• Title/Summary/Keyword: vascular dysfunction

Search Result 152, Processing Time 0.019 seconds

Nafamostat Mesilate Inhibits TNF-${\alpha}$-Induced Vascular Endothelial Cell Dysfunction by Inhibiting Reactive Oxygen Species Production

  • Kang, Min-Woong;Song, Hee-Jung;Kang, Shin Kwang;Kim, Yonghwan;Jung, Saet-Byel;Jee, Sungju;Moon, Jae Young;Suh, Kwang-Sun;Lee, Sang Do;Jeon, Byeong Hwa;Kim, Cuk-Seong
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.19 no.3
    • /
    • pp.229-234
    • /
    • 2015
  • Nafamostat mesilate (NM) is a serine protease inhibitor with anticoagulant and anti-inflammatory effects. NM has been used in Asia for anticoagulation during extracorporeal circulation in patients undergoing continuous renal replacement therapy and extra corporeal membrane oxygenation. Oxidative stress is an independent risk factor for atherosclerotic vascular disease and is associated with vascular endothelial function. We investigated whether NM could inhibit endothelial dysfunction induced by tumor necrosis factor-${\alpha}$ (TNF-${\alpha}$ ). Human umbilical vein endothelial cells (HUVECs) were treated with TNF-${\alpha}$ for 24 h. The effects of NM on monocyte adhesion, vascular cell adhesion molecule-1 (VCAM-1) and intracellular adhesion molecule-1 (ICAM-1) protein expression, p38 mitogenactivated protein kinase (MAPK) activation, and intracellular superoxide production were then examined. NM ($0.01{\sim}100{\mu}g/mL$) did not affect HUVEC viability; however, it inhibited the increases in reactive oxygen species (ROS) production and p66shc expression elicited by TNF-${\alpha}$ (3 ng/mL), and it dose dependently prevented the TNF-${\alpha}$ -induced upregulation of endothelial VCAM-1 and ICAM-1. In addition, it mitigated TNF-${\alpha}$ -induced p38 MAPK phosphorylation and the adhesion of U937 monocytes. These data suggest that NM mitigates TNF-${\alpha}$ -induced monocyte adhesion and the expression of endothelial cell adhesion molecules, and that the anti-adhesive effect of NM is mediated through the inhibition of p66shc, ROS production, and p38 MAPK activation.

Effects of Radix Angelicae Gigantis and Resina Ferulae on the Relaxation of Smooth Muscle and Expression of iNOS (당귀 및 아위가 평활근 이완과 iNOS 발현에 미치는 영향)

  • 김성재;송봉근;이언정;김형균;김중길
    • The Journal of Korean Medicine
    • /
    • v.21 no.2
    • /
    • pp.60-67
    • /
    • 2000
  • Objectives : Radix Angelicae Gigantis(RAG) and Resina Ferulae(RF) have been used in oriental medicine or folk medicine to increase stamina. The aim of this study was the characterization of the mechanism of action of RAG and RF on smooth muscle and macrophages in rats to find new substances for the treatment of erectile dysfunction, cardiovascular diseases and immune dysfunction. Methods : We investigated the effects of the water extracts of RAG and RF on phenylephrine or KCl-contracted rat endothelium-denuded aorta, the production of NO in vascular smooth muscle cell (VSMC) and the production of NO and induction of iNOS in the $IFN-{\gamma}-primed$ RAW 264.7 cells. Results : The water extracts of the RAG and RF showed significant concentration-dependent relaxation effects on phenylephrine or KCl-contracted rat endothelium-denuded aorta. It also reduced the tension of the rat endothelium denuded aorta which was contracted in $Ca^{2+}-free$ media. On the other hand, it increased production of NO in VSMC which was stimulated with $IL-{\beta}$ or $IL-{\beta}$ plus $IFN-{\gamma}$. The water extracts of RAG and RF increased production of NO and induction of iNOS in the $IFN-{\gamma}-primed$ RAW 264.7 cells. Conclusions : According to the above results, the water extracts of RAG and RF relaxed the smooth muscle effectively and increased the production of NO in VSMC and macrophages. So, these herbs can be applied to erectile dysfunction, hypertension, angina pectoris, artherosclerosis and a defense defect for virus or microbe.

