were higher than the corresponding levels in normal control and NP groups (p<0.05). In Tau-prevention and Tau-treatment groups, the serum levels of AST and triglycerides were lower than in HFD rats (p<0.05). In HFD rats, diffuse fatty degeneration and infiltration with inflammatory cells was observed in the liver; in the ileal mucosa, the villi were fractured or absent, the epithelium was exfoliated and infiltrated with inflammatory cells. The levels of TGF-β, IL-9, and their mRNA in the liver and ileal mucosa of HFD rats were significantly higher than in normal control and NP groups (p<0.05). In Tau-prevention and Tau-treatment groups, these levels were significantly lower than in HFD rats (p<0.05). Thus, TGF-β and IL-9 can be implicated in NAFLD genesis, while Tau can preventively or therapeutically diminish the damage to the liver and ileal mucosa in rats with this disease by down-regulating the expression of TGF-β and IL-9.
Collapse
Affiliation(s)
- Z Yao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - G Liang
- Department of Pathophysiology, Basic Medicine College of Guangxi Medical University, Nanning, China
| | - Z L Lv
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - L C Lan
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - F L Zhu
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Q Tang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - L Huang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - X Q Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - M X Yang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Q W Shan
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
31
|
Baliou S, Adamaki M, Ioannou P, Pappa A, Panayiotidis MI, Spandidos DA, Christodoulou I, Kyriakopoulos AM, Zoumpourlis V. Protective role of taurine against oxidative stress (Review). Mol Med Rep 2021; 24:605. [PMID: 34184084 PMCID: PMC8240184 DOI: 10.3892/mmr.2021.12242] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Taurine is a fundamental mediator of homeostasis that exerts multiple roles to confer protection against oxidant stress. The development of hypertension, muscle/neuro‑associated disorders, hepatic cirrhosis, cardiac dysfunction and ischemia/reperfusion are examples of some injuries that are linked with oxidative stress. The present review gives a comprehensive description of all the underlying mechanisms of taurine, with the aim to explain its anti‑oxidant actions. Taurine is regarded as a cytoprotective molecule due to its ability to sustain normal electron transport chain, maintain glutathione stores, upregulate anti‑oxidant responses, increase membrane stability, eliminate inflammation and prevent calcium accumulation. In parallel, the synergistic effect of taurine with other potential therapeutic modalities in multiple disorders are highlighted. Apart from the results derived from research findings, the current review bridges the gap between bench and bedside, providing mechanistic insights into the biological activity of taurine that supports its potential therapeutic efficacy in clinic. In the future, further clinical studies are required to support the ameliorative effect of taurine against oxidative stress.
Collapse
Affiliation(s)
- Stella Baliou
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Adamaki
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Petros Ioannou
- Department of Internal Medicine and Infectious Diseases, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Mihalis I. Panayiotidis
- Department of Cancer Genetics, Therapeutics and Ultrastructural Pathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- The Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
| | - Demetrios A. Spandidos
- Department of Internal Medicine and Infectious Diseases, University Hospital of Heraklion, 71110 Heraklion, Greece
| | | | | | | |
Collapse
|
32
|
Stachowicz A, Wiśniewska A, Kuś K, Białas M, Łomnicka M, Totoń-Żurańska J, Kiepura A, Stachyra K, Suski M, Bujak-Giżycka B, Jawień J, Olszanecki R. Diminazene Aceturate Stabilizes Atherosclerotic Plaque and Attenuates Hepatic Steatosis in apoE-Knockout Mice by Influencing Macrophages Polarization and Taurine Biosynthesis. Int J Mol Sci 2021; 22:5861. [PMID: 34070749 PMCID: PMC8199145 DOI: 10.3390/ijms22115861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis and nonalcoholic fatty liver disease are leading causes of morbidity and mortality in the Western countries. The renin-angiotensin system (RAS) with its two main opposing effectors, i.e., angiotensin II (Ang II) and Ang-(1-7), is widely recognized as a major regulator of cardiovascular function and body metabolic processes. Angiotensin-converting enzyme 2 (ACE2) by breaking-down Ang II forms Ang-(1-7) and thus favors Ang-(1-7) actions. Therefore, the aim of our study was to comprehensively evaluate the influence of prolonged treatment with ACE2 activator, diminazene aceturate (DIZE) on the development of atherosclerotic lesions and hepatic steatosis in apoE-/- mice fed a high-fat diet (HFD). We have shown that DIZE stabilized atherosclerotic lesions and attenuated hepatic steatosis in apoE-/- mice fed an HFD. Such effects were associated with decreased total macrophages content and increased α-smooth muscle actin levels in atherosclerotic plaques. Moreover, DIZE changed polarization of macrophages towards increased amount of anti-inflammatory M2 macrophages in the atherosclerotic lesions. Interestingly, the anti-steatotic action of DIZE in the liver was related to the elevated levels of HDL in the plasma, decreased levels of triglycerides, and increased biosynthesis and concentration of taurine in the liver of apoE-/- mice. However, exact molecular mechanisms of both anti-atherosclerotic and anti-steatotic actions of DIZE require further investigations.
