1
|
Goyal A, Jain H, Usman M, Zuhair V, Sulaiman SA, Javed B, Mubbashir A, Abozaid AM, Passey S, Yakkali S. A comprehensive exploration of novel biomarkers for the early diagnosis of aortic dissection. Hellenic J Cardiol 2025; 82:74-85. [PMID: 38909846 DOI: 10.1016/j.hjc.2024.06.006] [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: 02/16/2024] [Revised: 05/23/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
Aortic dissection (AD) is a catastrophic life-threatening cardiovascular emergency with a 1-2% per hour mortality rate post-diagnosis, characterized physiologically by the separation of aortic wall layers. AD initially presents as intense pain that can then radiate to the back, arms, neck, or jaw along with neurological deficits like difficulty in speaking, and unilateral weakness in some patients. This spectrum of clinical features associated with AD is often confused with acute myocardial infarction, hence leading to a delay in AD diagnosis. Cardiac and vascular biomarkers are structural proteins and microRNAs circulating in the bloodstream that correlate to tissue damage and their levels become detectable even before symptom onset. Timely diagnosis of AD using biomarkers, in combination with advanced imaging diagnostics, will significantly improve prognosis by allowing earlier vascular interventions. This comprehensive review aims to investigate emerging biomarkers in the diagnosis of AD, as well as provide future directives for creating advanced diagnostic tools and imaging techniques.
Collapse
Affiliation(s)
- Aman Goyal
- Department of Internal Medicine, Seth GS Medical College and KEM Hospital, Mumbai, India.
| | - Hritvik Jain
- All India Institute of Medical Sciences (AIIMS), Jodhpur, India.
| | | | | | | | - Binish Javed
- Atal Bihari Vajpayee Institute of Medical Sciences & Dr Ram Manohar Lohia Hospital, New Delhi, India.
| | | | | | - Siddhant Passey
- Department of Internal Medicine, University of Connecticut Health Center, Connecticut, USA.
| | - Shreyas Yakkali
- Department of Internal Medicine, NYC Health+Hospitals / Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
2
|
Yang G, Khan A, Liang W, Xiong Z, Stegbauer J. Aortic aneurysm: pathophysiology and therapeutic options. MedComm (Beijing) 2024; 5:e703. [PMID: 39247619 PMCID: PMC11380051 DOI: 10.1002/mco2.703] [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: 12/19/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Aortic aneurysm (AA) is an aortic disease with a high mortality rate, and other than surgery no effective preventive or therapeutic treatment have been developed. The renin-angiotensin system (RAS) is an important endocrine system that regulates vascular health. The ACE2/Ang-(1-7)/MasR axis can antagonize the adverse effects of the activation of the ACE/Ang II/AT1R axis on vascular dysfunction, atherosclerosis, and the development of aneurysms, thus providing an important therapeutic target for the prevention and treatment of AA. However, products targeting the Ang-(1-7)/MasR pathway still lack clinical validation. This review will outline the epidemiology of AA, including thoracic, abdominal, and thoracoabdominal AA, as well as current diagnostic and treatment strategies. Due to the highest incidence and most extensive research on abdominal AA (AAA), we will focus on AAA to explain the role of the RAS in its development, the protective function of Ang-(1-7)/MasR, and the mechanisms involved. We will also describe the roles of agonists and antagonists, suggest improvements in engineering and drug delivery, and provide evidence for Ang-(1-7)/MasR's clinical potential, discussing risks and solutions for clinical use. This study will enhance our understanding of AA and offer new possibilities and promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Guang Yang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Institute of Translational Medicine Shenzhen Second People's Hospital The First Affiliated Hospital of Shenzhen University Shenzhen China
- Department of Life Sciences Yuncheng University Yuncheng China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Abbas Khan
- Department of Nutrition and Health Promotion University of Home Economics Lahore Pakistan Lahore Pakistan
| | - Wei Liang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Zibo Xiong
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Johannes Stegbauer
- Department of Nephrology Medical Faculty University Hospital Düsseldorf Heinrich Heine University Düsseldorf Düsseldorf Germany
| |
Collapse
|
3
|
Cao Z, Gao J, Wu J, Zheng Y. The Impact of COVID-19 Infection on Abdominal Aortic Aneurysms: Mechanisms and Clinical Implications. Cardiovasc Ther 2024; 2024:7288798. [PMID: 39742024 PMCID: PMC11300061 DOI: 10.1155/2024/7288798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 01/03/2025] Open
Abstract
Background: The COVID-19 virus not only has significant pathogenicity but also influences the progression of many diseases, altering patient prognosis. Cardiovascular diseases, particularly aortic aneurysms, are among the most life-threatening conditions. Main Idea: COVID-19 infection is reported to accelerate the progression of abdominal aortic aneurysms (AAAs) and increase the risk of rupture; however, a comprehensive understanding of the underlying mechanisms remains elusive. This article primarily reviews the relevant foundational research, focusing on disruptions in the renin-angiotensin-aldosterone system (RAAS), immune system activation, and coagulation disorders. Furthermore, we summarize related clinical research, including the epidemiology of aortic aneurysms during the pandemic and specific case studies. Conclusion: COVID-19 infection can influence the onset and progression of aortic aneurysms by affecting the RAAS, triggering inflammation and immune dysregulation in the arterial wall, and inducing a hypercoagulation state. It is crucial to comprehensively understand the impact of pandemic viral infections on aortic diseases at the foundational and clinical levels, thereby identifying potential preventative or therapeutic approaches and preparing for potential future outbreaks.
Collapse
Affiliation(s)
- Zenghan Cao
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianhang Gao
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianqiang Wu
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Institute of Clinical MedicineNational Infrastructure for Translational MedicinePeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Complex Severe and Rare DiseasePeking Union Medical College Hospital, Beijing 100730, China
| | - Yuehong Zheng
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
4
|
Okuno N, Maruyama S, Wada D, Komemushi A, Shimazu H, Kanayama S, Saito F, Nakamori Y, Kuwagata Y. Retroperitoneal hemorrhage due to ruptured artery induced by median arcuate ligament syndrome in patients with COVID-19: A case series. Acute Med Surg 2024; 11:e70015. [PMID: 39575224 PMCID: PMC11578928 DOI: 10.1002/ams2.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/20/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Background Median arcuate ligament syndrome describes a clinical presentation associated with direct compression of the celiac artery by the median arcuate ligament. Decreased blood flow from the celiac artery and increased flow from superior mesenteric artery causes blood flow in the pancreatic arcade to increase, and aneurysms can form. We report our experience with six cases of retroperitoneal hemorrhage induced by median arcuate ligament syndrome in patients during the COVID-19 period. Case Presentation The time from the onset of COVID-19 to that of abdominal pain in the patients ranged from 3 to 9 days. None of the patients required oxygen for COVID-19. We used contrast-enhanced computed tomography to diagnose retroperitoneal hemorrhage and formation of a visceral pseudoaneurysm or aneurysm due to median arcuate ligament syndrome. Five patients underwent transcatheter arterial embolization. One patient suffered complications of duodenal stenosis and another suffered portal vein thrombosis. Conclusions Retroperitoneal hemorrhage associated with median arcuate ligament syndrome is rare. However, it might be triggered by COVID-19 disease, resulting in arterial disruption and hemorrhage.
Collapse
Affiliation(s)
- Nao Okuno
- Department of Emergency and Critical Care MedicineKansai Medical University Medical CenterMoriguchiOsakaJapan
| | - Shuhei Maruyama
- Department of Emergency and Critical Care MedicineKansai Medical University Medical CenterMoriguchiOsakaJapan
| | - Daiki Wada
- Department of Emergency and Critical Care MedicineKansai Medical University Medical CenterMoriguchiOsakaJapan
| | - Atsushi Komemushi
- Department of RadiologyKansai Medical University Medical CenterMoriguchiOsakaJapan
| | - Haruka Shimazu
- Department of Emergency and Critical Care MedicineKansai Medical University Medical CenterMoriguchiOsakaJapan
| | - Shuji Kanayama
- Department of Emergency and Critical Care MedicineKansai Medical University Medical CenterMoriguchiOsakaJapan
| | - Fukuki Saito
- Department of Emergency and Critical Care MedicineKansai Medical University Medical CenterMoriguchiOsakaJapan
| | - Yasushi Nakamori
- Department of Emergency and Critical Care MedicineKansai Medical University Medical CenterMoriguchiOsakaJapan
| | - Yasuyuki Kuwagata
- Department of Emergency and Critical Care MedicineKansai Medical University HospitalHirakataOsakaJapan
| |
Collapse
|
5
|
Hulme J. COVID-19 and Diarylamidines: The Parasitic Connection. Int J Mol Sci 2023; 24:6583. [PMID: 37047556 PMCID: PMC10094973 DOI: 10.3390/ijms24076583] [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: 03/15/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
As emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants (Omicron) continue to outpace and negate combinatorial vaccines and monoclonal antibody therapies targeting the spike protein (S) receptor binding domain (RBD), the appetite for developing similar COVID-19 treatments has significantly diminished, with the attention of the scientific community switching to long COVID treatments. However, treatments that reduce the risk of "post-COVID-19 syndrome" and associated sequelae remain in their infancy, particularly as no established criteria for diagnosis currently exist. Thus, alternative therapies that reduce infection and prevent the broad range of symptoms associated with 'post-COVID-19 syndrome' require investigation. This review begins with an overview of the parasitic-diarylamidine connection, followed by the renin-angiotensin system (RAS) and associated angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSSR2) involved in SARS-CoV-2 infection. Subsequently, the ability of diarylamidines to inhibit S-protein binding and various membrane serine proteases associated with SARS-CoV-2 and parasitic infections are discussed. Finally, the roles of diarylamidines (primarily DIZE) in vaccine efficacy, epigenetics, and the potential amelioration of long COVID sequelae are highlighted.
Collapse
Affiliation(s)
- John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
6
|
Zhang M, Sui W, Zhang M, Zhang Y, Zhang C. An animal model of EPO-induced abdominal aortic aneurysm in WT and Apoe -/- mice. STAR Protoc 2023; 4:101929. [PMID: 36527710 PMCID: PMC9792537 DOI: 10.1016/j.xpro.2022.101929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/21/2022] [Accepted: 11/20/2022] [Indexed: 12/23/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a potentially fatal vascular disease, but the underlying mechanisms remain obscure. Here, we provide a protocol using erythropoietin (EPO) to induce the formation of AAA in both wild-type (WT) and apolipoprotein E (Apoe-/-) mice. We describe the dose, manner, and timing of EPO administration. We also detail mice dissection, aorta isolation, and histological analysis. The animal model of EPO-induced AAA provides a useful tool for exploring the mechanism of AAA in experimental studies. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2021).1.
