Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Rev. bras. cir. cardiovasc ; 37(4): 439-446, Jul.-Aug. 2022. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1394715

ABSTRACT

ABSTRACT Introduction: A weak venous wall is one of the major reasons contributing to vein graft failure after coronary artery bypass grafting (CABG). We investigated whether adventitial collagen cross-linking by glutaraldehyde reinforces venous wall, preserving the endothelium of veins during high-pressure distention. Methods: Human saphenous veins (SVs) were collected from 40 patients undergoing CABG, and adventitia cross-linking was performed with 0.3% glutaraldehyde for five minutes. The cross-linked SVs were accessed by biodegradation assay, immunofluorescent staining, and tensile test. Native SVs and cross-linked SVs from another 20 patients received the 200 mmHg pressure distention for two minutes. Pressure-induced injury of SVs were accessed by immunohistochemistry and electron microscopy. Results: Time to digestion was 97±13 minutes for native SVs and 720±0 minutes for cross-linked SVs (P<0.05). After adventitial cross-linking, the collagen I fibres of the vein remarkably presented with compact and nonporous arrangement. In the high-stretch region (stretch ratio 1.4-1.8), the Young's elastic modulus of stress-stretch ratio curve in cross-linked SVs was larger than that in native SVs (13.88 vs. 5.83, P<0.05). The cross-linked SVs had a lower extent of endothelial denudation without fibre fracture during high-pressure distension than native SVs. Comparing with the non-cross-linked SVs, the percentage of endothelial nitric oxide synthase staining length on the endothelium of cross-linked SVs was significantly preserved after high-pressure distension (85.2% vs. 64.7%, P<0.05). Conclusion: Adventitial collagen cross-linking by glutaraldehyde reinforced venous wall by increasing stiffness and decreasing extensibility of SVs and mitigated the endothelial damage under high-pressure distension.

2.
Rev. Assoc. Med. Bras. (1992) ; 66(8): 1128-1133, Aug. 2020. tab
Article in English | SES-SP, LILACS | ID: biblio-1136334

ABSTRACT

SUMMARY AIM The aim of this study was to examine the roles of nitric oxide (NOx), endothelial nitric oxide synthetase (eNOS), and asymmetric dimethylarginine (ADMA), which is the major endogenous inhibitor of nitric oxide synthases (NOS), in the pathophysiology of hemorrhoidal disease. METHODS This study included 54 patients with grades 3 and 4 internal hemorrhoidal disease and 54 patients without the disease who attended the General Surgery Clinic. NOx, eNOS, and ADMA levels were measured with the Enzyme-Linked ImmunoSorbent Assay (ELISA) method. RESULTS The patients had higher NO and eNOS levels and lower ADMA levels than the control subjects (p<0.001). A significant highly positive correlation was found between NO and eNOS (p<0.001). Nevertheless, there was a highly negative correlation between ADMA and NO-eNOS(p<0.001, p<0.001). CONCLUSION This preliminary study reveals that higher NOx and eNOS activities and lower ADMA levels in the rectal mucosa are observed in patients with hemorrhoidal disease than in those with normal rectal tissue. The imbalance between endothelium-derived relaxing factors, such as NO and endogenous competitive inhibitor of NOS, ADMA, may cause hemorrhoidal disease. Our study proposes that hemorrhoids display apparent vascular dilatation and present with bleeding or swelling. ADMA is an effective NOS inhibitor and may be a promising therapeutic option for hemorrhoidal disease.


