1
|
Bibha K, Akhigbe TM, Hamed MA, Akhigbe RE. Metabolic Derangement by Arsenic: a Review of the Mechanisms. Biol Trace Elem Res 2024; 202:1972-1982. [PMID: 37670201 DOI: 10.1007/s12011-023-03828-4] [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: 05/22/2023] [Accepted: 08/21/2023] [Indexed: 09/07/2023]
Abstract
Studies have implicated arsenic exposure in various pathological conditions, including metabolic disorders, which have become a global phenomenon, affecting developed, developing, and under-developed nations. Despite the huge risks associated with arsenic exposure, humans remain constantly exposed to it, especially through the consumption of contaminated water and food. This present study provides an in-depth insight into the mechanistic pathways involved in the metabolic derangement by arsenic. Compelling pieces of evidence demonstrate that arsenic induces metabolic disorders via multiple pathways. Apart from the initiation of oxidative stress and inflammation, arsenic prevents the phosphorylation of Akt at Ser473 and Thr308, leading to the inhibition of PDK-1/Akt insulin signaling, thereby reducing GLUT4 translocation through the activation of Nrf2. Also, arsenic downregulates mitochondrial deacetylase Sirt3, decreasing the ability of its associated transcription factor, FOXO3a, to bind to the agents that support the genes for manganese superoxide dismutase and PPARg co-activator (PGC)-1a. In addition, arsenic activates MAPKs, modulates p53/ Bcl-2 signaling, suppresses Mdm-2 and PARP, activates NLRP3 inflammasome and caspase-mediated apoptosis, and induces ER stress, and ox-mtDNA-dependent mitophagy and autophagy. More so, arsenic alters lipid metabolism by decreasing the presence of 3-hydroxy-e-methylglutaryl-CoA synthase 1 and carnitine O-octanoyl transferase (Crot) and increasing the presence of fatty acid-binding protein-3 mRNA. Furthermore, arsenic promotes atherosclerosis by inducing endothelial damage. This cascade of pathophysiological events promotes metabolic derangement. Although the pieces of evidence provided by this study are convincing, future studies evaluating the involvement of other likely mechanisms are important. Also, epidemiological studies might be necessary for the translation of most of the findings in animal models to humans.
Collapse
Affiliation(s)
- K Bibha
- Department of Zoology, Magadh Mahila College, Patna University, Patna, India
| | - T M Akhigbe
- Breeding and Plant Genetics Unit, Department of Agronomy, Osun State University, Osogbo, Osun State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - M A Hamed
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
- Department of Medical Laboratory Science, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
- The Brainwill Laboratory, Osogbo, Osun State, Nigeria
| | - R E Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria.
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria.
| |
Collapse
|
2
|
Huang Q, Liu Y, Cheng Y, Jia F, Pu C, Yan Q, Chang Z, Liao P, Ma D, Xu L, Zhang H, Lu Y, Liu X, Liu K. High-throughput quantitation of serological dimethylarginines by LC/MS/MS: Potential cardiovascular biomarkers for rheumatoid arthritis. J Pharm Biomed Anal 2023; 232:115336. [PMID: 37159983 DOI: 10.1016/j.jpba.2023.115336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 05/11/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by systemic inflammation of the joints and extra-articular tissues. The incidence of cardiovascular disease (CVD) remains the main cause of morbidity and mortality in patients with RA. Despite the development of new therapeutics targeting the articular manifestations, the relief of the cardiovascular burden is still an unmet medical need during the management of RA. So, the early prognosis of RA-associated CVD plays a crucial role in improving the clinical outcomes of RA patients. Recently, circulating dimethylarginines have gained attention as potential biomarkers for CVDs. Here, we present the development and validation of a high-throughput liquid chromatography-tandem mass spectrometric (LC/MS/MS) method for simultaneous quantification of creatinine, arginine, and dimethylarginines in human serum within 2 mins by isotope dilution mass spectrometry. This method employed a protein precipitation method for rapid sample preparation, trichloroacetic acid (TCA)-based ion pairing chromatography for fast analyte separation, and multiple reaction monitoring (MRM) with stable isotope-labeled internal standards (ISs) for simultaneous quantitation. To assure the quality, our method was validated against the FDA guidelines for lower limit of quantitation (0.2 µM), linearity (square of coefficient correlation>0.99), precision (intra-&inter-assay imprecision < 10 %), accuracy (intra-&inter-assay inaccuracy < 10 %), sample preparation recovery (recovery ≥ 90 %), stability (instability < 10 %), matrix effect (signal suppression < 55 %), and carryover ( < 0.01 %). Afterward, we applied the validated method to a retrospective cross-sectional study. We aimed to evaluate the utility of serological dimethylarginines as potential cardiovascular biomarkers in the development of RA-associated CVD. Our results revealed that the serological ratio of asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA), an indicator of physiological arginine methylation status, was significantly elevated in patients with RA. This finding might provide value in detecting CVD to improve clinical outcomes in RA management.
Collapse
Affiliation(s)
- Qianyang Huang
- Shenzhen Kanghua Juntai Biotech Co. Ltd., B 215, Unit No.7, Shahe Rd W, Nanshan, Shenzhen, Guangdong Province 518063, China
| | - Ying Liu
- Physical Examination Center, Yuebei People's Hospital, Wu Jiang Qu, Shao Guan Shi, Guangzhou 512027, China
| | - Yuna Cheng
- Shanghai Center for Disease Control and Prevention, Chang Ning Qu, Shang Hai Shi, 200051, China
| | - Fujian Jia
- Shenzhen Kanghua Juntai Biotech Co. Ltd., B 215, Unit No.7, Shahe Rd W, Nanshan, Shenzhen, Guangdong Province 518063, China
| | - Chunchao Pu
- Shenzhen Kanghua Juntai Biotech Co. Ltd., B 215, Unit No.7, Shahe Rd W, Nanshan, Shenzhen, Guangdong Province 518063, China
| | - Qinghua Yan
- Shanghai Center for Disease Control and Prevention, Chang Ning Qu, Shang Hai Shi, 200051, China
| | - Zhaoyu Chang
- Shanghai Center for Disease Control and Prevention, Chang Ning Qu, Shang Hai Shi, 200051, China
| | - Ping Liao
- Shanghai Center for Disease Control and Prevention, Chang Ning Qu, Shang Hai Shi, 200051, China
| | - Dandan Ma
- Shenzhen Kanghua Juntai Biotech Co. Ltd., B 215, Unit No.7, Shahe Rd W, Nanshan, Shenzhen, Guangdong Province 518063, China
| | - Lei Xu
- Shenzhen Kanghua Juntai Biotech Co. Ltd., B 215, Unit No.7, Shahe Rd W, Nanshan, Shenzhen, Guangdong Province 518063, China
| | - Hua Zhang
- Shenzhen Kanghua Juntai Biotech Co. Ltd., B 215, Unit No.7, Shahe Rd W, Nanshan, Shenzhen, Guangdong Province 518063, China
| | - Yuanzhi Lu
- Department of Pathology, The First Affiliated Hospital of Jinan University, Tianhe Qu, Guangzhou 510632, China
| | - Xin Liu
- Shenzhen Kanghua Juntai Biotech Co. Ltd., B 215, Unit No.7, Shahe Rd W, Nanshan, Shenzhen, Guangdong Province 518063, China.
| | - Kang Liu
- Shenzhen Kanghua Juntai Biotech Co. Ltd., B 215, Unit No.7, Shahe Rd W, Nanshan, Shenzhen, Guangdong Province 518063, China.
| |
Collapse
|
3
|
Sun Y, Gao Y, Zhou L, Lu Y, Zong Y, Zhu H, Tang Y, Zheng F, Sun Y, Li Y. A multi-target protective effect of Danggui-Shaoyao-San on the vascular endothelium of atherosclerotic mice. BMC Complement Med Ther 2023; 23:60. [PMID: 36803348 PMCID: PMC9940384 DOI: 10.1186/s12906-023-03883-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/11/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Atherosclerosis (AS) is a chronic disease characterized by abnormal blood lipid metabolism, inflammation and vascular endothelial injury. Vascular endothelial injury is the initial stage during the occurrence of AS. However, the function and mechanism of anti-AS are not well characterized. Danggui-Shaoyao-San (DGSY) is a classic Traditional Chinese Medicine (TCM) prescription for the treatment of gynecological diseases, and has been widely used in the treatment of AS in recent years. METHODS ApoE-/- atherosclerosis male mice were established by feeding with high-fat diet, and then randomly divided into three groups: Atherosclerosis group (AS), Danggui-Shaoyao-San group (DGSY), and Atorvastatin calcium group (X). The mice were administered with the drugs for 16 weeks. Pathological changes in aortic vessels were examined by staining with Oil red O, Masson and hematoxylin-eosin. In addition, blood lipids were analyzed. The level of IL-6 and IL-8 in aortic vessels were detected by ELISA and the expression of ICAM-1 and VCAM-1 in the aortic vascular endothelium were measured by Immunohistochemical. The mRNA expression of interα5β1/c-Abl/YAP in the aortic vessels were measured by Real-time quantitative PCR and location of expression was assessed by immunofluorescence. RESULTS DGSY can significantly reduce the content of TC,TG and LDL-C and increase the level of HDL-C in the serum, reduce the plaque area and inhibit the concentration of IL-6 and IL-8, down-regulate the expression of IVAM-1,VCAM-1 and interα5β1/ c-Abl/YAP in the aortic vessels. CONCLUSIONS Collectively, DGSY can alleviate vascular endothelium damage and delay the occurrence of AS, and the underlying mechanism may be related to the multi-target protective of DGSY.
Collapse
Affiliation(s)
- Yuemeng Sun
- grid.24695.3c0000 0001 1431 9176School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yushan Gao
- grid.24695.3c0000 0001 1431 9176School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Lu Zhou
- grid.24695.3c0000 0001 1431 9176School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yixing Lu
- grid.24695.3c0000 0001 1431 9176School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yulin Zong
- grid.24695.3c0000 0001 1431 9176School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Haoyu Zhu
- grid.24695.3c0000 0001 1431 9176School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yang Tang
- grid.24695.3c0000 0001 1431 9176School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Fengjie Zheng
- grid.24695.3c0000 0001 1431 9176School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yan Sun
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yuhang Li
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
4
|
Chen CH, Guo BC, Hu PA, Lee HT, Hu HY, Hsu MC, Chen WH, Lee TS. Ractopamine at legal residue dosage accelerates atherosclerosis by inducing endothelial dysfunction and promoting macrophage foam cell formation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 313:120080. [PMID: 36057326 DOI: 10.1016/j.envpol.2022.120080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/08/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Ractopamine, a synthetic β-adrenoreceptor agonist, is used as an animal feed additive to increase food conversion efficiency and accelerate lean mass accretion in farmed animals. The U.S. Food and Drug Administration claimed that ingesting products containing ractopamine residues at legal dosages might not cause short-term harm to human health. However, the effect of ractopamine on chronic inflammatory diseases and atherosclerosis is unclear. Therefore, we investigated the effects of ractopamine on atherosclerosis and its action mechanism in apolipoprotein E-null (apoe-/-) mice and human endothelial cells (ECs) and macrophages. Daily treatment with ractopamine for four weeks increased the body weight and the weight of brown adipose tissues and gastrocnemius muscles. However, it decreased the weight of white adipose tissues in apoe-/- mice. Additionally, ractopamine exacerbated hyperlipidemia and systemic inflammation, deregulated aortic cholesterol metabolism and inflammation, and accelerated atherosclerosis. In ECs, ractopamine treatment induced endothelial dysfunction and increased monocyte adhesion and transmigration across ECs. In macrophages, ractopamine dysregulated cholesterol metabolism by increasing oxidized low-density lipoprotein (oxLDL) internalization and decreasing reverse cholesterol transporters, increasing oxLDL-induced lipid accumulation. Collectively, our findings revealed that ractopamine induces EC dysfunction and deregulated cholesterol metabolism of macrophages, which ultimately accelerates atherosclerosis progression.
