1
|
Kamdar A, Sykes R, Thomson CR, Mangion K, Ang D, Lee MAW, Van Agtmael T, Berry C. Vascular fibrosis and extracellular matrix remodelling in post-COVID 19 conditions. INFECTIOUS MEDICINE 2024; 3:100147. [PMID: 39649442 PMCID: PMC11621938 DOI: 10.1016/j.imj.2024.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/02/2024] [Accepted: 09/23/2024] [Indexed: 12/10/2024]
Abstract
Causal associations between viral infections and acute myocardial injury are not fully understood, with mechanisms potentially involving direct cardiovascular involvement or systemic inflammation. This review explores plausible mechanisms of vascular fibrosis in patients with post-COVID-19 syndrome, focusing on extracellular matrix remodelling. Despite global attention, significant mechanistic or translational breakthroughs in the management of post-viral syndromes remain limited. No effective pharmacological or non-pharmacological interventions are currently available for patients experiencing persistent symptoms following COVID-19 infection. The substantial expansion of scientific knowledge resulting from collaborative efforts by medical experts, scientists, and government organisations in undertaking COVID-19 research could inform treatment strategies for other post-viral syndromes and respiratory illnesses. There is a critical need for clinical trials to evaluate potential therapeutic candidates, providing evidence to guide treatment decisions for post-COVID-19 syndromes.
Collapse
Affiliation(s)
- Anna Kamdar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Robert Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Cameron R. Thomson
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK
| | - Daniel Ang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Michelle AW Lee
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Tom Van Agtmael
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK
| |
Collapse
|
2
|
Liu L, Li Y, Li JX, Xiao X, Wan TT, Li HH, Guo SB. ACE2 Expressed on Myeloid Cells Alleviates Sepsis-Induced Acute Liver Injury via the Ang-(1-7)-Mas Receptor Axis. Inflammation 2024; 47:891-908. [PMID: 38240986 DOI: 10.1007/s10753-023-01949-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 06/04/2024]
Abstract
Sepsis-induced acute liver injury (ALI) is common in intensive care units. Angiotensin-converting enzyme 2 (ACE2) plays a vital role in hepatic fibrosis and steatosis; however, its role in sepsis-induced ALI remains unclear. This study found that hepatic ACE2 expression in cecal ligation and puncture (CLP)-treated mice significantly decreased 24 h after CLP. ACE2-transgenic (TG) mice exhibited a significant improvement in CLP-induced ALI, accompanied by the inhibition of hepatocyte apoptosis, oxidative stress, and inflammation, while ACE2-knockout mice demonstrated an opposite trend. During sepsis-induced ALI, ACE2-TG could also elevate the Ang-(1-7) and Mas receptor (MasR) levels in liver tissues. Interestingly, the MasR inhibitor A779 abrogated the favorable effects of ACE2 on CLP-induced ALI. In a bone marrow transplantation experiment, the ACE2-TG transplantation group showed significantly improved inflammation and liver dysfunction, less hepatocyte apoptosis, and reduced oxidative stress after CLP compared with the wild-type transplantation group. In contrast, the ACE2-knockout group showed poor inflammatory response and liver dysfunction, significantly more hepatocyte apoptosis, and elevated oxidative stress than the wild-type transplantation group after CLP. ACE2 protects against sepsis-induced ALI by inhibiting hepatocyte apoptosis, oxidative stress, and inflammation via the Ang-(1-7)-Mas receptor axis. Thus, targeting ACE2 may be a promising novel strategy for preventing and treating sepsis-induced ALI.
Collapse
Affiliation(s)
- Lei Liu
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing, 100020, China
| | - Ya Li
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing, 100020, China
| | - Jia-Xin Li
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing, 100020, China
| | - Xue Xiao
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing, 100020, China
| | - Tian-Tian Wan
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing, 100020, China
| | - Hui-Hua Li
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing, 100020, China.
| | - Shu-Bin Guo
- Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Department of Emergency Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
3
|
Wan TT, Li Y, Li JX, Xiao X, Liu L, Li HH, Guo SB. ACE2 activation alleviates sepsis-induced cardiomyopathy by promoting MasR-Sirt1-mediated mitochondrial biogenesis. Arch Biochem Biophys 2024; 752:109855. [PMID: 38097099 DOI: 10.1016/j.abb.2023.109855] [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: 08/24/2023] [Revised: 10/17/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023]
Abstract
Sepsis-induced cardiomyopathy (SIC), caused by a dysregulated host response to infection, is a major contributor to high mortality. Angiotensin-converting enzyme 2 (ACE2), a crucial component of the renin-angiotensin system (RAS), has protective effects against several cardiovascular diseases, such as myocardial infarction and heart failure. However, the role of ACE2 in the pathogenesis of SIC and underlying mechanisms remain unknown. The present study was designed to examine the effects of ACE2 activation or inhibition on SIC in C57BL/6 mice. The ACE2 activator diminazene aceturate (DIZE) and ACE2 inhibitor MLN-4760 were applied for treatment. Myocardial function, inflammatory response, oxidative stress, apoptosis and mitochondrial biogenesis were investigated. Major assays were echocardiography, H&E staining, immunofluorescence staining, DHE staining, TUNEL staining, Western blot, qPCR analysis, ELISA and corresponding kits. We confirmed that ACE2 was markedly downregulated in septic heart tissues. Pharmacological activation of ACE2 by DIZE ameliorated cecal ligation puncture (CLP)-induced mortality, cardiac dysfunction, inflammatory response, oxidative stress and the cardiomyocyte apoptosis by promoting MasR-Sirt1-mediated mitochondrial biogenesis. In contrast, SIC was aggravated via inhibiting MasR-Sirt1-mediated mitochondrial biogenesis by the use of ACE2 inhibitor MLN-4760. Consequently, activation of ACE2 may protect against SIC by promoting MasR-Sirt1-mediated mitochondrial biogenesis.
Collapse
Affiliation(s)
- Tian-Tian Wan
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Ya Li
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Jia-Xin Li
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Xue Xiao
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Lei Liu
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China.
| | - Shu-Bin Guo
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China.
| |
Collapse
|
4
|
Kim D, Jeong W, Kim Y, Lee J, Cho SW, Oh CM, Park R. Pharmacologic Activation of Angiotensin-Converting Enzyme II Alleviates Diabetic Cardiomyopathy in db/db Mice by Reducing Reactive Oxidative Stress. Diabetes Metab J 2023; 47:487-499. [PMID: 37096378 PMCID: PMC10404524 DOI: 10.4093/dmj.2022.0125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/29/2022] [Indexed: 04/26/2023] Open
Abstract
BACKGRUOUND Diabetes mellitus is one of the most common chronic diseases worldwide, and cardiovascular disease is the leading cause of morbidity and mortality in diabetic patients. Diabetic cardiomyopathy (DCM) is a phenomenon characterized by a deterioration in cardiac function and structure, independent of vascular complications. Among many possible causes, the renin-angiotensin-aldosterone system and angiotensin II have been proposed as major drivers of DCM development. In the current study, we aimed to investigate the effects of pharmacological activation of angiotensin-converting enzyme 2 (ACE2) on DCM. METHODS The ACE2 activator diminazene aceturate (DIZE) was administered intraperitoneally to male db/db mice (8 weeks old) for 8 weeks. Transthoracic echocardiography was used to assess cardiac mass and function in mice. Cardiac structure and fibrotic changes were examined using histology and immunohistochemistry. Gene and protein expression levels were examined using quantitative reverse transcription polymerase chain reaction and Western blotting, respectively. Additionally, RNA sequencing was performed to investigate the underlying mechanisms of the effects of DIZE and identify novel potential therapeutic targets for DCM. RESULTS Echocardiography revealed that in DCM, the administration of DIZE significantly improved cardiac function as well as reduced cardiac hypertrophy and fibrosis. Transcriptome analysis revealed that DIZE treatment suppresses oxidative stress and several pathways related to cardiac hypertrophy. CONCLUSION DIZE prevented the diabetes mellitus-mediated structural and functional deterioration of mouse hearts. Our findings suggest that the pharmacological activation of ACE2 could be a novel treatment strategy for DCM.