  • PDF

Effects of Antioxidant Tempol on Systematic Inflammation and Endothelial Apoptosis in Emphysematous Rats Exposed to Intermittent Hypoxia

  • Zhao, Haiyan;Zhao, Yaping;Li, Xin;Xu, Leiqian;Jiang, Fangxin;Hou, Wanju;Dong, Lixia;Cao, Jie
    • Yonsei Medical Journal
    • /
    • v.59 no.9
    • /
    • pp.1079-1087
    • /
    • 2018
  • Purpose: Obstructive sleep apnea and chronic obstructive pulmonary disease are independent risk factors of cardiovascular disease (CVD), and their coexistence is known as overlap syndrome (OS). Endothelial dysfunction is the initial stage of CVD; however, underlying mechanisms linking OS and CVD are not well understood. The aim of this study was to explore whether OS can lead to more severe inflammation and endothelial apoptosis by promoting endothelial dysfunction, and to assess the intervention effects of antioxidant tempol. Materials and Methods: Male Wistar rats (n=66) were exposed to normal oxygen [normal control (NC) group], intermittent hypoxia (IH group), cigarette smoke (CH group), as well as cigarette smoke and IH (OS group). Tempol intervention was assessed in OS group treated with tempol (OST group) or NaCl (OSN group). After an 8-week challenge, lung tissues, serum, and fresh blood were harvested for analysis of endothelial markers and apoptosis. Results: The levels of intracellular adhesion molecule-1, vascular cellular adhesion molecule-1, and apoptosis in circulating epithelial cells were the highest in OS group and the lowest in NC group. These levels were all greater in IH group than in CH group, and were lower in OST group than in OS and OSN groups (all p<0.001). Conclusion: Synergistic effects of IH with cigarette smoke-induced emphysema produce a greater inflammatory status and endothelial apoptosis. OS-related inflammation and endothelial cell apoptosis may play important roles in promoting cardiovascular dysfunction, and antioxidant tempol could achieve a partial protective effect.

Whole body hypoxic preconditioning-mediated multiorgan protection in db/db mice via nitric oxide-BDNF-GSK-3β-Nrf2 signaling pathway

  • Li, Yuefang;Huang, Yan;Cheng, Xi;He, Youjun;Hu, Xin
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.25 no.4
    • /
    • pp.281-296
    • /
    • 2021
  • The beneficial effects of hypoxic preconditioning are abolished in the diabetes. The present study was designed to investigate the protective effects and mechanisms of repeated episodes of whole body hypoxic preconditioning (WBHP) in db/db mice. The protective effects of preconditioning were explored on diabetes-induced vascular dysfunction, cognitive impairment and ischemia-reperfusion (IR)-induced increase in myocardial injury. Sixteen-week old db/db (diabetic) and C57BL/6 (non-diabetic) mice were employed. There was a significant impairment in cognitive function (Morris Water Maze test), endothelial function (acetylcholine-induced relaxation in aortic rings) and a significant increase in IR-induced heart injury (Langendorff apparatus) in db/db mice. WBHP stimulus was given by exposing mice to four alternate cycles of low (8%) and normal air O2 for 10 min each. A single episode of WBHP failed to produce protection; however, two and three episodes of WBHP significantly produced beneficial effects on the heart, brain and blood vessels. There was a significant increase in the levels of brain-derived neurotrophic factor (BDNF) and nitric oxide (NO) in response to 3 episodes of WBHP. Moreover, pretreatment with the BDNF receptor, TrkB antagonist (ANA-12) and NO synthase inhibitor (L-NAME) attenuated the protective effects imparted by three episodes of WBHP. These pharmacological agents abolished WBHP-induced restoration of p-GSK-3β/GSK-3β ratio and Nrf2 levels in IR-subjected hearts. It is concluded that repeated episodes of WHBP attenuate cognitive impairment, vascular dysfunction and enhancement in IR-induced myocardial injury in diabetic mice be due to increase in NO and BDNF levels that may eventually activate GSK-3β and Nrf2 signaling pathway to confer protection.

Different effects of prolonged β-adrenergic stimulation on heart and cerebral artery

