1
|
Guo X, Ou T, Yang X, Song Q, Zhu L, Mi S, Zhang J, Zhang Y, Chen W, Guo J. Untargeted metabolomics based on ultra-high performance liquid chromatography-mass spectrometry/MS reveals the lipid-lowering mechanism of taurine in hyperlipidemia mice. Front Nutr 2024; 11:1367589. [PMID: 38706565 PMCID: PMC11066166 DOI: 10.3389/fnut.2024.1367589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/03/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction Taurine has a prominent lipid-lowering effect on hyperlipidemia. However, a comprehensive analysis of the effects of taurine on endogenous metabolites in hyperlipidemia has not been documented. This study aimed to explore the impact of taurine on multiple metabolites associated with hyperlipidemia. Methods The hyperlipidemic mouse model was induced by high-fat diet (HFD). Taurine was administered via oral gavage at doses of 700 mg/kg/day for 14 weeks. Evaluation of body weight, serum lipid levels, and histopathology of the liver and adipose tissue was performed to confirm the lipid-lowering effect of taurine. Ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS)-based metabonomics analyses of serum, urine, feces, and liver, coupled with multivariate data analysis, were conducted to assess changes in the endogenous metabolites. Results and discussion Biochemical and histological examinations demonstrated that taurine administration prevented weight gain and dyslipidemia, and alleviated lipid deposition in the liver and adipose tissue in hyperlipidemic mice. A total of 76 differential metabolites were identified by UPLC-MS-based metabolomics approach, mainly involving BAs, GPs, SMs, DGs, TGs, PUFAs and amino acids. Taurine was found to partially prevent HFDinduced abnormalities in the aforementioned metabolites. Using KEGG database and MetaboAnalyst software, it was determined that taurine effectively alleviates metabolic abnormalities caused by HFD, including fatty acid metabolism, sphingolipid metabolism, glycerophospholipid metabolism, diacylglycerol metabolism, amino acid metabolism, bile acid and taurine metabolism, taurine and hypotaurine metabolism. Moreover, DGs, GPs and SMs, and taurine itself may serve as active metabolites in facilitating various anti-hyperlipidemia signal pathways associated with taurine. This study provides new evidence for taurine to prevent hyperlipidemia.
Collapse
Affiliation(s)
- Xinzhe Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Tong Ou
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Xinyu Yang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Qi Song
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Lin Zhu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Shengquan Mi
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Jing Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Yanzhen Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Wen Chen
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Junxia Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| |
Collapse
|
2
|
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
|
3
|
Du Q, Teng M, Yang L, Meng C, Qiu Y, Wang C, Chen J, Wang T, Chen S, Luo Y, Sun J, Dong Y. Metabolic characteristics of voriconazole - Induced liver injury in rats. Chem Biol Interact 2023; 383:110693. [PMID: 37659626 DOI: 10.1016/j.cbi.2023.110693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/14/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
Voriconazole (VOR) - induced liver injury is a common adverse reaction, and can lead to serious clinical outcomes. It is of great significance to describe the metabolic characteristics of VOR - induced liver injury and to elucidate the potential mechanisms. This study investigated the changes of plasma metabolic profiles in a rat model of VOR - induced liver injury by non - targeted metabolomics. Correlation analysis was performed between differentially expressed metabolites and plasma liver function indexes. The metabolites with strong correlation were determined for their predictive performance for liver injury using receiver operating characteristic (ROC) curve analysis. Potential biomarkers were then screened combined with liver pathological scores. Finally, the expression level of genes that involved in lipid metabolism were determined in rat liver to verify the mechanism of VOR - induced liver injury we proposed. VOR - induced liver injury in rats was characterized by plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) elevation, the lipid droplets accumulation in liver, as well as inflammation and fibrosis. Significant changes of plasma metabolites were observed, with a decrease in lipid metabolites accounting for over 50% of all changed metabolites, and alterations of cholesterol and bile acids metabolites. The decrease of 3 phosphatidylcholine (PC) in plasma could indicate the occurrence of VOR - induced liver injury. Decreased fatty acids (FA) oxidation and bile acid excretion might be the potential mechanisms of VOR - induced liver injury. This study provided new insights into the molecular characterization of VOR - induced liver injury.
Collapse
Affiliation(s)
- Qian Du
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Mengmeng Teng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Luting Yang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Chao Meng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yulan Qiu
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Chuhui Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiaojiao Chen
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Taotao Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Siying Chen
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yu Luo
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jinyao Sun
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
4
|
Yang H, Li X, Jin H, Turkez H, Ozturk G, Doganay HL, Zhang C, Nielsen J, Uhlén M, Borén J, Mardinoglu A. Longitudinal metabolomics analysis reveals the acute effect of cysteine and NAC included in the combined metabolic activators. Free Radic Biol Med 2023:S0891-5849(23)00429-X. [PMID: 37245532 DOI: 10.1016/j.freeradbiomed.2023.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/30/2023]
Abstract
Growing evidence suggests that the depletion of plasma NAD+ and glutathione (GSH) may play an important role in the development of metabolic disorders. The administration of Combined Metabolic Activators (CMA), consisting of GSH and NAD+ precursors, has been explored as a promising therapeutic strategy to target multiple altered pathways associated with the pathogenesis of the diseases. Although studies have examined the therapeutic effect of CMA that contains N-acetyl-l-cysteine (NAC) as a metabolic activator, a system-wide comparison of the metabolic response to the administration of CMA with NAC and cysteine remains lacking. In this placebo-controlled study, we studied the acute effect of the CMA administration with different metabolic activators, including NAC or cysteine with/without nicotinamide or flush free niacin, and performed longitudinal untargeted-metabolomics profiling of plasma obtained from 70 well-characterized healthy volunteers. The time-series metabolomics data revealed the metabolic pathways affected after the administration of CMAs showed high similarity between CMA containing nicotinamide and NAC or cysteine as metabolic activators. Our analysis also showed that CMA with cysteine is well-tolerated and safe in healthy individuals throughout the study. Last, our study systematically provided insights into a complex and dynamics landscape involved in amino acid, lipid and nicotinamide metabolism, reflecting the metabolic responses to CMA administration containing different metabolic activators.
Collapse
Affiliation(s)
- Hong Yang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden.
| | - Xiangyu Li
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden; Bash Biotech Inc, 600 West Broadway, Suite 700, San Diego, CA, 92101, USA
| | - Han Jin
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Gurkan Ozturk
- Research Institute for Health Sciences and Technologies (SABITA), International School of Medicine, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Hamdi Levent Doganay
- Gastroenterology and Hepatology Unit, VM Pendik Medicalpark Teaching Hospital, İstanbul, Turkey; Department of Internal Medicine, Bahçeşehir University (BAU), Istanbul, Turkey
| | - Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden; BioInnovation Institute, Ole Maaløes Vej 3, DK2200, Copenhagen, Denmark
| | - Mathias Uhlén
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
5
|
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: 2] [Impact Index Per Article: 2.0] [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
|
6
|
Xu M, Lan R, Qiao L, Lin X, Hu D, Zhang S, Yang J, Zhou J, Ren Z, Li X, Liu G, Liu L, Xu J. Bacteroides vulgatus Ameliorates Lipid Metabolic Disorders and Modulates Gut Microbial Composition in Hyperlipidemic Rats. Microbiol Spectr 2023; 11:e0251722. [PMID: 36625637 PMCID: PMC9927244 DOI: 10.1128/spectrum.02517-22] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hyperlipidemia is a risk factor and key indicator for cardiovascular diseases, and the gut microbiota is highly associated with hyperlipidemia. Bacteroides vulgatus is a prevalent mutualist across human populations and confers multiple health benefits such as immunoregulation, antiobesity, and coronary artery disease intervention. However, its role in antihyperlipidemia has not been systematically characterized. This study sought to identify the effect of B. vulgatus Bv46 on hyperlipidemia. Hyperlipidemic rats were modeled by feeding them a high-fat diet for 6 weeks. The effect of B. vulgatus Bv46 supplementation was evaluated by measuring anthropometric parameters, lipid and inflammation markers, and the liver pathology. Multi-omics was used to explore the underlying mechanisms. The ability of B. vulgatus Bv46 to produce bile salt hydrolase was confirmed by gene annotation and in vitro experiments. Oral administration of B. vulgatus Bv46 in hyperlipidemic rats significantly reduced the body weight gain, food efficiency, and liver index, improved the serum lipid profile, lowered the levels of serum inflammatory cytokines, promoted the loss of fecal bile acids (BAs), and extended the fecal pool of short-chain fatty acids (SCFAs), especially propionate and butyrate. B. vulgatus Bv46 induced compositional shifts of the gut microbial community of hyperlipidemic rats, characterized by a lower ratio of Firmicutes to Bacteroidetes with an increase of genera Bacteroides and Parabacteroides. After intervention, serum metabolite profiling exhibited an adaptation in amino acids and glycerophospholipid metabolism. Transcriptomics further detected altered biological processes, including primary bile acid biosynthesis and fatty acid metabolic process. Taken together, the findings suggest that B. vulgatus Bv46 could be a promising candidate for interventions against hyperlipidemia. IMPORTANCE As a core microbe of the human gut ecosystem, Bacteroides vulgatus has been linked to multiple aspects of metabolic disorders in a collection of associative studies, which, while indicative, warrants more direct experimental evidence to verify. In this study, we experimentally demonstrated that oral administration of B. vulgatus Bv46 ameliorated the serum lipid profile and systemic inflammation of high-fat diet-induced hyperlipidemic rats in a microbiome-regulated manner, which appears to be associated with changes of bile acid metabolism, short-chain fatty acid biosynthesis, and serum metabolomic profile. This finding supports the causal contribution of B. vulgatus in host metabolism and helps to form the basis of novel therapies for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Mingchao Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Lei Qiao
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoying Lin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dalong Hu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Suping Zhang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jing Yang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
| | - Juan Zhou
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhihong Ren
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xianping Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guoxing Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liyun Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianguo Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Public Health, Nankai University, Tianjin, China
| |
Collapse
|
7
|
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: 2.0] [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
|
8
|
Ruminal Degradation of Taurine and Its Effects on Rumen Fermentation In Vitro. FERMENTATION-BASEL 2023. [DOI: 10.3390/fermentation9010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Taurine accounts for approximately 0.1% of an animal’s body. It cannot be used for protein synthesis but plays a wide range of important roles in the animal body. Taurine does not exist in plants, while mammals can only synthesize 30–40% of the taurine they need. Supplementing taurine to beef cattle may be necessary to improve their nutrient utilization and health status. However, no data are available regarding the metabolism of taurine in the rumen. Two in vitro trials were conducted to investigate the ruminal degradability of taurine and its effects on rumen fermentation. In Trial 1, Tilley and Terry’s in vitro rumen fermentation technique was used for incubation. As treatments, two levels of taurine, i.e., 0 and 10 mg, were added into plastic tubes containing 0.4000 g of feed mixture with a calibrated volume of 50 mL. Three adult cattle fitted with rumen cannulas were used as the donors for rumen fluid. The incubation was carried out at 39 °C for 48 h. The results showed that the taurine degradability increased with incubation time (p < 0.001) while its 2 h-degradability reached 99%. Taurine decreased the 48 h-dry matter degradability (DMD) (p = 0.008) and increased the 24 h- and 48 h-pH (p = 0.005; p = 0.018), respectively. In Trial 2, the Hohenheim gas test was used for incubation. Four levels of taurine, i.e., 0, 5, 10 and 20 mg, were added into glass syringes containing 0.2000 g feed mixture with a calibrated volume of 100 mL as treatments. The rumen fluid donors were the same as in Trial 1. The incubation was carried out at 39 °C for 48 h. The results showed that taurine increased the 48 h-pH (p < 0.001) linearly, decreased the cumulative gas production (p < 0.001) and the total volatile fatty acids (VFA) concentration (p = 0.014), and quadratically affected the ammonia–nitrogen (p < 0.001) and microbial crude protein (MCP) concentrations (p < 0.001). It was concluded that taurine was highly degradable in rumen fermentation. Taurine inhibits ruminal fermentation by decreasing DMD, VFA and gas production while improving MCP synthesis on a dose-dependent basis.
