101
|
Chen F, Li YM, Yang LQ, Zhong CG, Zhuang ZX. Association of NOS2 and NOS3 gene polymorphisms with susceptibility to type 2 diabetes mellitus and diabetic nephropathy in the Chinese Han population. IUBMB Life 2016; 68:516-25. [PMID: 27192959 DOI: 10.1002/iub.1513] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/20/2016] [Indexed: 11/10/2022]
Abstract
Inducible nitric oxide synthase (NOS2) and endothelial nitric oxide synthase (NOS3) gene play important roles in the susceptibility to type 2 diabetes mellitus (T2DM). The present study aims to detect the potential association of NOS2 and NOS3 gene polymorphisms with the susceptibility toT2DM and diabetic nephropathy (DN) in the Chinese Han population. Four hundred and ninety T2DM patients and 485 healthy controls were enrolled in this case-control study. The genotypes of NOS2 and NOS3 gene polymorphisms were analyzed by the polymerase chain reaction (PCR)-ligase detection reaction (LDR) method. Our data demonstrated that the NOS2 rs2779248 and NOS2 rs1137933 genetic polymorphisms were significantly associated with the increased susceptibility to T2DM in the heterozygote comparison, dominant model, and allele contrast; and NOS3 rs3918188 genetic polymorphism was significantly associated with the increased susceptibility to T2DM in the homozygote comparison and recessive model. The allele-C and genotype-TC of NOS2 rs2779248, allele-A and genotype-GA of NOS2 rs1137933 and genotype-AA of NOS3 rs3918188 genetic polymorphisms might be the risk factors for increasing the susceptibility to T2DM. And a significant haplotype effect of NOS2 rs10459953/C- rs1137933/G- rs2779248/T was found between T2DM cases and controls. Moreover, NOS3 rs1800783 polymorphism was significantly associated with the increased susceptibility to DN in the heterozygote comparison, recessive model and allele contrast. At last, a positive correlation of family history of diabetes with NOS3 rs11771443 polymorphism was found in DN. These preliminary findings indicate that the NOS2 rs2779248, NOS2 rs1137933, and NOS3 rs3918188 genetic polymorphisms are potentially related to the susceptibility to T2DM, and the rs1800783 polymorphism might be considered as genetic risk factors for diabetic nephropathy, and family history of diabetes was closely associated with rs11771443 polymorphism in DN, and the genetic variants might be used as molecular markers for evaluating the risk of T2DM and diabetic nephropathy. © 2016 IUBMB Life, 68(7):516-525, 2016.
Collapse
Affiliation(s)
- Feng Chen
- Department of Toxicology, XiangYa School of Public Health, Central South University, Changsha, Hunan, China.,Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China.,Department of Pharmacy, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu-Mei Li
- Department of Pharmacy, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lin-Qing Yang
- Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Cai-Gao Zhong
- Department of Toxicology, XiangYa School of Public Health, Central South University, Changsha, Hunan, China
| | - Zhi-Xiong Zhuang
- Department of Toxicology, XiangYa School of Public Health, Central South University, Changsha, Hunan, China.,Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| |
Collapse
|
102
|
Smith RM, Kruzliak P, Adamcikova Z, Zulli A. Role of Nox inhibitors plumbagin, ML090 and gp91ds-tat peptide on homocysteine thiolactone induced blood vessel dysfunction. Clin Exp Pharmacol Physiol 2016; 42:860-4. [PMID: 25998981 DOI: 10.1111/1440-1681.12427] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/29/2015] [Accepted: 05/15/2015] [Indexed: 02/05/2023]
Abstract
Antioxidants have not reduced the burden of cardiovascular disease, and current evidence suggests a beneficial role of oxidative stress, via NADPH oxidase (Nox) upregulation, in endothelial function. Homocysteine thiolactone (HcyT) induces blood vessel dysfunction and this correlates with increased vascular oxidative stress. This study aimed to determine if pharmacological inhibition of Nox could impair HcyT induced blood vessel dysfunction. Abdominal aorta were excised from New Zealand White rabbits (n = 6), cut into rings and sequentially mounted in organ baths. Rings were preincubated with 0.55 μmol/L homocysteine thiolactone for 1 h, or combinations of putative Nox inhibitors (plumbagin for Nox4, gp91ds-tat for Nox2, and ML090 for Nox1), 30 min prior to the addition of HcyT, followed by a dose response curve to acetylcholine on phenylephrine preconstricted rings. Plumbagin, ML090 + gp91ds-tat and HcyT reduced responses to acetylcholine, and Plumbagin + Hcyt caused constriction to acetylcholine, which was normalised to plumbagin by ML090. Plumbagin + ML090 or plumbagin + gp91ds-tat completely impaired the effect of acetylcholine. ML090 inhibited the effect of HcyT on reduced response to acetylcholine, whereas gp91ds-tat had no effect. This study concludes that inhibition of Nox1 prevents, whereas inhibition of Nox4 worsens, acetylcholine induced blood vessel relaxation caused by HcyT, while Nox2 inhibition has no effect. However combinations of Nox inhibitors worsen acetylcholine induced blood vessel relaxation. These results suggest that there is cross-talk between Nox isoforms during physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Renee M Smith
- Centre for Chronic Disease Prevention and Management, Western CHRE, College of Health and Biomedicine, Victoria University, St Albans, Vic., Australia
| | - Peter Kruzliak
- Department of Cardiovascular Diseases, International Clinical Research Center, St Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Zuzana Adamcikova
- Department of Public Health, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovak Republic
| | - Anthony Zulli
- Centre for Chronic Disease Prevention and Management, Western CHRE, College of Health and Biomedicine, Victoria University, St Albans, Vic., Australia
| |
Collapse
|
103
|
CTRP9 Ameliorates Pulmonary Arterial Hypertension Through Attenuating Inflammation and Improving Endothelial Cell Survival and Function. J Cardiovasc Pharmacol 2016; 67:394-401. [DOI: 10.1097/fjc.0000000000000364] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
104
|
Genetic Deletion of ACE2 Induces Vascular Dysfunction in C57BL/6 Mice: Role of Nitric Oxide Imbalance and Oxidative Stress. PLoS One 2016; 11:e0150255. [PMID: 27070147 PMCID: PMC4829150 DOI: 10.1371/journal.pone.0150255] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 02/11/2016] [Indexed: 12/18/2022] Open
Abstract
Accumulating evidence indicates that angiotensin-converting enzyme 2 (ACE2) plays a critical role in cardiovascular homeostasis, and its altered expression is associated with major cardiac and vascular disorders. The aim of this study was to evaluate the regulation of vascular function and assess the vascular redox balance in ACE2-deficient (ACE2-/y) animals. Experiments were performed in 20–22 week-old C57BL/6 and ACE2-/y male mice. Evaluation of endothelium-dependent and -independent relaxation revealed an impairment of in vitro and in vivo vascular function in ACE2-/y mice. Drastic reduction in eNOS expression at both protein and mRNA levels, and a decrease in •NO concentrations were observed in aortas of ACE2-/y mice in comparison to controls. Consistently, these mice presented a lower plasma and urine nitrite concentration, confirming reduced •NO availability in ACE2-deficient animals. Lipid peroxidation was significantly increased and superoxide dismutase activity was decreased in aorta homogenates of ACE2-/y mice, indicating impaired antioxidant capacity. Taken together, our data indicate, that ACE2 regulates vascular function by modulating nitric oxide release and oxidative stress. In conclusion, we elucidate mechanisms by which ACE2 is involved in the maintenance of vascular homeostasis. Furthermore, these findings provide insights into the role of the renin-angiotensin system in both vascular and systemic redox balance.
Collapse
|
105
|
Truran S, Weissig V, Madine J, Davies HA, Guzman-Villanueva D, Franco DA, Karamanova N, Burciu C, Serrano G, Beach TG, Migrino RQ. Nanoliposomes protect against human arteriole endothelial dysfunction induced by β-amyloid peptide. J Cereb Blood Flow Metab 2016; 36:405-12. [PMID: 26661197 PMCID: PMC4759678 DOI: 10.1177/0271678x15610134] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 09/02/2015] [Indexed: 12/13/2022]
Abstract
We tested whether nanoliposomes containing phosphatidylcholine, cholesterol and phosphatidic acid (NLPA) prevent β-amyloid 1-42 (Aβ42) fibrillation and Aβ42-induced human arteriole endothelial dysfunction. NLPA abolished Aβ42 fibril formation (thioflavin-T fluorescence/electron microscopy). In ex-vivo human adipose and leptomeningeal arterioles, Aβ42 impaired dilator response to acetylcholine that was reversed by NLPA; this protection was abolished by L-NG-nitroarginine methyl ester. Aβ42 reduced human umbilical vein endothelial cell NO production that was restored by NLPA. Nanoliposomes prevented Aβ42 amyloid formation, reversed Aβ42-induced human microvascular endothelial dysfunction and may be useful in Alzheimer's disease.
Collapse
Affiliation(s)
- Seth Truran
- Phoenix Veterans Affair Health Care System, Phoenix, AZ, USA
| | | | | | | | | | - Daniel A Franco
- Phoenix Veterans Affair Health Care System, Phoenix, AZ, USA
| | - Nina Karamanova
- Phoenix Veterans Affair Health Care System, Phoenix, AZ, USA
| | - Camelia Burciu
- Phoenix Veterans Affair Health Care System, Phoenix, AZ, USA
| | - Geidy Serrano
- Banner Sun Health Research Institute, Sun City, AZ, USA
| | | | - Raymond Q Migrino
- Phoenix Veterans Affair Health Care System, Phoenix, AZ, USA University of Arizona College of Medicine, Phoenix, AZ, USA
| |
Collapse
|
106
|
Kirby PL, Buerk DG, Parikh J, Barbee KA, Jaron D. Mathematical model for shear stress dependent NO and adenine nucleotide production from endothelial cells. Nitric Oxide 2016; 52:1-15. [PMID: 26529478 PMCID: PMC4703509 DOI: 10.1016/j.niox.2015.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 10/21/2015] [Accepted: 10/27/2015] [Indexed: 11/19/2022]
Abstract
We developed a mass transport model for a parallel-plate flow chamber apparatus to predict the concentrations of nitric oxide (NO) and adenine nucleotides (ATP, ADP) produced by cultured endothelial cells (ECs) and investigated how the net rates of production, degradation, and mass transport for these three chemical species vary with changes in wall shear stress (τw). These simulations provide an improved understanding of experimental results obtained with parallel-plate flow chambers and allows quantitative analysis of the relationship between τw, adenine nucleotide concentrations, and NO produced by ECs. Experimental data obtained after altering ATP and ADP concentrations with apyrase were analyzed to quantify changes in the rate of NO production (RNO). The effects of different isoforms of apyrase on ATP and ADP concentrations and nucleotide-dependent changes in RNO could be predicted with the model. A decrease in ATP was predicted with apyrase, but an increase in ADP was simulated due to degradation of ATP. We found that a simple proportional relationship relating a component of RNO to the sum of ATP and ADP provided a close match to the fitted curve for experimentally measured changes in RNO with apyrase. Estimates for the proportionality constant ranged from 0.0067 to 0.0321 μM/s increase in RNO per nM nucleotide concentration, depending on which isoform of apyrase was modeled, with the largest effect of nucleotides on RNO at low τw (<6 dyn/cm(2)).
Collapse
Affiliation(s)
- Patrick L Kirby
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Jaimit Parikh
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
107
|
Dormanns K, Brown RG, David T. The role of nitric oxide in neurovascular coupling. J Theor Biol 2016; 394:1-17. [PMID: 26796228 DOI: 10.1016/j.jtbi.2016.01.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/09/2015] [Accepted: 01/03/2016] [Indexed: 11/29/2022]
Abstract
Nitric oxide (NO) is a neurotransmitter known to act as a potent cerebral vasodilator. Its role in neurovascular coupling (NVC) is discussed controversially and one of the main unanswered questions is which cell type provides the governing source of NO for the regulation of vasodynamics. Mathematical modelling can be an appropriate tool to investigate the contribution of NO towards the key components of NVC and analyse underlying mechanisms. The lumped parameter model of a neurovascular unit, including neurons (NE), astrocytes (AC), smooth muscle cells (SMC) and endothelial cells (EC), was extended to model the NO signalling pathway. Results show that NO leads to a general shift of the resting regional blood flow by dilating the arteriolar radius. Furthermore, dilation during neuronal activation is enhanced. Simulations show that potassium release is responsible for the fast onset of vascular response, whereas NO-modulated mechanisms maintain dilation. Wall shear stress-activated NO release from the EC leads to a delayed return to the basal state of the arteriolar radius. The governing source of vasodilating NO that diffuses into the SMC, which determine the arteriolar radius, depends on neuronal activation. In the resting state the EC provides the major contribution towards vasorelaxation, whereas during neuronal stimulation NO produced by the NE dominates.
Collapse
Affiliation(s)
- K Dormanns
- UC HPC Unit, University of Canterbury, Christchurch, New Zealand
| | - R G Brown
- Institute of Fundamental Sciences, Massey University, Palmerston North, New Zealand
| | - T David
- UC HPC Unit, University of Canterbury, Christchurch, New Zealand.
| |
Collapse
|
108
|
Seo J, Cho DH, Lee HJ, Sung MS, Lee JY, Won KJ, Park JH, Jo I. Citron Rho-interacting kinase mediates arsenite-induced decrease in endothelial nitric oxide synthase activity by increasing phosphorylation at threonine 497: Mechanism underlying arsenite-induced vascular dysfunction. Free Radic Biol Med 2016; 90:133-44. [PMID: 26593676 DOI: 10.1016/j.freeradbiomed.2015.11.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 11/05/2015] [Accepted: 11/12/2015] [Indexed: 01/27/2023]
Abstract
We reported that arsenite causes an acute decrease in nitric oxide (NO) production by increasing phosphorylation of endothelial NO synthase at threonine 497 (eNOS-Thr(497)); however, the detailed mechanism has not yet been clarified. Here, we investigated the kinase involving in arsenite-stimulated eNOS-Thr(497) phosphorylation. Although treatment with H-89, a known protein kinase A (PKA) inhibitor, inhibited arsenite-stimulated eNOS-Thr(497) phosphorylation, no inhibition was found in cells treated with other PKA inhibitors, including Rp-8-Br-cAMPS or PKI. Based on previous reports, we also tested whether RhoA mediates arsenite-stimulated eNOS-Thr(497) phosphorylation and found that arsenite causes an acute increase in RhoA activity. Ectopic expression of dominant negative (DN)-RhoA significantly reversed arsenite-stimulated eNOS-Thr(497) phosphorylation. An in vitro phosphorylation assay also revealed that the well-known Rho effectors, Rho-associated protein kinase 1/2 (ROCK1/2), directly phosphorylate eNOS-Thr(497). Y27632, a selective ROCK inhibitor, reversed arsenite-stimulated eNOS-Thr(497) phosphorylation. However, overexpression of a small interfering RNA (siRNA) against ROCK1/2 or DN-ROCK did not reverse arsenite-stimulated eNOS-Thr(497) phosphorylation, thereby providing no conclusive evidence of a role for ROCK1/2. Knockdown of PKC-related protein kinase 1/2, another Rho effector, also did not reverse arsenite-stimulated eNOS-Thr(497) phosphorylation. In contrast, we found that transfection with an siRNA against citron Rho-interacting kinase (CRIK), the other downstream effector of Rho, significantly reversed the arsenite-induced eNOS-Thr(497) phosphorylation that was accompanied by restoration of eNOS enzymatic activity repressed by arsenite. Moreover, CRIK directly phosphorylated eNOS-Thr(497)in vitro. Finally, we also found that arsenite increased eNOS-Thr(497) phosphorylation and decreased acetylcholine-induced vessel relaxation in rat aortas. In conclusion, we demonstrate that arsenite acutely inhibits eNOS enzymatic activity and vessel relaxation in part by increasing the RhoA/CRIK/eNOS-Thr(497) phosphorylation signaling axis, which provides a molecular mechanism underlying arsenite-induced impaired vascular diseases.
Collapse
Affiliation(s)
- Jungwon Seo
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, South Korea; Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, Chonbuk, South Korea
| | - Du-Hyong Cho
- Department of Pharmacology, School of Medicine, Eulji University, Jung-gu, Daejeon 301-746, South Korea
| | - Hyeon-Ju Lee
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, South Korea
| | - Min-Sun Sung
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, South Korea
| | - Jee Young Lee
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, South Korea
| | - Kyung-Jong Won
- Department of Medical Science, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Jung-Hyun Park
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, South Korea
| | - Inho Jo
- Department of Molecular Medicine, Ewha Womans University Medical School, Yangcheon-gu, Seoul 158-710, South Korea.
| |
Collapse
|
109
|
Taguchi K, Hida M, Hasegawa M, Matsumoto T, Kobayashi T. Dietary polyphenol morin rescues endothelial dysfunction in a diabetic mouse model by activating the Akt/eNOS pathway. Mol Nutr Food Res 2015; 60:580-8. [DOI: 10.1002/mnfr.201500618] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/07/2015] [Accepted: 11/23/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry; Hoshi University; Tokyo Japan
| | - Mari Hida
- Department of Physiology and Morphology, Institute of Medicinal Chemistry; Hoshi University; Tokyo Japan
| | - Mami Hasegawa
- Department of Physiology and Morphology, Institute of Medicinal Chemistry; Hoshi University; Tokyo Japan
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry; Hoshi University; Tokyo Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry; Hoshi University; Tokyo Japan
| |
Collapse
|
110
|
Dexmedetomidine protects the heart against ischemia-reperfusion injury by an endothelial eNOS/NO dependent mechanism. Pharmacol Res 2015; 103:318-27. [PMID: 26607864 DOI: 10.1016/j.phrs.2015.11.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 10/27/2015] [Accepted: 11/10/2015] [Indexed: 02/06/2023]
Abstract
The alpha2-adrenergic receptor agonist Dexmedetomidine (Dex) is a sedative medication used by anesthesiologists. Dex protects the heart against ischemia-reperfusion (IR) and can also act as a preconditioning mimetic. The mechanisms involved in Dex-dependent cardiac preconditioning, and whether this action occurs directly or indirectly on cardiomyocytes, still remain unclear. The endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway and endothelial cells are known to play key roles in cardioprotection against IR injury. Therefore, the aims of this work were to evaluate whether the eNOS/NO pathway mediates the pharmacological cardiac effect of Dex, and whether endothelial cells are required in this cardioprotective action. Isolated adult rat hearts were treated with Dex (10nM) for 25min and the dimerization of eNOS and production of NO were measured. Hearts were then subjected to global IR (30/120min) and the role of the eNOS/NO pathway was evaluated. Dex promoted the activation of eNOS and production of NO. Dex reduced the infarct size and improved the left ventricle function recovery, but this effect was reversed when Dex was co-administered with inhibitors of the eNOS/NO/PKG pathway. In addition, Dex was unable to reduce cell death in isolated adult rat cardiomyocytes subjected to simulated IR. Cardiomyocyte death was attenuated by co-culturing them with endothelial cells pre-treated with Dex. In summary, our results show that Dex triggers cardiac protection by activating the eNOS/NO signaling pathway. This pharmacological effect of Dex requires its interaction with the endothelium.
Collapse
|
111
|
Clifton P. From sodium intake restriction to nitrate supplementation: Different measures with converging mechanistic pathways? Nutr Metab Cardiovasc Dis 2015; 25:1079-1086. [PMID: 26614018 DOI: 10.1016/j.numecd.2015.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 09/01/2015] [Accepted: 09/16/2015] [Indexed: 02/05/2023]
Abstract
Endothelial nitric oxide synthase is at the centre of endothelial physiology producing nitric oxide which dilates blood vessels, inhibits platelet aggregation and smooth muscle cell proliferation and reduces adhesion molecule production. The laminar shear stress is a common test used usually as the flow mediated dilatation test (FMD) which is sensitive to saturated fat, sodium and potassium although with the latter ion it is possible potassium has direct effects on ion channels in the smooth muscle cell as well as the endothelial cell. High blood pressure and blood cholesterol both reduce nitric oxide production, the latter probably by increasing caveolin-1 which binds nitric oxide synthase. Saturated fat reduces nitric oxide by elevating LDL cholesterol and caveolin-1 while insulin stimulates nitric oxide synthase activity by serine phosphorylation. Polyphenols from tea, coffee and cocoa and virgin olive oil enhance FMD and eNOS activity is essential for this activity. Wine polyphenols produce mixed results and it is not clear at present that they are beneficial. Blackberries and other polyphenol-rich fruit also enhance FMD. Dietary nitrate from beetroot and green leafy vegetables is converted to nitrite by salivary microbes and then to nitric oxide and this acts directly on the smooth muscle to lower blood pressure particularly in a low oxygen environment. Dietary nitrate also improves work efficiency and improves flow mediated dilatation.
Collapse
Affiliation(s)
- P Clifton
- University of South Australia, P5-16, GPO Box 2471, Adelaide SA 5000, Australia.
| |
Collapse
|
112
|
Carnitine palmitoyltransferase-1 up-regulation by PPAR-β/δ prevents lipid-induced endothelial dysfunction. Clin Sci (Lond) 2015; 129:823-37. [PMID: 26253087 DOI: 10.1042/cs20150111] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Fatty acids cause endothelial dysfunction involving increased ROS (reactive oxygen species) and reduced NO (nitric oxide) bioavailability. We show that in MAECs (mouse aortic endothelial cells), the PPARβ/δ (peroxisome- proliferator-activated receptor β/δ) agonist GW0742 prevented the decreased A23187-stimulated NO production, phosphorylation of eNOS (endothelial nitric oxide synthase) at Ser1177 and increased intracellular ROS levels caused by exposure to palmitate in vitro. The impaired endothelium-dependent relaxation to acetylcholine in mouse aorta induced by palmitate was restored by GW0742. In vivo, GW0742 treatment prevented the reduced aortic relaxation, phosphorylation of eNOS at Ser1177, and increased ROS production and NADPH oxidase in mice fed on a high-fat diet. The PPARβ/δ antagonist GSK0660 abolished all of these protective effects induced by GW0742. This agonist enhanced the expression of CPT (carnitine palmitoyltransferase)-1. The effects of GW0742 on acetylcholine- induced relaxation in aorta and on NO and ROS production in MAECs exposed to palmitate were abolished by the CPT-1 inhibitor etomoxir or by siRNA targeting CPT-1. GW0742 also inhibited the increase in DAG (diacylglycerol), PKCα/βII (protein kinase Cα/βII) activation, and phosphorylation of eNOS at Thr495 induced by palmitate in MAECs, which were abolished by etomoxir. In conclusion, PPARβ/δ activation restored the lipid-induced endothelial dysfunction by up-regulation of CPT-1, thus reducing DAG accumulation and the subsequent PKC-mediated ROS production and eNOS inhibition.
Collapse
|
113
|
Oliveira-Paula GH, Lacchini R, Tanus-Santos JE. Endothelial nitric oxide synthase: From biochemistry and gene structure to clinical implications of NOS3 polymorphisms. Gene 2015; 575:584-99. [PMID: 26428312 DOI: 10.1016/j.gene.2015.09.061] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/10/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is an important vasodilator with a well-established role in cardiovascular homeostasis. While mediator is synthesized from L-arginine by neuronal, endothelial, and inducible nitric oxide synthases (NOS1,NOS3 and NOS2 respectively), NOS3 is the most important isoform for NO formation in the cardiovascular system. NOS3 is a dimeric enzyme whose expression and activity are regulated at transcriptional, posttranscriptional,and posttranslational levels. The NOS3 gene, which encodes NOS3, exhibits a number of polymorphic sites including single nucleotide polymorphisms (SNPs), variable number of tandem repeats (VNTRs), microsatellites, and insertions/deletions. Some NOS3 polymorphisms show functional effects on NOS3 expression or activity, thereby affecting NO formation. Interestingly, many studies have evaluated the effects of functional NOS3 polymorphisms on disease susceptibility and drug responses. Moreover, some studies have investigated how NOS3 haplotypes may impact endogenous NO formation and disease susceptibility. In this article,we carried out a comprehensive review to provide a basic understanding of biochemical mechanisms involved in NOS3 regulation and how genetic variations in NOS3 may translate into relevant clinical and pharmacogenetic implications.
Collapse
Affiliation(s)
- Gustavo H Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
114
|
Vascular nitric oxide: Beyond eNOS. J Pharmacol Sci 2015; 129:83-94. [PMID: 26499181 DOI: 10.1016/j.jphs.2015.09.002] [Citation(s) in RCA: 494] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/11/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
As the first discovered gaseous signaling molecule, nitric oxide (NO) affects a number of cellular processes, including those involving vascular cells. This brief review summarizes the contribution of NO to the regulation of vascular tone and its sources in the blood vessel wall. NO regulates the degree of contraction of vascular smooth muscle cells mainly by stimulating soluble guanylyl cyclase (sGC) to produce cyclic guanosine monophosphate (cGMP), although cGMP-independent signaling [S-nitrosylation of target proteins, activation of sarco/endoplasmic reticulum calcium ATPase (SERCA) or production of cyclic inosine monophosphate (cIMP)] also can be involved. In the blood vessel wall, NO is produced mainly from l-arginine by the enzyme endothelial nitric oxide synthase (eNOS) but it can also be released non-enzymatically from S-nitrosothiols or from nitrate/nitrite. Dysfunction in the production and/or the bioavailability of NO characterizes endothelial dysfunction, which is associated with cardiovascular diseases such as hypertension and atherosclerosis.
Collapse
|
115
|
Barrera-Chimal J, Prince S, Fadel F, El Moghrabi S, Warnock DG, Kolkhof P, Jaisser F. Sulfenic Acid Modification of Endothelin B Receptor is Responsible for the Benefit of a Nonsteroidal Mineralocorticoid Receptor Antagonist in Renal Ischemia. J Am Soc Nephrol 2015; 27:398-404. [PMID: 26361797 DOI: 10.1681/asn.2014121216] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 04/07/2015] [Indexed: 12/13/2022] Open
Abstract
AKI is associated with high mortality rates and the development of CKD. Ischemia/reperfusion (IR) is an important cause of AKI. Unfortunately, there is no available pharmacologic approach to prevent or limit renal IR injury in common clinical practice. Renal IR is characterized by diminished nitric oxide bioavailability and reduced renal blood flow; however, the mechanisms leading to these alterations are poorly understood. In a rat model of renal IR, we investigated whether the administration of the novel nonsteroidal mineralocorticoid receptor (MR) antagonist BR-4628 can prevent or treat the renal dysfunction and tubular injury induced by IR. Renal injury induced by ischemia was associated with increased oxidant damage, which led to a cysteine sulfenic acid modification in endothelin B receptor and consequently decreased endothelial nitric oxide synthase activation. These modifications were efficiently prevented by nonsteroidal MR antagonism. Furthermore, we demonstrated that the protective effect of BR-4628 against IR was lost when a selective endothelin B receptor antagonist was coadministered. These data describe a new mechanism for reduced endothelial nitric oxide synthase activation during renal IR that can be blocked by MR antagonism with BR-4628.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, Paris, France
| | - Sonia Prince
- INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, Paris, France
| | - Fouad Fadel
- INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, Paris, France
| | - Soumaya El Moghrabi
- INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, Paris, France
| | - David G Warnock
- University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Peter Kolkhof
- Bayer Healthcare, Cardiology Research, Wuppertal, Germany
| | - Frédéric Jaisser
- INSERM, UMRS 1138, Team 1, Centre de Recherche des Cordeliers, Pierre et Marie Curie University, Paris Descartes University, Paris, France
| |
Collapse
|
116
|
Han L, Su L, Chen D, Zhang S, Zhang Y, Zhao B, Zhao J, Miao J. ZnS nanoarchitectures induced dysfunction of vascular endothelial cells in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY 2015; 30:755-768. [PMID: 24449189 DOI: 10.1002/tox.21955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 01/07/2014] [Accepted: 01/08/2014] [Indexed: 06/03/2023]
Abstract
ZnS nanoarchitectures have been intensively investigated recently because of their applications in optoelectronics and adsorption capacity. The potential hazard of ZnS nanoarchitectures is not well known. In this study, we investigated the toxicity of ZnS nanoarchitectures on vascular endothelial cell (VEC) in vitro and in vivo. The results showed that ZnS could inhibit human umbilical vein endothelial cell (HUVEC) proliferation at 50 and 200 μg/mL. Endothelial nitric oxide synthase (eNOS) activity, nitric oxide (NO), and reactive oxygen species productions were increased, which was companied with the decrease in caveolin-1 level. The endothelium of the aortic root was damaged and the NO levels in serum were elevated in the mice treated with 5 or 10 mg/kg ZnS for 3 and 6 days, but the body could repair the damage. The data suggested that the high concentration of ZnS could induce dysfunction of VECs through decreasing caveolin-1 and elevation of the eNOS activity and thus present toxicity.
Collapse
Affiliation(s)
- Lei Han
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China
| | - Le Su
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China
| | - Dagui Chen
- Key Laboratory of Optoelectronic Materials Chemistry and Physics, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
| | - ShangLi Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, 250012, China
| | - BaoXiang Zhao
- Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Jing Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China
| | - JunYing Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, 250012, China
| |
Collapse
|
117
|
Mutchler SM, Straub AC. Compartmentalized nitric oxide signaling in the resistance vasculature. Nitric Oxide 2015; 49:8-15. [PMID: 26028569 DOI: 10.1016/j.niox.2015.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 01/23/2023]
Abstract
Nitric oxide (NO) was first described as a bioactive molecule through its ability to stimulate soluble guanylate cyclase, but the revelation that NO was the endothelium derived relaxation factor drove the field to its modern state. The wealth of research conducted over the past 30 years has provided us with a picture of how diverse NO signaling can be within the vascular wall, going beyond simple vasodilation to include such roles as signaling through protein S-nitrosation. This expanded view of NO's actions requires highly regulated and compartmentalized production. Importantly, resistance arteries house multiple proteins involved in the production and transduction of NO allowing for efficient movement of the molecule to regulate vascular tone and reactivity. In this review, we focus on the many mechanisms regulating NO production and signaling action in the vascular wall, with a focus on the control of endothelial nitric oxide synthase (eNOS), the enzyme responsible for synthesizing most of the NO within these confines. We also explore how cross talk between the endothelium and smooth muscle in the microcirculation can modulate NO signaling, illustrating that this one small molecule has the capability to produce a plethora of responses.
Collapse
Affiliation(s)
- Stephanie M Mutchler
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|
118
|
Gonzalez C, Rosas-Hernandez H, Jurado-manzano B, Ramirez-Lee MA, Salazar-Garcia S, Martinez-Cuevas PP, Velarde-salcedo AJ, Morales-Loredo H, Espinosa-Tanguma R, Ali SF, Rubio R. The prolactin family hormones regulate vascular tone through NO and prostacyclin production in isolated rat aortic rings. Acta Pharmacol Sin 2015; 36:572-86. [PMID: 25891087 DOI: 10.1038/aps.2014.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 09/26/2014] [Indexed: 01/17/2023] Open
Abstract
AIM Prolactin family hormones include growth hormone, placental lactogen and prolactin, which are able to regulate angiogenesis via NO and prostaglandins. However, their effects on vascular tone are not fully understood. The aim of this study was to evaluate the effects of prolactin family hormones on rat vascular tone in vitro. METHODS Aortic rings were prepared from adult male rats and precontracted with phenylephrine, then treated with the hormones and drugs. The tension was measured with isometric force displacement transducer connected to a polygraph. NO production and prostacyclin release in physiological solution was determined. Cultured rat aortic endothelial cells (RAECs) were treated with the hormones and drugs, and the phosphorylation of eNOS at serine 1177 was assessed using Western bolt analysis. RESULTS Administration of growth hormone or placental lactogen (0.01-100 nmol/L) induced endothelium-dependent vasodilation. Both the hormones significantly increased the phosphorylation of eNOS in RAECs and NO level in physiological solution. Preincubation with L-NAME blocked growth hormone- or placental lactogen-induced vasodilation and NO production. Preincubation with an antibody against growth hormone receptors blocked growth hormone- and placental lactogen-induced vasodilation. Addition of a single dose of prolactin (0.01 nmol/L) induced sustained vessel relaxation, whereas multiple doses of prolactin induced a biphasic contraction-relaxation effect. The vascular effects of prolactin depended on endothelium. Prolactin significantly increased the level of prostacyclin I2 in physiological solution. Preincubation with indomethacin or an antibody against prolactin receptors blocked prolactin-induced vasodilation. CONCLUSION The prolactin family hormones regulate rat vascular tone, selectively promoting either relaxation or contraction of vascular smooth muscle via activation of either growth hormone receptors or prolactin receptors within the endothelium.
Collapse
|
119
|
Glushko AA, Voronkov AV, Chernikov MV. [Molecular targets for searching of endothelial-protective substances]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2015; 40:515-27. [PMID: 25895347 DOI: 10.1134/s1068162014050069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Endothelial dysfunction underlies the development of many cardiovascular diseases. Thus endothelium becomes an independent therapeutic target, and the search of new substances with endothelial-protective action to date is one of the promising tasks for pharmacotherapy and medicinal chemistry. Molecular modeling is an effective tool for solving this problem. Computer chemistry methods use is only possible in combination with detailed information on three dimensional structure and functions of molecular targets: receptors and enzymes, involved in signal transduction inside and outside of endothelial cells. Information on structure and function of various macromolecules involved in vascular tone regulation is collected in the review. The structure of endothelial NO-synthase (EC 1.14.13.39) (eNOS)--enzyme, responsible for the nitric oxide synthesis and involved in vascular tone regulation process is reviewed. The importance of eNOS substrate--L-arginine is underlined in the review in terms of this enzyme activity, regulation, the information on structure and functions of L-arginine transport system is provided. Also different ways of eNOS activity regulation are reviewed, among which are enzyme activation and concurrent inhibition by substances interaction with active center of enzyme, inhibition by caveoline binding with oxigenase domain, and also regulation by phosphorylation of certain amino acids of eNOS by proteinkinase and dephoshphorylation of them by phosphatases. The importance of membrane receptors of endothelial cells as targets for endothelial-protective substances is underlined. Among them are receptors of endothelin, platelet activation factor, prostanoids, bradykinin, histamine, serotonin and protease activated receptors. The important role of potassium and calcium ion channels of vascular cells in endothelial-protective activity is underlined. Macromolecules presented in the review finally are considered as targets for searching for medicinal substances with endothelial-protective activity using proposed ways and methods of molecular modeling.