Collapse
Affiliation(s)
- Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bei-Chia Guo
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-An Hu
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsueh-Te Lee
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsuan-Yun Hu
- International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Man-Chen Hsu
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Hua Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
5
|
Sabbir MG, Wigle JT, Taylor CG, Zahradka P. Growth State-Dependent Expression of Arachidonate Lipoxygenases in the Human Endothelial Cell Line EA.hy926. Cells 2022; 11:cells11162478. [PMID: 36010555 PMCID: PMC9406857 DOI: 10.3390/cells11162478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 11/30/2022] Open
Abstract
Endothelial cells regulate vascular homeostasis through the secretion of various paracrine molecules, including bioactive lipids, but little is known regarding the enzymes responsible for generating these lipids under either physiological or pathophysiological conditions. Arachidonate lipoxygenase (ALOX) expression was therefore investigated in confluent and nonconfluent EA.h926 endothelial cells, which represent the normal quiescent and proliferative states, respectively. mRNAs for ALOX15, ALOX15B, and ALOXE3 were detected in EA.hy926 cells, with the highest levels present in confluent cells compared to nonconfluent cells. In contrast, ALOX5, ALOX12, and ALOX12B mRNAs were not detected. At the protein level, only ALOX15B and ALOXE3 were detected but only in confluent cells. ALOXE3 was also observed in confluent human umbilical artery endothelial cells (HUAEC), indicating that its expression, although previously unreported, may be a general feature of endothelial cells. Exposure to laminar flow further increased ALOXE3 levels in EA.hy926 cells and HUAECs. The evidence obtained in this study indicates that proliferative status and shear stress are both important factors that mediate endothelial ALOX gene expression. The presence of ALOX15B and ALOXE3 exclusively in quiescent human endothelial cells suggests their activity likely contributes to the maintenance of a healthy endothelium.
Collapse
Affiliation(s)
- Mohammad G. Sabbir
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Jeffrey T. Wigle
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Carla G. Taylor
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Peter Zahradka
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Correspondence: ; Tel.: +204-235-3507; Fax: +204-237-4018
| |
Collapse
|
6
|
Xu L, Xu C, Lin X, Lu H, Cai Y. Interference with lysophosphatidic acid receptor 5 ameliorates oxidized low-density lipoprotein-induced human umbilical vein endothelial cell injury by inactivating NOD-like receptor family, pyrin domain containing 3 inflammasome signaling. Bioengineered 2021; 12:8089-8099. [PMID: 34662522 PMCID: PMC8806909 DOI: 10.1080/21655979.2021.1983975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
Endothelial cell damage induced by oxidized low-density lipoprotein (ox-LDL) plays an important role in the pathogenesis of atherosclerosis (AS). We aimed to explore the effects of lysophosphatidic acid receptor 5 (LPAR5) on ox-LDL-induced damage of human umbilical vein endothelial cells (HUVECs). After HUVECs exposed to ox-LDL, LPAR5 expression was detected by RT-qPCR and western blotting. Then, LPAR5 was silenced and cell viability was determined with a CCK-8 assay. ELISA was employed to analyze the contents of inflammatory factors. The levels of oxidative stress markers were examined by kits. The expression of proteins related to endothelium function, including CD31, α-SMA, iNOS and eNOS, was evaluated with RT-qPCR and western blotting. Additionally, the effects of LPAR5 deletion on the NLRP3 inflammasome signaling in HUVECs under ox-LDL condition were assessed by determining NLRP3, caspase-1 and ASC expression. Afterward, NLRP3 agonist MSU was adopted for exploring the regulation of LPAR5 on NLRP3 inflammasome signaling in ox-LDL HUVECs injury. Results revealed that ox-LDL led to a significant upregulation in LPAR5 expression. NLRP3 knockdown enhanced cell viability, inhibited inflammation and oxidative stress in HUVECs after ox-LDL exposure. Besides, the expression of CD31 and eNOS was increased while that of α-SMA and iNOS was decreased after LPAR5 silencing. Moreover, interference with LPAR5 remarkably downregulated NLRP3, caspase-1 and ASC expression. Furthermore, MSU addition partially abrogated the inhibitory effects of LPAR5 deletion on the inflammation, oxidative stress and endothelium dysfunction of HUVECs. To conclude, we demonstrated that LPAR5 silencing alleviates ox-LDL-induced HUVECs injury by inhibiting NLRP3 inflammasome signaling.
Collapse
Affiliation(s)
- Ling Xu
- Department of Cardiovascular Medicine, The Second Attached Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Chaoxiang Xu
- Department of Cardiovascular Medicine, The Second Attached Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xiaoxin Lin
- Department of Cardiovascular Medicine, The Second Attached Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Huiyao Lu
- Department of Cardiovascular Medicine, The Second Attached Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yinlian Cai
- Department of Cardiovascular Medicine, The Second Attached Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
7
|
Lee TS, Lu TM, Chen CH, Guo BC, Hsu CP. Hyperuricemia induces endothelial dysfunction and accelerates atherosclerosis by disturbing the asymmetric dimethylarginine/dimethylarginine dimethylaminotransferase 2 pathway. Redox Biol 2021; 46:102108. [PMID: 34438260 PMCID: PMC8390558 DOI: 10.1016/j.redox.2021.102108] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/28/2022] Open
Abstract
Hyperuricemia is closely associated with the mobility and mortality of patients with cardiovascular diseases. However, how hyperuricemia accelerates atherosclerosis progression is not well understood. The balance between asymmetric dimethylarginine (ADMA) and dimethylarginine dimethylaminotransferases (DDAHs) is crucial to regulate vascular homeostasis. Therefore, we investigated the role of the ADMA/DDAH pathway in hyperuricemia-induced endothelial dysfunction and atherosclerosis and the underlying molecular mechanisms in endothelial cells (ECs) and apolipoprotein E-knockout (apoe-/-) mice. Our results demonstrated that uric acid at pathological concentrations increased the intracellular levels of ADMA and downregulated DDAH-2 expression without affecting DDAH-1 expression. Excess uric acid also reduced NO bioavailability and increased monocyte adhesion to ECs, which were abolished by using the antioxidant N-acetylcysteine, the nicotinamide adenine dinucleotide phosphate oxidase inhibitor apocynin, or DDAH-2 overexpression. In apoe-/- mice, treatment with oxonic acid, a uricase inhibitor, increased the circulating level of uric acid, cholesterol, and lipid peroxidation; exacerbated systemic and aortic inflammation; and worsened atherosclerosis compared with vehicle-treated apoe-/- mice. Furthermore, oxonic acid-treated apoe-/- mice exhibited elevated ADMA plasma level and downregulated aortic expression of DDAH-2 protein. Notably, DDAH-2 overexpression in the ECs of apoe-/- mice prevented hyperuricemia-induced deleterious effects from influencing ADMA production, lipid peroxidation, inflammation, and atherosclerosis. Collectively, our findings suggest that hyperuricemia disturbs the balance of the ADMA/DDAH-2 axis, results in EC dysfunction, and, consequently, accelerates atherosclerosis.
Collapse
Affiliation(s)
- Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Tse-Min Lu
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bei-Chia Guo
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chiao-Po Hsu
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
8
|
Arlouskaya Y, Sawicka A, Głowala M, Giebułtowicz J, Korytowska N, Tałałaj M, Nowicka G, Wrzosek M. Asymmetric Dimethylarginine (ADMA) and Symmetric Dimethylarginine (SDMA) Concentrations in Patients with Obesity and the Risk of Obstructive Sleep Apnea (OSA). J Clin Med 2019; 8:jcm8060897. [PMID: 31234586 PMCID: PMC6616493 DOI: 10.3390/jcm8060897] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 12/25/2022] Open
Abstract
Asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) are endogenous inhibitors of nitric oxide (NO) synthesis, and play a critical role in the process of endothelial dysfunction, and are considered markers of oxidative stress. The aim of the present study was to explore relationships between ADMA and/or SDMA and the occurrence of OSA in obese patients as well as the effect of the endothelial nitric oxide synthase (eNOS) gene polymorphism, which may modify the influence of ADMA or SDMA on NO production. A total of 518 unrelated obese subjects were included in this study. Body weight, height and blood pressure were measured and data on self-reported smoking status were collected. Obstructive sleep apnea (OSA) was assessed by the apnea hypopnea index (AHI). Blood samples were collected to measure serum concentrations of glucose, total cholesterol, LDL-cholesterol, HDL-cholesterol, triglycerides, creatinine, HbA1c (%), folic acid, vitamin B12, C-reactive protein (CRP), aspartate aminotransferase (ASP), alanine aminotransferase (ALT) and IL-6 by routine methods. The NOS3 gene G894T and 4a/4b polymorphisms were detected by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis. ADMA, SDMA and arginine concentrations were assessed simultaneously using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) method. Adjusted multivariate logistic regression analysis showed a significant association between the occurrence of OSA and high serum ADMA levels, BMI above 40, age > 43 years, hypertension and male sex. Heterozygotes for the G894T eNOS polymorphism have the lowest serum concentrations of ADMA and SDMA, while no effect of the 4a/4b variants was observed. The results indicate that OSA in obese individuals can coexist with high ADMA levels, which appear as a potential OSA predictor.
Collapse
Affiliation(s)
- Yana Arlouskaya
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy and Laboratory of Biochemistry and Clinical Chemistry at the Preclinical Research Center, Medical University of Warsaw, 02-097 Warsaw, Poland.
| | - Ada Sawicka
- Department of Family Medicine, Internal Medicine and Metabolic Bone Diseases, Medical Centre of Postgraduate Education, 00-416 Warsaw, Poland.
| | - Marek Głowala
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy and Laboratory of Biochemistry and Clinical Chemistry at the Preclinical Research Center, Medical University of Warsaw, 02-097 Warsaw, Poland.
| | - Joanna Giebułtowicz
- Department of Bioanalysis and Drug Analysis, Medical University of Warsaw, 02-097 Warsaw, Poland.
| | - Natalia Korytowska
- Department of Bioanalysis and Drug Analysis, Medical University of Warsaw, 02-097 Warsaw, Poland.
| | - Marek Tałałaj
- Department of Family Medicine, Internal Medicine and Metabolic Bone Diseases, Medical Centre of Postgraduate Education, 00-416 Warsaw, Poland.
| | - Grażyna Nowicka
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy and Laboratory of Biochemistry and Clinical Chemistry at the Preclinical Research Center, Medical University of Warsaw, 02-097 Warsaw, Poland.
| | - Małgorzata Wrzosek
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy and Laboratory of Biochemistry and Clinical Chemistry at the Preclinical Research Center, Medical University of Warsaw, 02-097 Warsaw, Poland.
| |
Collapse
|
9
|
Babic M, Schuchardt M, Tölle M, van der Giet M. In times of tobacco-free nicotine consumption: The influence of nicotine on vascular calcification. Eur J Clin Invest 2019; 49:e13077. [PMID: 30721530 DOI: 10.1111/eci.13077] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 01/11/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Smoking remains the most important avoidable cause of global mortality. Even though the number of cigarette smokers declines in first world countries, the uses of alternative nicotine delivery products increase and may even surpass the sells of cigarettes. In this light, the explicit role of nicotine in the development of cardiovascular diseases should be elucidated. OBJECTIVES This narrative review attempts to connect current literature about possible effects of nicotine on the environment of the vasculature to the pathogenesis of vascular calcification, focusing on the tunica media of the vessel wall. METHODS For this review, papers found on Pubmed and Medline until December 2018 by searching for the keywords nicotine, vascular calcification, oxidative stress, osteoblastic transdifferentiation and matrix degradation were considered. RESULTS Nicotine creates an environment that probably facilitates and maybe even induces osteogenic transdifferentiation of VSMC by inflammation, endothelial dysfunction and reactive oxygen species. This process is believed to be a key event in calcification of the tunica media of the vessel wall. Furthermore, nicotine could lead to the formation of nucleation sites for hydroxyapatite by facilitating matrix vesicles and extracellular matrix degradation. CONCLUSIONS There is a growing body of evidence implicating that nicotine alone could impair vascular function and lead to vascular calcification. Further research is necessary to elucidate the explicit influence of nicotine on arteriosclerosis.