Collapse
|
9
|
Moludi J, Qaisar SA, Kadhim MM, Ahmadi Y, Davari M. Protective and therapeutic effectiveness of taurine supplementation plus low calorie diet on metabolic parameters and endothelial markers in patients with diabetes mellitus: a randomized, clinical trial. Nutr Metab (Lond) 2022; 19:49. [PMID: 35870947 PMCID: PMC9308194 DOI: 10.1186/s12986-022-00684-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 05/31/2022] [Indexed: 12/22/2022] Open
Abstract
Background Taurine supplementation as a sulfur-containing amino acid may attenuate and/or alleviate diabetes-induced complications and endothelial dysfunction via its anti-inflammatory and antioxidant activities. Our purpose was to investigate the effect of Taurine supplementation on endothelial dysfunction markers, oxidative stress, inflammation, and glycemic control in patients with type 2 diabetes mellitus (T2DM). Methods In the current clinical trial, 120 patients with T2DM were randomly allocated to take either Taurine (containing 1 g Taurine, n = 60) or placebo (n = 60) three times per day for an eight-week period. Moreover, all patients were on a low-calorie diet. The primary outcome was fasting blood glucose (FBG) and endothelial markers including sera intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule (VCAM), and matrix metallopeptidase 9 (MMP-9). The secondary outcome was dietary intake, anthropometric indices, serum insulin and Homeostatic Model Assessment of Insulin Resistance (HOMA-IR), total antioxidant capacity (TAC), tumor necrosis factor (TNF), high-sensitivity C-reactive protein (hs-CRP), malondialdehyde (MDA), and lipid profile.
Results After 8 weeks, Taurine-supplemented patients had a considerable decrease in serum insulin and HOMA-IR compared to placebo group. However, Taurine supplementation did not improve other metabolic parameters including lipid profiles, glycated hemoglobin, and fasting blood glucose (FBG). There was a significant decline in MDA, TNF, and hs-CRP levels after these eight-week period of Taurine supplementation. In addition, the Taurine group had fewer serum levels of endothelial dysfunction markers than the placebo group. Conclusions The evidence from our study revealed that Taurine supplementation significantly reduced insulin and HOMA-IR, as well as oxidative stress, inflammation, and endothelial markers in individuals with T2DM.
Trial registration The protocol of the study was recorded in the Iranian Registry of Clinical Trials (IRCT20180712040438N3).
Collapse
|
10
|
Chen M, Bai F, Song T, Niu X, Wang X, Wang K, Ye J. Hepatic Transcriptome Analysis Provides New Insight into the Lipid-Reducing Effect of Dietary Taurine in High-Fat Fed Groupers ( Epinephelus coioides). Metabolites 2022; 12:670. [PMID: 35888794 PMCID: PMC9318954 DOI: 10.3390/metabo12070670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 12/18/2022] Open
Abstract
A transcriptome analysis was conducted to provide the first detailed overview of dietary taurine intervention on liver lipid accumulation caused by high-fat in groupers. After an eight-week feeding, the fish fed 15% fat diet (High-fat diet) had higher liver lipid contents vs. fish fed 10% fat diet (Control diet). 15% fat diet with 1% taurine (Taurine diet) improved weight gain and feed utilization, and decreased hepatosomatic index and liver lipid contents vs. the High-fat diet. In the comparison of the Control vs. High-fat groups, a total of 160 differentially expressed genes (DEGs) were identified, of which up- and down-regulated genes were 72 and 88, respectively. There were 49 identified DEGs with 26 and 23 of up- and down-regulated in the comparison to High-fat vs. Taurine. Several key genes, such as cysteine dioxygenase (CDO1), ADP-ribosylation factor 1/2 (ARF1_2), sodium/potassium-transporting ATPase subunit alpha (ATP1A), carnitine/acylcarnitine translocase (CACT), and calcium/calmodulin-dependent protein kinase II (CAMK) were obtained by enrichment for the above DEGs. These genes were enriched in taurine and hypotaurine metabolism, bile secretion, insulin secretion, phospholipase D signaling pathway, and thermogenesis pathways, respectively. The present study will also provide a new insight into the nutritional physiological function of taurine in farmed fish.
Collapse
Affiliation(s)
- Mingfan Chen
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen 361021, China; (M.C.); (F.B.); (T.S.); (X.N.); (K.W.)
| | - Fakai Bai
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen 361021, China; (M.C.); (F.B.); (T.S.); (X.N.); (K.W.)
| | - Tao Song
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen 361021, China; (M.C.); (F.B.); (T.S.); (X.N.); (K.W.)
| | - Xingjian Niu
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen 361021, China; (M.C.); (F.B.); (T.S.); (X.N.); (K.W.)
| | - Xuexi Wang
- Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China or
| | - Kun Wang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen 361021, China; (M.C.); (F.B.); (T.S.); (X.N.); (K.W.)
| | - Jidan Ye
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen 361021, China; (M.C.); (F.B.); (T.S.); (X.N.); (K.W.)
| |
Collapse
|
11
|
Macroalgal Proteins: A Review. Foods 2022; 11:foods11040571. [PMID: 35206049 PMCID: PMC8871301 DOI: 10.3390/foods11040571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/08/2022] [Indexed: 12/11/2022] Open
Abstract
Population growth is the driving change in the search for new, alternative sources of protein. Macroalgae (otherwise known as seaweeds) do not compete with other food sources for space and resources as they can be sustainably cultivated without the need for arable land. Macroalgae are significantly rich in protein and amino acid content compared to other plant-derived proteins. Herein, physical and chemical protein extraction methods as well as novel techniques including enzyme hydrolysis, microwave-assisted extraction and ultrasound sonication are discussed as strategies for protein extraction with this resource. The generation of high-value, economically important ingredients such as bioactive peptides is explored as well as the application of macroalgal proteins in human foods and animal feed. These bioactive peptides that have been shown to inhibit enzymes such as renin, angiotensin-I-converting enzyme (ACE-1), cyclooxygenases (COX), α-amylase and α-glucosidase associated with hypertensive, diabetic, and inflammation-related activities are explored. This paper discusses the significant uses of seaweeds, which range from utilising their anthelmintic and anti-methane properties in feed additives, to food techno-functional ingredients in the formulation of human foods such as ice creams, to utilising their health beneficial ingredients to reduce high blood pressure and prevent inflammation. This information was collated following a review of 206 publications on the use of seaweeds as foods and feeds and processing methods to extract seaweed proteins.
Collapse
|
12
|
Tagawa R, Kobayashi M, Sakurai M, Yoshida M, Kaneko H, Mizunoe Y, Nozaki Y, Okita N, Sudo Y, Higami Y. Long-Term Dietary Taurine Lowers Plasma Levels of Cholesterol and Bile Acids. Int J Mol Sci 2022; 23:ijms23031793. [PMID: 35163722 PMCID: PMC8836270 DOI: 10.3390/ijms23031793] [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: 12/25/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022] Open
Abstract
Cholesterol is an essential lipid in vertebrates, but excess blood cholesterol promotes atherosclerosis. In the liver, cholesterol is metabolized to bile acids by cytochrome P450, family 7, subfamily a, polypeptide 1 (CYP7A1), the transcription of which is negatively regulated by the ERK pathway. Fibroblast growth factor 21 (FGF21), a hepatokine, induces ERK phosphorylation and suppresses Cyp7a1 transcription. Taurine, a sulfur-containing amino acid, reportedly promotes cholesterol metabolism and lowers blood and hepatic cholesterol levels. However, the influence of long-term feeding of taurine on cholesterol levels and metabolism remains unclear. Here, to evaluate the more chronic effects of taurine on cholesterol levels, we analyzed mice fed a taurine-rich diet for 14-16 weeks. Long-term feeding of taurine lowered plasma cholesterol and bile acids without significantly changing other metabolic parameters, but hardly affected these levels in the liver. Moreover, taurine upregulated Cyp7a1 levels, while downregulated phosphorylated ERK and Fgf21 levels in the liver. Likewise, taurine-treated Hepa1-6 cells, a mouse hepatocyte line, exhibited downregulated Fgf21 levels and upregulated promoter activity of Cyp7a1. These results indicate that taurine promotes cholesterol metabolism by suppressing the FGF21/ERK pathway followed by upregulating Cyp7a1 expression. Collectively, this study shows that long-term feeding of taurine lowers both plasma cholesterol and bile acids, reinforcing that taurine effectively prevents hypercholesterolemia.