Collapse
Affiliation(s)
- Donghyun Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Wooju Jeong
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jibeom Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, College of Medicine, Inje University, Goyang, Korea
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University, Busan, Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
5
|
Kim K, Moon JH, Ahn CH, Lim S. Effect of olmesartan and amlodipine on serum angiotensin-(1-7) levels and kidney and vascular function in patients with type 2 diabetes and hypertension. Diabetol Metab Syndr 2023; 15:43. [PMID: 36899369 PMCID: PMC10005920 DOI: 10.1186/s13098-023-00987-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/27/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Recent studies suggest that angiotensin-converting enzyme 2 (ACE2) and angiotensin-(1-7) [Ang-(1-7)] might have beneficial effects on the cardiovascular system. We investigated the effects of olmesartan on the changes in serum ACE2 and Ang-(1-7) levels as well as kidney and vascular function in patients with type 2 diabetes and hypertension. METHODS This was a prospective, randomized, active comparator-controlled trial. Eighty participants with type 2 diabetes and hypertension were randomized to receive 20 mg of olmesartan (N = 40) or 5 mg of amlodipine (N = 40) once daily. The primary endpoint was changes of serum Ang-(1-7) from baseline to week 24. RESULTS Both olmesartan and amlodipine treatment for 24 weeks decreased systolic and diastolic blood pressures significantly by > 18 mmHg and > 8 mmHg, respectively. Serum Ang-(1-7) levels were more significantly increased by olmesartan treatment (25.8 ± 34.5 pg/mL → 46.2 ± 59.4 pg/mL) than by amlodipine treatment (29.2 ± 38.9 pg/mL → 31.7 ± 26.0 pg/mL), resulting in significant between-group differences (P = 0.01). Serum ACE2 levels showed a similar pattern (6.31 ± 0.42 ng/mL → 6.74 ± 0.39 ng/mL by olmesartan treatment vs. 6.43 ± 0.23 ng/mL → 6.61 ± 0.42 ng/mL by amlodipine treatment; P < 0.05). The reduction in albuminuria was significantly associated with the increases in ACE2 and Ang-(1-7) levels (r = - 0.252 and r = - 0.299, respectively). The change in Ang-(1-7) levels was positively associated with improved microvascular function (r = 0.241, P < 0.05). Multivariate regression analyses showed that increases in serum Ang-(1-7) levels were an independent predictor of a reduction in albuminuria. CONCLUSIONS These findings suggest that the beneficial effects of olmesartan on albuminuria may be mediated by increased ACE2 and Ang-(1-7) levels. These novel biomarkers may be therapeutic targets for the prevention and treatment of diabetic kidney disease. TRIAL REGISTRATION ClinicalTrials.gov NCT05189015.
Collapse
Affiliation(s)
- Kyuho Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-707, South Korea
| | - Ji Hye Moon
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-707, South Korea
| | - Chang Ho Ahn
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-707, South Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-707, South Korea.
| |
Collapse
|
6
|
Zhao K, Xu T, Mao Y, Wu X, Hua D, Sheng Y, Li P. Alamandine alleviated heart failure and fibrosis in myocardial infarction mice. Biol Direct 2022; 17:25. [PMID: 36167556 PMCID: PMC9516792 DOI: 10.1186/s13062-022-00338-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/20/2022] [Indexed: 11/10/2022] Open
Abstract
Alamandine (Ala) is the newest identified peptide of the renin-angiotensin system and has protective effect on myocyte hypertrophy. However, it is still unclear whether Ala can alleviate heart failure (HF). The aim of this study was to explore the effects of Ala on HF and the related cardiac fibrosis, and to probe the mechanism. HF model was induced by myocardial infarction (MI) in mice. Four weeks after MI, Ala was administrated by intraperitoneal injection for two weeks. Ala injection significantly improved cardiac dysfunction of MI mice in vivo. The cardiac fibrosis and the related biomarkers were attenuated after Ala administration in HF mice in vivo. The increases of collagen I, alpha-smooth muscle actin and transforming growth factor-beta induced by oxygen–glucose deprivation (OGD) in neonatal rat cardiac fibroblasts (NRCFs) were inhibited by Ala treatment in vitro. The biomarkers of apoptosis were elevated in NRCFs induced by OGD, which were attenuated after treating with Ala in vitro. The enhancement of oxidative stress in the heart of MI mice or in the NRCFs treated with OGD was suppressed by treating with Ala in vivo and in vitro. These effects of Ala were reversed by tBHP, an exogenous inducer of oxidative stress in vitro. These results demonstrated that Ala could alleviate cardiac dysfunction and attenuate cardiac fibrosis via inhibition of oxidative stress.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Tianhua Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yukang Mao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Xiaoguang Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Dongxu Hua
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China. .,Department of Cardiology, Jiangsu Province Hospital, Nanjing, Jiangsu, China.
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
7
|
Dörr K, Kammer M, Reindl-Schwaighofer R, Lorenz M, Marculescu R, Poglitsch M, Beitzke D, Oberbauer R. The Effect of FGF23 on Cardiac Hypertrophy Is Not Mediated by Systemic Renin-Angiotensin- Aldosterone System in Hemodialysis. Front Med (Lausanne) 2022; 9:878730. [PMID: 35559350 PMCID: PMC9086596 DOI: 10.3389/fmed.2022.878730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/05/2022] [Indexed: 01/08/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) is elevated in patients with chronic kidney disease and contributes to left ventricular hypertrophy (LVH). The aim of the analysis was to determine whether this effect is mediated by the renin-angiotensin-aldosterone system (RAAS) in hemodialysis. Serum samples from 62 randomized hemodialysis patients with LVH were analyzed for plasma renin activity (PRA-S), angiotensin II (AngII), and metabolites, angiotensin-converting enzyme-2 (ACE2) and aldosterone using a high throughput mass spectrometry assay. Compared to healthy individuals, levels of the RAAS parameters PRA-S, AngII and aldosterone were generally lower [median (IQR) PRA-S 130 (46–269) vs. 196 (98, 238) pmol/L; AngII 70 (28–157) vs. 137 (76, 201) pmol/L; Aldosterone 130 (54, 278) vs. 196 (98, 238) pmol/L]. We did not find an indication that the effect of FGF23 on LVH was mediated by RAAS parameters, with all estimated indirect effects virtually zero. Furthermore, FGF23 was not associated with RAAS parameter levels throughout the study. While there was a clear association between FGF23 levels and left ventricular mass index (LVMI) at the end of the study and in the FGF23 fold change and LVMI change analysis, no association between RAAS and LVMI was observed. Serum concentrations of PRA-S, AngII, and aldosterone were below the ranges measured in healthy controls suggesting that RAAS is not systemically activated in hemodialysis patients. The effect of FGF23 on LVMI was not mediated by systemic RAAS activity. These findings challenge the current paradigm of LVH progression and treatment with RAAS blockers in dialysis.
Collapse
Affiliation(s)
- Katharina Dörr
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| | - Michael Kammer
- Department of Nephrology, Medical University of Vienna, Vienna, Austria.,Center for Medical Statistics, Informatics, and Intelligent Systems, Section for Clinical Biometrics, Medical University of Vienna, Vienna, Austria
| | | | | | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Cardiovascular and Interventional Radiology, Medical University of Vienna, Vienna, Austria
| | - Rainer Oberbauer
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Megiorni F, Pontecorvi P, Gerini G, Anastasiadou E, Marchese C, Ceccarelli S. Sex-Related Factors in Cardiovascular Complications Associated to COVID-19. Biomolecules 2021; 12:biom12010021. [PMID: 35053169 PMCID: PMC8773922 DOI: 10.3390/biom12010021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), the pandemic infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents with an extremely heterogeneous spectrum of symptoms and signs. The clinical manifestations seem to be correlated with disease severity. COVID-19 susceptibility and mortality show a significant sex imbalance, with men being more prone to infection and showing a higher rate of hospitalization and mortality compared to women. Such variability can be ascribed to both sex-related biological factors and gender-related behavioral cues. This review will discuss the potential mechanisms accounting for sex/gender influence in vulnerability to COVID-19. Cardiovascular diseases play a central role in determining COVID-19 outcome, whether they are pre-existent or arose upon infection. We will pay particular attention to the impact of sex and gender on cardiovascular manifestations related to COVID-19. Finally, we will discuss the sex-dependent variability in some biomarkers for the evaluation of COVID-19 infection and prognosis. The aim of this work is to highlight the significance of gendered medicine in setting up personalized programs for COVID-19 prevention, clinical evaluation and treatment.