Collapse
Affiliation(s)
- Meng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenhai Sui
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Meng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
7
|
Sawada H, Lu HS, Cassis LA, Daugherty A. Twenty Years of Studying AngII (Angiotensin II)-Induced Abdominal Aortic Pathologies in Mice: Continuing Questions and Challenges to Provide Insight Into the Human Disease. Arterioscler Thromb Vasc Biol 2022; 42:277-288. [PMID: 35045728 PMCID: PMC8866209 DOI: 10.1161/atvbaha.121.317058] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AngII (angiotensin II) infusion in mice has been used to provide mechanistic insight into human abdominal aortic aneurysms for over 2 decades. This is a technically facile animal model that recapitulates multiple facets of the human disease. Although numerous publications have reported abdominal aortic aneurysms with AngII infusion in mice, there remain many fundamental unanswered questions such as uniformity of describing the pathological characteristics and which cell type is stimulated by AngII to promote abdominal aortic aneurysms. Extrapolation of the findings to provide insight into the human disease has been hindered by the preponderance of studies designed to determine the effects on initiation of abdominal aortic aneurysms, rather than a more clinically relevant scenario of determining efficacy on the established disease. The purpose of this review is to enhance understanding of AngII-induced abdominal aortic pathologies in mice, thereby providing greater insight into the human disease.
Collapse
Affiliation(s)
- Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY,Saha Aortic Center, University of Kentucky, Lexington, KY,Department of Physiology, University of Kentucky, Lexington, KY
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY,Saha Aortic Center, University of Kentucky, Lexington, KY,Department of Physiology, University of Kentucky, Lexington, KY
| | - Lisa A. Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY,Saha Aortic Center, University of Kentucky, Lexington, KY,Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
8
|
Li T, Huang HY, Wang H, Gao CC, Liang H, Deng CL, Zhao X, Han YL, Zhou ML. Restoration of Brain Angiotensin-Converting Enzyme 2 Alleviates Neurological Deficits after Severe Traumatic Brain Injury via Mitigation of Pyroptosis and Apoptosis. J Neurotrauma 2021; 39:423-434. [PMID: 34861788 DOI: 10.1089/neu.2021.0382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Clinically, the renin-angiotensin-aldosterone system is intensely activated in moderate to severe traumatic brain injury (TBI) patients. Increased angiotensin II in circulatory blood after TBI can enter the brain through the disrupted blood-brain barrier. Angiotensin-converting enzyme 2 (ACE2) is an enzyme that metabolizes angiotensin II into angiotensin (1-7), which has been shown to have neuroprotective results. However, the expression and role of ACE2 in the brain after TBI remains elusive. We found that ACE2 protein abundance was downregulated around the contusional area in the brains of both humans and mice. Endogenous ACE2 was expressed in neurons, astrocytes, and microglia in the cortex of the mouse brain. Administration of recombinant human ACE2 intracerebroventricularly alleviated neurological defects after TBI in mice. Treatment of recombinant human ACE2 suppressed TBI-induced increase of angiotensin II and the decrease of angiotensin (1-7) in the brain, mitigated neural cell death, reduced the activation of NLRP3 and Caspase3, decreased phosphorylation of mitogen-activated protein kinases, and nuclear factor kappa B, and reduced inflammatory cytokines TNF-α and IL-1β. Administration of ACE2 enzyme activator diminazene aceturate intraperitoneally rescued downregulation of ACE2 enzymatic activity and protein abundance in the brain. Diminazene aceturate treatment once per day in the acute stage after TBI alleviated long-term cognitive defects and neuronal loss in mice. Collectively, these results indicated that restoration of ACE2 alleviated neurological deficits after TBI by mitigation of pyroptosis and apoptosis.
Collapse
Affiliation(s)
- Tao Li
- Nanjing Medical University, 12461, Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine,, Nanjing, Jiangsu, China;
| | - Han-Yu Huang
- Nanjing Medical University, 12461, Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine,, Nanjing, Jiangsu, China;
| | - Handong Wang
- Nanjing Medical University, 12461, neurosurgery of jinling hospital, Nanjing, Jiangsu, China;
| | - Chao-Chao Gao
- Nanjing Medical University, 12461, Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine,, Nanjing, Jiangsu, China;
| | - Hui Liang
- Nanjing University, 12581, Department of Neurosurgery, Jinling Hospital, Nanjing, Jiangsu, China;
| | - Chu-Lei Deng
- Nanjing Jinling Hospital, 144990, Department of Neurosurgery, Nanjing, Jiangsu, China;
| | - Xin Zhao
- Nanjing University, 12581, Department of Neurosurgery, Jinling Hospital, Nanjing, Jiangsu, China;
| | - Yan-Lin Han
- Nanjing University, 12581, Department of Neurosurgery, Jinling Hospital, Nanjing, Jiangsu, China;
| | - Meng-Liang Zhou
- Nanjing University, 12581, Department of Neurosurgery, Jinling Hospital, Nanjing, Jiangsu, China;
| |
Collapse
|
9
|
Xu B, Li G, Guo J, Ikezoe T, Kasirajan K, Zhao S, Dalman RL. Angiotensin-converting enzyme 2, coronavirus disease 2019, and abdominal aortic aneurysms. J Vasc Surg 2021; 74:1740-1751. [PMID: 33600934 PMCID: PMC7944865 DOI: 10.1016/j.jvs.2021.01.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the etiologic agent of the current, world-wide coronavirus disease 2019 (COVID-19) pandemic. Angiotensin-converting enzyme 2 (ACE2) is the SARS-CoV-2 host entry receptor for cellular inoculation and target organ injury. We reviewed ACE2 expression and the role of ACE2-angiotensin 1-7-Mas receptor axis activity in abdominal aortic aneurysm (AAA) pathogenesis to identify potential COVID-19 influences on AAA disease pathogenesis. METHODS A comprehensive literature search was performed on PubMed, National Library of Medicine. Key words included COVID-19, SARS-CoV-2, AAA, ACE2, ACE or angiotensin II type 1 (AT1) receptor inhibitor, angiotensin 1-7, Mas receptor, age, gender, respiratory diseases, diabetes, and autoimmune diseases. Key publications on the epidemiology and pathogenesis of COVID-19 and AAAs were identified and reviewed. RESULTS All vascular structural cells, including endothelial and smooth muscle cells, fibroblasts, and pericytes express ACE2. Cigarette smoking, diabetes, chronic obstructive pulmonary disease, lupus, certain types of malignancies, and viral infection promote ACE2 expression and activity, with the magnitude of response varying by sex and age. Genetic deficiency of AT1 receptor, or pharmacologic ACE or AT1 inhibition also increases ACE2 and its catalytic product angiotensin 1-7. Genetic ablation or pharmacologic inhibition of ACE2 or Mas receptor augments, whereas ACE2 activation or angiotensin 1-7 treatment attenuates, progression of experimental AAAs. The potential influences of SARS-CoV-2 on AAA pathogenesis include augmented ACE-angiotensin II-AT1 receptor activity resulting from decreased reciprocal ACE2-angiotensin 1-7-Mas activation; increased production of proaneurysmal mediators stimulated by viral spike proteins in ACE2-negative myeloid cells or by ACE2-expressing vascular structural cells; augmented local or systemic cross-talk between viral targeted nonvascular, nonleukocytic ACE2-expressing cells via ligand recognition of their cognate leukocyte receptors; and hypoxemia and increased systemic inflammatory tone experienced during severe COVID-19 illness. CONCLUSIONS COVID-19 may theoretically influence AAA disease through multiple SARS-CoV-2-induced mechanisms. Further investigation and clinical follow-up will be necessary to determine whether and to what extent the COVID-19 pandemic will influence the prevalence, progression, and lethality of AAA disease in the coming decade.
Collapse
Affiliation(s)
- Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif.
| | - Gang Li
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Jia Guo
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Toru Ikezoe
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| | | | - Sihai Zhao
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Ronald L Dalman
- Department of Surgery, Stanford University School of Medicine, Stanford, Calif
| |
Collapse
|
10
|
Miličić Stanić B, Maddox S, de Souza AMA, Wu X, Mehranfard D, Ji H, Speth RC, Sandberg K. Male bias in ACE2 basic science research: missed opportunity for discovery in the time of COVID-19. Am J Physiol Regul Integr Comp Physiol 2021; 320:R925-R937. [PMID: 33848207 PMCID: PMC8203415 DOI: 10.1152/ajpregu.00356.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 11/22/2022]
Abstract
Throughout the world, including the United States, men have worse outcomes from COVID-19 than women. SARS-CoV-2, the causative virus of the COVID-19 pandemic, uses angiotensin-converting enzyme 2 (ACE2) to gain cellular entry. ACE2 is a member of the renin-angiotensin system (RAS) and plays an important role in counteracting the harmful effects mediated by the angiotensin type 1 receptor. Therefore, we conducted Ovid MEDLINE and Embase database searches of basic science studies investigating the impact of the biological variable of sex on ACE2 expression and regulation from 2000, the year ACE2 was discovered, through December 31, 2020. Out of 2,131 publications, we identified 853 original research articles on ACE2 conducted in primary cells, tissues, and/or whole mammals excluding humans. The majority (68.7%) of these studies that cited the sex of the animal were conducted in males, while 11.2% were conducted solely in females; 9.26% compared ACE2 between the sexes, while 10.8% did not report the sex of the animals used. General findings are that sex differences are tissue-specific and when present, are dependent upon gonadal state. Renal, cardiac, and adipose ACE2 is increased in both sexes under experimental conditions that model co-morbidities associated with worse COVID-19 outcomes including hypertension, obesity, and renal and cardiovascular diseases; however, ACE2 protein was generally higher in the males. Studies in Ace2 knockout mice indicate ACE2 plays a greater role in protecting the female from developing hypertension than the male. Studying the biological variable of sex in ACE2 research provides an opportunity for discovery in conditions involving RAS dysfunction and will shed light on sex differences in COVID-19 severity.