RESUMO OBJETIVO O objetivo deste estudo foi examinar os papéis do óxido nítrico (NOx), do óxido nítrico sintetase endotelial (eNOS) e da dimetilarginina assimétrica (ADMA), que é o principal inibidor endógeno das óxido nítrico sintase (NOS) na fisiopatologia da doença hemorróida. MÉTODOS Este estudo incluiu 54 pacientes com doença hemorróida interna de grau 3 e 4 e 54 pacientes sem a doença que se inscreveram na Clínica Geral de Cirurgia. Os níveis de NOx, eNOS e ADMA foram medidos com o método de Ensaio Imuno absorvente ligado a enzima (ELISA). RESULTADOS Os pacientes têm níveis mais altos de NO e eNOS e níveis mais baixos de ADMA do que os indivíduos controle (p <0,001). Uma correlação altamente positiva significativa foi encontrada entre o NO-eNOS (p <0,001). No entanto, houve uma correlação negativa muito séria entre ADMA e NO-eNOS (p <0,001, p <0,001). CONCLUSÃO Este estudo preliminar revela que os pacientes com doença hemorróida têm atividades mais altas de NOx e eNOS e níveis mais baixos de ADMA na mucosa retal do que os pacientes com tecido retal normal. Desequilíbrio entre o fator relaxante derivado do endotélio, como; O NO e o inibidor competitivo endógeno da NOS, ADMA, podem causar doenças hemorróidas. Nosso estudo propõe que as hemorróidas exibam aparente dilatação vascular e apresentam sangramento ou inchaço, a ADMA é um inibidor eficaz da NOS e pode ser uma opção terapêutica promissora para a doença hemorróida.


Subject(s)
Humans , Hemorrhoids , Arginine/analogs & derivatives , Nitric Oxide Synthase Type III , Nitric Oxide
3.
Acta cir. bras ; 34(8): e201900802, 2019. tab, graf
Article in English | LILACS | ID: biblio-1038128

ABSTRACT

Abstract Purpose To reveal the function of miR-134 in myocardial ischemia. Methods Real-time PCR and western blotting were performed to measure the expression of miR-134, nitric oxide synthase 3 (NOS3) and apoptotic-associated proteins. Lactic dehydrogenase (LDH) assay, cell counting kit-8 (CCK-8), Hoechst 33342/PI double staining and flow cytometry assay were implemented in H9c2 cells, respectively. MiR-134 mimic/inhibitor was used to regulate miR-134 expression. Bioinformatic analysis and luciferase reporter assay were utilized to identify the interrelation between miR-134 and NOS3. Rescue experiments exhibited the role of NOS3. The involvement of PI3K/AKT was assessed by western blot analysis. Results MiR-134 was high regulated in the myocardial ischemia model, and miR-134 mimic/inhibitor transfection accelerated/impaired the speed of cell apoptosis and attenuated/exerted the cell proliferative prosperity induced by H/R regulating active status of PI3K/AKT signaling. LDH activity was also changed due to the different treatments. Moreover, miR-134 could target NOS3 directly and simultaneously attenuated the expression of NOS3. Co-transfection miR-134 inhibitor and pcDNA3.1-NOS3 highlighted the inhibitory effects of miR-134 on myocardial H/R injury. Conclusion This present work puts insights into the crucial effects of the miR-134/NOS3 axis in myocardial H/R injury, delivering a potential therapeutic technology in future.


Subject(s)
Animals , Rats , Myocardial Reperfusion Injury/metabolism , MicroRNAs/metabolism , Nitric Oxide Synthase Type III/metabolism , Hypoxia/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Apoptosis/drug effects , Apoptosis/physiology , Phosphatidylinositol 3-Kinases/metabolism , MicroRNAs/genetics , MicroRNAs/therapeutic use , Cell Proliferation/drug effects , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism
4.
Vascular Specialist International ; : 105-112, 2016.
Article in English | WPRIM | ID: wpr-79756

ABSTRACT

PURPOSE: The endothelial nitric oxide synthase (eNOS) G894T polymorphism has been reported to cause endothelial dysfunction and may have a role in the development of coronary artery disease (CAD). The aim of the present study was to investigate the association of eNOS G894T genetic polymorphism and plasma levels of nitric oxide (NO) with CAD risk in an Iranian population. MATERIALS AND METHODS: We studied 200 patients with angiographically documented CAD and 100 matched controls. Analysis of G894T genetic polymorphism of eNOS was performed by polymerase chain reaction-restriction fragment length polymorphism method. Plasma levels of NO were determined using Griess method. Biochemical analysis was conducted by routine colorimetric methods. RESULTS: Plasma levels of NO were significantly lower in CAD patients than control subjects (41.60±12.70 vs. 55.48±16.57, P=0.001). Also, the mean plasma levels of NO were significantly lower in T allele carriers of eNOS G894T polymorphism than G allele carriers (P0.05). CONCLUSION: Reduced plasma level of NO is associated with increased risk of CAD in our population. Moreover, eNOS G894T polymorphism is a significant risk factor for CAD development via reducing the plasma levels of NO. However, eNOS G894T polymorphism is not a contributing factor for the severity of CAD.