Collapse
|
9
|
Rajtik T, Galis P, Bartosova L, Paulis L, Goncalvesova E, Klimas J. Alternative RAS in Various Hypoxic Conditions: From Myocardial Infarction to COVID-19. Int J Mol Sci 2021; 22:ijms222312800. [PMID: 34884604 PMCID: PMC8657827 DOI: 10.3390/ijms222312800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 12/28/2022] Open
Abstract
Alternative branches of the classical renin–angiotensin–aldosterone system (RAS) represent an important cascade in which angiotensin 2 (AngII) undergoes cleavage via the action of the angiotensin-converting enzyme 2 (ACE2) with subsequent production of Ang(1-7) and other related metabolites eliciting its effects via Mas receptor activation. Generally, this branch of the RAS system is described as its non-canonical alternative arm with counterbalancing actions to the classical RAS, conveying vasodilation, anti-inflammatory, anti-remodeling and anti-proliferative effects. The implication of this branch was proposed for many different diseases, ranging from acute cardiovascular conditions, through chronic respiratory diseases to cancer, nonetheless, hypoxia is one of the most prominent common factors discussed in conjugation with the changes in the activity of alternative RAS branches. The aim of this review is to bring complex insights into the mechanisms behind the various forms of hypoxic insults on the activity of alternative RAS branches based on the different duration of stimuli and causes (acute vs. intermittent vs. chronic), localization and tissue (heart vs. vessels vs. lungs) and clinical relevance of studied phenomenon (experimental vs. clinical condition). Moreover, we provide novel insights into the future strategies utilizing the alternative RAS as a diagnostic tool as well as a promising pharmacological target in serious hypoxia-associated cardiovascular and cardiopulmonary diseases.
Collapse
Affiliation(s)
- Tomas Rajtik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
- Correspondence: ; Tel.: +42-12-501-17-391
| | - Peter Galis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| | - Linda Bartosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| | - Ludovit Paulis
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia;
| | - Eva Goncalvesova
- Department of Heart Failure, Clinic of Cardiology, National Institute of Cardiovascular Diseases, 831 01 Bratislava, Slovakia;
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| |
Collapse
|
10
|
Wang LF, Sun YY, Pan Q, Yin YQ, Tian XM, Liu Y, Bu T, Zhang Q, Wang YA, Zhao J, Luo Y. Diminazen Aceturate Protects Pulmonary Ischemia-Reperfusion Injury via Inhibition of ADAM17-Mediated Angiotensin-Converting Enzyme 2 Shedding. Front Pharmacol 2021; 12:713632. [PMID: 34712133 PMCID: PMC8546118 DOI: 10.3389/fphar.2021.713632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 09/16/2021] [Indexed: 01/30/2023] Open
Abstract
Lung ischemia-reperfusion (IR) injury is induced by pulmonary artery occlusion and reperfusion. Lung IR injury commonly happens after weaning from extracorporeal circulation, lung transplantation, and pulmonary thromboendarterectomy; it is a lethal perioperative complication. A definite therapeutic intervention remains to be determined. It is known that the enzyme activity of angiotensin-converting enzyme 2 (ACE2) is critical in maintaining pulmonary vascular tone and epithelial integrity. In a noxious environment to the lungs, inactivation of ACE2 is mainly due to a disintegrin and metalloprotease 17 (ADAM17) protein-mediated ACE2 shedding. Thus, we assumed that protection of local ACE2 in the lung against ADAM17-mediated shedding would be a therapeutic target for lung IR injury. In this study, we established both in vivo and in vitro models to demonstrate that the damage degree of lung IR injury depends on the loss of ACE2 and ACE2 enzyme dysfunction in lung tissue. Treatment with ACE2 protectant diminazen aceturate (DIZE) maintained higher ACE2 enzyme activity and reduced angiotensin II, angiotensin type 1 receptor, and ADAM17 levels in the lung tissue. Concurrently, DIZE-inhibited oxidative stress and nitrosative stress via p38MAPK and NF-κB pathways consequently reduced release of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β. The underlying molecular mechanism of DIZE contributed to its protective effect against lung IR injury and resulted in the improvement of oxygenation index and ameliorating pulmonary pathological damage. We concluded that DIZE protects the lungs from IR injury via inhibition of ADAM17-mediated ACE2 shedding.
Collapse
Affiliation(s)
| | - Yang-Yang Sun
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Qian Pan
- China-Japan Friendship Hospital, Beijing, China
| | - Yi-Qing Yin
- China-Japan Friendship Hospital, Beijing, China
| | | | - Yue Liu
- China-Japan Friendship Hospital, Beijing, China
| | - Tegeleqi Bu
- China-Japan Friendship Hospital, Beijing, China
| | - Qingy Zhang
- China-Japan Friendship Hospital, Beijing, China
| | - Yong-An Wang
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Jing Zhao
- China-Japan Friendship Hospital, Beijing, China
| | - Yuan Luo
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences (AMMS), Beijing, China
| |
Collapse
|
11
|
Zhang M, Sui W, Xing Y, Cheng J, Cheng C, Xue F, Zhang J, Wang X, Zhang C, Hao P, Zhang Y. Angiotensin IV attenuates diabetic cardiomyopathy via suppressing FoxO1-induced excessive autophagy, apoptosis and fibrosis. Theranostics 2021; 11:8624-8639. [PMID: 34522203 PMCID: PMC8419053 DOI: 10.7150/thno.48561] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/17/2021] [Indexed: 12/27/2022] Open
Abstract
Rationale: The rennin-angiotensin-aldosterone system (RAAS) plays a critical role in the pathogenesis of diabetic cardiomyopathy, but the role of a member of RAAS, angiotensin IV (Ang IV), in this disease and its underlying mechanism are unclear. This study was aimed to clarify the effects of Ang IV and its downstream mediator forkhead box protein O1 (FoxO1) on diabetic cardiomyopathy. Methods:In vivo, diabetic mice were treated with low-, medium- and high-dose Ang IV, AT4R antagonist divalinal, FoxO1 inhibitor AS1842856 (AS), or their combinations. In vitro, H9C2 cardiomyocytes and cardiac fibroblasts were treated with different concentrations of glucose, low-, medium- and high-dose Ang IV, divalinal, FoxO1-overexpression plasmid (FoxO1-OE), AS, or their combinations. Results: Ang IV treatment dose-dependently attenuated left ventricular dysfunction, fibrosis, and myocyte apoptosis in diabetic mice. Besides, enhanced autophagy and FoxO1 protein expression by diabetes were dose-dependently suppressed by Ang IV treatment. However, these cardioprotective effects of Ang IV were completely abolished by divalinal administration. Bioinformatics analysis revealed that the differentially expressed genes were enriched in autophagy, apoptosis, and FoxO signaling pathways among control, diabetes, and diabetes+high-dose Ang IV groups. Similar to Ang IV, AS treatment ameliorated diabetic cardiomyopathy in mice. In vitro, high glucose stimulation increased collagen expression, apoptosis, overactive autophagy flux and FoxO1 nuclear translocation in cardiomyocytes, and upregulated collagen and FoxO1 expression in cardiac fibroblasts, which were substantially attenuated by Ang IV treatment. However, these protective effects of Ang IV were completely blocked by the use of divalinal or FoxO1-OE, and these detrimental effects were reversed by the additional administration of AS. Conclusions: Ang IV treatment dose-dependently attenuated left ventricular dysfunction and remodeling in a mouse model of diabetic cardiomyopathy, and the mechanisms involved stimulation of AT4R and suppression of FoxO1-mediated fibrosis, apoptosis, and overactive autophagy.
Collapse
|
12
|
ACE2 as therapeutic agent. Clin Sci (Lond) 2021; 134:2581-2595. [PMID: 33063820 DOI: 10.1042/cs20200570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
The angiotensin-converting enzyme 2 (ACE2) has emerged as a critical regulator of the renin-angiotensin system (RAS), which plays important roles in cardiovascular homeostasis by regulating vascular tone, fluid and electrolyte balance. ACE2 functions as a carboxymonopeptidase hydrolyzing the cleavage of a single C-terminal residue from Angiotensin-II (Ang-II), the key peptide hormone of RAS, to form Angiotensin-(1-7) (Ang-(1-7)), which binds to the G-protein-coupled Mas receptor and activates signaling pathways that counteract the pathways activated by Ang-II. ACE2 is expressed in a variety of tissues and overwhelming evidence substantiates the beneficial effects of enhancing ACE2/Ang-(1-7)/Mas axis under many pathological conditions in these tissues in experimental models. This review will provide a succinct overview on current strategies to enhance ACE2 as therapeutic agent, and discuss limitations and future challenges. ACE2 also has other functions, such as acting as a co-factor for amino acid transport and being exploited by the severe acute respiratory syndrome coronaviruses (SARS-CoVs) as cellular entry receptor, the implications of these functions in development of ACE2-based therapeutics will also be discussed.