Collapse
Affiliation(s)
- Ryoma Tagawa
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (R.T.); (M.S.); (M.Y.); (H.K.); (Y.N.); (Y.S.)
| | - Masaki Kobayashi
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (R.T.); (M.S.); (M.Y.); (H.K.); (Y.N.); (Y.S.)
- Correspondence: (M.K.); (Y.H.); Tel.: +81-4-7121-3676 (M.K.); +81-4-7121-3675 (Y.H.)
| | - Misako Sakurai
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (R.T.); (M.S.); (M.Y.); (H.K.); (Y.N.); (Y.S.)
| | - Maho Yoshida
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (R.T.); (M.S.); (M.Y.); (H.K.); (Y.N.); (Y.S.)
| | - Hiroki Kaneko
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (R.T.); (M.S.); (M.Y.); (H.K.); (Y.N.); (Y.S.)
| | - Yuhei Mizunoe
- Department of Internal Medicine Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
| | - Yuka Nozaki
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (R.T.); (M.S.); (M.Y.); (H.K.); (Y.N.); (Y.S.)
| | - Naoyuki Okita
- Division of Pathological Biochemistry, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan;
| | - Yuka Sudo
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (R.T.); (M.S.); (M.Y.); (H.K.); (Y.N.); (Y.S.)
| | - Yoshikazu Higami
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (R.T.); (M.S.); (M.Y.); (H.K.); (Y.N.); (Y.S.)
- Division of Integrated Research, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
- Correspondence: (M.K.); (Y.H.); Tel.: +81-4-7121-3676 (M.K.); +81-4-7121-3675 (Y.H.)
| |
Collapse
|
13
|
Chen M, Zhu JY, Mu WJ, Guo L. Cysteine dioxygenase type 1 (CDO1): its functional role in physiological and pathophysiological processes. Genes Dis 2022. [DOI: 10.1016/j.gendis.2021.12.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
14
|
Roşca AE, Vlădăreanu AM, Mirica R, Anghel-Timaru CM, Mititelu A, Popescu BO, Căruntu C, Voiculescu SE, Gologan Ş, Onisâi M, Iordan I, Zăgrean L. Taurine and Its Derivatives: Analysis of the Inhibitory Effect on Platelet Function and Their Antithrombotic Potential. J Clin Med 2022; 11:jcm11030666. [PMID: 35160118 PMCID: PMC8837186 DOI: 10.3390/jcm11030666] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/23/2022] [Accepted: 01/26/2022] [Indexed: 11/16/2022] Open
Abstract
Taurine is a semi-essential, the most abundant free amino acid in the human body, with a six times higher concentration in platelets than any other amino acid. It is highly beneficial for the organism, has many therapeutic actions, and is currently approved for heart failure treatment in Japan. Taurine has been repeatedly reported to elicit an inhibitory action on platelet activation and aggregation, sustained by in vivo, ex vivo, and in vitro animal and human studies. Taurine showed effectiveness in several pathologies involving thrombotic diathesis, such as diabetes, traumatic brain injury, acute ischemic stroke, and others. As human prospective studies on thrombosis outcome are very difficult to carry out, there is an obvious need to validate existing findings, and bring new compelling data about the mechanisms underlying taurine and derivatives antiplatelet action and their antithrombotic potential. Chloramine derivatives of taurine proved a higher stability and pronounced selectivity for platelet receptors, raising the assumption that they could represent future potential antithrombotic agents. Considering that taurine and its analogues display permissible side effects, along with the need of finding new, alternative antithrombotic drugs with minimal side effects and long-term action, the potential clinical relevance of this fascinating nutrient and its derivatives requires further consideration.
Collapse
Affiliation(s)
- Adrian Eugen Roşca
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.-M.A.-T.); (C.C.); (S.E.V.); (L.Z.)
- Department of Cardiology, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania
- Correspondence: (A.E.R.); (A.-M.V.)
| | - Ana-Maria Vlădăreanu
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (A.M.); (M.O.); (I.I.)
- Correspondence: (A.E.R.); (A.-M.V.)
| | - Radu Mirica
- Department of Surgery, “Carol Davila” University of Medicine and Pharmacy, “Sf. Ioan” Clinical Hospital, 042122 Bucharest, Romania;
| | - Cristina-Mihaela Anghel-Timaru
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.-M.A.-T.); (C.C.); (S.E.V.); (L.Z.)
| | - Alina Mititelu
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (A.M.); (M.O.); (I.I.)
| | - Bogdan Ovidiu Popescu
- Department of Neurology, “Carol Davila” University of Medicine and Pharmacy, Colentina Clinical Hospital, 020125 Bucharest, Romania;
| | - Constantin Căruntu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.-M.A.-T.); (C.C.); (S.E.V.); (L.Z.)
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Suzana Elena Voiculescu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.-M.A.-T.); (C.C.); (S.E.V.); (L.Z.)
| | - Şerban Gologan
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, Elias Clinical Hospital, 011461 Bucharest, Romania;
| | - Minodora Onisâi
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (A.M.); (M.O.); (I.I.)
| | - Iuliana Iordan
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (A.M.); (M.O.); (I.I.)
- Department of Medical Semiology and Nephrology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Leon Zăgrean
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.-M.A.-T.); (C.C.); (S.E.V.); (L.Z.)
| |
Collapse
|
15
|
Rais N, Parveen K, Ahmad R, Siddiqui WA, Nadeem A, Ved A. S-allyl Cysteine and Taurine revert peripheral metabolic and lipid profile in non-insulin-dependent diabetes mellitus animals: Combination vs Monotherapy. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e201183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
| | | | | | | | | | - Akash Ved
- Goel Institute of Pharmaceutical Sciences, India
| |
Collapse
|
16
|
The Disease-Modifying Role of Taurine and Its Therapeutic Potential in Coronavirus Disease 2019 (COVID-19). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:3-21. [DOI: 10.1007/978-3-030-93337-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
17
|
The Effects of TRX Suspension Training Combined with Taurine Supplementation on Body Composition, Glycemic and Lipid Markers in Women with Type 2 Diabetes. Nutrients 2021; 13:nu13113958. [PMID: 34836211 PMCID: PMC8621658 DOI: 10.3390/nu13113958] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 11/24/2022] Open
Abstract
Background: We aimed to investigate the effects of an 8-week total-body resistance exercise (TRX) suspension training intervention combined with taurine supplementation on body composition, blood glucose, and lipid markers in T2D females. Methods: Forty T2D middle-aged females (age: 53 ± 5 years, body mass = 84.3 ± 5.1 kg) were randomly assigned to four groups, TRX suspension training + placebo (TP; n = 10), TRX suspension training + taurine supplementation (TT; n = 10), taurine supplementation (T; n = 10), or control (C; n = 10). Body composition (body mass, body mass index (BMI), body fat percentage (BFP)), blood glucose (fasting blood sugar (FBS)), hemoglobin A1c (HbA1c), Insulin, and Insulin resistance (HOMA-IR), and lipid markers (low-density lipoprotein (LDL), high-density lipoprotein (HDL), triglyceride (TG), and total cholesterol (TC)) were evaluated prior to and after interventions. Results: All three interventions significantly decreased body mass, BMI, and BFP with no changes between them for body mass and BMI; however, BFP changes in the TT group were significantly greater than all other groups. FBS was significantly reduced in TP and TT. Insulin concentrations’ decrement were significantly greater in all experimental groups compared to C; however, no between group differences were observed between TT, TP, and T. In regards to HOMA-IR, decreases in TT were significantly greater than all other groups TG, HbA1c, and LDL were reduced following all interventions. HDL values significantly increased only in the TT group, while TC significantly decreased in TP and TT groups. Changes in HbA1c, TG, HDL, and TC were significantly greater in the TT compared to all other groups. Conclusions: TRX training improved glycemic and lipid profiles, while taurine supplementation alone failed to show hypoglycemic and hypolipidemic properties. Notably, the synergic effects of TRX training and taurine supplementation were shown in HbA1c, HOMA-IR, TG, TC, HDL, and BFP changes. Our outcomes suggest that TRX training + taurine supplementation may be an effective adjuvant therapy in individuals with T2D.
Collapse
|
18
|
Miyata M, Tanaka T, Takahashi K, Funaki A, Sugiura Y. Cholesterol-lowering effects of taurine through the reduction of ileal FXR signaling due to the alteration of ileal bile acid composition. Amino Acids 2021; 53:1523-1532. [PMID: 34596761 DOI: 10.1007/s00726-021-03068-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/14/2021] [Indexed: 01/05/2023]
Abstract
Studies using animal models of hypercholesterolemia have established that taurine reduces cholesterol levels; however, the precise mechanism underlying this cholesterol-lowering effect is unclear. This study addressed this issue by investigating whether bile acid/farnesoid X receptor (FXR) signaling is involved in taurine-mediated cholesterol-lowering effect. Fxr-null and wild-type mice were administered 2% (w/v) taurine in their drinking water and fed a control diet or control diet supplemented with 1% (w/w) cholesterol (cholesterol diet) for 10 days. Taurine intake did not significantly alter hepatic and serum total cholesterol (TC) levels and bile acid compositions of the liver and intestinal lumen in Fxr-null and wild-type mice fed the control diet. By changing to a cholesterol diet, taurine intake significantly decreased hepatic and serum cholesterol levels in wild-type mice. In contrast, it significantly decreased hepatic, not serum, cholesterol levels in Fxr-null mice. Taurine intake significantly altered the bile acid composition of the intestinal lumen in wild-type mice fed a cholesterol diet, but not in Fxr-null mice. An increase in FXR antagonistic bile acids was detected in the intestinal lumen of taurine-treated wild-type mice fed a cholesterol diet. Taurine intake reduced the ileal expression of FXR target genes fibroblast growth factor 15 (Fgf15) and small heterodimer partner (Shp). In contrast, it enhanced the hepatic expression of cholesterol 7α-hydroxylase (Cyp7a1) in wild-type mice fed a cholesterol diet, but not in Fxr-null mice. These results suggest that taurine is partially involved in cholesterol lowering by reducing the ileal FXR signaling due to the alteration of ileal bile acid composition.