Collapse
Affiliation(s)
- Branka Miličić Stanić
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Sydney Maddox
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Aline M A de Souza
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Xie Wu
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Danial Mehranfard
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida
| | - Hong Ji
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Robert C Speth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, District of Columbia
| | - Kathryn Sandberg
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| |
Collapse
|
11
|
AlSiraj Y, Thatcher SE, Liang CL, Ali H, Ensor M, Cassis LA. Therapeutic Assessment of Combination Therapy with a Neprilysin Inhibitor and Angiotensin Type 1 Receptor Antagonist on Angiotensin II-Induced Atherosclerosis, Abdominal Aortic Aneurysms, and Hypertension. J Pharmacol Exp Ther 2021; 377:326-335. [PMID: 33707301 PMCID: PMC8140395 DOI: 10.1124/jpet.121.000525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 12/21/2022] Open
Abstract
Combined neprilysin (NEP) inhibition (sacubitril) and angiotensin type 1 receptor (AT1R) antagonism (valsartan) is used in the treatment of congestive heart failure and is gaining interest for other angiotensin II (AngII)-related cardiovascular diseases. In addition to heart failure, AngII promotes hypertension, atherosclerosis, and abdominal aortic aneurysms (AAAs). Similarly, NEP substrates or products have broad effects on the cardiovascular system. In this study, we examined NEP inhibition (with sacubitril) and AT1R antagonism (with valsartan) alone or in combination on AngII-induced hypertension, atherosclerosis, or AAAs in male low-density lipoprotein receptor-deficient mice. Preliminary studies assessed drug delivery via osmotic minipumps for simultaneous release of sacubitril and/or valsartan with AngII over 28 days. Mice were infused with AngII (1000 ng/kg per minute) in the absence (vehicle) or presence of sacubitril (1, 6, or 9 mg/kg per day), valsartan (0.3, 0.5, 1, 6, or 20 mg/kg per day), or the combination thereof (1 and 0.3, or 9 or 0.5 mg/kg per day of sacubitril and valsartan, respectively). Plasma AngII and renin concentrations increased 4-fold at higher valsartan doses, indicative of removal of AngII negative feedback on renin. Sacubitril doubled plasma AngII concentrations at lower doses (1 mg/kg per day). Valsartan dose-dependently decreased systolic blood pressure, aortic atherosclerosis, and AAAs of AngII-infused mice, whereas sacubitril had no effect on atherosclerosis or AAAs but reduced blood pressure of AngII-infused mice. Combination therapy with sacubitril and valsartan did not provide additive benefits. These results suggest limited effects of combination therapy with NEP inhibition and AT1R antagonism against AngII-induced hypertension, atherosclerosis, or AAAs. SIGNIFICANCE STATEMENT: The combination of valsartan (angiotensin type 1 receptor antagonist) and sacubitril (neprilysin inhibitor) did not provide benefit above valsartan alone on AngII-induced hypertension, atherosclerosis, or abdominal aortic aneurysms in low-density lipoprotein receptor-deficient male mice. These results do not support this drug combination in therapy of these AngII-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Yasir AlSiraj
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Sean E Thatcher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Ching Ling Liang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Heba Ali
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Mark Ensor
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
12
|
Shi J, Guo J, Li Z, Xu B, Miyata M. Importance of NLRP3 Inflammasome in Abdominal Aortic Aneurysms. J Atheroscler Thromb 2021; 28:454-466. [PMID: 33678767 PMCID: PMC8193780 DOI: 10.5551/jat.rv17048] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic inflammatory degenerative aortic disease, which particularly affects older people. Nucleotide-binding oligomerization domain-like receptor family protein 3 (NLRP3) inflammasome is a multi-protein complex and mediates inflammatory responses by activating caspase 1 for processing premature interleukin (IL)-1β and IL-18. In this review, we first summarize the principle of NLRP3 inflammasome activation and the functionally distinct classes of small molecule NLRP3 inflammasome inhibitors. Next, we provide a comprehensive literature review on the expression of NLRP3 inflammasome effector mediators (IL-1β and IL-18) and components (caspase 1, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and NLRP3) in clinical and experimental AAAs. Finally, we discuss the influence of genetic deficiency or pharmacological inhibition of individual effector mediators and components of NLRP3 inflammasome on experimental AAAs. Accumulating clinical and experimental evidence suggests that NLRP3 inflammasome may be a promise therapeutic target for developing pharmacological strategies for clinical AAA management.
Collapse
Affiliation(s)
- Jinyun Shi
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan, Shanxi Province, P. R. China
| | - Jia Guo
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan, Shanxi Province, P. R. China
| | - Zhidong Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi Province, P. R. China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masaaki Miyata
- School of Health Science, Faculty of Medicine, Kagoshima University, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
13
|
Poznyak AV, Bezsonov EE, Eid AH, Popkova TV, Nedosugova LV, Starodubova AV, Orekhov AN. ACE2 Is an Adjacent Element of Atherosclerosis and COVID-19 Pathogenesis. Int J Mol Sci 2021; 22:ijms22094691. [PMID: 33946649 PMCID: PMC8124184 DOI: 10.3390/ijms22094691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/17/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a highly contagious new infection caused by the single-stranded RNA Sars-CoV-2 virus. For the first time, this infection was recorded in December 2019 in the Chinese province of Wuhan. The virus presumably crossed the interspecies barrier and passed to humans from a bat. Initially, the disease was considered exclusively in the context of damage to the respiratory system, but it quickly became clear that the disease also entails serious consequences from various systems, including the cardiovascular system. Among these consequences are myocarditis, myocardial damage, subsequent heart failure, myocardial infarction, and Takotsubo syndrome. On the other hand, clinical data indicate that the presence of chronic diseases in a patient aggravates the course and outcome of coronavirus infection. In this context, the relationship between COVID-19 and atherosclerosis, a condition preceding cardiovascular disease and other disorders of the heart and blood vessels, is particularly interesting. The renin-angiotensin system is essential for the pathogenesis of both coronavirus disease and atherosclerosis. In particular, it has been shown that ACE2, an angiotensin-converting enzyme 2, plays a key role in Sars-CoV-2 infection due to its receptor activity. It is noteworthy that this enzyme is important for the normal functioning of the cardiovascular system. Disruptions in its production and functioning can lead to various disorders, including atherosclerosis.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| | - Evgeny E. Bezsonov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Tatyana V. Popkova
- V.A. Nasonova Institute of Rheumatology, 34A Kashirskoye Shosse, 115522 Moscow, Russia;
| | - Ludmila V. Nedosugova
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of the Russian Federation 8/2 Trubenskaya Street, 119991 Moscow, Russia;
| | - Antonina V. Starodubova
- Federal Research Centre for Nutrition, Biotechnology and Food Safety, 2/14 Ustinsky Passage, 109240 Moscow, Russia;
- Pirogov Russian National Research Medical University, 1 Ostrovitianov Street, 117997 Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
14
|
Kasirajan K. Acute upper extremity ischemia and symptomatic popliteal artery aneurysm secondary to coronavirus disease 2019. JOURNAL OF VASCULAR SURGERY CASES INNOVATIONS AND TECHNIQUES 2021; 7:267-270. [PMID: 33681541 PMCID: PMC7923860 DOI: 10.1016/j.jvscit.2021.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/21/2021] [Indexed: 12/01/2022]
Abstract
We report the cases of two patients with coronavirus disease 2019 (COVID-19). One patient had presented with acute right upper extremity ischemia (axillary artery thrombosis) and one patient with a symptomatic popliteal artery aneurysm (7.5 × 7.2 cm). Both patients had tested positive for COVID-19 but had no systemic symptoms. The patients were successfully treated using percutaneous techniques and subsequently discharged with oral anticoagulation therapy. A review of the pathogenesis of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2)–related arterial thrombosis and aneurysmal disease was performed and discussed.
Collapse
Affiliation(s)
- Karthikeshwar Kasirajan
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Stanford, Pleasanton, Calif
| |
Collapse
|
15
|
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
|
16
|
Liu X, Liu X, Li M, Zhang Y, Chen W, Zhang M, Zhang C, Zhang M. Mechanical Stretch Induces Smooth Muscle Cell Dysfunction by Regulating ACE2 via P38/ATF3 and Post-transcriptional Regulation by miR-421. Front Physiol 2021; 11:540591. [PMID: 33536929 PMCID: PMC7848200 DOI: 10.3389/fphys.2020.540591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 12/08/2020] [Indexed: 12/25/2022] Open
Abstract
Mechanical stretch promotes deregulation of vascular smooth muscle cell (VSMC) functions during hypertension-induced vascular remodeling. ACE2 has a wide range of cardiovascular and renal protective effects. Loss of ACE2 is associated with cardiovascular disease, but little is known about the regulation of its expression, especially by abnormal mechanical stretch during hypertension. The present study was designed to investigate the contribution of ACE2 to vascular remodeling under mechanical stretch and to assess the possible underlying mechanisms. The abdominal aortic constriction model was established to mimic the environment in vivo. FX-5000T Strain Unit provided mechanical stretch in vitro. Overexpression was used to analyze the role of ACE2 played in the proliferation, migration, apoptosis, and collagen metabolism of the VSMCs. RT-qPCR, Western blot, luciferase assay, and ChIP assay were used to elucidate the molecular mechanism of ACE2 expression regulated by stretch. We found that mechanical stretch modulated the expression of the ACE2/Ang-(1–7) and ACE/AngII axis. ACE2 was mechanically sensitive and was involved in the stretch-induced dysfunction of VSMCs. The p38 MAPK/ATF3 pathway and miR-421 participated in the regulation of ACE2. Thus, ACE2 may contribute to the development of vascular remodeling under conditions of mechanical stretch.
Collapse
Affiliation(s)
- Xiaolin Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Xinxin Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Mengmeng Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Weijia Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Mei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Sharma R, Li J, Krishnan S, Richards E, Raizada M, Mohandas R. Angiotensin-converting enzyme 2 and COVID-19 in cardiorenal diseases. Clin Sci (Lond) 2021; 135:1-17. [PMID: 33399851 PMCID: PMC7796300 DOI: 10.1042/cs20200482] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 01/08/2023]
Abstract
The rapid spread of the novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has brought into focus the key role of angiotensin-converting enzyme 2 (ACE2), which serves as a cell surface receptor required for the virus to enter cells. SARS-CoV-2 can decrease cell surface ACE2 directly by internalization of ACE2 bound to the virus and indirectly by increased ADAM17 (a disintegrin and metalloproteinase 17)-mediated shedding of ACE2. ACE2 is widely expressed in the heart, lungs, vasculature, kidney and the gastrointestinal (GI) tract, where it counteracts the deleterious effects of angiotensin II (AngII) by catalyzing the conversion of AngII into the vasodilator peptide angiotensin-(1-7) (Ang-(1-7)). The down-regulation of ACE2 by SARS-CoV-2 can be detrimental to the cardiovascular system and kidneys. Further, decreased ACE2 can cause gut dysbiosis, inflammation and potentially worsen the systemic inflammatory response and coagulopathy associated with SARS-CoV-2. This review aims to elucidate the crucial role of ACE2 both as a regulator of the renin-angiotensin system and a receptor for SARS-CoV-2 as well as the implications for Coronavirus disease 19 and its associated cardiovascular and renal complications.
Collapse
Affiliation(s)
- Ravindra K. Sharma
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Jing Li
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Suraj Krishnan
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Elaine M. Richards
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Mohan K. Raizada
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Rajesh Mohandas
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| |
Collapse
|
18
|
Jia H, Yue X, Lazartigues E. ACE2 mouse models: a toolbox for cardiovascular and pulmonary research. Nat Commun 2020; 11:5165. [PMID: 33057007 PMCID: PMC7560817 DOI: 10.1038/s41467-020-18880-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been identified as the host entry receptor for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) responsible for the COVID-19 pandemic. ACE2 is a regulatory enzyme of the renin-angiotensin system and has protective functions in many cardiovascular, pulmonary and metabolic diseases. This review summarizes available murine models with systemic or organ-specific deletion of ACE2, or with overexpression of murine or human ACE2. The purpose of this review is to provide researchers with the genetic tools available for further understanding of ACE2 biology and for the investigation of ACE2 in the pathogenesis and treatment of COVID-19.
Collapse
Affiliation(s)
- Hongpeng Jia
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xinping Yue
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Eric Lazartigues
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA. .,Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA. .,Southeast Louisiana Veterans Health Care Systems, New Orleans, LA, 70119, USA.
| |
Collapse
|
19
|
Silva GM, França-Falcão MS, Calzerra NTM, Luz MS, Gadelha DDA, Balarini CM, Queiroz TM. Role of Renin-Angiotensin System Components in Atherosclerosis: Focus on Ang-II, ACE2, and Ang-1-7. Front Physiol 2020; 11:1067. [PMID: 33013457 PMCID: PMC7494970 DOI: 10.3389/fphys.2020.01067] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/04/2020] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is the leading cause of vascular disease worldwide and contributes significantly to deaths from cardiovascular complications. There is a remarkably close relationship between atherosclerotic plaque formation and the activation of renin-angiotensin system (RAS). However, depending on which RAS pathway is activated, pro- or anti-atherogenic outcomes may be observed. This brief review focuses on the role of three of the most important pieces of RAS axis, angiotensin II (Ang-II), angiotensin converting enzyme type 2 (ACE2), and angiotensin 1-7 (Ang-1-7) and their involvement in atherosclerosis. We focused on the effects of these molecules on vascular function and inflammation, which are important determinants of atherogenesis. Furthermore, we highlighted potential pharmacological approaches to treat this disorder.