Collapse
|
13
|
Yi Y, Xu Y, Jiang H, Wang J. Cardiovascular Disease and COVID-19: Insight From Cases With Heart Failure. Front Cardiovasc Med 2021; 8:629958. [PMID: 33791346 PMCID: PMC8005527 DOI: 10.3389/fcvm.2021.629958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/22/2021] [Indexed: 12/21/2022] Open
Abstract
Recent evidence indicates that a large proportion of deaths from coronavirus disease 2019 (COVID-19) can be attributed to cardiovascular disease, including acute myocardial infarction, arrhythmias and heart failure. Indeed, severe infection increases the risk of heart failure among patients with COVID-19. In most patients, heart failure arises from complex interactions between pre-existing conditions, cardiac injury, renin-angiotensin system activation, and the effects of systemic inflammation on the cardiovascular system. In this review, we summarize current knowledge regarding pathogen-driven heart failure occurring during treatment for COVID-19, the potential effects of commonly used cardiovascular and anti-infective drugs in these patients, and possible directions for establishing a theoretical basis for clinical treatment.
Collapse
Affiliation(s)
- Yang Yi
- Department of Cardiology Fourth Ward, Xinjiang Medical University Affiliated Hospital of Traditional Chinese Medicine, Urumqi, China
| | - Yanan Xu
- Department of Respiratory Medicine, The People's Hospital of Xuancheng City, Xuancheng, China
| | - Haibing Jiang
- Department of Cardiology Fourth Ward, Xinjiang Medical University Affiliated Hospital of Traditional Chinese Medicine, Urumqi, China
| | - Jun Wang
- Department of Cardiology, The People's Hospital of Xuancheng City, Anhui, China
| |
Collapse
|
14
|
Zhu H, Zhang L, Ma Y, Zhai M, Xia L, Liu J, Yu S, Duan W. The role of SARS-CoV-2 target ACE2 in cardiovascular diseases. J Cell Mol Med 2021; 25:1342-1349. [PMID: 33443816 PMCID: PMC7875924 DOI: 10.1111/jcmm.16239] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/03/2020] [Accepted: 12/14/2020] [Indexed: 12/24/2022] Open
Abstract
SARS-CoV-2, the virus responsible for the global coronavirus disease (COVID-19) pandemic, attacks multiple organs of the human body by binding to angiotensin-converting enzyme 2 (ACE2) to enter cells. More than 20 million people have already been infected by the virus. ACE2 is not only a functional receptor of COVID-19 but also an important endogenous antagonist of the renin-angiotensin system (RAS). A large number of studies have shown that ACE2 can reverse myocardial injury in various cardiovascular diseases (CVDs) as well as is exert anti-inflammatory, antioxidant, anti-apoptotic and anticardiomyocyte fibrosis effects by regulating transforming growth factor beta, mitogen-activated protein kinases, calcium ions in cells and other major pathways. The ACE2/angiotensin-(1-7)/Mas receptor axis plays a decisive role in the cardiovascular system to combat the negative effects of the ACE/angiotensin II/angiotensin II type 1 receptor axis. However, the underlying mechanism of ACE2 in cardiac protection remains unclear. Some approaches for enhancing ACE2 expression in CVDs have been suggested, which may provide targets for the development of novel clinical therapies. In this review, we aimed to identify and summarize the role of ACE2 in CVDs.
Collapse
Affiliation(s)
- Hanzhao Zhu
- Department of Cardiovascular SurgeryThe First Affiliated HospitalThe Air Force Medical UniversityXi’anChina
| | - Liyun Zhang
- Department of Cardiovascular SurgeryThe First Affiliated HospitalThe Air Force Medical UniversityXi’anChina
| | - Yubo Ma
- Department of Dermatology and VenereologyPeking University First HospitaBeijingChina
| | - Mengen Zhai
- Department of Cardiovascular SurgeryThe First Affiliated HospitalThe Air Force Medical UniversityXi’anChina
| | - Lin Xia
- Department of Cardiovascular SurgeryThe First Affiliated HospitalThe Air Force Medical UniversityXi’anChina
| | - Jincheng Liu
- Department of Cardiovascular SurgeryThe First Affiliated HospitalThe Air Force Medical UniversityXi’anChina
| | - Shiqiang Yu
- Department of Cardiovascular SurgeryThe First Affiliated HospitalThe Air Force Medical UniversityXi’anChina
| | - Weixun Duan
- Department of Cardiovascular SurgeryThe First Affiliated HospitalThe Air Force Medical UniversityXi’anChina
| |
Collapse
|
15
|
Xu YW, Xu ZD, An R, Zhang H, Wang XH. Revealing the synergistic mechanism of Shenfu Decoction for anti-heart failure through network pharmacology strategy. Chin J Nat Med 2021; 18:536-549. [PMID: 32616194 DOI: 10.1016/s1875-5364(20)30064-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Indexed: 12/09/2022]
Abstract
The present study was designed to investigate the targets and synergistic mechanism of Shenfu Decoction (SFD) in the treatment of heart failure. A heart failure animal models was established to evaluate the pharmacological effects of SFD for anti-heart failure, then constructed ingredient-target interaction network by developing ingredient and target databases, the Discovery sdudio software was used for molecular docking. In addition, we validated the predicted protein targets of active ingredients in SFD by using surface plasmon resonance (SPR) technology. Our results demonstrated that SFD could enhance ejection fraction, alleviate myocardial histopathological characteristics, and reduce the level of angiotensin converting enzyme (ACE), aldosterone (ALD), atrial natriuretic polypeptide (ANP) and Renin (REN) in heart failure rat model. In addition, the ingredient database including 349 constituents and target database including 236 proteins were established, and 75 proteins were screened and identified by molecular docking strategy. 22 core target proteins were identified through network pharmacology, and the component-core target network was constructed. Finally, the affinity between the compounds and targets were verified by the SPR analysis method. The present study suggested that SFD may act on ACE 2, REN, ACE, ICAM-1, EGF, HTR2B, PARP1, NPPB and other proteins through AC, BAC, ACN, Re, Rg1, Rb1 to exert synergistic effects against heart failure.
Collapse
Affiliation(s)
- Yan-Wen Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhen-Dong Xu
- Department of Anesthesiology and Critical Care, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Rui An
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hai Zhang
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China.
| | - Xin-Hong Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
16
|
Gul R, Kim UH, Alfadda AA. Renin-angiotensin system at the interface of COVID-19 infection. Eur J Pharmacol 2021; 890:173656. [PMID: 33086029 PMCID: PMC7568848 DOI: 10.1016/j.ejphar.2020.173656] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 11/30/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been recognized as a potential entry receptor for SARS-CoV-2 infection. Binding of SARS-CoV-2 to ACE2 allows engagement with pulmonary epithelial cells and pulmonary infection with the virus. ACE2 is an essential component of renin-angiotensin system (RAS), and involved in promoting protective effects to counter-regulate angiotensin (Ang) II-induced pathogenesis. The use of angiotensin receptor blockers (ARBs) and ACE inhibitors (ACEIs) was implicitly negated during the early phase of COVID-19 pandemic, considering the role of these antihypertensive agents in enhancing ACE2 expression thereby promoting the susceptibility to SARS-CoV-2. However, no clinical data has supported this assumption, but indeed evidence demonstrates that ACEIs and ARBs, besides their cardioprotective effects in COVID-19 patients with cardiovascular diseases, might also be beneficial in acute lung injuries by preserving the ACE2 function and switching the balance from deleterious ACE/Ang II/AT1 receptor axis towards a protective ACE2/Ang (1-7)/Mas receptor axis.