Collapse
Affiliation(s)
- Aneta Stachowicz
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Anna Wiśniewska
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Katarzyna Kuś
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Magdalena Białas
- Chair of Pathomorphology, Jagiellonian University Medical College, 31-531 Krakow, Poland;
| | - Magdalena Łomnicka
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Justyna Totoń-Żurańska
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Anna Kiepura
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Kamila Stachyra
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Maciej Suski
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Beata Bujak-Giżycka
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Jacek Jawień
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Rafał Olszanecki
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| |
Collapse
|
33
|
Dong Y, Li X, Liu Y, Gao J, Tao J. The molecular targets of taurine confer anti-hyperlipidemic effects. Life Sci 2021; 278:119579. [PMID: 33961852 DOI: 10.1016/j.lfs.2021.119579] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/16/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
Hyperlipidemia, an independent risk factor for atherosclerosis, is regarded as a lipid metabolism disorder associated with elevated plasma triglyceride and/or cholesterol. Genetic factors and unhealthy lifestyles, such as excess caloric intake and physical inactivity, can result in hyperlipidemia. Taurine, a sulfur-containing non-essential amino acid, is abundant in marine foods and has been associated with wide-ranging beneficial physiological effects, with special reference to regulating aberrant lipid metabolism. Its anti-hyperlipidemic mechanism is complex, which is related to many enzymes in the process of fat anabolism and catabolism (e.g., HMGCR, CYP7A1, LDLR, FXR, FAS and ACC). Anti-inflammatory and antioxidant molecular targets, lipid autophagy, metabolic reprogramming and gut microbiota will also be reviewed.
Collapse
Affiliation(s)
- Yuanyuan Dong
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China
| | - Xiaoling Li
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China
| | - Yaling Liu
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China
| | - Jie Gao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China.
| |
Collapse
|
34
|
Lazebnik LB, Golovanova EV, Turkina SV, Raikhelson KL, Okovityy SV, Drapkina OM, Maev IV, Martynov AI, Roitberg GE, Khlynova OV, Abdulganieva DI, Alekseenko SA, Ardatskaya MD, Bakulin IG, Bakulina NV, Bueverov AO, Vinitskaya EV, Volynets GV, Eremina EY, Grinevich VB, Dolgushina AI, Kazyulin AN, Kashkina EI, Kozlova IV, Konev YV, Korochanskaya NV, Kravchuk YA, Li ED, Loranskaya ID, Makhov VM, Mekhtiev SN, Novikova VP, Ostroumova OD, Pavlov CS, Radchenko VG, Samsonov AA, Sarsenbaeva AS, Sayfutdinov RG, Seliverstov PV, Sitkin SI, Stefanyuk OV, Tarasova LV, Tkachenko EI, Uspensky YP, Fominykh YA, Khavkin AI, Tsyganova YV, Sharhun OO. Non-alcoholic fatty liver disease in adults: clinic, diagnostics, treatment. Guidelines for therapists, third version. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2021; 1:4-52. [DOI: 10.31146/1682-8658-ecg-185-1-4-52] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Affiliation(s)
- L. B. Lazebnik
- Federal State Budgetary Educational Institution of Higher Education “A. I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russion Federation
| | - E. V. Golovanova
- Federal State Budgetary Educational Institution of Higher Education “A. I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russion Federation
| | - S. V. Turkina
- State-funded Educational Establishment of Higher Professional Education «Volgograd State Medical University of the Ministry of Public Health of the Russian Federation»
| | | | - S. V. Okovityy
- Saint Petersburg State Chemical Pharmaceutical University (SPCPA)
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine of the Russian Ministry of Health
| | - I. V. Maev
- Federal State Budgetary Educational Institution of Higher Education “A. I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russion Federation
| | - A. I. Martynov
- Federal State Budgetary Educational Institution of Higher Education “A. I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russion Federation
| | - G. E. Roitberg
- Pirogov Russian National Research Medical University; JSC «Medicine»
| | - O. V. Khlynova
- Perm State Medical University named after academician E. A. Vagner Ministry of Health care of Russia
| | | | | | - M. D. Ardatskaya
- Federal State Budgetary Institution “Central Clinical Hospital”, of the Russian Federation Presidential Administration