Collapse
Affiliation(s)
- Milen Babic
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mirjam Schuchardt
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus Tölle
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus van der Giet
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
10
|
Li B, He J, Lv H, Liu Y, Lv X, Zhang C, Zhu Y, Ai D. c-Abl regulates YAPY357 phosphorylation to activate endothelial atherogenic responses to disturbed flow. J Clin Invest 2019; 129:1167-1179. [PMID: 30629551 DOI: 10.1172/jci122440] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 01/03/2019] [Indexed: 12/26/2022] Open
Abstract
Local flow patterns determine the uneven distribution of atherosclerotic lesions. This research aims to elucidate the mechanism of regulation of nuclear translocation of Yes-associated protein (YAP) under oscillatory shear stress (OSS) in the atheroprone phenotype of endothelial cells (ECs). We report here that OSS led to tyrosine phosphorylation and strong, continuous nuclear translocation of YAP in ECs that is dependent on integrin α5β1 activation. YAP overexpression in ECs blunted the anti-atheroprone effect of an integrin α5β1-blocking peptide (ATN161) in Apoe-/- mice. Activation of integrin α5β1 induced tyrosine, but not serine, phosphorylation of YAP in ECs. Blockage of integrin α5β1 with ATN161 abolished the phosphorylation of YAP at Y357 induced by OSS. Mechanistic studies showed that c-Abl inhibitor attenuated the integrin α5β1-induced YAP tyrosine phosphorylation. Furthermore, the phosphorylation of c-Abl and YAPY357 was significantly increased in ECs in atherosclerotic vessels of mice and in human plaques versus normal vessels. Finally, bosutinib, a tyrosine kinase inhibitor, markedly reduced the level of YAPY357 and the development of atherosclerosis in Apoe-/- mice. The c-Abl/YAPY357 pathway serves as a mechanism for the activation of integrin α5β1 and the atherogenic phenotype of ECs in response to OSS, and provides a potential therapeutic strategy for atherogenesis.
Collapse
|
11
|
Wang F, Li G, Guan X, Han Z, Yu X, You Q. Overexpression of eNOS decrease tissue factor (TF) level in CD34+ cells exhibit increased antithrombogenic property in small caliber vascular graft. THE JOURNAL OF CARDIOVASCULAR SURGERY 2019; 60:136-143. [DOI: 10.23736/s0021-9509.18.08951-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
12
|
Geng J, Xu H, Yu X, Xu G, Cao H, Lin G, Sui D. Rosuvastatin protects against oxidized low‑density lipoprotein‑induced endothelial cell injury of atherosclerosis in vitro. Mol Med Rep 2018; 19:432-440. [PMID: 30483737 PMCID: PMC6297788 DOI: 10.3892/mmr.2018.9666] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis‑induced cardiovascular diseases (CVDs) are accompanied by substantial morbidity and mortality. The loss and injury of endothelial cells is the primary cause of atherosclerosis. Rosuvastatin is an alternative agent used to reduce the risk of cardiovascular disease. Subsequently, the present study aimed to investigate the protective effects of rosuvastatin on oxidized‑low‑density lipoprotein (ox‑LDL)‑induced human umbilical vein endothelial cell (HUVEC) injury. The viability of ox‑LDL‑cultured HUVECs with or without rosuvastatin (0.01, 0.1 and 1 µmol/l) pretreatment, and pretreatment at different time points (3, 6, 12 and 24 h) was determined using an MTT assay. Morphological changes and the extent of apoptosis were detected; the anti‑oxidase activity, including superoxide dismutase (SOD) and catalase (CAT), was examined, and the contents of malondiahdehyde (MDA) and nitric oxide (NO) were measured. The phosphorylation levels of endothelial nitric oxide synthase (eNOS), protein kinase B (Akt) and phosphoinositide 3 kinase (PI3K) were detected using western blot analysis. The results demonstrated that pretreatment with 0.01‑1 µmol/l rosuvastatin decreased cell apoptosis caused by ox‑LDL. Notably, pretreatment with 1 µmol/l rosuvastatin for >12 h increased cell viability. Additionally, DAPI staining revealed that rosuvastatin inhibited HUVEC apoptosis. Rosuvastatin treatment also resulted in increased SOD and CAT activities and decreased MDA content in ox‑LDL‑stimulated HUVECs. Furthermore, pretreatment with 0.01‑1 µmol/l rosuvastatin significantly increased` the NO content compared with HUVECs treated with ox‑LDL alone. Western blot analyses demonstrated that rosuvastatin upregulated the phosphorylation of eNOS, Akt and PI3K. These findings indicated that rosuvastatin could protect HUVECs against ox‑LDL‑induced injury through its anti‑oxidant effect and its ability to upregulate the expression of vascular endotheliocyte‑protecting factors.
Collapse
Affiliation(s)
- Jianan Geng
- Department of Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Huali Xu
- Department of Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaofeng Yu
- Department of Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guoliang Xu
- Department of Cardiovascular Medicine, The Eastern Division of First Hospital, Jilin University, Changchun, Jilin 130031, P.R. China
| | - Hongyan Cao
- Department of Cardiovascular Medicine, The Eastern Division of First Hospital, Jilin University, Changchun, Jilin 130031, P.R. China
| | - Guangzhu Lin
- Department of Cardiovascular Medicine, The Eastern Division of First Hospital, Jilin University, Changchun, Jilin 130031, P.R. China
| | - Dayun Sui
- Department of Pharmacology, College of Pharmacy, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
13
|
Niu H, Wei Z, Zhang Y, He J, Jia D. Atorvastatin improves coronary flow and endothelial function in patients with coronary slow flow. Exp Ther Med 2017; 15:904-908. [PMID: 29399097 PMCID: PMC5772870 DOI: 10.3892/etm.2017.5484] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/31/2017] [Indexed: 01/16/2023] Open
Abstract
The underlying mechanisms behind the effect of atorvastatin on patients with coronary slow flow (CSF) remain largely unknown. To investigate the possible underlying molecular mechanisms 108 patients were divided into atorvastatin group and control group. Coronary flow was quantified according to corrected TIMI frame count (CTFC). Serum high sensitivity C-reactive protein (hs-CRP), lipids, ET-1, interleukin (IL)-6, NO, circulating endothelial progenitor cell (cEPC) count, adhesion, migration and proliferation were measured in pretreatment and post-treatment. After respective treatment, the atorvastatin group had significantly decreased levels of TC, TG, LDL-C, hs-CRP, ET-1 and IL-6 and increased NO compared to the control group. The atorvastatin group had a more significant improvement of CTFC, effective rate, cEPC number, EPC adhesion, migration and proliferation compared to the control group. In conclusion, atorvastatin can be used in treatment of CSF by suppressing inflammation and improving endothelial function.
Collapse
Affiliation(s)
- Hongmei Niu
- Department of Cardiovascular Medicine, Shandong Provincial Third Hospital, Jinan, Shandong 250000, P.R. China
| | - Zhenzhen Wei
- Department of Cardiovascular Medicine, The First People's Hospital of Jinan, Jinan, Shandong 250000, P.R. China
| | - Yanling Zhang
- Department of Cardiovascular Medicine, The First People's Hospital of Jinan, Jinan, Shandong 250000, P.R. China
| | - Jian He
- Digestive Disease Department of Internal Medicine, The First People's Hospital of Jinan, Jinan, Shandong 250000, P.R. China
| | - Danyan Jia
- Jinan First Aid Center, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
14
|
SCF-KIT signaling induces endothelin-3 synthesis and secretion: Thereby activates and regulates endothelin-B-receptor for generating temporally- and spatially-precise nitric oxide to modulate SCF- and or KIT-expressing cell functions. PLoS One 2017; 12:e0184154. [PMID: 28880927 PMCID: PMC5589172 DOI: 10.1371/journal.pone.0184154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/19/2017] [Indexed: 01/11/2023] Open
Abstract
We demonstrate that SCF-KIT signaling induces synthesis and secretion of endothelin-3 (ET3) in human umbilical vein endothelial cells and melanoma cells in vitro, gastrointestinal stromal tumors, human sun-exposed skin, and myenteric plexus of human colon post-fasting in vivo. This is the first report of a physiological mechanism of ET3 induction. Integrating our finding with supporting data from literature leads us to discover a previously unreported pathway of nitric oxide (NO) generation derived from physiological endothelial NO synthase (eNOS) or neuronal NOS (nNOS) activation (referred to as the KIT-ET3-NO pathway). It involves: (1) SCF-expressing cells communicate with neighboring KIT-expressing cells directly or indirectly (cleaved soluble SCF). (2) SCF-KIT signaling induces timely local ET3 synthesis and secretion. (3) ET3 binds to ETBR on both sides of intercellular space. (4) ET3-binding-initiated-ETBR activation increases cytosolic Ca2+, activates cell-specific eNOS or nNOS. (5) Temporally- and spatially-precise NO generation. NO diffuses into neighboring cells, thus acts in both SCF- and KIT-expressing cells. (6) NO modulates diverse cell-specific functions by NO/cGMP pathway, controlling transcriptional factors, or other mechanisms. We demonstrate the critical physiological role of the KIT-ET3-NO pathway in fulfilling high demand (exceeding basal level) of endothelium-dependent NO generation for coping with atherosclerosis, pregnancy, and aging. The KIT-ET3-NO pathway most likely also play critical roles in other cell functions that involve dual requirement of SCF-KIT signaling and NO. New strategies (e.g. enhancing the KIT-ET3-NO pathway) to harness the benefit of endogenous eNOS and nNOS activation and precise NO generation for correcting pathophysiology and restoring functions warrant investigation.
Collapse
|
15
|
García-Villalón A, Amor S, Monge L, Fernández N, Prodanov M, Muñoz M, Inarejos-García A, Granado M. In vitro studies of an aged black garlic extract enriched in S -allylcysteine and polyphenols with cardioprotective effects. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.08.062] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
16
|
Possible vasculoprotective role of linagliptin against sodium arsenite-induced vascular endothelial dysfunction. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:167-75. [PMID: 26497187 DOI: 10.1007/s00210-015-1184-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/09/2015] [Indexed: 01/09/2023]
Abstract
Vascular endothelial dysfunction (VED) interrupts the integrity and function of endothelial lining through enhanced markers of oxidative stress and decrease endothelial nitric oxide synthase (eNOS) expression. The main aim of the present study has been designed to investigate the possible vasculoprotective role of linagliptin against sodium arsenite-induced VED. Sodium arsenite (1.5 mg/kg, i.p., 2 weeks) abrogated the acetylcholine-induced, endothelium-dependent vasorelaxation by depicting the decrease in serum nitrite/nitrate concentration, reduced glutathione level, and simultaneously enhance the thiobarbituric acid reactive substances (TBARS) level, superoxide level, and tumor necrosis factor-alpha. These elevated markers interrupt the integrity of endothelial lining of thoracic aorta which was assessed histologically. The study elicits dose dependent effect of linagliptin (1.5 mg/kg, i.p. and 3 mg/kg, i.p.) or atorvastatin (30 mg/kg, p.o.) treatment, improved the endothelium-dependent independent relaxation, improve the integrity of endothelium lining which was assessed histologically by enhancing the serum nitrite/nitrate level, reduced glutathione level and simultaneously decreasing the TBARS level, superoxide anion level and tumor necrosis factor-alpha (TNF-α) level. L-NAME (25 mg/kg, i.p.), eNOS inhibitor, abrogated the ameliorative potential of linagliptin. However, the ameliorative potential of linagliptin has been enhanced by l-arginine (200 mg/kg, i.p.) which elicits that ameliorative potential of linagliptin was through eNOS signaling cascade and it may be concluded that linagliptin 3 mg/kg, i.p. has more significantly activated the eNOS and decreased the oxidative markers than linagliptin 1.5 mg/kg, i.p. and prevented sodium arsenite-induced VED.