Subject(s)
Humans , Alleles , Coronary Artery Disease , Coronary Vessels , Gene Frequency , Genotype , Methods , Nitric Oxide Synthase Type III , Nitric Oxide , Plasma , Polymorphism, Genetic , Risk Factors
5.
Medical Journal of Chinese People's Liberation Army ; (12): 502-506, 2015.
Article in Chinese | WPRIM | ID: wpr-850231

ABSTRACT

Objective To explore the effects of unsafe decompression on the endothelial function of pulmonary artery in rat and its possible related mechanism. Methods Sixty male SD rats (260±35g) were randomly divided into two groups (30 each): control group and decompression (DCS) group. Decompression sickness (DCS) model was reproduced by placing the rats in a compression chamber with air pressure of 600kPa for 60min, followed by decompression at a rate of 100kPa/min to normal pressure. The surviving rats in both control and DCS groups were sacrificed and their pulmonary artery was harvested. The endothelium dependent vasodilatation capacity of isolated pulmonary artery was assessed. The expression and uncoupling of endothelial nitric oxide synthetase (eNOS), as well as the nitration level of each kind of protein in the pulmonary artery tissue, were analyzed by Western blotting. The concentration of reactive oxygen species (ROS) in the pulmonary artery was determined with superoxide anion probe dihematoporphyrin ether (DHE) staining. Results Ten of 30 rats in DC group died of unsafe decompression, and the endothelium dependent vasodilatation capacity of excised pulmonary artery in survived rats was found to decline obviously (P0.05), but the ratio of eNOS monomer/dimer increased significantly in DC group than in control group (P<0.05). The tyrosine nitration level of each kind of protein in the pulmonary artery tissues was higher significantly in DC group than that in control group (P<0.05). DHE showed that the generated amount of DCS in pulmonary artery tissues was obviously higher in DC group than in control group (P<0.05). Conclusions Unsafe decompression may lead to uncoupling of eNOS dimers in the endothelium of pulmonary artery. Uncoupled eNOS monomers may inhibit the synthesis of NO, thereby affect the endothelium dependent vasodilatation function. On the other hand, the eNOS monomers may facilitate the anabolism of ONOO-, leading to an increase in tyrosine nitration level of each kind of protein in the pulmonary artery tissues, thereby cause the regulation disorder of cell information system. The eNOS monomers may also increase the production of ROC, there by mediate the peroxide injuries.

6.
Tumor ; (12): 135-140, 2014.
Article in Chinese | WPRIM | ID: wpr-848805

ABSTRACT

Objective: To evaluate the association between the 894G>T polymorphism of nitric oxide synthase 3 (NOS3) gene and the susceptibility to prostate cancer. Methods: A computer-based online search was performed by using Cochrane Library, PubMed, EMBase, CNKI (China National Knowledge Infrasrtucture), Wanfang database and VIP database. The case-control studies were selected according to defined inclusion and exclusion criteria. After quality evaluation and data abstraction, a Meta-analysis was performed by using STATA 12.0 software. Odds ratio (OR) of the association between NOS3 894G>T and prostate cancer susceptibility was pooled. Then the subgroup analysis, sensitivity analysis and publication bias test were performed. Results: A total of 5 case-control studies were eligible for this analysis, including 3 078 cases and 3 677 healthy controls. Meta-analysis showed that NOS3 894G>T polymorphism didn't increase the risk of prostate cancer [TT vs GG, OR = 0.95, 95% confidence interval (CI): 0.80-1.14; TT vs GT, OR = 0.88, 95% CI: 0.73-1.05; TT+GT vs GG, OR = 1.07, 95% CI: 0.97-1.18; TT vs GG+GT, OR = 0.92, 95% CI: 0.77-1.09]. In the subgroup analysis of ethnicity, NOS3 894G>T polymorphism didn't increase the risk of prostate cancer in Europeans (TT vs GG, OR = 0.87, 95% CI: 0.73-1.04; TT vs GT, OR = 0.85, 95% CI: 0.71-1.02; TT+GT vs GG, OR = 1.00, 95% CI: 0.90-1.11; TT vs GG+GT, OR = 0.86, 95% CI: 0.72-1.02). Conclusion: NOS3 894G>T polymorphism is not associated with the prostate cancer susceptibility as well as in Europeans. Copyright © 2014 by TUMOR.