Collapse
|
120
|
Cocks M, Wagenmakers AJM. The effect of different training modes on skeletal muscle microvascular density and endothelial enzymes controlling NO availability. J Physiol 2015; 594:2245-57. [PMID: 25809076 DOI: 10.1113/jp270329] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/16/2015] [Indexed: 01/12/2023] Open
Abstract
It is becoming increasingly apparent that a high vasodilator response of the skeletal muscle microvasculature to insulin and exercise is of critical importance for adequate muscle perfusion and long-term microvascular and muscle metabolic health. Previous research has shown that a sedentary lifestyle, obesity and ageing lead to impairments in the vasodilator response, while a physically active lifestyle keeps both microvascular density and vasodilator response high. To investigate the molecular mechanisms behind these impairments and the benefits of exercise training interventions, our laboratory has recently developed quantitative immunofluorescence microscopy methods to measure protein content of eNOS and NAD(P)Hoxidase specifically in the endothelial layer of capillaries and arterioles of human skeletal muscle. As eNOS produces nitric oxide (NO) and NAD(P)Hoxidase produces superoxide anions (O2 (-) , quenching NO) we propose that the eNOS/NAD(P)Hoxidase protein ratio is a marker of vasodilator capacity. The novel methods show that endurance training (ET) and high intensity interval training (HIT), generally regarded as a time-efficient alternative to ET, increase eNOS protein content and the eNOS/NADP(H)oxidase protein ratio in previously sedentary lean and obese young men. Resistance exercise training had smaller but qualitatively similar effects. Western blot data of other laboratories suggest that endurance exercise training leads to similar changes in sedentary elderly men. Future research will be required to investigate the relative importance of other sources and tissues in the balance between NO and O2 (-) production seen by the vascular smooth muscle layer of terminal arterioles.
Collapse
Affiliation(s)
- Matthew Cocks
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool, L3 3AF, UK
| | - Anton J M Wagenmakers
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool, L3 3AF, UK
| |
Collapse
|
121
|
Huseynova SA, Panakhova NF, Orujova PA, Hasanov SS, Guliyev MR, Yagubova VI. Altered endothelial nitric oxide synthesis in preterm and small for gestational age infants. Pediatr Int 2015; 57:269-75. [PMID: 25294660 DOI: 10.1111/ped.12520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 09/14/2014] [Accepted: 09/17/2014] [Indexed: 12/01/2022]
Abstract
BACKGROUND Preterm infants are often exposed to neuronal and endothelial damage. The aim of the present study was to investigate the correlation between endothelial dysfunction and neuronal injury in preterm infants. METHODS We compared serum nitric oxide (NO), endothelial nitric oxide synthase (eNOS) and neuron-specific enolase (NSE) concentrations in 33 moderate preterm (MP) and 47 late preterm (LP) infants using standard ELISA. Each group was classified as appropriate for gestational age (AGA) or small for gestational age (SGA). RESULTS Compared to the AGA infants, the SGA infants had higher NO on day 1 (MP: mean, 72.3 ng/mL, range, 50.9-99.5 ng/mL vs 52.2 ng/mL, range, 28.1-68.2 ng/mL, P < 0.05; LP: mean, 58.4 ng/mL, range, 25.7-66.4 ng/mL vs 43.7 ng/mL, range, 21.2-60.6 ng/mL, P < 0.05), lower eNOS concentration on day 3 in the MP group (mean, 5.8 IU/mL, range, 1.2-7.9 IU/mL vs 8.9 IU/mL, range, 4.2-14.6 IU/mL, P < 0.05), and on day 1 in the LP group (mean, 5.5 IU/mL, range, 1.5-8.1 IU/mL vs 7.7 IU/mL, range, 4.4-13.8 IU/mL, P < 0.05). The NO/eNOS ratio was higher in SGA infants compared with the AGA subgroups (MP: mean, 13.8, range, 9.9-20.2 vs mean, 9.9, range, 4.7-13.1, P < 0.05; LP: mean, 12.2, range, 9.2-19.9 vs mean, 9.9, range, 5.4-14.4, P < 0.05). AGA infants had lower NSE concentration compared with the SGA infants on day 1 in the LP group (mean, 27.4 ng/mL, range, 20-43 ng/mL vs mean, 40.89 ng/mL, range, 34-51 ng/mL, P < 0.05). A positive correlation was found between NO/eNOS ratio and NSE concentration (r = 0.75, P < 0.05 and r = 0.64, P < 0.05 on days 1 and 3, respectively). CONCLUSION High NO concentration in the context of low eNOS activity suggests a possible role of NO in the development of neuronal injury in SGA infants.
Collapse
|
122
|
Nano-Mg(OH)2-induced proliferation inhibition and dysfunction of human umbilical vein vascular endothelial cells through caveolin-1-mediated endocytosis. Cell Biol Toxicol 2015; 31:15-27. [PMID: 25575676 DOI: 10.1007/s10565-014-9291-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 12/22/2014] [Indexed: 12/17/2022]
Abstract
Nano-Mg(OH)2 is efficiently used in pollutant adsorption and removal due to its high adsorption capability, low-cost, and recyclability. A recent research from our group showed that Mg(OH)2 nanoflakes are not evidently internalized by cancer cells and are not cytotoxic. But the biocompatibility and potential toxicity of nano-Mg(OH)2 in a normal biological system are largely unclear. Nanoparticles could affect the function of endothelial cells, and endothelial dysfunction represents an early sign of lesion within the vasculature. Here, we applied the human umbilical vein vascular endothelial cells (HUVECs) as an in vitro model of the endothelium to study the cytotoxicity of nano-Mg(OH)2. Our results showed that nano-Mg(OH)2 at 200 μg/ml impaired proliferation and induced dysfunction of HUVECs, but did not result in cell necrosis and apoptosis. Transmission electron microscopy images and immunofluorescence results showed that the nano-Mg(OH)2 could enter HUVECs through caveolin-1-mediated endocytosis. Nano-Mg(OH)2 at high concentrations decreased the level of caveolin-1 and increased the activity of endothelial nitric oxide synthase (eNOS), thus leading to the production of excess nitric oxide (NO). In this work, we provide the cell damage concentrations of nano-Mg(OH)2 nanoparticles, and we propose a mechanism of injury induced by nano-Mg(OH)2 in HUVECs.
Collapse
|
123
|
Activation of eNOS in endothelial cells exposed to ionizing radiation involves components of the DNA damage response pathway. Biochem Biophys Res Commun 2015; 456:541-6. [DOI: 10.1016/j.bbrc.2014.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/01/2014] [Indexed: 01/29/2023]
|
124
|
Seo J, Lee JY, Sung MS, Byun CJ, Cho DH, Lee HJ, Park JH, Cho HS, Cho SJ, Jo I. Arsenite Acutely Decreases Nitric Oxide Production via the ROS-Protein Phosphatase 1-Endothelial Nitric Oxide Synthase-Thr(497) Signaling Cascade. Biomol Ther (Seoul) 2014; 22:510-8. [PMID: 25489418 PMCID: PMC4256030 DOI: 10.4062/biomolther.2014.106] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 11/17/2022] Open
Abstract
Chronic (>24 h) exposure of arsenite, an environmental toxicant, has shown the decreased nitric oxide (NO) production in endothelial cells (EC) by decreasing endothelial NO synthase (eNOS) expression and/or its phosphorylation at serine 1179 (eNOS-Ser1179 in bovine sequence), which is associated with increased risk of vascular diseases. Here, we investigated the acute (<24 h) effect of arsenite on NO production using bovine aortic EC (BAEC). Arsenite acutely increased the phosphorylation of eNOS-Thr497, but not of eNOS-Ser116 or eNOS-Ser1179, which was accompanied by decreased NO production. The level of eNOS expression was unaltered under this condition. Treatment with arsenite also induced reactive oxygen species (ROS) production, and pretreatment with a ROS scavenger N-acetyl-L-cysteine (NAC) completely reversed the observed effect of arsenite on eNOS-Thr497 phosphorylation. Although protein kinase C (PKC) and protein phosphatase 1 (PP1) were reported to be involved in eNOS-Thr497 phosphorylation, treatment with PKC inhibitor, Ro318425, and overexpression of various PKC isoforms did not affect the arsenite-stimulated eNOS-Thr497 phosphorylation. In contrast, treatment with PP1 inhibitor, calyculin A, mimicked the observed effect of arsenite on eNOS-Thr497 phosphorylation. Lastly, we found decreased cellular PP1 activity in arsenite-treated cells, which was reversed by NAC. Overall, our study demonstrates firstly that arsenite acutely decreases NO production at least in part by increasing eNOS-Thr497 phosphorylation via ROS-PP1 signaling pathway, which provide the molecular mechanism underlying arsenite-induced increase in vascular disease.
Collapse
Affiliation(s)
- Jungwon Seo
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710 ; Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan 570-749
| | - Jee Young Lee
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| | - Min-Sun Sung
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| | | | - Du-Hyong Cho
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 301-768
| | - Hyeon-Ju Lee
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| | - Jung-Hyun Park
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| | - Ho-Seong Cho
- Biosafety Research Institute and College of Veterinary Medicine, Chonbuk National University, Jeonju 561-756
| | - Sung-Jin Cho
- Department of Biology, College of Natural Sciences, Chungbuk National University, Cheongju 362-763, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710
| |
Collapse
|
125
|
Shao Y, Cheng Z, Li X, Chernaya V, Wang H, Yang XF. Immunosuppressive/anti-inflammatory cytokines directly and indirectly inhibit endothelial dysfunction--a novel mechanism for maintaining vascular function. J Hematol Oncol 2014; 7:80. [PMID: 25387998 PMCID: PMC4236671 DOI: 10.1186/s13045-014-0080-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022] Open
Abstract
Endothelial dysfunction is a pathological status of the vascular system, which can be broadly defined as an imbalance between endothelium-dependent vasoconstriction and vasodilation. Endothelial dysfunction is a key event in the progression of many pathological processes including atherosclerosis, type II diabetes and hypertension. Previous reports have demonstrated that pro-inflammatory/immunoeffector cytokines significantly promote endothelial dysfunction while numerous novel anti-inflammatory/immunosuppressive cytokines have recently been identified such as interleukin (IL)-35. However, the effects of anti-inflammatory cytokines on endothelial dysfunction have received much less attention. In this analytical review, we focus on the recent progress attained in characterizing the direct and indirect effects of anti-inflammatory/immunosuppressive cytokines in the inhibition of endothelial dysfunction. Our analyses are not only limited to the importance of endothelial dysfunction in cardiovascular disease progression, but also expand into the molecular mechanisms and pathways underlying the inhibition of endothelial dysfunction by anti-inflammatory/immunosuppressive cytokines. Our review suggests that anti-inflammatory/immunosuppressive cytokines serve as novel therapeutic targets for inhibiting endothelial dysfunction, vascular inflammation and cardio- and cerebro-vascular diseases.