| | - I. G. Bakulin
- North- Western state medical University named after I. I. Mechnikov, Ministry of health of the Russian Federation
| | - N. V. Bakulina
- North- Western state medical University named after I. I. Mechnikov, Ministry of health of the Russian Federation
| | - A. O. Bueverov
- Moscow regional research and clinical Institute of M. F. Vladimirsky
| | | | | | | | | | | | - A. N. Kazyulin
- Federal State Budgetary Educational Institution of Higher Education “A. I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russion Federation
| | | | - I. V. Kozlova
- Saratov State Medical University n. a. V. I. Razumovsky
| | - Yu. V. Konev
- Federal State Budgetary Educational Institution of Higher Education “A. I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russion Federation
| | - N. V. Korochanskaya
- Federal State Budgetary Educational Institution of Higher Education Kuban State Medical University Health Ministry of Russian Federation
| | | | - E. D. Li
- Multifunctional medical center of the Bank of Russia
| | - I. D. Loranskaya
- Federal State Budgetary Educational Institution of Further Professional Education “Russian Medical Academy of Continuous Professional Education” of the Ministry of Healthcare of the Russian Federation
| | - V. M. Makhov
- I. M. Sechenov First Moscow Medical State University
| | - S. N. Mekhtiev
- Institute of Professional Retraining of the International Medical Center “SOGAZ”
| | | | - O. D. Ostroumova
- Federal State Budgetary Educational Institution of Further Professional Education “Russian Medical Academy of Continuous Professional Education” of the Ministry of Healthcare of the Russian Federation
| | - Ch. S. Pavlov
- I. M. Sechenov First Moscow Medical State University
| | | | - A. A. Samsonov
- Federal State Budgetary Educational Institution of Higher Education “A. I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russion Federation
| | | | - R. G. Sayfutdinov
- Kazan State Medical Academy — Branch Campus of the Federal State Budgetary Educational Institution of Further Professional Education “Russian Medical Academy of Continuous Professional Education” of the Ministry of Healthcare of the Russian Federation, central scientifi c research laboratory
| | - P. V. Seliverstov
- North-Western state medical University named after I. I. Mechnikov, Ministry of health of the Russian Federation
| | - S. I. Sitkin
- North-Western state medical University named after I. I. Mechnikov, Ministry of health of the Russian Federation
| | - O. V. Stefanyuk
- Federal State Budgetary Educational Institution of Higher Education “A. I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russion Federation; National Medical Research Center for Therapy and Preventive Medicine of the Russian Ministry of Health
| | | | | | | | | | - A. I. Khavkin
- Pirogov Russian National Research Medical University
| | | | - O. O. Sharhun
- Pirogov Russian National Research Medical University
| |
Collapse
|
35
|
The Role of the Transsulfuration Pathway in Non-Alcoholic Fatty Liver Disease. J Clin Med 2021; 10:jcm10051081. [PMID: 33807699 PMCID: PMC7961611 DOI: 10.3390/jcm10051081] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/21/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing and approximately 25% of the global population may have NAFLD. NAFLD is associated with obesity and metabolic syndrome, but its pathophysiology is complex and only partly understood. The transsulfuration pathway (TSP) is a metabolic pathway regulating homocysteine and cysteine metabolism and is vital in controlling sulfur balance in the organism. Precise control of this pathway is critical for maintenance of optimal cellular function. The TSP is closely linked to other pathways such as the folate and methionine cycles, hydrogen sulfide (H2S) and glutathione (GSH) production. Impaired activity of the TSP will cause an increase in homocysteine and a decrease in cysteine levels. Homocysteine will also be increased due to impairment of the folate and methionine cycles. The key enzymes of the TSP, cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE), are highly expressed in the liver and deficient CBS and CSE expression causes hepatic steatosis, inflammation, and fibrosis in animal models. A causative link between the TSP and NAFLD has not been established. However, dysfunctions in the TSP and related pathways, in terms of enzyme expression and the plasma levels of the metabolites (e.g., homocysteine, cystathionine, and cysteine), have been reported in NAFLD and liver cirrhosis in both animal models and humans. Further investigation of the TSP in relation to NAFLD may reveal mechanisms involved in the development and progression of NAFLD.
Collapse
|
36
|
Song Q, Guo J, Zhang Y, Chen W. The beneficial effects of taurine in alleviating fatty liver disease. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
37
|
Shu Y, Liu X, Huang H, Wen Q, Shu J. Research progress of natural compounds in anti-liver fibrosis by affecting autophagy of hepatic stellate cells. Mol Biol Rep 2021; 48:1915-1924. [PMID: 33609264 PMCID: PMC7925445 DOI: 10.1007/s11033-021-06171-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
Chronic liver diseases caused by various pathogenesis are marked by inflammatory infiltration and wound healing reaction, while their normal regeneration ability is impaired. The unbalance between the generation and the degradation of extracellular matrix (ECM) leads to collagen accumulation and develops into liver fibrosis. Inflammation, oxidative stress, and autophagy interact closely in the pathogenesis of hepatic fibrosis. Reactive Oxygen Species (ROS) can not only stimulate Kupffer cells to release massive inflammatory factors, but induce autophagy. However, the latter may suppress inflammatory reaction by inhibiting proinflammatory complex formation directly, and removing damaged organelles or pathogenic microorganism indirectly. At present, effective anti-fibrosis drugs are still lacking. Previous studies have found various natural compounds enabled liver protection through anti-inflammatory, antioxidant, and other mechanisms. In recent years, autophagy, a vital life activity, has been found to be involved in the mechanism of liver fibrosis. As a new target, developing anti-liver fibrosis drugs that regulate the activity of autophagy is very promising. In this review, we summarize the latest studies about natural compounds in the treatment of liver fibrosis by regulating autophagy.
Collapse
Affiliation(s)
- Yongxiang Shu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Xuyou Liu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Haifeng Huang
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Qi Wen
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Jianchang Shu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| |
Collapse
|
38
|
Rives C, Fougerat A, Ellero-Simatos S, Loiseau N, Guillou H, Gamet-Payrastre L, Wahli W. Oxidative Stress in NAFLD: Role of Nutrients and Food Contaminants. Biomolecules 2020; 10:E1702. [PMID: 33371482 PMCID: PMC7767499 DOI: 10.3390/biom10121702] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is often the hepatic expression of metabolic syndrome and its comorbidities that comprise, among others, obesity and insulin-resistance. NAFLD involves a large spectrum of clinical conditions. These range from steatosis, a benign liver disorder characterized by the accumulation of fat in hepatocytes, to non-alcoholic steatohepatitis (NASH), which is characterized by inflammation, hepatocyte damage, and liver fibrosis. NASH can further progress to cirrhosis and hepatocellular carcinoma. The etiology of NAFLD involves both genetic and environmental factors, including an unhealthy lifestyle. Of note, unhealthy eating is clearly associated with NAFLD development and progression to NASH. Both macronutrients (sugars, lipids, proteins) and micronutrients (vitamins, phytoingredients, antioxidants) affect NAFLD pathogenesis. Furthermore, some evidence indicates disruption of metabolic homeostasis by food contaminants, some of which are risk factor candidates in NAFLD. At the molecular level, several models have been proposed for the pathogenesis of NAFLD. Most importantly, oxidative stress and mitochondrial damage have been reported to be causative in NAFLD initiation and progression. The aim of this review is to provide an overview of the contribution of nutrients and food contaminants, especially pesticides, to oxidative stress and how they may influence NAFLD pathogenesis.