Collapse
|
17
|
Besedina A. NO-Synthase Activity in Patients with Coronary Heart Disease Associated with Hypertension of Different Age Groups. J Med Biochem 2015; 35:43-49. [PMID: 28356863 PMCID: PMC5346800 DOI: 10.1515/jomb-2015-0008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/16/2015] [Indexed: 11/26/2022] Open
Abstract
Background Coronary heart disease is the leading cause of death and disability worldwide. Hypertension is a major independent risk factor for the development of CHD. Abnormalities in NO generation or activity have been proposed as a major mechanism of CHD. The purpose of this article is to determine the activity of eNOS and iNOS in patients with isolated CHD and CHD associated with HT of different age groups. Methods Fifty patients with isolated CHD and 42 patients with CHD associated with HT were enrolled in this study. NOS activity was determined by nitrite anion formed in the reaction. Results A statistically significant increase in iNOS activity is observed in elderly donors. In patients with isolated coronary heart disease cNOS activity is statistically significantly reduced with respect to the control group. The reduction of enzymatic activity of cNOS is more expressed in elderly patients than in middle-aged patients with coronary heart disease. Alterations in eNOS activity are more expressed in patients with coronary heart disease associated with hypertension than in patients with isolated coronary heart disease. Against the background of cNOS inhibition in the patients, a sharp increase in iNOS activity is observed. Conclusions It has been shown that disturbance of endothelial function in patients with coronary heart disease associated with hypertension is characterized by reduced endothelial NO synthesis by cNOS and increased systemic NO synthesis due to increased iNOS activity. It has been found that the lack of endothelial NO and hyperproduction of »harmful« NO by iNOS are more expressed in elderly patients.
Collapse
Affiliation(s)
- Anna Besedina
- Department of Family Medicine, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
18
|
Babbitt DM, Kim JS, Forrester SJ, Brown MD, Park JY. Effect of Interleukin-10 and Laminar Shear Stress on Endothelial Nitric Oxide Synthase and Nitric Oxide in African American Human Umbilical Vein Endothelial Cells. Ethn Dis 2015; 25:413-8. [PMID: 26674844 DOI: 10.18865/ed.25.4.413] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND African Americans have a predisposition to heightened systemic inflammation and a high prevalence of hypertension. OBJECTIVE The purpose of this study was to evaluate the influence of interleukin-10 (IL-10) and laminar shear stress (LSS) on African American endothelial cells by measuring total endothelial nitric oxide synthase (eNOS) protein expression and its phosphorylated form (p-eNOS) at Serine 1177, and nitric oxide (NO) levels, in response to IL-10 incubation and high physiological levels of LSS, used as an in vitro mimetic for aerobic exercise training (AEXT). DESIGN Human umbilical vein endothelial cells (HUVEC) from an African American donor were cultured. The experimental conditions included Static, Static with IL-10 Incubation, LSS at 20 dynes/cm², and LSS at 20 dynes/cm² with IL-10 Incubation. Western blotting was used to measure eNOS and p-eNOS protein expression in the cells. A modified Griess assay was used to measure NO metabolites in the cell culture media. RESULTS There were significant increases in p-eNOS, eNOS, and NO in the LSS at 20 dynes/cm² and LSS at 20 dynes/cm² with IL-10 Incubation experimental conditions when compared to the Static experimental condition. There were no other statistically significant differences demonstrating that IL-10 did not have an additive effect on eNOS activity in our study. CONCLUSION The significant increases in p-eNOS, eNOS, and NO as a result of LSS in African American HUVECs suggest that AEXT may be a viable, nonpharmacologic method to improve vascular inflammation status and vasodilation, and thereby contribute to hypertension reduction in the African American population.
Collapse
Affiliation(s)
| | - Ji-Seok Kim
- 2. Department of Kinesiology, Temple University
| | | | - Michael D Brown
- 3. Vascular Health Laboratory, Department of Kinesiology & Nutrition, University of Illinois at Chicago
| | - Joon-Young Park
- 2. Department of Kinesiology, Temple University ; 4.Cardiovascular Research Center, Temple University School of Medicine
| |
Collapse
|
19
|
Angelone T, Quintieri AM, Pasqua T, Filice E, Cantafio P, Scavello F, Rocca C, Mahata SK, Gattuso A, Cerra MC. The NO stimulator, Catestatin, improves the Frank-Starling response in normotensive and hypertensive rat hearts. Nitric Oxide 2015; 50:10-19. [PMID: 26241941 DOI: 10.1016/j.niox.2015.07.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 07/08/2015] [Accepted: 07/29/2015] [Indexed: 10/23/2022]
Abstract
The myocardial response to mechanical stretch (Frank-Starling law) is an important physiological cardiac determinant. Modulated by many endogenous substances, it is impaired in the presence of cardiovascular pathologies and during senescence. Catestatin (CST:hCgA352-372), a 21-amino-acid derivate of Chromogranin A (CgA), displays hypotensive/vasodilatory properties and counteracts excessive systemic and/or intra-cardiac excitatory stimuli (e.g., catecholamines and endothelin-1). CST, produced also by the myocardium, affects the heart by modulating inotropy, lusitropy and the coronary tone through a Nitric Oxide (NO)-dependent mechanism. This study evaluated the putative influence elicited by CST on the Frank-Starling response of normotensive Wistar-Kyoto (WKY) and hypertensive (SHR) hearts by using isolated and Langendorff perfused cardiac preparations. Functional changes were evaluated on aged (18-month-old) WKY rats and SHR which mimic human chronic heart failure (HF). Comparison to WKY rats, SHR showed a reduced Frank-Starling response. In both rat strains, CST administration improved myocardial mechanical response to increased end-diastolic pressures. This effect was mediated by EE/IP3K/NOS/NO/cGMP/PKG, as revealed by specific inhibitors. CST-dependent positive Frank-Starling response is paralleled by an increment in protein S-Nitrosylation. Our data suggested CST as a NO-dependent physiological modulator of the stretch-induced intrinsic regulation of the heart. This may be of particular importance in the aged hypertrophic heart, whose function is impaired because of a reduced systolic performance accompanied by delayed relaxation and increased diastolic stiffness.
Collapse
Affiliation(s)
- T Angelone
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy; National Institute of Cardiovascular Research, Bologna, Italy
| | - A M Quintieri
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy
| | - T Pasqua
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy
| | - E Filice
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy
| | - P Cantafio
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy
| | - F Scavello
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy
| | - C Rocca
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy
| | - S K Mahata
- Department of Medicine, University of California & VA San Diego Healthcare System, San Diego, USA
| | - A Gattuso
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy.
| | - M C Cerra
- Dept of Biology, Ecology and Earth Sciences, University of Calabria, Rende, CS, Italy; National Institute of Cardiovascular Research, Bologna, Italy.
| |
Collapse
|
20
|
Shen J, Wilmot KA, Ghasemzadeh N, Molloy DL, Burkman G, Mekonnen G, Gongora MC, Quyyumi AA, Sperling LS. Mediterranean Dietary Patterns and Cardiovascular Health. Annu Rev Nutr 2015; 35:425-49. [DOI: 10.1146/annurev-nutr-011215-025104] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jia Shen
- Emory Clinical Cardiovascular Research Institute,
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| | - Kobina A. Wilmot
- Emory Clinical Cardiovascular Research Institute,
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| | - Nima Ghasemzadeh
- Emory Clinical Cardiovascular Research Institute,
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| | - Daniel L. Molloy
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| | - Gregory Burkman
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| | - Girum Mekonnen
- Emory Clinical Cardiovascular Research Institute,
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| | - Maria C. Gongora
- Emory Clinical Cardiovascular Research Institute,
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| | - Arshed A. Quyyumi
- Emory Clinical Cardiovascular Research Institute,
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| | - Laurence S. Sperling
- Emory Clinical Cardiovascular Research Institute,
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia 30322;
| |
Collapse
|
21
|
Hartaigh BÓ, Allore HG, Trentalange M, McAvay G, Pilz S, Dodson JA, Gill TM. Elevations in time-varying resting heart rate predict subsequent all-cause mortality in older adults. Eur J Prev Cardiol 2015; 22:527-34. [PMID: 24445263 PMCID: PMC4156557 DOI: 10.1177/2047487313519932] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND An increased resting heart rate (RHR) has long been associated with unhealthy life. Nevertheless, it remains uncertain whether time-varying measurements of RHR are predictive of mortality in older persons. DESIGN The purpose of this study was to assess the relationship between repeated measurements of RHR and risk of death from all causes among older adults. METHODS We evaluated repeat measurements of resting heart rate among 5691 men and women (aged 65 years or older) enrolled in the Cardiovascular Health Study. RHR was measured annually for six consecutive years by validated electrocardiogram. All-cause mortality was confirmed by a study-wide Mortality Review Committee using reviews of obituaries, death certificates and hospital records, interviews with attending physicians, and next-of-kin. RESULTS Of the study cohort, 974 (17.1%) participants died. Each 10 beat/min increment in RHR increased the risk of death by 33% (adjusted hazard ratio, 95% confidence interval (CI) = 1.33, 1.26-1.40). Similar results were observed (adjusted hazard ratio, 95% CI = 2.21, 1.88-2.59) when comparing the upper-most quartile of RHR (mean = 81 beats/min) with the lowest (mean = 53 beats/min). Compared with participants whose RHR was consistently ≤65 beats/min during the study period, the risk of death increased monotonically for each 10 beat/min (consistent) increment in RHR, with adjusted hazard ratios (95% CI) ranging from 1.30 (1.23-1.37) for 75 beats/min to 4.78 (3.49-6.52) for 125 beats/min. CONCLUSIONS Elevations in the RHR over the course of six years are associated with an increased risk of all-cause mortality among older adults.
Collapse
Affiliation(s)
- Bríain ó Hartaigh
- Department of Internal Medicine/Geriatrics, Yale School of Medicine, USA
| | - Heather G Allore
- Department of Internal Medicine/Geriatrics, Yale School of Medicine, USA
| | - Mark Trentalange
- Department of Internal Medicine/Geriatrics, Yale School of Medicine, USA
| | - Gail McAvay
- Department of Internal Medicine/Geriatrics, Yale School of Medicine, USA
| | - Stefan Pilz
- Department of Internal Medicine, Medical University of Graz, Austria
| | - John A Dodson
- Department of Internal Medicine, Brigham and Women's Hospital, USA
| | - Thomas M Gill
- Department of Internal Medicine/Geriatrics, Yale School of Medicine, USA
| |
Collapse
|
22
|
Guo BC, Wei J, Su KH, Chiang AN, Zhao JF, Chen HY, Shyue SK, Lee TS. Transient receptor potential vanilloid type 1 is vital for (-)-epigallocatechin-3-gallate mediated activation of endothelial nitric oxide synthase. Mol Nutr Food Res 2015; 59:646-57. [PMID: 25581901 DOI: 10.1002/mnfr.201400699] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/31/2014] [Accepted: 01/02/2015] [Indexed: 11/06/2022]
Abstract
SCOPE Epigallocatechin-3-gallate (EGCG), the most abundant catechin of green tea, has beneficial effects on physiological functions of endothelial cells (ECs), yet the detailed mechanisms are not fully understood. In this study, we investigated the role of transient receptor potential vanilloid type 1 (TRPV1), a ligand-gated nonselective calcium channel, in EGCG-mediated endothelial nitric oxide (NO) synthase (eNOS) activation and angiogenesis. METHODS AND RESULTS In ECs, treatment with EGCG time-dependently increased the intracellular level of Ca(2+) . Removal of extracellular calcium (Ca(2+) ) by EGTA or EDTA or inhibition of TRPV1 by capsazepine or SB366791 abrogated EGCG-increased intracellular Ca(2+) level in ECs or TRPV1-transfected HEK293 cells. Additionally, EGCG increased the phsophorylation of eNOS at Ser635 and Ser1179, Akt at Ser473, calmodulin-dependent protein kinase II (CaMKII) at Thr286 and AMP-activated protein kinase (AMPK) at Thr172, all abolished by the TRPV1 antagonist capsazepine. EGCG-induced NO production was diminished by pretreatment with LY294002 (an Akt inhibitor), KN62 (a CaMKII inhibitor), and compound C (an AMPK inhibitor). Moreover, blocking TRPV1 activation prevented EGCG-induced EC proliferation, migration, and tube formation, as well as angiogenesis in Matrigel plugs in mice. CONCLUSION EGCG may trigger activation of TRPV1-Ca(2+) signaling, which leads to phosphorylation of Akt, AMPK, and CaMKII; eNOS activation; NO production; and, ultimately, angiogenesis in ECs.
Collapse
Affiliation(s)
- Bei-Chia Guo
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
It is a need to define the line between pathological and physiological functions of reactive oxygen species (ROS) in order to understand their beneficial role over their injurious consequences.