Collapse
Affiliation(s)
- Masaaki Miyata
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan.
| | - Tomoyuki Tanaka
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan
| | - Kazuho Takahashi
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan
| | - Akihiro Funaki
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan
| | - Yoshimasa Sugiura
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan
| |
Collapse
|
19
|
Tolerable amounts of amino acids for human supplementation: summary and lessons from published peer-reviewed studies. Amino Acids 2021; 53:1313-1328. [PMID: 34338884 PMCID: PMC8416832 DOI: 10.1007/s00726-021-03054-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/21/2021] [Indexed: 12/31/2022]
Abstract
Amino acid supplementation may be indicated to correct for insufficient amino acid intake in healthy individuals, and in specific physiological or pathophysiological situations. However, there is a concern to not supplement beyond the tolerable upper intake level (UL) by determining parameters of no-observed-adverse-effect level (NOAEL) or lowest-observed-adverse-effect level (LOAEL) for each amino acid. Since the NOAEL and LOAEL values are at least one order of magnitude different when comparing the values obtained in rats and humans, the aim of this review is to evaluate to what extent the amino acid UL measured in the rat model, when referenced to the dietary usual consumption (UC) and dietary requirement (RQ) for indispensable amino acids, may be used as an approximation of the UL in humans. This review then compares the ratios of the NOAEL or LOAEL over UC and RQ in the rat model with the same ratios calculated in humans for the nine amino acids (arginine, serine, glycine, histidine, leucine, lysine, methionine, phenylalanine, and tryptophan) for which this comparison can be done. From the calculations made, it appears that for these 9 amino acids, the calculated ratios for rats and humans, although rather different for several amino acids, remains for all of them in the same order of magnitude. For tryptophan, tyrosine, and valine, the ratios calculated in rats are markedly different according to the sex of animals, raising the view that it may be also the case in humans.
Collapse
|
20
|
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: 84] [Impact Index Per Article: 28.0] [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
|
21
|
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: 6.3] [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
|
22
|
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: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
23
|
Chen G, Chen J, Wu J, Ren X, Li L, Lu S, Cheng T, Tan L, Liu M, Luo Q, Liang S, Nie Q, Zhang X, Luo W. Integrative Analyses of mRNA Expression Profile Reveal SOCS2 and CISH Play Important Roles in GHR Mutation-Induced Excessive Abdominal Fat Deposition in the Sex-Linked Dwarf Chicken. Front Genet 2021; 11:610605. [PMID: 33519913 PMCID: PMC7841439 DOI: 10.3389/fgene.2020.610605] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/30/2020] [Indexed: 01/28/2023] Open
Abstract
Sex-linked dwarf (SLD) chicken, which is caused by a recessive mutation of the growth hormone receptor (GHR), has been widely used in the Chinese broiler industry. However, it has been found that the SLD chicken has more abdominal fat deposition than normal chicken. Excessive fat deposition not only reduced the carcass quality of the broilers but also reduced the immunity of broilers to diseases. To find out the key genes and the precise regulatory pathways that were involved in the GHR mutation-induced excessive fat deposition, we used high-fat diet (HFD) and normal diet to feed the SLD chicken and normal chicken and analyzed the differentially expressed genes (DEGs) among the four groups. Results showed that the SLD chicken had more abdominal fat deposition and larger adipocytes size than normal chicken and HFD can promote abdominal fat deposition and induce adipocyte hypertrophy. RNA sequencing results of the livers and abdominal fats from the above chickens revealed that many DEGs between the SLD and normal chickens were enriched in fat metabolic pathways, such as peroxisome proliferator-activated receptor (PPAR) signaling, extracellular matrix (ECM)-receptor pathway, and fatty acid metabolism. Importantly, by constructing and analyzing the GHR-downstream regulatory network, we found that suppressor of cytokine signaling 2 (SOCS2) and cytokine-inducible SH2-containing protein (CISH) may involve in the GHR mutation-induced abdominal fat deposition in chicken. The ectopic expression of SOCS2 and CISH in liver-related cell line leghorn strain M chicken hepatoma (LMH) cell and immortalized chicken preadipocytes (ICP) revealed that these two genes can regulate fatty acid metabolism, adipocyte differentiation, and lipid droplet accumulation. Notably, overexpression of SOCS2 and CISH can rescue the hyperactive lipid metabolism and excessive lipid droplet accumulation of primary liver cell and preadipocytes that were isolated from the SLD chicken. This study found some genes and pathways involved in abdominal fat deposition of the SLD chicken and reveals that SOCS2 and CISH are two key genes involved in the GHR mutation-induced excessive fat deposition of the SLD chicken.
Collapse
Affiliation(s)
- Genghua Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Jiahui Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Jingwen Wu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Xueyi Ren
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Limin Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Shiyi Lu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Tian Cheng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Liangtian Tan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Manqing Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Qingbin Luo
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Shaodong Liang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Qinghua Nie
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Xiquan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| | - Wen Luo
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, South China Agricultural University, Guangzhou, China
| |
Collapse
|
24
|
Beutner F, Ritter C, Scholz M, Teren A, Holdt LM, Teupser D, Becker S, Thiele H, Gielen S, Thiery J, Ceglarek U. A metabolomic approach to identify the link between sports activity and atheroprotection. Eur J Prev Cardiol 2020; 29:436-444. [PMID: 33624084 DOI: 10.1093/eurjpc/zwaa122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/15/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
AIMS Physical activity (PA) is a mainstay of cardiovascular prevention. This study aimed to identify metabolic mediators of PA that protect against the development of atherosclerosis. METHODS AND RESULTS A total of 2160 participants in the LIFE heart study were analysed with data on PA and vascular phenotyping. In a targeted metabolomic approach, 61 metabolites (amino acids and acylcarnitines) were measured using liquid chromatography-tandem mass spectrometry. We investigated the interactions between PA, metabolites and markers of atherosclerosis in order to uncover possible mediation effects. Intended sports activity, but no daily PA, was associated with a lower degree of atherosclerosis, odds ratio (OR) for total atherosclerotic burden of 0.76 (95% confidence interval 0.62-0.94), carotid artery plaque OR 0.79 (0.66-0.96), and peripheral artery disease OR 0.74 (0.56-0.98). Twelve amino acids, free carnitine, five acylcarnitines were associated with sports activity. Of these, eight metabolites were also associated with the degree of atherosclerosis. In the mediation analyses, a cluster of amino acids (arginine, glutamine, pipecolic acid, taurine) were considered as possible mediators of atheroprotection. In contrast, a group of members of the carnitine metabolism (free carnitine, acetyl carnitine, octadecenoyl carnitine) were associated with inactivity and higher atherosclerotic burden. CONCLUSION Our metabolomic approach, which is integrated into a mediation model, provides transformative insights into the complex metabolic processes involved in atheroprotection. Metabolites with antioxidant and endothelial active properties are believed to be possible mediators of atheroprotection. The metabolomic mediation approach can support the understanding of complex diseases in order to identify targets for prevention and therapy.
Collapse
Affiliation(s)
- Frank Beutner
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany.,Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, 04289 Leipzig, Germany
| | - Christian Ritter
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany
| | - Markus Scholz
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany.,Institute of Medical Informatics, Statistics and Epidemiology, University Leipzig, Leipzig, Germany
| | - Andrej Teren
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany.,Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, 04289 Leipzig, Germany
| | - Lesca Miriam Holdt
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany.,Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Daniel Teupser
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany.,Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Susen Becker
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany.,Institute of Laboratory Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, 04289 Leipzig, Germany
| | - Stephan Gielen
- Department of Cardiology, Angiology and Intensive Care, Klinikum Lippe, Detmold, Germany
| | - Joachim Thiery
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany.,Institute of Laboratory Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- LIFE Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany.,Institute of Laboratory Medicine, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
25
|
Herring Milt and Herring Milt Protein Hydrolysate Are Equally Effective in Improving Insulin Sensitivity and Pancreatic Beta-Cell Function in Diet-Induced Obese- and Insulin-Resistant Mice. Mar Drugs 2020; 18:md18120635. [PMID: 33322303 PMCID: PMC7763884 DOI: 10.3390/md18120635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
Although genetic predisposition influences the onset and progression of insulin resistance and diabetes, dietary nutrients are critical. In general, protein is beneficial relative to carbohydrate and fat but dependent on protein source. Our recent study demonstrated that 70% replacement of dietary casein protein with the equivalent quantity of protein derived from herring milt protein hydrolysate (HMPH; herring milt with proteins being enzymatically hydrolyzed) significantly improved insulin resistance and glucose homeostasis in high-fat diet-induced obese mice. As production of protein hydrolysate increases the cost of the product, it is important to determine whether a simply dried and ground herring milt product possesses similar benefits. Therefore, the current study was conducted to investigate the effect of herring milt dry powder (HMDP) on glucose control and the associated metabolic phenotypes and further to compare its efficacy with HMPH. Male C57BL/6J mice on a high-fat diet for 7 weeks were randomized based on body weight and blood glucose into three groups. One group continued on the high-fat diet and was used as the insulin-resistant/diabetic control and the other two groups were given the high-fat diet modified to have 70% of casein protein being replaced with the same amount of protein from HMDP or HMPH. A group of mice on a low-fat diet all the time was used as the normal control. The results demonstrated that mice on the high-fat diet increased weight gain and showed higher blood concentrations of glucose, insulin, and leptin, as well as impaired glucose tolerance and pancreatic β-cell function relative to those on the normal control diet. In comparison with the high-fat diet, the replacement of 70% dietary casein protein with the same amount of HMDP or HMPH protein decreased weight gain and significantly improved the aforementioned biomarkers, insulin sensitivity or resistance, and β-cell function. The HMDP and HMPH showed similar effects on every parameter except blood lipids where HMDP decreased total cholesterol and non-HDL-cholesterol levels while the effect of HMPH was not significant. The results demonstrate that substituting 70% of dietary casein protein with the equivalent amount of HMDP or HMPH protein protects against obesity and diabetes, and HMDP is also beneficial to cholesterol homeostasis.