Collapse
Affiliation(s)
- Rukhsana Gul
- Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia.
| | - Uh-Hyun Kim
- Department of Biochemistry & National Creative Research Laboratory for Ca(2+) Signaling, Chonbuk National University Medical School, Jeonju, 54907, Republic of Korea
| | - Assim A Alfadda
- Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia; Department of Medicine, College of Medicine, King Saud University, PO Box 2925, Riyadh, 11461, Saudi Arabia; Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Moin ASM, Sathyapalan T, Atkin SL, Butler AE. Renin-Angiotensin System overactivation in polycystic ovary syndrome, a risk for SARS-CoV-2 infection? Metabol Open 2020; 7:100052. [PMID: 32838280 PMCID: PMC7434315 DOI: 10.1016/j.metop.2020.100052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 01/08/2023] Open
Abstract
Background The SARS-CoV-2 coronavirus gains entry to target cells via the angiotensin-converting enzyme 2 (ACE2) receptor present on cells in blood vessels, lungs, heart, intestines, and kidneys. Renin-Angiotensin System (RAS) overactivity has also been described in metabolic syndrome, type 2 diabetes (T2D) and obesity, conditions shared by women with polycystic ovary syndrome (PCOS) We hypothesized that RAS overactivity may be present in PCOS. Methods We determined plasma levels of RAS-related proteins in a cohort of age matched control women (n = 97) and women with PCOS (n = 146). Plasma levels of RAS-related proteins (ACE2, Renin and Angiotensinogen (AGT)) were determined by Slow Off-rate Modified Aptamer (SOMA)-scan plasma protein measurement. Results PCOS women had a higher BMI (p < 0.001), systolic (p < 0.0001) and diastolic (p < 0.05) blood pressure, waist circumference (p < 0.0001), testosterone (p < 0.0001), free androgen index (p < 0.0001) and CRP (p < 0.0001). Renin was elevated in PCOS (p < 0.05) and angiotensinogen was lower in PCOS (p < 0.05), indicating overactivity of the RAS system in PCOS. ACE2 levels were lower in PCOS (p < 0.05), suggesting that PCOS women are at risk for development of hypertension. Conclusion RAS proteins levels differed between PCOS and control women, suggesting that the insulin resistance inherent in PCOS may predispose these women to more severe COVID-19 infection.
Collapse
Affiliation(s)
- Abu Saleh Md Moin
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | | | | | - Alexandra E. Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
- Corresponding author.
| |
Collapse
|
18
|
Guzik TJ, Mohiddin SA, Dimarco A, Patel V, Savvatis K, Marelli-Berg FM, Madhur MS, Tomaszewski M, Maffia P, D’Acquisto F, Nicklin SA, Marian AJ, Nosalski R, Murray EC, Guzik B, Berry C, Touyz RM, Kreutz R, Wang DW, Bhella D, Sagliocco O, Crea F, Thomson EC, McInnes IB. COVID-19 and the cardiovascular system: implications for risk assessment, diagnosis, and treatment options. Cardiovasc Res 2020; 116:1666-1687. [PMID: 32352535 PMCID: PMC7197627 DOI: 10.1093/cvr/cvaa106] [Citation(s) in RCA: 898] [Impact Index Per Article: 179.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
The novel coronavirus disease (COVID-19) outbreak, caused by SARS-CoV-2, represents the greatest medical challenge in decades. We provide a comprehensive review of the clinical course of COVID-19, its comorbidities, and mechanistic considerations for future therapies. While COVID-19 primarily affects the lungs, causing interstitial pneumonitis and severe acute respiratory distress syndrome (ARDS), it also affects multiple organs, particularly the cardiovascular system. Risk of severe infection and mortality increase with advancing age and male sex. Mortality is increased by comorbidities: cardiovascular disease, hypertension, diabetes, chronic pulmonary disease, and cancer. The most common complications include arrhythmia (atrial fibrillation, ventricular tachyarrhythmia, and ventricular fibrillation), cardiac injury [elevated highly sensitive troponin I (hs-cTnI) and creatine kinase (CK) levels], fulminant myocarditis, heart failure, pulmonary embolism, and disseminated intravascular coagulation (DIC). Mechanistically, SARS-CoV-2, following proteolytic cleavage of its S protein by a serine protease, binds to the transmembrane angiotensin-converting enzyme 2 (ACE2) -a homologue of ACE-to enter type 2 pneumocytes, macrophages, perivascular pericytes, and cardiomyocytes. This may lead to myocardial dysfunction and damage, endothelial dysfunction, microvascular dysfunction, plaque instability, and myocardial infarction (MI). While ACE2 is essential for viral invasion, there is no evidence that ACE inhibitors or angiotensin receptor blockers (ARBs) worsen prognosis. Hence, patients should not discontinue their use. Moreover, renin-angiotensin-aldosterone system (RAAS) inhibitors might be beneficial in COVID-19. Initial immune and inflammatory responses induce a severe cytokine storm [interleukin (IL)-6, IL-7, IL-22, IL-17, etc.] during the rapid progression phase of COVID-19. Early evaluation and continued monitoring of cardiac damage (cTnI and NT-proBNP) and coagulation (D-dimer) after hospitalization may identify patients with cardiac injury and predict COVID-19 complications. Preventive measures (social distancing and social isolation) also increase cardiovascular risk. Cardiovascular considerations of therapies currently used, including remdesivir, chloroquine, hydroxychloroquine, tocilizumab, ribavirin, interferons, and lopinavir/ritonavir, as well as experimental therapies, such as human recombinant ACE2 (rhACE2), are discussed.
Collapse
Affiliation(s)
- Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Saidi A Mohiddin
- Barts Heart Center, St Bartholomew’s NHS Trust, London, UK
- William Harvey Institute Queen Mary University of London, London, UK
| | | | - Vimal Patel
- Barts Heart Center, St Bartholomew’s NHS Trust, London, UK
| | | | | | - Meena S Madhur
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Stuart A Nicklin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ali J Marian
- Department of Medicine, Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ryszard Nosalski
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Eleanor C Murray
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Bartlomiej Guzik
- Jagiellonian University Medical College, Institute of Cardiology, Department of Interventional Cardiology; John Paul II Hospital, Krakow, Poland
| | - Colin Berry
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Reinhold Kreutz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institut für Klinische Pharmakologie und Toxikologie, Germany
| | - Dao Wen Wang
- Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - David Bhella
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, UK
| | - Orlando Sagliocco
- Emergency Department, Intensive Care Unit; ASST Bergamo Est Bolognini Hospital Bergamo, Italy
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Emma C Thomson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, UK
- Department of Infectious Diseases, Queen Elizabeth University Hospital, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
19
|
Dörr K, Kammer M, Reindl-Schwaighofer R, Lorenz M, Loewe C, Marculescu R, Erben R, Oberbauer R. Effect of etelcalcetide on cardiac hypertrophy in hemodialysis patients: a randomized controlled trial (ETECAR-HD). Trials 2019; 20:601. [PMID: 31651370 PMCID: PMC6813957 DOI: 10.1186/s13063-019-3707-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/06/2019] [Indexed: 02/15/2023] Open
Abstract
Background Fibroblast growth factor 23 (FGF23) is associated with left ventricular hypertrophy (LVH) in patients with chronic kidney disease, and calcimimetic therapy reduces plasma concentrations of FGF23. It remains unknown whether treatment with the calcimimetic etelcalcetide (ETL) reduces LVH in patients on hemodialysis. Methods/design This single-blinded randomized trial of 12 months duration will test the effects of ETL compared with alfacalcidol on LVH and cardiac fibrosis in maintenance hemodialysis patients with secondary hyperparathyroidism. Both treatment regimens will be titrated to equally suppress secondary hyperparathyroidism while alfacalcidol treatment causes an increase and ETL a decrease in FGF23, respectively. Patients treated thrice weekly with hemodialysis for ≥ 3 months and ≤ 3 years with parathyroid hormone levels ≥ 300 pg/ml and LVH will be enrolled in the study. The primary study endpoint is change from baseline to 12 months in left ventricular mass index (LVMI; g/m2) measured by cardiac magnetic resonance imaging. Sample size calculations showed that 62 randomized patients will be necessary to detect a difference in LVMI of at least 20 g/m2 between the two groups at 12 months. Due to the strong association of volume overload and LVH, randomization will be stratified by residual kidney function, and regular body composition monitoring will be performed to control the volume status of patients. Study medication will be administered intravenously by the dialysis nurses after every hemodialysis session, thus omitting adherence issues. Secondary study endpoints are cardiac parameters measured by echocardiography, biomarker concentrations of bone metabolism (FGF23, vitamin D, parathyroid hormone, calcium, phosphate, s-Klotho), cardiac markers (pro-brain natriuretic peptide, pre- and postdialysis troponin T) and metabolites of the renin–angiotensin–aldosterone cascade (angiotensin I (Ang I), Ang II, Ang-(1–7), Ang-(1–5), Ang-(1–9), and aldosterone). Discussion The causal inference and pathophysiology of LVH regression by FGF23 reduction using calcimimetic treatment has not yet been shown. This intervention study has the potential to discover a new strategy for the treatment of cardiac hypertrophy and fibrosis in patients on maintenance hemodialysis. It might be speculated that successful treatment of cardiac morphology will also reduce the risk of cardiac death in this population. Trial registration European Clinical Trials Database, EudraCT number 2017-000222-35; ClinicalTrials.gov, NCT03182699. Registered on
Collapse
Affiliation(s)
- Katharina Dörr
- Department of Nephrology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Michael Kammer
- Department of Nephrology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.,Center for Medical Statistics, Informatics and Intelligent Systems (CeMSIIS), Section for Clinical Biometrics, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | | | - Matthias Lorenz
- Vienna Dialysis Center, Kapellenweg 37, 1220, Vienna, Austria
| | - Christian Loewe
- Division of Cardiovascular and Interventional Radiology, Department of Bioimaging and Image-Guided Intervention, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Rodrig Marculescu
- Laboratory Medicine, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Reinhold Erben
- Physiology, Pathophysiology, and Experimental Endocrinology, VetMed Vienna, Veterinärplatz 1, Vienna, Austria
| | - Rainer Oberbauer
- Department of Nephrology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.