Collapse
Affiliation(s)
- Arun Kumar Sharma
- Department of Pharmacology
- Amity Institute of Pharmacy
- Amity University
- Noida-201303
- India
| | - Gourav Taneja
- Department of Pharmacology
- Amity Institute of Pharmacy
- Amity University
- Noida-201303
- India
| | - Deepa Khanna
- Department of Pharmacology
- Rajendra Institute of Technology and Sciences
- Sirsa-125 055
- India
| | - Satyendra K. Rajput
- Department of Pharmacology
- Amity Institute of Pharmacy
- Amity University
- Noida-201303
- India
| |
Collapse
|
24
|
Cho DH, Park JH, Joo Lee E, Jong Won K, Lee SH, Kim YH, Hwang S, Ja Kwon K, Young Shin C, Song KH, Jo I, Han SH. Valproic acid increases NO production via the SH-PTP1-CDK5-eNOS-Ser(116) signaling cascade in endothelial cells and mice. Free Radic Biol Med 2014; 76:96-106. [PMID: 25150199 DOI: 10.1016/j.freeradbiomed.2014.07.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 07/19/2014] [Accepted: 07/31/2014] [Indexed: 02/07/2023]
Abstract
Valproic acid (VPA) with its inhibitory activity of histone deacetylase has been used in the treatment of epilepsy and bipolar disorder associated with cerebrovascular dysfunction. Because nitric oxide (NO) produced by endothelial NO synthase (eNOS) plays a role in the maintenance of vascular function, NO is likely to mediate VPA׳s drug effect, but its effect on NO production remains controversial. We investigated whether and how VPA regulates NO production in bovine aortic endothelial cells (BAECs) and mice. VPA increased NO production in BAECs, which was accompanied by a decrease in phosphorylation of eNOS at serine 116 (eNOS-Ser(116)) and cyclin-dependent kinase 5 at tyrosine 15 (CDK5-Tyr(15)). Ectopic expression of p25, a CDK5 activator, restored the VPA-inhibited eNOS-Ser(116) phosphorylation. In silico analysis revealed that the CDK5-Tyr(15) residue might be a substrate for SH2 domain-containing protein tyrosine phosphatase 1 (SH-PTP1), and CDK5 actually interacted with SH-PTP1. VPA increased SH-PTP1 expression and its activity. Stibogluconate, a specific SH-PTP1 inhibitor, reversed the VPA-inhibited phosphorylation of CDK5-Tyr(15) and eNOS-Ser(116). Knockdown of SH-PTP1 using small interfering RNA also reversed all the observed effects of VPA. Finally, both serum NO level and acetylcholine-induced aortic relaxation increased in VPA-medicated male mice. These increases were accompanied by increased SH-PTP1 expression and decreased phosphorylation of CDK5-Tyr(15) and eNOS-Ser(116) in mouse aortas. In conclusion, VPA increases NO production by inhibiting the CDK5-Tyr(15)-eNOS-Ser(116) phosphorylation axis; this process is mediated by SH-PTP1. VPA may be useful in the treatment of NO-related cerebrocardiovascular diseases.
Collapse
Affiliation(s)
- Du-Hyong Cho
- Department of Neurology, Konkuk University Medical Center, and Department of Pharmacology, Center for Geriatric Neuroscience Research, SMART Institute of Advanced Biomedical Science, and Gwangjin-gu, Seoul 143-701, Korea; Department of Pharmacology, School of Medicine, Eulji University, Jung-gu, Daejeon 301-746, Korea
| | - Jung-Hyun Park
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, Korea
| | - Eun Joo Lee
- Department of Neurology, Konkuk University Medical Center, and Department of Pharmacology, Center for Geriatric Neuroscience Research, SMART Institute of Advanced Biomedical Science, and Gwangjin-gu, Seoul 143-701, Korea
| | - Kyung Jong Won
- Department of Medical Science, Institute of Functional Genomics, Konkuk University School of Medicine, Chungju 380-701, Korea
| | - Sang-Hee Lee
- Department of Microbiology, Chungbuk National University, Heungduk-gu, Cheongju 361-763, Korea
| | - Yang-Hoon Kim
- Department of Microbiology, Chungbuk National University, Heungduk-gu, Cheongju 361-763, Korea
| | - Soojin Hwang
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, Korea
| | - Kyoung Ja Kwon
- Department of Neurology, Konkuk University Medical Center, and Department of Pharmacology, Center for Geriatric Neuroscience Research, SMART Institute of Advanced Biomedical Science, and Gwangjin-gu, Seoul 143-701, Korea
| | - Chan Young Shin
- Department of Neurology, Konkuk University Medical Center, and Department of Pharmacology, Center for Geriatric Neuroscience Research, SMART Institute of Advanced Biomedical Science, and Gwangjin-gu, Seoul 143-701, Korea
| | - Kee-Ho Song
- Department of Internal Medicine, Konkuk University School of Medicine, Gwangjin-gu, Seoul 143-701, Korea
| | - Inho Jo
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, Korea.
| | - Seol-Heui Han
- Department of Neurology, Konkuk University Medical Center, and Department of Pharmacology, Center for Geriatric Neuroscience Research, SMART Institute of Advanced Biomedical Science, and Gwangjin-gu, Seoul 143-701, Korea.
| |
Collapse
|
25
|
Superparamagnetic iron oxide nanoparticles impair endothelial integrity and inhibit nitric oxide production. Acta Biomater 2014; 10:4896-4911. [PMID: 25123083 DOI: 10.1016/j.actbio.2014.07.027] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/26/2014] [Accepted: 07/22/2014] [Indexed: 01/08/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPION) are widely used both clinically and experimentally for diverse in vivo applications, such as contrast enhancement in magnetic resonance imaging, hyperthermia and drug delivery. Biomedical applications require particles to have defined physical and chemical properties, and to be stable in biological media. Despite a suggested low cytotoxic action, adverse reactions of SPION in concentrations relevant for biomedical use have not yet been studied in sufficient detail. In the present work we employed Endorem®, dextran-stabilized SPION approved as an intravenous contrast agent, and compared its action to a set of other nanoparticles with potential for magnetic resonance imaging applications. SPION in concentrations relevant for in vivo applications were rapidly taken up by endothelial cells and exhibited no direct cytotoxicity. Electric cell impedance sensing measurements demonstrated that SPION, but not BaSO4/Gd nanoparticles, impaired endothelial integrity, as was confirmed by increased intercellular gap formation in endothelial monolayers. These structural changes induced the subcellular translocation and inhibition of the cytoprotective and anti-atherosclerotic enzyme endothelial NO-synthase and reduced NO production. Lipopolysaccharide-induced inflammatory NO production of macrophages was not affected by SPION. In conclusion, our data suggest that SPION might substantially alter endothelial integrity and function at therapeutically relevant doses, which are not cytotoxic.
Collapse
|
26
|
Wang S, Lin LM, Wu YN, Fang M, Yu YQ, Zhou J, Gong ZY. Angiotensin I Converting Enzyme (ACE) inhibitory activity and antihypertensive effects of grass carp peptides. Food Sci Biotechnol 2014. [DOI: 10.1007/s10068-014-0226-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
27
|
Posada-Duque RA, Barreto GE, Cardona-Gomez GP. Protection after stroke: cellular effectors of neurovascular unit integrity. Front Cell Neurosci 2014; 8:231. [PMID: 25177270 PMCID: PMC4132372 DOI: 10.3389/fncel.2014.00231] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/24/2014] [Indexed: 12/16/2022] Open
Abstract
Neurological disorders are prevalent worldwide. Cerebrovascular diseases (CVDs), which account for 55% of all neurological diseases, are the leading cause of permanent disability, cognitive and motor disorders and dementia. Stroke affects the function and structure of blood-brain barrier, the loss of cerebral blood flow regulation, oxidative stress, inflammation and the loss of neural connections. Currently, no gold standard treatments are available outside the acute therapeutic window to improve outcome in stroke patients. Some promising candidate targets have been identified for the improvement of long-term recovery after stroke, such as Rho GTPases, cell adhesion proteins, kinases, and phosphatases. Previous studies by our lab indicated that Rho GTPases (Rac and RhoA) are involved in both tissue damage and survival, as these proteins are essential for the morphology and movement of neurons, astrocytes and endothelial cells, thus playing a critical role in the balance between cell survival and death. Treatment with a pharmacological inhibitor of RhoA/ROCK blocks the activation of the neurodegeneration cascade. In addition, Rac and synaptic adhesion proteins (p120 catenin and N-catenin) play critical roles in protection against cerebral infarction and in recovery by supporting the neurovascular unit and cytoskeletal remodeling activity to maintain the integrity of the brain parenchyma. Interestingly, neuroprotective agents, such as atorvastatin, and CDK5 silencing after cerebral ischemia and in a glutamate-induced excitotoxicity model may act on the same cellular effectors to recover neurovascular unit integrity. Therefore, future efforts must focus on individually targeting the structural and functional roles of each effector of neurovascular unit and the interactions in neural and non-neural cells in the post-ischemic brain and address how to promote the recovery or prevent the loss of homeostasis in the short, medium and long term.
Collapse
Affiliation(s)
- Rafael Andres Posada-Duque
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria (SIU), University of Antioquia UdeA Medellín, Colombia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá D.C., Colombia
| | - Gloria Patricia Cardona-Gomez
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Faculty of Medicine, Sede de Investigación Universitaria (SIU), University of Antioquia UdeA Medellín, Colombia
| |
Collapse
|
28
|
Delgado-Lista J, Perez-Martinez P, Garcia-Rios A, Perez-Caballero AI, Perez-Jimenez F, Lopez-Miranda J. Mediterranean Diet and Cardiovascular Risk: Beyond Traditional Risk Factors. Crit Rev Food Sci Nutr 2014; 56:788-801. [DOI: 10.1080/10408398.2012.726660] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
29
|
Guan XM, Cheng M, Li H, Cui XD, Li X, Wang YL, Sun JL, Zhang XY. Biological properties of bone marrow-derived early and late endothelial progenitor cells in different culture media. Mol Med Rep 2013; 8:1722-8. [PMID: 24126824 DOI: 10.3892/mmr.2013.1718] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 09/26/2013] [Indexed: 11/05/2022] Open
Abstract
Ex vivo expansion of endothelial progenitor cells (EPCs) may be a promising strategy to overcome the clinical problem of limited cell numbers. As the culture medium is the key for the cell characteristics, the effects of different culture media on EPCs were investigated in the present study. Rat bone marrow mononuclear cells were cultured in different media, including M-199 media with 20% fetal bovine serum (FBS) and bovine pituitary extract (M1); M-199 media with 10% FBS, 20 ng/ml vascular endothelial growth factor (VEGF) and 10 ng/ml basic fibroblast growth factor (bFGF; M2) or epidermal growth medium (EGM)-2MV media. The cell morphology and biological functions, such as proliferation, adhesion, migration, tube formation and nitric oxide (NO) production were subsequently assayed in vitro. Moreover, endothelial biomarkers and apoptosis were also analyzed. The results showed that endothelial‑like cells appeared in all of the culture systems. First‑passage cells, namely early EPCs, tended to form colonies in M2 and EGM-2MV media but showed a fusiform shape in M1 media. The 3rd or 4th generation EPCs, namely late EPCs, cultured in EGM-2MV media exhibited increased adhesion, migration, tube formation and NO production as compared with EPCs in M1 or M2 media. Furthermore, late EPCs cultured in EGM-2MV expressed higher levels of endothelial cell markers, such as von Willibrand factor (vWF)and CD31, but relatively greater levels of apoptosis were observed. In conclusion, cell culture conditions, for example the medium used, affects the biological properties of bone marrow-derived early and late EPCs.
Collapse
Affiliation(s)
- Xiu M Guan
- Medicine Research Center, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
The role of the vessel wall. Methods Mol Biol 2013; 992:31-46. [PMID: 23546703 DOI: 10.1007/978-1-62703-339-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The role of the vessel wall is complex and its effects are wide-ranging. The vessel wall, specifically the endothelial monolayer that lines the inner lumen, possesses the ability to influence various physiological states both locally and systemically by controlling vascular tone, basement membrane component synthesis, angiogenesis, haemostatic properties, and immunogenicity. This is an overview of the function and structure of the vessel wall and how disruption and dysfunction in any of these regulatory roles can lead to disease states.
Collapse
|
31
|
Ni H, Chen J, Pan M, Zhang M, Zhang J, Chen P, Liu B. FTY720 prevents progression of renal fibrosis by inhibiting renal microvasculature endothelial dysfunction in a rat model of chronic kidney disease. J Mol Histol 2013; 44:693-703. [PMID: 23907620 DOI: 10.1007/s10735-013-9521-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/22/2013] [Indexed: 02/05/2023]
Abstract
Recent studies have shown that chronic endothelial dysfunction can impair multiple aspects of renal physiology and, in turn, contribute to renal fibrosis. Sphingosine 1-phosphate (S1P) has been highlighted as an endothelial barrier-stabilizing mediator. The aim of our study was to investigate the effect of FTY720, an S1P analog, on the progression of renal fibrosis by inhibiting renal microvasculature endothelial dysfunction in a rat model of chronic kidney disease. Thirty male Sprague-Dawley rats were used in this study. Seven days after surgery, we placed the animals into three groups: sham surgery; 5/6 nephrectomized (Nx) rats; and 5/6Nx + FTY720 (1 mg/kg/day). All of the animals were sacrificed 12 weeks after surgery. We obtained and analyzed blood and kidney tissue samples from all of the groups. Glomerular capillary density and peritubular capillary (PTC) density were determined by CD31 immunostaining. The expression of transforming growth factor beta 1 (TGF-β1), collagen IV, fibronectin, endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) were analyzed by immunohistochemistry, reverse transcription-polymerase chain reaction and western blotting. The 5/6Nx group exhibited increased blood urea nitrogen and serum creatinine, visible renal histological changes, pro-fibrotic molecule (TGF-β1) and production of extracellular matrix proteins such as collagen IV and fibronectin and decreased glomerular and PTC density, compared to the sham controls (P < 0.01). We observed that treatment with FTY720 reduced these abnormalities. Furthermore, the level of NO, the expression levels of eNOS and VEGF were downregulated in the kidney tissue in 5/6Nx rats, FTY720 treatment significantly attenuated this decrease. FTY720 prevents the progression of renal fibrosis by inhibiting renal microvasculature endothelial dysfunction in a rat model of chronic kidney disease.
Collapse
Affiliation(s)
- Haifeng Ni
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | | | | | | | | | | | | |
Collapse
|
32
|
Cabrales P, Friedman JM. HBOC vasoactivity: interplay between nitric oxide scavenging and capacity to generate bioactive nitric oxide species. Antioxid Redox Signal 2013; 18:2284-97. [PMID: 23249305 PMCID: PMC3638560 DOI: 10.1089/ars.2012.5099] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
SIGNIFICANCE Despite many advances in blood substitute research, the development of materials that are effective in maintaining blood volume and oxygen delivery remains a priority for emergency care and trauma. Clinical trials on hemoglobin (Hb)-based oxygen carriers (HBOCs) have not provided information on the mechanism of toxicity, although all commercial formulations have safety concerns. Specifically, it is important to reconcile the different hypotheses of Hb toxicity, such as nitric oxide (NO) depletion and oxidative reactions, to provide a coherent molecular basis for designing a safe HBOC. RECENT ADVANCES HBOCs with different sizes often exhibit differences in the degree of HBOC-induced vasoactivity. This has been attributed to differences in the degree of NO scavenging and in the extent of Hb extravasation. Additionally, it is appears that Hb can undergo reactions that compensate for NO scavenging by generating bioactive forms of NO. CRITICAL ISSUES Engineering modifications to enhance bioactive NO production can result in diminished oxygen delivery by virtue of increased oxygen affinity. This strategy can prevent the HBOC from fulfilling the intended goal on preserving oxygenation; however, the NO production effects will increase perfusion and oxygen transport. FUTURE DIRECTIONS Hb modifications influence NO scavenging and the capacity of certain HBOCs to compensate for NO scavenging through nitrite-mediated reactions that generate bioactive NO. Based on the current understanding of these NO-related factors, possible synthetic strategies are presented that address how HBOC formulations can be prepared that: (i) effectively deliver oxygen, (ii) maintain tissue perfusion, and (iii) limit/reverse underlying inflammation within the vasculature.
Collapse
Affiliation(s)
- Pedro Cabrales
- Department of Bioengineering, University of California, San Diego, CA, USA.
| | | |
Collapse
|
33
|
Kathuria S, Mahadevan N, Balakumar P. Possible involvement of PPARγ-associated eNOS signaling activation in rosuvastatin-mediated prevention of nicotine-induced experimental vascular endothelial abnormalities. Mol Cell Biochem 2013; 374:61-72. [PMID: 23149826 DOI: 10.1007/s11010-012-1505-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/25/2012] [Indexed: 12/30/2022]
Abstract
Nicotine exposure via cigarette smoking and tobacco chewing is associated with vascular complications. The present study investigated the effect of rosuvastatin in nicotine (2 mg/kg/day, i.p., 4 weeks)-induced vascular endothelial dysfunction (VED) in rats. The development of VED was assessed by employing isolated aortic ring preparation and estimating aortic and serum nitrite/nitrate concentration. Further, scanning electron microscopy and hematoxylin-eosin staining of thoracic aorta were performed to assess the vascular endothelial integrity. Moreover, oxidative stress was assessed by estimating aortic superoxide anion generation and serum thiobarbituric acid-reactive substances. The nicotine administration produced VED by markedly reducing acetylcholine-induced endothelium-dependent relaxation, impairing the integrity of vascular endothelium, decreasing aortic and serum nitrite/nitrate concentration, increasing oxidative stress, and inducing lipid alteration. However, treatment with rosuvastatin (10 mg/kg/day, i.p., 4 weeks) markedly attenuated nicotine-induced vascular endothelial abnormalities, oxidative stress, and lipid alteration. Interestingly, the co-administration of peroxisome proliferator-activated receptor γ (PPARγ) antagonist, GW9662 (1 mg/kg/day, i.p., 2 weeks) submaximally, significantly prevented rosuvastatin-induced improvement in vascular endothelial integrity, endothelium-dependent relaxation, and nitrite/nitrate concentration in rats administered nicotine. However, GW9662 co-administration did not affect rosuvastatin-associated vascular anti-oxidant and lipid-lowering effects. The incubation of aortic ring, isolated from rosuvastatin-treated nicotine-administered rats, with L-NAME (100 μM), an inhibitor of nitric oxide synthase (NOS), significantly attenuated rosuvastatin-induced improvement in acetylcholine-induced endothelium-dependent relaxation. Rosuvastatin prevents nicotine-induced vascular endothelial abnormalities by activating PPARγ and endothelial NOS signaling pathways. Moreover, the PPARγ-independent anti-oxidant and lipid-lowering effects of rosuvastatin might additionally play a role in the improvement of vascular endothelial function.
Collapse
Affiliation(s)
- Sonam Kathuria
- Cardiovascular Pharmacology Division, Department of Pharmacology, Rajendra Institute of Technology and Sciences, Sirsa, 125 055, India
| | | | | |
Collapse
|
34
|
Filho AG, Kinote A, Pereira DJ, Rennó A, dos Santos RC, Ferreira-Melo SE, Velloso LA, Bordin S, Anhê GF, Junior HM. Infliximab prevents increased systolic blood pressure and upregulates the AKT/eNOS pathway in the aorta of spontaneously hypertensive rats. Eur J Pharmacol 2013; 700:201-9. [DOI: 10.1016/j.ejphar.2012.11.059] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 11/09/2012] [Accepted: 11/20/2012] [Indexed: 01/10/2023]
|
35
|
Zhang J, Zhang X, Li H, Cui X, Guan X, Tang K, Jin C, Cheng M. Hyperglycaemia exerts deleterious effects on late endothelial progenitor cell secretion actions. Diab Vasc Dis Res 2013; 10:49-56. [PMID: 22561229 DOI: 10.1177/1479164112444639] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Endothelial progenitor cells (EPCs) play a fundamental role in tissue regeneration and vascular repair both by differentiating into endothelial cells and by secretion of vasoactive substances that promote angiogenesis and maintain vascular homeostasis. It has previously been shown that hyperglycaemia impairs early and late EPC functions, such as differentiation, proliferation and adhesion. However, its role in the regulation of the production of vasoactive substances in EPCs, especially in late EPCs, is less well defined. We investigated the effects of hyperglycaemia on the production of vasodilator, fibrinolytic and angiogenic growth factors, and also on the activity of superoxide dismutase (SOD) in late EPCs. For this purpose, late EPCs were incubated with different concentrations of D-glucose (5-40 mmol/L) for 24 hr. Levels of nitric oxide (NO), tissue plasminogen activator (t-PA), plasminogen activator inhibitor-1 (PAI-1), prostaglandin I(2) (PGI(2)), vascular endothelial growth factor (VEGF) and the activity of SOD were measured by enzyme-linked immunosorbent assay (ELISA). Under high glucose stress conditions, late EPCs exhibited lower levels of NO, t-PA, PAI-1, PGI(2) and VEGF compared to control medium (5 mmol/L glucose). Moreover, high glucose was also observed to decrease the activity of SOD in late EPCs. These results suggest that hyperglycaemia-induced impairment of late EPC secretion functions could contribute to the development of vascular disease in diabetes.
Collapse
Affiliation(s)
- Jie Zhang
- Weifang Medical College Medicine Research Center, Weifang, Shandong, 261053, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kim KW, Won YL, Ko KS, Heo KH, Chung YH. The effects of hazardous chemical exposure on cardiovascular disease in chemical products manufacturing workers. Toxicol Res 2012; 28:269-77. [PMID: 24278620 PMCID: PMC3834428 DOI: 10.5487/tr.2012.28.4.269] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 11/26/2012] [Accepted: 12/03/2012] [Indexed: 11/20/2022] Open
Abstract
The purpose of this study was to understand the mechanism of cardiovascular disease (CVD) caused by exposure to hazardous chemicals. We investigated changes in the symptoms of metabolic syndrome, which is strongly related to CVD, and in levels of other CVD risk factors, with a special emphasis on the roles of catecholamines and oxidative stress. The results revealed that neither body mass index (BMI) nor waist and hip circumferences were associated with exposure to hazardous chemicals. Among metabolic syndrome criteria, only HDL-cholesterol level increased on exposure to hazardous chemicals. Levels of epinephrine (EP) and norepinephrine (NEP) were not influenced by exposure to hazardous chemicals; however, the total antioxidative capacity (TAC) reduced because of increased oxidative stress. Both hazardous chemical exposure level and metabolite excretion were related to EP, NEP, and the oxidative stress index (OSI). Logistic regression analysis with these factors as independent variables and metabolic syndrome criteria as dependent variables revealed that EP was associated with blood pressure, and NEP with metabolic syndrome in the chemicalexposed group. In conclusion, the results suggest that reactive oxygen species generated and oxidative stress due to exposure to hazardous chemicals act as mediators and cause changes in the physiological levels of EP and NEP to increase blood pressure. This ultimately leads to the development of CVD through increase in cholesterol, triglyceride, and blood glucose levels by lipid peroxidation.