  • Shin, Eunji;Ko, Kyung Soo;Rhee, Byoung Doo;Han, Jin;Kim, Nari
    • Integrative Medicine Research
    • /
    • v.3 no.4
    • /
    • pp.204-210
    • /
    • 2014
  • The aim of this review was to understand the effects of ${\beta}$-adrenergic stimulation on oxidative stress, structural remodeling, and functional alterations in the heart and cerebral artery. Diverse stimuli activate the sympathetic nervous system, leading to increased levels of catecholamines. Long-term overstimulation of the ${\beta}$-adrenergic receptor (${\beta}AR$) in response to catecholamines causes cardiovascular diseases, including cardiac hypertrophy, stroke, coronary artery disease, and heartfailure. Although catecholamines have identical sites of action in the heart and cerebral artery, the structural and functional modifications differentially activate intracellular signaling cascades. ${\beta}AR$-stimulation can increase oxidative stress in the heart and cerebral artery, but has also been shown to induce different cytoskeletal and functional modifications by modulating various components of the ${\beta}AR$ signal transduction pathways. Stimulation of ${\beta}AR$ leads to cardiac dysfunction due to an overload of intracellular $Ca^{2+}$ in cardiomyocytes. However, this stimulation induces vascular dysfunction through disruption of actin cytoskeleton in vascular smooth muscle cells. Many studies have shown that excessive concentrations of catecholamines during stressful conditions can produce coronary spasms or arrhythmias by inducing $Ca^{2+}$-handling abnormalities and impairing energy production in mitochondria, In this article, we highlight the different fates caused by excessive oxidative stress and disruptions in the cytoskeletal proteome network in the heart and the cerebral artery in responsed to prolonged ${\beta}AR$-stimulation.

Endothelial Ca2+ signaling-dependent vasodilation through transient receptor potential channels

  • Hong, Kwang-Seok;Lee, Man-Gyoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.24 no.4
    • /
    • pp.287-298
    • /
    • 2020
  • Ca2+ signaling of endothelial cells plays a critical role in controlling blood flow and pressure in small arteries and arterioles. As the impairment of endothelial function is closely associated with cardiovascular diseases (e.g., atherosclerosis, stroke, and hypertension), endothelial Ca2+ signaling mechanisms have received substantial attention. Increases in endothelial intracellular Ca2+ concentrations promote the synthesis and release of endothelial-derived hyperpolarizing factors (EDHFs, e.g., nitric oxide, prostacyclin, or K+ efflux) or directly result in endothelial-dependent hyperpolarization (EDH). These physiological alterations modulate vascular contractility and cause marked vasodilation in resistance arteries. Transient receptor potential (TRP) channels are nonselective cation channels that are present in the endothelium, vascular smooth muscle cells, or perivascular/sensory nerves. TRP channels are activated by diverse stimuli and are considered key biological apparatuses for the Ca2+ influx-dependent regulation of vasomotor reactivity in resistance arteries. Ca2+-permeable TRP channels, which are primarily found at spatially restricted microdomains in endothelial cells (e.g., myoendothelial projections), have a large unitary or binary conductance and contribute to EDHFs or EDH-induced vasodilation in concert with the activation of intermediate/small conductance Ca2+-sensitive K+ channels. It is likely that endothelial TRP channel dysfunction is related to the dysregulation of endothelial Ca2+ signaling and in turn gives rise to vascular-related diseases such as hypertension. Thus, investigations on the role of Ca2+ dynamics via TRP channels in endothelial cells are required to further comprehend how vascular tone or perfusion pressure are regulated in normal and pathophysiological conditions.

Ginsenoside Rg3 protects against iE-DAP-induced endothelial-to-mesenchymal transition by regulating the miR-139-5p-NF-κB axis

  • Lee, Aram;Yun, Eunsik;Chang, Woochul;Kim, Jongmin
    • Journal of Ginseng Research
    • /
    • v.44 no.2
    • /
    • pp.300-307
    • /
    • 2020
  • Background: Emerging evidence suggests that endothelial-to-mesenchymal transition (EndMT) in endothelial dysfunction due to persistent inflammation is a key component and emerging concept in the pathogenesis of vascular diseases. Ginsenoside Rg3 (Rg3), an active compound from red ginseng, has been known to be important for vascular homeostasis. However, the effect of Rg3 on inflammation-induced EndMT has never been reported. Here, we hypothesize that Rg3 might reverse the inflammation-induced EndMT and serve as a novel therapeutic strategy for vascular diseases. Methods: EndMT was examined under an inflammatory condition mediated by the NOD1 agonist, γ-d-glutamyl-meso-diaminopimelic acid (iE-DAP), treatment in human umbilical vein endothelial cells. The expression of EndMT markers was determined by Western blot analysis, real-time polymerase chain reaction, and immunocytochemistry. The underlying mechanisms of Rg3-mediated EndMT regulation were investigated by modulating the microRNA expression. Results: The NOD1 agonist, iE-DAP, led to a fibroblast-like morphology change with a decrease in the expression of endothelial markers and an increase in the expression of the mesenchymal marker, namely EndMT. On the other hand, Rg3 markedly attenuated the iE-DAP-induced EndMT and preserved the endothelial phenotype. Mechanically, miR-139 was downregulated in cells with iE-DAP-induced EndMT and partly reversed in response to Rg3 via the regulation of NF-κB signaling, suggesting that the Rg3-miR-139-5p-NF-κB axis is a key mediator in iE-DAP-induced EndMT. Conclusion: These results suggest, for the first time, that Rg3 can be used to inhibit inflammation-induced EndMT and may be a novel therapeutic option against EndMT-associated vascular diseases.

