首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 218 毫秒
1.
肾素-血管紧张素系统的新调节分子:ACE2   总被引:2,自引:0,他引:2  
Li YT  Cheng GF 《生理科学进展》2006,37(2):179-181
血管紧张素转化酶(angiotensin—converting enzyme,ACE)为含锌的金属蛋白酶,是肾素-血管紧张素系统(renin—angiotensin system,RAS)重要的调节分子。血管紧张素转化酶2(angiotensin—con—verting enzyme2,ACE2)是迄今发现的唯一的ACE同系物(homologue),它主要分布于睾丸、肾脏和心脏。ACE2可水解血管紧张素Ⅰ(angiotensinⅠ,AngⅠ)和血管紧张素Ⅱ(angiotensinⅡ,AngⅡ)羧基端的1个氨基酸残基,分别形成Ang1-9和有血管舒张作用的Ang1-7。ACE2的生理病理作用还不甚明了,传统的ACE抑制剂不能抑制ACE2的活性。ACE2在心血管、肾脏系统的作用可能与ACE相反.与ACE共同调节心脏、肾脏等脏器的正常功能。  相似文献   

2.
血管紧张素-(1-7)[angiotensin(1-7), Ang-(1-7) ]是肾素-血管紧张素系 统(renin-angiotensin system, RAS)的新成员之一. 与血管紧张素Ⅱ(Ang Ⅱ)比 较,Ang-(1-7)在高血压及动脉粥样硬化等疾病中发挥着心血管保护作用. 最新研究 发现,Ang-(1-7)在平滑肌细胞增殖及迁移、内皮细胞功能、脂质代谢的这些动脉粥样硬化相关病理发展过程中起着重要作用,能够抑制动脉粥样硬化斑块的形成及加强斑块的稳定性. 本文就Ang-(1-7)与动脉粥样硬化疾病相关的最新研究进展进行综述,讨论Ang-(1-7)在动脉粥样硬化中的作用机制,也为Ang-(1-7)相关的ACE2/Ang -(1-7)/Mas轴作为未来临床治疗途径提供思路.  相似文献   

3.
血管紧张素转换酶2(angiotensin—converting enzyme 2,ACE2)是新发现的与血管紧张素转换酶(ACE)相关的羧肽酶,在肾素-血管紧张素系统(rennin-angiotensin system,RAS)中ACE2可以使AngⅡ转换为Ang1-7,从而产生与血管紧张素Ⅱ相反的效应,同时ACE2还可使Ang I转换为Ang1-9。研究发现:ACE2与高血压、SARS以及肾脏、生殖等系统的疾病有着密切的关系。  相似文献   

4.
肾脏中肾素-血管紧张素系统的生理和病理生理作用   总被引:14,自引:0,他引:14  
肾脏中肾素-血管紧张素系统(RAS)在肾脏生理功能的调节中有重要作用.近年来,肾脏RAS的新成分及新作用机制不断被发现.转基因动物研究使肾脏血管紧张素Ⅱ(AngⅡ)在血压及水钠平衡调节中的作用进一步阐明;AngⅡ的非血流动力学作用已经确立;血管紧张素转换酶2(ACE2)及Ang 1~7对肾功能的调节作用也已得到认可.肾素/前肾素特异性受体、ACE的信号转导功能,以及AT1受体的转激活功能等,已成为肾脏生理科学研究的热点.这些研究对于人们认识肾脏局部RAS功能,探讨延缓慢性肾脏病的进展的治疗策略具有重要意义.  相似文献   

5.
血管紧张素转换酶2(angiotensin-converting enzyme 2,ACE2)是肾素-血管紧张素系统(renin-angio-tensin system,RAS)的重要成员,广泛分布于肺、心血管系统、肠道、肾脏、中枢神经系统和脂肪组织中.ACE2具有多种生理功能,在防止高血压、动脉粥样硬化、心力衰竭、肺...  相似文献   

6.
肾素-血管紧张素-醛固酮系统起初被认为是较简单的神经体液调节机制之一。但是,这一想法随着RAAS阻滞剂:肾素阻滞剂、血管紧张素转换酶抑制剂(ACEI)、AT1受体拮抗剂及盐皮质激素受体拮抗剂的深入研究而受到挑战。因此,RAAS的组成、以上药物发挥作用的具体通路及副作用均得到重新定义。在RAAS阻滞剂的应用过程中,机体肾素水平升高,并刺激肾素原受体(即无活性的肾素前体,PRR),进而对机体造成不良影响。同理,在AT1受体拮抗剂的应用过程中,血浆血管紧张素II的水平升高,并与2型血管紧张素II(AT2)受体结合,进而对机体产生有利作用。此外,随着ACEI及ARB的应用,血管紧张素1-7水平升高,其与Mas受体结合,发挥心脏及肾脏保护的作用,还可通过刺激干细胞发挥组织修复作用。  相似文献   