Collapse
Affiliation(s)
- Gabriela M Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| | | | | | - Mickael S Luz
- Center of Biotechnology, Federal University of Paraiba, João Pessoa, Brazil
| | | | - Camille M Balarini
- Health Sciences Center, Federal University of Paraiba, João Pessoa, Brazil
| | - Thyago M Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| |
Collapse
|
20
|
Sex differences in cardiovascular actions of the renin-angiotensin system. Clin Auton Res 2020; 30:393-408. [PMID: 32860555 DOI: 10.1007/s10286-020-00720-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) remains a worldwide public health concern despite decades of research and the availability of numerous targeted therapies. While the intrinsic physiological mechanisms regulating cardiovascular function are similar between males and females, marked sex differences have been established in terms of CVD onset, pathophysiology, manifestation, susceptibility, prevalence, treatment responses and outcomes in animal models and clinical populations. Premenopausal females are generally protected from CVD in comparison to men of similar age, with females tending to develop cardiovascular complications later in life following menopause. Emerging evidence suggests this cardioprotection in females is, in part, attributed to sex differences in hormonal regulators, such as the renin-angiotensin system (RAS). To date, research has largely focused on canonical RAS pathways and shown that premenopausal females are protected from cardiovascular derangements produced by activation of angiotensin II pathways. More recently, a vasodilatory arm of the RAS has emerged that is characterized by angiotensin-(1-7) [(Ang-(1-7)], angiotensin-converting enzyme 2 and Mas receptors. Emerging studies provide evidence for a shift towards these cardioprotective Ang-(1-7) pathways in females, with effects modulated by interactions with estrogen. Despite well-established sex differences, female comparison studies on cardiovascular outcomes are lacking at both the preclinical and clinical levels. Furthermore, there are no specific guidelines in place for the treatment of cardiovascular disease in men versus women, including therapies targeting the RAS. This review summarizes current knowledge on sex differences in the cardiovascular actions of the RAS, focusing on interactions with gonadal hormones, emerging data for protective Ang-(1-7) pathways and potential clinical implications for established and novel therapies.
Collapse
|
21
|
Ielapi N, Licastro N, Provenzano M, Andreucci M, Franciscis SD, Serra R. Cardiovascular disease as a biomarker for an increased risk of COVID-19 infection and related poor prognosis. Biomark Med 2020; 14:713-716. [PMID: 32426991 PMCID: PMC7236792 DOI: 10.2217/bmm-2020-0201] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Nicola Ielapi
- Interuniversity Center of Phlebolymphology (CIFL), International Research & Educational Program in Clinical & Experimental Biotechnology at the Department of Medical & Surgical Sciences University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy.,Sapienza University of Rome, Department of Public Health & Infectious Disease, Roma, Italy
| | - Noemi Licastro
- Interuniversity Center of Phlebolymphology (CIFL), International Research & Educational Program in Clinical & Experimental Biotechnology at the Department of Medical & Surgical Sciences University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy.,Department of Medical & Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Michele Provenzano
- Interuniversity Center of Phlebolymphology (CIFL), International Research & Educational Program in Clinical & Experimental Biotechnology at the Department of Medical & Surgical Sciences University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy.,Department of Health Sciences, University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Michele Andreucci
- Interuniversity Center of Phlebolymphology (CIFL), International Research & Educational Program in Clinical & Experimental Biotechnology at the Department of Medical & Surgical Sciences University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy.,Department of Health Sciences, University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Stefano de Franciscis
- Interuniversity Center of Phlebolymphology (CIFL), International Research & Educational Program in Clinical & Experimental Biotechnology at the Department of Medical & Surgical Sciences University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy.,Department of Medical & Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research & Educational Program in Clinical & Experimental Biotechnology at the Department of Medical & Surgical Sciences University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy.,Department of Medical & Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, Catanzaro 88100, Italy
| |
Collapse
|
22
|
Xue F, Yang J, Cheng J, Sui W, Cheng C, Li H, Zhang M, Zhang J, Xu X, Ma J, Lu L, Xu J, Zhang M, Zhang Y, Zhang C. Angiotensin-(1-7) mitigated angiotensin II-induced abdominal aortic aneurysms in apolipoprotein E-knockout mice. Br J Pharmacol 2020; 177:1719-1734. [PMID: 31658493 DOI: 10.1111/bph.14906] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/19/2019] [Accepted: 10/06/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE To test the hypothesis that angiotensin-(1-7) [Ang-(1-7)] may attenuate abdominal aortic aneurysm (AAA) via inhibiting vascular inflammation, extracellular matrix degradation, and smooth muscle cell (SMC) apoptosis, an animal model of AAA was established by angiotensin II (Ang II) infusion to apolipoprotein E-knockout (ApoE-/- ) mice. EXPERIMENTAL APPROACH All mice and cultured SMCs or macrophages were divided into control, Ang II-treated, Ang II + Ang-(1-7)-treated, Ang II + Ang-(1-7) + A779-treated and Ang II + Ang-(1-7) + PD123319-treated groups respectively. In vivo, aortic mechanics and serum lipids were assessed. Ex vivo, AAA were examined by histology, immunohistochemistry and zymography. Cultured cells were analysed by RT-PCR, western blots and TUNEL assays. KEY RESULTS In vivo, Ang-(1-7) reduced the incidence and severity of AAA induced by Ang II infusion, by inhibiting macrophage infiltration, attenuating expression of IL-6, TNF-α, CCL2 and MMP2, and decreasing SMC apoptosis in abdominal aortic tissues. Addition of A779 or PD123319 reversed Ang-(1-7)-mediated beneficial effects on AAA. In vitro, Ang-(1-7) decreased expression of mRNA for IL-6, TNF-α, and CCL2 induced by Ang II in macrophages. In addition, Ang-(1-7) suppressed apoptosis and MMP2 expression and activity in Ang II-treated SMCs. These effects were accompanied by inhibition of the ERK1/2 signalling pathways via Ang-(1-7) stimulation of Mas and AT2 receptors. CONCLUSION AND IMPLICATIONS Ang-(1-7) treatment attenuated Ang II-induced AAA via inhibiting vascular inflammation, extracellular matrix degradation, and SMC apoptosis. Ang-(1-7) may provide a novel and promising approach to the prevention and treatment of AAA.
Collapse
Affiliation(s)
- Fei Xue
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Cheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhai Sui
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Cheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hongxuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xingli Xu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Ma
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jinfeng Xu
- Department of Ultrasound, Shenzhen People's Hospital, Shenzhen, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
23
|
Zhou M, Shi Z, Cai L, Li X, Ding Y, Xie T, Fu W. Circular RNA expression profile and its potential regulative role in human abdominal aortic aneurysm. BMC Cardiovasc Disord 2020; 20:70. [PMID: 32039711 PMCID: PMC7008530 DOI: 10.1186/s12872-020-01374-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 01/31/2020] [Indexed: 11/10/2022] Open
Abstract
Background This study aimed to identify the differentially expressed circular RNAs (circRNAs) between human abdominal aortic aneurysm (AAA) and the control group. Methods High-throughput sequencing was applied to determine the circRNA expression profiles of 4 paired aortic samples. Real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was carried out to testify 6 randomly selected dysregulated circRNAs. Kyoto Encyclopedia of Genes and Genomes and Gene ontology (GO) analysis were conducted for functional annotation of the parental genes. Additionally, interaction networks between circRNA and 5 putative microRNA (miRNA) partners were constructed. Results Finally, 411 differentially expressed circRNAs were discovered, including 266 downregulated and 145 upregulated circRNAs. Compared with the control group, the expression level of hsa (Homo sapiens) _circ_0005360 (LDLR) and hsa_circ_0002168 (TMEM189) were proved significantly lower in the AAA group by qRT-PCR. Regarding upregulated circRNAs, the most enriched GO molecular function, biological process and cellular component terms were poly(A) RNA binding, negative regulation of transcription from RNA polymerase II promoter and nucleoplasm, respectively. Moreover, circRNA/miRNA interaction networks showed that hsa_circ_0005360/miR-181b and hsa_circ_0002168/miR-15a axis might have a regulative role in human AAA. Conclusions This study revealed new circRNAs potentially related to the pathogenesis of AAA. Further experimental studies are warranted to clarify the potential molecular mechanisms.
Collapse
Affiliation(s)
- Min Zhou
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhenyu Shi
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 180 Fenglin Road, Shanghai, 200032, China
| | - Liang Cai
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 180 Fenglin Road, Shanghai, 200032, China
| | - Xu Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 180 Fenglin Road, Shanghai, 200032, China
| | - Yong Ding
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 180 Fenglin Road, Shanghai, 200032, China
| | - Tianchen Xie
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 180 Fenglin Road, Shanghai, 200032, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
24
|
Khoury MK, Liu B. Invited commentary. J Vasc Surg 2019; 70:1668. [DOI: 10.1016/j.jvs.2019.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/04/2019] [Indexed: 10/25/2022]
|
25
|
Stegbauer J, Thatcher SE, Yang G, Bottermann K, Rump LC, Daugherty A, Cassis LA. Mas receptor deficiency augments angiotensin II-induced atherosclerosis and aortic aneurysm ruptures in hypercholesterolemic male mice. J Vasc Surg 2019; 70:1658-1668.e1. [PMID: 30850299 PMCID: PMC6728232 DOI: 10.1016/j.jvs.2018.11.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Previous studies demonstrated that deficiency of angiotensin-converting enzyme 2 (ACE2) augmented angiotensin II (AngII)-induced atherosclerosis and abdominal aortic aneurysm (AAA) formation in hypercholesterolemic mice. Effects of ACE2 deficiency could arise from increased concentrations of its substrate, AngII, or decreased concentrations of its product, angiotensin-(1-7) [Ang-(1-7)]. Infusion of Ang-(1-7), a Mas receptor (MasR) ligand, to hypercholesterolemic male mice reduced AngII-induced atherosclerosis, suggesting a protective role of the Ang-(1-7)/MasR axis. However, it is unclear whether endogenous Ang-(1-7) acts at MasR to influence AngII-induced vascular diseases. The purpose of this study was to define the role of MasR deficiency in AngII-induced atherosclerosis and AAA formation and severity in hypercholesterolemic male mice. METHODS MasR+/+ and MasR-/- male mice on a low-density lipoprotein receptor-deficient (Ldlr-/-) or apolipoprotein E-deficient (Apoe-/-) background were infused with AngII at either 600 or 1000 ng/kg/min by osmotic minipump for 28 days. Atherosclerosis was quantified at study end point as percentage lesion surface area of the aortic arch in Ldlr-/- mice. Abdominal aortic internal diameters were quantified by ultrasound, and maximal external AAA diameters were quantified at study end point. Blood pressure was quantified by radiotelemetry and a tail cuff-based technique. Serum cholesterol concentrations and vascular tissue characterization were examined at study end point. RESULTS MasR deficiency did not influence body weight, systolic blood pressure at baseline and during AngII infusion, or serum cholesterol concentrations in either Apoe-/- or Ldlr-/- mice. MasR deficiency increased AngII-induced atherosclerosis in aortic arches of Ldlr-/- mice (P < .05), associated with increased oxidative stress and apoptosis in aortic root sections (P < .05). MasR deficiency also augmented internal and external AAA diameters and increased aortic ruptures of both Ldlr-/- and Apoe-/- mice (P < .05). These effects were associated with increased elastin breaks and T-lymphocyte and macrophage accumulation into abdominal aortas of AngII-infused MasR-deficient mice (P < .05). CONCLUSIONS These results demonstrate that MasR deficiency augmented AngII-induced atherosclerosis and AAA rupture through mechanisms involving increased oxidative stress, inflammation, and apoptosis, suggesting that MasR activation may provide therapeutic efficacy against vascular diseases.