Altered Vascular Expression of Nitric Oxide Synthase Isozymes in Hypertension (고혈압에서 혈관 산화질소 합성 동위 효소 발현 변화)

  • 김인광;강대길;이종은;오봉석
    • Journal of Chest Surgery
    • /
    • v.32 no.2
    • /
    • pp.138-143
    • /
    • 1999
  • Background: The endothelium-dependent vasorelaxation has been largely accounted for by the release of nitric oxide (NO). Three distinct isoforms of NO synthases (NOS) have been characterized, i.e., brain(bNOS), inducible (iNOS), and endothelial constitutive (ecNOS). Although hypertension hasbeen associated with a vascular endothelial dysfunction, changes in the vascular expression of NOS isoforms have not been established. The present study was aimed at exploring the vascular expression of NOS isozymes in hypertension. Material and Method: Two-kidney, one clip (2K1C) and deoxycorticosterone acetate (DOCA)-salt hypertension were induced in rats. The expression of different NOS isozymes in the thoracic aorta was determined by Western blot analysis. The vascular tissue contents of nitrites were measured by colorimetric assay. Result: Arterial blood pressure was significantly higher in experimental groups of 2K1C and DOCA-salt rats compared with their corresponding control rats. The vascular expression of bNOS as well as that of ecNOS was decreased in both models of hypertension. iNOS was not changed in DOCA-salt hypertension, but was also decreased in 2K1C hypertension. The vascular contents of nitrites were significantly decreased in DOCA-salt as well as in 2K1C hypertension. Conclusion: These results suggest that 2K1C and DOCA-salt hypertension are associated with decreases in the vascular expression of NOS isozymes and nitrite contents.

  • PDF

Study on Infrared Thermography (적외석 체열촬영에 관한 고찰)

  • Kim, Ho-Bong
    • The Journal of Korean Academy of Orthopedic Manual Physical Therapy
    • /
    • v.1 no.1
    • /
    • pp.9-14
    • /
    • 1995
  • Thermography is a diagnostic procedures that measures infrared energy emitted by the skin. Thermography detects body temperature change which are controlled by the autonomic nervous system. It does show the thermal dysfunction that correlates closely with pain syndromes as well as normalization when the healing process takes place. Experienced clinical interpretation of the thermal pattern is necessary to diagnose and establish causation. Thermography is useful in the diagnosis of painful conditions such as herniated disc diseases, myofascial syndrome, myositis, musculoligamentous injury, reflex sympathetic dystrophy, athretic injuries, vascular diseases, arthritis, inflammation and breast tumors.

  • PDF

Resveratrol attenuates lipopolysaccharide-induced dysfunction of blood-brain barrier in endothelial cells via AMPK activation

  • Hu, Min;Liu, Bo
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.20 no.4
    • /
    • pp.325-332
    • /
    • 2016
  • Resveratrol, a phytoalexin, is reported to activate AMP-activated protein kinase (AMPK) in vascular cells. The blood-brain barrier (BBB), formed by specialized brain endothelial cells that are interconnected by tight junctions, strictly regulates paracellular permeability to maintain an optimal extracellular environment for brain homeostasis. The aim of this study was to elucidate the effects of resveratrol and the role of AMPK in BBB dysfunction induced by lipopolysaccharide (LPS). Exposure of human brain microvascular endothelial cells (HBMECs) to LPS ($1{\mu}g/ml$) for 4 to 24 hours week dramatically increased the permeability of the BBB in parallel with lowered expression levels of occluding and claudin-5, which are essential to maintain tight junctions in HBMECs. In addition, LPS significantly increased the reactive oxygen species (ROS) productions. All effects induced by LPS in HBVMCs were reversed by adenoviral overexpression of superoxide dismutase, inhibition of NAD(P) H oxidase by apocynin or gain-function of AMPK by adenoviral overexpression of constitutively active mutant (AMPK-CA) or by resveratrol. Finally, upregulation of AMPK by either AMPK-CA or resveratrol abolished the levels of LPS-enhanced NAD(P)H oxidase subunits protein expressions. We conclude that AMPK activation by resveratrol improves the integrity of the BBB disrupted by LPS through suppressing the induction of NAD(P)H oxidase-derived ROS in HBMECs.