7.
Journal of the Korean Society of Hypertension ; : 75-87, 2012.
Article in English | WPRIM | ID: wpr-51850

ABSTRACT

There are several aspects of blood pressure. Clinically, how to best assess blood pressure average and variability is still a matter of the ongoing debate. Besides office blood pressure, we must pay more careful attention focused on hypertension with blood pressure fluctuation. Impaired endothelial function is intimately associated with the development of hypertension and atherosclerosis. In this review, we describe the relation between endothelial dysfunction and hypertension, the effect of gene polymorphism on endothelial dysfunction, the effects of antihypertensive agents and dietary supplementation on impaired endothelial function in hypertension. In order to predict the future atherosclerosis and cardiovascular events in subjects with hypertension, the adequate assessment of endothelial function is one of the most reliable markers. Furthermore, we discuss the close relationship between blood pressure variability and endothelial function. Blood pressure variability during a day or a week is an important, new risk factor for cardiovascular disease and restoring impaired endothelial function might be a target to prevent blood pressure variation and future cardiovascular events.


Subject(s)
Antihypertensive Agents , Atherosclerosis , Blood Pressure , Cardiovascular Diseases , Dietary Supplements , Hypertension , Nitric Oxide , Nitric Oxide Synthase Type III , Risk Factors
8.
Experimental & Molecular Medicine ; : 191-201, 2012.
Article in English | WPRIM | ID: wpr-194084

ABSTRACT

Nitric oxide (NO) produced by endothelial NO synthase (eNOS) plays an important role in vascular functions, including vasorelaxation. We here investigated the pharmacological effect of the natural product syringaresinol on vascular relaxation and eNOS-mediated NO production as well as its underlying biochemical mechanism in endothelial cells. Treatment of aortic rings from wild type, but not eNOS-/- mice, with syringaresinol induced endothelium-dependent relaxation, which was abolished by addition of the NOS inhibitor NG-monomethyl-L-arginine. Treatment of human endothelial cells and mouse aortic rings with syringaresinol increased NO production, which was correlated with eNOS phosphorylation via the activation of Akt and AMP kinase (AMPK) as well as elevation of intracellular Ca2+ levels. A phospholipase C (PLC) inhibitor blocked the increases in intracellular Ca2+ levels, AMPK-dependent eNOS phosphorylation, and NO production, but not Akt activation, in syringaresinol-treated endothelial cells. Syringaresinol-induced AMPK activation was inhibited by co-treatment with PLC inhibitor, Ca2+ chelator, calmodulin antagonist, and CaMKKbeta siRNA. This compound also increased eNOS dimerization, which was inhibited by a PLC inhibitor and a Ca2+-chelator. The chemicals that inhibit eNOS phosphorylation and dimerization attenuated vasorelaxation and cGMP production. These results suggest that syringaresinol induces vasorelaxation by enhancing NO production in endothelial cells via two distinct mechanisms, phosphatidylinositol 3-kinase/Akt- and PLC/Ca2+/CaMKKbeta-dependent eNOS phosphorylation and Ca2+-dependent eNOS dimerization.