7.
人血管紧张素转换酶2(ACE2)是肾素-血管紧张素系统(RAS)的重要调节分子,它在控制心血管和血压的正常生理活动中具有重要的作用。此外,ACE2作为SARS病毒的受体,对于病毒的入侵起关键作用。目前ACE2已经被用于高血压和心血管相关疾病的药物靶标设计和基因治疗,随着研究的深入,ACE2在临床上的应用将更加广泛。  相似文献   

8.
人血管紧张素转换酶2(ACE2)是目前已知的惟一的人血管紧张素转换酶(ACE)的同源物,是一种新型的金属羧肽酶,很多特性与ACE截然不同.ACE2在肾素-血管紧张素系统(RAS)中具有独特的作用,调节心脏功能和机体血压.最近ACE2被鉴定为SARS病毒的功能受体.ACE2已经成为目前药物研发的新靶点.对ACE2的认识才刚刚开始,有待进-步深入研究.  相似文献   

9.
糖尿病时,肾素-血管紧张素系统(renin-angiotensin system,RAS)被激活,升高的血管紧张素Ⅱ(Ang Ⅱ)通过细胞表面的AT1受体,刺激心肌成纤维细胞增生及胶原代谢改变,引起心脏结构重塑,导致心肌间质及血管周围纤维化,胶原含量增多和排列紊乱,造成心室肌僵硬而影响舒张功能,出现糖尿病心肌病(diabetic cardiomyopathy,DCM)的临床症状.本文从RAS的主要成分Ang Ⅱ、Ang-(1-7)、Ac-SDKP和血管紧张素受体(ATR)与内皮素、活性氧、转化生长因子-β1、核因子-κB、信号转导系统以及细胞凋亡之间的相互作用,阐述RAS在糖尿病心肌病发生发展中所起的重要作用.  相似文献   

10.
本文旨在探究β-CM7对糖尿病大鼠心肌组织肾素-血管紧张素系统(Renin angiotensin system,RAS)的影响及其保护机制。32只雄性SD大鼠通过相应处理被分为正常对照组、模型对照组、胰岛素治疗组(3.7×10~(–8) mol/d)及β-CM7干预组(7.5×10~(–8) mol/d)。连续饲养30 d后,处死大鼠取心肌。β-CM7在干预糖尿病模型后,组织中AngⅡ含量显著降低,Ang1-7含量极显著升高;AT1受体和Mas受体mRNA表达均显著升高;ACE和ACE2的mRNA表达均显著升高,且酶活均显著升高。综上可得,β-CM7可以通过激活RAS的负性调节通路"ACE2-Ang1-7-Mas轴"显著抑制大鼠心肌ACE mRNA和蛋白的强表达,缓解AngⅡ对心肌组织的损伤,提示β-CM7抑制心肌损伤的作用可能与ACE/ACE2通路有关。  相似文献   

11.
The authors’ previous studies have indicated that angiotensin(Ang)-(1-7) protects the heart against reperfusion arrhythmias. The aim of this study was to determine whether a functional angiotensin-converting enzyme2 (ACE2)/Ang-(1-7)/Mas receptor axis is present in the sinoatrial node (SAN) of Wistar rats. SAN cells were identified by Masson’s trichrome staining, HCN4 expression, and lack of connexin43 expression. Immunohistochemistry technique was used to detect the expression of ACE2, Ang-(1-7), and Mas in the SAN. To evaluate the role of this axis in the SAN function, atrial tachyarrhythmias (ATs) were induced in isolated rat atria perfused with Krebs-Ringer solution (KRS) alone (control) or KRS containing Ang-(1-7). The specific Mas antagonist, A-779, was used to evaluate the role of Mas in the Ang-(1-7) effects. The findings showed that all components of the ACE2/Ang-(1-7)/Mas branch are present in the SAN of rats. Importantly, it was found that this axis is functional because perfusion of atria with Ang-(1-7) significantly reduced the duration of ATs. Additionally, this anti-arrhythmogenic effect was attenuated by A-779. No significant changes were observed in heart rate, contractile tension, or ±dT/dt. These observations demonstrate that the ACE2/Ang-(1-7)/Mas axis is expressed in SAN cells of rats. They provide the morphological support to the anti-arrhythmogenic effect of Ang-(1-7).  相似文献   