Collapse
MESH Headings
- Angiotensin I/metabolism
- Angiotensin II/administration & dosage
- Angiotensin II/metabolism
- Animals
- Aorta/pathology
- Aortic Aneurysm, Abdominal/blood
- Aortic Aneurysm, Abdominal/etiology
- Aortic Aneurysm, Abdominal/pathology
- Aortic Rupture/blood
- Aortic Rupture/etiology
- Aortic Rupture/pathology
- Apoptosis/genetics
- Atherosclerosis/blood
- Atherosclerosis/complications
- Cholesterol
- Disease Models, Animal
- Humans
- Male
- Mice
- Mice, Knockout, ApoE
- Oxidative Stress/genetics
- Peptide Fragments/metabolism
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
Collapse
Affiliation(s)
- Johannes Stegbauer
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Sean E Thatcher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Ky.
| | - Guang Yang
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Katharina Bottermann
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Lars Christian Rump
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Alan Daugherty
- Department of Pharmacology and Saha Cardiovascular Research Center, University of Kentucky, Lexington, Ky
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Ky
| |
Collapse
|
26
|
Potential Medication Treatment According to Pathological Mechanisms in Abdominal Aortic Aneurysm. J Cardiovasc Pharmacol 2019; 71:46-57. [PMID: 28953105 DOI: 10.1097/fjc.0000000000000540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a vascular disease with high mortality. Because of the lack of effective medications to stop or reverse the progression of AAA, surgical operation has become the most predominant recommendation of treatment for patients. There are many potential mechanisms, including inflammation, smooth muscle cell apoptosis, extracellular matrix degradation, oxidative stress, and so on, involving in AAA pathogenesis. According to those mechanisms, some potential therapeutic drugs have been proposed and tested in animal models and even in clinical trials. This review focuses on recent advances in both pathogenic mechanisms and potential pharmacologic therapies of AAA.
Collapse
|
27
|
Santos RAS. Genetic Models. ANGIOTENSIN-(1-7) 2019. [PMCID: PMC7120897 DOI: 10.1007/978-3-030-22696-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Genetically altered rat and mouse models have been instrumental in the functional analysis of genes in a physiological context. In particular, studies on the renin-angiotensin system (RAS) have profited from this technology in the past. In this review, we summarize the existing animal models for the protective axis of the RAS consisting of angiotensin-converting enzyme 2 (ACE2), angiotensin-(1-7)(Ang-(1-7), and its receptor Mas. With the help of models with altered expression of the components of this axis in the brain and cardiovascular organs, its physiological and pathophysiological functions have been elucidated. Thus, novel opportunities for therapeutic interventions in cardiovascular diseases were revealed targeting ACE2 or Mas.
Collapse
|
28
|
Alenina N, Bader M. ACE2 in Brain Physiology and Pathophysiology: Evidence from Transgenic Animal Models. Neurochem Res 2018; 44:1323-1329. [PMID: 30443713 PMCID: PMC7089194 DOI: 10.1007/s11064-018-2679-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/15/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a protein consisting of two domains, the N-terminus is a carboxypeptidase homologous to ACE and the C-terminus is homologous to collectrin and responsible for the trafficking of the neutral amino acid transporter B(0)AT1 to the plasma membrane of gut epithelial cells. The carboxypeptidase domain not only metabolizes angiotensin II to angiotensin-(1–7), but also other peptide substrates, such as apelin, kinins and morphins. In addition, the collectrin domain regulates the levels of some amino acids in the blood, in particular of tryptophan. Therefore it is of no surprise that animals with genetic alterations in the expression of ACE2 develop a diverse pattern of phenotypes ranging from hypertension, metabolic and behavioural dysfunctions, to impairments in serotonin synthesis and neurogenesis. This review summarizes the phenotypes of such animals with a particular focus on the central nervous system.
Collapse
Affiliation(s)
- Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany.
- Charité - University Medicine, Berlin, Germany.
- Institute for Biology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
29
|
Wu CH, Mohammadmoradi S, Chen JZ, Sawada H, Daugherty A, Lu HS. Renin-Angiotensin System and Cardiovascular Functions. Arterioscler Thromb Vasc Biol 2018; 38:e108-e116. [PMID: 29950386 PMCID: PMC6039412 DOI: 10.1161/atvbaha.118.311282] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Chia-Hua Wu
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
| | - Shayan Mohammadmoradi
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
| | - Jeff Z Chen
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| | - Hisashi Sawada
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| | - Hong S Lu
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| |
Collapse
|
30
|
Rossignoli A, Vorkapic E, Wanhainen A, Länne T, Skogberg J, Folestad E, Wågsäter D. Plasma cholesterol lowering in an AngII‑infused atherosclerotic mouse model with moderate hypercholesterolemia. Int J Mol Med 2018; 42:471-478. [PMID: 29658561 DOI: 10.3892/ijmm.2018.3619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/22/2018] [Indexed: 11/06/2022] Open
Abstract
Atherosclerosis is the main underlying causes of cardiovascular disease. There is a well‑established association between high blood cholesterol levels and the extent of atherosclerosis. Furthermore, atherosclerosis has been proposed to augment abdominal aortic aneurysm (AAA) formation. As patients with AAA often have parallel atherosclerotic disease and are therefore often on cholesterol‑lowering therapy, it is not possible to fully address the independent effects of plasma cholesterol lowering (PCL) treatment on AAA. The present study investigated the effect of angiotensin II (AngII)‑infusion in modestly hypercholesterolemic Ldlr‑/‑Apob100/100Mttpflox/floxMx1‑Cre mice with or without PCL treatment on a morphological and molecular level, in terms of atherosclerosis and AAA development. AngII infusion in the study mice resulted in an increased atherosclerotic lesion area and increased infiltration of inflammatory leukocytes, which was not observed in mice with PCL induced prior to AngII infusion. This suggested that AngII infusion in this mouse model induced atherosclerosis development, and that plasma cholesterol levels represent a controlling factor. Furthermore, AngII infusion in Ldlr‑/‑Apob100/100Mttpflox/floxMx1‑Cre mice caused a modest aneurysmal phenotype, and no differences in AAA development were observed between the different study groups. However, the fact that modest hypercholesterolemic mice did not develop AAA in a classical aneurysmal model indicated that plasma cholesterol levels are important for disease development.
Collapse
Affiliation(s)
- Aránzazu Rossignoli
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Emina Vorkapic
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, 58183 Linköping, Sweden
| | - Anders Wanhainen
- Department of Surgical Sciences, Section of Vascular Surgery, Uppsala University, 75185 Uppsala, Sweden
| | - Toste Länne
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, 58183 Linköping, Sweden
| | - Josefin Skogberg
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Erika Folestad
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Dick Wågsäter
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, 58183 Linköping, Sweden
| |
Collapse
|
31
|
Angiotensin converting enzyme 2 and diminazene: role in cardiovascular and blood pressure regulation. Curr Opin Nephrol Hypertens 2018; 25:384-95. [PMID: 27367913 DOI: 10.1097/mnh.0000000000000254] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW Angiotensin converting enzyme 2 (ACE2) is an important regulator of the renin-angiotensin system through actions to degrade angiotensin II. Loss of ACE2 can contribute to the development and progression of cardiovascular disease, and experimental studies have highlighted a beneficial role for novel therapeutic approaches that activate or replenish tissue ACE2. This review focuses on experimental studies that have used the off-target effects of the antitrypanosomal agent, diminazene aceturate (DIZE) to activate ACE2. RECENT FINDINGS In cardiovascular disease, activation of the classical renin-angiotensin system and depletion of ACE2 leads to pathophysiological changes. One approach to activate ACE2 involves the drug DIZE, which has been shown to have beneficial effects in experimental models of hypertension, pulmonary hypertension, myocardial infarction, stroke, atherosclerosis, type 1 diabetes, and eye disease. The precise mechanism of action of DIZE to activate ACE2 remains under scrutiny. SUMMARY Activation of ACE2 may represent an important therapeutic approach in cardiovascular disease. To date, most studies have focused on the off-target actions of DIZE, in experimental models of disease. More research is required to determine the exact mechanism of action of DIZE and evaluate its therapeutic potential in comparison with currently available clinical interventions. There are no clinical studies of DIZE, and its side-effects, and toxicity make such studies unlikely. Hence, new methods of selectively activating or replenishing ACE2 will be needed in the future if this approach is to be used in a clinical context.
Collapse
|
32
|
Santos RAS, Sampaio WO, Alzamora AC, Motta-Santos D, Alenina N, Bader M, Campagnole-Santos MJ. The ACE2/Angiotensin-(1-7)/MAS Axis of the Renin-Angiotensin System: Focus on Angiotensin-(1-7). Physiol Rev 2018; 98:505-553. [PMID: 29351514 PMCID: PMC7203574 DOI: 10.1152/physrev.00023.2016] [Citation(s) in RCA: 768] [Impact Index Per Article: 109.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 05/09/2017] [Accepted: 06/18/2017] [Indexed: 12/16/2022] Open
Abstract
The renin-angiotensin system (RAS) is a key player in the control of the cardiovascular system and hydroelectrolyte balance, with an influence on organs and functions throughout the body. The classical view of this system saw it as a sequence of many enzymatic steps that culminate in the production of a single biologically active metabolite, the octapeptide angiotensin (ANG) II, by the angiotensin converting enzyme (ACE). The past two decades have revealed new functions for some of the intermediate products, beyond their roles as substrates along the classical route. They may be processed in alternative ways by enzymes such as the ACE homolog ACE2. One effect is to establish a second axis through ACE2/ANG-(1-7)/MAS, whose end point is the metabolite ANG-(1-7). ACE2 and other enzymes can form ANG-(1-7) directly or indirectly from either the decapeptide ANG I or from ANG II. In many cases, this second axis appears to counteract or modulate the effects of the classical axis. ANG-(1-7) itself acts on the receptor MAS to influence a range of mechanisms in the heart, kidney, brain, and other tissues. This review highlights the current knowledge about the roles of ANG-(1-7) in physiology and disease, with particular emphasis on the brain.
Collapse
Affiliation(s)
- Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Walkyria Oliveira Sampaio
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Andreia C Alzamora
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Daisy Motta-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Natalia Alenina
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Michael Bader
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Maria Jose Campagnole-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
33
|
Ramadan A, Singh KK, Quan A, Plant PJ, Al-Omran M, Teoh H, Verma S. Loss of vascular smooth muscle cell autophagy exacerbates angiotensin II-associated aortic remodeling. J Vasc Surg 2017; 68:859-871. [PMID: 29273297 DOI: 10.1016/j.jvs.2017.08.086] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 08/25/2017] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The pathophysiologic processes of abdominal aortic aneurysms (AAAs) and atherosclerosis often intersect. Given that anomalies in vascular smooth muscle cell (SMC) autophagy have been noted in models of atherosclerosis, we sought to evaluate the potential role that SMC autophagy may play in the initiation and progression of AAAs. METHODS Studies were conducted in ATG7flx/flxSM22α-Cretg/+ (SMC ATG7 knockout [SMC-ATG7-KO]) and ATG7WT/WT; SM22α-Cretg/+ (SMC ATG7 wild-type [SMC-ATG7-WT]) littermates that were continuously infused with angiotensin II (Ang II; 1.5 mg/kg/d) for up to 12 weeks. Mortality, morbidity, hemodynamics, and aortic remodeling were documented. RESULTS During the 12-week observation window, all of the Ang II-treated SMC-ATG-WT mice (n = 6) survived, whereas 10 of the 19 Ang II-treated SMC-ATG-KO mice had died by week 7 (log-rank test, P < .001). Mean arterial pressure (128.07 ± 3.4 mm Hg for Ang II-treated SMC-ATG-KO vs 138.5 ± 5.87 mm Hg for Ang II-treated SMC-ATG-WT mice) and diastolic arterial pressure (109.7 ± 2.55 mm Hg for Ang II-treated SMC-ATG7-KO vs 119.4 ± 2.12 mm Hg for Ang II-treated SMC-ATG7-WT mice) were significantly different between the two groups (P < .01). Cardiac rupture, myocardial infarct, end-organ damage, pleural effusion, and venous distention were noted in Ang II-treated SMC-ATG7-KO but not in Ang II-treated SMC-ATG7-WT mice. Although the suprarenal aortic diameters of the Ang II-treated SMC-ATG7-KO group demonstrated a trending increase (at week 4, 1.26 ± 0.06 mm [n = 14] for Ang II-treated SMC-ATG-KO mice vs 1.09 ± 0.02 mm [n = 5] for Ang II-treated SMC-ATG-WT mice; P < .05), only 2 of the 19 developed abdominal aortic dissections. CONCLUSIONS Mice with SMC ATG7 deficiency that are chronically infused with Ang II do not tend to develop dissecting AAA but do exhibit adverse aortic remodeling and appreciable cardiac failure-associated mortality.
Collapse
Affiliation(s)
- Azza Ramadan
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Krishna K Singh
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Division of Vascular Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Quan
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Pamela J Plant
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Mohammed Al-Omran
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Division of Vascular Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, King Saud University, Riyadh, Kingdom of Saudi Arabia; King Saud University-Li Ka Shing Collaborative Research Program, Riyadh, Kingdom of Saudi Arabia
| | - Hwee Teoh
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
34
|
Alsiraj Y, Thatcher SE, Blalock E, Fleenor B, Daugherty A, Cassis LA. Sex Chromosome Complement Defines Diffuse Versus Focal Angiotensin II-Induced Aortic Pathology. Arterioscler Thromb Vasc Biol 2017; 38:143-153. [PMID: 29097367 DOI: 10.1161/atvbaha.117.310035] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/19/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Aortic pathologies exhibit sexual dimorphism, with aneurysms in both the thoracic and abdominal aorta (ie, abdominal aortic aneurysm [AAA]) exhibiting higher male prevalence. Women have lower prevalence of aneurysms, but when they occur, aneurysms progress rapidly. To define mechanisms for these sex differences, we determined the role of sex chromosome complement and testosterone on the location and progression of angiotensin II (AngII)-induced aortic pathologies. APPROACH AND RESULTS We used transgenic male mice expressing Sry (sex-determining region Y) on an autosome to create Ldlr (low-density lipoprotein receptor)-deficient male mice with an XY or XX sex chromosome complement. Transcriptional profiling was performed on abdominal aortas from XY or XX males, demonstrating 1746 genes influenced by sex chromosomes or sex hormones. Males (XY or XX) were either sham-operated or orchiectomized before AngII infusions. Diffuse aortic aneurysm pathology developed in XY AngII-infused males, whereas XX males developed focal AAAs. Castration reduced all AngII-induced aortic pathologies in XY and XX males. Thoracic aortas from AngII-infused XY males exhibited adventitial thickening that was not present in XX males. We infused male XY and XX mice with either saline or AngII and quantified mRNA abundance of key genes in both thoracic and abdominal aortas. Regional differences in mRNA abundance existed before AngII infusions, which were differentially influenced by AngII between genotypes. Prolonged AngII infusions resulted in aortic wall thickening of AAAs from XY males, whereas XX males had dilated focal AAAs. CONCLUSIONS An XY sex chromosome complement mediates diffuse aortic pathology, whereas an XX sex chromosome complement contributes to focal AngII-induced AAAs.
Collapse
Affiliation(s)
- Yasir Alsiraj
- From the Department of Pharmacology and Nutritional Sciences (Y.A., S.E.T., E.B., L.A.C.), Department of Kinesiology (B.F.), Department of Physiology (A.D.), and Saha Cardiovascular Research Center (A.D.), University of Kentucky, Lexington
| | - Sean E Thatcher
- From the Department of Pharmacology and Nutritional Sciences (Y.A., S.E.T., E.B., L.A.C.), Department of Kinesiology (B.F.), Department of Physiology (A.D.), and Saha Cardiovascular Research Center (A.D.), University of Kentucky, Lexington
| | - Eric Blalock
- From the Department of Pharmacology and Nutritional Sciences (Y.A., S.E.T., E.B., L.A.C.), Department of Kinesiology (B.F.), Department of Physiology (A.D.), and Saha Cardiovascular Research Center (A.D.), University of Kentucky, Lexington
| | - Bradley Fleenor
- From the Department of Pharmacology and Nutritional Sciences (Y.A., S.E.T., E.B., L.A.C.), Department of Kinesiology (B.F.), Department of Physiology (A.D.), and Saha Cardiovascular Research Center (A.D.), University of Kentucky, Lexington
| | - Alan Daugherty
- From the Department of Pharmacology and Nutritional Sciences (Y.A., S.E.T., E.B., L.A.C.), Department of Kinesiology (B.F.), Department of Physiology (A.D.), and Saha Cardiovascular Research Center (A.D.), University of Kentucky, Lexington
| | - Lisa A Cassis
- From the Department of Pharmacology and Nutritional Sciences (Y.A., S.E.T., E.B., L.A.C.), Department of Kinesiology (B.F.), Department of Physiology (A.D.), and Saha Cardiovascular Research Center (A.D.), University of Kentucky, Lexington.
| |
Collapse
|
35
|
Moran CS, Biros E, Krishna SM, Wang Y, Tikellis C, Morton SK, Moxon JV, Cooper ME, Norman PE, Burrell LM, Thomas MC, Golledge J. Resveratrol Inhibits Growth of Experimental Abdominal Aortic Aneurysm Associated With Upregulation of Angiotensin-Converting Enzyme 2. Arterioscler Thromb Vasc Biol 2017; 37:2195-2203. [DOI: 10.1161/atvbaha.117.310129] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/31/2017] [Indexed: 12/16/2022]
Abstract
Objective—
Recent evidence suggests an important role for angiotensin-converting enzyme 2 (ACE2) in limiting abdominal aortic aneurysm (AAA). This study examined the effect of ACE2 deficiency on AAA development and the efficacy of resveratrol to upregulate ACE2 in experimental AAA.
Approach and Results—
Ace2
deletion in apolipoprotein-deficient mice (
ApoE
−/−
Ace2
−/y
) resulted in increased aortic diameter and spontaneous aneurysm of the suprarenal aorta associated with increased expression of inflammation and proteolytic enzyme markers. In humans, serum ACE2 activity was negatively associated with AAA diagnosis.
ACE2
expression was lower in infrarenal biopsies of patients with AAA than organ donors. AAA was more severe in
ApoE
−/−
Ace2
−/y
mice compared with controls in 2 experimental models. Resveratrol (0.05/100-g chow) inhibited growth of pre-established AAAs in
ApoE
−/−
mice fed high-fat chow and infused with angiotensin II continuously for 56 days. Reduced suprarenal aorta dilatation in mice receiving resveratrol was associated with elevated serum ACE2 and increased suprarenal aorta tissue levels of ACE2 and sirtuin 1 activity. In addition, the relative phosphorylation of Akt and ERK (extracellular signal-regulated kinase) 1/2 within suprarenal aorta tissue and gene expression for nuclear factor of kappa light polypeptide gene enhancer in B cells 1, angiotensin type-1 receptor, and metallopeptidase 2 and 9 were significantly reduced. Upregulation of ACE2 in human aortic smooth muscle cells by resveratrol in vitro was sirtuin 1-dependent.
Conclusions—
This study provides experimental evidence of an important role for ACE2 in limiting AAA development and growth. Resveratrol upregulated ACE2 and inhibited AAA growth in a mouse model.
Collapse
Affiliation(s)
- Corey S. Moran
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Erik Biros
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Smriti M. Krishna
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Yutang Wang
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Chris Tikellis
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Susan K. Morton
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Joseph V. Moxon
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Mark E. Cooper
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Paul E. Norman
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Louise M. Burrell
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Merlin C. Thomas
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| | - Jonathan Golledge
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (C.S.M., E.B., S.M.K., S.K.M., J.V.M., J.G.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, Victoria (Y.W.); Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia (C.T., M.E.C., M.C.T.); School of Surgery,
| |
Collapse
|
36
|
Kawai T, Takayanagi T, Forrester SJ, Preston KJ, Obama T, Tsuji T, Kobayashi T, Boyer MJ, Cooper HA, Kwok HF, Hashimoto T, Scalia R, Rizzo V, Eguchi S. Vascular ADAM17 (a Disintegrin and Metalloproteinase Domain 17) Is Required for Angiotensin II/β-Aminopropionitrile-Induced Abdominal Aortic Aneurysm. Hypertension 2017; 70:959-963. [PMID: 28947615 DOI: 10.1161/hypertensionaha.117.09822] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 06/22/2017] [Accepted: 08/31/2017] [Indexed: 01/05/2023]
Abstract
Angiotensin II (AngII)-activated epidermal growth factor receptor has been implicated in abdominal aortic aneurysm (AAA) development. In vascular smooth muscle cells (VSMCs), AngII activates epidermal growth factor receptor via a metalloproteinase, ADAM17 (a disintegrin and metalloproteinase domain 17). We hypothesized that AngII-dependent AAA development would be prevented in mice lacking ADAM17 in VSMCs. To test this concept, control and VSMC ADAM17-deficient mice were cotreated with AngII and a lysyl oxidase inhibitor, β-aminopropionitrile, to induce AAA. We found that 52.4% of control mice did not survive because of aortic rupture. All other surviving control mice developed AAA and demonstrated enhanced expression of ADAM17 in the AAA lesions. In contrast, all AngII and β-aminopropionitrile-treated VSMC ADAM17-deficient mice survived and showed reduction in external/internal diameters (51%/28%, respectively). VSMC ADAM17 deficiency was associated with lack of epidermal growth factor receptor activation, interleukin-6 induction, endoplasmic reticulum/oxidative stress, and matrix deposition in the abdominal aorta of treated mice. However, both VSMC ADAM17-deficient and control mice treated with AngII and β-aminopropionitrile developed comparable levels of hypertension. Treatment of C57Bl/6 mice with an ADAM17 inhibitory antibody but not with control IgG also prevented AAA development. In conclusion, VSMC ADAM17 silencing or systemic ADAM17 inhibition seems to protect mice from AAA formation. The mechanism seems to involve suppression of epidermal growth factor receptor activation.