Subject(s)
Animals , Humans , Mice , Aorta/drug effects , Enzyme Activation/drug effects , Furans/pharmacology , Gene Deletion , Human Umbilical Vein Endothelial Cells/drug effects , Lignans/pharmacology , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide Phospholipase C/metabolism , Phosphorylation/drug effects , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Vasodilation/drug effects
9.
Arq. bras. cardiol ; 96(1): 68-75, jan. 2011. ilus
Article in Portuguese | LILACS | ID: lil-573607

ABSTRACT

O óxido nítrico (NO), primariamente identificado como um fator relaxante derivado do endotélio, é um radical livre atuante na sinalização de diferentes processos biológicos. A identificação das isoformas das sintases do NO (NOS) e a subsequente caracterização dos mecanismos de ativação celulares das enzimas possibilitaram tanto a compreensão de parte das interações fisiológicas como a compreensão de parte dos mecanismos de doença, na qual o NO está envolvido. A isoforma endotelial da NOS (eNOS), expressa principalmente no endotélio vascular, desempenha importante papel na regulação da reatividade vascular e no desenvolvimento e na progressão da aterosclerose. Esta revisão tem o propósito de contextualizar o leitor sobre a estrutura da eNOS e seus mecanismos de ativação celular. Tendo em vista os avanços da biologia molecular, trataremos ainda dos conhecidos mecanismos de regulação da expressão gênica e do papel de variantes no código genético da eNOS associados a fenótipos cardiovasculares. Embora se reconheça a importância do NO como molécula ateroprotetora, nossa atenção estará voltada à revisão de literatura envolvendo NO e sua participação na modulação do fenótipo de vasodilatação muscular.


Nitric oxide (NO), primarily identified as an endothelium-derived relaxing factor, is a free radical that signals different biological processes. The identification of NO synthase (NOS) isoforms and the subsequent characterization of the mechanisms of cell activation of the enzymes permitted the partial understanding of both the physiological interactions and of the mechanisms of the diseases in which NO is involved. Mainly expressed in the vascular endothelium, the endothelial NOS isoform (eNOS) plays an important role in the regulation of vascular reactivity and in the development and progression of atherosclerosis. The purpose of this review is to contextualize the reader about the eNOS structure and its mechanisms of cell activation. In view of the advances in molecular biology, we will also address the known mechanisms of gene expression regulation and the role of variants on the genetic code of eNOS associated with cardiovascular phenotypes. Although the importance of NO as an atheroprotective molecule is recognized, our focus will be the review of the literature on NO and its participation in the modulation of the muscle vasodilatation phenotype.


El óxido nítrico (NO), primariamente identificado como un factor relajante derivado del endotelio, es un radical libre actuante en la señalización de diferentes procesos biológicos. La identificación de las isoformas de las sintasas del NO (NOS) y la subsecuente caracterización de los mecanismos de activación celulares de las enzimas posibilitaron tanto la comprensión de parte de las interacciones fisiológicas como la comprensión de parte de los mecanismos de enfermedad, en la cual el NO está envuelto. La isoforma endotelial de la NOS (eNOS), expresada principalmente en el endotelio vascular, desempeña importante papel en la regulación de la reactividad vascular y en el desarrollo y en la progresión de la aterosclerosis. Esta revisión tiene el propósito de contextualizar al lector sobre la estructura de la eNOS y sus mecanismos de activación celular. Teniendo en vista los avances de la biología molecular, trataremos aun de los conocidos mecanismos de regulación de la expresión génica y del papel de variantes en el código genético de la eNOS asociados a fenotipos cardiovasculares. Aunque se reconozca la importancia del NO como molécula ateroprotectora, nuestra atención estará volcada a la revisión de literatura envolviendo NO y su participación en la modulación del fenotipo de vasodilatación muscular.


Subject(s)
Humans , Cardiovascular Diseases/physiopathology , Endothelium, Vascular/physiopathology , Nitric Oxide Synthase Type III/physiology , Nitric Oxide Synthase/physiology , Nitric Oxide/physiology , Enzyme Activation , Endothelium-Dependent Relaxing Factors/metabolism , Gene Expression Regulation, Enzymologic/physiology , Muscles/blood supply , Nitric Oxide Synthase Type III/genetics , Nitric Oxide/genetics , Phenotype , Vasoconstriction/physiology , Vasodilation/physiology
10.
Experimental & Molecular Medicine ; : 768-776, 2010.
Article in English | WPRIM | ID: wpr-122639