12.
The last decade has seen the discovery of several new components of the renin-angiotensin system (RAS). Among them, angiotensin converting enzyme-2 (ACE2) and the Mas receptor have forced a reevaluation of the original cascade and led to the emergence of a new arm of the RAS: the ACE2/ANG-(1-7)/Mas axis. Accordingly, the new system is now seen as a balance between a provasoconstrictor, profibrotic, progrowth axis (ACE/ANG-II/AT(1) receptor) and a provasodilatory, antifibrotic, antigrowth arm (ACE2/ANG-(1-7)/Mas receptor). Already, this simplistic vision is evolving and new components are branching out upstream [ANG-(1-12) and (pro)renin receptor] and downstream (angiotensin-IV and other angiotensin peptides) of the classical cascade. In this review, we will summarize the role of the ACE2/ANG-(1-7)/Mas receptor, focusing on the central nervous system with respect to cardiovascular diseases such as hypertension, chronic heart failure, and stroke, as well as neurological diseases. In addition, we will discuss the new pharmacological (antagonists, agonists, activators) and genomic (knockout and transgenic animals) tools that are currently available. Finally, we will review the latest data regarding the various signaling pathways downstream of the Mas receptor.  相似文献   

13.
Xue H  Zhou L  Yuan P  Wang Z  Ni J  Yao T  Wang J  Huang Y  Yu C  Lu L 《Regulatory peptides》2012,177(1-3):12-20
In the updated concept of renin-angiotensin system (RAS), it contains the angiotensin converting enzyme (ACE)-angiotensin (Ang) II-angtiogensin type 1 receptor (AT1) axis and the angiotensin-converting enzyme-related carboxypeptidase (ACE2)-Ang-(1-7)-Mas axis. The former axis has been well demonstrated performing the vasoconstrictive, proliferative and pro-inflammatory functions by activation of AT1 receptors, while the later new identified axis is considered counterbalancing the effects of the former. The present study is aimed at observing the interaction between Ang-(1-7) and Ang II on cultured rat renal mesangial cells (MCs). RT-PCR, Western blot and immunofluorescent staining and confocal microscopy results showed that both AT1 and Mas receptor were co-distributed in rat renal MCs. Ang-(1-7) showed similar effects on Ang II in cultured MCs that stimulated phosphorylated extracellular signal-regulated kinase (ERK)1/2 phosphorylation and transforms growth factor-β1 synthesis, and cell proliferation and extracellular matrix synthesis. Co-treatment of the cell with Ang-(1-7) and Ang II, Ang-(1-7) counteracted AngII-induced effects in a concentration dependent manner, but failed to alter the changes induced by endothelin-1. The stimulating effect of Ang II was mediated by AT1 receptor while all the effects of Ang-(1-7) were blocked by Mas receptor antagonist A-779, but not by AT1 receptor antagonist losartan or AT2 receptor antagonist PD123319. These results suggest that Ang-(1-7) and Ang II specifically interact with each other on rat renal MCs via activation of their specific receptors, Mas and AT1 receptor respectively.  相似文献   

14.
SARS-CoV-2, the virus responsible for the global coronavirus disease (COVID-19) pandemic, attacks multiple organs of the human body by binding to angiotensin-converting enzyme 2 (ACE2) to enter cells. More than 20 million people have already been infected by the virus. ACE2 is not only a functional receptor of COVID-19 but also an important endogenous antagonist of the renin-angiotensin system (RAS). A large number of studies have shown that ACE2 can reverse myocardial injury in various cardiovascular diseases (CVDs) as well as is exert anti-inflammatory, antioxidant, anti-apoptotic and anticardiomyocyte fibrosis effects by regulating transforming growth factor beta, mitogen-activated protein kinases, calcium ions in cells and other major pathways. The ACE2/angiotensin-(1-7)/Mas receptor axis plays a decisive role in the cardiovascular system to combat the negative effects of the ACE/angiotensin II/angiotensin II type 1 receptor axis. However, the underlying mechanism of ACE2 in cardiac protection remains unclear. Some approaches for enhancing ACE2 expression in CVDs have been suggested, which may provide targets for the development of novel clinical therapies. In this review, we aimed to identify and summarize the role of ACE2 in CVDs.  相似文献   