Collapse
Affiliation(s)
- Ying Shao
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Zhongjian Cheng
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Xinyuan Li
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Valeria Chernaya
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Hong Wang
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Xiao-feng Yang
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA. .,Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|
126
|
Adekola H, Romero R, Chaemsaithong P, Korzeniewski SJ, Dong Z, Yeo L, Hassan SS, Chaiworapongsa T. Endocan, a putative endothelial cell marker, is elevated in preeclampsia, decreased in acute pyelonephritis, and unchanged in other obstetrical syndromes. J Matern Fetal Neonatal Med 2014; 28:1621-32. [PMID: 25211383 PMCID: PMC4412749 DOI: 10.3109/14767058.2014.964676] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objective: Endocan, a dermatan sulphate proteoglycan produced by endothelial cells, is considered a biomarker for endothelial cell activation/dysfunction. Preeclampsia is characterized by systemic vascular inflammation, and endothelial cell activation/dysfunction. Therefore, the objectives of this study were to determine whether: (1) plasma endocan concentrations in preeclampsia differ from those in uncomplicated pregnancies; (2) changes in plasma endocan concentration relate to the severity of preeclampsia, and whether these changes are specific or observed in other obstetrical syndromes such as small-for-gestational age (SGA), fetal death (FD), preterm labor (PTL) or preterm prelabor rupture of membranes (PROM); (3) a correlation exists between plasma concentration of endocan and angiogenic (placental growth factor or PlGF)/anti-angiogenic factors (soluble vascular endothelial growth factor receptor or sVEGFR-1, and soluble endoglin or sEng) among pregnancies complicated by preeclampsia; and (4) plasma endocan concentrations in patients with preeclampsia and acute pyelonephritis (both conditions in which there is endothelial cell activation) differ. Method: This cross-sectional study included the following groups: (1) uncomplicated pregnancy (n = 130); (2) preeclampsia (n = 102); (3) pregnant women without preeclampsia who delivered an SGA neonate (n = 51); (4) FD (n = 49); (5) acute pyelonephritis (AP; n = 35); (6) spontaneous PTL (n = 75); and (7) preterm PROM (n = 64). Plasma endocan concentrations were determined in all groups, and PIGF, sEng and VEGFR-1 plasma concentrations were measured by ELISA in the preeclampsia group. Results: (1) Women with preeclampsia had a significantly higher median plasma endocan concentration than those with uncomplicated pregnancies (p = 0.004); (2) among women with preeclampsia, the median plasma endocan concentration did not differ significantly according to disease severity (p = 0.1), abnormal uterine artery Doppler velocimetry (p = 0.7) or whether diagnosis was made before or after 34 weeks gestational age (p = 0.3); (3) plasma endocan concentration in women with preeclampsia correlated positively with plasma anti-angiogenic factor concentrations [sVEGFR-1: Spearman rho 0.34, p = 0.001 and sEng: Spearman rho 0.30, p = 0.003]; (4) pregnancies complicated by acute pyelonephritis with bacteremia had a lower median plasma endocan concentration than pregnancies complicated by acute pyelonephritis without bacteremia (p = 0.004), as well as uncomplicated pregnancies (p = 0.001); and (5) there was no significant difference in the median plasma endocan concentration between uncomplicated pregnancies and those complicated by FD, delivery of an SGA neonate, PTL or preterm PROM (other members of the “great obstetrical syndromes”; each p > 0.05). Conclusion: Median maternal plasma endocan concentrations were higher preeclampsia and lower in acute pyelonephritis with bacteremia than in uncomplicated pregnancy. No significant difference was observed in the median plasma endocan concentration between other great obstetrical syndromes and uncomplicated pregnancies. The difference in the direction of change of endocan in preeclampsia and acute pyelonephritis with bacteremia may be consistent with the view that both disease entities differ in pathogenic mechanisms, despite their associations with systemic vascular inflammation and endothelial cell activation/dysfunction.
Collapse
Affiliation(s)
- Henry Adekola
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH , Bethesda , MD (Detroit, MI) , USA
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Molecular modeling and simulation of the human eNOS reductase domain, an enzyme involved in the release of vascular nitric oxide. J Mol Model 2014; 20:2470. [DOI: 10.1007/s00894-014-2470-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 09/14/2014] [Indexed: 01/19/2023]
|
128
|
Zamberlam G, Sahmi F, Price CA. Nitric oxide synthase activity is critical for the preovulatory epidermal growth factor-like cascade induced by luteinizing hormone in bovine granulosa cells. Free Radic Biol Med 2014; 74:237-44. [PMID: 24992832 DOI: 10.1016/j.freeradbiomed.2014.06.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 06/16/2014] [Accepted: 06/19/2014] [Indexed: 01/22/2023]
Abstract
In rabbits and rodents, nitric oxide (NO) is generally considered to be critical for ovulation. In monovulatory species, however, the importance of NO has not been determined, nor is it clear where in the preovulatory cascade NO may act. The objectives of this study were (1) to determine if nitric oxide synthase (NOS) enzymes are regulated by luteinizing hormone (LH) and (2) to determine if and where endogenous NO is critical for expression of genes essential for the ovulatory cascade in bovine granulosa cells in serum-free culture. Time- and dose-response experiments demonstrated that LH had a significant stimulatory effect on endothelial NOS (NOS3) mRNA abundance, but in a prostaglandin-dependent manner. NO production was stimulated by LH before a detectable increase in NOS3 mRNA levels was observed. Pretreatment of cells with the NOS inhibitor L-NAME blocked the effect of LH on the epidermal growth factor (EGF)-like ligands epiregulin and amphiregulin, as well as prostaglandin-endoperoxide synthase-2 mRNA abundance and protein levels. Similarly, EGF treatment increased mRNA encoding epiregulin, amphiregulin, and the early response gene EGR1, and this was inhibited by pretreatment with L-NAME. Interestingly, pretreatment with L-NAME had no effect on either ERK1/2 or AKT activation. Taken together, these results suggest that endogenous NOS activity is critical for the LH-induced ovulatory cascade in granulosa cells of a monotocous species and acts downstream of EGF receptor activation but upstream of the EGF-like ligands.
Collapse
Affiliation(s)
- Gustavo Zamberlam
- Centre de Recherche en Reproduction Animale, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC J2S 7C6, Canada
| | - Fatiha Sahmi
- Centre de Recherche en Reproduction Animale, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC J2S 7C6, Canada
| | - Christopher A Price
- Centre de Recherche en Reproduction Animale, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC J2S 7C6, Canada.
| |
Collapse
|
129
|
Endothelial nitric oxide synthase dimerization is regulated by heat shock protein 90 rather than by phosphorylation. PLoS One 2014; 9:e105479. [PMID: 25153129 PMCID: PMC4143281 DOI: 10.1371/journal.pone.0105479] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 07/23/2014] [Indexed: 11/29/2022] Open
Abstract
Endothelial nitric oxide synthase (eNOS) is a multifunctional enzyme with roles in diverse cellular processes including angiogenesis, tissue remodeling, and the maintenance of vascular tone. Monomeric and dimeric forms of eNOS exist in various tissues. The dimeric form of eNOS is considered the active form and the monomeric form is considered inactive. The activity of eNOS is also regulated by many other mechanisms, including amino acid phosphorylation and interactions with other proteins. However, the precise mechanisms regulating eNOS dimerization, phosphorylation, and activity remain incompletely characterized. We utilized purified eNOS and bovine aorta endothelial cells (BAECs) to investigate the mechanisms regulating eNOS degradation. Both eNOS monomer and dimer existed in purified bovine eNOS. Incubation of purified bovine eNOS with protein phosphatase 2A (PP2A) resulted in dephosphorylation at Serine 1179 (Ser1179) in both dimer and monomer and decrease in eNOS activity. However, the eNOS dimer∶monomer ratio was unchanged. Similarly, protein phosphatase 1 (PP1) induced dephosphorylation of eNOS at Threonine 497 (Thr497), without altering the eNOS dimer∶monomer ratio. Different from purified eNOS, in cultured BAECs eNOS existed predominantly as dimers. However, eNOS monomers accumulated following treatment with the proteasome inhibitor lactacystin. Additionally, treatment of BAECs with vascular endothelial growth factor (VEGF) resulted in phosphorylation of Ser1179 in eNOS dimers without altering the phosphorylation status of Thr497 in either form. Inhibition of heat shock protein 90 (Hsp90) or Hsp90 silencing destabilized eNOS dimers and was accompanied by dephosphorylation both of Ser1179 and Thr497. In conclusion, our study demonstrates that eNOS monomers, but not eNOS dimers, are degraded by ubiquitination. Additionally, the dimeric eNOS structure is the predominant condition for eNOS amino acid modification and activity regulation. Finally, destabilization of eNOS dimers not only results in eNOS degradation, but also causes changes in eNOS amino acid modifications that further affect eNOS activity.
Collapse
|
130
|
Sun X, Kumar S, Sharma S, Aggarwal S, Lu Q, Gross C, Rafikova O, Lee SG, Dasarathy S, Hou Y, Meadows ML, Han W, Su Y, Fineman JR, Black SM. Endothelin-1 induces a glycolytic switch in pulmonary arterial endothelial cells via the mitochondrial translocation of endothelial nitric oxide synthase. Am J Respir Cell Mol Biol 2014; 50:1084-95. [PMID: 24392990 DOI: 10.1165/rcmb.2013-0187oc] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recent studies have indicated that, during the development of pulmonary hypertension (PH), there is a switch from oxidative phosphorylation to glycolysis in the pulmonary endothelium. However, the mechanisms underlying this phenomenon have not been elucidated. Endothelin (ET)-1, an endothelial-derived vasoconstrictor peptide, is increased in PH, and has been shown to play an important role in the oxidative stress associated with PH. Thus, in this study, we investigated whether there was a potential link between increases in ET-1 and mitochondrial remodeling. Our data indicate that ET-1 induces the redistribution of endothelial nitric oxide synthase (eNOS) from the plasma membrane to the mitochondria in pulmonary arterial endothelial cells, and that this was dependent on eNOS uncoupling. We also found that ET-1 disturbed carnitine metabolism, resulting in the attenuation of mitochondrial bioenergetics. However, ATP levels were unchanged due to a compensatory increase in glycolysis. Further mechanistic investigations demonstrated that ET-1 mediated the redistribution of eNOS via the phosphorylation of eNOS at Thr495 by protein kinase C δ. In addition, the glycolytic switch appeared to be dependent on mitochondrial-derived reactive oxygen species that led to the activation of hypoxia-inducible factor signaling. Finally, the cell culture data were confirmed in vivo using the monocrotaline rat model of PH. Thus, we conclude that ET-1 induces a glycolytic switch in pulmonary arterial endothelial cells via the redistribution of uncoupled eNOS to the mitochondria, and that preventing this event may be an approach for the treatment of PH.
Collapse
Affiliation(s)
- Xutong Sun
- 1 Pulmonary Disease Program, Vascular Biology Center, and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
The N-terminal portion of autoinhibitory element modulates human endothelial nitric-oxide synthase activity through coordinated controls of phosphorylation at Thr495 and Ser1177. Biosci Rep 2014; 34:BSR20140079. [PMID: 24993645 PMCID: PMC4122979 DOI: 10.1042/bsr20140079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
NO production catalysed by eNOS (endothelial nitric-oxide synthase) plays an important role in the cardiovascular system. A variety of agonists activate eNOS through the Ser1177 phosphorylation concomitant with Thr495 dephosphorylation, resulting in increased ·NO production with a basal level of calcium. To date, the underlying mechanism remains unclear. We have previously demonstrated that perturbation of the AIE (autoinhibitory element) in the FMN-binding subdomain can also lead to eNOS activation with a basal level of calcium, implying that the AIE might regulate eNOS activation through modulating phosphorylation at Thr495 and Ser1177. Here we generated stable clones in HEK-293 (human embryonic kidney 293) cells with a series of deletion mutants in both the AIE (Δ594-604, Δ605-612 and Δ626-634) and the C-terminal tail (Δ14; deletion of 1164-1177). The expression of Δ594-604 and Δ605-612 mutants in non-stimulated HEK-293 cells substantially increased nitrate/nitrite release into the culture medium; the other two mutants, Δ626-634 and Δ1164-1177, displayed no significant difference when compared with WTeNOS (wild-type eNOS). Intriguingly, mutant Δ594-604 showed close correlation between Ser1177 phosphorylation and Thr495 dephosphorylation, and NO production. Our results have indicated that N-terminal portion of AIE (residues 594-604) regulates eNOS activity through coordinated phosphorylation on Ser1177 and Thr495.
Collapse
|
132
|
Sansbury BE, Hill BG. Regulation of obesity and insulin resistance by nitric oxide. Free Radic Biol Med 2014; 73:383-99. [PMID: 24878261 PMCID: PMC4112002 DOI: 10.1016/j.freeradbiomed.2014.05.016] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/16/2014] [Accepted: 05/17/2014] [Indexed: 02/07/2023]
Abstract
Obesity is a risk factor for developing type 2 diabetes and cardiovascular disease and has quickly become a worldwide pandemic with few tangible and safe treatment options. Although it is generally accepted that the primary cause of obesity is energy imbalance, i.e., the calories consumed are greater than are utilized, understanding how caloric balance is regulated has proven a challenge. Many "distal" causes of obesity, such as the structural environment, occupation, and social influences, are exceedingly difficult to change or manipulate. Hence, molecular processes and pathways more proximal to the origins of obesity-those that directly regulate energy metabolism or caloric intake-seem to be more feasible targets for therapy. In particular, nitric oxide (NO) is emerging as a central regulator of energy metabolism and body composition. NO bioavailability is decreased in animal models of diet-induced obesity and in obese and insulin-resistant patients, and increasing NO output has remarkable effects on obesity and insulin resistance. This review discusses the role of NO in regulating adiposity and insulin sensitivity and places its modes of action into context with the known causes and consequences of metabolic disease.