Collapse
|
26
|
The effects of taurine supplementation on obesity, blood pressure and lipid profile: A meta-analysis of randomized controlled trials. Eur J Pharmacol 2020; 885:173533. [DOI: 10.1016/j.ejphar.2020.173533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022]
|
27
|
Tan W, Zhao K, Xiang J, Zhou X, Cao F, Song W, Liu Q, Zhang X, Li X, Tan Z. Pyrazinamide alleviates rifampin-induced steatohepatitis in mice by regulating the activities of cholesterol-activated 7α-hydroxylase and lipoprotein lipase. Eur J Pharm Sci 2020; 151:105402. [DOI: 10.1016/j.ejps.2020.105402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/06/2020] [Accepted: 05/27/2020] [Indexed: 11/28/2022]
|
28
|
Dietary taurine stimulates the hepatic biosynthesis of both bile acids and cholesterol in the marine teleost, tiger puffer ( Takifugu rubripes). Br J Nutr 2020; 123:1345-1356. [PMID: 31959268 DOI: 10.1017/s0007114520000161] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Taurine (TAU) plays important roles in the metabolism of bile acids, cholesterol and lipids. However, little relevant information has been available in fish where TAU has been identified as a conditionally essential nutrient. The present study aimed to investigate the effects of dietary TAU on the metabolism of bile acids, cholesterol and lipids in tiger puffer, which is both an important aquaculture species and a good research model, having a unique lipid storage pattern. An 8-week feeding trial was conducted in a flow-through seawater system. Three experimental diets differed only in TAU level, that is, 1·7, 8·2 and 14·0 mg/kg. TAU supplementation increased the total bile acid content in liver but decreased the content in serum. TAU supplementation also increased the contents of total cholesterol and HDL-cholesterol in both liver and serum. The hepatic bile acid profile mainly includes taurocholic acid (94·48 %), taurochenodeoxycholic acid (4·17 %) and taurodeoxycholic acid (1·35 %), and the contents of all these conjugated bile acids were increased by dietary TAU. The hepatic lipidomics analysis showed that TAU tended to decrease the abundance of individual phospholipids and increase those of some individual TAG and ceramides. The hepatic mRNA expression study showed that TAU stimulated the biosynthesis of both bile acids and cholesterol, possibly via regulation of farnesoid X receptor and HDL metabolism. TAU also stimulated the hepatic expression of lipogenic genes. In conclusion, dietary TAU stimulated the hepatic biosynthesis of both bile acids and cholesterol and tended to regulate lipid metabolism in multiple ways.
Collapse
|
29
|
Maleki V, Alizadeh M, Esmaeili F, Mahdavi R. The effects of taurine supplementation on glycemic control and serum lipid profile in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled trial. Amino Acids 2020; 52:905-914. [PMID: 32472292 DOI: 10.1007/s00726-020-02859-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Previous studies have suggested that taurine has hypoglycemic and hypolipidemic effects on experimental diabetic models. Therefore, this clinical trial was designed to explore the impacts of taurine supplementation on glycemic control and lipid profile in the patients with T2DM. This study was conducted on 45 patients with T2DM in Tabriz Sheikhor-raees Polyclinic and Imam-Reza Hospital Endocrine Center. Subjects were randomly divided into taurine and placebo groups. Accordingly, the taurine group (n = 23) received taurine 3000 mg/daily and the placebo group (n = 22) took crystalline microcellulose/daily for the duration of 8 weeks. At baseline and after the trial completion, fasting blood samples were obtained from the patients to assess the glycemic indicators and lipid profile. Independent t test, paired t test, Pearson's correlation, and analysis of covariance was used for analysis. At the end of the study, levels of FBS (p = 0.01), insulin (p = 0.01), HOMA-IR (p = 0.003), TC (p = 0.013), and LDL-C (p = 0.041) significantly decreased in the taurine group compared to the placebo group. In addition, there was no significant changes in HbA1c, triglyceride, HDL-C, anthropometric indicators or dietary intakes by passing 8 weeks from the intervention. In conclusion, the findings of the current study indicated that taurine supplementation (3000 mg/day) for 8 weeks could improve the glycemic indexes and lipid profiles including TC and LDL-C in the patients with T2DM.
Collapse
Affiliation(s)
- Vahid Maleki
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Alizadeh
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Esmaeili
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mahdavi
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Velazquez-Torres G, Fuentes-Mattei E, Choi HH, Yeung SCJ, Meng X, Lee MH. Diabetes mellitus type 2 drives metabolic reprogramming to promote pancreatic cancer growth. Gastroenterol Rep (Oxf) 2020; 8:261-276. [PMID: 32843973 PMCID: PMC7434590 DOI: 10.1093/gastro/goaa018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/17/2020] [Accepted: 03/01/2020] [Indexed: 02/07/2023] Open
Abstract
Background Diabetes mellitus type 2 (DM2) is a modifiable risk factor associated with pancreatic carcinogenesis and tumor progression on the basis of epidemiology studies, but the biological mechanisms are not completely understood. The purpose of this study is to demonstrate direct evidence for the mechanisms mediating these epidemiologic phenomena. Our hypothesis is that DM2 accelerates pancreatic cancer growth and that metformin treatment has a beneficial impact. Methods To determine the effect of glucose and insulin in pancreatic cancer proliferation, we used conditioned media to mimic DM2 conditions. Also, we studied the effect of anti-diabetic drugs, particularly metformin and rosiglitazone on pancreatic cancer growth. We established orthotopic/syngeneic (Leprdb/db) mouse cancer models to evaluate the effect of diabetes on pancreatic tumor growth and aggressiveness. Results Our results showed that diabetes promotes pancreatic tumor growth. Furthermore, enhanced tumor growth and aggressiveness (e.g. epithelial–mesenchymal transition) can be explained by functional transcriptomic and metabolomic changes in the mice with diabetes, namely via activation of the AKT/mTOR pathway. Metformin treatment suppressed the diabetes-induced AKT/mTOR pathway activation and tumor growth. The metabolic profile determined by mass spectrum showed important changes of metabolites in the pancreatic cancer derived from diabetic mice treated with metformin. Conclusions Diabetes mellitus type 2 has critical effects that promote pancreatic cancer progression via transcriptomic and metabolomic changes. Our animal models provide strong evidence for the causal relationship between diabetes and accelerated pancreatic cancers. This study sheds a new insight into the effects of metformin and its potential as part of therapeutic interventions for pancreatic cancer in diabetic patients.
Collapse
Affiliation(s)
- Guermarie Velazquez-Torres
- Department of Molecular and Cellular Oncology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enrique Fuentes-Mattei
- Department of Molecular and Cellular Oncology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hyun Ho Choi
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, Sun Yat-sen University, Guangzhou, P. R. China.,Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Sai-Ching J Yeung
- Department of Emergency Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiangqi Meng
- Guangdong Provincial Key laboratory of Colorectal and Pelvic Floor Disease, Sun Yat-sen University, Guangzhou, P. R. China
| | - Mong-Hong Lee
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
31
|
Effect of Saffron Extract and Crocin in Serum Metabolites of Induced Obesity Rats. BIOMED RESEARCH INTERNATIONAL 2020. [DOI: 10.1155/2020/1247946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The effect of saffron extract (Crocus sativus L.) and its primary compound crocin was studied on an induced obesity rat model. Our study is aimed at investigating and comparing the metabolite changes in obese and obese treated with saffron extract and crocin and at improving the understanding of the therapeutic effect of saffron extract and crocin. Two different doses of saffron extracts and crocin (40 and 80 mg/kg) were incorporated in a high-fat diet (HFD) and were given for eight weeks to the obese rats. The changes in metabolite profiles of the serum were determined using proton nuclear magnetic resonance (1H-NMR). Pattern recognition by multivariate data analysis (MVDA) showed that saffron extract and crocin at 80 mg/kg was the best dosage compared to 40 mg/kg. It also showed that both treatments work in different pathways, especially concerning glucose, lipid, and creatinine metabolism. In conclusion, although the pure compound, crocin, is superior to the saffron crude extract, this finding suggested that the saffron extract can be considered as an alternative aside from crocin in treating obesity.
Collapse
|
32
|
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: 2.5] [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
|
33
|
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: 0.8] [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
|
34
|
Miyazaki T, Sasaki SI, Toyoda A, Shirai M, Ikegami T, Matsuzaki Y, Honda A. Influences of Taurine Deficiency on Bile Acids of the Bile in the Cat Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1155:35-44. [PMID: 31468384 DOI: 10.1007/978-981-13-8023-5_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Taurine content in the body is maintained by both biosynthesis from sulfur-contained amino acids in the liver and ingestion from usual foods, mainly seafoods and meat. Contrary to the rodents, the maintenance of taurine content in the body depends on the oral taurine ingestion in cats as well as humans because of the low ability of the biosynthesis. Therefore, insufficient of dietary taurine intake increases the risks of various diseases such as blind and expanded cardiomyopathy in the cats. One of the most established physiological roles of taurine is the conjugation with bile acid in the liver. In addition, taurine has effect to increase the expression and activity of bile acid synthesis rate-limiting enzyme CYP7A1. Present study purposed to evaluate the influence of taurine deficiency on bile acids in the cats fed taurine-lacking diet. Adult cats were fed the soybean protein-based diet with 0.15% taurine or without taurine for 30 weeks. Taurine concentration in serum and liver was undetectable, and bile acids in the bile were significantly decreased in the taurine-deficient cats. Taurine-conjugated bile acids in the bile were significantly decreased, and instead, unconjugated bile acids were significantly increased in the taurine-deficient cats. Present results suggested that the taurine may play an important role in the synthesis of bile acids in the liver.