15.
Arecoline induces oral submucous fibrosis (OSF) via promoting the reactive oxygen species (ROS). Angiotensin (1–7) (Ang-(1–7)) protects against fibrosis by counteracting angiotensin II (Ang-II) via the Mas receptor. However, the effects of Ang-(1–7) on OSF remain unknown. NOD-like receptors (NLRs) family pyrin domain containing 3 (NLRP3) inflammasome is identified as the novel mechanism of fibrosis. Whereas the effects of arecoline on NLRP3 inflammasome remain unclear. We aimed to explore the effect of Ang-(1–7) on NLRP3 inflammasome in human oral myofibroblasts. In vivo, activation of NLRP3 inflammasomes with an increase of Ang-II type 1 receptor (AT1R) protein level and ROS production in human oral fibrosis tissues. Ang-(1–7) improved arecoline-induced rats OSF, reduced protein levels of NADPH oxidase 4 (NOX4) and the NLRP3 inflammasome. In vitro, arecoline increased ROS along with upregulation of the angiotensin-converting enzyme (ACE)/Ang-II/AT1R axis and NLRP3 inflammasome/interleukin-1β axis in human oral myofibroblasts, which were reduced by NOX4 inhibitor VAS2870, ROS scavenger N-acetylcysteine, and NOX4 small interfering RNA (siRNA). Furthermore, arecoline induced collagen synthesis or migration via the Smad or RhoA-ROCK pathway respectively, which could be inhibited by NLRP3 siRNA or caspase-1 blocker VX-765. Ang-(1–7) shifted the balance of RAS toward the ACE2/Ang-(1–7)/Mas axis, inhibited arecoline-induced ROS and NLRP3 inflammasome activation, leading to attenuation of migration or collagen synthesis. In summary, Ang-(1–7) attenuates arecoline-induced migration and collagen synthesis via inhibiting NLRP3 inflammasome in human oral myofibroblasts.  相似文献   

16.
Renin angiotensin system (RAS) is an endocrine system widely known for its physiological roles in electrolyte homeostasis, body fluid volume regulation and cardiovascular control in peripheral circulation. However, brain RAS is an independent form of RAS expressed locally in the brain, which is known to be involved in brain functions and disorders. There is strong evidence for a major involvement of excessive brain angiotensin converting enzyme (ACE)/Angiotensin II (Ang II)/Angiotensin type-1 receptor (AT-1R) axis in increased activation of oxidative stress, apoptosis and neuroinflammation causing neurodegeneration in several brain disorders. Numerous studies have demonstrated strong neuroprotective effects by blocking AT1R in these brain disorders. Additionally, the angiotensin converting enzyme 2 (ACE2)/Angiotensin (1–7)/Mas receptor (MASR), is another axis of brain RAS which counteracts the damaging effects of ACE/Ang II/AT1R axis on neurons in the brain. Thus, angiotensin II receptor blockers (ARBs) and activation of ACE2/Angiotensin (1–7)/MASR axis may serve as an exciting and novel method for neuroprotection in several neurodegenerative diseases. Here in this review article, we discuss the expression of RAS in the brain and highlight how altered RAS level may cause neurodegeneration. Understanding the pathophysiology of RAS and their links to neurodegeneration has enormous potential to identify potentially effective pharmacological tools to treat neurodegenerative diseases in the brain.  相似文献   

17.
The renin–angiotensin system (RAS) is involved in the cardiac and vascular remodeling associated with cardiovascular diseases. Angiotensin (Ang) II/AT1 axis is known to promote cardiac hypertrophy and collagen deposition. In contrast, Ang-(1–7)/Mas axis opposes Ang II effects in the heart producing anti-trophic and anti-fibrotic effects. Exercise training is known to induce cardiac remodeling with physiological hypertrophy without fibrosis. We hypothesize that cardiac remodeling induced by chronic exercise depends on the action of Ang-(1–7)/Mas axis. Thus, we evaluated the effect of exercise training on collagen deposition and RAS components in the heart of FVB/N mice lacking Mas receptor (Mas-KO). Male wild-type and Mas-KO mice were subjected to a moderate-intense swimming exercise training for 6 weeks. The left ventricle (LV) of the animals was sectioned and submitted to qRT-PCR and histological analysis. Circulating and tissue angiotensin peptides were measured by RIA. Sedentary Mas-KO presented a higher circulating Ang II/Ang-(1–7) ratio and an increased ACE2 expression in the LV. Physical training induced in Mas-KO and WT a similar cardiac hypertrophy accompanied by a pronounced increase in collagen I and III mRNA expression. Trained Mas-KO and trained WT presented increased Ang-(1–7) in the blood. However, only in trained-WT there was an increase in Ang-(1–7) in the LV. In summary, we showed that deletion of Mas in FVB/N mice produced an unbalance in RAS equilibrium increasing Ang II/AT1 arm and inducing deleterious cardiac effects as deposition of extracellular matrix proteins. These data indicate that Ang-(1–7)/Mas axis is an important counter-regulatory mechanism in physical training mediate cardiac adaptations.  相似文献   