Collapse
Affiliation(s)
- Clémence Rives
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Anne Fougerat
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Laurence Gamet-Payrastre
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Walter Wahli
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
39
|
Leonardi BF, Gosmann G, Zimmer AR. Modeling Diet-Induced Metabolic Syndrome in Rodents. Mol Nutr Food Res 2020; 64:e2000249. [PMID: 32978870 DOI: 10.1002/mnfr.202000249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 08/24/2020] [Indexed: 12/17/2022]
Abstract
Standardized animal models represent one of the most valuable tools available to understand the mechanism underlying the metabolic syndrome (MetS) and to seek for new therapeutic strategies. However, there is considerable variability in the studies conducted with this essential purpose. This review presents an updated discussion of the most recent studies using diverse experimental conditions to induce MetS in rodents with unbalanced diets, discusses the key findings in metabolic outcomes, and critically evaluates what we have been learned from them and how to advance in the field. The study includes scientific reports sourced from the Web of Science and PubMed databases, published between January 2013 and June 2020, which used hypercaloric diets to induce metabolic disorders, and address the impact of the diet on metabolic parameters. The collected data are used as support to discuss variables such as sex, species, and age of the animals, the most favorable type of diet, and the ideal diet length to generate metabolic changes. The experimental characteristics propose herein improve the performance of a preclinical model that resembles the human MetS and will guide researchers to investigate new therapeutic alternatives with confidence and higher translational validity.
Collapse
Affiliation(s)
- Bianca F Leonardi
- Phytochemistry and Organic Synthesis Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), 2752 Ipiranga avenue, Porto Alegre, RS, 90610-000, Brazil
| | - Grace Gosmann
- Phytochemistry and Organic Synthesis Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), 2752 Ipiranga avenue, Porto Alegre, RS, 90610-000, Brazil
| | - Aline R Zimmer
- Phytochemistry and Organic Synthesis Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), 2752 Ipiranga avenue, Porto Alegre, RS, 90610-000, Brazil
| |
Collapse
|
40
|
Yang J, Suo H, Song J. Protective role of mitoquinone against impaired mitochondrial homeostasis in metabolic syndrome. Crit Rev Food Sci Nutr 2020; 61:3857-3875. [PMID: 32815398 DOI: 10.1080/10408398.2020.1809344] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mitochondria control various processes in cellular metabolic homeostasis, such as adenosine triphosphate production, generation and clearance of reactive oxygen species, control of intracellular Ca2+ and apoptosis, and are thus a critical therapeutic target for metabolic syndrome (MetS). The mitochondrial targeted antioxidant mitoquinone (MitoQ) reduces mitochondrial oxidative stress, prevents impaired mitochondrial dynamics, and increases mitochondrial turnover by promoting autophagy (mitophagy) and mitochondrial biogenesis, which ultimately contribute to the attenuation of MetS conditions, including obesity, insulin resistance, hypertension and cardiovascular disease. The regulatory effect of MitoQ on mitochondrial homeostasis is mediated through AMPK and its downstream signaling pathways, including MTOR, SIRT1, Nrf2 and NF-κB. However, there are few reviews focusing on the critical role of MitoQ as a therapeutic agent in the treatment of MetS. The purpose of this review is to summarize the mitochondrial role in the pathogenesis of MetS, especially in obesity and type 2 diabetes, and discuss the effect and underlying mechanism of MitoQ on mitochondrial homeostasis in MetS.
Collapse
Affiliation(s)
- Jing Yang
- Chongqing Engineering Research Center for Processing & Storage of Distinct Agricultural Products, Chongqing Technology and Business University, Chongqing, China.,Graduate School, Chongqing Technology and Business University, Chongqing, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing, China
| |
Collapse
|
41
|
Huang L, Ren P, Ouyang Z, Wei T, Kong X, Li T, Yin Y, He S, Yang C, He Q. Effect of fermented feed on growth performance, holistic metabolism and fecal microbiota in weanling piglets. Anim Feed Sci Technol 2020. [DOI: 10.1016/j.anifeedsci.2020.114505] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
42
|
Beyond Body Weight-Loss: Dietary Strategies Targeting Intrahepatic Fat in NAFLD. Nutrients 2020; 12:nu12051316. [PMID: 32384593 PMCID: PMC7284418 DOI: 10.3390/nu12051316] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/21/2020] [Accepted: 04/29/2020] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as the most prevalent liver disease in industrialized countries. It is regarded as the hepatic manifestation of the metabolic syndrome (MetS) resulting from insulin resistance. Moreover, insulin resistance impairs glycogen synthesis, postprandially diverting a substantial amount of carbohydrates to the liver and storing them there as fat. NAFLD has far-reaching metabolic consequences involving glucose and lipoprotein metabolism disorders and risk of cardiovascular disease, the leading cause of death worldwide. No pharmaceutical options are currently approved for the treatment of NAFLD. Exercise training and dietary interventions remain the cornerstone of NAFLD treatment. Current international guidelines state that the primary goal of nutritional therapy is to reduce energy intake to achieve a 7%-10% reduction in body weight. Meal replacement therapy (formula diets) results in more pronounced weight loss compared to conventional calorie-restricted diets. However, studies have shown that body mass index (BMI) or weight reduction is not obligatory for decreasing hepatic fat content or to restore normal liver function. Recent studies have achieved significant reductions in liver fat with eucaloric diets and without weight loss through macronutrient modifications. Based on this evidence, an integrative nutritional therapeutic concept was formulated that combines the most effective nutrition approaches termed "liver-fasting." It involves the temporary use of a low calorie diet (total meal replacement with a specific high-protein, high-soluble fiber, lower-carbohydrate formula), followed by stepwise food reintroduction that implements a Mediterranean style low-carb diet as basic nutrition.