Collapse
Affiliation(s)
- Brian E Sansbury
- Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Bradford G Hill
- Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
133
|
Hyndman KA, Ho DH, Sega MF, Pollock JS. Histone deacetylase 1 reduces NO production in endothelial cells via lysine deacetylation of NO synthase 3. Am J Physiol Heart Circ Physiol 2014; 307:H803-9. [PMID: 25015965 DOI: 10.1152/ajpheart.00243.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The lysine acetylation state of nonhistone proteins may be regulated through histone deacetylases (HDACs). Evidence suggests that nitric oxide (NO) synthase 3 (NOS3; endothelial NOS) is posttranslationally lysine acetylated, leading to increased NO production in the endothelium. We tested the hypothesis that NOS3 is lysine acetylated and that upregulated HDAC1-mediated deacetylation leads to reduced NO production in endothelial cells. We determined that NOS3 is basally lysine acetylated in cultured bovine aortic endothelial cells (BAECs). In BAECs, HDAC1 is expressed in the nucleus and cytosol and forms a novel protein-protein interaction with NOS3. Overexpression of HDAC1 in BAECs resulted in a significant reduction in NOS3 lysine acetylation (control = 1.0 ± 0.1 and HDAC1 = 0.59 ± 0.08 arbitrary units, P < 0.01) and significantly blunted basal nitrite production (control 287.7 ± 29.1 and HDAC1 172.4 ± 31.7 pmol·mg(-1)·h(-1), P < 0.05) as well as attenuating endothelin-1-stimulated nitrite production (control = 481.8 ± 50.3 and HDAC1 243.1 ± 48.2 pmol·mg(-1)·h(-1), P < 0.05). While HDAC1 knockdown with small-interfering RNA resulted in no change in NOS3 acetylation level, yet increased basal nitrite production (730.6 ± 99.1 pmol·mg(-1)·h(-1)) and further exaggerated increases in endothelin-1 stimulated nitrite production (1276.9 ± 288.2 pmol·mg(-1)·h(-1)) was observed. Moreover, overexpression or knockdown of HDAC1 resulted in no significant effect on NOS3 protein expression or NOS3 phosphorylation sites T497, S635, or S1179. Thus these data indicate that upregulated HDAC1 decreases NOS3 activity, most likely through direct lysine deacetylation of NOS3. We propose that HDAC1-mediated deacetylation of NOS3 may represent a novel target for endothelial dysfunction.
Collapse
Affiliation(s)
- Kelly A Hyndman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Dao H Ho
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Martiana F Sega
- Department of Medicine, Georgia Regents University, Augusta, Georgia
| | - Jennifer S Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Medicine, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
134
|
Chang ML, Chang JS, Yu WY, Cheah KP, Li JS, Cheng HW, Hu CM. Polygonum viviparum L. induces vasorelaxation in the rat thoracic aorta via activation of nitric oxide synthase in endothelial cells. Altern Ther Health Med 2014; 14:150. [PMID: 24885446 PMCID: PMC4030279 DOI: 10.1186/1472-6882-14-150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 04/17/2014] [Indexed: 11/10/2022]
Abstract
Background In the past several decades, Polygonum viviparum L. (PV) was reported to have antibacterial, antiulcer, antioxidant, antitumor, anti-inflammatory, and antiarthritic properties. The anti-inflammatory pathway was recently elucidated through cytosolic nuclear factor E2-related factor 2 (Nrf2) activation and heme oxygenase (HO)-1 protein expression. PV is a perennial herb and widely distributed in high-elevation mountain regions, such as the Tibetan Plateau. In Tibetan traditional medicine, PV is usually used to boost the blood circulation to dissipate blood stasis. Therefore, this study focused on how PV improves the vascular circulation and acts on vascular tissues. Methods In this study, we isolated aortas from Sprague-Dawley rats (male, weight about 250 ~ 350 g), and detected the effects of PV on phenylephrine (PE)-induced contraction and cyclic guanosine 3′,5′-monophosphate (cGMP) formation using aortic rings. In addition, human umbilical vein endothelial cells (HUVECs) were used to exam nitric oxygen (NO) synthase (NOS) activity by directly measuring NO production in the culture medium. Endothelial (e) NOS phosphorylation, and cytosolic Nrf2 and HO-1 expressions were measured using a Western blot analysis. Results PV dose-dependently relaxed PE-induced contractions in endothelial-intact but not -denuded aorta. The concentration to produce 50% relaxation was 22.04 ± 1.77 μg/ml. PV-induced vasorelaxation was markedly blocked by pretreatment with NG-nitro-L-arginine methyl ester (L-NAME), an NOS inhibitor, methylene blue (MB), a guanylyl cyclase inhibitor, and hemoglobin, an NO scavenger. PV increased cGMP formation; however, this effect was also suppressed by co-pretreatment with l-NAME, MB, hemoglobin, and Ca2+-free medium. In HUVECs, PV increased NO formation, which was greatly attenuated by NOS inhibitors (L-NAME and L-NMMA) and by removing extracellular Ca2+ and chelating intracellular Ca2+ with BAPTA-AM. In addition, PV promoted eNOS phosphorylation, Nrf2 degradation, and HO-1 protein expression according to a Western blot analysis. Conclusions The results suggest that PV possesses vasorelaxing action in an endothelium-dependent manner and works through activating Ca2+/calmodulin- dependent NO synthesis; when NO is released and then transferred to smooth muscle cells, NO activates guanylyl cyclase and increases cGMP formation, ultimately resulting in vasorelaxation. Thus, PV can be considered for application as a potential therapeutic approach for vascular-associated disorders.
Collapse
|
135
|
Liu S, Premont RT, Rockey DC. Endothelial nitric-oxide synthase (eNOS) is activated through G-protein-coupled receptor kinase-interacting protein 1 (GIT1) tyrosine phosphorylation and Src protein. J Biol Chem 2014; 289:18163-74. [PMID: 24764294 DOI: 10.1074/jbc.m113.521203] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) is a critical regulator of vascular tone and plays an especially prominent role in liver by controlling portal blood flow and pressure within liver sinusoids. Synthesis of NO in sinusoidal endothelial cells by endothelial nitric-oxide synthase (eNOS) is regulated in response to activation of endothelial cells by vasoactive signals such as endothelins. The endothelin B (ETB) receptor is a G-protein-coupled receptor, but the mechanisms by which it regulates eNOS activity in sinusoidal endothelial cells are not well understood. In this study, we built on two previous strands of work, the first showing that G-protein βγ subunits mediated activation of phosphatidylinositol 3-kinase and Akt to regulate eNOS and the second showing that eNOS directly bound to the G-protein-coupled receptor kinase-interacting protein 1 (GIT1) scaffold protein, and this association stimulated NO production. Here we investigated the mechanisms by which the GIT1-eNOS complex is formed and regulated. GIT1 was phosphorylated on tyrosine by Src, and Y293F and Y554F mutations reduced GIT1 phosphorylation as well as the ability of GIT1 to bind to and activate eNOS. Akt phosphorylation activated eNOS (at Ser(1177)), and Akt also regulated the ability of Src to phosphorylate GIT1 as well as GIT1-eNOS association. These pathways were activated by endothelin-1 through the ETB receptor; inhibiting receptor-activated G-protein βγ subunits blocked activation of Akt, GIT1 tyrosine phosphorylation, and ET-1-stimulated GIT1-eNOS association but did not affect Src activation. These data suggest a model in which Src and Akt cooperate to regulate association of eNOS with the GIT1 scaffold to facilitate NO production.
Collapse
Affiliation(s)
- Songling Liu
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and
| | - Richard T Premont
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710
| | - Don C Rockey
- From the Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and
| |
Collapse
|
136
|
Honavar J, Bradley E, Bradley K, Oh JY, Vallejo MO, Kelley EE, Cantu-Medellin N, Doran S, Dell'italia LJ, Matalon S, Patel RP. Chlorine gas exposure disrupts nitric oxide homeostasis in the pulmonary vasculature. Toxicology 2014; 321:96-102. [PMID: 24769334 DOI: 10.1016/j.tox.2014.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/04/2014] [Accepted: 04/15/2014] [Indexed: 10/25/2022]
Abstract
Exposure to chlorine (Cl2) gas during industrial accidents or chemical warfare leads to significant airway and distal lung epithelial injury that continues post exposure. While lung epithelial injury is prevalent, relatively little is known about whether Cl2 gas also promotes injury to the pulmonary vasculature. To determine this, rats were subjected to a sub-lethal Cl2 gas exposure (400 ppm, 30 min) and then brought back to room air. Pulmonary arteries (PA) were isolated from rats at various times post-exposure and contractile (phenylephrine) and nitric oxide (NO)-dependent vasodilation (acetylcholine and mahmanonoate) responses measured ex vivo. PA contractility did not change, however significant inhibition of NO-dependent vasodilation was observed that was maximal at 24-48 h post exposure. Superoxide dismutase restored NO-dependent vasodilation suggesting a role for increased superoxide formation. This was supported by ∼2-fold increase in superoxide formation (measured using 2-hydroethidine oxidation to 2-OH-E+) from PA isolated from Cl2 exposed rats. We next measured PA pressures in anesthetized rats. Surprisingly, PA pressures were significantly (∼4 mmHg) lower in rats that had been exposed to Cl2 gas 24 h earlier suggesting that deficit in NO-signaling observed in isolated PA experiments did not manifest as increased PA pressures in vivo. Administration of the iNOS selective inhibitor 1400W, restored PA pressures to normal in Cl2 exposed, but not control rats suggesting that any deficit in NO-signaling due to increased superoxide formation in the PA, is offset by increased NO-formation from iNOS. These data indicate that disruption of endogenous NO-signaling mechanisms that maintain PA tone is an important aspect of post-Cl2 gas exposure toxicity.
Collapse
Affiliation(s)
- Jaideep Honavar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Eddie Bradley
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Kelley Bradley
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Joo Yeun Oh
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Matthew O Vallejo
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Eric E Kelley
- Department of Anesthesiology, University of Pittsburgh, PA, United States
| | | | - Stephen Doran
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Louis J Dell'italia
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Birmingham VA Medical Center, United States
| | - Sadis Matalon
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Centers for Free Radical Biology and Lung Injury and Repair, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Rakesh P Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Centers for Free Radical Biology and Lung Injury and Repair, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
137
|
Endothelial dysfunction in experimental models of arterial hypertension: cause or consequence? BIOMED RESEARCH INTERNATIONAL 2014. [PMID: 24738065 DOI: 10.1155/2014/598271.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hypertension is a risk factor for other cardiovascular diseases and endothelial dysfunction was found in humans as well as in various commonly employed animal experimental models of arterial hypertension. Data from the literature indicate that, in general, endothelial dysfunction would not be the cause of experimental hypertension and may rather be secondary, that is, resulting from high blood pressure (BP). The initial mechanism of endothelial dysfunction itself may be associated with a lack of endothelium-derived relaxing factors (mainly nitric oxide) and/or accentuation of various endothelium-derived constricting factors. The involvement and role of endothelium-derived factors in the development of endothelial dysfunction in individual experimental models of hypertension may vary, depending on the triggering stimulus, strain, age, and vascular bed investigated. This brief review was focused on the participation of endothelial dysfunction, individual endothelium-derived factors, and their mechanisms of action in the development of high BP in the most frequently used rodent experimental models of arterial hypertension, including nitric oxide deficient models, spontaneous (pre)hypertension, stress-induced hypertension, and selected pharmacological and diet-induced models.
Collapse
|
138
|
Chen X, Mak IT. Mg supplementation protects against ritonavir-mediated endothelial oxidative stress and hepatic eNOS downregulation. Free Radic Biol Med 2014; 69:77-85. [PMID: 24434120 PMCID: PMC3960338 DOI: 10.1016/j.freeradbiomed.2014.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 02/07/2023]
Abstract
Ritonavir (RTV), a prototypical protease inhibitor currently used as a key component of anti-HIV therapy, is known for its endothelial and hepatic toxicity. The effects of RTV and magnesium supplementation on cultured bovine endothelial cell (EC) and rat hepatic endothelial nitric oxide synthase (eNOS) status were investigated. RTV dose-dependently (5-30 µM) decreased EC viability after 48 h; high Mg (2mM) significantly attenuated the lost viability. ECs incubated with 15 µM RTV for 6 to 24h resulted in two- to fourfold elevation of oxidized glutathione and a 25% loss of total glutathione. At 24h, EC superoxide production due to RTV was detected by dihydroethidium staining and increased 41% when quantified by flow cytometry; altered glutathione status and superoxide levels were both substantially reversed by 2mM Mg. RTV reduced eNOS mRNA (-25% at 24 h) and led to decreased eNOS dimer/monomer ratios; nitric oxide-derived products decreased 40%; both changes were attenuated by Mg supplementation. In male Lewis-Brown Norway rats, RTV administration (75 mg/kg/day, 5 weeks) resulted in an 85% increase in plasma 8-isoprostane and a 23% decrease in hepatic eNOS mRNA; concomitantly, eNOS protein decreased 75%, whereas plasma nitrite level was reduced 48%. Dietary Mg supplementation (sixfold higher than control) prevented the eNOS mRNA decrease along with lowering 8-isoprostane and restored the eNOS protein and plasma nitrite levels comparable to controls. In conclusion, Mg attenuates RTV-mediated EC oxidative eNOS dysfunction and downregulation of hepatic eNOS expression; we suggest that Mg can serve as a beneficial adjunct therapeutic against RTV-mediated eNOS toxicity.
Collapse
Affiliation(s)
- Xi Chen
- Department of Biochemistry & Molecular Medicine, The George Washington University Medical Center, Washington, DC 20037, USA
| | - I Tong Mak
- Department of Biochemistry & Molecular Medicine, The George Washington University Medical Center, Washington, DC 20037, USA.
| |
Collapse
|
139
|
Endothelial dysfunction in experimental models of arterial hypertension: cause or consequence? BIOMED RESEARCH INTERNATIONAL 2014; 2014:598271. [PMID: 24738065 PMCID: PMC3971506 DOI: 10.1155/2014/598271] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 01/31/2014] [Indexed: 02/07/2023]
Abstract
Hypertension is a risk factor for other cardiovascular diseases and endothelial dysfunction was found in humans as well as in various commonly employed animal experimental models of arterial hypertension. Data from the literature indicate that, in general, endothelial dysfunction would not be the cause of experimental hypertension and may rather be secondary, that is, resulting from high blood pressure (BP). The initial mechanism of endothelial dysfunction itself may be associated with a lack of endothelium-derived relaxing factors (mainly nitric oxide) and/or accentuation of various endothelium-derived constricting factors. The involvement and role of endothelium-derived factors in the development of endothelial dysfunction in individual experimental models of hypertension may vary, depending on the triggering stimulus, strain, age, and vascular bed investigated. This brief review was focused on the participation of endothelial dysfunction, individual endothelium-derived factors, and their mechanisms of action in the development of high BP in the most frequently used rodent experimental models of arterial hypertension, including nitric oxide deficient models, spontaneous (pre)hypertension, stress-induced hypertension, and selected pharmacological and diet-induced models.
Collapse
|
140
|
García C, Nuñez-Anita RE, Thebault S, Arredondo Zamarripa D, Jeziorsky MC, Martínez de la Escalera G, Clapp C. Requirement of phosphorylatable endothelial nitric oxide synthase at Ser-1177 for vasoinhibin-mediated inhibition of endothelial cell migration and proliferation in vitro. Endocrine 2014; 45:263-70. [PMID: 23640371 DOI: 10.1007/s12020-013-9964-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/16/2013] [Indexed: 12/23/2022]
Abstract
Endothelial nitric oxide synthase (eNOS)-derived nitric oxide is a major vasorelaxing factor and a mediator of vasopermeability and angiogenesis. Vasoinhibins, a family of antiangiogenic prolactin fragments that include 16 K prolactin, block most eNOS-mediated vascular effects. Vasoinhibins activate protein phosphatase 2A, causing eNOS inactivation through dephosphorylation of eNOS at serine residue 1179 in bovine endothelial cells and thereby blocking vascular permeability. In this study, we examined whether human eNOS phosphorylation at S1177 (analogous to bovine S1179) influences other actions of vasoinhibins. Bovine umbilical vein endothelial cells were stably transfected with human wild-type eNOS (WT) or with phospho-mimetic (S1177D) or non-phosphorylatable (S1177A) eNOS mutants. Vasoinhibins inhibited the increases in eNOS activity, migration, and proliferation following the overexpression of WT eNOS but did not affect these responses in cells expressing S1177D and S1177A eNOS mutants. We conclude that eNOS inhibition by dephosphorylation of S1177 is fundamental for the inhibition of endothelial cell migration and proliferation by vasoinhibins.
Collapse
Affiliation(s)
- Celina García
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM-Juriquilla, 76230, Querétaro, QRO, Mexico
| | | | | | | | | | | | | |
Collapse
|
141
|
Kang JJ, Shu L, Park JL, Shayman JA, Bodary PF. Endothelial nitric oxide synthase uncoupling and microvascular dysfunction in the mesentery of mice deficient in α-galactosidase A. Am J Physiol Gastrointest Liver Physiol 2014; 306:G140-6. [PMID: 24232002 PMCID: PMC3920075 DOI: 10.1152/ajpgi.00185.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A defect in the gene for the lysosomal enzyme α-galactosidase A (Gla) results in globotriaosylceramide (Gb3) accumulation in Fabry disease and leads to premature death from cardiac and cerebrovascular events. However, gastrointestinal symptoms are often first observed during childhood in these patients and are not well understood. In this study, we demonstrate an age-dependent microvasculopathy of the mesenteric artery (MA) in a murine model of Fabry disease (Gla-knockout mice) resulting from dysregulation of the vascular homeostatic enzyme endothelial nitric oxide synthase (eNOS). The progressive accumulation of Gb3 in the MA was confirmed by thin-layer chromatographic analysis. A total absence of endothelium-dependent dilation was observed in MAs from mice at 8 mo of age, while suppression of ACh-mediated vasodilation was evident from 2 mo of age. Endothelium-independent dilation with sodium nitroprusside was normal compared with age-matched wild-type mice. The microvascular defect in MAs from Fabry mice was endothelium-dependent and associated with suppression of the active homodimer of eNOS. Phosphorylation of eNOS at the major activation site (Ser(1179)) was significantly downregulated, while phosphorylation at the major inhibitory site (Thr(495)) was remarkably enhanced in MAs from aged Fabry mice. These profound alterations in eNOS bioavailability at 8 mo of age were observed in parallel with high levels of 3-nitrotyrosine, suggesting increased reactive oxygen species along with eNOS uncoupling in this vascular bed. Overall, the mesenteric microvessels in the setting of Fabry disease were observed to have an early and profound endothelial dysfunction associated with elevated reactive nitrogen species and decreased nitric oxide bioavailability.
Collapse
Affiliation(s)
- Justin J. Kang
- 1School of Kinesiology, University of Michigan, Ann Arbor, Michigan;
| | - Liming Shu
- 2Division of Nephrology, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan; and
| | - James L. Park
- 2Division of Nephrology, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan; and ,3DMT-USA, Inc., Ann Arbor, Michigan
| | - James A. Shayman
- 2Division of Nephrology, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan; and
| | - Peter F. Bodary
- 1School of Kinesiology, University of Michigan, Ann Arbor, Michigan;
| |
Collapse
|
142
|
Rafikov R, Kumar S, Aggarwal S, Pardo D, Fonseca FV, Ransom J, Rafikova O, Chen Q, Springer ML, Black SM. Protein engineering to develop a redox insensitive endothelial nitric oxide synthase. Redox Biol 2014; 2:156-64. [PMID: 25460726 PMCID: PMC4297941 DOI: 10.1016/j.redox.2013.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 11/25/2022] Open
Abstract
The zinc tetrathiolate (ZnS4) cluster is an important structural feature of endothelial nitric oxide synthase (eNOS). The cluster is located on the dimeric interface and four cysteine residues (C94 and C99 from two adjacent subunits) form a cluster with a Zn ion in the center of a tetrahedral configuration. Due to its high sensitivity to oxidants this cluster is responsible for eNOS dimer destabilization during periods of redox stress. In this work we utilized site directed mutagenesis to replace the redox sensitive cysteine residues in the ZnS4 cluster with redox stable tetra-arginines. Our data indicate that this C94R/C99R eNOS mutant is active. In addition, this mutant protein is insensitive to dimer disruption and inhibition when challenged with hydrogen peroxide (H2O2). Further, the overexpression of the C94R/C99R mutant preserved the angiogenic response in endothelial cells challenged with H2O2. The over-expression of the C94R/C99R mutant preserved the ability of endothelial cells to migrate towards vascular endothelial growth factor (VEGF) and preserved the endothelial monolayer in a scratch wound assay. We propose that this dimer stable eNOS mutant could be utilized in the treatment of diseases in which there is eNOS dysfunction due to high levels of oxidative stress. The ZnS4 cluster is an important structural feature of eNOS. This cluster is responsible for eNOS dimer destabilization during redox stress. Site directed mutagenesis replaced ZnS4 clusters with redox stable tetra-arginines. This eNOS mutant is insensitive to dimer disruption during redox stress. This eNOS mutant continues to produce NO during redox stress.
Collapse
Affiliation(s)
- Ruslan Rafikov
- Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - Sanjiv Kumar
- Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - Saurabh Aggarwal
- Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - Daniel Pardo
- Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - Fabio V Fonseca
- Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - Jessica Ransom
- Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - Olga Rafikova
- Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - Qiumei Chen
- The Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew L Springer
- The Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephen M Black
- Pulmonary Vascular Disease, Vascular Biology Center, Georgia Regents University, Augusta, GA, USA.
| |
Collapse
|
143
|
Nemoto S, Matsumoto T, Taguchi K, Kobayashi T. Relationships among protein tyrosine phosphatase 1B, angiotensin II, and insulin-mediated aortic responses in type 2 diabetic Goto-Kakizaki rats. Atherosclerosis 2014; 233:64-71. [PMID: 24529124 DOI: 10.1016/j.atherosclerosis.2013.12.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 11/26/2013] [Accepted: 12/09/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVE We investigated the relationships among protein tyrosine phosphatase 1B (PTP1B), angiotensin II (Ang II), and insulin signaling in the presence of endothelial dysfunction in type 2 diabetic Goto-Kakizaki (GK) rat aortas. METHODS AND RESULTS Aortas isolated from GK or control Wistar rats were examined in the presence or absence of Ang II with or without a selective antagonist of the Ang II type 1 (AT1) receptor or a PTP1B inhibitor to evaluate vascular functional and molecular mechanisms, such as insulin-induced relaxation, nitric oxide (NO) production, phosphorylation of insulin receptor substrate (IRS)-1, endothelial NO synthase (eNOS), and phosphorylation, and the subcellular localization of PTP1B. GK aortas exhibited reductions of: 1) insulin-induced relaxation, 2) NO production, 3) Ser(1177)-p-eNOS, and 4) Tyr(612)-p-IRS-1. Pre-incubation with a PTP1B inhibitor normalized these reductions. In Wistar aortas, the four above-mentioned parameters were reduced by Ang II, but were completely inhibited by co-treatment with the PTP1B inhibitor. The membrane expression of PTP1B was greater in GK than in Wistar aortas, and it was increased by Ang II in Wistar rats. The membrane PTP1B expression in the presence of insulin + Ang II was reduced by the PTP1B inhibitor or AT1-receptor antagonist. CONCLUSIONS These results suggest that the membrane PTP1B suppressed insulin-mediated aortic relaxation, and this was due to the Ang II-AT1-receptor signaling pathway. The inhibition of PTP1B warrants further investigation as a potential therapeutic target for endothelial dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- Shingo Nemoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| |
Collapse
|
144
|
Abstract
The prevalence of obesity has increased remarkably in the past four decades. Because obesity can promote the development of type 2 diabetes and cardiovascular disease, understanding the mechanisms that engender weight gain and discovering safe antiobesity therapies are of critical importance. In particular, the gaseous signaling molecule, nitric oxide (NO), appears to be a central factor regulating adiposity and systemic metabolism. Obese and diabetic states are characterized by a deficit in bioavailable NO, with such decreases commonly attributed to downregulation of endothelial NO synthase (eNOS), loss of eNOS activity, or quenching of NO by its reaction with oxygen radicals. Gain-of-function studies, in which vascular-derived NO has been increased pharmacologically or genetically, reveal remarkable actions of NO on body composition and systemic metabolism. This review addresses the metabolic actions of eNOS and the potential therapeutic utility of harnessing its antiobesogenic effects.