Collapse
Affiliation(s)
- Ki-Woong Kim
- Occupational Safety and Health Research Institute, KOSHA, Daejeon, Korea
| | - Yong Lim Won
- Occupational Safety and Health Research Institute, KOSHA, Incheon, Korea
| | - Kyung Sun Ko
- Occupational Safety and Health Research Institute, KOSHA, Incheon, Korea
| | - Kyung-Hwa Heo
- Occupational Safety and health Training Institute, KOSHA, Incheon, Korea
| | - Yong Hyun Chung
- Occupational Safety and Health Research Institute, KOSHA, Daejeon, Korea
| |
Collapse
|
37
|
Balakumar P, Kathuria S. Submaximal PPARγ activation and endothelial dysfunction: new perspectives for the management of cardiovascular disorders. Br J Pharmacol 2012; 166:1981-92. [PMID: 22404217 DOI: 10.1111/j.1476-5381.2012.01938.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PPARγ activation plays an important role in glucose metabolism by enhancing insulin sensitization. PPARγ is a primary target for thiazolidinedione-structured insulin sensitizers like pioglitazone and rosiglitazone employed for the treatment of type 2 diabetes mellitus. Additionally, PPARγ activation inhibits adhesion cascades and detrimental vascular inflammatory events. Importantly, activation of PPARγ plays a distinctive role in regulating the physiology and expression of endothelial nitric oxide synthase (eNOS) in the endothelium, resulting in enhanced generation of vascular nitric oxide. The PPARγ activation-mediated vascular anti-inflammatory and direct endothelial functional regulatory actions could, therefore, be beneficial in improving the vascular function in patients with atherosclerosis and hypertension with or without diabetes mellitus. Despite the disappointing cardiac side effect profile of rosiglitazone-like PPARγ full agonists, the therapeutic potential of novel pharmacological agents targeting PPARγ submaximally cannot be ruled out. This review discusses the potential regulatory role of PPARγ on eNOS expression and activation in improving the function of vascular endothelium. We argue that partial/submaximal activation of PPARγ could be a major target for vascular endothelial functional improvement. Interestingly, newly synthesized partial agonists of PPARγ such as balaglitazone, MBX-102, MK-0533, PAR-1622, PAM-1616, KR-62776 and SPPARγM5 are devoid of or have a reduced tendency to cause the adverse effects associated with full agonists of PPARγ. We propose that the vascular protective properties of pharmacological agents, which submaximally activate PPARγ, should be investigated. Moreover, the therapeutic opportunities of agents that submaximally activate PPARγ in preventing vascular endothelial dysfunction (VED) and VED-associated cardiovascular disorders are discussed.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Cardiovascular Pharmacology Division, Department of Pharmacology, Institute of Pharmacy, Rajendra Institute of Technology and Sciences-RITS, Sirsa, India.
| | | |
Collapse
|
38
|
Balakumar P, Taneja G. Fish oil and vascular endothelial protection: bench to bedside. Free Radic Biol Med 2012; 53:271-9. [PMID: 22584102 DOI: 10.1016/j.freeradbiomed.2012.05.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Revised: 03/16/2012] [Accepted: 05/03/2012] [Indexed: 02/07/2023]
Abstract
Fish oil is recommended for the management of hypertriglyceridemia and to prevent secondary cardiovascular disorders. Fish oil is a major source of ω-3-polyunsaturated fatty acids (PUFAs) such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). Clinical studies suggest that fish oil not only prevents the incidence of detrimental cardiovascular events, but also lowers the cardiovascular mortality rate. In addition to a classic lipid-lowering action, ω-3-PUFAs in fish oil could regulate blood pressure and enhance vascular integrity and compliance. Additionally, ω-3-PUFAs have the ability to protect vascular endothelial cells by decreasing oxidative stress, halting atherosclerotic events, and preventing vascular inflammatory and adhesion cascades. Intriguingly, recent studies have demonstrated that ω-3-PUFAs improve the function of vascular endothelium by enhancing the generation and bioavailability of endothelium-derived relaxing factor (nitric oxide) through upregulation and activation of endothelial nitric oxide synthase (eNOS). This certainly opens up a new area of research identifying potential mechanisms influencing fish oil-mediated functional regulatory action on vascular endothelium. We address in this review the potential of fish oil to prevent vascular endothelial dysfunction and associated cardiovascular disorders. Moreover, the mechanisms pertaining to fish oil-mediated eNOS activation and nitric oxide generation in improving endothelial function are delineated. We finally suggest the importance of further studies to determine the dose adjustment of fish oil with an optimal ratio of EPA and DHA for achieving consistent cardiovascular protection.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Cardiovascular Pharmacology Division, Department of Pharmacology, Institute of Pharmacy, Rajendra Institute of Technology and Sciences, Sirsa 125 055, India.
| | | |
Collapse
|
39
|
Balakumar P, Kathuria S, Taneja G, Kalra S, Mahadevan N. Is targeting eNOS a key mechanistic insight of cardiovascular defensive potentials of statins? J Mol Cell Cardiol 2012; 52:83-92. [DOI: 10.1016/j.yjmcc.2011.09.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Accepted: 09/16/2011] [Indexed: 01/14/2023]
|
40
|
Katz DL, Doughty K, Ali A. Cocoa and chocolate in human health and disease. Antioxid Redox Signal 2011; 15:2779-811. [PMID: 21470061 PMCID: PMC4696435 DOI: 10.1089/ars.2010.3697] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 04/05/2011] [Accepted: 04/06/2011] [Indexed: 01/26/2023]
Abstract
Cocoa contains more phenolic antioxidants than most foods. Flavonoids, including catechin, epicatechin, and procyanidins predominate in antioxidant activity. The tricyclic structure of the flavonoids determines antioxidant effects that scavenge reactive oxygen species, chelate Fe2+ and Cu+, inhibit enzymes, and upregulate antioxidant defenses. The epicatechin content of cocoa is primarily responsible for its favorable impact on vascular endothelium via its effect on both acute and chronic upregulation of nitric oxide production. Other cardiovascular effects are mediated through anti-inflammatory effects of cocoa polyphenols, and modulated through the activity of NF-κB. Antioxidant effects of cocoa may directly influence insulin resistance and, in turn, reduce risk for diabetes. Further, cocoa consumption may stimulate changes in redox-sensitive signaling pathways involved in gene expression and the immune response. Cocoa can protect nerves from injury and inflammation, protect the skin from oxidative damage from UV radiation in topical preparations, and have beneficial effects on satiety, cognitive function, and mood. As cocoa is predominantly consumed as energy-dense chocolate, potential detrimental effects of overconsumption exist, including increased risk of weight gain. Overall, research to date suggests that the benefits of moderate cocoa or dark chocolate consumption likely outweigh the risks.
Collapse
Affiliation(s)
- David L Katz
- Yale University Prevention Research Center, Griffin Hospital, Derby, Connecticut 06418, USA.
| | | | | |
Collapse
|
41
|
Angelone T, Quintieri AM, Pasqua T, Gentile S, Tota B, Mahata SK, Cerra MC. Phosphodiesterase type-2 and NO-dependent S-nitrosylation mediate the cardioinhibition of the antihypertensive catestatin. Am J Physiol Heart Circ Physiol 2011; 302:H431-42. [PMID: 22058158 DOI: 10.1152/ajpheart.00491.2011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The chromogranin A (CHGA)-derived peptide catestatin (CST: hCHGA(352-372)) is a noncompetitive catecholamine-release inhibitor that exerts vasodilator, antihypertensive, and cardiosuppressive actions. We have shown that CST directly influences the basal performance of the vertebrate heart where CST dose dependently induced a nitric oxide-cGMP-dependent cardiosuppression and counteracted the effects of adrenergic stimulation through a noncompetitive antagonism. Here, we sought to determine the specific intracardiac signaling activated by CST in the rat heart. Physiological analyses performed on isolated, Langendorff-perfused cardiac preparations revealed that CST-induced negative inotropism and lusitropism involve β(2)/β(3)-adrenergic receptors (β(2)/β(3)-AR), showing a higher affinity for β(2)-AR. Interaction with β(2)-AR activated phosphatidylinositol 3-kinase/endothelial nitric oxide synthase (eNOS), increased cGMP levels, and induced activation of phosphodiesterases type 2 (PDE2), which was found to be involved in the antiadrenergic action of CST as evidenced by the decreased cAMP levels. CST-dependent negative cardiomodulation was abolished by functional denudation of the endothelium with Triton. CST also increased the eNOS expression in cardiac tissue and human umbilical vein endothelial cells. cells, confirming the involvement of the vascular endothelium. In ventricular extracts, CST increased S-nitrosylation of both phospholamban and β-arrestin, suggesting an additional mechanism for intracellular calcium modulation and β-adrenergic responsiveness. We conclude that PDE2 and S-nitrosylation play crucial roles in the CST regulation of cardiac function. Our results are of importance in relation to the putative application of CST as a cardioprotective agent against stress, including excessive sympathochromaffin overactivation.
Collapse
Affiliation(s)
- Tommaso Angelone
- Department of Cell Biology, Laboratory of Cardiovascular Pathophysiology, University of Calabria, Arcavacata di Rende (CS), Italy.
| | | | | | | | | | | | | |
Collapse
|
42
|
Looft-Wilson RC, Billaud M, Johnstone SR, Straub AC, Isakson BE. Interaction between nitric oxide signaling and gap junctions: effects on vascular function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1895-902. [PMID: 21835160 DOI: 10.1016/j.bbamem.2011.07.031] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 07/14/2011] [Accepted: 07/19/2011] [Indexed: 02/07/2023]
Abstract
Nitric oxide signaling, through eNOS (or possibly nNOS), and gap junction communication are essential for normal vascular function. While each component controls specific aspects of vascular function, there is substantial evidence for cross-talk between nitric oxide signaling and the gap junction proteins (connexins), and more recently, protein-protein association between eNOS and connexins. This review will examine the evidence for interaction between these pathways in normal and diseased arteries, highlight the questions that remain about the mechanisms of their interaction, and explore the possible interaction between nitric oxide signaling and the newly discovered pannexin channels. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.
Collapse
Affiliation(s)
- R C Looft-Wilson
- Department of Kinesiology and Health Sciences, College of William and Mary, Williamsburg, VA 23187, USA
| | | | | | | | | |
Collapse
|
43
|
Delgado-Lista J, Garcia-Rios A, Perez-Martinez P, Fuentes F, Jiménez-Gomez Y, Gomez-Luna MJ, Parnell LD, Marin C, Lai CQ, Perez-Jimenez F, Ordovas JM, Lopez-Miranda J. Gene variations of nitric oxide synthase regulate the effects of a saturated fat rich meal on endothelial function. Clin Nutr 2011; 30:234-8. [DOI: 10.1016/j.clnu.2010.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 08/17/2010] [Accepted: 08/23/2010] [Indexed: 10/19/2022]
|
44
|
Verma S, Reddy K, Balakumar P. The defensive effect of benfotiamine in sodium arsenite-induced experimental vascular endothelial dysfunction. Biol Trace Elem Res 2010; 137:96-109. [PMID: 19943121 DOI: 10.1007/s12011-009-8567-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 11/03/2009] [Indexed: 10/20/2022]
Abstract
The present study has been designed to investigate the effect of benfotiamine, a thiamine derivative, in sodium arsenite-induced vascular endothelial dysfunction (VED) in rats. Sodium arsenite (1.5 mg(-1) kg(-1) day(-1) i.p., 2 weeks) was administered in rats to produce VED. The development of VED was assessed by employing isolated aortic ring preparation and estimating the serum and aortic concentrations of nitrite/nitrate. Further, the integrity of vascular endothelium in thoracic aorta was assessed by scanning electron microscopy. Moreover, the oxidative stress was assessed by estimating serum thiobarbituric acid reactive substances (TBARS) and aortic superoxide anion generation. The administration of sodium arsenite markedly produced VED by attenuating acetylcholine-induced endothelium-dependent relaxation, decreasing serum and aortic concentrations of nitrite/nitrate, and impairing the integrity of vascular endothelium. Further, sodium arsenite produced oxidative stress by increasing serum TBARS and aortic superoxide generation. The treatment with benfotiamine (25, 50, and 100 mg(-1) kg(-1) day(-1) p.o.) or atorvastatin (30 mg(-1) kg(-1) day(-1) p.o., a standard agent) prevented sodium arsenite-induced VED and oxidative stress. However, the beneficial effects of benfotiamine in preventing the sodium arsenite-induced VED were attenuated by co-administration with N-omega-nitro-L: -arginine methyl ester (L: -NAME) (25 mg(-1) kg(-1) day(-1), i.p.), an inhibitor of NOS. Thus, it may be concluded that benfotiamine reduces oxidative stress and activates endothelial nitric oxide synthase to enhance the generation and bioavailability of NO and subsequently improves the integrity of vascular endothelium to prevent sodium arsenite-induced experimental VED.