| |
Collapse
|
20
|
Fan Z, Wu G, Yue M, Ye J, Chen Y, Xu B, Shu Z, Zhu J, Lu N, Tan X. Hypertension and hypertensive left ventricular hypertrophy are associated with ACE2 genetic polymorphism. Life Sci 2019; 225:39-45. [PMID: 30917908 DOI: 10.1016/j.lfs.2019.03.059] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/19/2019] [Accepted: 03/23/2019] [Indexed: 02/05/2023]
Abstract
AIMS Renin-angiotensin system modulates cardiac structure independent of blood pressure. The present study aimed at investigating whether single nucleotide polymorphism (SNP) and haplotype of angiotensin converting enzyme 2 (ACE2) could influence blood pressure and the susceptibility to hypertensive left ventricular hypertrophy (LVH). SUBJECTS AND METHODS A total of 647 patients (347 females and 300 males) with newly diagnosed mild to moderate essential hypertension were enrolled in a blood pressure matched, case-control study. Four ACE2 tagSNPs (rs2074192, rs4646176, rs4646155 and rs2106809) were genotyped and major haplotypes consisting of these four SNPs were reconstructed for all subjects. KEY FINDINGS In females, minor alleles of ACE2 rs2074192 and rs2106809 respectively conferred a 2.1 and 2.0 fold risk for LVH. ACE2 haplotype TCGT increased the risk for LVH while another haplotype CCGC decreased the risk in females. The covariates-adjusted mean left ventricular mass index was 11% greater in TCGT haplotype carriers than in noncarriers in women. In females, the covariates-adjusted mean systolic blood pressure was 3.4 mm Hg lower in CCGC haplotype carriers than in noncarriers. In males, the covariates-adjusted mean systolic blood pressure was 2.4 mm Hg lower in CCGC haplotype carriers than in noncarriers. SIGNIFICANCE ACE2 tagSNPs rs2074192 and rs2106809 as well as major haplotypes CCGC and TCGT may serve as novel risk markers for LVH in hypertensive patients.
Collapse
Affiliation(s)
- Zhimin Fan
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Guihai Wu
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Minghui Yue
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jianfeng Ye
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yequn Chen
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Bayi Xu
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Zhouwu Shu
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jinxiu Zhu
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Nan Lu
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xuerui Tan
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China.
| |
Collapse
|
21
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 693] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
22
|
Castardeli C, Sartório CL, Pimentel EB, Forechi L, Mill JG. The ACE 2 activator diminazene aceturate (DIZE) improves left ventricular diastolic dysfunction following myocardial infarction in rats. Biomed Pharmacother 2018; 107:212-218. [DOI: 10.1016/j.biopha.2018.07.170] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 01/10/2023] Open
|
23
|
Araújo AAD, Araújo LDS, Medeiros CACXD, Leitão RFDC, Brito GADC, Costa DVDS, Guerra GCB, Garcia VB, Lima MLDS, Araújo Junior RFD. Protective effect of angiotensin II receptor blocker against oxidative stress and inflammation in an oral mucositis experimental model. J Oral Pathol Med 2018; 47:972-984. [DOI: 10.1111/jop.12775] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/23/2018] [Accepted: 07/30/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Aurigena Antunes de Araújo
- Postgraduate Programs in Public Health and Pharmaceutical Science Department of Biophysics and Pharmacology Federal University of Rio Grande Norte Natal Brazil
| | | | - Caroline Addison Carvalho Xavier de Medeiros
- Department of Biophysics and Pharmacology UFRN Natal Brazil
- Postgraduate Program in Biological Science and Rede Nordeste de Biotecnologia/Renorbio Federal University of Rio Grande Norte Natal Brazil
| | | | - Gerly Anne de Castro Brito
- Postgraduate Programs in Pharmacology and Morphology Department of Morphology/Pharmacology Federal University of Ceará Fortaleza Brazil
| | | | - Gerlane Coelho Bernardo Guerra
- Postgraduate Programs in Postgraduate Program in Biological Science/Pharmaceutical Science Department of Biophysical and Pharmacology UFRN Natal Brazil
| | | | | | | |
Collapse
|
24
|
Angiotensin-converting enzyme 2 overexpression protects against doxorubicin-induced cardiomyopathy by multiple mechanisms in rats. Oncotarget 2018; 8:24548-24563. [PMID: 28445944 PMCID: PMC5421869 DOI: 10.18632/oncotarget.15595] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/13/2017] [Indexed: 11/25/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is considered a potential therapeutic target of the renin-angiotensin system (RAS) for the treatment of cardiovascular diseases. We aimed to explore the effects of ACE2 overexpression on doxorubicin-induced cardiomyopathy in rats. Rats were randomly divided into treatment and control groups. The rats of treatment group were injected intraperitoneally with 6 doses of doxorubicin (2.5 mg/kg) within a period of two weeks. Two weeks after the initial injection of doxorubicin, these rats were randomly divided into Mock, Ad-EGFP, Ad-ACE2, and Cilazapril groups. The rats of Ad-EGFP and Ad-ACE2 groups received intramyocardial injection of Ad-EGFP and Ad-ACE2, respectively. The rats of Cilazapril group received cilazapril (10 mg/kg/day) via intragastric intubation. Apoptosis, inflammation, oxidative stress, cardiac function, the extent of myocardial fibrosis, and levels of ACE2, ACE, angiotensin II (AngII), and angiotensin (1–7) were evaluated. Four weeks after ACE2 gene transfer, the Ad-ACE2 group showed not only reduced apoptosis, inflammatory response, oxidative stress, left ventricular (LV) volume, extent of myocardial fibrosis and mortality of rats, but also increased LV ejection fraction and ACE2 expression level compared with the Mock and Ad-EGFP groups. ACE2 overexpression was superior to cilazapril in improving doxorubicin-induced cardiomyopathy. The putative mechanisms may involve activation of the AMPK and PI3K-AKT pathways, inhibition of the ERK pathway, decrease of TGF-β1 expression, and interactions of shifting RAS components, such as decreased myocardium AngII levels, increased myocardium Ang (1–7) levels, and reduced ACE expression. Thus, ACE2 may be a novel therapeutic approach to prevent and treat doxorubicin-induced cardiomyopathy.
Collapse
|
25
|
Dai H, Zhang X, Yang Z, Li J, Zheng J. Effects of Baicalin on Blood Pressure and Left Ventricular Remodeling in Rats with Renovascular Hypertension. Med Sci Monit 2017. [PMID: 28622281 PMCID: PMC5484556 DOI: 10.12659/msm.902536] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background This study aimed to explore the effect of baicalin, which is a kind of bioactive flavonoid, on blood pressure and left ventricular remodeling in rats with renovascular hypertension. Material/Methods A total of 40 male Wistar rats were randomly assigned into sham-operation (n=10) and renal hypertension model groups (2-kidney-1 clip; 2K-1C, n=30). The rats in the renal hypertension model group were randomly subdivided into 2K-1C (n=13) and 2K-1C/Baicalin groups (n=14). The cardiac function indexes were determined after 4 weeks. The morphological changes in the myocardial tissue were observed using hematoxylin and eosin and Masson staining. The myocardial apoptosis was detected using the terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling method, and the expression of C/EBP homologous protein and caspase-3 was monitored by Western blot. The expression of GRP78 and GRP94 in myocardial cells of rats was detected by qPCR and Western blot technology. Results No significant change in blood pressure was observed in the 2K-1C/Baicalin group compared with the 2K-1C group, but the indexes of left ventricular remodeling significantly improved. Pathological myocardial fibrosis and expression of fibrosis-related factors significantly decreased in the 2K-1C/Baicalin group compared with the 2K-1C group. The expression of glucose-regulated protein (GRP)78, GRP94, CHOP, and caspase-3, and apoptosis of cardiomyocytes also decreased in the 2K-1C/Baicalin group. Conclusions Baicalin has no significant antihypertensive effect, but reduced pathological changes in the myocardium, alleviated endoplasmic reticulum stress, and reduced myocardial apoptosis, reverting left ventricular remodeling in rats with renovascular hypertension.
