1
|
Shen Y, Fan J, Li C, Wu F, Wu X, Tao L, Yang Q, Shen X. Restorative mechanisms of Shugan Yiyang capsule on male infertility through 'pharmaco-metabo-net' tripartite correlation analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156706. [PMID: 40220430 DOI: 10.1016/j.phymed.2025.156706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Shugan Yiyang capsule (SGYY), a commonly used traditional Chinese medicine formulation, is primarily indicated for the treatment of erectile dysfunction, yet existing studies on the therapeutic effects on male infertility (MI) are insufficient and the specific mechanisms remain poorly understood. Given the close relationship between MI, sperm quality, and erectile function, this study aims to investigate the role of SGYY in the restoration of MI and explore the underlying mechanisms. METHODS The efficacy of SGYY is comprehensively evaluated through pharmacodynamic, metabolomic, and network pharmacology. Sperm parameters, reproductive hormones, sexual behavior, neural enzymes, oxidative stress markers, pro-inflammatory cytokines, and testicular histopathology are measured to reveal the restorative effects of MI. Furthermore, urine and serum metabolomics, along with network pharmacology and surface plasmon resonance, are employed to explore the molecular mechanisms and predict core targets. RESULTS SGYY significantly improved overall health parameters, including body weight, water intake, urine output, food consumption, and spontaneous activity. Specifically, SGYY prominently recovered sexual behavior, ameliorated sperm quality, increased mitochondrial membrane potential, normalized reproductive hormones, upregulated endothelial nitric oxide synthase, attenuated oxidative stress markers, and pro-inflammatory cytokines, and repaired testicular pathological damage. Metabolomic analysis identified 47 candidate biomarkers, among which SGYY significantly modulated 39 potential biomarkers, encompassing 8 main metabolic pathways such as histidine metabolism, cysteine and methionine metabolism, propanoate metabolism, and taurine and hypotaurine metabolism. Additionally, network pharmacology predicted 8 core targets, comprising HSP90AA1, ESR1, MAPK1, CASP3, IL6, TNF, BCL2, and MAPK8. CONCLUSION SGYY improves sperm quality and erectile function by regulating oxidative stress, energy metabolism, and neurological function, thereby exerting a restorative effect on MI, as evidenced by the modulation of 8 main metabolic pathways, 39 potential biomarkers, and 8 core targets. Pharmacodynamic provides foundational validation, metabolomic uncovers intrinsic metabolic changes, and network pharmacology predicts therapeutic targets, with findings from the 'Pharmaco-Metabo-Net' tripartite correlation analysis providing a solid theoretical strategy and scientific evidence to support the clinical application of SGYY in restoring MI.
Collapse
Affiliation(s)
- Ying Shen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China; National Engineering Research Center of Miao's Medicines, Guizhou Yibai Pharmaceutical Co., Ltd., Guiyang 550008, China
| | - Jian Fan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Chunmei Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Fuli Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Xiangli Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Ling Tao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China.
| | - Qingbo Yang
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China; National Engineering Research Center of Miao's Medicines, Guizhou Yibai Pharmaceutical Co., Ltd., Guiyang 550008, China.
| | - Xiangchun Shen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang 561113, China; High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China.
| |
Collapse
|
2
|
Morikura T, Sakaguchi K, Tanaka RI, Yoshida A, Takahashi H, Iwasaki K, Shimizu T. Conditioned serum-free culture medium accomplishes adhesion and proliferation of bovine myogenic cells on uncoated dishes. NPJ Sci Food 2024; 8:108. [PMID: 39715787 DOI: 10.1038/s41538-024-00355-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
To establish a sustainable cultured meat technology, a low-cost culture medium must be developed without expensive biological materials such as serum and coating substances. However, even adhering bovine myogenic cells to uncoated culture dishes in the serum-free medium is challenging. We found that serum-free culture medium conditioned by HepG2 and NIH/3T3 cells not only accomplished the cell adhesion on uncoated culture dishes (the serum-containing medium : the serum-free medium : the conditioned medium = 6722 ± 1500 : 2210 ± 319 : 5985 ± 1558 cells/cm2), but also induced proliferation comparable to that observed in a serum-containing medium (the serum-containing medium : the serum-free medium : the conditioned medium = 10,050 ± 2814 : 2200 ± 707 : 8998 ± 3890 cells/cm2). Interestingly, although the nutrient composition of the developed medium differed significantly from that of the serum-containing medium, it tended to coordinate the expression of cell adhesion, proliferation, and myogenic differentiation markers as serum-containing medium. Component analysis and validation experiments suggested that pyridoxamine, asparagine, and glutamic acid contributed to the acquisition of the culture function of the developed medium. Our study paves the way to realize a low-cost and sustainable cultured meat technology.
Collapse
Affiliation(s)
- Takashi Morikura
- Graduate School of Science and Technology, Keio University, Yokohama, Japan
| | - Katsuhisa Sakaguchi
- Department of Medical Engineering, Faculty of Science and Engineering, Tokyo City University, Tokyo, Japan.
| | - Ryu-Ichiro Tanaka
- Institute of Advanced Biomedical Engineering and Sciences, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Azumi Yoshida
- Institute of Advanced Biomedical Engineering and Sciences, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Sciences, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Kiyotaka Iwasaki
- Department of Modern Mechanical Engineering, Waseda University, Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Sciences, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
3
|
Chen W, Xiao L, Guo W, Li H, Chen R, Duan Z, Chen Q, Lei Q. Research progress of traditional Chinese medicine regulating intestinal flora in the treatment of hypertension. Front Pharmacol 2024; 15:1449972. [PMID: 39717555 PMCID: PMC11664361 DOI: 10.3389/fphar.2024.1449972] [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: 06/20/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Hypertension is a common disease; however, it is more prevalent in older adults, and its prevalence is increasing in younger populations. Numerous studies have revealed that hypertension and the composition and functionality of the intestinal flora are closely correlated. The balance of the intestinal flora, intestinal barrier integrity, and metabolite content of the intestinal flora play significant roles in the occurrence and progression of hypertension. Therefore, we performed a comprehensive review of Traditional Chinese medicine (TCM) for hypertension, focusing on the role of the intestinal flora to understand the mechanism by which TCM regulates hypertension through its effects on the intestinal flora. We analyzed the findings using the terms "traditional Chinese medicine," "hypertension," "high blood pressure," "blood pressure," "intestinal flora," "intestinal barrier function," "intestinal flora metabolites," and other keywords from the China National Knowledge Infrastructure, VIP Chinese Science and Technology, Wanfang Data, PubMed, and ScienceDirect databases. We found that TCM treats hypertension by regulating the balance of the intestinal microbiota, increasing the abundance of beneficial bacteria, reducing the abundance of harmful bacteria, improving intestinal barrier function, increasing compact proteins, reducing intestinal permeability, and regulating the content of intestinal flora metabolites. The use of TCM to treat hypertension by regulating the intestinal flora is a promising therapeutic strategy. However, most studies are limited by small sample sizes and there is a lack of large-scale randomized controlled trials. In the future, multi-center controlled clinical trials are needed to verify the efficacy and safety of TCM, optimize therapeutic protocols, and establish a foundation for the standardized and personalized application of TCM in hypertension management.
Collapse
Affiliation(s)
- Wenjun Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Longfei Xiao
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Wenlong Guo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Hailin Li
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Rong Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhongyu Duan
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qinghua Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qing Lei
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
4
|
Dicks LMT. Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation. Int J Mol Sci 2024; 25:10634. [PMID: 39408963 PMCID: PMC11476619 DOI: 10.3390/ijms251910634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease (CVD) may be inherited, as recently shown with the identification of single nucleotide polymorphisms (SNPs or "snips") on a 250 kb DNA fragment that encodes 92 proteins associated with CVD. CVD is also triggered by microbial dysbiosis, microbial metabolites, metabolic disorders, and inflammatory intestinal epithelial cells (IECs). The epithelial cellular adhesion molecule (Ep-CAM) and trefoil factor 3 (TFF3) peptide keeps the gut wall intact and healthy. Variations in Ep-CAM levels are directly linked to changes in the gut microbiome. Leptin, plasminogen activator inhibitor 1 (PAI1), and alpha-1 acid glycoprotein 1 (AGP1) are associated with obesity and may be used as biomarkers. Although contactin 1 (CNTN1) is also associated with obesity and adiposity, it regulates the bacterial metabolism of tryptophan (Trp) and thus appetite. A decrease in CNTN1 may serve as an early warning of CVD. Short-chain fatty acids (SCFAs) produced by gut microbiota inhibit pro-inflammatory cytokines and damage vascular integrity. Trimethylamine N-oxide (TMAO), produced by gut microbiota, activates inflammatory Nod-like receptors (NLRs) such as Nod-like receptor protein 3 (NLRP3), which increase platelet formation. Mutations in the elastin gene (ELN) cause supra valvular aortic stenosis (SVAS), defined as the thickening of the arterial wall. Many of the genes expressed by human cells are regulated by gut microbiota. The identification of new molecular markers is crucial for the prevention of CVD and the development of new therapeutic strategies. This review summarizes the causes of CVD and identifies possible CVD markers.
Collapse
Affiliation(s)
- Leon M T Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
5
|
Peris-Fernández M, Roca-Marugán MI, Amengual JL, Balaguer-Timor Á, Viejo-Boyano I, Soldevila-Orient A, Devesa-Such R, Sánchez-Pérez P, Hernández-Jaras J. Metabolic Pathways Affected in Patients Undergoing Hemodialysis and Their Relationship with Inflammation. Int J Mol Sci 2024; 25:9364. [PMID: 39273311 PMCID: PMC11394964 DOI: 10.3390/ijms25179364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Worldwide, 3.9 million individuals rely on kidney replacement therapy. They experience heightened susceptibility to cardiovascular diseases and mortality, alongside an increased risk of infections and malignancies, with inflammation being key to explaining this intensified risk. This study utilized semi-targeted metabolomics to explore novel metabolic pathways related to inflammation in this population. We collected pre- and post-session blood samples of patients who had already undergone one year of chronic hemodialysis and used liquid chromatography and high-resolution mass spectrometry to perform a metabolomic analysis. Afterwards, we employed both univariate (Mann-Whitney test) and multivariate (logistic regression with LASSO regularization) to identify metabolites associated with inflammation. In the univariate analysis, indole-3-acetaldehyde, 2-ketobutyric acid, and urocanic acid showed statistically significant decreases in median concentrations in the presence of inflammation. In the multivariate analysis, metabolites positively associated with inflammation included allantoin, taurodeoxycholic acid, norepinephrine, pyroglutamic acid, and L-hydroorotic acid. Conversely, metabolites showing negative associations with inflammation included benzoic acid, indole-3-acetaldehyde, methionine, citrulline, alphaketoglutarate, n-acetyl-ornithine, and 3-4-dihydroxibenzeneacetic acid. Non-inflamed patients exhibit preserved autophagy and reduced mitochondrial dysfunction. Understanding inflammation in this group hinges on the metabolism of arginine and the urea cycle. Additionally, the microbiota, particularly uricase-producing bacteria and those metabolizing tryptophan, play critical roles.