Collapse
Affiliation(s)
- Sanjali Verma
- Cardiovascular Pharmacology Division, ISF College of Pharmacy, Moga, 142 001, Punjab, India
| | | | | |
Collapse
|
45
|
Kaur J, Reddy K, Balakumar P. The Novel Role of Fenofibrate in Preventing Nicotine- and Sodium Arsenite-Induced Vascular Endothelial Dysfunction in the Rat. Cardiovasc Toxicol 2010; 10:227-38. [DOI: 10.1007/s12012-010-9086-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
46
|
Kaur T, Goel RK, Balakumar P. Effect of rosiglitazone in sodium arsenite-induced experimental vascular endothelial dysfunction. Arch Pharm Res 2010; 33:611-8. [PMID: 20422371 DOI: 10.1007/s12272-010-0416-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2009] [Revised: 01/23/2010] [Accepted: 01/25/2010] [Indexed: 10/19/2022]
Abstract
The present study has been designed to investigate the effect of rosiglitazone, a peroxisome proliferator activated receptor gamma agonist in sodium arsenite-induced vascular endothelial dysfunction (VED) in rats. The rats were administered sodium arsenite (1.5 mg/kg/day, i.p., 2 weeks) to induce VED. The development of VED was assessed by employing isolated aortic ring preparation and estimating serum nitrite/nitrate concentration. Further, the integrity of the aortic endothelium was assessed histologically using haematoxylin-eosin staining. Moreover, the oxidative stress was assessed by estimating serum thiobarbituric acid reactive substances, aortic reactive oxygen species and reduced form of glutathione. The administration of sodium arsenite produced VED by impairing acetylcholine-induced endothelium dependent relaxation, diminishing the integrity of vascular endothelium and decreasing the serum nitrite/nitrate concentration. In addition, sodium arsenite was noted to produce oxidative stress as it increased serum thiobarbituric acid reactive substances and aortic reactive oxygen species and consequently decreased glutathione. Treatment with rosiglitazone (3 mg/kg/day, p.o., 2 weeks and 5 mg/kg/day, p.o., 2 weeks) significantly prevented sodium arsenite-induced VED by enhancing acetylcholine-induced endothelium dependent relaxation, improving the integrity of vascular endothelium, increasing the nitrite/nitrate concentration and decreasing the oxidative stress. However, the vascular protective effect of rosiglitazone was markedly abolished by co-administration of nitric oxide synthase inhibitor, N-Omega-Nitro-L-Arginine Methyl Ester (L-NAME) (25 mg/kg/day, i.p., 2 weeks). Thus, it may be concluded that rosiglitazone reduces oxidative stress, activates eNOS and enhances the generation of nitric oxide to prevent sodium arsenite-induced VED in rats.
Collapse
Affiliation(s)
- Tajpreet Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, 147002, India
| | | | | |
Collapse
|
47
|
Xu X, Zhao CX, Wang L, Tu L, Fang X, Zheng C, Edin ML, Zeldin DC, Wang DW. Increased CYP2J3 expression reduces insulin resistance in fructose-treated rats and db/db mice. Diabetes 2010; 59:997-1005. [PMID: 20068141 PMCID: PMC2844847 DOI: 10.2337/db09-1241] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Accumulating evidence suggests that cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid into epoxyeicosatrienoic acids (EETs), which play crucial and diverse roles in cardiovascular homeostasis. The anti-inflammatory, antihypertensive, and pro-proliferative effects of EETs suggest a possible beneficial role for EETs on insulin resistance and diabetes. RESEARCH DESIGN AND METHODS This study investigated the effects of CYP2J3 epoxygenase gene therapy on insulin resistance and blood pressure in diabetic db/db mice and in a model of fructose-induced hypertension and insulin resistance in rats. RESULTS CYP2J3 gene delivery in vivo increased EET generation, reduced blood pressure, and reversed insulin resistance as determined by plasma glucose levels, homeostasis model assessment insulin resistance index, and glucose tolerance test. Furthermore, CYP2J3 treatment prevented fructose-induced decreases in insulin receptor signaling and phosphorylation of AMP-activated protein kinases (AMPKs) in liver, muscle, heart, kidney, and aorta. Thus, overexpression of CYP2J3 protected against diabetes and insulin resistance in peripheral tissues through activation of insulin receptor and AMPK pathways. CONCLUSIONS These results highlight the beneficial roles of the CYP epoxygenase-EET system in diabetes and insulin resistance.
Collapse
Affiliation(s)
- Xizhen Xu
- Department of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Chun Xia Zhao
- Department of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Luyun Wang
- Department of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ling Tu
- Department of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaosai Fang
- Department of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Changlong Zheng
- Department of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Dao Wen Wang
- Department of Internal Medicine and The Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Corresponding author: Dao Wen Wang,
| |
Collapse
|
48
|
Gu X, Herrera GA. Expression of eNOS in kidneys from hypertensive patients. Int J Nephrol Renovasc Dis 2010; 3:11-9. [PMID: 21694923 PMCID: PMC3108783 DOI: 10.2147/ijnrd.s6572] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Indexed: 01/16/2023] Open
Abstract
Endothelium-derived nitric oxide (NO) is essential for maintenance and regulation of blood pressure. In animal models, altered endothelium-derived nitric oxide synthase (eNOS) expression and impaired NO generation are important factors for renal injury. However, the pattern of eNOS expression in the kidneys from hypertensive patients has not been well established. We have studied the eNOS immuno-expression in kidney biopsies from hypertensive patients. Compared to kidneys from normotensive individuals, there were no significant alterations of eNOS immuno-expression in the vasculature of patients with chronic essential hypertension. In contrast, the expression of eNOS was significantly decreased in the glomeruli and arterioles/small arteries of patients with malignant hypertension, particularly in those with significant intimal edema and myxoid degeneration or thrombi. Endothelial dysfunction is an important pathogenetic factor for chronic primary hypertension and eNOS plays a major role in the regulation of vascular tone and function. Unchanged eNOS in the kidney vasculature in chronic primary hypertension indicates that these patients have an ability to compensate. In patients with malignant hypertension, the expression of eNOS protein was diminished in the injured vasculature. Loss of the compensatory mechanism via continued release of NO to prevent vascular injury may be responsible for morphological changes typically seen in the renal vasculature in patients with accelerated hypertension.
Collapse
Affiliation(s)
- Xin Gu
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| | | |
Collapse
|
49
|
Cho DH, Seo J, Park JH, Jo C, Choi YJ, Soh JW, Jo I. Cyclin-Dependent Kinase 5 Phosphorylates Endothelial Nitric Oxide Synthase at Serine 116. Hypertension 2010; 55:345-52. [DOI: 10.1161/hypertensionaha.109.140210] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nitric oxide (NO) production in endothelial cells (EC) is regulated by multisite phosphorylation of specific serine and threonine residues in endothelial NO synthase (eNOS). Among these, eNOS-Ser116 is phosphorylated in the basal state, and its phosphorylation contributes to basal NO production. Here, we investigated the mechanism by which eNOS-Ser116 is phosphorylated during the basal state using bovine aortic EC. Although a previous study suggested that protein kinase C was involved in eNOS-Ser116 phosphorylation, overexpression of various protein kinase C isoforms did not affect eNOS-Ser116 phosphorylation. An in silico analysis using a motif scan revealed that the eNOS-Ser116 residue might be a substrate for proline-directed protein kinases. Roscovitine, a specific inhibitor of cyclin-dependent kinase (CDK), 1, 2, and 5, but not an inhibitor of mitogen-activated protein kinase kinase or glycogen synthase kinase 3β, inhibited eNOS-Ser116 phosphorylation dose dependently. Furthermore, purified CDK1, 2, or 5 directly phosphorylated eNOS-Ser116 in vitro. Ectopic expression of the dominant-negative CDK5 but not dominant-negative CDK1 or dominant-negative CDK2 repressed eNOS-Ser116 phosphorylation and increased NO production. In addition, CDK5 activity was detected in bovine aortic EC, and coimmunoprecipitation and confocal microscopy studies revealed a colocalization of eNOS and CDK5. Cotransfection of CDK5 and p25, the specific CDK5 activator, increased eNOS-Ser116 phosphorylation and decreased NO production, but its parent molecule, p35, and p39, another activator, were not detected in bovine aortic EC, which suggests the existence of a novel CDK5 activator. Overall, this is the first study to find that CDK5 is a physiological kinase responsible for eNOS-Ser116 phosphorylation and regulation of NO production.
Collapse
Affiliation(s)
- Du-Hyong Cho
- From the School of Medicine (D.-H.C.), Konkuk University, Seoul, South Korea; Department of Molecular Medicine and Ewha Medical Research Institute (J.S., I.J.), Ewha Womans University Medical School, Seoul, South Korea; Department of Biotechnology (J.-H.P.), Korea University, Seoul, South Korea; Center for Biomedical Sciences (C.J.), National Institute of Health, Seoul, South Korea; Department of Biochemistry (Y.J.C.), Seoul National University School of Medicine, Seoul, South Korea; Department of
| | - Jungwon Seo
- From the School of Medicine (D.-H.C.), Konkuk University, Seoul, South Korea; Department of Molecular Medicine and Ewha Medical Research Institute (J.S., I.J.), Ewha Womans University Medical School, Seoul, South Korea; Department of Biotechnology (J.-H.P.), Korea University, Seoul, South Korea; Center for Biomedical Sciences (C.J.), National Institute of Health, Seoul, South Korea; Department of Biochemistry (Y.J.C.), Seoul National University School of Medicine, Seoul, South Korea; Department of
| | - Jung-Hyun Park
- From the School of Medicine (D.-H.C.), Konkuk University, Seoul, South Korea; Department of Molecular Medicine and Ewha Medical Research Institute (J.S., I.J.), Ewha Womans University Medical School, Seoul, South Korea; Department of Biotechnology (J.-H.P.), Korea University, Seoul, South Korea; Center for Biomedical Sciences (C.J.), National Institute of Health, Seoul, South Korea; Department of Biochemistry (Y.J.C.), Seoul National University School of Medicine, Seoul, South Korea; Department of
| | - Chulman Jo
- From the School of Medicine (D.-H.C.), Konkuk University, Seoul, South Korea; Department of Molecular Medicine and Ewha Medical Research Institute (J.S., I.J.), Ewha Womans University Medical School, Seoul, South Korea; Department of Biotechnology (J.-H.P.), Korea University, Seoul, South Korea; Center for Biomedical Sciences (C.J.), National Institute of Health, Seoul, South Korea; Department of Biochemistry (Y.J.C.), Seoul National University School of Medicine, Seoul, South Korea; Department of
| | - Yoon Jung Choi
- From the School of Medicine (D.-H.C.), Konkuk University, Seoul, South Korea; Department of Molecular Medicine and Ewha Medical Research Institute (J.S., I.J.), Ewha Womans University Medical School, Seoul, South Korea; Department of Biotechnology (J.-H.P.), Korea University, Seoul, South Korea; Center for Biomedical Sciences (C.J.), National Institute of Health, Seoul, South Korea; Department of Biochemistry (Y.J.C.), Seoul National University School of Medicine, Seoul, South Korea; Department of
| | - Jae-Won Soh
- From the School of Medicine (D.-H.C.), Konkuk University, Seoul, South Korea; Department of Molecular Medicine and Ewha Medical Research Institute (J.S., I.J.), Ewha Womans University Medical School, Seoul, South Korea; Department of Biotechnology (J.-H.P.), Korea University, Seoul, South Korea; Center for Biomedical Sciences (C.J.), National Institute of Health, Seoul, South Korea; Department of Biochemistry (Y.J.C.), Seoul National University School of Medicine, Seoul, South Korea; Department of
| | - Inho Jo
- From the School of Medicine (D.-H.C.), Konkuk University, Seoul, South Korea; Department of Molecular Medicine and Ewha Medical Research Institute (J.S., I.J.), Ewha Womans University Medical School, Seoul, South Korea; Department of Biotechnology (J.-H.P.), Korea University, Seoul, South Korea; Center for Biomedical Sciences (C.J.), National Institute of Health, Seoul, South Korea; Department of Biochemistry (Y.J.C.), Seoul National University School of Medicine, Seoul, South Korea; Department of
| |
Collapse
|
50
|
Titushkin IA, Rao VS, Pickard WF, Moros EG, Shafirstein G, Cho MR. Altered Calcium Dynamics Mediates P19-Derived Neuron-Like Cell Responses to Millimeter-Wave Radiation. Radiat Res 2009; 172:725-36. [DOI: 10.1667/rr1760.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|