Collapse
Affiliation(s)
- Hualei Dai
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Xinjin Zhang
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Zhigang Yang
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Jianmei Li
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Jialin Zheng
- Department of Cardiology, The Second People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| |
Collapse
|
26
|
Tamargo M, Tamargo J. Future drug discovery in renin-angiotensin-aldosterone system intervention. Expert Opin Drug Discov 2017; 12:827-848. [PMID: 28541811 DOI: 10.1080/17460441.2017.1335301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Renin-angiotensin-aldosterone system inhibitors (RAASIs), including angiotensin-converting enzyme inhibitors, angiotensin AT1 receptor blockers and mineralocorticoid receptor antagonists (MRAs), are the cornerstone for the treatment of cardiovascular and renal diseases. Areas covered: The authors searched MEDLINE, PubMed and ClinicalTrials.gov to identify eligible full-text English language papers. Herein, the authors discuss AT2-receptor agonists and ACE2/angiotensin-(1-7)/Mas-receptor axis modulators, direct renin inhibitors, brain aminopeptidase A inhibitors, biased AT1R blockers, chymase inhibitors, multitargeted drugs, vaccines and aldosterone receptor antagonists as well as aldosterone synthase inhibitors. Expert opinion: Preclinical studies have demonstrated that activation of the protective axis of the RAAS represents a novel therapeutic strategy for treating cardiovascular and renal diseases, but there are no clinical trials supporting our expectations. Non-steroidal MRAs might become the third-generation of MRAs for the treatment of heart failure, diabetes mellitus and chronic kidney disease. The main challenge for these new drugs is that conventional RAASIs are safe, effective and cheap generics. Thus, the future of new RAASIs will be directed by economical/strategic reasons.
Collapse
Affiliation(s)
- Maria Tamargo
- a Department of Cardiology , Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV , Madrid , Spain
| | - Juan Tamargo
- b Department of Pharmacology , School of Medicine, University Complutense, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV , Madrid , Spain
| |
Collapse
|
27
|
Abstract
Chronic kidney disease (CKD) is associated with an increased risk of cardiovascular mortality, infections, and impaired cognitive function. It is characterized by excessively increased levels of the phosphaturic hormone fibroblast growth factor 23 (FGF23) and a deficiency of its co-receptor Klotho. Despite the important physiological effect of FGF23 in maintaining phosphate homeostasis, there is increasing evidence that higher FGF23 levels are a risk factor for mortality and cardiovascular disease. FGF23 directly induces left ventricular hypertrophy via activation of the FGF receptor 4/calcineurin/nuclear factor of activated T cells signaling pathway. By contrast, the impact of FGF23 on endothelial function and the development of atherosclerosis are poorly understood. The results of recent experimental studies indicate that FGF23 directly impacts on hippocampal neurons and may thereby impair learning and memory function in CKD patients. Finally, it has been shown that FGF23 interferes with the immune system by directly acting on polymorphonuclear leukocytes and macrophages. In this review, we discuss recent data from clinical and experimental studies on the extrarenal effects of FGF23 with respect to the cardiovascular, central nervous, and immune systems.
Collapse
|
28
|
MiRNA-30e mediated cardioprotection of ACE2 in rats with Doxorubicin-induced heart failure through inhibiting cardiomyocytes autophagy. Life Sci 2017; 169:69-75. [DOI: 10.1016/j.lfs.2016.09.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 08/26/2016] [Accepted: 09/11/2016] [Indexed: 12/13/2022]
|
29
|
Angiotensin-converting enzyme 2 inhibits high-mobility group box 1 and attenuates cardiac dysfunction post-myocardial ischemia. J Mol Med (Berl) 2016; 94:37-49. [PMID: 26498282 DOI: 10.1007/s00109-015-1356-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 08/03/2015] [Accepted: 09/24/2015] [Indexed: 12/23/2022]
Abstract
High-mobility group box 1 (HMGB1) triggers and amplifies inflammation cascade following ischemic injury, and its elevated levels are associated with adverse clinical outcomes in patients with myocardial infarction (MI). Angiotensin-converting enzyme 2 (ACE2), a key member of vasoprotective axis of the renin-angiotensin system (RAS), regulates cardiovascular functions and exerts beneficial effects in cardiovascular disease. However, the association between HMGB1 and ACE2 has not been studied. We hypothesized that overexpression of ACE2 provides cardioprotective effects against MI via inhibiting HMGB1 and inflammation. ACE2 knock-in (KI) mice and littermate wild-type (WT) controls were subjected to either sham or coronary artery ligation surgery to induce MI. Heart function was assessed 4 weeks after surgery using echocardiography and Millar catheterization. Tissues were collected for histology and analysis of the expression of HMGB1, RAS components, and inflammatory cytokines. ACE2 in the heart of the ACE2 KI mice was 58-fold higher than WT controls. ACE2-MI mice exhibited a remarkable preservation of cardiac function and reduction of infarct size in comparison to WT-MI mice. Notably, ACE2 overexpression significantly reduced the MI-induced increase in apoptosis, macrophage infiltration, and HMGB1 and proinflammatory cytokine expression (TNF-α and IL-6). Moreover, in an in vitro study, ACE2 activation prevented the hypoxia-induced cell death and upregulation of HMGB1 in adult cardiomyocytes. This protective effect is correlated with downregulation of HMGB1 and downstream proinflammatory cascades, which could be useful for the development of novel treatment for ischemic heart disease.
Collapse
|
30
|
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
|
31
|
Patel VB, Zhong JC, Grant MB, Oudit GY. Role of the ACE2/Angiotensin 1-7 Axis of the Renin-Angiotensin System in Heart Failure. Circ Res 2016; 118:1313-26. [PMID: 27081112 DOI: 10.1161/circresaha.116.307708] [Citation(s) in RCA: 604] [Impact Index Per Article: 67.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/19/2016] [Indexed: 11/16/2022]
Abstract
Heart failure (HF) remains the most common cause of death and disability, and a major economic burden, in industrialized nations. Physiological, pharmacological, and clinical studies have demonstrated that activation of the renin-angiotensin system is a key mediator of HF progression. Angiotensin-converting enzyme 2 (ACE2), a homolog of ACE, is a monocarboxypeptidase that converts angiotensin II into angiotensin 1-7 (Ang 1-7) which, by virtue of its actions on the Mas receptor, opposes the molecular and cellular effects of angiotensin II. ACE2 is widely expressed in cardiomyocytes, cardiofibroblasts, and coronary endothelial cells. Recent preclinical translational studies confirmed a critical counter-regulatory role of ACE2/Ang 1-7 axis on the activated renin-angiotensin system that results in HF with preserved ejection fraction. Although loss of ACE2 enhances susceptibility to HF, increasing ACE2 level prevents and reverses the HF phenotype. ACE2 and Ang 1-7 have emerged as a key protective pathway against HF with reduced and preserved ejection fraction. Recombinant human ACE2 has been tested in phase I and II clinical trials without adverse effects while lowering and increasing plasma angiotensin II and Ang 1-7 levels, respectively. This review discusses the transcriptional and post-transcriptional regulation of ACE2 and the role of the ACE2/Ang 1-7 axis in cardiac physiology and in the pathophysiology of HF. The pharmacological and therapeutic potential of enhancing ACE2/Ang 1-7 action as a novel therapy for HF is highlighted.