Collapse
Affiliation(s)
- María Peris-Fernández
- Health Research Institute Hospital La Fe, 46026 Valencia, Spain
- University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| | | | - Julià L Amengual
- Big Data AI and Biostatistics Platform, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
| | - Ángel Balaguer-Timor
- Big Data AI and Biostatistics Platform, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
| | | | | | | | | | - Julio Hernández-Jaras
- Health Research Institute Hospital La Fe, 46026 Valencia, Spain
- University and Polytechnic La Fe Hospital, 46026 Valencia, Spain
| |
Collapse
|
6
|
Wang S, Zhou H, Cui W, Zhang J, Wu D, Zhang N, Xu X. Qi Wei Anti-burn Tincture Remodels Liver Metabolic Pathways and Treats Burn Wounds Efficiently. J Burn Care Res 2024; 45:916-925. [PMID: 36402740 DOI: 10.1093/jbcr/irac175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Indexed: 02/17/2024]
Abstract
This work aims to elucidate the molecular mechanism of Qi Wei anti-burn Tincture (QW) on wound healing in burnt mice using metabolomics and molecular biology techniques. A scald model was first established in Kunming mice. After treatment, biochemical indicators for liver function and burnt skin tissues were then evaluated via biochemical detection and HE staining, respectively. Liver tissues were further analyzed for differential metabolites, inflammatory factors, and mRNA levels of cytokines using metabolomics and molecular biology techniques. Involved metabolic pathways were also identified using software. QW treatment did promote the healing of the burn wounds in Kunming mice with a downregulation of ALP, ALT, and AST to normal levels. In mouse liver tissue, the contents of glutamine, aspartic acid, and citrulline were significantly reduced, while the contents of 5-hydroxyproline, taurine, hypotaurine, and glutamic acid significantly increased. These major differential compounds are involved in the arginine metabolic pathway, nitrogen excretion, and the metabolism of taurine and hypotaurine, suggesting that QW reprogramed the above metabolic processes in the liver. Furthermore, the application of QW increased the expression of TGF-β1 and FGF-2 and reduced the levels of TNF-α, IL-1β, IL-6, and reactive oxygen species in the liver of mice induced by burn injury. This study found that QW treatment promoted metabolic pathway remodeling in the liver, which might be a potential mechanism for QW to treat burn wounds.
Collapse
Affiliation(s)
- Shuai Wang
- The First People's Hospital of Zhengzhou, P. R. China
| | - Hui Zhou
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, P. R. China
| | - Weiqi Cui
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, P. R. China
| | - Junwei Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, P. R. China
| | - Deqiao Wu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, P. R. China
| | - Nan Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, P. R. China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Zhengzhou University, P. R. China
| | - Xia Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, P. R. China
| |
Collapse
|
7
|
Li Y, Gao YN, Zhu YB, Lu WF, Yu JY, Dong YY, Xu MY, Peng B, Wu JZ, Su Q, Bai J, Shi XL, Kang YM, Li HB, Xu ML. Taurocholic acid ameliorates hypertension through the activation of TGR5 in the hypothalamic paraventricular nucleus. Food Funct 2024; 15:5088-5102. [PMID: 38666497 DOI: 10.1039/d4fo00808a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Diets rich in taurine can increase the production of taurine-conjugated bile acids, which are known to exert antihypertensive effects. Despite their benefits to the heart, kidney and arteries, their role in the central nervous system during the antihypertensive process remains unclear. Since hypothalamic paraventricular nucleus (PVN) plays a key role in blood pressure regulation, we aimed to investigate the function of bile acids in the PVN. The concentration of bile acids in the PVN of spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto rats (WKY) fed with normal chow was measured using LC-MS/MS, which identified taurocholic acid (TCA) as the most down-regulated bile acid. To fully understand the mechanism of TCA's functions in the PVN, bi-lateral PVN micro-infusion of TCA was carried out. TCA treatment in the PVN led to a significant reduction in the blood pressure of SHRs, with decreased plasma levels of norepinephrine and improved morphology of cardiomyocytes. It also decreased the number of c-fos+ neurons, reduced the inflammatory response, and suppressed oxidative stress in the PVN of the SHRs. Most importantly, the TGR5 receptors in neurons and microglia were activated. PVN infusion of SBI-115, a TGR5 specific antagonist, was able to counteract with TCA in the blood pressure regulation of SHRs. In conclusion, TCA supplementation in the PVN of SHRs can activate TGR5 in neurons and microglia, reduce the inflammatory response and oxidative stress, suppress activated neurons, and attenuate hypertension.
Collapse
Affiliation(s)
- Ying Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Ya-Nan Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Ying-Bao Zhu
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi, 712000, China
| | - Wen-Fang Lu
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi, 712000, China
| | - Jia-Yue Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Yuan-Yuan Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Meng-Yue Xu
- The Second Clinical College of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712000, China
| | - Bo Peng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Jun-Zhe Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Qing Su
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Juan Bai
- Department of Anesthesiology, Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiao-Lian Shi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Cardiovascular Sciences, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Meng-Lu Xu
- Department of Nephrology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China.
| |
Collapse
|
8
|
Jin W, Zheng M, Chen Y, Xiong H. Update on the development of TGR5 agonists for human diseases. Eur J Med Chem 2024; 271:116462. [PMID: 38691888 DOI: 10.1016/j.ejmech.2024.116462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/20/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
The G protein-coupled bile acid receptor 1 (GPBAR1) or TGR5 is widely distributed across organs, including the small intestine, stomach, liver, spleen, and gallbladder. Many studies have established strong correlations between TGR5 and glucose homeostasis, energy metabolism, immune-inflammatory responses, and gastrointestinal functions. These results indicate that TGR5 has a significant impact on the progression of tumor development and metabolic disorders such as diabetes mellitus and obesity. Targeting TGR5 represents an encouraging therapeutic approach for treating associated human ailments. Notably, the GLP-1 receptor has shown exceptional efficacy in clinical settings for diabetes management and weight loss promotion. Currently, numerous TGR5 agonists have been identified through natural product-based approaches and virtual screening methods, with some successfully progressing to clinical trials. This review summarizes the intricate relationships between TGR5 and various diseases emphasizing recent advancements in research on TGR5 agonists, including their structural characteristics, design tactics, and biological activities. We anticipate that this meticulous review could facilitate the expedited discovery and optimization of novel TGR5 agonists.
Collapse
Affiliation(s)
- Wangrui Jin
- Institute for Advanced Study, and College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China; Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mingyue Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yihua Chen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, China; Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Hai Xiong
- Institute for Advanced Study, and College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
9
|
Sheng JY, Meng ZF, Li Q, Yang YS. Recent advances in promising drugs for primary prevention of gastroesophageal variceal bleeding with cirrhotic portal hypertension. Hepatobiliary Pancreat Dis Int 2024; 23:4-13. [PMID: 37580228 DOI: 10.1016/j.hbpd.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Gastroesophageal variceal bleeding is one of the most severe complications of patients with cirrhosis. Although primary prevention drugs, including non-selective β-blockers, have effectively reduced the incidence of bleeding, their efficacy is limited due to side effects and related contraindications. With recent advances in precision medicine, precise drug treatment provides better treatment efficacy. DATA SOURCES Literature search was conducted in PubMed, MEDLINE and Web of Science for relevant articles published up to May 2022. Information on clinical trials was obtained from https://clinicaltrials.gov/ and http://www.chictr.org.cn/. RESULTS The in-depth understanding of the pathogenesis and advances of portal hypertension has enabled the discovery of multiple molecular targets for promising drugs. According to the site of action, these drugs could be classified into four classes: intrahepatic, extrahepatic, both intrahepatic and extrahepatic targets and others. All these classes of drugs offer advantages over traditional treatments in prevention of gastroesophageal variceal bleeding in patients with cirrhotic portal hypertension. CONCLUSIONS This review classified and summarized the promising drugs, which prevent gastroesophageal variceal bleeding by targeting specific markers of pathogenesis of portal hypertension, demonstrating the significance of using the precision medicine strategy to discover and develop promising drugs for the primary prevention of gastroesophageal variceal bleeding in patients with cirrhotic portal hypertension.
Collapse
Affiliation(s)
- Ji-Yao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130041, China; Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, the Second Hospital of Jilin University, Changchun 130041, China
| | - Zi-Fan Meng
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130041, China; Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, the Second Hospital of Jilin University, Changchun 130041, China
| | - Qiao Li
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130041, China
| | - Yong-Sheng Yang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun 130041, China; Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, the Second Hospital of Jilin University, Changchun 130041, China.
| |
Collapse
|
10
|
Chen J, Zhang X, Zhang Y, Jiang S, Han Y, Zhang L, Zhang Y, Du H. Taurine enhances growth performance by improving intestinal integrity and antioxidant capacity of weaned piglets. J Anim Sci 2024; 102:skae311. [PMID: 39394665 PMCID: PMC11604117 DOI: 10.1093/jas/skae311] [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: 05/17/2024] [Accepted: 10/10/2024] [Indexed: 10/14/2024] Open
Abstract
Taurine is an amino acid that has been considered by animal husbandry as a feed additive due to its abundant biological functions. However, the effective dose of taurine added to feed is unknown. The aim of the current study was to determine the optimal taurine supplementation level by investigating its effects on growth performance, diarrhea index, intestinal health, and antioxidant capacity of weaned piglets. A total of 160 crossbred piglets (Landrace × Yorkshire, initially 8.39 ± 0.11 kg) were assigned to 4 groups (10 pigs/pen and 4 replicates/group). Basal diets containing 0 (control, CON), 0.1%, 0.3%, and 0.5% taurine were respectively provided to the piglets for a duration of 28 d. Six piglets from each group were selected for euthanasia and subsequent sample collection on day 29. The results showed that dietary 0.3% or 0.5% taurine supplementation increased average daily gain (P < 0.05), feed-to-gain ratio (P < 0.01), and serum albumin (P < 0.05), and decreased diarrhea index (P < 0.01) and diamine oxidase (DAO) level in the serum (P < 0.05). The greater expression of tight junction-related genes, including ZO-1 (P < 0.05) and Claudin-1 (P < 0.01), were observed in the duodenum after supplementation with 0.5% taurine. The supplementation of 0.3% or 0.5% taurine resulted in a significant reduction of crypt depth (P < 0.01) and an increase of villus height-to-crypt depth ratio (P < 0.01) in the duodenum. A greater abundance of goblet cells was detected in the duodenum and jejunum of piglets fed 0.5% taurine (P < 0.05). In addition, serum superoxide dismutase (SOD) level, liver catalase (CAT) level, and liver total antioxidant capacity level were all significantly (P < 0.05) increased with 0.1%, 0.3% or 0.5% dietary taurine supplementation. On the whole, dietary supplementation with 0.3% or 0.5% taurine has the potential to significantly enhance the growth performance of piglets by improving the integrity of the intestinal barrier and boosting their antioxidant capacity.
Collapse
Affiliation(s)
- Jianjun Chen
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaofeng Zhang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310004, China
| | - Yuhui Zhang
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shouchuan Jiang
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yu Han
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lei Zhang
- Department of Animal Nutrition, Zhejiang NHU Group Corporation, Xinchang 312500, China
| | - Yuanyuan Zhang
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huahua Du
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
11
|
Zhou Y, Wang T, Fan H, Liu S, Teng X, Shao L, Shen Z. Research Progress on the Pathogenesis of Aortic Aneurysm and Dissection in Metabolism. Curr Probl Cardiol 2024; 49:102040. [PMID: 37595858 DOI: 10.1016/j.cpcardiol.2023.102040] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Aortic aneurysm and dissection are complicated diseases having both high prevalence and mortality. It is usually diagnosed at advanced stages and posing diagnostic and therapeutic challenges due to the limitations of current detecting methods for aortic dissection used in clinics. Metabonomics demonstrated its great potential capability in the early diagnosis and personalized treatment of several diseases. Emerging evidence suggests that metabolic disorders including amino acid metabolism, glycometabolism, and lipid metabolism disturbance are involved in the pathogenesis of aortic aneurysm and dissection by affecting multiple functional aortic cells. The purpose of this review is to provide new insights into the metabolism alterations and their related regulatory mechanisms with a focus on recent advances and findings and provide a theoretical basis for the diagnosis, prevention, and drug development for aortic aneurysm and dissection.
Collapse
Affiliation(s)
- Yihong Zhou
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Tingyu Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Hongyou Fan
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Shan Liu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Xiaomei Teng
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China.
| |
Collapse
|
12
|
Chen S, Shao Q, Chen J, Lv X, Ji J, Liu Y, Song Y. Bile acid signalling and its role in anxiety disorders. Front Endocrinol (Lausanne) 2023; 14:1268865. [PMID: 38075046 PMCID: PMC10710157 DOI: 10.3389/fendo.2023.1268865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Anxiety disorder is a prevalent neuropsychiatric disorder that afflicts 7.3%~28.0% of the world's population. Bile acids are synthesized by hepatocytes and modulate metabolism via farnesoid X receptor (FXR), G protein-coupled receptor (TGR5), etc. These effects are not limited to the gastrointestinal tract but also extend to tissues and organs such as the brain, where they regulate emotional centers and nerves. A rise in serum bile acid levels can promote the interaction between central FXR and TGR5 across the blood-brain barrier or activate intestinal FXR and TGR5 to release fibroblast growth factor 19 (FGF19) and glucagon-like peptide-1 (GLP-1), respectively, which in turn, transmit signals to the brain via these indirect pathways. This review aimed to summarize advancements in the metabolism of bile acids and the physiological functions of their receptors in various tissues, with a specific focus on their regulatory roles in brain function. The contribution of bile acids to anxiety via sending signals to the brain via direct or indirect pathways was also discussed. Different bile acid ligands trigger distinct bile acid signaling cascades, producing diverse downstream effects, and these pathways may be involved in anxiety regulation. Future investigations from the perspective of bile acids are anticipated to lead to novel mechanistic insights and potential therapeutic targets for anxiety disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuehan Song
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Santulli G, Kansakar U, Varzideh F, Mone P, Jankauskas SS, Lombardi A. Functional Role of Taurine in Aging and Cardiovascular Health: An Updated Overview. Nutrients 2023; 15:4236. [PMID: 37836520 PMCID: PMC10574552 DOI: 10.3390/nu15194236] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Taurine, a naturally occurring sulfur-containing amino acid, has attracted significant attention in recent years due to its potential health benefits. Found in various foods and often used in energy drinks and supplements, taurine has been studied extensively to understand its impact on human physiology. Determining its exact functional roles represents a complex and multifaceted topic. We provide an overview of the scientific literature and present an analysis of the effects of taurine on various aspects of human health, focusing on aging and cardiovascular pathophysiology, but also including athletic performance, metabolic regulation, and neurological function. Additionally, our report summarizes the current recommendations for taurine intake and addresses potential safety concerns. Evidence from both human and animal studies indicates that taurine may have beneficial cardiovascular effects, including blood pressure regulation, improved cardiac fitness, and enhanced vascular health. Its mechanisms of action and antioxidant properties make it also an intriguing candidate for potential anti-aging strategies.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Urna Kansakar
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| | - Fahimeh Varzideh
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Pasquale Mone
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Stanislovas S. Jankauskas
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| | - Angela Lombardi
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| |
Collapse
|
14
|
Stojanović NM, Randjelović PJ, Maslovarić A, Kostić M, Raičević V, Sakač M, Bjedov S. How do different bile acid derivatives affect rat macrophage function - Friends or foes? Chem Biol Interact 2023; 383:110688. [PMID: 37648052 DOI: 10.1016/j.cbi.2023.110688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/13/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
Due to an increased need for new immunomodulatory agents, many previously known molecules have been structurally modified in order to obtain new drugs, preserving at the same time some of the benevolent characteristics of the parent molecule. This study aimed to evaluate the immunomodulatory potential of a selected library of bile acid derivatives (BAD) using a broad spectrum of assays, evaluating rat peritoneal macrophages viability, cell membrane damage, lysosomal and adhesion function, and nitric oxide and cytokine production as a response to lipopolysaccharide stimulation. Also, in silico studies on two bile acid-activated receptors were conducted and the results were related to the observed in vitro effects. All tested BAD exerted significant toxicity in concentrations higher than 10 μM, which was determined based on mitochondria and cell membrane damage in a panel of assays. On the other hand, at lower concentrations, the tested BAD proved to be immunomodulatory since they affected lysosomal function, cell adhesion capacities and the ability to produce inflammatory cytokines in response to a stimulus. One of the compounds proved to exhibit significant toxicity toward macrophages, but also caused a concentration-dependent decrease in nitric oxide levels and was identified as a potential farnesoid X receptor agonist.
Collapse
Affiliation(s)
- Nikola M Stojanović
- Department of Physiology, Faculty of Medicine, University of Niš, 18000, Niš, Serbia.
| | - Pavle J Randjelović
- Department of Physiology, Faculty of Medicine, University of Niš, 18000, Niš, Serbia
| | | | - Miloš Kostić
- Department of Immunology, Faculty of Medicine, University of Niš, 18000, Niš, Serbia
| | - Vidak Raičević
- Department of Chemistry, Biochemistry, and Environmental Protection, Faculty of Sciences, University of Novi Sad, 21000, Novi Sad, Serbia
| | - Marija Sakač
- Department of Chemistry, Biochemistry, and Environmental Protection, Faculty of Sciences, University of Novi Sad, 21000, Novi Sad, Serbia
| | - Srđan Bjedov
- Department of Chemistry, Biochemistry, and Environmental Protection, Faculty of Sciences, University of Novi Sad, 21000, Novi Sad, Serbia
| |
Collapse
|
15
|
Gou X, Qin L, Wu D, Xie J, Lu Y, Zhang Q, He Y. Research Progress of Takeda G Protein-Coupled Receptor 5 in Metabolic Syndrome. Molecules 2023; 28:5870. [PMID: 37570840 PMCID: PMC10421342 DOI: 10.3390/molecules28155870] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Bile acids are acknowledged as signaling molecules involved in metabolic syndrome. The Takeda G protein-coupled receptor 5 (TGR5) functions as a significant bile acid receptor. The accumulated evidence suggests that TGR5 involves lipid homeostasis, glucose metabolism, and inflammation regulation. In line with this, recent preclinical studies also demonstrate that TGR5 plays a significant role in the generation and progression of metabolic syndrome, encompassing type 2 diabetes mellitus, obesity, atherosclerosis, and non-alcoholic fatty liver disease (NAFLD). In this review, we discuss the role of TGR5 in metabolic syndrome, illustrating the underlying mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Xianmei Gou
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Lin Qin
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Di Wu
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Jian Xie
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Yanliu Lu
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Qianru Zhang
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Yuqi He
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
16
|
Swiderski J, Sakkal S, Apostolopoulos V, Zulli A, Gadanec LK. Combination of Taurine and Black Pepper Extract as a Treatment for Cardiovascular and Coronary Artery Diseases. Nutrients 2023; 15:nu15112562. [PMID: 37299525 DOI: 10.3390/nu15112562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The shift in modern dietary regimens to "Western style" and sedentary lifestyles are believed to be partly responsible for the increase in the global burden of cardiovascular diseases. Natural products have been used throughout human history as treatments for a plethora of pathological conditions. Taurine and, more recently, black pepper have gained attention for their beneficial health effects while remaining non-toxic even when ingested in excess. Taurine, black pepper, and the major terpene constituents found in black pepper (i.e., β-caryophyllene; α-pinene; β-pinene; α-humulene; limonene; and sabinene) that are present in PhytoCann BP® have been shown to have cardioprotective effects based on anti-inflammatory, antioxidative, anti-hypertensive and anti-atherosclerotic mechanisms. This comprehensive review of the literature focuses on determining whether the combination of taurine and black pepper extract is an effective natural treatment for reducing cardiovascular diseases risk factors (i.e., hypertension and hyperhomocysteinemia) and for driving anti-inflammatory, antioxidative and anti-atherosclerotic mechanisms to combat coronary artery disease, heart failure, myocardial infarction, and atherosclerotic disease.
Collapse
Affiliation(s)
- Jordan Swiderski
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Samy Sakkal
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
- Immunology Program, Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| |
Collapse
|
17
|
Chen X, Hu C, Yan C, Tao E, Zhu Z, Shu X, Guo R, Jiang M. Maternal separation leads to dynamic changes of visceral hypersensitivity and fecal metabolomics from childhood to adulthood. Sci Rep 2023; 13:7670. [PMID: 37169847 PMCID: PMC10175246 DOI: 10.1038/s41598-023-34792-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023] Open
Abstract
We assessed dynamic changes in visceral hypersensitivity and fecal metabolomics through a mouse model of irritable bowel syndrome (IBS) from childhood to adulthood. A mouse model of IBS was constructed with maternal separation (MS) in early life. Male mice aged 25, 40, and 70 days were used. Visceral sensitivity was assessed by recording the reaction between the abdominal withdrawal reflex and colorectal distension. Metabolomics was identified and quantified by liquid chromatography-tandem mass spectrometry. The visceral sensitivity of the MS group was significantly higher than that of the non-separation (NS) group in the three age groups. The top four fecal differential metabolites in the different age groups were lipids, lipid molecules, organic heterocyclic compounds, organic acids and derivatives, and benzenoids. Five identical differential metabolites were detected in the feces and ileal contents of the MS and NS groups at different ages, namely, benzamide, taurine, acetyl-L-carnitine, indole, and ethylbenzene. Taurine and hypotaurine metabolism were the most relevant pathways at P25, whereas histidine metabolism was the most relevant pathway at P40 and P70. Visceral hypersensitivity in the MS group lasted from childhood to adulthood. The different metabolites and metabolic pathways detected in MS groups of different ages provide a theoretical basis for IBS pathogenesis.
Collapse
Affiliation(s)
- Xiaolong Chen
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
- Department of Pediatrics, The First People's Hospital of Jiashan, Jiashan, 314100, China
| | - Chenmin Hu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Chenxi Yan
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Enfu Tao
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Zhenya Zhu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Xiaoli Shu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Rui Guo
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China
| | - Mizu Jiang
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China.
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, 310052, China.
| |
Collapse
|
18
|
Ommati MM, Mobasheri A, Ma Y, Xu D, Tang Z, Manthari RK, Abdoli N, Azarpira N, Lu Y, Sadeghian I, Mousavifaraz A, Nadgaran A, Nikoozadeh A, Mazloomi S, Mehrabani PS, Rezaei M, Xin H, Mingyu Y, Niknahad H, Heidari R. Taurine mitigates the development of pulmonary inflammation, oxidative stress, and histopathological alterations in a rat model of bile duct ligation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1557-1572. [PMID: 36097067 DOI: 10.1007/s00210-022-02291-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
Lung injury is a significant complication associated with cholestasis/cirrhosis. This problem significantly increases the risk of cirrhosis-related morbidity and mortality. Hence, finding effective therapeutic options in this field has significant clinical value. Severe inflammation and oxidative stress are involved in the mechanism of cirrhosis-induced lung injury. Taurine (TAU) is an abundant amino acid with substantial anti-inflammatory and antioxidative properties. The current study was designed to evaluate the role of TAU in cholestasis-related lung injury. For this purpose, bile duct ligated (BDL) rats were treated with TAU (0.5 and 1% w: v in drinking water). Significant increases in the broncho-alveolar lavage fluid (BALF) level of inflammatory cells (lymphocytes, neutrophils, basophils, monocytes, and eosinophils), increased IgG, and TNF-α were detected in the BDL animals (14 and 28 days after the BDL surgery). Alveolar congestion, hemorrhage, and fibrosis were the dominant pulmonary histopathological changes in the BDL group. Significant increases in the pulmonary tissue biomarkers of oxidative stress, including reactive oxygen species formation, lipid peroxidation, increased oxidized glutathione levels, and decreased reduced glutathione, were also detected in the BDL rats. Moreover, significant myeloperoxidase activity and nitric oxide levels were seen in the lung of BDL rats. It was found that TAU significantly blunted inflammation, alleviated oxidative stress, and mitigated lung histopathological changes in BDL animals. These data suggest TAU as a potential protective agent against cholestasis/cirrhosis-related lung injury.