Collapse
Affiliation(s)
- Tatsuo Kawai
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Takehiko Takayanagi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Steven J Forrester
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Kyle J Preston
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Takashi Obama
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Toshiyuki Tsuji
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Tomonori Kobayashi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Michael J Boyer
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Hannah A Cooper
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Hang Fai Kwok
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Tomoki Hashimoto
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Rosario Scalia
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Victor Rizzo
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.).
| | - Satoru Eguchi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.).
| |
Collapse
|
37
|
Abstract
The health of the cardiovascular and pulmonary systems is inextricably linked to the renin-angiotensin system (RAS). Physiologically speaking, a balance between the vasodeleterious (Angiotensin-converting enzyme [ACE]/Angiotensin II [Ang II]/Ang II type 1 receptor [AT1R]) and vasoprotective (Angiotensin-converting enzyme 2 [ACE2]/Angiotensin-(1-7) [Ang-(1-7)]/Mas receptor [MasR]) components of the RAS is critical for cardiopulmonary homeostasis. Upregulation of the ACE/Ang II/AT1R axis shifts the system toward vasoconstriction, proliferation, hypertrophy, inflammation, and fibrosis, all factors that contribute to the development and progression of cardiopulmonary diseases. Conversely, stimulation of the vasoprotective ACE2/Ang-(1-7)/MasR axis produces a counter-regulatory response that promotes cardiovascular health. Current research is investigating novel strategies to augment actions of the vasoprotective RAS components, particularly ACE2, in order to treat various pathologies. Although multiple approaches to increase the activity of ACE2 have displayed beneficial effects against experimental disease models, the mechanisms behind its protective actions remain incompletely understood. Recent work demonstrating a non-catalytic role for ACE2 in amino acid transport in the gut has led us to speculate that the therapeutic effects of ACE2 can be mediated, in part, by its actions on the gastrointestinal tract and/or gut microbiome. This is consistent with emerging data which suggest that dysbiosis of the gut and lung microbiomes is associated with cardiopulmonary disease. This review highlights new developments in the protective actions of ACE2 against cardiopulmonary disorders, discusses innovative approaches to targeting ACE2 for therapy, and explores an evolving role for gut and lung microbiota in cardiopulmonary health.
Collapse
|
38
|
Velkoska E, Patel SK, Griggs K, Burrell LM. Diminazene Aceturate Improves Cardiac Fibrosis and Diastolic Dysfunction in Rats with Kidney Disease. PLoS One 2016; 11:e0161760. [PMID: 27571511 PMCID: PMC5003360 DOI: 10.1371/journal.pone.0161760] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 08/11/2016] [Indexed: 12/02/2022] Open
Abstract
Angiotensin converting enzyme (ACE) 2 is a negative regulator of the renin angiotensin system (RAS) through its role to degrade angiotensin II. In rats with subtotal nephrectomy (STNx), adverse cardiac remodelling occurs despite elevated cardiac ACE2 activity. We hypothesised that diminazene aceturate (DIZE), which has been described as having an off-target effect to activate ACE2, would have beneficial cardiac effects in STNx rats. STNx led to hypertension, diastolic dysfunction, left ventricular hypertrophy, cardiac fibrosis, and increased cardiac ACE, ACE2, Ang II and Ang 1-7 levels. Cardiac gene expression of ADAM17 was also increased. In STNx, two-weeks of subcutaneous DIZE (15mg/kg/d) had no effect on blood pressure but improved diastolic dysfunction and cardiac fibrosis, reduced ADAM17 mRNA and shifted the cardiac RAS balance to a cardioprotective profile with reduced ACE and Ang II. There was no change in cardiac ACE2 activity or in cardiac Ang 1-7 levels with DIZE. In conclusion, our results suggest that DIZE exerts a protective effect on the heart under the pathological condition of kidney injury. This effect was not due to improved kidney function, a fall in blood pressure or a reduction in LVH but was associated with a reduction in cardiac ACE and cardiac Ang II levels. As in vitro studies showed no direct effect of DIZE on ACE2 or ACE activity, the precise mechanism of action of DIZE remains to be determined.
Collapse
Affiliation(s)
- Elena Velkoska
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Sheila K. Patel
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Karen Griggs
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | - Louise M. Burrell
- Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
39
|
Diminazene aceturate, an angiotensin-converting enzyme II activator, prevents gastric mucosal damage in mice: Role of the angiotensin-(1-7)/Mas receptor axis. Biochem Pharmacol 2016; 112:50-9. [PMID: 27241079 DOI: 10.1016/j.bcp.2016.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/24/2016] [Indexed: 01/15/2023]
Abstract
The angiotensin (Ang) II converting enzyme (ACE II) pathway has recently been shown to be associated with several beneficial effects in various organisms, including gastroprotection. ACE II is responsible for converting Ang II into an active peptide, Ang-(1-7), which in turn binds the Mas receptor. Recent studies have shown that diminazene aceturate (Dize) a trypanocidal used in animals, activates ACE II. Thus, in this study, we aimed to evaluate the gastroprotective effects of Dize via the ACE II/Ang-(1-7)/Mas receptor pathway against gastric lesions induced by ethanol and acetic acid in mice. The results showed that Dize could promote gastric protection via several mechanisms, including increased levels of antioxidants and anti-inflammatory factors (e.g., decreasing tumor necrosis factor and interleukin-6 expression and reducing myeloperoxidase activity), maturation of collagen fibers, and promotion of re-epithelialization and regeneration of gastric tissue in different injury models. Thus, Dize represents a novel potential gastroprotective agent.
Collapse
|
40
|
Diminazene aceturate—An antiparasitic drug of antiquity: Advances in pharmacology & therapeutics. Pharmacol Res 2015; 102:138-57. [DOI: 10.1016/j.phrs.2015.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/28/2015] [Accepted: 10/09/2015] [Indexed: 12/31/2022]
|
41
|
Singh N, Joshi S, Guo L, Baker MB, Li Y, Castellano RK, Raizada MK, Jarajapu YPR. ACE2/Ang-(1-7)/Mas axis stimulates vascular repair-relevant functions of CD34+ cells. Am J Physiol Heart Circ Physiol 2015; 309:H1697-H1707. [PMID: 26386115 PMCID: PMC4666983 DOI: 10.1152/ajpheart.00854.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 09/04/2015] [Indexed: 01/08/2023]
Abstract
CD34(+) stem/progenitor cells have been identified as a promising cell population for the autologous cell-based therapies in patients with cardiovascular disease. The counter-regulatory axes of renin angiotensin system, angiotensin converting enzyme (ACE)/Ang II/angiotensin type 1 (AT1) receptor and ACE2/Ang-(1-7)/Mas receptor, play an important role in the cardiovascular repair. This study evaluated the expression and vascular repair-relevant functions of these two pathways in human CD34(+) cells. CD34(+) cells were isolated from peripheral blood mononuclear cells (MNCs), obtained from healthy volunteers. Expression of ACE, ACE2, AT1, and angiotensin type 2 and Mas receptors were determined. Effects of Ang II, Ang-(1-7), Norleu(3)-Ang-(1-7), and ACE2 activators, xanthenone (XNT) and diminazene aceturate (DIZE) on proliferation, migration, and adhesion of CD34(+) cells were evaluated. ACE2 and Mas were relatively highly expressed in CD34(+) cells compared with MNCs. Ang-(1-7) or its analog, Norleu(3)-Ang-(1-7), stimulated proliferation of CD34(+) cells that was associated with decrease in phosphatase and tensin homologue deleted on chromosome 10 levels and was inhibited by triciribin, an AKT inhibitor. Migration of CD34(+) cells was enhanced by Ang-(1-7) or Norleu(3)-Ang-(1-7) that was decreased by a Rho-kinase inhibitor, Y-27632. In the presence of Ang II, XNT or DIZE enhanced proliferation and migration that were blocked by DX-600, an ACE2 inhibitor. Treatment of MNCs with Ang II, before the isolation of CD34(+) cells, attenuated the proliferation and migration to stromal derived factor-1α. This attenuation was reversed by apocynin, an NADPH oxidase inhibitor. Adhesion of MNCs or CD34(+) cells to fibronectin was enhanced by Ang II and was unaffected by Ang-(1-7). This study suggests that ACE2/Ang-(1-7)/Mas pathway stimulates functions of CD34(+) cells that are cardiovascular protective, whereas Ang II attenuates these functions by acting on MNCs. These findings imply that activation of ACE2/Ang-(1-7)/Mas axis is a promising approach for enhancing reparative outcomes of cell-based therapies.
Collapse
MESH Headings
- Angiotensin I/pharmacology
- Angiotensin II/pharmacology
- Angiotensin-Converting Enzyme 2
- Antigens, CD34
- Cell Adhesion/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Diminazene/analogs & derivatives
- Diminazene/pharmacology
- Humans
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Peptide Fragments/pharmacology
- Peptidyl-Dipeptidase A/drug effects
- Peptidyl-Dipeptidase A/metabolism
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/drug effects
- Proto-Oncogene Proteins/metabolism
- Reactive Oxygen Species/metabolism
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/drug effects
- Receptor, Angiotensin, Type 2/metabolism
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/metabolism
- Vasoconstrictor Agents/pharmacology
- Vasodilator Agents/pharmacology
- Wound Healing/drug effects
- Xanthones/pharmacology
Collapse
Affiliation(s)
- Neha Singh
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Shrinidh Joshi
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Lirong Guo
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota; Department of Pathophysiology, College of Basic Medical Science, Jilin University, Changchun, Jilin, China
| | - Matthew B Baker
- Department of Chemistry, University of Florida, Gainesville, Florida; and
| | - Yan Li
- Department of Chemistry, University of Florida, Gainesville, Florida; and
| | | | - Mohan K Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida
| | - Yagna P R Jarajapu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota;
| |
Collapse
|
42
|
Fraga-Silva RA, Montecucco F, Costa-Fraga FP, Nencioni A, Caffa I, Bragina ME, Mach F, Raizada MK, Santos RAS, da Silva RF, Stergiopulos N. Diminazene enhances stability of atherosclerotic plaques in ApoE-deficient mice. Vascul Pharmacol 2015; 74:103-113. [PMID: 26304699 PMCID: PMC5589185 DOI: 10.1016/j.vph.2015.08.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/22/2015] [Accepted: 08/20/2015] [Indexed: 11/23/2022]
Abstract
Angiotensin (Ang) II contributes to the development of atherosclerosis, while Ang-(1-7) has atheroprotective actions. Accordingly, angiotensin-converting enzyme 2 (ACE2), which breaks-down Ang II and forms Ang-(1-7), has been suggested as a target against atherosclerosis. Here we investigated the actions of diminazene, a recently developed ACE2 activator compound, in a model of vulnerable atherosclerotic plaque. Atherosclerotic plaque formation was induced in the carotid artery of ApoE-deficient mice by a shear stress (SS) modifier device. The animals were treated with diminazene (15mg/kg/day) or vehicle. ACE2 was strongly expressed in the aortic root and low SS-induced carotid plaques, but poorly expressed in the oscillatory SS-induced carotid plaques. Diminazene treatment did not change the lesion size, but ameliorated the composition of aortic root and low SS-induced carotid plaques by increasing collagen content and decreasing both MMP-9 expression and macrophage infiltration. Interestingly, these beneficial effects were not observed in the oscillatory SS-induced plaque. Additionally, diminazene treatment decreased intraplaque ICAM-1 and VCAM-1 expression, circulating cytokine and chemokine levels and serum triglycerides. In summary, ACE2 was distinctively expressed in atherosclerotic plaques, which depends on the local pattern of shear stress. Moreover, diminazene treatment enhances the stability of atherosclerotic plaques.