18.
Zhuo JL  Li XC 《Peptides》2011,32(7):1551-1565
Although renin, the rate-limiting enzyme of the renin-angiotensin system (RAS), was first discovered by Robert Tigerstedt and Bergman more than a century ago, the research on the RAS still remains stronger than ever. The RAS, once considered to be an endocrine system, is now widely recognized as dual (circulating and local/tissue) or multiple hormonal systems (endocrine, paracrine and intracrine). In addition to the classical renin/angiotensin I-converting enzyme (ACE)/angiotensin II (Ang II)/Ang II receptor (AT1/AT2) axis, the prorenin/(Pro)renin receptor (PRR)/MAP kinase axis, the ACE2/Ang (1-7)/Mas receptor axis, and the Ang IV/AT4/insulin-regulated aminopeptidase (IRAP) axis have recently been discovered. Furthermore, the roles of the evolving RAS have been extended far beyond blood pressure control, aldosterone synthesis, and body fluid and electrolyte homeostasis. Indeed, novel actions and underlying signaling mechanisms for each member of the RAS in physiology and diseases are continuously uncovered. However, many challenges still remain in the RAS research field despite of more than one century's research effort. It is expected that the research on the expanded RAS will continue to play a prominent role in cardiovascular, renal and hypertension research. The purpose of this article is to review the progress recently being made in the RAS research, with special emphasis on the local RAS in the kidney and the newly discovered prorenin/PRR/MAP kinase axis, the ACE2/Ang (1-7)/Mas receptor axis, the Ang IV/AT4/IRAP axis, and intracrine/intracellular Ang II. The improved knowledge of the expanded RAS will help us better understand how the classical renin/ACE/Ang II/AT1 receptor axis, extracellular and/or intracellular origin, interacts with other novel RAS axes to regulate blood pressure and cardiovascular and kidney function in both physiological and diseased states.  相似文献   

19.
Brain renin‐angiotensin (Ang) system (RAS) is implicated in neuroinflammation, a major characteristic of aging process. Angiotensin (Ang) II, produced by angiotensin‐converting enzyme (ACE), activates immune system via angiotensin type 1 receptor (AT1), whereas Ang(1–7), generated by ACE2, binds with Mas receptor (MasR) to restrain excessive inflammatory response. Therefore, the present study aims to explore the relationship between RAS and neuroinflammation. We found that repeated lipopolysaccharide (LPS) treatment shifted the balance between ACE/Ang II/AT1 and ACE2/Ang(1–7)/MasR axis to the deleterious side and treatment with either MasR agonist, AVE0991 (AVE) or ACE2 activator, diminazene aceturate, exhibited strong neuroprotective actions. Mechanically, activation of ACE2/Ang(1–7)/MasR axis triggered the Forkhead box class O1 (FOXO1)‐autophagy pathway and induced superoxide dismutase (SOD) and catalase (CAT), the FOXO1‐targeted antioxidant enzymes. Meanwhile, knockdown of MasR or FOXO1 in BV2 cells, or using the selective FOXO1 inhibitor, AS1842856, in animals, suppressed FOXO1 translocation and compromised the autophagic process induced by MasR activation. We further used chloroquine (CQ) to block autophagy and showed that suppressing either FOXO1 or autophagy abrogated the anti‐inflammatory action of AVE. Likewise, Ang(1–7) also induced FOXO1 signaling and autophagic flux following LPS treatment in BV2 cells. Cotreatment with AS1842856 or CQ all led to autophagic inhibition and thereby abolished Ang(1–7)‐induced remission on NLRP3 inflammasome activation caused by LPS exposure, shifting the microglial polarization from M1 to M2 phenotype. Collectively, these results firstly illustrated the mechanism of ACE2/Ang(1–7)/MasR axis in neuroinflammation, strongly indicating the involvement of FOXO1‐mediated autophagy in the neuroimmune‐modulating effects triggered by MasR activation.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号