Collapse
Affiliation(s)
- Teruo Miyazaki
- Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan.
| | - Sei-Ich Sasaki
- Ibaraki Prefectural University of Health Sciences, Ibaraki, Japan.,Toyo Public Health College, Tokyo, Japan
| | - Atsushi Toyoda
- College of Agriculture, Ibaraki University, Ibaraki, Japan
| | - Mutsumi Shirai
- Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Tadashi Ikegami
- Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | | | - Akira Honda
- Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| |
Collapse
|
35
|
Wen C, Li F, Zhang L, Duan Y, Guo Q, Wang W, He S, Li J, Yin Y. Taurine is Involved in Energy Metabolism in Muscles, Adipose Tissue, and the Liver. Mol Nutr Food Res 2018; 63:e1800536. [DOI: 10.1002/mnfr.201800536] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/13/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Chaoyue Wen
- Laboratory of Animal Nutrition and Human HealthHunan international joint laboratory of Animal Intestinal Ecology and HealthCollege of Life ScienceHunan Normal University Changsha Hunan 410081 China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
- Hunan Co‐Innovation Center of Animal Production SafetyCICAPSHunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients Changsha 410128 China
| | - Lingyu Zhang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
- University of Chinese Academy of Sciences Beijing 100039 China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
- University of Chinese Academy of Sciences Beijing 100039 China
| | - Wenlong Wang
- Laboratory of Animal Nutrition and Human HealthHunan international joint laboratory of Animal Intestinal Ecology and HealthCollege of Life ScienceHunan Normal University Changsha Hunan 410081 China
| | - Shanping He
- Laboratory of Animal Nutrition and Human HealthHunan international joint laboratory of Animal Intestinal Ecology and HealthCollege of Life ScienceHunan Normal University Changsha Hunan 410081 China
| | - Jianzhong Li
- Laboratory of Animal Nutrition and Human HealthHunan international joint laboratory of Animal Intestinal Ecology and HealthCollege of Life ScienceHunan Normal University Changsha Hunan 410081 China
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
- Hunan Co‐Innovation Center of Animal Production SafetyCICAPSHunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients Changsha 410128 China
| |
Collapse
|
36
|
Kim KS, Bang E. Metabolomics Profiling of the Effects of Taurine Supplementation on Dyslipidemia in a High-Fat-Diet-Induced Rat Model by 1H NMR Spectroscopy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:329-336. [PMID: 28849467 DOI: 10.1007/978-94-024-1079-2_29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Metabolomics, the comprehensive study of metabolites, has merged as a potent tool for analyzing complex phenotypes and identifying biomarkers of specific physiological responses and has the potential to lead to innovative therapeutic and diagnostic schemes for many diseases. In a former report, we showed that taurine supplementation considerably ameliorated dyslipidemia in rats fed a high-caloric diet. In this work, we examined the metabolic changes that occur in rat serum after they were fed a normal diet, a high-fat diet, and a high-fat diet containing 2% taurine (tau) by NMR spectroscopy combined with a multivariate statistical analysis containing PCA, PLS-DA, and OPLS-DA. We obtained 1H-NMR spectra of rat serum and used pattern recognition to identify key metabolites related to taurine supplementation. We found significant changes in creatine, methionine, glutamine, and threonine as well as in lipids, all of which decreased in the Tau group. To use these changes in metabolites as novel therapeutic and diagnostic markers, it should first be investigated whether these results are reproducible in future experiments. Next, researchers should determine how these changes affect serum lipid changes. This study identified some changes in serum metabolites and demonstrated the possibility of using an NMR-based metabolomics method to explore the effects of a taurine supplement on dyslipidemia in a high-fat-diet-induced rat model.
Collapse
Affiliation(s)
- Kyoung Soo Kim
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, South Korea.
- East-West Bone and Joint Disease Research Institute, Kyung Hee University Hospital at Gangdong, 149 Sangil-dong, Gangdong-gu, Seoul, South Korea.
| | - Eunjung Bang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, South Korea.
| |
Collapse
|
37
|
Choi WS, Lee JS. The Effect of Taurine Intake among Korean College Students: Serum Biochemistry and Blood Hematology. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2018. [DOI: 10.15324/kjcls.2018.50.3.236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Woo-Soon Choi
- Department of Clinical Laboratory Science, Songho University, Hoengseong, Korea
| | - Jae-Sik Lee
- Department of Clinical Laboratory Science, Hyejeon College, Hongseong, Korea
| |
Collapse
|
38
|
Vikøren LA, Drotningsvik A, Bergseth MT, Mjøs SA, Austgulen MH, Mellgren G, Gudbrandsen OA. Intake of Baked Cod Fillet Resulted in Lower Serum Cholesterol and Higher Long Chain n-3 PUFA Concentrations in Serum and Tissues in Hypercholesterolemic Obese Zucker fa/fa Rats. Nutrients 2018; 10:E840. [PMID: 29958397 PMCID: PMC6073601 DOI: 10.3390/nu10070840] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/15/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence indicates that lean fish consumption may benefit cardiovascular health. High cholesterol and low n-3 PUFA concentrations in serum are associated with an increased risk of coronary heart disease; therefore, it is of interest to investigate effects of cod intake on cholesterol and n-3 PUFAs in serum and tissues. Hypercholesterolemic obese Zucker fa/fa rats were fed diets containing 25% protein from baked cod fillet and 75% protein from casein (Baked Cod Diet), or casein as the sole protein source (Control Diet) for four weeks. Consuming Baked Cod Diet resulted in lower serum cholesterol and lower hepatic mRNA concentrations of HMG-CoA reductase and sterol O-acyltransferase-2 without affecting serum bile acid concentration, faecal excretion of cholesterol and bile acid, and hepatic concentrations of bile acids, cholesterol and cholesterol 7 alpha-hydroxylase mRNA when compared to Control Diet. Rats fed Baked Cod Diet had higher concentrations of n-3 PUFAs in serum, liver, skeletal muscle and adipose tissue. To conclude, baked cod fillet intake resulted in lower serum cholesterol, which was probably caused by lower endogenous cholesterol synthesis, and higher n-3 PUFA in serum and tissues in obese Zucker fa/fa rats. These findings support the evidence that lean fish consumption might benefit cardiovascular health.
Collapse
Affiliation(s)
- Linn A Vikøren
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
| | - Aslaug Drotningsvik
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
| | - Marthe T Bergseth
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
| | - Svein A Mjøs
- Department of Chemistry, University of Bergen, P.O. Box 7803, 5020 Bergen, Norway.
- Nofima BioLab, P.O. Box 1425 Oasen, 5828 Bergen, Norway.
| | - Maren H Austgulen
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
| | - Gunnar Mellgren
- Department of Clinical Science, KG Jebsen Center for Diabetes Research, University of Bergen, Haukeland University Hospital, 5021 Bergen, Norway.
- Hormone Laboratory, Haukeland University Hospital, 5021 Bergen, Norway.
| | - Oddrun A Gudbrandsen
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
| |
Collapse
|
39
|
Drotningsvik A, Vikøren LA, Mjøs SA, Oterhals Å, Pampanin D, Flesland O, Gudbrandsen OA. Water-Soluble Fish Protein Intake Led to Lower Serum and Liver Cholesterol Concentrations in Obese Zucker fa/fa Rats. Mar Drugs 2018; 16:md16050149. [PMID: 29724010 PMCID: PMC5983280 DOI: 10.3390/md16050149] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/16/2018] [Accepted: 04/26/2018] [Indexed: 12/25/2022] Open
Abstract
Proteins from different fish species and different raw materials such as fish fillets and by-products have shown promising cardioprotective effects in rodents and humans, including effects on cholesterol metabolism. Blue whiting is used mainly to produce fish meal for the feed industry and during this production, a water-soluble protein fraction, containing small peptides that are easily absorbed and may hold bioactive properties, is isolated. The effects of water-soluble fish protein on cholesterol metabolism were investigated in twelve male obese Zucker fa/fa rats. Rats were fed diets with water-soluble protein from blue whiting (BWW) as 1/3 of the total protein and the remaining 2/3 as casein (BWW group) or with casein as the sole protein source (control group). After 5 weeks intervention, the BWW group had lower serum total, high-density lipoprotein (HDL), and low-density lipoprotein (LDL) cholesterol concentrations and lower cholesteryl ester concentration compared to controls. Hepatic concentrations of cholesterol, 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, and LDL receptors were also lower in the BWW group. The groups had a similar concentration of serum total bile acids and similar fecal excretions of cholesterol and bile acids. To conclude, the BWW diet led to lower concentrations of serum and liver cholesterol in obese Zucker fa/fa rats, probably due to lower hepatic cholesterol synthesis.
Collapse
Affiliation(s)
- Aslaug Drotningsvik
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
- Vedde AS, TripleNine Group, 6030 Langevåg, Norway.
| | - Linn Anja Vikøren
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
| | - Svein Are Mjøs
- Department of Chemistry, University of Bergen, P.O. Box 7803, 5020 Bergen, Norway.