Collapse
Affiliation(s)
- Rodrigo A Fraga-Silva
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Fabrizio Montecucco
- Division of Cardiology, Faculty of Medicine, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
| | - Fabiana P Costa-Fraga
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alessio Nencioni
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
| | - Irene Caffa
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
| | - Maiia E Bragina
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - François Mach
- Division of Cardiology, Faculty of Medicine, Foundation for Medical Researches, University of Geneva, Geneva, Switzerland
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, United States
| | - Robson A S Santos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rafaela F da Silva
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nikolaos Stergiopulos
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
43
|
Pafili K, Gouni-Berthold I, Papanas N, Mikhailidis DP. Abdominal aortic aneurysms and diabetes mellitus. J Diabetes Complications 2015; 29:1330-6. [PMID: 26440573 DOI: 10.1016/j.jdiacomp.2015.08.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/16/2015] [Accepted: 08/17/2015] [Indexed: 11/16/2022]
Abstract
There is accumulating evidence that risk profiles differ between coronary artery disease and abdominal aortic aneurysms (AAAs). However, diabetes mellitus (DM) appears to be negatively associated with AAA formation. The underlying mechanisms for this negative relationship are far from defined, but may include: increased arterial wall matrix formation via advanced glycation end products; suppression of plasmin and reduction of levels and activity of matrix metalloproteinases (MMP)-2 and 9; diminished aortic wall macrophage infiltration, elastolysis and neovascularization. In addition, the effect of pharmacological agents used for the treatment of patients with DM on AAA formation has been studied with rather controversial results. Statins, angiotensin converting enzyme inhibitors, angiotensin receptor blockers, fenofibrate, antibiotics and some hypoglycemic agents are beginning to be appreciated for a potential modest protection from AAAs, but further studies are needed.
Collapse
Affiliation(s)
- Kalliopi Pafili
- Diabetes Clinic, Second Department of Internal Medicine, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioanna Gouni-Berthold
- Center for Endocrinology, Diabetes and Preventive Medicine, University of Cologne, Cologne, Germany
| | - Nikolaos Papanas
- Diabetes Clinic, Second Department of Internal Medicine, Medical School, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry (Vascular Disease Prevention Clinics), Royal Free Hospital campus, University College London Medical School, University College London (UCL), London NW3 2QG, UK
| |
Collapse
|
44
|
Miner GH, Faries PL, Costa KD, Hanss BG, Marin ML. An update on the etiology of abdominal aortic aneurysms: implications for future diagnostic testing. Expert Rev Cardiovasc Ther 2015; 13:1079-90. [PMID: 26401919 DOI: 10.1586/14779072.2015.1082906] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abdominal aortic aneurysm (AAA) disease is multifactorial with both environmental and genetic risk factors. The current research in AAA revolves around genetic profiles and expression studies in both human and animal models. Variants in genes involved in extracellular matrix degradation, inflammation, the renin-angiotensin system, cell growth and proliferation and lipid metabolism have been associated with AAA using a variety of study designs. However, the results have been inconsistent and without a standard animal model for validation. Thus, despite the growing body of knowledge, the specific variants responsible for AAA development, progression and rupture have yet to be determined. This review explores some of the more significant genetic studies to provide an overview of past studies that have influenced the current understanding of AAA etiology. Expanding our understanding of disease pathogenesis will inform research into novel diagnostics and therapeutics and ultimately to improve outcomes for patients with AAA.
Collapse
Affiliation(s)
- Grace H Miner
- a Icahn school of Medicine at Mount Sinai, New York, USA
| | - Peter L Faries
- a Icahn school of Medicine at Mount Sinai, New York, USA
| | - Kevin D Costa
- a Icahn school of Medicine at Mount Sinai, New York, USA
| | - Basil G Hanss
- a Icahn school of Medicine at Mount Sinai, New York, USA
| | | |
Collapse
|
45
|
Thatcher SE, Zhang X, Woody S, Wang Y, Alsiraj Y, Charnigo R, Daugherty A, Cassis LA. Exogenous 17-β estradiol administration blunts progression of established angiotensin II-induced abdominal aortic aneurysms in female ovariectomized mice. Biol Sex Differ 2015; 6:12. [PMID: 26131353 PMCID: PMC4485333 DOI: 10.1186/s13293-015-0030-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/15/2015] [Indexed: 11/24/2022] Open
Abstract
Background Abdominal aortic aneurysms (AAAs) occur predominately in males. However, AAAs in females have rapid growth rates and rupture at smaller sizes. Mechanisms contributing to AAA progression in females are undefined. We defined effects of ovariectomy, with and without 17-β estradiol (E2), on progression of established angiotensin II (AngII)-induced AAAs in female mice. Methods We used neonatal testosterone exposures at 1 day of age to promote susceptibility to AngII-induced AAAs in adult female Ldlr−/− mice. Females were infused with AngII for 28 days to induce AAAs, and then stratified into groups that were sham, ovariectomized (Ovx, vehicle), or Ovx with E2 administration for 2 months of continued AngII infusions. Aortic lumen diameters were quantified by ultrasound and analyzed by linear mixed model, and maximal AAA diameters were analyzed by one-way ANOVA. Atherosclerosis was quantified en face in the aortic arch. AAA tissue sections were analyzed for cellular composition. We quantified effects of E2 on abdominal aortic smooth muscle cell (SMC) growth, α-actin and transforming growth factor-beta (TGF-β) production, and wound healing. Results Serum E2 concentrations were increased significantly by E2. Aortic lumen diameters increased over time in sham-operated and Ovx (vehicle) females, but not in Ovx females administered E2. At day 70, E2 administration decreased significantly aortic lumen diameters compared to Ovx vehicle and sham-operated females. Compared to Ovx females (vehicle), maximal AAA diameters were reduced significantly by E2. AAA tissue sections from Ovx females administered E2 exhibited significant increases in α-actin and decreases in neutrophils compared to Ovx females administered vehicle. In abdominal aortic SMCs, E2 resulted in a concentration-dependent increase in α-actin, elevated TGF-β, and more rapid wound healing. E2 administration to Ovx females also significantly reduced atherosclerotic lesions compared to sham-operated females. This effect was accompanied by significant reductions in serum cholesterol concentrations. Conclusions E2 administration to Ovx females abolished progressive growth and decreased severity of AngII-induced AAAs. These effects were accompanied by increased SMC α-actin, elevated TGF-β, and reduced neutrophils. Similarly, E2 administration reduced AngII-induced atherosclerosis. These results suggest that loss of E2 in post-menopausal females may contribute to progressive growth of AAAs. Electronic supplementary material The online version of this article (doi:10.1186/s13293-015-0030-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sean E Thatcher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Room 521b, Charles T. Wethington Bldg, Lexington, KY 40536-0200 USA
| | - Xuan Zhang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Room 521b, Charles T. Wethington Bldg, Lexington, KY 40536-0200 USA
| | - Shannon Woody
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Room 521b, Charles T. Wethington Bldg, Lexington, KY 40536-0200 USA
| | - Yu Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Room 521b, Charles T. Wethington Bldg, Lexington, KY 40536-0200 USA
| | - Yasir Alsiraj
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Room 521b, Charles T. Wethington Bldg, Lexington, KY 40536-0200 USA
| | - Richard Charnigo
- Department of Statistics, University of Kentucky, Lexington, KY 40536 USA
| | - Alan Daugherty
- Saha Cardiovascular Center, University of Kentucky, Lexington, KY 40536 USA ; Department of Physiology, University of Kentucky, Lexington, KY 40536 USA
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Room 521b, Charles T. Wethington Bldg, Lexington, KY 40536-0200 USA
| |
Collapse
|
46
|
Short-term treatment with diminazene aceturate ameliorates the reduction in kidney ACE2 activity in rats with subtotal nephrectomy. PLoS One 2015; 10:e0118758. [PMID: 25786223 PMCID: PMC4364975 DOI: 10.1371/journal.pone.0118758] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 01/06/2015] [Indexed: 01/28/2023] Open
Abstract
Angiotensin converting enzyme (ACE) 2 is an important modulator of the renin angiotensin system (RAS) through its role to degrade angiotensin (Ang) II. Depletion of kidney ACE2 occurs following kidney injury due to renal mass reduction and may contribute to progressive kidney disease. This study assessed the effect of diminazine aceturate (DIZE), which has been described as an ACE2 activator, on kidney ACE2 mRNA and activity in rats with kidney injury due to subtotal nephrectomy (STNx). Sprague Dawley rats were divided into Control groups or underwent STNx; rats then received vehicle or the DIZE (s.c. 15 mg/kg/day) for 2 weeks. STNx led to hypertension (P<0.01), kidney hypertrophy (P<0.001) and impaired kidney function (P<0.001) compared to Control rats. STNx was associated with increased kidney cortical ACE activity, and reduced ACE2 mRNA in the cortex (P<0.01), with reduced cortical and medullary ACE2 activity (P<0.05), and increased urinary ACE2 excretion (P<0.05) compared to Control rats. Urinary ACE2 activity correlated positively with urinary protein excretion (P<0.001), and negatively with creatinine clearance (P=0.04). In STNx rats, DIZE had no effect on blood pressure or kidney function, but was associated with reduced cortical ACE activity (P<0.01), increased cortical ACE2 mRNA (P<0.05) and increased cortical and medullary ACE2 activity (P<0.05). The precise in vivo mechanism of action of DIZE is not clear, and its effects to increase ACE2 activity may be secondary to an increase in ACE2 mRNA abundance. In ex vivo studies, DIZE did not increase ACE2 activity in either Control or STNx kidney cortical membranes. It is not yet known if chronic administration of DIZE has long-term benefits to slow the progression of kidney disease.
Collapse
|