Collapse
Affiliation(s)
- Brian E Sansbury
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Louisville, Kentucky, USA; Department of Physiology and Biophysics, Louisville, Kentucky, USA
| | - Bradford G Hill
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Louisville, Kentucky, USA; Department of Physiology and Biophysics, Louisville, Kentucky, USA; Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky, USA.
| |
Collapse
|
145
|
Eghbalzadeh K, Brixius K, Bloch W, Brinkmann C. Skeletal muscle nitric oxide (NO) synthases and NO-signaling in "diabesity"--what about the relevance of exercise training interventions? Nitric Oxide 2013; 37:28-40. [PMID: 24368322 DOI: 10.1016/j.niox.2013.12.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/30/2013] [Accepted: 12/17/2013] [Indexed: 12/30/2022]
Abstract
Type 2 diabetes mellitus associated with obesity, or "diabesity", coincides with an altered nitric oxide (NO) metabolism in skeletal muscle. Three isoforms of nitric oxide synthase (NOS) exist in human skeletal muscle tissue. Both neuronal nitric oxide synthase (nNOS) and endothelial nitric oxide synthase (eNOS) are constitutively expressed under physiological conditions, producing low levels of NO, while the inducible nitric oxide synthase (iNOS) is strongly up-regulated only under pathophysiological conditions, excessively increasing NO concentrations. Due to chronic inflammation, overweight/obese type 2 diabetic patients exhibit up-regulated protein contents of iNOS and concomitant elevated amounts of NO in skeletal muscle. Low muscular NO levels are important for attaining an adequate cellular redox state--thereby maintaining metabolic integrity--while high NO levels are believed to destroy cellular components and to disturb metabolic processes, e.g., through strongly augmented posttranslational protein S-nitrosylation. Physical training with submaximal intensity has been shown to attenuate inflammatory profiles and iNOS protein contents in the long term. The present review summarizes signaling pathways which induce iNOS up-regulation under pathophysiological conditions and describes molecular mechanisms by which high NO concentrations are likely to contribute to triggering skeletal muscle insulin resistance and to reducing mitochondrial capacity during the development and progression of type 2 diabetes. Based on this information, it discusses the beneficial effects of regular physical exercise on the altered NO metabolism in the skeletal muscle of overweight/obese type 2 diabetic subjects, thus unearthing new perspectives on training strategies for this particular patient group.
Collapse
Affiliation(s)
- Kaveh Eghbalzadeh
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Germany
| | - Klara Brixius
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Germany
| | - Christian Brinkmann
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Germany.
| |
Collapse
|
146
|
Than A, Zhang X, Leow MKS, Poh CL, Chong SK, Chen P. Apelin attenuates oxidative stress in human adipocytes. J Biol Chem 2013; 289:3763-74. [PMID: 24362107 DOI: 10.1074/jbc.m113.526210] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
It has been recently recognized that the increased oxidative stress (ROS overproduction) in obese condition is a key contributor to the pathogenesis of obesity-associated metabolic diseases. Apelin is an adipocytokine secreted by adipocytes, and known for its anti-obesity and anti-diabetic properties. In obesity, both oxidative stress and plasma level of apelin are increased. However, the regulatory roles of apelin on oxidative stress in adipocytes remain unknown. In the present study, we provide evidence that apelin, through its interaction with apelin receptor (APJ), suppresses production and release of reactive oxygen species (ROS) in adipocytes. This is further supported by the observations that apelin promotes the expression of anti-oxidant enzymes via MAPK kinase/ERK and AMPK pathways, and suppresses the expression of pro-oxidant enzyme via AMPK pathway. We further demonstrate that apelin is able to relieve oxidative stress-induced dysregulations of the expression of anti- and pro-oxidant enzymes, mitochondrial biogenesis and function, as well as release of pro- and anti-inflammatory adipocytokines. This study, for the first time, reveals the antioxidant properties of apelin in adipocytes, and suggests its potential as a novel therapeutic target for metabolic diseases.
Collapse
Affiliation(s)
- Aung Than
- From the Division of Bioengineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | | | | | | | | | | |
Collapse
|
147
|
Raber PL, Thevenot P, Sierra R, Wyczechowska D, Halle D, Ramirez ME, Ochoa AC, Fletcher M, Velasco C, Wilk A, Reiss K, Rodriguez PC. Subpopulations of myeloid-derived suppressor cells impair T cell responses through independent nitric oxide-related pathways. Int J Cancer 2013; 134:2853-64. [PMID: 24259296 DOI: 10.1002/ijc.28622] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/17/2013] [Accepted: 11/05/2013] [Indexed: 12/13/2022]
Abstract
The accumulation of myeloid-derived suppressor cells (MDSC) in tumor-bearing hosts is a hallmark of malignancy-associated inflammation and a major mediator for the induction of T cell suppression in cancer. MDSC can be divided phenotypically into granulocytic (G-MDSC) and monocytic (Mo-MDSC) subgroups. Several mechanisms mediate the induction of T cell anergy by MDSC; however, the specific role of these pathways in the inhibitory activity of MDSC subpopulations remains unclear. Therefore, we aimed to determine the effector mechanisms by which subsets of tumor-infiltrating MDSC block T cell function. We found that G-MDSC had a higher ability to impair proliferation and expression of effector molecules in activated T cells, as compared to Mo-MDSC. Interestingly, both MDSC subgroups inhibited T cells through nitric oxide (NO)-related pathways, but expressed different effector inhibitory mechanisms. Specifically, G-MDSC impaired T cells through the production of peroxynitrites (PNT), while Mo-MDSC suppressed by the release of NO. The production of PNT in G-MDSC depended on the expression of gp91(phox) and endothelial NO synthase (eNOS), while inducible NO synthase (iNOS) mediated the generation of NO in Mo-MDSC. Deletion of eNOS and gp91(phox) or scavenging of PNT blocked the suppressive function of G-MDSC and induced anti-tumoral effects, without altering Mo-MDSC inhibitory activity. Furthermore, NO-scavenging or iNOS knockdown prevented Mo-MDSC function, but did not affect PNT production or suppression by G-MDSC. These results suggest that MDSC subpopulations utilize independent effector mechanisms to regulate T cell function. Inhibition of these pathways is expected to specifically block MDSC subsets and overcome immune suppression in cancer.
Collapse
Affiliation(s)
- Patrick L Raber
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA; Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Aggarwal S, Gross CM, Sharma S, Fineman JR, Black SM. Reactive oxygen species in pulmonary vascular remodeling. Compr Physiol 2013; 3:1011-34. [PMID: 23897679 DOI: 10.1002/cphy.c120024] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The pathogenesis of pulmonary hypertension is a complex multifactorial process that involves the remodeling of pulmonary arteries. This remodeling process encompasses concentric medial thickening of small arterioles, neomuscularization of previously nonmuscular capillary-like vessels, and structural wall changes in larger pulmonary arteries. The pulmonary arterial muscularization is characterized by vascular smooth muscle cell hyperplasia and hypertrophy. In addition, in uncontrolled pulmonary hypertension, the clonal expansion of apoptosis-resistant endothelial cells leads to the formation of plexiform lesions. Based upon a large number of studies in animal models, the three major stimuli that drive the vascular remodeling process are inflammation, shear stress, and hypoxia. Although, the precise mechanisms by which these stimuli impair pulmonary vascular function and structure are unknown, reactive oxygen species (ROS)-mediated oxidative damage appears to play an important role. ROS are highly reactive due to their unpaired valence shell electron. Oxidative damage occurs when the production of ROS exceeds the quenching capacity of the antioxidant mechanisms of the cell. ROS can be produced from complexes in the cell membrane (nicotinamide adenine dinucleotide phosphate-oxidase), cellular organelles (peroxisomes and mitochondria), and in the cytoplasm (xanthine oxidase). Furthermore, low levels of tetrahydrobiopterin (BH4) and L-arginine the rate limiting cofactor and substrate for endothelial nitric oxide synthase (eNOS), can cause the uncoupling of eNOS, resulting in decreased NO production and increased ROS production. This review will focus on the ROS generation systems, scavenger antioxidants, and oxidative stress associated alterations in vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | |
Collapse
|
149
|
Yusuf K, Wilson RD, Kamaluddeen M, Franta J, Hasan SU, Akierman A. Methemoglobin levels in umbilical cord blood of women with intrauterine growth restriction and preeclampsia. J Matern Fetal Neonatal Med 2013; 27:789-94. [DOI: 10.3109/14767058.2013.838949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
150
|
Taylor SY, Dixon HM, Yoganayagam S, Price N, Lang D. Folic acid modulates eNOS activity via effects on posttranslational modifications and protein-protein interactions. Eur J Pharmacol 2013; 714:193-201. [PMID: 23796957 PMCID: PMC3769861 DOI: 10.1016/j.ejphar.2013.05.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 05/10/2013] [Accepted: 05/24/2013] [Indexed: 02/07/2023]
Abstract
Folic acid enhances endothelial function and improves outcome in primary prevention of cardiovascular disease. The exact intracellular signalling mechanisms involved remain elusive and were therefore the subject of this study. Particular focus was placed on folic acid-induced changes in posttranslational modifications of endothelial nitric oxide synthase (eNOS). Cultured endothelial cells were exposed to folic acid in the absence or presence of phosphatidylinositol-3' kinase/Akt (PI3K/Akt) inhibitors. The phosphorylation status of eNOS was determined via western blotting. The activities of eNOS and PI3K/Akt were evaluated. The interaction of eNOS with caveolin-1, Heat-Shock Protein 90 and calmodulin was studied using co-immunoprecipitation. Intracellular localisation of eNOS was investigated using sucrose gradient centrifugation and confocal microscopy. Folic acid promoted eNOS dephosphorylation at negative regulatory sites, and increased phosphorylation at positive regulatory sites. Modulation of phosphorylation status was concomitant with increased cGMP concentrations, and PI3K/Akt activity. Inhibition of PI3K/Akt revealed specific roles for this kinase pathway in folic acid-mediated eNOS phosphorylation. Regulatory protein and eNOS protein associations were altered in favour of a positive regulatory effect in the absence of bulk changes in intracellular eNOS localisation. Folic acid-mediated eNOS activation involves the modulation of eNOS phosphorylation status at multiple residues and positive changes in important protein-protein interactions. Such intracellular mechanisms may in part explain improvements in clinical vascular outcome following folic acid treatment.
Collapse
Affiliation(s)
| | | | | | | | - Derek Lang
- Department of Pharmacology, Therapeutics & Toxicology, Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park Campus, Cardiff CF14 4XN, UK
| |
Collapse
|