Collapse
Affiliation(s)
- Mohammad Mehdi Ommati
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mobasheri
- Physics, and Technology, Faculty of Medicine, Research Unit of Medical Imaging, University of Oulu, 90014, Oulu, Finland
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
- Department of Regenerative Medicine, State Research Institute Center for Innovative Medicine, 08406, Vilnius, Lithuania
| | - Yanqin Ma
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Dongmei Xu
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Zhongwei Tang
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Ram Kumar Manthari
- Department of Biotechnology, GITAM Institute of Science, Gandhi Institute of Technology and Management, Visakhapatnam-530045, Andhra Pradesh, India
| | - Narges Abdoli
- Food and Drug Administration, Iran Ministry of Health and Medical Education, Tehran, Iran
| | - Negar Azarpira
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yu Lu
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abolghasem Mousavifaraz
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Nadgaran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Nikoozadeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahra Mazloomi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooria Sayar Mehrabani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Rezaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hu Xin
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Yang Mingyu
- College of Life Sciences, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
19
|
Wang L, Wang S, Zhang Q, He C, Fu C, Wei Q. The role of the gut microbiota in health and cardiovascular diseases. MOLECULAR BIOMEDICINE 2022; 3:30. [PMID: 36219347 PMCID: PMC9554112 DOI: 10.1186/s43556-022-00091-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiota is critical to human health, such as digesting nutrients, forming the intestinal epithelial barrier, regulating immune function, producing vitamins and hormones, and producing metabolites to interact with the host. Meanwhile, increasing evidence indicates that the gut microbiota has a strong correlation with the occurrence, progression and treatment of cardiovascular diseases (CVDs). In patients with CVDs and corresponding risk factors, the composition and ratio of gut microbiota have significant differences compared with their healthy counterparts. Therefore, gut microbiota dysbiosis, gut microbiota-generated metabolites, and the related signaling pathway may serve as explanations for some of the mechanisms about the occurrence and development of CVDs. Several studies have also demonstrated that many traditional and latest therapeutic treatments of CVDs are associated with the gut microbiota and its generated metabolites and related signaling pathways. Given that information, we summarized the latest advances in the current research regarding the effect of gut microbiota on health, the main cardiovascular risk factors, and CVDs, highlighted the roles and mechanisms of several metabolites, and introduced corresponding promising treatments for CVDs regarding the gut microbiota. Therefore, this review mainly focuses on exploring the role of gut microbiota related metabolites and their therapeutic potential in CVDs, which may eventually provide better solutions in the development of therapeutic treatment as well as the prevention of CVDs.
Collapse
Affiliation(s)
- Lu Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Shiqi Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Qing Zhang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chengqi He
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chenying Fu
- grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,grid.412901.f0000 0004 1770 1022Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Quan Wei
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| |
Collapse
|
20
|
Duan Y, Xing Y, Zeng S, Dan X, Mo Z, Zhang J, Li Y. Integration of metagenomic and metabolomic insights into the effects of microcystin-LR on intestinal microbiota of Litopenaeus vannamei. Front Microbiol 2022; 13:994188. [PMID: 36212851 PMCID: PMC9537473 DOI: 10.3389/fmicb.2022.994188] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/05/2022] [Indexed: 12/04/2022] Open
Abstract
Microcystin-LR (MC-LR) is a hazardous substance that threaten the health of aquatic animals. Intestinal microbes and their metabolites can interact with hosts to influence physiological homeostasis. In this study, the shrimp Litopenaeus vannamei were exposed to 1.0 μg/l MC-LR for 72 h, and the toxic effects of MC-LR on the intestinal microbial metagenomic and metabolomic responses of the shrimp were investigated. The results showed that MC-LR stress altered the gene functions of intestinal microbial, including ABC transporter, sulfur metabolism and riboflavin (VB2) metabolism, and induced a significant increase of eight carbohydrate metabolism enzymes. Alternatively, intestinal metabolic phenotypes were also altered, especially ABC transporters, protein digestion and absorption, and the biosynthesis and metabolism of amino acid. Furthermore, based on the integration of intestinal microbial metagenomic and metabolome, four bacteria species (Demequina globuliformis, Demequina sp. NBRC 110055, Sphingomonas taxi and Sphingomonas sp. RIT328) and three metabolites (yangonin, α-hederin and soyasaponin ii) biomarkers were identified. Overall, our study provides new insights into the effects of MC-LR on the intestinal microbial functions of L. vannamei.
Collapse
Affiliation(s)
- Yafei Duan
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, Sanya Tropical Fisheries Research Institute, Sanya, China
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Yifu Xing
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Shimin Zeng
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Xueming Dan
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Zequan Mo
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Jiasong Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- *Correspondence: Jiasong Zhang,
| | - Yanwei Li
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, China
- Yanwei Li,
| |
Collapse
|
21
|
Effects of taurine on vascular tone. Amino Acids 2022; 54:1527-1540. [DOI: 10.1007/s00726-022-03198-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 08/05/2022] [Indexed: 11/01/2022]
|
22
|
Valorization of avocado seeds with antioxidant capacity using pressurized hot water extraction. Sci Rep 2022; 12:13036. [PMID: 35906278 PMCID: PMC9338084 DOI: 10.1038/s41598-022-17326-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 07/25/2022] [Indexed: 02/07/2023] Open
Abstract
The pulp of avocado (Persea Americana) is widely consumed as the primary food source, while the seed is often discarded as food waste. Increased consumption of avocado would inevitably results in production of waste by-products such as avocado seeds, hence the ability to extract phytochemicals from such waste, and upcycling to potential nutraceutical products is of great interest. The overall aim of this study is to explore avocado seeds as potential functional food through the combined use of a green extraction method, chemical standardization and pattern recognition tools, and biological characterization assays. Specifically, this study utilized an organic solvent-free extraction method, pressurized hot water extraction (PHWE) to extract phytochemicals from avocado seeds and liquid chromatography mass spectrometry (LCMS) was used to identify the phytochemicals present in the avocado seeds. Our results demonstrated that avocado seed extracts have antioxidant activity and inhibited oxidative stress-induced metabolomics changes in endothelial cells, suggesting that avocado seed extracts have vasoprotective actions.
Collapse
|
23
|
Yang JY, Zhang TT, Yu ZL, Wang CC, Zhao YC, Wang YM, Xue CH. Taurine Alleviates Trimethylamine N-Oxide-Induced Atherosclerosis by Regulating Bile Acid Metabolism in ApoE -/- Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5738-5747. [PMID: 35486890 DOI: 10.1021/acs.jafc.2c01376] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Trimethylamine N-oxide (TMAO) widely exists in seafood and is associated with the atherosclerosis progress, but dietary seafood reduced the cardiovascular risk. This intimates that there may be some ingredients in seafood to offset the cardiovascular risk caused by TMAO. Taurine is a marker ingredient in seafood. Thus, this study determined the influences of taurine on TMAO-induced atherosclerosis in apolipoprotein-E-deficient mice. The results showed that dietary taurine significantly reduced the TMAO-induced atherosclerotic lesion area. Further studies found that taurine increased the hepatic- and serum-conjugated bile acid/unconjugated bile acid ratio via increasing hepatic gene expression of conjugated bile acid synthesis. Meanwhile, taurine changed TMAO-induced abnormal bile acid profiles in the gallbladder. Moreover, taurine increased bile acid deconjugation by enhancing the genera Ruminiclostridium level and increased excretion of fecal neutral sterols. Additionally, taurine attenuated inflammation in the serum and artery. These results indicate that taurine alleviated TMAO-induced atherosclerosis via regulating bile acid metabolism.
Collapse
Affiliation(s)
- Jin-Yue Yang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Zhu-Lin Yu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Ying-Cai Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
- Laboratory of Marine Drugs & Biological Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong 266237, People's Republic of China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
- Laboratory of Marine Drugs & Biological Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong 266237, People's Republic of China
| |
Collapse
|
24
|
Stepien M, Lopez-Nogueroles M, Lahoz A, Kühn T, Perlemuter G, Voican C, Ciocan D, Boutron-Ruault MC, Jansen E, Viallon V, Leitzmann M, Tjønneland A, Severi G, Mancini FR, Dong C, Kaaks R, Fortner RT, Bergmann MM, Boeing H, Trichopoulou A, Karakatsani A, Peppa E, Palli D, Krogh V, Tumino R, Sacerdote C, Panico S, Bueno-de-Mesquita HB, Skeie G, Merino S, Ros RZ, Sánchez MJ, Amiano P, Huerta JM, Barricarte A, Sjöberg K, Ohlsson B, Nyström H, Werner M, Perez-Cornago A, Schmidt JA, Freisling H, Scalbert A, Weiderpass E, Christakoudi S, Gunter MJ, Jenab M. Prediagnostic alterations in circulating bile acid profiles in the development of hepatocellular carcinoma. Int J Cancer 2022; 150:1255-1268. [PMID: 34843121 DOI: 10.1002/ijc.33885] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022]
Abstract
Bile acids (BAs) play different roles in cancer development. Some are carcinogenic and BA signaling is also involved in various metabolic, inflammatory and immune-related processes. The liver is the primary site of BA synthesis. Liver dysfunction and microbiome compositional changes, such as during hepatocellular carcinoma (HCC) development, may modulate BA metabolism increasing concentration of carcinogenic BAs. Observations from prospective cohorts are sparse. We conducted a study (233 HCC case-control pairs) nested within a large observational prospective cohort with blood samples taken at recruitment when healthy with follow-up over time for later cancer development. A targeted metabolomics method was used to quantify 17 BAs (primary/secondary/tertiary; conjugated/unconjugated) in prediagnostic plasma. Odd ratios (OR) for HCC risk associations were calculated by multivariable conditional logistic regression models. Positive HCC risk associations were observed for the molar sum of all BAs (ORdoubling = 2.30, 95% confidence intervals [CI]: 1.76-3.00), and choline- and taurine-conjugated BAs. Relative concentrations of BAs showed positive HCC risk associations for glycoholic acid and most taurine-conjugated BAs. We observe an association between increased HCC risk and higher levels of major circulating BAs, from several years prior to tumor diagnosis and after multivariable adjustment for confounders and liver functionality. Increase in BA concentration is accompanied by a shift in BA profile toward higher proportions of taurine-conjugated BAs, indicating early alterations of BA metabolism with HCC development. Future studies are needed to assess BA profiles for improved stratification of patients at high HCC risk and to determine whether supplementation with certain BAs may ameliorate liver dysfunction.
Collapse
Affiliation(s)
- Magdalena Stepien
- Nutrition and Metabolism Branch (NME), International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | | | - Agustin Lahoz
- Analytical Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gabriel Perlemuter
- INSERM U996, Intestinal Microbiota, Macrophages and Liver Inflammation, DHU Hepatinov, Labex LERMIT, Clamart, France
- Faculté de Médecine Paris-Sud, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Service d'hépato-Gastroentérologie, Hôpital Antoine-Béclère, Hôpitaux Universitaires Paris-Sud, Assistance Publique-Hôpitaux de Paris, Clamart, France
| | - Cosmin Voican
- INSERM U996, Intestinal Microbiota, Macrophages and Liver Inflammation, DHU Hepatinov, Labex LERMIT, Clamart, France
- Faculté de Médecine Paris-Sud, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Service d'hépato-Gastroentérologie, Hôpital Antoine-Béclère, Hôpitaux Universitaires Paris-Sud, Assistance Publique-Hôpitaux de Paris, Clamart, France
| | - Dragos Ciocan
- INSERM U996, Intestinal Microbiota, Macrophages and Liver Inflammation, DHU Hepatinov, Labex LERMIT, Clamart, France
- Faculté de Médecine Paris-Sud, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Service d'hépato-Gastroentérologie, Hôpital Antoine-Béclère, Hôpitaux Universitaires Paris-Sud, Assistance Publique-Hôpitaux de Paris, Clamart, France
| | - Marie-Christine Boutron-Ruault
- CESP, Faculté de Médecine-Université Paris-Saclay, Faculté de Médecine-UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Eugene Jansen
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Vivian Viallon
- Nutrition and Metabolism Branch (NME), International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Michael Leitzmann
- Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Anne Tjønneland
- Diet, Genes and Environment Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Gianluca Severi
- CESP, Faculté de Médecine-Université Paris-Saclay, Faculté de Médecine-UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Francesca Romana Mancini
- CESP, Faculté de Médecine-Université Paris-Saclay, Faculté de Médecine-UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Catherine Dong
- CESP, Faculté de Médecine-Université Paris-Saclay, Faculté de Médecine-UVSQ, INSERM, Université Paris-Saclay, Villejuif, France
- Gustave Roussy, Villejuif, France
- Department of Gastroenterology, Hôpital de Bicêtre, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Manuela M Bergmann
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Germany
| | | | - Anna Karakatsani
- Hellenic Health Foundation, Athens, Greece
- 2nd Pulmonary Medicine Department, School of Medicine, National and Kapodistrian University of Athens, "ATTIKON" University Hospital, Haidari, Greece
| | | | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network-ISPRO, Florence, Italy
| | - Vittorio Krogh
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori Milano, Milan, Italy
| | - Rosario Tumino
- Department of Cancer Registry and Histopathology, "M.P. Arezzo" Hospital, ASP Ragusa, Ragusa, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - H Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, The Netherlands
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, UK
- Department of Social & Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Guri Skeie
- Department of Community Medicine, UIT-The Arctic University of Norway, Tromsø, Norway
| | | | - Raul Zamora Ros
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Maria Jose Sánchez
- Escuela Andaluza de Salud Pública, Instituto de Investigación Biosanitaria ibs.GRANADA, Universidad de Granada, Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Pilar Amiano
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Public Health Division of Gipuzkoa, BioDonostia Research Institute, San Sebastian, Spain
| | - Jose Mª Huerta
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | - Aurelio Barricarte
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Klas Sjöberg
- Department of Gastroenterology and Nutrition, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Bodil Ohlsson
- Department of Internal Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Hanna Nyström
- Department of Surgery, Umeå University, Umeå, Sweden
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Marten Werner
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Julie A Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Heinz Freisling
- Nutrition and Metabolism Branch (NME), International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Augustin Scalbert
- Nutrition and Metabolism Branch (NME), International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Elisabete Weiderpass
- Office of the Director, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Sofia Christakoudi
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Transplantation, King's College London, London, UK
| | - Marc J Gunter
- Nutrition and Metabolism Branch (NME), International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Mazda Jenab
- Nutrition and Metabolism Branch (NME), International Agency for Research on Cancer (IARC-WHO), Lyon, France
| |
Collapse
|
25
|
Oyagbemi AA, Adejumobi OA, Jarikre TA, Ajani OS, Asenuga ER, Gbadamosi IT, Adedapo ADA, Aro AO, Ogunpolu BS, Hassan FO, Falayi OO, Ogunmiluyi IO, Omobowale TO, Arojojoye OA, Ola-Davies OE, Saba AB, Adedapo AA, Emikpe BO, Oyeyemi MO, Nkadimeng SM, McGaw LJ, Kayoka-Kabongo PN, Oguntibeju OO, Yakubu MA. Clofibrate, a Peroxisome Proliferator-Activated Receptor-Alpha (PPARα) Agonist, and Its Molecular Mechanisms of Action against Sodium Fluoride-Induced Toxicity. Biol Trace Elem Res 2022; 200:1220-1236. [PMID: 33893992 DOI: 10.1007/s12011-021-02722-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/12/2021] [Indexed: 01/16/2023]
Abstract
Sodium fluoride (NaF) is one of the neglected environmental pollutants. It is ubiquitously found in the soil, water, and environment. Interestingly, fluoride has been extensively utilized for prevention of dental caries and tartar formation, and may be added to mouthwash, mouth rinse, and toothpastes. This study is aimed at mitigating fluoride-induced hypertension and nephrotoxicity with clofibrate, a peroxisome proliferator-activated receptor-alpha (PPARα) agonist. For this study, forty male Wistar rats were used and randomly grouped into ten rats per group, control, sodium fluoride (NaF; 300 ppm) only, NaF plus clofibrate (250 mg/kg) and NaF plus lisinopril (10 mg/kg), respectively, for 7 days. The administration of NaF was by drinking water ad libitum, while clofibrate and lisinopril were administered by oral gavage. Administration of NaF induced hypertension, and was accompanied with exaggerated oxidative stress; depletion of antioxidant defence system; reduced nitric oxide production; increased systolic, diastolic and mean arterial pressure; activation of angiotensin-converting enzyme activity and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB); and testicular apoptosis. Treatment of rats with clofibrate reduced oxidative stress, improved antioxidant status, lowered high blood pressure through the inhibition of angiotensin-converting enzyme activity, mineralocorticoid receptor over-activation, and abrogated testicular apoptosis. Taken together, clofibrate could offer exceptional therapeutic benefit in mitigating toxicity associated with sodium fluoride.
Collapse
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Olumuyiwa Abiola Adejumobi
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Theophilus Aghogho Jarikre
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumide Samuel Ajani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ebunoluwa Racheal Asenuga
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Benin, Benin, Nigeria
| | | | | | - Abimbola Obemisola Aro
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Florida, South Africa
| | - Blessing Seun Ogunpolu
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Fasilat Oluwakemi Hassan
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Olubunmi Falayi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Iyanuoluwa Omolola Ogunmiluyi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temidayo Olutayo Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Olufunke Eunice Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adebowale Benard Saba
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeolu Alex Adedapo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Benjamin Obukowho Emikpe
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Sanah Malomile Nkadimeng
- Phytomedicine Programme, Department of Paraclinical Science, Faculty of Veterinary Science, University of Pretoria, Old Soutpan Road, Onderstepoort, Pretoria, 0110, South Africa
| | - Lyndy Joy McGaw
- Phytomedicine Programme, Department of Paraclinical Science, Faculty of Veterinary Science, University of Pretoria, Old Soutpan Road, Onderstepoort, Pretoria, 0110, South Africa
| | - Prudence Ngalula Kayoka-Kabongo
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Florida, South Africa
| | - Oluwafemi Omoniyi Oguntibeju
- Phytomedicine and Phytochemistry Group, Oxidative Stress Research Centre, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, Cape Town, 7535, South Africa
| | - Momoh Audu Yakubu
- Department of Environmental & Interdisciplinary Sciences, College of Science, Engineering & Technology, Vascular Biology Unit, Center for Cardiovascular Diseases, Texas Southern University, Houston, TX, USA
| |
Collapse
|
26
|
Zou W, Liu B, Wang Y, Shi F, Pang S. Metformin attenuates high glucose-induced injury in islet microvascular endothelial cells. Bioengineered 2022; 13:4385-4396. [PMID: 35139776 PMCID: PMC8973819 DOI: 10.1080/21655979.2022.2033411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
As one of the most frequently prescribed antidiabetic drugs, metformin can lower glucose levels, improve insulin resistance manage body weight. However, the effect of metformin on islet microcirculation remains unclear. In the present study, to explore the effect of metformin on islet endothelial cells and investigated the underlying mechanism, we assessed the effects of metformin on islet endothelial cell survival, proliferation, oxidative stress and apoptosis. Our results suggest that metformin stimulates the proliferation of pancreatic islet endothelial cells and inhibits the apoptosis and oxidative stress caused by high glucose levels. By activating farnesoid X receptor (FXR), metformin increases the expression of vascular endothelial growth factor-A (VEGF-A) and endothelial nitric oxide synthase (eNOS), improves the production of nitric oxide (NO) and decreases the production of ROS. After the inhibition of FXR or VEGF-A, all of the effects disappeared. Thus, metformin appears to regulate islet microvascular endothelial cell (IMEC) proliferation, apoptosis and oxidative stress by activating the FXR/VEGF-A/eNOS pathway. These findings provide a new mechanism underlying the islet-protective effect of metformin.
Collapse
Affiliation(s)
- Wenyu Zou
- Department of endocrinologyEndocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bingkun Liu
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, China
| | - Yulu Wang
- Department of Internal Medicine, Weifang Medical University, Weifang, China
| | - Fangbin Shi
- Department of endocrinologyEndocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuguang Pang
- Department of endocrinologyEndocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
27
|
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: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
28
|
Kp AD, Martin A. Recent insights into the molecular regulators and mechanisms of taurine to modulate lipid metabolism: a review. Crit Rev Food Sci Nutr 2022; 63:6005-6017. [PMID: 35040723 DOI: 10.1080/10408398.2022.2026873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lipid metabolism disorders such as hypertriglyceridemia and hypercholesterolemia are risk factors for cardiovascular diseases and atherosclerosis that are grave public health issues. Taurine, a sulfur-containing non-essential amino acid exerts a wide range of physiological effects that regulate lipid metabolic disorders. Although the effects of taurine on lipid-lowering have been reported in animals and humans, mechanisms elucidating the lipid-lowering action of taurine remain unclear. A series of molecular regulators associated with lipid metabolism have been identified in the past few decades. These include nuclear receptors, transcription factors, and enzymes that undergo important changes during taurine treatment. In this review, we focus on the role of taurine in lipid metabolism and discuss taurine-related interventions in combating lipid disorders.
Collapse
Affiliation(s)
- Arya Devi Kp
- Department of Food Safety and Analytical Quality Control Laboratory, CSIR - Central Food Technological Research Institute, Mysore, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC, Ghaziabad, Uttar Pradesh, India
| | - Asha Martin
- Department of Food Safety and Analytical Quality Control Laboratory, CSIR - Central Food Technological Research Institute, Mysore, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
29
|
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]
|
30
|
Zhang S, Zhou J, Wu W, Zhu Y, Liu X. The Role of Bile Acids in Cardiovascular Diseases: from Mechanisms to Clinical Implications. Aging Dis 2022; 14:261-282. [PMID: 37008052 PMCID: PMC10017164 DOI: 10.14336/ad.2022.0817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022] Open
Abstract
Bile acids (BAs), key regulators in the metabolic network, are not only involved in lipid digestion and absorption but also serve as potential therapeutic targets for metabolic disorders. Studies have shown that cardiac dysfunction is associated with abnormal BA metabolic pathways. As ligands for several nuclear receptors and membrane receptors, BAs systematically regulate the homeostasis of metabolism and participate in cardiovascular diseases (CVDs), such as myocardial infarction, diabetic cardiomyopathy, atherosclerosis, arrhythmia, and heart failure. However, the molecular mechanism by which BAs trigger CVDs remains controversial. Therefore, the regulation of BA signal transduction by modulating the synthesis and composition of BAs is an interesting and novel direction for potential therapies for CVDs. Here, we mainly summarized the metabolism of BAs and their role in cardiomyocytes and noncardiomyocytes in CVDs. Moreover, we comprehensively discussed the clinical prospects of BAs in CVDs and analyzed the clinical diagnostic and application value of BAs. The latest development prospects of BAs in the field of new drug development are also prospected. We aimed to elucidate the underlying mechanism of BAs treatment in CVDs, and the relationship between BAs and CVDs may provide new avenues for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Shuwen Zhang
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Junteng Zhou
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
- Health Management Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Wenchao Wu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Ye Zhu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China.
- Correspondence should be addressed to: Prof. Xiaojing Liu (), and Prof. Ye Zhu (), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China.
- Correspondence should be addressed to: Prof. Xiaojing Liu (), and Prof. Ye Zhu (), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
31
|
Li Y, Zhao D, Qian M, Liu J, Pan C, Zhang X, Duan X, Zhang Y, Jia W, Wang L. Amlodipine, an anti-hypertensive drug, alleviates non-alcoholic fatty liver disease by modulating gut microbiota. Br J Pharmacol 2021; 179:2054-2077. [PMID: 34862599 DOI: 10.1111/bph.15768] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/08/2021] [Accepted: 11/21/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Non-alcoholic fatty liver disease (NAFLD) represents a severe public health problem. It often coexists with hypertension in the context of metabolic syndrome. Here, we investigated the effects of amlodipine on non-alcoholic fatty liver disease combined with hypertension and the underlying mechanism. EXPERIMENTAL APPROACH mice were fed with high-fat diet and 0.05% N-Nitro-L-arginine methylester sterile water to induce NAFLD with hypertension. Gut microbiota composition and function were assessed by 16S ribosomal DNA and metagenomic sequencing. Untargeted metabolome profiles were applied to identify differential metabolites in mice cecum. KEY RESULTS Amlodipine besylate (AB) and amlodipine aspartate (AA) significantly decreased liver injury, hepatic steatosis and improved lipid metabolism with a concomitant reduction in the expression of lipogenic genes in mice with NAFLD and hypertension. Mechanistically, AA and AB have potential in restoring intestinal barrier integrity and improving antimicrobial defense along with the elevated abundances of Akkermansia, Bacteroides and Lactobacillus. Noteworthily, the gut microbiota in AB and AA-treated mice had higher abundance of functional genes involved in taurine and hypotaurine metabolism. Consistently, the strengthened taurine and hypotaurine metabolism was confirmed by the untargeted metabolome analysis. Based on the correlation and causal analysis, the altered gut microbiota composition and the enhancement of taurine and hypotaurine metabolism may synergistically decreased ALT, liver triglycerides, lipogenic genes and plasma cholesterol in HFD-fed hypertensive mice. CONCLUSION AND IMPLICATIONS Collectively, AA and AB exert multi-factorial improvements in NAFLD and hypertension by modulating gut microbiota, and may serve as a promising therapeutic agent for treating these diseases.
Collapse
Affiliation(s)
- Yang Li
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Danyang Zhao
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Minyi Qian
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Chuyue Pan
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xinxin Zhang
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xubin Duan
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yufei Zhang
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wenxin Jia
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lirui Wang
- Institute of Modern Biology, Nanjing University, Nanjing, China
| |
Collapse
|
32
|
Determination of metabolic phenotype and potential biomarkers in the liver of heroin addicted mice with hepatotoxicity. Life Sci 2021; 287:120103. [PMID: 34743944 DOI: 10.1016/j.lfs.2021.120103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Heroin is a semi-synthetic opioid that is commonly abused drugs in the world. It can cause hepatic injury and lead to multiple organs dysfunction to its addicts. Only a few reports exist on the metabolic changes and mechanisms in the liver of heroin-addicted mice with hepatic injury. METHODS Twelve adult male Kunming mice (30-40 g) were divided into two groups randomly. The mice in the heroin-addicted group were injected subcutaneously in the first ten days with an increased dosage of heroin from 10 mg/kg to 55 mg/kg. The dosage was then stabilized at 55 mg/kg for three days. The control group was injected with the same amount of saline in the same manner. The hepatic injury was confirmed through the combination of histopathological observation and aminotransferase (AST) and alanine aminotransferase (ALT) determination. The withdrawal symptoms were recorded and used for assessment of heroin addiction. Eventually, liver metabolic biomarkers of heroin-addicted mice with hepatotoxicity were measured using UHPLC-MS/MS. RESULTS Biochemical analysis and histopathological observation showed that heroin-addicted mice had a liver injury. The liver metabolites of heroin-addicted mice changed significantly. Metabonomics analysis revealed 41 metabolites in the liver of addicted heroin mice as biomarkers involving 34 metabolic pathways. Among them, glutathione metabolism, taurine and hypotaurine metabolism, vitamin B2 metabolism, riboflavin metabolism, and single-carbon metabolism pathways were markedly dispruted. CONCLUSIONS Heroin damages the liver and disrupts the liver's metabolic pathways. Glutathione, taurine, riboflavin, 4-pyridoxate, folic acid, and methionine are important metabolic biomarkers, which may be key targets of heroin-induced liver damage. Thus, this study provides an in-depth understanding of the mechanisms of heroin-induced hepatotoxicity and potential biomarkers of liver damage.
Collapse
|
33
|
Qi L, Tian Y, Chen Y. Circulating Bile Acid Profiles: A Need for Further Examination. J Clin Endocrinol Metab 2021; 106:3093-3112. [PMID: 34279029 DOI: 10.1210/clinem/dgab531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Indexed: 12/15/2022]
Abstract
CONTEXT Bile acids (BAs) are increasingly recognized as metabolic and chronobiologic integrators that synchronize the systemic metabolic response to nutrient availability. Alterations in the concentration and/or composition of circulating BAs are associated with a number of metabolic disorders, such as obesity, type 2 diabetes mellitus (T2DM), insulin resistance (IR), and metabolic associated fatty liver disease (MAFLD). This review summarizes recent evidence that links abnormal circulating BA profiles to multiple metabolic disorders, and discusses the possible mechanisms underlying the connections to determine the role of BA profiling as a novel biomarker for these abnormalities. EVIDENCE ACQUISITION The review is based on a collection of primary and review literature gathered from a PubMed search of BAs, T2DM, IR, and MAFLD, among other keywords. EVIDENCE SYNTHESIS Obese and IR subjects appear to have elevated fasting circulating BAs but lower postprandial increase when compared with controls. The possible underlying mechanisms are disruption in the synchronization between the feeding/fasting cycle and the properties of BA-regulated metabolic pathways. Whether BA alterations are associated per se with MAFLD remains inconclusive. However, increased fasting circulating BAs level was associated with higher risk of advanced fibrosis stage. Thus, for patients with MAFLD, dynamically monitoring the circulating BA profiles may be a promising tool for the stratification of MAFLD. CONCLUSIONS Alterations in the concentration, composition, and rhythm of circulating BAs are associated with adverse events in systemic metabolism. Subsequent investigations regarding these aspects of circulating BA kinetics may help predict future metabolic disorders and guide therapeutic interventions.
Collapse
Affiliation(s)
- Li Qi
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning Province, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Yongsheng Chen
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| |
Collapse
|
34
|
Yu Z, Xue W, Zhou M, Wang L, Wu S, Zhao W, Ding L. Potential Antihypertensive Mechanisms of the Egg White-Derived Peptide QIGLF in Spontaneously Hypertensive Rats Revealed Using Untargeted Serum Metabolomics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:12063-12071. [PMID: 34581184 DOI: 10.1021/acs.jafc.1c05599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The angiotensin-converting enzyme (ACE) inhibitory peptide QIGLF derived from egg white was shown to have significant in vivo antihypertensive effects in our previous study, but the intervention mechanisms at the metabolic level are still unclear. The UPLC-QTOF/MS-based untargeted metabolomics approach was used to clarify the potential antihypertensive mechanisms of QIGLF in the serum of spontaneously hypertensive rats (SHRs). Multivariate statistical analysis showed a clear difference in the metabolite profiles between the QIGLF and model groups. The results suggested that eight potential biomarkers were identified, that is, adrenic acid, ursodeoxycholic acid, glycocholic acid, taurocholic acid, tryptophan, acetylindoxyl, tyrosine, and 2-phenylethanol, which were mainly involved in aromatic amino acid biosynthesis and metabolism, biosynthesis of bile acid, and biosynthesis of unsaturated fatty acids. QIGLF might exert antihypertensive effects by improving endothelial dysfunction. This study provides a theoretical basis for future research and application of ACE inhibitory peptides in the prevention and improvement of hypertension.
Collapse
Affiliation(s)
- Zhipeng Yu
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, P. R. China
| | - Wenjun Xue
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, P. R. China
| | - Mingjie Zhou
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, P. R. China
| | - Li Wang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, P. R. China
| | - Sijia Wu
- Lab of Nutrition and Functional Food, Jilin University, Changchun 130062, P. R. China
| | - Wenzhu Zhao
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, P. R. China
| | - Long Ding
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, P. R. China
| |
Collapse
|
35
|
Sauerbruch T, Hennenberg M, Trebicka J, Beuers U. Bile Acids, Liver Cirrhosis, and Extrahepatic Vascular Dysfunction. Front Physiol 2021; 12:718783. [PMID: 34393832 PMCID: PMC8358446 DOI: 10.3389/fphys.2021.718783] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
The bile acid pool with its individual bile acids (BA) is modulated in the enterohepatic circulation by the liver as the primary site of synthesis, the motility of the gallbladder and of the intestinal tract, as well as by bacterial enzymes in the intestine. The nuclear receptor farnesoid X receptor (FXR) and Gpbar1 (TGR5) are important set screws in this process. Bile acids have a vasodilatory effect, at least according to in vitro studies. The present review examines the question of the extent to which the increase in bile acids in plasma could be responsible for the hyperdynamic circulatory disturbance of liver cirrhosis and whether modulation of the bile acid pool, for example, via administration of ursodeoxycholic acid (UDCA) or via modulation of the dysbiosis present in liver cirrhosis could influence the hemodynamic disorder of liver cirrhosis. According to our analysis, the evidence for this is limited. Long-term studies on this question are lacking.
Collapse
Affiliation(s)
- Tilman Sauerbruch
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Martin Hennenberg
- Department of Urology I, University Hospital, LMU Munich, Munich, Germany
| | - Jonel Trebicka
- Translational Hepatology, Medical Department, University of Frankfurt, Frankfurt, Germany
| | - Ulrich Beuers
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, location AMC, Amsterdam, Netherlands
| |
Collapse
|
36
|
Nabi AA, Atta SA, El-Ahwany E, Elzayat E, Saleh H. Taurine Upregulates miRNA-122-5p Expression and Suppresses the Metabolizing Enzymes of Glycolytic Pathway in Hepatocellular Carcinoma. Mol Biol Rep 2021; 48:5549-5559. [PMID: 34313924 DOI: 10.1007/s11033-021-06571-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a complicated disease with a poor prognosis and high mortality rates. The prevention, control, diagnosis, and treatment of liver cancer have become vital focuses in healthcare research. AIM This study aimed to evaluate the in vitro effect of taurine (Tau) on the expression of miR-122-5p that targets some limiting glycolytic enzymes and affects the overall glycolytic pathway in HepG2 cells. METHOD IC50 and the inhibitory effect of Tau on cell proliferation were measured after 48 h by MTT assay. Then, the mRNA expressions of some apoptosis-related genes P53, BAX, Caspase-3, and Bcl-2 were measured using quantitative real-time (qRT-PCR) and the protein levels were confirmed by enzyme-linked immunosorbent assay (ELISA). The activities of some antioxidant's biomarkers were assessed. The gene expression of miR-122-5p that targets some limiting glycolytic enzymes; Aldolase and Lactate dehydrogenase (LDH), were evaluated after treatment with Tau for 48 h. RESULTS A Significant inhibition in the proliferation of HepG2 was encountered after treatment with Tau in a dose-dependent manner. Moreover, the expression of apoptotic genes p53, Bax, and Caspase-3 exhibited a significant upregulation, while Bcl-2 showed a significant downregulation. These alterations in the expression levels were also confirmed on the protein level. The antioxidant activities of GPx, CAT, and NO were significantly elevated versus untreated control. Also, a significant increase in the expression level of miR-122-5p was observed after treatment with Tau affecting the metabolic activity of HCC cells. Concomitantly, a significant inhibition in ALDOA protein and the hallmark of glycolytic enzymes LDH and Aldolase were observed. CONCLUSIONS These observations showed that taurine inhibits HepG2 cell proliferation and restores the expression of miR-122-5p which inhibits the hallmark glycolytic enzymes and ultimately the metabolic activity of HCC cells. Tau is assumed to be a promising and effective antitumor therapy of HCC.
Collapse
Affiliation(s)
- Asmaa Abdel Nabi
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Shimaa Attia Atta
- Immunology Department, Theodor Bilharz Research Institute, Cairo, Egypt
| | - Eman El-Ahwany
- Immunology Department, Theodor Bilharz Research Institute, Cairo, Egypt
| | - Emad Elzayat
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Hanan Saleh
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
37
|
Abstract
TGR5 (G protein-coupled bile acid receptor 1, GPBAR-1) is a G protein-coupled receptor with seven transmembrane domains and is widely distributed in various organs and tissues. As an important bile acid receptor, TGR5 can be activated by primary and secondary bile acids. Increased expression of TGR5 is a risk factor for polycystic liver disease and hepatobiliary cancer. However, there is evidence that the anti-inflammatory effect of the TGR5 receptor and its regulatory effect on hydrophobic bile acid confer protective effects against most liver diseases. Recent studies have shown that TGR5 receptor activation can alleviate the development of diabetic liver fibrosis, regulate the differentiation of natural killer T cells into NKT10 cells, increase the secretion of anti-inflammatory factors, inhibit the invasion of hepatitis B virus, promote white adipose tissue browning, improve arterial vascular dynamics, maintain tight junctions between bile duct cells, and protect against apoptosis. In portal hypertension, TGR5 receptor activation can inhibit the contraction of hepatic stellate cells and improve intrahepatic microcirculation. In addition, the discovery of the regulatory relationship between the TGR5 receptor and miRNA-26a provides a new direction for further studies of the molecular mechanism underlying the effects of TGR5. In this review, we describe recent findings linking TGR5 to various liver diseases, with a focus on the mechanisms underlying its effects and potential therapeutic implications.
Collapse
Affiliation(s)
- Ke Ma
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dan Tang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chang Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lijin Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
38
|
Stachowicz A, Wiśniewska A, Kuś K, Białas M, Łomnicka M, Totoń-Żurańska J, Kiepura A, Stachyra K, Suski M, Bujak-Giżycka B, Jawień J, Olszanecki R. Diminazene Aceturate Stabilizes Atherosclerotic Plaque and Attenuates Hepatic Steatosis in apoE-Knockout Mice by Influencing Macrophages Polarization and Taurine Biosynthesis. Int J Mol Sci 2021; 22:5861. [PMID: 34070749 PMCID: PMC8199145 DOI: 10.3390/ijms22115861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis and nonalcoholic fatty liver disease are leading causes of morbidity and mortality in the Western countries. The renin-angiotensin system (RAS) with its two main opposing effectors, i.e., angiotensin II (Ang II) and Ang-(1-7), is widely recognized as a major regulator of cardiovascular function and body metabolic processes. Angiotensin-converting enzyme 2 (ACE2) by breaking-down Ang II forms Ang-(1-7) and thus favors Ang-(1-7) actions. Therefore, the aim of our study was to comprehensively evaluate the influence of prolonged treatment with ACE2 activator, diminazene aceturate (DIZE) on the development of atherosclerotic lesions and hepatic steatosis in apoE-/- mice fed a high-fat diet (HFD). We have shown that DIZE stabilized atherosclerotic lesions and attenuated hepatic steatosis in apoE-/- mice fed an HFD. Such effects were associated with decreased total macrophages content and increased α-smooth muscle actin levels in atherosclerotic plaques. Moreover, DIZE changed polarization of macrophages towards increased amount of anti-inflammatory M2 macrophages in the atherosclerotic lesions. Interestingly, the anti-steatotic action of DIZE in the liver was related to the elevated levels of HDL in the plasma, decreased levels of triglycerides, and increased biosynthesis and concentration of taurine in the liver of apoE-/- mice. However, exact molecular mechanisms of both anti-atherosclerotic and anti-steatotic actions of DIZE require further investigations.
Collapse
Affiliation(s)
- Aneta Stachowicz
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Anna Wiśniewska
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Katarzyna Kuś
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Magdalena Białas
- Chair of Pathomorphology, Jagiellonian University Medical College, 31-531 Krakow, Poland;
| | - Magdalena Łomnicka
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Justyna Totoń-Żurańska
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Anna Kiepura
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Kamila Stachyra
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Maciej Suski
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Beata Bujak-Giżycka
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Jacek Jawień
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Rafał Olszanecki
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| |
Collapse
|
39
|
Guizoni DM, Freitas IN, Victorio JA, Possebom IR, Araujo TR, Carneiro EM, Davel AP. Taurine treatment reverses protein malnutrition-induced endothelial dysfunction of the pancreatic vasculature: The role of hydrogen sulfide. Metabolism 2021; 116:154701. [PMID: 33417894 DOI: 10.1016/j.metabol.2021.154701] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/11/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Protein malnutrition in childhood predisposes individuals to vascular and pancreatic endocrine dysfunction, thus increasing the risk of diabetes and hypertension. Because taurine may reduce cardiometabolic risk, we hypothesized that taurine treatment has a beneficial effect on the pancreatic vasculature during protein restriction. METHODS AND RESULTS Weaned mice were fed a normal or a low-protein diet and were treated with or without taurine for 3 months. The lieno-pancreatic artery (LPA) from low-protein diet-treated mice exhibited impaired endothelium-dependent relaxation to acetylcholine that was associated with decreased endothelium-derived hyperpolarization (EDH), hydrogen sulfide (H2S) production, and H2S-synthesizing CBS expression and impaired vasorelaxation to an H2S-donor, NaHS. These changes were prevented by taurine treatment. We compared the effects of taurine with the effects of the direct vasodilator hydralazine and found that both normalized blood pressure and the endothelial vasodilator function of the LPA in the mice fed a protein-restricted diet. However, only taurine restored the CBS expression in the LPA and insulin secretion in response to high glucose. The LPA supplies the pancreas and shares morphometry with the mesenteric resistance artery (MRA). However, in the MRA, low-protein diet-induced endothelial dysfunction is driven by impaired NOS-derived NO with no changes in H2S signaling. CONCLUSIONS The results suggest that taurine protects against protein malnutrition-induced endothelial dysfunction in the LPA by upregulating the CBS-H2S pathway. Considering the importance of the pancreatic vasculature for endocrine islet activity, taurine may be a potential therapy for the vascular and metabolic dysfunction associated with malnutrition and comorbidities.
Collapse
Affiliation(s)
- Daniele M Guizoni
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Israelle N Freitas
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Jamaira A Victorio
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Isabela R Possebom
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Thiago R Araujo
- Obesity and Comorbidities Research Center-OCRC, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Everardo M Carneiro
- Obesity and Comorbidities Research Center-OCRC, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Ana P Davel
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
40
|
Lu YA, Jiang Y, Yang HW, Hwang J, Jeon YJ, Ryu B. Diphlorethohydroxycarmalol Isolated from Ishige okamurae Exerts Vasodilatory Effects via Calcium Signaling and PI3K/Akt/eNOS Pathway. Int J Mol Sci 2021; 22:1610. [PMID: 33562632 PMCID: PMC7914902 DOI: 10.3390/ijms22041610] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Nitric oxide (NO) is released by endothelial cells in the blood vessel wall to enhance vasodilation. Marine polyphenols are known to have protective effects against vascular dysfunction and hypertension. The present study is the first to investigate how diphlorethohydroxycarmalol (DPHC) isolated from Ishige okamurae affects calcium levels, resulting in enhanced vasodilation. We examined calcium modulation with the well-known receptors, acetylcholine receptor (AchR) and vascular endothelial growth factor 2 (VEGFR2), which are related to NO formation, and further confirmed the vasodilatory effect of DPHC. We confirmed that DPHC stimulated NO production by increasing calcium levels and endothelial nitric oxide synthase (eNOS) expression. DPHC affected AchR and VEGFR2 expression, thereby influencing transient calcium intake. Specific antagonists, atropine and SU5416, were used to verify our findings. Furthermore, based on the results of in vivo experiments, we treated Tg(flk:EGFP) transgenic zebrafish with DPHC to confirm its vasodilatory effect. In conclusion, the present study showed that DPHC modulated calcium transit through AchR and VEGFR2, increasing endothelial-dependent NO production. Thus, DPHC, a natural marine component, can efficiently ameliorate cardiovascular diseases by improving vascular function.
Collapse
Affiliation(s)
- Yu An Lu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Yunfei Jiang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Hye-Won Yang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - Jin Hwang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| | - Bomi Ryu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (Y.A.L.); (Y.J.); (H.-W.Y.); (J.H.)
| |
Collapse
|
41
|
Zhang R, Ma WQ, Fu MJ, Li J, Hu CH, Chen Y, Zhou MM, Gao ZJ, He YL. Overview of bile acid signaling in the cardiovascular system. World J Clin Cases 2021; 9:308-320. [PMID: 33521099 PMCID: PMC7812903 DOI: 10.12998/wjcc.v9.i2.308] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Bile acids (BAs) are classically known to play a vital role in the metabolism of lipids and in absorption. It is now well established that BAs act as signaling molecules, activating different receptors (such as farnesoid X receptor, vitamin D receptor, Takeda G-protein-coupled receptor 5, sphingosine-1-phosphate, muscarinic receptors, and big potassium channels) and participating in the regulation of energy homeostasis and lipid and glucose metabolism. In addition, increased BAs can impair cardiovascular function in liver cirrhosis. Approximately 50% of patients with cirrhosis develop cirrhotic cardiomyopathy. Exposure to high concentrations of hydrophobic BAs has been shown to be related to adverse effects with respect to vascular tension, endothelial function, arrhythmias, coronary atherosclerotic heart disease, and heart failure. The BAs in the serum BA pool have relevant through their hydrophobicity, and the lipophilic BAs are more harmful to the heart. Interestingly, ursodeoxycholic acid is a hydrophilic BA, and it is used as a therapeutic drug to reverse and protect the harmful cardiac effects caused by hydrophobic elevated BAs. In order to elucidate the mechanism of BAs and cardiovascular function, abundant experiments have been conducted in vitro and in vivo. The aim of this review was to explore the mechanism of BAs in the cardiovascular system.
Collapse
Affiliation(s)
- Rou Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Wen-Qi Ma
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Meng-Jun Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Chun-Hua Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Yi Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Mi-Mi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Zhi-Jie Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Ying-Li He
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
42
|
Montagnana M, Danese E, Giontella A, Bonafini S, Benati M, Tagetti A, Dalbeni A, Cavarzere P, Gaudino R, Pucci M, Salvagno GL, Antoniazzi F, Lippi G, Maffeis C, Fava C. Circulating Bile Acids Profiles in Obese Children and Adolescents: A Possible Role of Sex, Puberty and Liver Steatosis. Diagnostics (Basel) 2020; 10:diagnostics10110977. [PMID: 33233601 PMCID: PMC7699673 DOI: 10.3390/diagnostics10110977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 01/04/2023] Open
Abstract
Background. Childhood obesity is becoming a major health issue and contributes to increasing the risk of cardiovascular disease in adulthood. Since dysregulated metabolism of bile acids (BAs) plays a role in progression of obesity-related disorders, including steatosis and hypertension, this study aimed to investigate BAs profiles in obese children with and without steatosis and hypertension, as well as exploring the interplay between BAs profile and vascular function. Methods. BAs concentrations were quantified with liquid chromatography-tandem mass spectrometry in 69 overweight/obese children and adolescents (mean age, 11.6 ± 2.5 years; 30 females). Liver steatosis was defined with abdomen ultrasonography, whilst hypertension was defined according to the current European guidelines. Vascular function was assessed with ultrasound technique, by measuring carotid intima media thickness (cIMT) and common carotid artery distensibility (cDC). Results. Total and individual glycine-conjugated BAs concentrations were found to be significantly higher in males compared to females, as well as in pre-pubertal compared to pubertal stage (p < 0.05 for both). No difference in BAs concentration was observed between hypertensive and normotensive subjects. Total BAs and glycine conjugated BAs were significantly higher in participants with steatosis compared to those without (p = 0.004 for both). The values of total glycine-conjugate acids were positively correlated with cDC and this association remained significant in linear regression after adjusting for sex, age, pubertal stage, body mass index and aspartate aminotransferase. Conclusion. The results suggest a possible role of BAs in the pathogenesis of liver and/or vascular damage in children and adolescent. Further studies are hence needed to validate these preliminary findings.
Collapse
Affiliation(s)
- Martina Montagnana
- Section of Clinical Biochemistry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (E.D.); (M.B.); (M.P.); (G.L.S.); (G.L.)
- Correspondence:
| | - Elisa Danese
- Section of Clinical Biochemistry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (E.D.); (M.B.); (M.P.); (G.L.S.); (G.L.)
| | - Alice Giontella
- “General Medicine and Hypertension” Unit, Department of Medicine, University of Verona, 37134 Verona, Italy; (A.G.); (S.B.); (A.T.); (A.D.); (C.F.)
| | - Sara Bonafini
- “General Medicine and Hypertension” Unit, Department of Medicine, University of Verona, 37134 Verona, Italy; (A.G.); (S.B.); (A.T.); (A.D.); (C.F.)
| | - Marco Benati
- Section of Clinical Biochemistry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (E.D.); (M.B.); (M.P.); (G.L.S.); (G.L.)
| | - Angela Tagetti
- “General Medicine and Hypertension” Unit, Department of Medicine, University of Verona, 37134 Verona, Italy; (A.G.); (S.B.); (A.T.); (A.D.); (C.F.)
| | - Andrea Dalbeni
- “General Medicine and Hypertension” Unit, Department of Medicine, University of Verona, 37134 Verona, Italy; (A.G.); (S.B.); (A.T.); (A.D.); (C.F.)
| | - Paolo Cavarzere
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37126 Verona, Italy; (P.C.); (R.G.); (F.A.); (C.M.)
| | - Rossella Gaudino
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37126 Verona, Italy; (P.C.); (R.G.); (F.A.); (C.M.)
| | - Mairi Pucci
- Section of Clinical Biochemistry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (E.D.); (M.B.); (M.P.); (G.L.S.); (G.L.)
| | - Gian Luca Salvagno
- Section of Clinical Biochemistry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (E.D.); (M.B.); (M.P.); (G.L.S.); (G.L.)
| | - Franco Antoniazzi
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37126 Verona, Italy; (P.C.); (R.G.); (F.A.); (C.M.)
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37134 Verona, Italy; (E.D.); (M.B.); (M.P.); (G.L.S.); (G.L.)
| | - Claudio Maffeis
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37126 Verona, Italy; (P.C.); (R.G.); (F.A.); (C.M.)
| | - Cristiano Fava
- “General Medicine and Hypertension” Unit, Department of Medicine, University of Verona, 37134 Verona, Italy; (A.G.); (S.B.); (A.T.); (A.D.); (C.F.)
| |
Collapse
|
43
|
Oyagbemi AA, Bolaji-Alabi FB, Ajibade TO, Adejumobi OA, Ajani OS, Jarikre TA, Omobowale TO, Ola-Davies OE, Soetan KO, Aro AO, Emikpe BO, Saba AB, Adedapo AA, Oyeyemi MO, Nkadimeng SM, Kayoka-Kabongo PN, McGaw LJ, Oguntibeju OO, Yakubu MA. Novel antihypertensive action of rutin is mediated via inhibition of angiotensin converting enzyme/mineralocorticoid receptor/angiotensin 2 type 1 receptor (ATR1) signaling pathways in uninephrectomized hypertensive rats. J Food Biochem 2020; 44:e13534. [PMID: 33089540 DOI: 10.1111/jfbc.13534] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/18/2020] [Accepted: 10/02/2020] [Indexed: 12/01/2022]
Abstract
Hypertension is the most common cardiovascular disease that affects approximately 26% of adult population, worldwide. Rutin is one of the important flavonoids that is consumed in the daily diet, and found in many food items, vegetables, and beverages. Uninephrectomy (UNX) of the left kidney was performed, followed by induction of hypertension. The rats were randomly divided into four groups of 10 rats: group 1-Sham-operated rats; group 2-UNX rats, group 3-UNX-L-NAME (40 mg/kg) plus rutin (100 mg/kg bwt), and groups 4-UNX-L-NAME plus lisinopril (10 mg/kg bwt), orally for 3 weeks. Results revealed significant heightening of arterial pressure and oxidative stress indices, while hypertensive rats treated with rutin had lower expressions of angiotensin converting enzyme (ACE) and mineralocorticoid receptor in uninephrectomized rats. Together, rutin as a novel antihypertensive flavonoid could provide an unimaginable benefits for the management of hypertension through inhibition of angiotensin converting enzyme and mineralocorticoid receptor. PRACTICAL APPLICATIONS: Hypertension has been reported to be the most common cardiovascular disease, affecting approximately 26% of the adult population worldwide with predicted prevalence to increase by 60% by 2025. Recent advances in phytomedicine have shown flavonoids to be very helpful in the treatment of many diseases. Flavonoids have been used in the treatment and management of cardiovascular diseases, obesity and hypertension. The study revealed that rutin, a known flavonoid inhibited angiotensin converting enzyme (ACE), angiotensin 2 type 1 receptor (ATR1), and mineralocorticoid receptor (MCR), comparable to the classic ACE inhibitor, Lisinopril, indicating the novel antihypertensive property of rutin. Therefore, flavonoids such as rutin found in fruits and vegetables could, therefore, serve as an antihypertensive drug regimen. Combining all, functional foods rich in flavonoids could be used as potential therapeutic candidates for managing uninephrectomized hypertensive patients.
Collapse
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Foluso Bolawaye Bolaji-Alabi
- Department of Veterinary Surgery and Radiology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temitayo Olabisi Ajibade
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumuyiwa Abiola Adejumobi
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumide Samuel Ajani
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Theophilus Aghogho Jarikre
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temidayo Olutayo Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Eunice Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Kehinde Olugboyega Soetan
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Abimbola Obemisola Aro
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Benjamin Obukowho Emikpe
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adebowale Benard Saba
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Gauteng, South Africa
| | - Adeolu Alex Adedapo
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Gauteng, South Africa
| | | | - Sanah Malomile Nkadimeng
- Phytomedicine Programme, Department of Paraclinical Science, University of Pretoria Faculty of Veterinary Science, Pretoria, South Africa
| | | | - Lyndy Joy McGaw
- Phytomedicine Programme, Department of Paraclinical Science, University of Pretoria Faculty of Veterinary Science, Pretoria, South Africa
| | - Oluwafemi Omoniyi Oguntibeju
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Momoh Audu Yakubu
- Department of Environmental & Interdisciplinary Sciences, College of Science, Engineering & Technology, Vascular Biology Unit, Center for Cardiovascular Diseases, Texas Southern University, Houston, TX, USA
| |
Collapse
|
44
|
Kumar G, Dey SK, Kundu S. Functional implications of vascular endothelium in regulation of endothelial nitric oxide synthesis to control blood pressure and cardiac functions. Life Sci 2020; 259:118377. [PMID: 32898526 DOI: 10.1016/j.lfs.2020.118377] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 11/29/2022]
Abstract
The endothelium is the innermost vascular lining performing significant roles all over the human body while maintaining the blood pressure at physiological levels. Malfunction of endothelium is thus recognized as a biomarker linked with many vascular diseases including but not limited to atherosclerosis, hypertension and thrombosis. Alternatively, prevention of endothelial malfunctioning or regulating the functions of its associated physiological partners like endothelial nitric oxide synthase can prevent the associated vascular disorders which account for the highest death toll worldwide. While many anti-hypertensive drugs are available commercially, a comprehensive description of the key physiological roles of the endothelium and its regulation by endothelial nitric oxide synthase or vice versa is the need of the hour to understand its contribution in vascular homeostasis. This, in turn, will help in designing new therapeutics targeting endothelial nitric oxide synthase or its interacting partners present in the cellular pool. This review describes the central role of vascular endothelium in the regulation of endothelial nitric oxide synthase while outlining the emerging drug targets present in the vasculature with potential to treat vascular disorders including hypertension.
Collapse
Affiliation(s)
- Gaurav Kumar
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India
| | - Sanjay Kumar Dey
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India; Center for Advanced Biotechnology and Medicine, Rutgers University, NJ 08854, USA
| | - Suman Kundu
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India.
| |
Collapse
|
45
|
Yang J, Wang R, Cheng X, Qu H, Qi J, Li D, Xing Y, Bai Y, Zheng X. The vascular dilatation induced by Hydroxysafflor yellow A (HSYA) on rat mesenteric artery through TRPV4-dependent calcium influx in endothelial cells. JOURNAL OF ETHNOPHARMACOLOGY 2020; 256:112790. [PMID: 32234595 DOI: 10.1016/j.jep.2020.112790] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/20/2020] [Accepted: 03/21/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hydroxysafflor yellow A (HSYA) is the principal constituent of the flowers of Carthamus tinctorius L., a traditional Chinese herbal medicine, which has been used for the treatment of cerebrovascular and cardiovascular diseases due to its property of promoting blood circulation and removing blood stasis. It is dominated in the water extract of Carthamus tinctorius L., which has been used in the clinical treatment for cardiovascular diseases. HSYA exerts a variety of pharmacological efficacy upon the vascular system. However, the underlying mechanisms remain unclear. AIM OF THE STUDY To investigate the vascular dilatation effect of HSYA on rat mesenteric artery (MA) and its potential mechanism. MATERIALS AND METHODS Adult male Wistar rats were applied to the study. Tension studies were conducted to determine the dilatation activity of HSYA against pre-contracted mesenteric arterial (MA) rings by U 46619 and Phenylephrine (PE). The vascular activities were measured with or without incubation with some selective inhibitors, including L-N(ω)-nitro-L-arginine methyl ester (L-NAME, a nitro oxide synthase inhibitor), HC-067047 (a selective TRPV4 antagonist), BaCl2 (a Kir channel blocker), and Indomethacin (Indo, a nonselective cyclooxygenase inhibitor), respectively. Immunocytochemistry, Calcium Imaging, NO Production detection, and Western Blot were also employed to further study the underlying mechanism. RESULTS HSYA reversed the constriction of MAs induced by U 46619 in a manner of concentration dependency, and the dilatation capability was reversed by L-NAME. This effect was significantly dependent on the intactness of MA endothelium, accompanying an increment of NO production in mesenteric arterial endothelium cells. The increment of NO production was reversed by inhibiting the PKA. Also, the expression of p-eNOS was activated by HSYA shown in Western Blot assays. The cells imaging revealed a significant increase and drop of the influx of Ca2+ before and after treatment with HC-067047. CONCLUSIONS These findings suggest that HSYA exerts vessel dilation effect on MAs via a TRPV4-dependent influx of Ca2+ in endothelium cells, PKA-dependent eNOS phosphorylation and NO production mechanism. The present study indicates that HSYA has the potential to be a future candidate for the treatment of hypertension.
Collapse
Affiliation(s)
- Jianfeng Yang
- College of Pharmacy, Harbin Medical University - Daqing, Daqing, 163319, China.
| | - Rui Wang
- College of Pharmacy, Harbin Medical University - Daqing, Daqing, 163319, China.
| | - Xiaohan Cheng
- College of Pharmacy, Harbin Medical University - Daqing, Daqing, 163319, China.
| | - HuiChong Qu
- College of Pharmacy, Harbin Medical University - Daqing, Daqing, 163319, China.
| | - Jing Qi
- College of Basic Medicine, Harbin Medical University - Daqing, Daqing, Heilongjiang, 163319, PR China.
| | - Dan Li
- College of Pharmacy, Harbin Medical University - Daqing, Daqing, 163319, China.
| | - Yan Xing
- College of Basic Medicine, Harbin Medical University - Daqing, Daqing, Heilongjiang, 163319, PR China.
| | - Yuhua Bai
- College of Pharmacy, Harbin Medical University - Daqing, Daqing, 163319, China.
| | - Xiaodong Zheng
- College of Basic Medicine, Harbin Medical University - Daqing, Daqing, Heilongjiang, 163319, PR China.
| |
Collapse
|
46
|
Mensegue MF, Burgueño AL, Tellechea ML. Perinatal taurine exerts a hypotensive effect in male spontaneously hypertensive rats and down-regulates endothelial oxide nitric synthase in the aortic arch. Clin Exp Pharmacol Physiol 2020; 47:780-789. [PMID: 31958174 DOI: 10.1111/1440-1681.13260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/17/2019] [Accepted: 01/13/2020] [Indexed: 12/25/2022]
Abstract
Essential hypertension is considered to be a result of the interaction between genetic and environmental factors, including perinatal factors. Different advantageous perinatal factors proved to have beneficial long-lasting effects against an abnormal genetic background. Taurine is a ubiquitous sulphur-containing amino acid present in foods such as seafood. The antihypertensive effects of taurine have been reported in experimental studies and in human hypertension. We aimed to investigate the effects of perinatal treatment with taurine in spontaneously hypertensive rats (SHR), a known model of genetic hypertension. Female SHR were administered with taurine (3 g/L) during gestation and lactation (SHR-TAU). Untreated SHR and Wistar-Kyoto rats (WKY) were used as controls. Long-lasting effects in offspring were investigated. Addition of taurine to the mother's drinking water reduced blood pressure in adult offspring. No differences were observed in cardiac hypertrophy. Findings on morphometric evaluations suggest that perinatal treatment with taurine would be partially effective in improving structural alterations of the aorta. Modifications in gene expression of Bcl-2 family members and upregulation of endothelial nitric oxide synthase in the aorta of 22-week-old male offspring were found. No differences were observed on relative telomere length in different cardiovascular tissues between SHR and SHR-TAU. Altogether results suggest that taurine programming, albeit sex specific, is associated with gene expression changes which ultimately may lead to improvement of aortic remodelling and enhanced endothelial function because of augmented nitric oxide (NO) production.
Collapse
Affiliation(s)
- Melisa F Mensegue
- Institute of Medical Research A. Lanari, University of Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.,Department of Molecular Genetics and Biology of Complex Diseases, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Adriana L Burgueño
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pontificia Universidad Católica Argentina, Ciudad Autónoma de Buenos Aires, Argentina
| | - Mariana L Tellechea
- Institute of Medical Research A. Lanari, University of Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.,Department of Molecular Genetics and Biology of Complex Diseases, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|