ABSTRACT

Deregulation of endothelial nitric oxide synthase (eNOS) plays an important role in the development of multiple cardiovascular diseases. Our recent study demonstrated that genistein supplementation attenuates pulmonary arterial hypertension in broilers by restoration of endothelial function. In this study, we investigated the molecular mechanism by using broiler pulmonary arterial endothelial cells (PAECs). Our results showed that genistein stimulated a rapid phosphorylation of eNOS at Ser(1179) which was associated with activation of eNOS/NO axis. Further study indicated that the activation of eNOS was not mediated through estrogen receptors or tyrosine kinase inhibition, but via a phosphatidylinositol 3-kinase (PI3K)/Akt-dependent signaling pathway, as the eNOS activity and related NO release were largely abolished by pharmacological inhibitors of PI3K or Akt. Thus, our findings revealed a critical function of Akt in mediating genistein-stimulated eNOS activity in PAECs, partially accounting for the beneficial effects of genistein on the development of cardiovascular diseases observed in animal models.


Subject(s)
Animals , Female , Humans , Cell Line , Chickens , Disease Models, Animal , Endothelium, Vascular/drug effects , Enzyme Activation/drug effects , Genistein/pharmacology , Hypertension, Pulmonary/drug therapy , Nitric Oxide Synthase Type III/genetics , Oncogene Protein v-akt/metabolism , Phosphorylation , Signal Transduction/drug effects
11.
Experimental & Molecular Medicine ; : 524-532, 2010.
Article in English | WPRIM | ID: wpr-214627

ABSTRACT

Arginase competitively inhibits nitric oxide synthase (NOS) via use of the common substrate L-arginine. Arginase II has recently reported as a novel therapeutic target for the treatment of cardiovascular diseases such as atherosclerosis. Here, we demonstrate that piceatannol-3'-O-beta-D-glucopyranoside (PG), a potent component of stilbenes, inhibits the activity of arginase I and II prepared from mouse liver and kidney lysates, respectively, in a dose-dependent manner. In human umbilical vein endothelial cells, incubation of PG markedly blocked arginase activity and increased NOx production, as measured by Griess assay. The PG effect was associated with increase of eNOS dimer ratio, although the protein levels of arginase II or eNOS were not changed. Furthermore, isolated mice aortic rings treated with PG showed inhibited arginase activity that resulted in increased nitric oxide (NO) production upto 78%, as measured using 4-amino-5-methylamino-2',7'-difluorescein (DAF-FM) and a decreased superoxide anions up to 63%, as measured using dihydroethidine (DHE) in the intact endothelium. PG showed IC50 value of 11.22 microM and 11.06 microM against arginase I and II, respectively. PG as an arginase inhibitor, therefore, represents a novel molecule for the therapy of cardiovascular diseases derived from endothelial dysfunction and may be used for the design of pharmaceutical compounds.


Subject(s)
Animals , Humans , Mice , Aorta/drug effects , Arginase/antagonists & inhibitors , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Glucosides/chemistry , Mice, Inbred C57BL , Nitrates/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Nitrites/metabolism , Reactive Oxygen Species/metabolism , Rheum/chemistry , Stilbenes/chemistry
12.
Journal of Korean Medical Science ; : 16-23, 2010.
Article in English | WPRIM | ID: wpr-225001

ABSTRACT

Hypertension is associated with endothelial dysfunction and increased cardiovascular risk. Caveolin-1 regulates nitric oxide (NO) signaling by modulating endothelial nitric oxide synthase (eNOS). The purpose of this study was to examine whether HMG-CoA reductase inhibitor improves impaired endothelial function of the aorta in spontaneous hypertensive rat (SHR) and to determine the underlying mechanisms involved. Eight-week-old male SHR were assigned to either a control group (CON, n=11) or a rosuvastatin group (ROS, n=12), rosuvastatin (10 mg/kg/day) administered for eight weeks. Abdominal aortic rings were prepared and responses to acetylcholine (10-9-10-4 M) were determined in vitro. To evaluate the potential role of NO and caveolin-1, we examined the plasma activity of NOx, eNOS, phosphorylated-eNOS and expression of caveolin-1. The relaxation in response to acetylcholine was significantly enhanced in ROS compared to CON. Expression of eNOS RNA was unchanged, whereas NOx level and phosphorylated-eNOS at serine-1177 was increased accompanied with depressed level of caveolin-1 in ROS. We conclude that 3-Hydroxy-3-methylglutaryl Coenzyme-A (HMG-CoA) reductase inhibitor can improve impaired endothelial dysfunction in SHR, and its underlying mechanisms are associated with increased NO production. Furthermore, HMG-CoA reductase inhibitor can activate the eNOS by phosphorylation related to decreased caveolin-1 abundance. These results imply the therapeutic strategies for the high blood pressure-associated endothelial dysfunction through modifying caveolin status.