Collapse
|
43
|
Zhang Q, Fan X, Ye R, Hu Y, Zheng T, Shi R, Cheng W, Lv X, Chen L, Liang P. The Effect of Simvastatin on Gut Microbiota and Lipid Metabolism in Hyperlipidemic Rats Induced by a High-Fat Diet. Front Pharmacol 2020; 11:522. [PMID: 32410994 PMCID: PMC7201051 DOI: 10.3389/fphar.2020.00522] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
The objective of this study was to investigate the effects of simvastatin (SIM) on lipid metabolism disorders and gut microbiota in high-fat diet-induced hyperlipidemic rats. The obtained results revealed that feeding rats with SIM (20 mg/kg/day) significantly decreased serum lipid level and inhibited hepatic lipid accumulation and steatosis. Histological analysis further indicated that SIM reduced lipid deposition in adipocytes and hepatocytes in comparison with that of the HFD group. The underlying mechanisms of SIM administration against HFD-induced hyperlipidemia were also studied by UPLC-Q-TOF/MS-based liver metabonomics coupled with pathway analysis. Metabolic pathway enrichment analysis of liver metabolites with significant difference in abundance indicated that fatty acids metabolism and amino acid metabolism were the main metabolic pathways altered by SIM administration. Meanwhile, operational taxonomic units (OTUs) analysis revealed that oral administration of SIM altered the composition of gut microbiota, including Ruminococcaceae (OTU960) and Lactobacillus (OTU152), and so on. Furthermore, SIM treatment also regulated the mRNA levels of the genes involved in lipid and cholesterol metabolism. Immunohistochemistry (IHC) analysis of the liver-related proteins (CD36, CYP7A1 and SREBP-1C) showed that oral administration of SIM could regulate the levels of the protein expression related to hepatic lipid metabolism.
Collapse
Affiliation(s)
- Qing Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaoyun Fan
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Rui Ye
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yuzhong Hu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Tingting Zheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Rui Shi
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Wenjian Cheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xucong Lv
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, China
| | - Lijiao Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Peng Liang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
44
|
Yong HY, Larrouy-Maumus G, Zloh M, Smyth R, Ataya R, Benton CM, Munday MR. Early detection of metabolic changes in drug-induced steatosis using metabolomics approaches. RSC Adv 2020; 10:41047-41057. [PMID: 35519189 PMCID: PMC9057704 DOI: 10.1039/d0ra06577c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/23/2020] [Indexed: 12/26/2022] Open
Abstract
Steatosis is the accumulation of triglycerides in hepatic cells wherein fats exceed 5% of the entire liver weight. Although steatotic liver damage is reversible due to the liver's regenerative capability, protracted damage often and typically leads to irreversible conditions such as cirrhosis and hepatocellular carcinoma (HCC). Therefore, early steatotic detection is critical for preventing progression to advanced liver diseases. This also becomes particularly important given the higher prevalence of drug usage, as drugs are a frequent cause of liver damage. Currently, the recommendation to diagnose steatosis is using liver enzymes and performing a liver biopsy. Liver biopsy remains the gold standard method of detection, but the procedure is invasive and an unreliable diagnostic tool. Non-invasive, specific and sensitive diagnostic solutions such as biomarkers are therefore needed for the early detection of steatosis. Our aim is to identify changes in urinary metabolites in tetracycline-induced hepatic steatotic rats at different stages of the diseases using metabolomic-based techniques. Sprague Dawley male rats are treated by intraperitoneal injection (I.P.) with either 62.5 mg kg−1 or 125 mg kg−1 tetracycline, an antibiotic previously known to induce steatosis. We analyse the metabolic profile of the urinary tetracycline induced hepatic steatotic rats using 1H nuclear magnetic resonance (NMR), 2D 1H–1H TOCSY (total correlation spectroscopy) and electrospray liquid chromatography-mass spectrometry (ESI-LC-MS/MS) based metabolomics. The combined analysis of haematoxylin & eosin (H&E), oil red O (ORO) and direct measurement of triglyceride content in the liver tissues of the control samples against 125 mg kg−1 and 62.5 mg kg−1 treated samples, reveals that 125 mg kg−1 tetracycline exposure potentially induces steatosis. The combination of 1H NMR, 2D 1H–1H TOCSY and ESI-LC-MS/MS alongside multivariate statistical analysis, detected a total of 6 urinary metabolites changes, across 6 metabolic pathways. Furthermore, lysine concentration correlates with liver damage as tetracycline dose concentration increases, whilst both H&E and ORO fail to detect hepatocellular damage at the lowest dose concentration. We conclude that the combination of 1H NMR and ESI-LC-MS/MS suggests that these are suitable platforms for studying the pathogenesis of steatosis development, prior to morphological alterations observed in staining techniques and offer a more detailed description of the severity of the steatotic disease. Urinary metabolic profiling of tetracycline induced hepatic steatotic rats were investigated using 1H nuclear magnetic resonance, 2D 1H–1H total correlation spectroscopy and electrospray liquid chromatography-mass spectrometry based metabolomics.![]()
Collapse
Affiliation(s)
- Helena Y Yong
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| | - Gerald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection
- Department of Life Science
- Faculty of Natural Sciences
- Imperial College London
- UK
| | - Mire Zloh
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| | - Rosemary Smyth
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| | - Rayan Ataya
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| | | | - Michael R. Munday
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| |
Collapse
|
45
|
Miyata M, Funaki A, Fukuhara C, Sumiya Y, Sugiura Y. Taurine attenuates hepatic steatosis in a genetic model of fatty liver disease. J Toxicol Sci 2020; 45:87-94. [DOI: 10.2131/jts.45.87] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Masaaki Miyata
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University
| | - Akihiro Funaki
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University
| | - Chiaki Fukuhara
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University
| | - Yukino Sumiya
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University
| | - Yoshimasa Sugiura
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University
| |
Collapse
|
46
|
Liu Y, Zhang X, Guan T, Jia S, Liu Y, Zhao X. Effects of quercetin on cadmium-induced toxicity in rat urine using metabonomics techniques. Hum Exp Toxicol 2019; 39:524-536. [PMID: 31876187 DOI: 10.1177/0960327119895811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This study aimed to analyse the protective effects of quercetin on the toxicity of cadmium (Cd) using metabonomics techniques. Sixty male Sprague-Dawley rats were randomly divided into six groups (n = 10): control group (C), low-dose quercetin-treated group (Q1; 10 mg/kg bw/day), high-dose quercetin-treated group (Q2; 50 mg/kg bw/day), Cd-treated group (D; 4.89 mg/kg bw/day), low-dose quercetin plus Cd-treated group (DQ1) and high-dose quercetin plus Cd-treated group (DQ2). The rats continuously received quercetin and Cd via gavage and drinking water for 12 weeks, respectively. The rat urine samples were collected for metabonomics analysis. Finally, 10 metabolites were identified via the metabonomics profiles of the rat urine samples. Compared with the control group, the intensities of taurine, phosphocreatine, l-carnitine and uric acid were significantly decreased (p < 0.01) and those of LysoPC (18: 2 (9Z, 12Z)), guanidinosuccinic acid, dopamine, 2,5,7,8-tetramethyl-2(2'-carboxyethyl)-6-hydroxychroman and allantoic acid were significantly increased (p < 0.01) in the Cd-treated group. However, the intensities of the aforementioned metabolites had restorative changes in the high-dose quercetin plus Cd-treated groups unlike those in Cd-treated group (p < 0.01 or p < 0.05). Results indicated that quercetin exerts protective effects on Cd-induced toxicity by regulating energy and lipid metabolism, enhancing the antioxidant defence system and protecting liver and kidney function and so on.
Collapse
Affiliation(s)
- Y Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - X Zhang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - T Guan
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - S Jia
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Y Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - X Zhao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| |
Collapse
|
47
|
Lipid Profile Changes Induced by Chronic Administration of Anabolic Androgenic Steroids and Taurine in Rats. ACTA ACUST UNITED AC 2019; 55:medicina55090540. [PMID: 31462007 PMCID: PMC6780624 DOI: 10.3390/medicina55090540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 11/17/2022]
Abstract
Background and Objectives: Anabolic androgenic steroids (AAS), used as a therapy in various diseases and abused in sports, are atherogenic in supraphysiological administration, altering the plasma lipid profile. Taurine, a conditionally-essential amino acid often used in dietary supplements, was acknowledged to delay the onset and progression of atherogenesis, and to mitigate hyperlipidemia. The aim of the present study was to verify if taurine could prevent the alterations induced by concomitant chronic administration of high doses of AAS nandrolone decanoate (DECA) in rats. Materials and Methods: Thirty-two male Wistar rats, assigned to 4 equal groups, were treated for 12 weeks either with DECA (A group), taurine (T group), both DECA and taurine (AT group) or vehicle (C group). Plasma triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), hepatic triglycerides (TGh) and liver non-esterified fatty acids (NEFA) were then determined. Results: DECA elevated TG level in A group vs. control (p = 0.01), an increase prevented by taurine association in AT group (p = 0.04). DECA decreased HDL-C in A group vs. control (p = 0.02), while taurine tended to increase it in AT group. DECA decreased TGh (p = 0.02) in A group vs. control. Taurine decreased TGh in T (p = 0.004) and AT (p < 0.001) groups vs. control and tended to lower NEFA (p = 0.08) in AT group vs. A group. Neither DECA, nor taurine influenced TC and LDL-C levels. Conclusions: Taurine partially prevented the occurrence of DECA negative effects on lipid profile, suggesting a therapeutic potential in several conditions associated with chronic high levels of plasma androgens, such as endocrine disorders or AAS-abuse.
Collapse
|
48
|
Du Y, Liu H, Zhang M, Zhang S, Hu J, Wu G, Yang J. Taurine Increases Spermatozoa Quality and Function in Asthenospermia Rats Impaired by Ornidazole. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1155:507-520. [PMID: 31468427 DOI: 10.1007/978-981-13-8023-5_47] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Asthenospermia has been considered as one of the crucial causes of male infertility, which was closely related to epididymal dysfunction. Lots of documents have revealed that taurine palys an important role in male reproduction, including antioxidation, membrane stabilization, stimulation of sexual hormone secretion and elevation of sperm quality. The objective of this study was to expose the effect of taurine on spermatozoa quality and function in ornidazole-induced asthenospermia rats. We found that taurine treatment could obviously recover the decline of cauda epididymal sperm count, viability and motility, and the elevation of sperm abnormality in asthenospermia animals. Spermatozoa acrosin, LDH-X, SDH and CCO activities of model rats also were notably increased by taurine administration. The present data indicated that taurine could raise spermatozoa quality and function by elevating mitochondrial energy metabolism. Notably, taurine supplementation markedly raised serum GnRH, LH and T levels in asthenospermia rays, suggesting taurine rescued asthenosperm by means of stimulating hypothalamic-pituitary-testicular axis secretion. We also found that concentrations of asthenospermia epididymal carnitine, SA, α-Glu and ACP, and mRNA expression levels of MMP7 and IDO2 were significantly rised by taurine administration, indicating taurine may protect epididymal epithelium structure, improve secretion activity, and maintain intraluminal microenvironment homeostasis. Finally, the present results showed taurine effectively increased cauda epididymal SOD, GSH and γ-GT levels in model rats, reduced ROS and MDA production, suggesting epididymal antioxidant ability of asthenospermia rats could be elevated by taurine treatment. To sum up, our results indicated that taurine can promote spermatozoa quality and function in ornidazole-induced asthenospermia rats by facilitating epididymal epithelium secretion and luminal microenvironment homeostasis.
Collapse
Affiliation(s)
- Yanting Du
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Hong Liu
- Experimental Animal Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Meng Zhang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Shu Zhang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Jianmin Hu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Gaofeng Wu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Jiancheng Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| |
Collapse
|