- Nofima AS, P.O. Box 1425 Oasen, 5828 Bergen, Norway.
| | - Åge Oterhals
- Nofima AS, P.O. Box 1425 Oasen, 5828 Bergen, Norway.
| | - Daniela Pampanin
- International Research Institute of Stavanger, Mekjarvik 12, 4070 Randaberg, Norway.
| | - Ola Flesland
- Vedde AS, TripleNine Group, 6030 Langevåg, Norway.
| | - Oddrun Anita Gudbrandsen
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
| |
Collapse
|
40
|
Munteanu C, Rosioru C, Tarba C, Lang C. Long-term consumption of energy drinks induces biochemical and ultrastructural alterations in the heart muscle. Anatol J Cardiol 2018; 19:326-323. [PMID: 29724975 PMCID: PMC6280269 DOI: 10.14744/anatoljcardiol.2018.90094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Energy drinks (EDs) target young and active individuals and they are being marketed as enhancers of energy, concentration, and physical and cognitive performance. Their long-term consumption raises serious health concerns related to cardiovascular events. Here we investigate the effects of long-term Red Bull® consumption and its combination with alcohol on certain biochemical parameters and the ultrastructure of the myocardium. METHODS Male Wistar rats were categorized into four groups and given different treatments via oral administration. The Control (C) group received tap water, the Red Bull (RB) group received 1.5 ml/100 g body weight of Red Bull, the ethanol group (E) received 0.486 mg/100 g body weight of ethanol, and the Red Bull and ethanol (RBE) received a combination of the two beverages for 30 days. In the last 6 days of the experiment, the animals were tested for their physical performance by conducting a weight-loaded forced swim test. Immediately after swimming exhaustion, the animals were sacrificed under anesthesia and samples of the heart muscle were harvested for ultrastructural and biochemical analyses. RESULTS Our results showed a significant increase in the heart glucose and glycogen concentrations in the RB and RBE groups. Total cholesterol concentration significantly decreased in the RBE and RB groups. Total protein concentration and ALT and AST activities increased in all groups. The biochemical changes were accompanied by ultrastructural alterations. CONCLUSION Based on these results, we recommend that athletes and active persons should avoid the long-term consumption of the Red Bull ED and, particularly, its combination with alcohol.
Collapse
Affiliation(s)
| | | | | | - Camelia Lang
- Department of Molecular Biology and Biotechnology, Babes-Bolyai University; Cluj-Napoca-Romania.
| |
Collapse
|
41
|
Dong Z, Sinha R, Richie JP. Disease prevention and delayed aging by dietary sulfur amino acid restriction: translational implications. Ann N Y Acad Sci 2018; 1418:44-55. [PMID: 29399808 DOI: 10.1111/nyas.13584] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023]
Abstract
Sulfur amino acids (SAAs) play numerous critical roles in metabolism and overall health maintenance. Preclinical studies have demonstrated that SAA-restricted diets have many beneficial effects, including extending life span and preventing the development of a variety of diseases. Dietary sulfur amino acid restriction (SAAR) is characterized by chronic restrictions of methionine and cysteine but not calories and is associated with reductions in body weight, adiposity and oxidative stress, and metabolic changes in adipose tissue and liver resulting in enhanced insulin sensitivity and energy expenditure. SAAR-induced changes in blood biomarkers include reductions in insulin, insulin-like growth factor-1, glucose, and leptin and increases in adiponectin and fibroblast growth factor 21. On the basis of these preclinical data, SAAR may also have similar benefits in humans. While little is known of the translational significance of SAAR, its potential feasibility in humans is supported by findings of its effectiveness in rodents, even when initiated in adult animals. To date, there have been no controlled feeding studies of SAAR in humans; however, there have been numerous relevant epidemiologic and disease-based clinical investigations reported. Here, we summarize observations from these clinical investigations to provide insight into the potential effectiveness of SAAR for humans.
Collapse
Affiliation(s)
- Zhen Dong
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Raghu Sinha
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - John P Richie
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
42
|
Taurine does not affect the composition, diversity, or metabolism of human colonic microbiota simulated in a single-batch fermentation system. PLoS One 2017; 12:e0180991. [PMID: 28700670 PMCID: PMC5507302 DOI: 10.1371/journal.pone.0180991] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 06/23/2017] [Indexed: 01/12/2023] Open
Abstract
Accumulating evidence suggests that dietary taurine (2-aminoethanesulfonic acid) exerts beneficial anti-inflammatory effects in the large intestine. In this study, we investigated the possible impact of taurine on human colonic microbiota using our single-batch fermentation system (Kobe University Human Intestinal Microbiota Model; KUHIMM). Fecal samples from eight humans were individually cultivated with and without taurine in the KUHIMM. The results showed that taurine remained largely undegraded after 30 h of culturing in the absence of oxygen, although some 83% of the taurine was degraded after 30 h of culturing under aerobic conditions. Diversity in bacterial species in the cultures was analyzed by 16S rRNA gene sequencing, revealing that taurine caused no significant change in the diversity of the microbiota; both operational taxonomic unit and Shannon-Wiener index of the cultures were comparable to those of the respective source fecal samples. In addition, principal coordinate analysis indicated that taurine did not alter the composition of bacterial species, since the 16S rRNA gene profile of bacterial species in the original fecal sample was maintained in each of the cultures with and without taurine. Furthermore, metabolomic analysis revealed that taurine did not affect the composition of short-chain fatty acids produced in the cultures. These results, under these controlled but artificial conditions, suggested that the beneficial anti-inflammatory effects of dietary taurine in the large intestine are independent of the intestinal microbiota. We infer that dietary taurine may act directly in the large intestine to exert anti-inflammatory effects.
Collapse
|
43
|
Vikøren LA, Drotningsvik A, Bergseth MT, Mjøs SA, Mola N, Leh S, Mellgren G, Gudbrandsen OA. Effects of baked and raw salmon fillet on lipids and n-3 PUFAs in serum and tissues in Zucker fa/fa rats. Food Nutr Res 2017; 61:1333395. [PMID: 28659746 PMCID: PMC5475302 DOI: 10.1080/16546628.2017.1333395] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/18/2017] [Indexed: 01/13/2023] Open
Abstract
Knowledge of the health impact of consuming heat-treated versus raw fish fillet is limited. To investigate effects of baked or raw salmon fillet intake on lipids and n-3 PUFAs in serum and tissues, obese Zucker fa/fa rats were fed diets containing 25% of protein from baked or raw salmon fillet and 75% of protein from casein, or casein as the sole protein source (control group) for four weeks. Salmon diets had similar composition of amino and fatty acids. Growth and energy intake were similar in all groups. Amounts of lipids and n-3 PUFAs in serum, liver and skeletal muscle were similar between rats fed baked or raw salmon fillet. When compared to the control group, rats fed baked salmon had lower serum total and LDL cholesterol and higher serum triacylglycerol levels. Both raw and baked salmon groups had lower HDL cholesterol level when compared to control rats. In conclusion, baking as a preparation method does not alter protein and fat qualities of salmon fillets, and intake of baked and raw salmon fillets gave similar effects on lipids and n-3 PUFAs in serum and tissues from rats.
Collapse
Affiliation(s)
- Linn A Vikøren
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Clinical Science, University of Bergen, Haukeland University Hospital, Bergen, Norway
| | - Aslaug Drotningsvik
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Marthe T Bergseth
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Svein A Mjøs
- Department of Chemistry, University of Bergen, Bergen, Norway.,Nofima BioLab, Bergen, Norway
| | - Nazanin Mola
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Sabine Leh
- Department of Pathology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Department of Clinical Science, KG Jebsen Center for Diabetes Research, University of Bergen, Haukeland University Hospital, 5021Bergen, Norway.,Hormone Laboratory, Haukeland University Hospital, 5021Bergen, Norway
| | - Oddrun A Gudbrandsen
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
44
|
Zhu RG, Sun YD, Hou YT, Fan JG, Chen G, Li TP. Pectin penta-oligogalacturonide reduces cholesterol accumulation by promoting bile acid biosynthesis and excretion in high-cholesterol-fed mice. Chem Biol Interact 2017; 272:153-159. [PMID: 28549616 DOI: 10.1016/j.cbi.2017.05.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/15/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022]
Abstract
Haw pectin penta-oligogalacturonide (HPPS) has important role in improving cholesterol metabolism and promoting the conversion of cholesterol to bile acids (BA) in mice fed high-cholesterol diet (HCD). However, the mechanism is not clear. This study aims to investigate the effects of HPPS on cholesterol accumulation and the regulation of hepatic BA synthesis and transport in HCD-fed mice. Results showed that HPPS significantly decreased plasma and hepatic TC levels but increased plasma high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I (apoA-I) levels, compared to HCD. BA analysis showed that HPPS markedly decreased hepatic and small intestine BA levels but increased the gallbladder BA levels, and finally decreased the total BA pool size, compared to HCD. Studies of molecular mechanism revealed that HPPS promoted hepatic ATP-binding cassette transporter A1 (ABCA1), ATP-binding cassette transporter G1 (ABCG1), and scavenger receptor BI (SR-BI) expression but did not affect ATB binding cassette transporter G5/G8 (ABCG5/8) expression. HPPS inactivated hepatic farnesoid X receptor (FXR) and target genes expression, which resulted in significant increase of cholesterol 7α-hydroxylase 1 (CYP7A1) and sterol 12α-hydroxylase (CYP8B1) expression, with up-regulations of 204.2% and 33.5% for mRNA levels, respectively, compared with HCD. In addition, HPPS markedly enhanced bile salt export pump (BSEP) expression but didn't affect the sodium/taurocholate co-transporting polypeptide (NTCP) expression. In conclusion, the study revealed that HPPS reduced cholesterol accumulation by promoting BA synthesis in the liver and excretion in the feces, and might promote macrophage-to-liver reverse cholesterol transport (RCT) but did not liver-to-fecal RCT.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1/genetics
- ATP Binding Cassette Transporter 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- Animals
- Apolipoprotein A-I/blood
- Bile Acids and Salts/metabolism
- Cholesterol/blood
- Cholesterol 7-alpha-Hydroxylase/genetics
- Cholesterol 7-alpha-Hydroxylase/metabolism
- Cholesterol, HDL/blood
- Diet, High-Fat
- Gene Expression/drug effects
- Intestine, Small/drug effects
- Intestine, Small/metabolism
- Liver/drug effects
- Liver/metabolism
- Male
- Mice
- Oligosaccharides/pharmacology
- Pectins/chemistry
- Pectins/pharmacology
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Steroid 12-alpha-Hydroxylase/genetics
- Steroid 12-alpha-Hydroxylase/metabolism
Collapse
Affiliation(s)
- Ru-Gang Zhu
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang 110036, China.