Subject(s)
Animals , Male , Rats , Acetylcholine/metabolism , Aorta/metabolism , Blood Pressure/drug effects , Caveolin 1/metabolism , Down-Regulation , Drug Administration Schedule , Endothelium, Vascular/drug effects , Fluorobenzenes/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hypertension/enzymology , Nitric Oxide/blood , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Pyrimidines/administration & dosage , Rats, Inbred SHR , Sulfonamides/administration & dosage , Vasodilation/drug effects
13.
Tumor ; (12): 290-293, 2007.
Article in Chinese | WPRIM | ID: wpr-849598

ABSTRACT

Objective: Angiogenesis is a critical step in tumor growth and is regulated by multiple molecular pathways. We evaluted the expression of nitric oxide synthase (NOS) III and its relationship with the angiogenic phenotypes and expressions of the transcription factor Sp1, as well as their effect on survival rate of patients with gastric cancer. Methods: The expression levels of NOS III and Sp1, and tumor microvessel density (MVD) status in 86 specimens of resected gastric cancer tissues were determined by immunohistochemistry. Results: NOS III protein expression was significantly higher in both primary tumors and lymph node metastases than in normal gastric mucosa. In primary tumors, NOS III expression had close correlation with Sp1 expression (P=0.001) and MVD status (P=0.001). Patients with positive Sp1 expression were more likely to have strong NOS III expression (15 times) and a high MVD (7 times) than those with negative Sp1 expression. Univariate survival analysis showed that strong NOS III expression, strong Sp1 expression, and a high MVD were associated with poor prognosis. In a Cox proportional hazards model, only strong NOS III and Sp1 expression as well as advanced disease stage were independent factors for prognosis of patients. Conclusion: Our results indicate that the expression of NOS III may play an important role in tumor angiogenesis and infiltration of gastric cancer.

14.
Experimental & Molecular Medicine ; : 63-71, 2006.
Article in English | WPRIM | ID: wpr-77900

ABSTRACT

Fluid shear stress plays a critical role in vascular health and disease. While protein kinase A (PKA) has been implicated in shear-stimulated signaling events in endothelial cells, it remains unclear whether and how PKA is stimulated in response to shear stress. This issue was addressed in the present study by monitoring the phosphorylation of endogenous substrates of PKA. Shear stress stimulated the phosphorylation of cAMP responsive element binding protein (CREB) in a PKA-dependent manner. Western blot analysis using the antibody reactive against the consensus motif of PKA substrates detected two proteins, P135 and P50, whose phosphorylation was increased by shear stress. The phosphorylation of P135 was blocked by a PKA inhibitor, H89, but not by a phosphoinositide 3-kinase inhibitor, wortmannin. Expression of a constitutively active PKA subunit stimulated P135 phosphorylation, supporting the potential of P135 as a PKA substrate. P135 was identified as endothelial nitric oxide synthase (eNOS) by immunoprecipitation study. PKA appeared to mediate shear stress-stimulated eNOS activation. Shear stress stimulated intracellular translocation of PKA activity from 'soluble' to 'particulate' fractions without involving cellular cAMP increase. Taken together, this study suggests that shear stress stimulates PKA-dependent phosphorylation of target proteins including eNOS, probably by enhancing intracellular site-specific interactions between protein kinase and substrates.


Subject(s)
Animals , Cattle , Aorta, Thoracic/cytology , Blotting, Western , Cell Culture Techniques , Cell Extracts , Cells, Cultured , Comparative Study , Cyclic AMP-Dependent Protein Kinases/analysis , Endothelium, Vascular/cytology , Nitric Oxide Synthase Type III/analysis , Phosphorylation , Precipitin Tests , Stress, Mechanical , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL