1
|
Brooker IA, Fisher JJ, Delforce SJ, Endacott SK, Lumbers ER, Sutherland JM, Pringle KG. Characterising the role of placental angiotensin-converting enzyme 2 (ACE2) during the onset of oxidative insult by hypoxia/reoxygenation: Implications for fetal growth restriction. Placenta 2025; 165:82-90. [PMID: 40222298 DOI: 10.1016/j.placenta.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/20/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION Fetal growth restriction (FGR) is a leading cause of infant morbidity and mortality. Approximately 60% of FGR cases result from placental dysfunction, often due to defective remodelling of the uterine vasculature and subsequent exposure to hypoxia/reoxygenation that induces oxidative stress. Angiotensin-converting enzyme 2 (ACE2) counteracts the ACE-driven axis of the renin-angiotensin system and is reduced in FGR placentae. We aimed to investigate the role of ACE2 in protecting against placental oxidative stress induced via a hypoxia/reoxygenation event. METHODS Term placental explants were exposed to normoxia (8% O2) for 6 hrs or were treated with media alone or recombinant human (rh)ACE2 and exposed to a hypoxia/reoxygenation insult (1 hr hypoxia (1% O2), 5 hrs normoxia). Oxidative stress markers, and ACE and ACE2 mRNA, protein, or activity were assessed. RESULTS ACE2 mRNA expression was increased with hypoxia/reoxygenation compared with normoxia (p=0.045). Hypoxia/reoxygenation significantly increased placental mRNA expression of the oxidative enzymes NOX4 and NOX5 compared with normoxia (p=0.021 and 0.023). NOX5 protein was not significantly different between normoxic controls and hypoxia/reoxygenation; however, rhACE2 significantly reduced NOX5 protein levels (p=0.015). Antioxidant activity of SOD decreased (p=0.028), while CAT increased with hypoxia/reoxygenation (p=0.010). Placental Nrf2 and NQO1 mRNA expression increased with rhACE2 compared with hypoxia/reoxygenation alone (p=0.038 and 0.013). CONCLUSION We have characterised the redox-sensitive changes that occur in the placenta due to defective placentation and hypoxia/reoxygenation and have shown for the first time the role of placental ACE2 in mitigating oxidative insult associated with hypoxia/reoxygenation.
Collapse
Affiliation(s)
- India A Brooker
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan (Awabakal Country), NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights (Awabakal Country), NSW, Australia
| | - Joshua J Fisher
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan (Awabakal Country), NSW, Australia; Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights (Awabakal Country), NSW, Australia
| | - Sarah J Delforce
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan (Awabakal Country), NSW, Australia
| | - Saije K Endacott
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan (Awabakal Country), NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights (Awabakal Country), NSW, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan (Awabakal Country), NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights (Awabakal Country), NSW, Australia
| | - Jessie M Sutherland
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan (Awabakal Country), NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights (Awabakal Country), NSW, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan (Awabakal Country), NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights (Awabakal Country), NSW, Australia.
| |
Collapse
|
2
|
Buonfiglio F, Böhm EW, Tang Q, Daiber A, Gericke A. Revisiting the renin-angiotensin-aldosterone system in the eye: mechanistic insights and pharmacological targets. Pharmacol Res 2025:107771. [PMID: 40348100 DOI: 10.1016/j.phrs.2025.107771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 04/22/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
The renin-angiotensin-aldosterone system (RAAS) plays a fundamental role in regulating blood pressure and fluid homeostasis through key effectors such as angiotensin II and aldosterone. These agents and their receptors have become crucial molecular targets in several cardiovascular and renal diseases. Over the past few decades, a growing body of evidence has revealed the presence of RAAS components in ocular structures, suggesting a tissue-specific RAAS within the eye. Building on this knowledge, studies have indicated that the ocular RAAS plays a significant role in the pathogenesis of various eye diseases. An impaired and overactivated RAAS contributes to the development of severe and widespread disorders affecting both the anterior and posterior segments of the eye. In this context, the current work aims to delve into the pivotal molecular pathways involving the RAAS, with an in-depth exploration of the ocular pathophysiology. It focuses on the relationship between overactivation of the RAAS and oxidative stress, as well as the exacerbation of neovascularization and inflammatory processes. The objective is to provide an updated and comprehensive understanding of the role of the RAAS in ophthalmological diseases, highlighting the therapeutic potential of RAAS modulators and discussing the controversies and challenges in this area of research.
Collapse
Affiliation(s)
- Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| | - Elsa Wilma Böhm
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| | - Qi Tang
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| | - Andreas Daiber
- Department of Cardiology I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany.
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| |
Collapse
|
3
|
Buonfiglio F, Pfeiffer N, Gericke A. Glaucoma and the ocular renin-angiotensin-aldosterone system: Update on molecular signalling and treatment perspectives. Cell Signal 2024; 122:111343. [PMID: 39127136 DOI: 10.1016/j.cellsig.2024.111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
Glaucoma, a leading cause of blindness worldwide, encompasses a group of pathological conditions affecting the optic nerve and is characterized by progressive retinal ganglion cell loss, cupping of the optic nerve head, and distinct visual field defects. While elevated intraocular pressure (IOP) is the main risk factor for glaucoma, many patients do not have elevated IOP. Consequently, other risk factors, such as ocular blood flow abnormalities and immunological factors, have been implicated in its pathophysiology. Traditional therapeutic strategies primarily aim to reduce IOP, but there is growing interest in developing novel treatment approaches to improve disease management and reduce the high rates of severe visual impairment. In this context, targeting the ocular renin-angiotensin-aldosterone system (RAAS) has been found as a potential curative strategy. The RAAS contributes to glaucoma development through key effectors such as prorenin, angiotensin II, and aldosterone. Recent evidence has highlighted the potential of using RAAS modulators to combat glaucoma, yielding encouraging results. Our study aims to explore the molecular pathways linking the ocular RAAS and glaucoma, summarizing recent advances that elucidate the role of the RAAS in triggering oxidative stress, inflammation, and remodelling in the pathogenesis of glaucoma. Additionally, we will present emerging therapeutic approaches that utilize RAAS modulators and antioxidants to slow the progression of glaucoma.
Collapse
Affiliation(s)
- Francesco Buonfiglio
- Departments of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| | - Norbert Pfeiffer
- Departments of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| | - Adrian Gericke
- Departments of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| |
Collapse
|
4
|
Chen H, Peng J, Wang T, Wen J, Chen S, Huang Y, Zhang Y. Counter-regulatory renin-angiotensin system in hypertension: Review and update in the era of COVID-19 pandemic. Biochem Pharmacol 2023; 208:115370. [PMID: 36481346 PMCID: PMC9721294 DOI: 10.1016/j.bcp.2022.115370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is the major cause of mortality and disability, with hypertension being the most prevalent risk factor. Excessive activation of the renin-angiotensin system (RAS) under pathological conditions, leading to vascular remodeling and inflammation, is closely related to cardiovascular dysfunction. The counter-regulatory axis of the RAS consists of angiotensin-converting enzyme 2 (ACE2), angiotensin (1-7), angiotensin (1-9), alamandine, proto-oncogene Mas receptor, angiotensin II type-2 receptor and Mas-related G protein-coupled receptor member D. Each of these components has been shown to counteract the effects of the overactivated RAS. In this review, we summarize the latest insights into the complexity and interplay of the counter-regulatory RAS axis in hypertension, highlight the pathophysiological functions of ACE2, a multifunctional molecule linking hypertension and COVID-19, and discuss the function and therapeutic potential of targeting this counter-regulatory RAS axis to prevent and treat hypertension in the context of the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Hongyin Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518000, Guangdong, China
| | - Jiangyun Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Jielu Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China,Corresponding authors
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518000, Guangdong, China,Corresponding authors
| |
Collapse
|
5
|
Chen XY, Lin C, Liu GY, Pei C, Xu GQ, Gao L, Wang SZ, Pan YX. ACE2 gene combined with exercise training attenuates central AngII/AT1 axis function and oxidative stress in a prehypertensive rat model. J Appl Physiol (1985) 2022; 132:1460-1467. [PMID: 35546127 PMCID: PMC11918447 DOI: 10.1152/japplphysiol.00459.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 04/13/2022] [Accepted: 04/29/2022] [Indexed: 11/22/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) or exercise training (ExT) is beneficial to hypertension, but their combined effects remain unknown. In this study, lentivirus containing enhanced green fluorescent protein (eGFP) and ACE2 were microinjected into the paraventricular nucleus (PVN) of young male spontaneous hypertensive rats (SHRs), and SHRs were assigned into five groups: sedentary (SHR), SHR-ExT, SHR-eGFP, ACE2 gene (SHR-ACE2), and ACE2 gene combined with ExT (SHR-ACE2-ExT). Wistar-Kyoto (WKY) rats were used as a control. ACE2 gene or ExT significantly delayed the elevation of blood pressure, and the combined effect prevented the development and progression of prehypertension. Either ACE2 overexpression or ExT improved arterial baroreflex sensitivity (BRS), whereas the combined effect normalized BRS in SHR. Compared with SHR, SHR-ACE2 and SHR-ExT displayed a significantly higher level of ACE2 protein but had lower plasma norepinephrine (NE) and angiotensin II (AngII) as well as angiotensin II type 1 receptor (AT1) protein expression in the PVN. SHR-ACE2-ExT showed the largest decrease in AngII and AT1 protein expression. Reactive oxygen species (ROS) level and NADPH oxidase (NOX2 and NOX4) protein expression in PVN were also decreased in SHR-ACE2-ExT group than in SHR-ACE2 and SHR-ExT groups. It was concluded that the combined effect has effectively prevented prehypertension progression and baroreflex dysfunction in SHR, which is associated with the reduction in AngII/AT1 axis function and oxidative stress in the PVN.NEW & NOTEWORTHY Angiotensin-converting enzyme 2 (ACE2) gene in combination with exercise training (ExT) delayed the progression of hypertension via normalizing the blunted baroreflex sensitivity (BRS) and inhibiting sympathetic nerve activity (SNA). Its underlying mechanism may be related to the inhibition of AngII/AT1 axis function and central oxidative stress in the paraventricular nucleus (PVN) of prehypertensive rats.
Collapse
Affiliation(s)
- Xiu-Yun Chen
- Department of Rehabilitation Medicine, Health School of Fujian Medical University, Fuzhou, China
| | - Cheng Lin
- Department of Rehabilitation Medicine, Health School of Fujian Medical University, Fuzhou, China
| | - Guo-Ying Liu
- Department of Rehabilitation Medicine, Health School of Fujian Medical University, Fuzhou, China
| | - Chun Pei
- Department of Rehabilitation Medicine, Health School of Fujian Medical University, Fuzhou, China
| | - Gui-Qing Xu
- Department of Rehabilitation Medicine, Health School of Fujian Medical University, Fuzhou, China
| | - Lie Gao
- Department of Cellular and Integrative, Physiology of University of Nebraska Medical Center, Omaha, Nebraska
| | - Shi-Zhong Wang
- Department of Rehabilitation Medicine, Health School of Fujian Medical University, Fuzhou, China
| | - Yan-Xia Pan
- Department of Rehabilitation Medicine, Health School of Fujian Medical University, Fuzhou, China
| |
Collapse
|
6
|
Circulating Soluble ACE2 Plays an Independent Role to Protect against Vascular Damage in Diabetic Mice. Antioxidants (Basel) 2022; 11:antiox11050987. [PMID: 35624851 PMCID: PMC9138042 DOI: 10.3390/antiox11050987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 12/05/2022] Open
Abstract
Circulating levels of soluble ACE2 are increased by diabetes. Although this increase is associated with the presence and severity of cardiovascular disease, the specific role of soluble ACE2 in atherogenesis is unclear. Previous studies suggested that, like circulating ACE, soluble ACE2 plays a limited role in vascular homeostasis. To challenge this hypothesis, we aimed to selectively increase circulating ACE2 and measure its effects on angiotensin II dependent atherogenesis. Firstly, in Ace2/ApoE DKO mice, restoration of circulating ACE2 with recombinant murine soluble (rmACE219-613; 1 mg/kg/alternate day IP) reduced plaque accumulation in the aortic arch, suggesting that the phenotype may be driven as much by loss of soluble ACE2 as the reduction in local ACE2. Secondly, in diabetic ApoE KO mice, where activation of the renin angiotensin system drives accelerated atherosclerosis, rmACE219-613 also reduced plaque accumulation in the aorta after 6 weeks. Thirdly, to ensure consistent long-term delivery of soluble ACE2, an intramuscular injection was used to deliver a DNA minicircle encoding ACE219-613. This strategy efficiently increased circulating soluble ACE2 and reduced atherogenesis and albuminuria in diabetic ApoE KO mice followed for 10 weeks. We propose that soluble ACE2 has independent vasculoprotective effects. Future strategies that increase soluble ACE2 may reduce accelerated atherosclerosis in diabetes and other states in which the renin angiotensin system is upregulated.
Collapse
|
7
|
Shawky NM, Dalmasso C, Ojeda NB, Zuchowski Y, Stachenfeld N, Alexander BT, Reckelhoff JF. Consequences of hyperandrogenemia during pregnancy in female offspring: attenuated response to angiotensin II. J Hypertens 2022; 40:712-722. [PMID: 34980865 PMCID: PMC8897268 DOI: 10.1097/hjh.0000000000003067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is characterized by reproductive and metabolic dysfunction, and elevated blood pressure (BP). The cardiometabolic consequences of maternal hyperandrogenemia on offspring, either as adults or with aging, have not been well studied. We previously found that male offspring of hyperandrogenemic female (HAF) rats, a model of PCOS, are normotensive but have an exaggerated pressor response to angiotensin (Ang) II. METHOD In this study, the hypothesis was tested that adult and aging female offspring of HAF rats develop a metabolic and hypertensive phenotype. Control and HAF rats were implanted prepubertally with placebo or dihydrotestosterone pellets, which continued throughout pregnancy and lactation. RESULTS Female offspring of HAF dams had lower birth weight than female control offspring. Although female HAF offspring (aged 16-24 weeks) had no differences in intrarenal Ang II, plasma lipids or proteinuria, they did have lower intrarenal Ang (1-7) and lower nitrate/nitrite excretion than controls. Adult HAF offspring had similar baseline BP as controls, but had an attenuated pressor response to Ang II. With aging (16-20 months), female HAF offspring remained normotensive with an attenuated pressor response to Ang II and high salt diet but more proteinuria and higher intrarenal Ang(1-7) than controls. CONCLUSION Taken together, these data suggest that female HAF offspring are protected from developing hypertension, but may be at risk for renal injury with aging. Future studies are necessary to determine whether adult and postmenopausal offspring of PCOS women are at increased risk for cardiovascular dysfunction.Graphical abstract:http://links.lww.com/HJH/B820.
Collapse
Affiliation(s)
- Noha M. Shawky
- Departments of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Carolina Dalmasso
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington
| | - Norma B. Ojeda
- Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yvonne Zuchowski
- Departments of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Nina Stachenfeld
- The John Pierce Laboratory, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT
| | - Barbara T. Alexander
- Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jane F. Reckelhoff
- Departments of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
8
|
Gallo G, Calvez V, Savoia C. Hypertension and COVID-19: Current Evidence and Perspectives. High Blood Press Cardiovasc Prev 2022; 29:115-123. [PMID: 35184271 PMCID: PMC8858218 DOI: 10.1007/s40292-022-00506-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) outbreak, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), represents a real challenge for health-care systems worldwide. Male sex, older age and the coexistence of chronic comorbidities have been described as the most relevant conditions associated with a worse prognosis. Early reports suggested that hypertension might represent a risk factor for susceptibility to SARS-CoV-2 infection, a more severe course of COVID-19 and increased COVID-19-related deaths. Nevertheless, the independent role of hypertension remains under debate, since hypertension is often associated with the older age and other cardiovascular (CV) risk factors in the general population, which may also contribute to the SARS-Cov-2 infection and COVID-19. Moreover, the role of antihypertensive drugs, primarily angiotensin-converting inhibitors (ACEIs) and ARBs (angiotensin receptor blockers) in COVID-19 development and outcome appears controversial. Indeed, preclinical studies using these classes of drugs have suggested a potential upregulation of angiotensin-converting-enzyme 2 (ACE2) which is the key binding receptor promoting cell entry of SARS-CoV-2 in the organism. Renin–angiotensin system (RAS) blockers may potentially upregulate ACE2, hence, it has been initially hypothesized that these agents might contribute to a higher risk of SARS-CoV-2 infection and progressive course of COVID-19. However, several clinical reports do not support a detrimental role of RAS blockers in COVID-19, and an intense debate about the withdrawal or maintenance of chronic therapy with ACEi/ARB has been developed. In this review we will discuss the available evidence on the role of hypertension and antihypertensive drugs on SARS-CoV-2 infection and COVID-19 development.
Collapse
Affiliation(s)
- Giovanna Gallo
- Cardiology Unit, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Valentin Calvez
- Cardiology Unit, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Carmine Savoia
- Cardiology Unit, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
9
|
Liu LP, Zhang XL, Li J. New perspectives on angiotensin-converting enzyme 2 and its related diseases. World J Diabetes 2021; 12:839-854. [PMID: 34168732 PMCID: PMC8192247 DOI: 10.4239/wjd.v12.i6.839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/30/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Since the worldwide outbreak of coronavirus disease 2019, angiotensin-converting enzyme 2 (ACE2) has received widespread attention as the cell receptor of the severe acute respiratory syndrome coronavirus 2 virus. At the same time, as a key enzyme in the renin-angiotensin-system, ACE2 is considered to be an endogenous negative regulator of vasoconstriction, proliferation, fibrosis, and proinflammation caused by the ACE-angiotensin II-angiotensin type 1 receptor axis. ACE2 is now implicated as being closely connected to diabetes, cardiovascular, kidney, and lung diseases, and so on. This review covers the available information on the host factors regulating ACE2 and discusses its role in a variety of pathophysiological conditions in animal models and humans.
Collapse
Affiliation(s)
- Li-Ping Liu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Xiao-Li Zhang
- TheFifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg 68135, Baden-Württemberg, Germany
| | - Jian Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, Hunan Province, China
| |
Collapse
|
10
|
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) associates with a considerable high rate of mortality and represents currently the most important concern in global health. The risk of more severe clinical manifestation of COVID-19 is higher in males and steeply raised with age but also increased by the presence of chronic comorbidities. Among the latter, early reports suggested that arterial hypertension associates with higher susceptibility to SARS-CoV-2 infection, more severe course and increased COVID-19-related deaths. Furthermore, experimental studies suggested that key pathophysiological hypertension mechanisms, such as activation of the renin-angiotensin system (RAS), may play a role in COVID-19. In fact, ACE2 (angiotensin-converting-enzyme 2) is the pivotal receptor for SARS-CoV-2 to enter host cells and provides thus a link between COVID-19 and RAS. It was thus anticipated that drugs modulating the RAS including an upregulation of ACE2 may increase the risk for infection with SARS-CoV-2 and poorer outcomes in COVID-19. Since the use of RAS-blockers, ACE inhibitors or angiotensin receptor blockers, represents the backbone of recommended antihypertensive therapy and intense debate about their use in the COVID-19 pandemic has developed. Currently, a direct role of hypertension, independent of age and other comorbidities, as a risk factor for the SARS-COV-2 infection and COVID-19 outcome, particularly death, has not been established. Similarly, both current experimental and clinical studies do not support an unfavorable effect of RAS-blockers or other classes of first line blood pressure lowering drugs in COVID-19. Here, we review available data on the role of hypertension and its management on COVID-19. Conversely, some aspects as to how the COVID-19 affects hypertension management and impacts on future developments are also briefly discussed. COVID-19 has and continues to proof the critical importance of hypertension research to address questions that are important for global health.
Collapse
Affiliation(s)
- Carmine Savoia
- Clinical and Molecular Medicine Department, Division of Cardiology, Sant' Andrea Hospital, Sapienza University of Rome, Rome, Italy (C.S., M.V.)
| | - Massimo Volpe
- Clinical and Molecular Medicine Department, Division of Cardiology, Sant' Andrea Hospital, Sapienza University of Rome, Rome, Italy (C.S., M.V.)
| | - Reinhold Kreutz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institut für Klinische Pharmakologie und Toxikologie (R.K.)
| |
Collapse
|
11
|
ACE2 as therapeutic agent. Clin Sci (Lond) 2021; 134:2581-2595. [PMID: 33063820 DOI: 10.1042/cs20200570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
The angiotensin-converting enzyme 2 (ACE2) has emerged as a critical regulator of the renin-angiotensin system (RAS), which plays important roles in cardiovascular homeostasis by regulating vascular tone, fluid and electrolyte balance. ACE2 functions as a carboxymonopeptidase hydrolyzing the cleavage of a single C-terminal residue from Angiotensin-II (Ang-II), the key peptide hormone of RAS, to form Angiotensin-(1-7) (Ang-(1-7)), which binds to the G-protein-coupled Mas receptor and activates signaling pathways that counteract the pathways activated by Ang-II. ACE2 is expressed in a variety of tissues and overwhelming evidence substantiates the beneficial effects of enhancing ACE2/Ang-(1-7)/Mas axis under many pathological conditions in these tissues in experimental models. This review will provide a succinct overview on current strategies to enhance ACE2 as therapeutic agent, and discuss limitations and future challenges. ACE2 also has other functions, such as acting as a co-factor for amino acid transport and being exploited by the severe acute respiratory syndrome coronaviruses (SARS-CoVs) as cellular entry receptor, the implications of these functions in development of ACE2-based therapeutics will also be discussed.
Collapse
|
12
|
The Tissue Renin-Angiotensin System and Its Role in the Pathogenesis of Major Human Diseases: Quo Vadis? Cells 2021; 10:cells10030650. [PMID: 33804069 PMCID: PMC7999456 DOI: 10.3390/cells10030650] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 01/18/2023] Open
Abstract
Evidence has arisen in recent years suggesting that a tissue renin-angiotensin system (tRAS) is involved in the progression of various human diseases. This system contains two regulatory pathways: a pathological pro-inflammatory pathway containing the Angiotensin Converting Enzyme (ACE)/Angiotensin II (AngII)/Angiotensin II receptor type 1 (AGTR1) axis and a protective anti-inflammatory pathway involving the Angiotensin II receptor type 2 (AGTR2)/ACE2/Ang1–7/MasReceptor axis. Numerous studies reported the positive effects of pathologic tRAS pathway inhibition and protective tRAS pathway stimulation on the treatment of cardiovascular, inflammatory, and autoimmune disease and the progression of neuropathic pain. Cell senescence and aging are known to be related to RAS pathways. Further, this system directly interacts with SARS-CoV 2 and seems to be an important target of interest in the COVID-19 pandemic. This review focuses on the involvement of tRAS in the progression of the mentioned diseases from an interdisciplinary clinical perspective and highlights therapeutic strategies that might be of major clinical importance in the future.
Collapse
|
13
|
Kuriakose J, Montezano A, Touyz R. ACE2/Ang-(1-7)/Mas1 axis and the vascular system: vasoprotection to COVID-19-associated vascular disease. Clin Sci (Lond) 2021; 135:387-407. [PMID: 33511992 PMCID: PMC7846970 DOI: 10.1042/cs20200480] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
The two axes of the renin-angiotensin system include the classical ACE/Ang II/AT1 axis and the counter-regulatory ACE2/Ang-(1-7)/Mas1 axis. ACE2 is a multifunctional monocarboxypeptidase responsible for generating Ang-(1-7) from Ang II. ACE2 is important in the vascular system where it is found in arterial and venous endothelial cells and arterial smooth muscle cells in many vascular beds. Among the best characterized functions of ACE2 is its role in regulating vascular tone. ACE2 through its effector peptide Ang-(1-7) and receptor Mas1 induces vasodilation and attenuates Ang II-induced vasoconstriction. In endothelial cells activation of the ACE2/Ang-(1-7)/Mas1 axis increases production of the vasodilator's nitric oxide and prostacyclin's and in vascular smooth muscle cells it inhibits pro-contractile and pro-inflammatory signaling. Endothelial ACE2 is cleaved by proteases, shed into the circulation and measured as soluble ACE2. Plasma ACE2 activity is increased in cardiovascular disease and may have prognostic significance in disease severity. In addition to its enzymatic function, ACE2 is the receptor for severe acute respiratory syndrome (SARS)-coronavirus (CoV) and SARS-Cov-2, which cause SARS and coronavirus disease-19 (COVID-19) respectively. ACE-2 is thus a double-edged sword: it promotes cardiovascular health while also facilitating the devastations caused by coronaviruses. COVID-19 is associated with cardiovascular disease as a risk factor and as a complication. Mechanisms linking COVID-19 and cardiovascular disease are unclear, but vascular ACE2 may be important. This review focuses on the vascular biology and (patho)physiology of ACE2 in cardiovascular health and disease and briefly discusses the role of vascular ACE2 as a potential mediator of vascular injury in COVID-19.
Collapse
Affiliation(s)
- Jithin Kuriakose
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Augusto C. Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Rhian M. Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| |
Collapse
|
14
|
Parit R, Jayavel S. Association of ACE inhibitors and angiotensin type II blockers with ACE2 overexpression in COVID-19 comorbidities: A pathway-based analytical study. Eur J Pharmacol 2021; 896:173899. [PMID: 33508281 PMCID: PMC7839513 DOI: 10.1016/j.ejphar.2021.173899] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023]
Abstract
Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) outbreak is a major public health concern, which has accounted for >1.7 million deaths across the world. A surge in the case fatality ratio as compared with the infection ratio has been observed in most of the countries. The novel Coronavirus SARS-CoV-2 shares the most common sequence with SARS-CoV, but it has a higher rate of transmission. The SARS-CoV-2 pathogenesis is initiated by the binding of viral spike protein with the target receptor Angiotensin-Converting Enzyme 2 (ACE2) facilitating virus internalization within host cells. SARS-CoV-2 mainly causes alveolar damage ranging from mild to severe clinical respiratory manifestations. Most of the cases have revealed the association of Coronavirus disease with patients having earlier comorbidities like Hypertension, Diabetes mellitus, and Cerebrovascular diseases. Pharmacological investigation of the SARS-Cov-2 patients has revealed the frequent use of drugs belongs to Angiotensin-converting enzyme inhibitors (ACEi) and/or Angiotensin II type I receptor blockers (ARBs). Interestingly, a significant increase in ACE2 expression was noticed in patients routinely treated with the above group of drugs were also reported. To date, the association of ACEi and/or ARBs with the up-regulation of ACE2 expression has not been defined distinctively. The proposed review will focus on the pathways which are responsible for the upregulation of ACE2 and its impact on gravity of SARS-CoV-2 disease.
Collapse
Affiliation(s)
- Rahul Parit
- Department of Biotechnology (DDE), Madurai Kamaraj University, Madurai, 625021, Tamilnadu, India
| | - Sridhar Jayavel
- Department of Biotechnology (DDE), Madurai Kamaraj University, Madurai, 625021, Tamilnadu, India.
| |
Collapse
|
15
|
Effects of L-/N-Type Calcium Channel Blockers on Angiotensin II-Renin Feedback in Hypertensive Patients. Int J Hypertens 2021; 2020:6653851. [PMID: 33489354 PMCID: PMC7803135 DOI: 10.1155/2020/6653851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/10/2020] [Indexed: 11/17/2022] Open
Abstract
Objectives Cilnidipine, an L-/N-type calcium channel blocker (CCB), has unique organ-protective properties due to suppression of hyperactivity in the sympathetic nervous system and renin-angiotensin system (RAS). In this study, we hypothesized that cilnidipine might exert a renoprotective effect by suppressing the RAS. Methods A total of 25 hypertensive patients receiving a RAS inhibitor were randomly assigned to a cilnidipine (n = 12) or amlodipine (n = 13) group. The effects of cilnidipine on proteinuria and angiotensin II-renin feedback were assessed. Results After 6 months of treatment, both systolic and diastolic blood pressures were significantly reduced to a similar extent in both groups. The urine albumin-to-creatinine ratio was significantly lower in the cilnidipine group (p < 0.05) than in the amlodipine group. Amlodipine increased plasma angiotensin I and angiotensin II levels (p < 0.05), whereas cilnidipine did not. Interestingly, the cilnidipine group had a higher ratio of angiotensin-(1-7) (Ang-(1-7)) to angiotensin II in plasma than the amlodipine group (p < 0.05). Conclusions The L-/N-type CCB cilnidipine, but not amlodipine, decreased urinary albumin excretion in hypertensive patients. Cilnidipine also increased the ratio of Ang-(1-7) to angiotensin II in plasma, which might be one factor underlying its beneficial effects.
Collapse
|
16
|
Zhang H, Viveiros A, Nikhanj A, Nguyen Q, Wang K, Wang W, Freed DH, Mullen JC, MacArthur R, Kim DH, Tymchak W, Sergi CM, Kassiri Z, Wang S, Oudit GY. The Human Explanted Heart Program: A translational bridge for cardiovascular medicine. Biochim Biophys Acta Mol Basis Dis 2021; 1867:165995. [PMID: 33141063 PMCID: PMC7581399 DOI: 10.1016/j.bbadis.2020.165995] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022]
Abstract
The progression of cardiovascular research is often impeded by the lack of reliable disease models that fully recapitulate the pathogenesis in humans. These limitations apply to both in vitro models such as cell-based cultures and in vivo animal models which invariably are limited to simulate the complexity of cardiovascular disease in humans. Implementing human heart tissue in cardiovascular research complements our research strategy using preclinical models. We established the Human Explanted Heart Program (HELP) which integrates clinical, tissue and molecular phenotyping thereby providing a comprehensive evaluation into human heart disease. Our collection and storage of biospecimens allow them to retain key pathogenic findings while providing novel insights into human heart failure. The use of human non-failing control explanted hearts provides a valuable comparison group for the diseased explanted hearts. Using HELP we have been able to create a tissue repository which have been used for genetic, molecular, cellular, and histological studies. This review describes the process of collection and use of explanted human heart specimens encompassing a spectrum of pediatric and adult heart diseases, while highlighting the role of these invaluable specimens in translational research. Furthermore, we highlight the efficient procurement and bio-preservation approaches ensuring analytical quality of heart specimens acquired in the context of heart donation and transplantation.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Anissa Viveiros
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anish Nikhanj
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Quynh Nguyen
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kaiming Wang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Wei Wang
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Darren H Freed
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John C Mullen
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Roderick MacArthur
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel H Kim
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Wayne Tymchak
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Consolato M Sergi
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Anatomical Pathology, Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shaohua Wang
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
17
|
Silva-Aguiar RP, Peruchetti DB, Rocco PRM, Schmaier AH, E Silva PMR, Martins MA, Carvalho VF, Pinheiro AAS, Caruso-Neves C. Role of the renin-angiotensin system in the development of severe COVID-19 in hypertensive patients. Am J Physiol Lung Cell Mol Physiol 2020; 319:L596-L602. [PMID: 32783619 PMCID: PMC7516382 DOI: 10.1152/ajplung.00286.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 02/08/2023] Open
Abstract
A new form of severe acute respiratory syndrome (SARS) caused by SARS-coronavirus 2 (CoV-2), called COVID-19, has become a global threat in 2020. The mortality rate from COVID-19 is high in hypertensive patients, making this association especially dangerous. There appears to be a consensus, despite the lack of experimental data, that angiotensin II (ANG II) is linked to the pathogenesis of COVID-19. This process may occur due to acquired deficiency of angiotensin-converting enzyme 2 (ACE2), resulting in reduced degradation of ANG II. Furthermore, ANG II has a critical role in the genesis and worsening of hypertension. In this context, the idea that there is a surge in the level of ANG II with COVID-19 infection, causing multiple organ injuries in hypertensive patients becomes attractive. However, the role of other components of the renin angiotensin system (RAS) in this scenario requires elucidation. The identification of other RAS components in COVID-19 hypertension may provide both diagnostic and therapeutic benefits. Here, we summarize the pathophysiologic contributions of different components of RAS in hypertension and their possible correlation with poor outcome observed in hypertensive patients with COVID-19.
Collapse
Affiliation(s)
| | - Diogo Barros Peruchetti
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Alvin H Schmaier
- Case Western Reserve University, Cleveland, Ohio
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Patrícia Machado Rodrigues E Silva
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Marco Aurélio Martins
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Vinícius Frias Carvalho
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ana Acacia Sá Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Li X, Liu Y, Song J, Zhong J. Increased plasma ACE2 concentration does not mean increased risk of SARS-CoV-2 infection and increased fatality rate of COVID-19. Acta Pharm Sin B 2020; 10:2010-2014. [PMID: 32923317 PMCID: PMC7476560 DOI: 10.1016/j.apsb.2020.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Xueting Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ying Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Juanjuan Song
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Corresponding author. Tel.: +86 10 89138377; fax: +86 10 85231937.
| |
Collapse
|
19
|
Ma A, Gao L, Wafi AM, Yu L, Rudebush T, Zhou W, Zucker IH. Overexpression of Central ACE2 (Angiotensin-Converting Enzyme 2) Attenuates the Pressor Response to Chronic Central Infusion of Ang II (Angiotensin II): A Potential Role for Nrf2 (Nuclear Factor [Erythroid-Derived 2]-Like 2). Hypertension 2020; 76:1514-1525. [PMID: 32895018 DOI: 10.1161/hypertensionaha.120.15681] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We investigated the mechanism by which ACE2 (angiotensin-converting enzyme 2) overexpression alters neurohumoral outflow and central oxidative stress. Nrf2 (nuclear factor [erythroid-derived 2]-like 2) is a master antioxidant transcription factor that regulates cytoprotective and antioxidant genes. We hypothesized that upregulation of central ACE2 inhibits the pressor response to Ang II (angiotensin II) by reducing reactive oxygen species through a Nrf2/antioxidant enzyme-mediated mechanism in the rostral ventrolateral medulla. Synapsin human Angiotensin Converting Enzyme 2 positive (SynhACE2+/+) mice and their littermate controls synhACE2-/- were used to evaluate the consequence of intracerebroventricular infusion of Ang II. In control mice, Ang II infusion evoked a significant increase in blood pressure and norepinephrine excretion, along with polydipsia and polyuria. The pressor effect of central Ang II was completely blocked in synhACE2+/+ mice. Polydipsia, norepinephrine excretion, and markers of oxidative stress in response to central Ang II were also reduced in synhACE2+/+ mice. The MasR (Mas receptor) agonist Ang 1-7 and blocker A779 had no effects on blood pressure. synhACE2+/+ mice showed enhanced expression of Nrf2 in the rostral ventrolateral medulla which was blunted following Ang II infusion. Ang II evoked nuclear translocation of Nrf2 in cultured Neuro 2A (N2A) cells. In synhACE2-/- mice, the central Ang II pressor response was attenuated by simultaneous intracerebroventricular infusion of the Nrf2 activator sulforaphane; blood pressure was enhanced by knockdown of Nrf2 in the rostral ventrolateral medulla in Nrf2 floxed (Nrf2f/f) mice. These data suggest that the hypertensive effects of intracerebroventricular Ang II are attenuated by selective overexpression of brain synhACE2 and may be mediated by Nrf2-upregulated antioxidant enzymes in the rostral ventrolateral medulla.
Collapse
Affiliation(s)
- Anyun Ma
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Lie Gao
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Ahmed M Wafi
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Li Yu
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Tara Rudebush
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Wenxian Zhou
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Irving H Zucker
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| |
Collapse
|
20
|
Wang K, Basu R, Poglitsch M, Bakal JA, Oudit GY. Elevated Angiotensin 1-7/Angiotensin II Ratio Predicts Favorable Outcomes in Patients With Heart Failure. Circ Heart Fail 2020; 13:e006939. [PMID: 32580658 DOI: 10.1161/circheartfailure.120.006939] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND ACE2 (angiotensin-converting enzyme 2) and Ang 1-7 (angiotensin 1-7) are endogenous negative regulators of the renin-angiotensin system exerting cardioprotective effects in models of heart failure. Recombinant human ACE2 markedly increased plasma Ang 1-7 and lowered Ang II levels in phase II clinical trials. We hypothesize that the dynamic state of this renin-angiotensin system protective arm could influence long-term outcomes in patients with heart failure. METHODS One hundred ten patients with heart failure were prospectively enrolled from our outpatient clinic and the emergency department. Comprehensive circulating and equilibrium levels of plasma angiotensin peptide profiles were assessed using novel liquid chromatography-mass spectrometry/mass spectroscopy techniques. Plasma aldosterone, B-type natriuretic peptide, active renin concentration, and clinical profiles were captured at baseline. During a median follow-up of 5.1 years (interquartile range, 4.7-5.7 years), composite clinical outcomes were assessed using all-cause in-patient hospitalizations and mortality. RESULTS Circulating and equilibrium angiotensin peptide levels strongly correlated in our patient cohort. Adjusting for covariates, elevated equilibrium (hazard ratio, 0.38 [95% CI, 0.18-0.81] P=0.012), and circulating (hazard ratio, 0.38 [95% CI, 0.18-0.80] P=0.011) Ang 1-7/Ang II ratios were associated with improved survival. Lower hospitalization duration was also associated with elevated equilibrium (P<0.001) and circulating (P=0.023) Ang 1-7/Ang II ratios. Importantly, individual Ang 1-7 and Ang II peptide levels failed to predict all-cause mortality or hospitalization duration in our patient cohort. CONCLUSIONS We extensively profiled plasma angiotensin peptides in patients with heart failure and identified elevated Ang 1-7/Ang II ratio, as an independent and incremental predictor of beneficial outcomes, higher survival rate, and decreased hospitalization duration. These findings provide important clinical evidence supporting strategies aiming to promote the beneficial Ang 1-7/Mas axis concurrent with renin-angiotensin system blockade therapies inhibiting the detrimental Ang II/AT1 receptor axis.
Collapse
Affiliation(s)
- Kaiming Wang
- Division of Cardiology, Department of Medicine (K.W., R.B., G.Y.O.), University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute (K.W., R.B., G.Y.O.), University of Alberta, Edmonton, Canada
| | - Ratnadeep Basu
- Division of Cardiology, Department of Medicine (K.W., R.B., G.Y.O.), University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute (K.W., R.B., G.Y.O.), University of Alberta, Edmonton, Canada
| | | | - Jeffrey A Bakal
- Alberta Strategy for Patient Oriented Research (SPOR) Unit (J.A.B.), University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine (K.W., R.B., G.Y.O.), University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute (K.W., R.B., G.Y.O.), University of Alberta, Edmonton, Canada
| |
Collapse
|
21
|
Liao W, Wu J. The ACE2/Ang (1-7)/MasR axis as an emerging target for antihypertensive peptides. Crit Rev Food Sci Nutr 2020; 61:2572-2586. [PMID: 32551837 DOI: 10.1080/10408398.2020.1781049] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Food protein-derived bioactive peptides, particularly antihypertensive peptides, are important constituents of functional foods or nutraceuticals. Most antihypertensive are identified as the inhibitors of angiotensin converting enzyme (ACE), a key enzyme responsible for the generation of angiotensin II (Ang II), which is a vasoconstricting peptide. Hence, ACE has long been used as a universal target to identify antihypertensive peptides. Angiotensin converting enzyme 2 (ACE2), is a homolog of ACE but uses Ang II as its key substrate to produce angiotensin (1-7), exerting vasodilatory activity via the mas receptor (MasR). Therefore, ACE2 functions in the opposite way as ACE and is an emerging novel target for cardiovascular therapy. The potential of food protein-derived bioactive peptides in targeting ACE2 has been rarely explored. While, recently we found that IRW, an egg white ovotransferrin-derived antihypertensive peptide, reduced blood pressure in spontaneously hypertensive rats via the ACE2/Ang (1-7)/MasR axis, indicating a new mechanism of food protein-derived bioactive peptides in reducing blood pressure. The objectives of this review are to summarize the functions of the ACE2/Ang (1-7)/MasR axis and to examine its potential roles in the actions of food protein-derived antihypertensive peptides. The interaction between antihypertensive peptides and the ACE2/Ang (1-7)/MasR axis will also be discussed.
Collapse
Affiliation(s)
- Wang Liao
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jianping Wu
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
22
|
Gargaglioni LH, Marques DA. Let's talk about sex in the context of COVID-19. J Appl Physiol (1985) 2020; 128:1533-1538. [PMID: 32437244 PMCID: PMC7303729 DOI: 10.1152/japplphysiol.00335.2020] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022] Open
Abstract
In recent months, the coronavirus disease 2019 (COVID-19) pandemic has sent many countries into crisis. Studies have shown that this virus causes worse outcomes and a higher mortality in men than in women. It has been recognized that sex can affect the immune response to a pathogenic agent, as well as the susceptibility for some respiratory diseases. These different responses in males and females may be related to the actions of sex hormones. Angiotensin-converting enzyme 2 (ACE2) acts as the receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes COVID-19. The expression of ACE2 is influenced by sex hormones; therefore, we discuss in this article that this could be one of the reasons why COVID-19 is more prevalent in men than in women.
Collapse
Affiliation(s)
- Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, FCAVJ-UNESP-São Paulo State University, Jaboticabal, Brazil
| | - Danuzia A Marques
- Department of Pediatrics, Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| |
Collapse
|
23
|
Oudit GY, Pfeffer MA. Plasma angiotensin-converting enzyme 2: novel biomarker in heart failure with implications for COVID-19. Eur Heart J 2020; 41:1818-1820. [PMID: 32388547 PMCID: PMC7224688 DOI: 10.1093/eurheartj/ehaa414] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Gavin Y Oudit
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Marc A Pfeffer
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Gheblawi M, Wang K, Viveiros A, Nguyen Q, Zhong JC, Turner AJ, Raizada MK, Grant MB, Oudit GY. Angiotensin-Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System: Celebrating the 20th Anniversary of the Discovery of ACE2. Circ Res 2020; 126:1456-1474. [PMID: 32264791 PMCID: PMC7188049 DOI: 10.1161/circresaha.120.317015] [Citation(s) in RCA: 1358] [Impact Index Per Article: 271.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ACE2 (angiotensin-converting enzyme 2) has a multiplicity of physiological roles that revolve around its trivalent function: a negative regulator of the renin-angiotensin system, facilitator of amino acid transport, and the severe acute respiratory syndrome-coronavirus (SARS-CoV) and SARS-CoV-2 receptor. ACE2 is widely expressed, including, in the lungs, cardiovascular system, gut, kidneys, central nervous system, and adipose tissue. ACE2 has recently been identified as the SARS-CoV-2 receptor, the infective agent responsible for coronavirus disease 2019, providing a critical link between immunity, inflammation, ACE2, and cardiovascular disease. Although sharing a close evolutionary relationship with SARS-CoV, the receptor-binding domain of SARS-CoV-2 differs in several key amino acid residues, allowing for stronger binding affinity with the human ACE2 receptor, which may account for the greater pathogenicity of SARS-CoV-2. The loss of ACE2 function following binding by SARS-CoV-2 is driven by endocytosis and activation of proteolytic cleavage and processing. The ACE2 system is a critical protective pathway against heart failure with reduced and preserved ejection fraction including, myocardial infarction and hypertension, and against lung disease and diabetes mellitus. The control of gut dysbiosis and vascular permeability by ACE2 has emerged as an essential mechanism of pulmonary hypertension and diabetic cardiovascular complications. Recombinant ACE2, gene-delivery of Ace2, Ang 1-7 analogs, and Mas receptor agonists enhance ACE2 action and serve as potential therapies for disease conditions associated with an activated renin-angiotensin system. rhACE2 (recombinant human ACE2) has completed clinical trials and efficiently lowered or increased plasma angiotensin II and angiotensin 1-7 levels, respectively. Our review summarizes the progress over the past 20 years, highlighting the critical role of ACE2 as the novel SARS-CoV-2 receptor and as the negative regulator of the renin-angiotensin system, together with implications for the coronavirus disease 2019 pandemic and associated cardiovascular diseases.
Collapse
Affiliation(s)
- Mahmoud Gheblawi
- From the Department of Physiology (M.G., A.V., G.Y.O.)
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (M.G., K.W., A.V., Q.N., G.Y.O.)
| | - Kaiming Wang
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada (K.W., Q.N., G.Y.O.)
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (M.G., K.W., A.V., Q.N., G.Y.O.)
| | - Anissa Viveiros
- From the Department of Physiology (M.G., A.V., G.Y.O.)
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (M.G., K.W., A.V., Q.N., G.Y.O.)
| | - Quynh Nguyen
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada (K.W., Q.N., G.Y.O.)
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (M.G., K.W., A.V., Q.N., G.Y.O.)
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, China (J.-C.Z.)
| | - Anthony J. Turner
- School of Biomedical Sciences, University of Leeds, United Kingdom (A.J.T.)
| | - Mohan K. Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (M.K.R.)
| | - Maria B. Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham (M.B.G.)
| | - Gavin Y. Oudit
- From the Department of Physiology (M.G., A.V., G.Y.O.)
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada (K.W., Q.N., G.Y.O.)
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (M.G., K.W., A.V., Q.N., G.Y.O.)
| |
Collapse
|
25
|
Rahimi O, Kirby J, Varagic J, Westwood B, Tallant EA, Gallagher PE. Angiotensin-(1–7) reduces doxorubicin-induced cardiac dysfunction in male and female Sprague-Dawley rats through antioxidant mechanisms. Am J Physiol Heart Circ Physiol 2020; 318:H883-H894. [DOI: 10.1152/ajpheart.00224.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Doxorubicin (Dox) is an effective chemotherapeutic for a variety of pediatric malignancies. Unfortunately, Dox administration often results in a cumulative dose-dependent cardiotoxicity that manifests with marked oxidative stress, leading to heart failure. Adjunct therapies are needed to mitigate Dox cardiotoxicity and enhance quality of life in pediatric patients with cancer. Angiotensin-(1–7) [Ang-(1–7)] is an endogenous hormone with cardioprotective properties. This study investigated whether adjunct Ang-(1–7) attenuates cardiotoxicity resulting from exposure to Dox in male and female juvenile rats. Dox significantly reduced body mass, and the addition of Ang-(1–7) had no effect. However, adjunct Ang-(1–7) prevented Dox-mediated diastolic dysfunction, including markers of decreased passive filling as measured by reduced early diastole mitral valve flow velocity peak ( E) ( P < 0.05) and early diastole mitral valve annulus peak velocity ( e′; P < 0.001) and increased E/e′ ( P < 0.001), an echocardiographic measure of diastolic dysfunction. Since Dox treatment increases reactive oxygen species (ROS), the effect of Ang-(1–7) on oxidative by-products and enzymes that generate or reduce ROS was investigated. In hearts of male and female juvenile rats, Dox increased NADPH oxidase 4 ( P < 0.05), a major cardiovascular NADPH oxidase isozyme that generates ROS, as well as 4-hydroxynonenal ( P < 0.001) and malondialdehyde ( P < 0.001), markers of lipid peroxidation; Ang-(1–7) prevented these effects of Dox. Cotreatment with Dox and Ang-(1–7) increased the antioxidant enzymes SOD1 (male: P < 0.05; female: P < 0.01) and catalase ( P < 0.05), which likely contributed to reduced ROS. These results demonstrate that Ang-(1–7) prevents diastolic dysfunction in association with a reduction in ROS, suggesting that the heptapeptide hormone may serve as an effective adjuvant to improve Dox-induced cardiotoxicity. NEW & NOTEWORTHY Ang-(1–7) is a clinically safe peptide hormone with cardioprotective and antineoplastic properties that could be used as an adjuvant therapy to improve cancer treatment and mitigate the long-term cardiotoxicity associated with doxorubicin in pediatric patients with cancer.
Collapse
Affiliation(s)
- Omeed Rahimi
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jay Kirby
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jasmina Varagic
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Brian Westwood
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - E. Ann Tallant
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Patricia E. Gallagher
- Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
26
|
Azzam O, Kiuchi MG, Ho JK, Matthews VB, Gavidia LML, Nolde JM, Carnagarin R, Schlaich MP. New Molecules for Treating Resistant Hypertension: a Clinical Perspective. Curr Hypertens Rep 2019; 21:80. [PMID: 31506798 DOI: 10.1007/s11906-019-0978-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW To review the findings of trials evaluating pharmacological treatment approaches for hypertension in general, and resistant hypertension (RH) in particular, and propose future research and clinical directions. RECENT FINDINGS RH is defined as blood pressure (BP) that remains above target levels despite adherence to at least three antihypertensive medications, including a diuretic. Thus far, clinical trials of pharmacological approaches in RH have focused on older molecules, with spironolactone being demonstrated as the most efficacious fourth-line agent. However, the use of spironolactone in clinical practice is hampered by its side effect profile and the risk of hyperkalaemia in important RH subgroups, such as patients with moderate-severe chronic kidney disease (CKD). Clinical trials of new molecules targeting both well-established and more recently elucidated pathophysiologic mechanisms of hypertension offer a multitude of potential treatment avenues that warrant further evaluation in the context of RH. These include selective mineralocorticoid receptor antagonists (MRAs), aldosterone synthase inhibitors (ASIs), activators of the counterregulatory renin-angiotensin-system (RAS), vaccines, neprilysin inhibitors alone and in combined formulations, natriuretic peptide receptor agonists A (NPRA-A) agonists, vasoactive intestinal peptide (VIP) agonists, centrally acting aminopeptidase A (APA|) inhibitors, antimicrobial suppression of central sympathetic outflow (minocycline), dopamine β-hydroxylase (DβH) inhibitors and Na+/H+ Exchanger 3 (NHE3) inhibitors. There is a paucity of data from trials evaluating newer molecules for the treatment of RH. Emergent novel molecules for non-resistant forms of hypertension heighten the prospects of identifying new, effective and well-tolerated pharmacological approaches to RH. There is a glaring need to undertake RH-focused trials evaluating their efficacy and clinical applicability.
Collapse
Affiliation(s)
- Omar Azzam
- Department of Internal Medicine, Royal Perth Hospital, Perth, Western Australia, Australia.,Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Marcio G Kiuchi
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Jan K Ho
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Vance B Matthews
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Leslie Marisol Lugo Gavidia
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Janis M Nolde
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit / Medical Research Foundation, University of Western Australia, Level 3, MRF Building, Rear 50 Murray St, Perth, WA, 6000, Australia. .,Departments of Cardiology and Nephrology, Royal Perth Hospital, Perth, Australia. .,Neurovascular Hypertension & Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
27
|
Chen Y, Zhao W, Liu C, Meng W, Zhao T, Bhattacharya SK, Sun Y. Molecular and Cellular Effect of Angiotensin 1-7 on Hypertensive Kidney Disease. Am J Hypertens 2019; 32:460-467. [PMID: 30715105 DOI: 10.1093/ajh/hpz009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/25/2018] [Accepted: 01/15/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Studies implicate that angiotensin 1-7 (Ang1-7) imparts protective effects in the kidney. However, its relevance in hypertensive kidney disease is not fully understood. The purpose of this study was to explore the role of Ang1-7 on renal damage/remodeling during hypertension and its potential underlying molecular-cellular mechanisms. METHODS Hypertension was induced in adult Sprague-Dawley rats by infusion of aldosterone (ALDO; 0.75 μg/hour) for 4 weeks with or without co-treatment of Ang1-7 (1 mg/kg/day). Untreated rats served as controls. Systolic blood pressure was monitored by tail-cuff technique. Renal fibrosis was evaluated by picrosirius red staining and renal collagen volume fraction was quantitated using imaging analyzing system. The expression of profibrotic factors [transforming growth factor-β1 (TGF-β1), platelet-derived growth factor-D (PDGF-D), fibroblast growth factor-1 (FGF-1), vascular endothelial growth factor-D (VEGF-D), and tissue inhibitors of metalloproteinases (TIMPs)] and free radical producing enzymes (inducible nitric oxide synthase and nicotinamide adenine dinucleotide phosphate [NADPH] oxidase) in the kidney were examined by reverse transcription-polymerase chain reaction and western blot. Renal oxidative stress was assessed by malondialdehyde (MDA) measurement. RESULTS Chronic ALDO infusion caused hypertension and hypertensive renal disease represented as glomerular damage/sclerosis. Ang1-7 co-treatment did not affect blood pressure in ALDO-treated rats, but significantly attenuated the glomerular damage/fibrosis. ALDO treatment significantly elevated renal expression of profibrogenic factors, including TGF-β1, TIMP-1/TIMP-2, FGF-1, PDGF-D, and VEGF-D, whereas Ang1-7 co-treatment significantly reduced renal TGF-β1, TIMP-1/TIMP-2, and FGF-1, but not PDGF-D and VEGF-D. Furthermore, ALDO infusion elevated NADPH oxidase (gp91phox) and MDA in the kidney, which was attenuated by Ang1-7 co-treatment. CONCLUSIONS Ang1-7 plays a protective role in the hypertensive kidney disease independent of blood pressure. The beneficial effects of Ang1-7 are likely mediated via suppressing TGF-β/FGF-1 pathways and oxidative stress.
Collapse
Affiliation(s)
- Yuanjian Chen
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Wenyuan Zhao
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chang Liu
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Weixin Meng
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Tieqiang Zhao
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Syamal K Bhattacharya
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Yao Sun
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
28
|
Santos RAS, Oudit GY, Verano-Braga T, Canta G, Steckelings UM, Bader M. The renin-angiotensin system: going beyond the classical paradigms. Am J Physiol Heart Circ Physiol 2019; 316:H958-H970. [PMID: 30707614 PMCID: PMC7191626 DOI: 10.1152/ajpheart.00723.2018] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thirty years ago, a novel axis of the renin-angiotensin system (RAS) was unveiled by the discovery of angiotensin-(1−7) [ANG-(1−7)] generation in vivo. Later, angiotensin-converting enzyme 2 (ACE2) was shown to be the main mediator of this reaction, and Mas was found to be the receptor for the heptapeptide. The functional analysis of this novel axis of the RAS that followed its discovery revealed numerous protective actions in particular for cardiovascular diseases. In parallel, similar protective actions were also described for one of the two receptors of ANG II, the ANG II type 2 receptor (AT2R), in contrast to the other, the ANG II type 1 receptor (AT1R), which mediates deleterious actions of this peptide, e.g., in the setting of cardiovascular disease. Very recently, another branch of the RAS was discovered, based on angiotensin peptides in which the amino-terminal aspartate was replaced by alanine, the alatensins. Ala-ANG-(1−7) or alamandine was shown to interact with Mas-related G protein-coupled receptor D, and the first functional data indicated that this peptide also exerts protective effects in the cardiovascular system. This review summarizes the presentations given at the International Union of Physiological Sciences Congress in Rio de Janeiro, Brazil, in 2017, during the symposium entitled “The Renin-Angiotensin System: Going Beyond the Classical Paradigms,” in which the signaling and physiological actions of ANG-(1−7), ACE2, AT2R, and alatensins were reported (with a focus on noncentral nervous system-related tissues) and the therapeutic opportunities based on these findings were discussed.
Collapse
Affiliation(s)
- Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Federal University of Minas Gerais , Belo Horizonte, Minas Gerais , Brazil
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta , Edmonton , Canada
| | - Thiago Verano-Braga
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Federal University of Minas Gerais , Belo Horizonte, Minas Gerais , Brazil
| | - Giovanni Canta
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Federal University of Minas Gerais , Belo Horizonte, Minas Gerais , Brazil
| | - Ulrike Muscha Steckelings
- Department of Molecular Medicine, Cardiovascular & Renal Research, University of Southern Denmark, Odense, Denmark
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine , Berlin , Germany.,Deutsches Zentrum für Herz-Kreislaufforschung, Partner Site Berlin, Berlin , Germany.,Berlin Institute of Health , Berlin , Germany.,Charité-University Medicine, Berlin , Germany.,Institute for Biology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
29
|
Jang IA, Kim EN, Lim JH, Kim MY, Ban TH, Yoon HE, Park CW, Chang YS, Choi BS. Effects of Resveratrol on the Renin-Angiotensin System in the Aging Kidney. Nutrients 2018; 10:E1741. [PMID: 30424556 PMCID: PMC6267480 DOI: 10.3390/nu10111741] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin system (RAS), especially the angiotensin II (Ang II)/angiotensin II type 1 receptor (AT1R) axis, plays an important role in the aging process of the kidney, through increased tissue reactive oxygen species production and progressively increased oxidative stress. In contrast, the angiotensin 1-7 (Ang 1-7)/Mas receptor (MasR) axis, which counteracts the effects of Ang II, is protective for end-organ damage. To evaluate the ability of resveratrol (RSV) to modulate the RAS in aging kidneys, eighteen-month-old male C57BL/6 mice were divided into two groups that received either normal mouse chow or chow containing resveratrol, for six months. Renal expressions of RAS components, as well as pro- and antioxidant enzymes, were measured and mouse kidneys were isolated for histopathology. Resveratrol-treated mice demonstrated better renal function and reduced albuminuria, with improved renal histologic findings. Resveratrol suppressed the Ang II/AT1R axis and enhanced the AT2R/Ang 1-7/MasR axis. Additionally, the expression of nicotinamide adenine dinucleotide phosphate oxidase 4, 8-hydroxy-2'-deoxyguanosine, 3-nitrotyrosine, collagen IV, and fibronectin was decreased, while the expression of endothelial nitric oxide synthase and superoxide dismutase 2 was increased by resveratrol treatment. These findings demonstrate that resveratrol exerts protective effects on aging kidneys by reducing oxidative stress, inflammation, and fibrosis, through Ang II suppression and MasR activation.
Collapse
Affiliation(s)
- In-Ae Jang
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Eun Nim Kim
- Division of Medical Cell Biology, Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Ji Hee Lim
- Division of Medical Cell Biology, Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Min Young Kim
- Division of Medical Cell Biology, Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Tae Hyun Ban
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul 06591, Korea.
| | - Hye Eun Yoon
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, Incheon St. Mary's Hospital, Incheon 21431, Korea.
| | - Cheol Whee Park
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul 06591, Korea.
| | - Yoon Sik Chang
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, Yeouido St. Mary's Hospital, Seoul 07345, Korea.
| | - Bum Soon Choi
- Department of Internal medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- Division of Nephrology, Department of Internal Medicine, St. Paul's Hospital, Seoul 02559, Korea.
| |
Collapse
|
30
|
The anti-inflammatory peptide Ac-SDKP: Synthesis, role in ACE inhibition, and its therapeutic potential in hypertension and cardiovascular diseases. Pharmacol Res 2018; 134:268-279. [DOI: 10.1016/j.phrs.2018.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/12/2018] [Accepted: 07/07/2018] [Indexed: 01/27/2023]
|
31
|
O’Connor AT, Clark MA. Astrocytes and the Renin Angiotensin System: Relevance in Disease Pathogenesis. Neurochem Res 2018; 43:1297-1307. [DOI: 10.1007/s11064-018-2557-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/21/2018] [Accepted: 05/23/2018] [Indexed: 12/29/2022]
|
32
|
Hong S, Kwon J, Hiep NT, Sim SJ, Kim N, Kim KH, Lee D, Mar W. The isoflavones and extracts from Maclura tricuspidata fruit protect against neuronal cell death in ischemic injury via induction of Nox4-targeting miRNA-25, miRNA-92a, and miRNA-146a. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
33
|
Silva SD, Jara ZP, Peres R, Lima LS, Scavone C, Montezano AC, Touyz RM, Casarini DE, Michelini LC. Temporal changes in cardiac oxidative stress, inflammation and remodeling induced by exercise in hypertension: Role for local angiotensin II reduction. PLoS One 2017; 12:e0189535. [PMID: 29232407 PMCID: PMC5726656 DOI: 10.1371/journal.pone.0189535] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/27/2017] [Indexed: 12/15/2022] Open
Abstract
Exercise training reduces renin-angiotensin system (RAS) activation, decreases plasma and tissue oxidative stress and inflammation in hypertension. However, the temporal nature of these phenomena in response to exercise is unknown. We sought to determine in spontaneously hypertensive rats (SHR) and age-matched WKY controls the weekly effects of training on blood pressure (BP), plasma and left ventricle (LV) Ang II and Ang-(1–7) content (HPLC), LV oxidative stress (DHE staining), gene and protein expression (qPCR and WB) of pro-inflammatory cytokines, antioxidant enzymes and their consequence on hypertension-induced cardiac remodeling. SHR and WKY were submitted to aerobic training (T) or maintained sedentary (S) for 8 weeks; measurements were made at weeks 0, 1, 2, 4 and 8. Hypertension-induced cardiac hypertrophy was accompanied by acute plasma Ang II increase with amplified responses during the late phase of LV hypertrophy. Similar pattern was observed for oxidative stress markers, TNF alpha and interleukin-1β, associated with cardiomyocytes’ diameter enlargement and collagen deposition. SHR-T exhibited prompt and marked decrease in LV Ang II content (T1vs T4 in WKY-T), normalized oxidative stress (T2), augmented antioxidant defense (T4) and reduced both collagen deposition and inflammatory profile (T8), without changing cardiomyocytes’ diameter and LV hypertrophy. These changes were accompanied by decreased plasma Ang II content (T2-T4) and reduced BP (T8). SHR-T and WKY-T showed parallel increases in LV and plasma Ang-(1–7) content. Our data indicate that early training-induced downregulation of LV ACE-AngII-AT1 receptor axis is a crucial mechanism to reduce oxidative/pro-inflammatory profile and improve antioxidant defense in SHR-T, showing in addition this effect precedes plasma RAS deactivation.
Collapse
Affiliation(s)
- Sebastião D. Silva
- Department of Physiology & Biophysics, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, SP, Brazil
- Institute of Cardiovascular and Medical Sciences, BHF GCRC, University of Glasgow, Glasgow, United Kingdom
| | - Zaira P. Jara
- Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Roseli Peres
- Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Larissa S. Lima
- Department of Pharmacology, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Cristóforo Scavone
- Department of Pharmacology, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Augusto C. Montezano
- Institute of Cardiovascular and Medical Sciences, BHF GCRC, University of Glasgow, Glasgow, United Kingdom
| | - Rhian M. Touyz
- Institute of Cardiovascular and Medical Sciences, BHF GCRC, University of Glasgow, Glasgow, United Kingdom
| | - Dulce E. Casarini
- Department of Medicine, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Lisete C. Michelini
- Department of Physiology & Biophysics, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, SP, Brazil
- * E-mail:
| |
Collapse
|
34
|
Wu J, Liao W, Udenigwe CC. Revisiting the mechanisms of ACE inhibitory peptides from food proteins. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.07.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
35
|
Tamargo M, Tamargo J. Future drug discovery in renin-angiotensin-aldosterone system intervention. Expert Opin Drug Discov 2017; 12:827-848. [PMID: 28541811 DOI: 10.1080/17460441.2017.1335301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Renin-angiotensin-aldosterone system inhibitors (RAASIs), including angiotensin-converting enzyme inhibitors, angiotensin AT1 receptor blockers and mineralocorticoid receptor antagonists (MRAs), are the cornerstone for the treatment of cardiovascular and renal diseases. Areas covered: The authors searched MEDLINE, PubMed and ClinicalTrials.gov to identify eligible full-text English language papers. Herein, the authors discuss AT2-receptor agonists and ACE2/angiotensin-(1-7)/Mas-receptor axis modulators, direct renin inhibitors, brain aminopeptidase A inhibitors, biased AT1R blockers, chymase inhibitors, multitargeted drugs, vaccines and aldosterone receptor antagonists as well as aldosterone synthase inhibitors. Expert opinion: Preclinical studies have demonstrated that activation of the protective axis of the RAAS represents a novel therapeutic strategy for treating cardiovascular and renal diseases, but there are no clinical trials supporting our expectations. Non-steroidal MRAs might become the third-generation of MRAs for the treatment of heart failure, diabetes mellitus and chronic kidney disease. The main challenge for these new drugs is that conventional RAASIs are safe, effective and cheap generics. Thus, the future of new RAASIs will be directed by economical/strategic reasons.
Collapse
Affiliation(s)
- Maria Tamargo
- a Department of Cardiology , Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV , Madrid , Spain
| | - Juan Tamargo
- b Department of Pharmacology , School of Medicine, University Complutense, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV , Madrid , Spain
| |
Collapse
|
36
|
Li S, Wang Z, Yang X, Hu B, Huang Y, Fan S. Association between circulating angiotensin-converting enzyme 2 and cardiac remodeling in hypertensive patients. Peptides 2017; 90:63-68. [PMID: 28223093 DOI: 10.1016/j.peptides.2017.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 02/11/2017] [Accepted: 02/16/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Angiotensin-converting enzyme 2 (ACE2) plays a vital role in the pathogenesis of hypertension-induced cardiac remodeling and exhibits cardioprotective properties in hypertensive animal models. Evidence that ACE2 is an important regulator of hypertensive cardiac remodeling in humans has not been addressed directly yet. METHODS A total of 161 patients with essential hypertension and 47 age- and sex-matched normotensive healthy subjects were consecutively recruited. Serum concentration levels of ACE2 were determined by enzyme-linked immunosorbent assay. Cardiac structural and functional parameters were measured by echocardiography. RESULTS Serum ACE2 concentrations were higher in hypertensive patients compared to healthy subjects (170.31 [83.50-707.12] pg/ml in patients versus 59.28 [39.71-81.81] pg/ml in healthy subjects, P<0.001). After adjustment for confounders, including age, sex, body mass index, snoring, smoking, duration of hypertension, comorbidities, medication use, mean arterial pressure and N-terminal pro-brain natriuretic peptide, serum ACE2 concentrations were positively correlated with left atrial diameter, left ventricular end-diastolic diameter and left ventricular mass in hypertensive patients. Moreover, multiple regression analyses adjusting for covariates revealed that serum ACE2 concentrations were also independently associated with left ventricular ejection fraction and late diastolic filling velocities of the mitral inflow. CONCLUSIONS This study reveals an elevated serum concentration of ACE2 and independent associations between serum ACE2 and echocardiographic parameters in hypertensive patients.
Collapse
Affiliation(s)
- Shichao Li
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, 57 Jianshe South Rd, Tangshan City, Hebei Province, 063000, China
| | - Zhijun Wang
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, 57 Jianshe South Rd, Tangshan City, Hebei Province, 063000, China.
| | - Xiuhong Yang
- Department of Physiology, School of Basic Medical Sciences, North China University of Science and Technology, 57 Jianshe South Rd, Tangshan City, Hebei Province, 063000, China.
| | - Bo Hu
- School of Public Health, North China University of Science and Technology, 57 Jianshe South Rd, Tangshan City, Hebei Province, 063000, China
| | - Yuling Huang
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, 57 Jianshe South Rd, Tangshan City, Hebei Province, 063000, China
| | - Sujing Fan
- Department of Physiology, School of Basic Medical Sciences, North China University of Science and Technology, 57 Jianshe South Rd, Tangshan City, Hebei Province, 063000, China
| |
Collapse
|
37
|
Hong S, Kwon J, Kim DW, Lee HJ, Lee D, Mar W. Mulberrofuran G Protects Ischemic Injury-induced Cell Death via Inhibition of NOX4-mediated ROS Generation and ER Stress. Phytother Res 2016; 31:321-329. [DOI: 10.1002/ptr.5754] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 09/19/2016] [Accepted: 11/07/2016] [Indexed: 01/25/2023]
Affiliation(s)
- Sungeun Hong
- Natural Products Research Institute, College of Pharmacy; Seoul National University; Seoul 08826 Korea
| | - Jaeyoung Kwon
- Department of Biosystems and Biotechnology; Korea University; Seoul 02841 Korea
| | - Dong-Woo Kim
- Natural Products Research Institute, College of Pharmacy; Seoul National University; Seoul 08826 Korea
| | - Hak Ju Lee
- Division of Wood Chemistry and Microbiology, Department of Forest Products; Korea Forest Research Institute; Seoul 02455 Korea
| | - Dongho Lee
- Department of Biosystems and Biotechnology; Korea University; Seoul 02841 Korea
| | - Woongchon Mar
- Natural Products Research Institute, College of Pharmacy; Seoul National University; Seoul 08826 Korea
| |
Collapse
|
38
|
Patel VB, Zhong JC, Grant MB, Oudit GY. Role of the ACE2/Angiotensin 1-7 Axis of the Renin-Angiotensin System in Heart Failure. Circ Res 2016; 118:1313-26. [PMID: 27081112 DOI: 10.1161/circresaha.116.307708] [Citation(s) in RCA: 611] [Impact Index Per Article: 67.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/19/2016] [Indexed: 11/16/2022]
Abstract
Heart failure (HF) remains the most common cause of death and disability, and a major economic burden, in industrialized nations. Physiological, pharmacological, and clinical studies have demonstrated that activation of the renin-angiotensin system is a key mediator of HF progression. Angiotensin-converting enzyme 2 (ACE2), a homolog of ACE, is a monocarboxypeptidase that converts angiotensin II into angiotensin 1-7 (Ang 1-7) which, by virtue of its actions on the Mas receptor, opposes the molecular and cellular effects of angiotensin II. ACE2 is widely expressed in cardiomyocytes, cardiofibroblasts, and coronary endothelial cells. Recent preclinical translational studies confirmed a critical counter-regulatory role of ACE2/Ang 1-7 axis on the activated renin-angiotensin system that results in HF with preserved ejection fraction. Although loss of ACE2 enhances susceptibility to HF, increasing ACE2 level prevents and reverses the HF phenotype. ACE2 and Ang 1-7 have emerged as a key protective pathway against HF with reduced and preserved ejection fraction. Recombinant human ACE2 has been tested in phase I and II clinical trials without adverse effects while lowering and increasing plasma angiotensin II and Ang 1-7 levels, respectively. This review discusses the transcriptional and post-transcriptional regulation of ACE2 and the role of the ACE2/Ang 1-7 axis in cardiac physiology and in the pathophysiology of HF. The pharmacological and therapeutic potential of enhancing ACE2/Ang 1-7 action as a novel therapy for HF is highlighted.
Collapse
Affiliation(s)
- Vaibhav B Patel
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Jiu-Chang Zhong
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Maria B Grant
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Gavin Y Oudit
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.).
| |
Collapse
|
39
|
Diniz GP, Senger N, Carneiro-Ramos MS, Santos RAS, Barreto-Chaves MLM. Cardiac ACE2/angiotensin 1-7/Mas receptor axis is activated in thyroid hormone-induced cardiac hypertrophy. Ther Adv Cardiovasc Dis 2016; 10:192-202. [PMID: 26715125 PMCID: PMC5942623 DOI: 10.1177/1753944715623228] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Thyroid hormone (TH) promotes marked effects on the cardiovascular system, including the development of cardiac hypertrophy. Some studies have demonstrated that the renin-angiotensin system (RAS) is a key mediator of the cardiac growth in response to elevated TH levels. Although some of the main RAS components are changed in cardiac tissue on hyperthyroid state, the potential modulation of the counter regulatory components of the RAS, such as angiotensin-converting enzyme type 2 (ACE2), angiotensin 1-7 (Ang 1-7) levels and Mas receptor induced by hyperthyroidism is unknown. The aim of this study was to investigate the effect of hyperthyroidism on cardiac Ang 1-7, ACE2 and Mas receptor levels. METHODS Hyperthyroidism was induced in Wistar rats by daily intraperitoneal injections of T4 for 14 days. RESULTS Although plasma Ang 1-7 levels were unchanged by hyperthyroidism, cardiac Ang 1-7 levels were increased in TH-induced cardiac hypertrophy. ACE2 enzymatic activity was significantly increased in hearts from hyperthyroid animals, which may be contributing to the higher Ang 1-7 levels observed in the T4 group. Furthermore, elevated cardiac levels of Ang 1-7 levels were accompanied by increased Mas receptor protein levels. CONCLUSION The counter-regulatory components of the RAS are activated in hyperthyroidism and may be contributing to modulate the cardiac hypertrophy in response to TH.
Collapse
Affiliation(s)
- Gabriela P Diniz
- Department of Anatomy, Laboratory of Cell Biology and Functional Anatomy, University of São Paulo, Sao Paulo, Brazil
| | - Nathalia Senger
- Department of Anatomy, Laboratory of Cell Biology and Functional Anatomy, University of São Paulo, Sao Paulo, Brazil
| | | | - Robson A S Santos
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, BrazilCardiology Institute of Rio Grande do Sul, University Foundation of Cardiology
| | - Maria Luiza M Barreto-Chaves
- Laboratory of Cellular Biology and Functional Anatomy, Department of Anatomy, Biomedical Sciences Institute, University of São Paulo, Av. Prof. Lineu Prestes 2415, Cidade Universitária, São Paulo, SP 05508-900, Brazil
| |
Collapse
|
40
|
Wang W, McKinnie SMK, Farhan M, Paul M, McDonald T, McLean B, Llorens-Cortes C, Hazra S, Murray AG, Vederas JC, Oudit GY. Angiotensin-Converting Enzyme 2 Metabolizes and Partially Inactivates Pyr-Apelin-13 and Apelin-17: Physiological Effects in the Cardiovascular System. Hypertension 2016; 68:365-77. [PMID: 27217402 DOI: 10.1161/hypertensionaha.115.06892] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/21/2016] [Indexed: 02/06/2023]
Abstract
Apelin peptides mediate beneficial effects on the cardiovascular system and are being targeted as potential new drugs. However, apelin peptides have extremely short biological half-lives, and improved understanding of apelin peptide metabolism may lead to the discovery of biologically stable analogues with therapeutic potential. We examined the ability of angiotensin-converting enzyme 2 (ACE2) to cleave and inactivate pyr-apelin 13 and apelin 17, the dominant apelin peptides. Computer-assisted modeling shows a conserved binding of pyr-apelin 13 and apelin 17 to the ACE2 catalytic site. In ACE2 knockout mice, hypotensive action of pyr-apelin 13 and apelin 17 was potentiated, with a corresponding greater elevation in plasma apelin levels. Similarly, pharmacological inhibition of ACE2 potentiated the vasodepressor action of apelin peptides. Biochemical analysis confirmed that recombinant human ACE2 can cleave pyr-apelin 13 and apelin 17 efficiently, and apelin peptides are degraded slower in ACE2-deficient plasma. The biological relevance of ACE2-mediated proteolytic processing of apelin peptides was further supported by the reduced potency of pyr-apelin 12 and apelin 16 on the activation of signaling pathways and nitric oxide production from endothelial cells. Importantly, although pyr-apelin 13 and apelin 17 rescued contractile function in a myocardial ischemia-reperfusion model, ACE2 cleavage products, pyr-apelin 12 and 16, were devoid of these cardioprotective effects. We designed and synthesized active apelin analogues that were resistant to ACE2-mediated degradation, thereby confirming that stable apelin analogues can be designed as potential drugs. We conclude that ACE2 represents a major negative regulator of apelin action in the vasculature and heart.
Collapse
Affiliation(s)
- Wang Wang
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Shaun M K McKinnie
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Maikel Farhan
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Manish Paul
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Tyler McDonald
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Brent McLean
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Catherine Llorens-Cortes
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Saugata Hazra
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Allan G Murray
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - John C Vederas
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.)
| | - Gavin Y Oudit
- From the Division of Cardiology, Department of Medicine (W.W., B.M., G.Y.O.), Mazankowski Alberta Heart Institute (W.W., B.M., G.Y.O.), Department of Chemistry, Faculty of Science (S.M.K.M., T.M., J.C.V.), and Division of Nephrology, Department of Medicine (M.F., A.G.M.), University of Alberta, Edmonton, AB, Canada; Department of Zoology, RBC College, West Bengal State University, West Bengal, India (M.P.); INSERM, Laboratory of Central Neuropeptides and Regulations of Water Balance and Cardiovascular Functions, Center for Interdisciplinary Research in Biology, College de France, Paris, France (C.L.-C.); Department of Biotechnology, Indian Institute of Technology, Roorkee, India (S.H.).
| |
Collapse
|
41
|
Husková Z, Kopkan L, Červenková L, Doleželová Š, Vaňourková Z, Škaroupková P, Nishiyama A, Kompanowska-Jezierska E, Sadowski J, Kramer HJ, Červenka L. Intrarenal alterations of the angiotensin-converting enzyme type 2/angiotensin 1-7 complex of the renin-angiotensin system do not alter the course of malignant hypertension in Cyp1a1-Ren-2 transgenic rats. Clin Exp Pharmacol Physiol 2016; 43:438-49. [DOI: 10.1111/1440-1681.12553] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/15/2016] [Accepted: 01/26/2016] [Indexed: 01/13/2023]
Affiliation(s)
- Zuzana Husková
- Centre for Experimental Medicine; Institute for Clinical and Experimental Medicine; Prague Czech Republic
| | - Libor Kopkan
- Centre for Experimental Medicine; Institute for Clinical and Experimental Medicine; Prague Czech Republic
| | - Lenka Červenková
- Centre for Experimental Medicine; Institute for Clinical and Experimental Medicine; Prague Czech Republic
| | - Šárka Doleželová
- Centre for Experimental Medicine; Institute for Clinical and Experimental Medicine; Prague Czech Republic
| | - Zdeňka Vaňourková
- Centre for Experimental Medicine; Institute for Clinical and Experimental Medicine; Prague Czech Republic
| | - Petra Škaroupková
- Centre for Experimental Medicine; Institute for Clinical and Experimental Medicine; Prague Czech Republic
| | | | - Elzbieta Kompanowska-Jezierska
- Department of Renal and Body Fluid Physiology; Mossakowski Medical Research Centre; Polish Academy of Science; Warsaw Poland
| | - Janusz Sadowski
- Department of Renal and Body Fluid Physiology; Mossakowski Medical Research Centre; Polish Academy of Science; Warsaw Poland
| | - Herbert J. Kramer
- Section of Nephrology; Department of Medicine; University of Bonn; Bonn Germany
| | - Luděk Červenka
- Centre for Experimental Medicine; Institute for Clinical and Experimental Medicine; Prague Czech Republic
- Department of Pathophysiology; 2nd Faculty of Medicine; Charles University; Prague Czech Republic
| |
Collapse
|
42
|
Mak KY, Chin R, Cunningham SC, Habib MR, Torresi J, Sharland AF, Alexander IE, Angus PW, Herath CB. ACE2 Therapy Using Adeno-associated Viral Vector Inhibits Liver Fibrosis in Mice. Mol Ther 2015; 23:1434-43. [PMID: 25997428 DOI: 10.1038/mt.2015.92] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/28/2015] [Indexed: 12/11/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) which breaks down profibrotic peptide angiotensin II to antifibrotic peptide angiotensin-(1-7) is a potential therapeutic target in liver fibrosis. We therefore investigated the long-term therapeutic effect of recombinant ACE2 using a liver-specific adeno-associated viral genome 2 serotype 8 vector (rAAV2/8-ACE2) with a liver-specific promoter in three murine models of chronic liver disease, including carbon tetrachloride-induced toxic injury, bile duct ligation-induced cholestatic injury, and methionine- and choline-deficient diet-induced steatotic injury. A single injection of rAAV2/8-ACE2 was administered after liver disease has established. Hepatic fibrosis, gene and protein expression, and the mechanisms that rAAV2/8-ACE2 therapy associated reduction in liver fibrosis were analyzed. Compared with control group, rAAV2/8-ACE2 therapy produced rapid and sustained upregulation of hepatic ACE2, resulting in a profound reduction in fibrosis and profibrotic markers in all diseased models. These changes were accompanied by reduction in hepatic angiotensin II levels with concomitant increases in hepatic angiotensin-(1-7) levels, resulting in significant reductions of NADPH oxidase assembly, oxidative stress and ERK1/2 and p38 phosphorylation. Moreover, rAAV2/8-ACE2 therapy normalized increased intrahepatic vascular tone in fibrotic livers. We conclude that rAAV2/8-ACE2 is an effective liver-targeted, long-term therapy for liver fibrosis and its complications without producing unwanted systemic effects.
Collapse
Affiliation(s)
- Kai Y Mak
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Ruth Chin
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Victoria, Australia
| | - Sharon C Cunningham
- Gene Therapy Research Unit, Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Miriam R Habib
- Transplantation Research Group, Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Joseph Torresi
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
| | - Alexandra F Sharland
- Transplantation Research Group, Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Peter W Angus
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia.,Department of Gastroenterology and Hepatology, Austin Health, Heidelberg, Victoria, Australia
| | - Chandana B Herath
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
43
|
Antagonism of angiotensin 1-7 prevents the therapeutic effects of recombinant human ACE2. J Mol Med (Berl) 2015; 93:1003-13. [PMID: 25874965 DOI: 10.1007/s00109-015-1285-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/04/2015] [Accepted: 03/27/2015] [Indexed: 01/24/2023]
Abstract
UNLABELLED Activation of the angiotensin 1-7/Mas receptor (MasR) axis counteracts angiotensin II (Ang II)-mediated cardiovascular disease. Recombinant human angiotensin-converting enzyme 2 (rhACE2) generates Ang 1-7 from Ang II. We hypothesized that the therapeutic effects of rhACE2 are dependent on Ang 1-7 action. Wild type male C57BL/6 mice (10-12 weeks old) were infused with Ang II (1.5 mg/kg/d) and treated with rhACE2 (2 mg/kg/d). The Ang 1-7 antagonist, A779 (200 ng/kg/min), was administered to a parallel group of mice. rhACE2 prevented Ang II-induced hypertrophy and diastolic dysfunction while A779 prevented these beneficial effects and precipitated systolic dysfunction. rhACE2 effectively antagonized Ang II-mediated myocardial fibrosis which was dependent on the action of Ang 1-7. Myocardial oxidative stress and matrix metalloproteinase 2 activity was further increased by Ang 1-7 inhibition even in the presence of rhACE2. Activation of Akt and endothelial nitric oxide synthase (eNOS) by rhACE2 were suppressed by the antagonism of Ang 1-7 while the activation of pathological signaling pathways was maintained. Blocking Ang 1-7 action prevents the therapeutic effects of rhACE2 in the setting of elevated Ang II culminating in systolic dysfunction. These results highlight a key cardioprotective role of Ang 1-7, and increased Ang 1-7 action represents a potential therapeutic strategy for cardiovascular diseases. KEY MESSAGES Activation of the renin-angiotensin system (RAS) plays a key pathogenic role in cardiovascular disease. ACE2, a monocarboxypeptidase, negatively regulates pathological effects of Ang II. Antagonizing Ang 1-7 prevents the therapeutic effects of recombinant human ACE2. Our results highlight a key protective role of Ang 1-7 in cardiovascular disease.
Collapse
|
44
|
Zhang Z, Chen L, Zhong J, Gao P, Oudit GY. ACE2/Ang-(1–7) signaling and vascular remodeling. SCIENCE CHINA-LIFE SCIENCES 2014; 57:802-8. [DOI: 10.1007/s11427-014-4693-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 05/20/2014] [Indexed: 02/06/2023]
|
45
|
Hamza SM, Dyck JRB. Systemic and renal oxidative stress in the pathogenesis of hypertension: modulation of long-term control of arterial blood pressure by resveratrol. Front Physiol 2014; 5:292. [PMID: 25140155 PMCID: PMC4122172 DOI: 10.3389/fphys.2014.00292] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/19/2014] [Indexed: 12/12/2022] Open
Abstract
Hypertension affects over 25% of the global population and is associated with grave and often fatal complications that affect many organ systems. Although great advancements have been made in the clinical assessment and treatment of hypertension, the cause of hypertension in over 90% of these patients is unknown, which hampers the development of targeted and more effective treatment. The etiology of hypertension involves multiple pathological processes and organ systems, however one unifying feature of all of these contributing factors is oxidative stress. Once the body's natural anti-oxidant defense mechanisms are overwhelmed, reactive oxygen species (ROS) begin to accumulate in the tissues. ROS play important roles in normal regulation of many physiological processes, however in excess they are detrimental and cause widespread cell and tissue damage as well as derangements in many physiological processes. Thus, control of oxidative stress has become an attractive target for pharmacotherapy to prevent and manage hypertension. Resveratrol (trans-3,5,4'-Trihydroxystilbene) is a naturally occurring polyphenol which has anti-oxidant effects in vivo. Many studies have shown anti-hypertensive effects of resveratrol in different pre-clinical models of hypertension, via a multitude of mechanisms that include its function as an anti-oxidant. However, results have been mixed and in some cases resveratrol has no effect on blood pressure. This may be due to the heavy emphasis on peripheral vasodilator effects of resveratrol and virtually no investigation of its potential renal effects. This is particularly troubling in the arena of hypertension, where it is well known and accepted that the kidney plays an essential role in the long term regulation of arterial pressure and a vital role in the initiation, development and maintenance of chronic hypertension. It is thus the focus of this review to discuss the potential of resveratrol as an anti-hypertensive treatment via amelioration of oxidative stress within the framework of the fundamental physiological principles of long term regulation of arterial blood pressure.
Collapse
Affiliation(s)
- Shereen M. Hamza
- Department of Pediatrics, Cardiovascular Research Centre, University of AlbertaEdmonton, AB, Canada
| | - Jason R. B. Dyck
- Department of Pediatrics, Cardiovascular Research Centre, University of AlbertaEdmonton, AB, Canada
- Department of Pharmacology, Cardiovascular Research Centre, University of AlbertaEdmonton, AB, Canada
| |
Collapse
|
46
|
Silva PA, Monnerat-Cahli G, Pereira-Acácio A, Luzardo R, Sampaio LS, Luna-Leite MA, Lara LS, Einicker-Lamas M, Panizzutti R, Madeira C, Vieira-Filho LD, Castro-Chaves C, Ribeiro VS, Paixão ADO, Medei E, Vieyra A. Mechanisms involving Ang II and MAPK/ERK1/2 signaling pathways underlie cardiac and renal alterations during chronic undernutrition. PLoS One 2014; 9:e100410. [PMID: 24983243 PMCID: PMC4077653 DOI: 10.1371/journal.pone.0100410] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 05/27/2014] [Indexed: 02/07/2023] Open
Abstract
Background Several studies have correlated protein restriction associated with other nutritional deficiencies with the development of cardiovascular and renal diseases. The driving hypothesis for this study was that Ang II signaling pathways in the heart and kidney are affected by chronic protein, mineral and vitamin restriction. Methodology/Principal Findings Wistar rats aged 90 days were fed from weaning with either a control or a deficient diet that mimics those used in impoverished regions worldwide. Such restriction simultaneously increased ouabain-insensitive Na+-ATPase and decreased (Na++K+)ATPase activity in the same proportion in cardiomyocytes and proximal tubule cells. Type 1 angiotensin II receptor (AT1R) was downregulated by that restriction in both organs, whereas AT2R decreased only in the kidney. The PKC/PKA ratio increased in both tissues and returned to normal values in rats receiving Losartan daily from weaning. Inhibition of the MAPK pathway restored Na+-ATPase activity in both organs. The undernourished rats presented expanded plasma volume, increased heart rate, cardiac hypertrophy, and elevated systolic pressure, which also returned to control levels with Losartan. Such restriction led to electrical cardiac remodeling represented by prolonged ventricular repolarization parameters, induced triggered activity, early after-depolarization and delayed after-depolarization, which were also prevented by Losartan. Conclusion/Significance The mechanisms responsible for these alterations are underpinned by an imbalance in the PKC- and PKA-mediated pathways, with participation of angiotensin receptors and by activation of the MAPK/ERK1/2 pathway. These cellular and molecular alterations culminate in cardiac electric remodeling and in the onset of hypertension in adulthood.
Collapse
Affiliation(s)
- Paulo A. Silva
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Gustavo Monnerat-Cahli
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Amaury Pereira-Acácio
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Ricardo Luzardo
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Luzia S. Sampaio
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Marcia A. Luna-Leite
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucienne S. Lara
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Einicker-Lamas
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Rogério Panizzutti
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline Madeira
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leucio D. Vieira-Filho
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Carmen Castro-Chaves
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Valdilene S. Ribeiro
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Ana D. O. Paixão
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Emiliano Medei
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
47
|
Huang W, Tang L, Cai Y, Zheng Y, Zhang L. Effect and mechanism of the Ang-(1-7) on human mesangial cells injury induced by low density lipoprotein. Biochem Biophys Res Commun 2014; 450:1051-7. [PMID: 24978313 DOI: 10.1016/j.bbrc.2014.06.107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 06/22/2014] [Indexed: 11/26/2022]
Abstract
Hyperlipidemia is an independent risk factor for renal disease, and lipid deposition is associated with glomerulosclerosis. The angiotensin converting enzyme 2-angiotensin-(1-7)-Mas axis (ACE2-Ang-(1-7)-Mas axis) has been reported to participate in lipid metabolic regulation but its mechanism remains unclear. We hypothesized Ang-(1-7) would reduce lipid uptake in human mesangial cells (HMCs) by regulating the low density lipoprotein receptor-sterol regulatory element binding proteins 2-SREBP cleavage activating protein (LDLr-SREBP2-SCAP) negative feedback system, and improve glomerulosclerosis by regulating the transforming growth factor-β1 (TGF-β1). In this study we found that ACE2 was undetected in HMCs. The administration of LDL caused normal LDLr-SREBPs-SCAP negative feedback effect. Exogenous Ang-(1-7) enhanced this negative feedback effect via down-regulating LDLr, SREBP2, and SCAP expression, and effectively inhibited LDL-induced lipid deposition and cholesterol increases. This enhanced inhibitory effect was reversed by the Mas receptor antagonist A-779. Meanwhile, Ang-(1-7) significantly decreased the high LDL-induced production of TGF-β1, an effect blocked by A-779. Interestingly, HMCs treated with Ang-(1-7) alone activated the TGF-β1 expression. Our results suggested that Ang-(1-7) inhibits LDL accumulation and decreases cholesterol levels via modulating the LDLr-SREBPs-SCAP negative feedback system through the Mas receptor. Moreover, Ang-(1-7) exhibits a dual regulatory effect on TGF-β1 in HMCs.
Collapse
Affiliation(s)
- Wenhan Huang
- Department of Nephrology of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Lin Tang
- Department of Nephrology of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Ying Cai
- Department of Nephrology of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yaning Zheng
- Department of Nephrology of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ling Zhang
- Department of Nephrology of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
48
|
Patel SK, Velkoska E, Freeman M, Wai B, Lancefield TF, Burrell LM. From gene to protein-experimental and clinical studies of ACE2 in blood pressure control and arterial hypertension. Front Physiol 2014; 5:227. [PMID: 25009501 PMCID: PMC4067757 DOI: 10.3389/fphys.2014.00227] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/02/2014] [Indexed: 12/13/2022] Open
Abstract
Hypertension is a major risk factor for stroke, coronary events, heart and renal failure, and the renin-angiotensin system (RAS) plays a major role in its pathogenesis. Within the RAS, angiotensin converting enzyme (ACE) converts angiotensin (Ang) I into the vasoconstrictor Ang II. An “alternate” arm of the RAS now exists in which ACE2 counterbalances the effects of the classic RAS through degradation of Ang II, and generation of the vasodilator Ang 1-7. ACE2 is highly expressed in the heart, blood vessels, and kidney. The catalytically active ectodomain of ACE2 undergoes shedding, resulting in ACE2 in the circulation. The ACE2 gene maps to a quantitative trait locus on the X chromosome in three strains of genetically hypertensive rats, suggesting that ACE2 may be a candidate gene for hypertension. It is hypothesized that disruption of tissue ACE/ACE2 balance results in changes in blood pressure, with increased ACE2 expression protecting against increased blood pressure, and ACE2 deficiency contributing to hypertension. Experimental hypertension studies have measured ACE2 in either the heart or kidney and/or plasma, and have reported that deletion or inhibition of ACE2 leads to hypertension, whilst enhancing ACE2 protects against the development of hypertension, hence increasing ACE2 may be a therapeutic option for the management of high blood pressure in man. There have been relatively few studies of ACE2, either at the gene or the circulating level in patients with hypertension. Plasma ACE2 activity is low in healthy subjects, but elevated in patients with cardiovascular risk factors or cardiovascular disease. Genetic studies have investigated ACE2 gene polymorphisms with either hypertension or blood pressure, and have produced largely inconsistent findings. This review discusses the evidence regarding ACE2 in experimental hypertension models and the association between circulating ACE2 activity and ACE2 polymorphisms with blood pressure and arterial hypertension in man.
Collapse
Affiliation(s)
- Sheila K Patel
- Department of Medicine, Austin Health, University of Melbourne Heidelberg, VIC, Australia
| | - Elena Velkoska
- Department of Medicine, Austin Health, University of Melbourne Heidelberg, VIC, Australia
| | - Melanie Freeman
- Department of Medicine, Austin Health, University of Melbourne Heidelberg, VIC, Australia
| | - Bryan Wai
- Department of Medicine, Austin Health, University of Melbourne Heidelberg, VIC, Australia ; Department of Cardiology, Austin Health, University of Melbourne Heidelberg, VIC, Australia
| | - Terase F Lancefield
- Department of Medicine, Austin Health, University of Melbourne Heidelberg, VIC, Australia
| | - Louise M Burrell
- Department of Medicine, Austin Health, University of Melbourne Heidelberg, VIC, Australia ; Department of Cardiology, Austin Health, University of Melbourne Heidelberg, VIC, Australia ; Department of Cardiology, The Northern Hospital, University of Melbourne Epping, VIC, Australia
| |
Collapse
|
49
|
Meng W, Zhao W, Zhao T, Liu C, Chen Y, Liu H, Sun Y. Autocrine and paracrine function of Angiotensin 1-7 in tissue repair during hypertension. Am J Hypertens 2014; 27:775-82. [PMID: 24429674 DOI: 10.1093/ajh/hpt270] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Angiotensin-converting enzyme 2 (ACE2) cleaves angiotensin (Ang) II to generate Ang1-7, which mediates cellular actions through Mas receptors (MasR). Hypertension is accompanied by high or low circulating AngII levels and cardiac/renal injury. The purpose of this study is to explore (i) whether circulating AngII affects ACE2/MasR expressions in the hypertensive heart and kidney; and (ii) whether Ang1-7 regulates cardiac repair/remodeling responses through MasR during hypertension. METHODS In the first portion of the study, rats received either an AngII infusion (400ng/kg/min) for 4 weeks, leading to hypertension with high circulating AngII, or an aldosterone (ALDO, 0.75 μg/h) infusion for 4 weeks, leading to hypertension with low/normal circulating AngII. Cardiac and renal ACE2/MasR expressions were examined. We found that cardiac ACE2 was increased and MasR attenuated in both AngII and ALDO groups. However, renal ACE2 and MasR remained unchanged in both AngII- and ALDO-treated animals. RESULTS In the second portion, rats received AngII infusion with/without MasR antagonist (A779, 1mg/kg/day) for 4 weeks. The roles of MasR blockade in cardiac inflammation, fibrosis, apoptosis, and ventricular function were examined. Chronic AngII infusion caused scattered cardiac injuries, and A779 cotreatment exacerbated cardiac injury, resulting in aggravated inflammatory, fibrogenic, and apoptotic responses compared with the AngII group. Cardiac function, however, was unaltered in the AngII and A779 groups. CONCLUSIONS ACE2 and MasR expressions in the hypertensive heart and kidney are not regulated by circulating AngII levels. Ang1-7 is involved in multiple repair responses, suggesting that therapeutic strategies aimed at administering Ang1-7 hold potential for the management of cardiac remodeling.
Collapse
Affiliation(s)
- Weixin Meng
- Division of Cardiac Surgery, Department of Surgery, First Affiliate Hospital of Harbin Medical University, Harbin, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, Zhang K, Zhang C. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol 2014; 11:413-26. [PMID: 24776703 PMCID: PMC7097196 DOI: 10.1038/nrcardio.2014.59] [Citation(s) in RCA: 300] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) 2 and its product angiotensin 1–7 are thought to have effects that counteract the adverse actions of other, better-known renin–angiotensin system (RAS) components Numerous experimental studies have suggested that ACE2 and angiotensin 1–7 have notable protective effects in the heart and blood vessels ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the functional importance and signalling mechanisms of angiotensin-1–7-induced actions remain unclear New pharmacological interventions targeting ACE2 are expected to be useful in clinical treatment of cardiovascular disease, especially those associated with overactivation of the conventional RAS More studies, especially randomized controlled clinical trials, are needed to clearly delineate the benefits of therapies targeting angiotensin 1–7 actions
Angiotensin-converting enzyme 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of the better-known members of the renin–angiotensin system and might, therefore, be useful therapeutic targets in patients with cardiovascular disease. Professor Jiang and colleagues review the evidence for the potential roles of these proteins in various cardiovascular conditions, including hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes. The renin–angiotensin system (RAS) has pivotal roles in the regulation of normal physiology and the pathogenesis of cardiovascular disease. Angiotensin-converting enzyme (ACE) 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of other, better known and understood, members of the RAS. The physiological and pathological importance of ACE2 and angiotensin 1–7 in the cardiovascular system are not completely understood, but numerous experimental studies have indicated that these components have protective effects in the heart and blood vessels. Here, we provide an overview on the basic properties of ACE2 and angiotensin 1–7 and a summary of the evidence from experimental and clinical studies of various pathological conditions, such as hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes mellitus. ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the relevant functions and signalling mechanisms of actions induced by angiotensin 1–7 have not been conclusively determined. The ACE2–angiotensin 1–7 pathway, however, might provide a useful therapeutic target for the treatment of cardiovascular disease, especially in patients with overactive RAS.
Collapse
Affiliation(s)
- Fan Jiang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Jianmin Yang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Yongtao Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Mei Dong
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Shuangxi Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Fang Fang Liu
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Kai Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| |
Collapse
|