| | - Yan-Di Sun
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang 110036, China
| | - Yu-Ting Hou
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang 110036, China
| | - Jun-Gang Fan
- Forestry Biotechnology and Analysis Test Center, Liaoning Academy of Forestry Sciences, Shenyang 110032, China
| | - Gang Chen
- Forestry Biotechnology and Analysis Test Center, Liaoning Academy of Forestry Sciences, Shenyang 110032, China
| | - Tuo-Ping Li
- College of Food Science, Shenyang Agriculture University, Shenyang 110032, China.
| |
Collapse
|
45
|
Zhu R, Hou Y, Sun Y, Li T, Fan J, Chen G, Wei J. Pectin Penta-Oligogalacturonide Suppresses Intestinal Bile Acids Absorption and Downregulates the FXR-FGF15 Axis in High-Cholesterol Fed Mice. Lipids 2017; 52:489-498. [PMID: 28474246 DOI: 10.1007/s11745-017-4258-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
Haw pectin penta-oligogalacturonide (HPPS), purified from the hydrolysates of haw pectin, has important role in decreasing hepatic cholesterol accumulation and promoting bile acids (BA) excretion in the feces of mice fed a high-cholesterol diet (HCD). However, the mechanism is not clear. This study aims to investigate the effects of HPPS on BA reabsorption in ileum and biosynthesis in liver of mice. Results showed that HPPS increased fecal BA output by approximately 110%, but decreased ileal BA and the total BA pool size by approximately 47 and 36%, respectively, compared to HCD. Studies of molecular mechanism revealed that HPPS significantly decreased the mRNA and protein levels of farnesoid X receptor (FXR) in the small intestine of mice and inactivated the fibroblast growth factor 15 (FXR-FGF15) axis, which increased the mRNA and protein levels of CYP7A1 by approximately 204 and 104%, respectively, compared to HCD. Interestingly, the mRNA and protein levels of apical sodium-dependent bile acid transporter (ASBT) in the small intestine were approximately 128 and 73% higher in HPPS-fed mice than those in HCD-fed mice, respectively. However, no significant difference was detected for ASBT expression between HCD group and BA sequestrant cholestyramine group. These findings indicate that HPPS can suppress intestinal BA reabsorption and promoting hepatic BA biosynthesis. We speculated that HPPS could be ASBT competitive inhibitor rather than BA sequestrant in inhibiting BA reabsorption in ileum and improving cholesterol metabolism.
Collapse
Affiliation(s)
- Rugang Zhu
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China.
| | - Yuting Hou
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China
| | - Yandi Sun
- Department of Food Science, College of Light Industry, Liaoning University, Liaoning Engineering Research Center for Food Bioprocessing, Shenyang Key Laboratory of Food Bioprocessing and Quality Control, Shenyang, 110036, China
| | - Tuoping Li
- College of Food Science, Shenyang Agriculture University, Shenyang, 110032, China
| | - Jungang Fan
- Forestry Biotechnology and Analysis Test Center, Liaoning Academy of Forestry Sciences, Shenyang, 110032, China
| | - Gang Chen
- Forestry Biotechnology and Analysis Test Center, Liaoning Academy of Forestry Sciences, Shenyang, 110032, China
| | - Junxiu Wei
- Electrical Engineering and Automation, College of Light Industry, Liaoning University, Shenyang, 110032, China
| |
Collapse
|
46
|
Awwad HM, Geisel J, Obeid R. Determination of trimethylamine, trimethylamine N-oxide, and taurine in human plasma and urine by UHPLC–MS/MS technique. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1038:12-18. [DOI: 10.1016/j.jchromb.2016.10.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 10/08/2016] [Accepted: 10/14/2016] [Indexed: 01/25/2023]
|
47
|
Li M, Xue S, Tan S, Qin X, Gu M, Wang D, Zhang Y, Guo L, Huang F, Yao Y, Zhou Z, Fan S, Huang C. Crabapple fruit extracts lower hypercholesterolaemia in high-fat diet-induced obese mice. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.09.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
48
|
Taurine zinc solid dispersions enhance bile-incubated L02 cell viability and improve liver function by inhibiting ERK2 and JNK phosphorylation during cholestasis. Toxicology 2016; 366-367:10-9. [PMID: 27501764 DOI: 10.1016/j.tox.2016.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/20/2016] [Accepted: 07/25/2016] [Indexed: 12/31/2022]
Abstract
Dietary intakes of taurine and zinc are associated with decreased risk of liver disease. In this study, solid dispersions (SDs) of a taurine zinc complex on hepatic injury were examined in vitro using the immortalized human hepatocyte cell line L02 and in a rat model of bile duct ligation. Sham-operated and bile duct ligated Sprague-Dawley rats were treated with the vehicle alone or taurine zinc (40, 80, 160mg/kg) for 17days. Bile duct ligation significantly increased blood lipid levels, and promoted hepatocyte apoptosis, inflammation and compensatory biliary proliferation. In vitro, incubation with bile significantly reduced L02 cell viability; this effect was significantly attenuated by pretreatment with SP600125 (a JNK inhibitor) and enhanced when co-incubated with taurine zinc SDs. In vivo, administration of taurine zinc SDs decreased serum alanine aminotransferase and aspartate aminotransferase activities in a dose-dependent manner and attenuated the increases in serum total bilirubin, total cholesterol and low density lipoprotein cholesterol levels after bile duct ligation. Additionally, taurine zinc SDs downregulated the expression of interleukin-1β and inhibited the phosphorylation of Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase2 (ERK2) in the liver after bile duct ligation. Moreover, taurine zinc SDs had more potent blood lipid regulatory and anti-apoptotic effects than the physical mixture of taurine and zinc acetate. Therefore, we speculate that taurine zinc SDs protect liver function at least in part via a mechanism linked to reduce phosphorylation of JNK and ERK2, which suppresses inflammation, apoptosis and cholangiocyte proliferation during cholestasis.
Collapse
|
49
|
Jensen IJ, Walquist M, Liaset B, Elvevoll EO, Eilertsen KE. Dietary intake of cod and scallop reduces atherosclerotic burden in female apolipoprotein E-deficient mice fed a Western-type high fat diet for 13 weeks. Nutr Metab (Lond) 2016; 13:8. [PMID: 26839578 PMCID: PMC4735963 DOI: 10.1186/s12986-016-0068-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/21/2016] [Indexed: 01/25/2023] Open
Abstract
Background It is now increasingly recognized that the beneficial effects of seafood consumption is not limited to lipids and fatty acid, but that the protein part, i.e., peptides and amino acids, together with vitamins and even unknown bioactive constituents also are important for disease prevention. This study was designed to evaluate the putative anti-atherogenic effects of different protein sources (a lean seafood and a nonseafood) in apolipoprotein E-deficient (apoE−/−) mice. Methods Twenty-four 5-week-old female apoE−/− mice were fed Western type diets containing chicken or a combination of cod and scallops as dietary protein sources for 13 weeks. Atherosclerotic plaque burden, weight, serum levels of leptin, glucose and LDL cholesterol as well as gene expressions from liver and heart were evaluated. Statistical analyses were performed using SPSS. Differences between the variables were evaluated using independent t-test or Mann–Whitney U test for normally and non-normally distributed variables, respectively. Normality was defined by the Shapiro-Wilk test. Results The mice fed cod-scallop had a 24 % (p < 0.05) reduced total aorta atherosclerotic plaque burden compared to the chicken fed group, whereas the reduction in plaque in the less lesion prone thoracic and abdominal parts of the descending aorta were 46 % (p < 0.05) and 56 % (p < 0.05), respectively. In addition, mice fed cod-scallop gained less weight, and had lower serum levels of leptin, glucose and LDL cholesterol, compared to those fed chicken. Analysis of expression of the genes from liver and heart showed that hepatic endogenous antioxidant paraoxonase 2 (Pon2 gene) and the vascular cell adhesion molecule VCAM-1 (Vcam1 gene) were down regulated in mice fed cod-scallop compared to mice fed chicken. Conclusion The present study revealed a metabolic beneficial effect of lean seafood compared to chicken, as atherosclerotic plaque burden, serum glucose, leptin and LDL cholesterol levels were reduced in mice fed cod-scallop. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0068-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ida-Johanne Jensen
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UIT - The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Mari Walquist
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UIT - The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, 5004 Bergen, Norway
| | - Edel O Elvevoll
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UIT - The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Karl-Erik Eilertsen
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UIT - The Arctic University of Norway, N-9037 Tromsø, Norway
| |
Collapse
|
50
|
Chen W, Guo J, Zhang Y, Zhang J. The beneficial effects of taurine in preventing metabolic syndrome. Food Funct 2016; 7:1849-63. [DOI: 10.1039/c5fo01295c] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A review of the data fromin vitro, animal and limited human studies of the beneficial effects of taurine on obesity, dyslipidaemia, diabetes mellitus and hypertension, as well as the possible metabolic and molecular mechanisms for the prevention of metabolic syndrome by taurine.
Collapse
Affiliation(s)
- Wen Chen
- Beijing Key Laboratory of Bioactive Substances and Functional Foods
- Beijing Union University
- Beijing 100191
- P.R. China
| | - Junxia Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods
- Beijing Union University
- Beijing 100191
- P.R. China
| | - Yanzhen Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods
- Beijing Union University
- Beijing 100191
- P.R. China
| | - Jing Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods
- Beijing Union University
- Beijing 100191
- P.R. China
| |
Collapse
|