Collapse
Affiliation(s)
- Vaibhav B Patel
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Jiu-Chang Zhong
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Maria B Grant
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.)
| | - Gavin Y Oudit
- From the Division of Cardiology, Department of Medicine (V.B.P., G.Y.O.), Mazankowski Alberta Heart Institute (V.B.P., G.Y.O.), and Department of Physiology (G.Y.O.), University of Alberta, Edmonton, Canada; State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.-C.Z.); Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China (J.-C.Z.); and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis (M.B.G.).
| |
Collapse
|
32
|
Cardioprotective effects of diminazene aceturate in pressure-overloaded rat hearts. Life Sci 2016; 155:63-9. [DOI: 10.1016/j.lfs.2016.04.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 04/29/2016] [Accepted: 04/30/2016] [Indexed: 12/12/2022]
|
33
|
Bomb R, Heckle MR, Sun Y, Mancarella S, Guntaka RV, Gerling IC, Weber KT. Myofibroblast secretome and its auto-/paracrine signaling. Expert Rev Cardiovasc Ther 2016; 14:591-8. [PMID: 26818589 DOI: 10.1586/14779072.2016.1147348] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myofibroblasts (myoFb) are phenotypically transformed, contractile fibroblast-like cells expressing α-smooth muscle actin microfilaments. They are integral to collagen fibrillogenesis with scar tissue formation at sites of repair irrespective of the etiologic origins of injury or tissue involved. MyoFb can persist long after healing is complete, where their ongoing turnover of collagen accounts for a progressive structural remodeling of an organ (a.k.a. fibrosis, sclerosis or cirrhosis). Such persistent metabolic activity is derived from a secretome consisting of requisite components in the de novo generation of angiotensin (Ang) II. Autocrine and paracrine signaling induced by tissue AngII is expressed via AT1 receptor ligand binding to respectively promote: i) regulation of myoFb collagen synthesis via the fibrogenic cytokine TGF-β1-Smad pathway; and ii) dedifferentiation and protein degradation of atrophic myocytes immobilized and ensnared by fibrillar collagen at sites of scarring. Several cardioprotective strategies in the prevention of fibrosis and involving myofibroblasts are considered. They include: inducing myoFb apoptosis through inactivation of antiapoptotic proteins; AT1 receptor antagonist to interfere with auto-/paracrine myoFb signaling or to induce counterregulatory expression of ACE2; and attacking the AngII-AT1R-TGF-β1-Smad pathway by antibody or the use of triplex-forming oligonucleotides.
Collapse
Affiliation(s)
- Ritin Bomb
- a Division of Cardiovascular Diseases , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Mark R Heckle
- b Department of Medicine , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Yao Sun
- a Division of Cardiovascular Diseases , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Salvatore Mancarella
- c Department of Physiology , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Ramareddy V Guntaka
- d Department of Microbiology, Immunology and Biochemistry , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Ivan C Gerling
- e Division of Endocrinology , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Karl T Weber
- a Division of Cardiovascular Diseases , University of Tennessee Health Science Center , Memphis , TN , USA
| |
Collapse
|
34
|
Kovesdy CP, Quarles LD. FGF23 from bench to bedside. Am J Physiol Renal Physiol 2016; 310:F1168-74. [PMID: 26864938 DOI: 10.1152/ajprenal.00606.2015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/04/2016] [Indexed: 12/31/2022] Open
Abstract
There is a strong association between elevated circulating fibroblast growth factor-23 (FGF23) levels and adverse outcomes in patients with chronic kidney disease (CKD) of all stages. Initially discovered as a regulator of phosphate and vitamin D homeostasis, FGF23 has now been implicated in several pathophysiological mechanisms that may negatively impact the cardiovascular and renal systems. FGF23 is purported to have direct (off-target) effects in the myocardium, as well as canonical effects on FGF receptor/α-klotho receptor complexes in the kidney to activate the renin-angiotensin-aldosterone system, modulate soluble α-klotho levels, and increase sodium retention, to cause left ventricular hypertrophy (LVH). Conversely, FGF23 could be an innocent bystander produced in response to chronic inflammation or other processes associated with CKD that cause LVH and adverse cardiovascular outcomes. Further exploration of these complex mechanisms is needed before modulation of FGF23 can become a legitimate clinical target in CKD.
Collapse
Affiliation(s)
- Csaba P Kovesdy
- University of Tennessee Health Science Center, Memphis, Tennessee; and Memphis Veterans Affairs Medical Center, Memphis, Tennessee
| | - L Darryl Quarles
- University of Tennessee Health Science Center, Memphis, Tennessee; and
| |
Collapse
|
35
|
Murphy KT. The pathogenesis and treatment of cardiac atrophy in cancer cachexia. Am J Physiol Heart Circ Physiol 2015; 310:H466-77. [PMID: 26718971 DOI: 10.1152/ajpheart.00720.2015] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/29/2015] [Indexed: 02/08/2023]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by a progressive loss of skeletal muscle mass associated with significant functional impairment. In addition to a loss of skeletal muscle mass and function, many patients with cancer cachexia also experience cardiac atrophy, remodeling, and dysfunction, which in the field of cancer cachexia is described as cardiac cachexia. The cardiac alterations may be due to underlying heart disease, the cancer itself, or problems initiated by the cancer treatment and, unfortunately, remains largely underappreciated by clinicians and basic scientists. Despite recent major advances in the treatment of cancer, little progress has been made in the treatment of cardiac cachexia in cancer, and much of this is due to lack of information regarding the mechanisms. This review focuses on the cardiac atrophy associated with cancer cachexia, describing some of the known mechanisms and discussing the current and future therapeutic strategies to treat this condition. Above all else, improved awareness of the condition and an increased focus on identification of mechanisms and therapeutic targets will facilitate the eventual development of an effective treatment for cardiac atrophy in cancer cachexia.
Collapse
Affiliation(s)
- Kate T Murphy
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
36
|
Chen C, Li R, Ross RS, Manso AM. Integrins and integrin-related proteins in cardiac fibrosis. J Mol Cell Cardiol 2015; 93:162-74. [PMID: 26562414 DOI: 10.1016/j.yjmcc.2015.11.010] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/07/2015] [Accepted: 11/07/2015] [Indexed: 12/21/2022]
Abstract
Cardiac fibrosis is one of the major components of the healing mechanism following any injury of the heart and as such may contribute to both systolic and diastolic dysfunction in a range of pathophysiologic conditions. Canonically, it can occur as part of the remodeling process that occurs following myocardial infarction or that follows as a response to pressure overload. Integrins are cell surface receptors which act in both cellular adhesion and signaling. Most importantly, in the context of the continuously contracting myocardium, they are recognized as mechanotransducers. They have been implicated in the development of fibrosis in several organs, including the heart. This review will focus on the involvement of integrins and integrin-related proteins, in cardiac fibrosis, outlining the roles of these proteins in the fibrotic responses in specific cardiac pathologies, discuss some of the common end effectors (angiotensin II, transforming growth factor beta 1 and mechanical stress) through which integrins function and finally discuss how manipulation of this set of proteins may lead to new treatments which could prove useful to alter the deleterious effects of cardiac fibrosis.
Collapse
Affiliation(s)
- Chao Chen
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Ruixia Li
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Robert S Ross
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| | - Ana Maria Manso
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA 92093-0613, USA; Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA.
| |
Collapse
|
37
|
Dalpiaz PLM, Lamas AZ, Caliman IF, Ribeiro RF, Abreu GR, Moyses MR, Andrade TU, Gouvea SA, Alves MF, Carmona AK, Bissoli NS. Sex Hormones Promote Opposite Effects on ACE and ACE2 Activity, Hypertrophy and Cardiac Contractility in Spontaneously Hypertensive Rats. PLoS One 2015; 10:e0127515. [PMID: 26010093 PMCID: PMC4444272 DOI: 10.1371/journal.pone.0127515] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 04/16/2015] [Indexed: 11/25/2022] Open
Abstract
Background There is growing interest in sex differences and RAS components. However, whether gender influences cardiac angiotensin I-converting enzyme (ACE) and angiotensin-converting enzyme 2 (ACE2) activity is still unknown. In the present work, we determined the relationship between ACE and ACE2 activity, left ventricular function and gender in spontaneously hypertensive rats (SHRs). Methodology / Principal Findings Twelve-week-old female (F) and male (M) SHRs were divided into 2 experimental groups (n = 7 in each group): sham (S) and gonadectomized (G). Fifty days after gonadectomy, we measured positive and negative first derivatives (dP/dt maximum left ventricle (LV) and dP/dt minimum LV, respectively), hypertrophy (morphometric analysis) and ACE and ACE2 catalytic activity (fluorimetrically). Expression of calcium handling proteins was measured by western blot. Male rats exhibited higher cardiac ACE and ACE2 activity as well as hypertrophy compared to female rats. Orchiectomy decreased the activity of these enzymes and hypertrophy, while ovariectomy increased hypertrophy and ACE2, but did not change ACE activity. For cardiac function, the male sham group had a lower +dP/dt than the female sham group. After gonadectomy, the +dP/dt increased in males and reduced in females. The male sham group had a lower -dP/dt than the female group. After gonadectomy, the -dP/dt increased in the male and decreased in the female groups when compared to the sham group. No difference was observed among the groups in SERCA2a protein expression. Gonadectomy increased protein expression of PLB (phospholamban) and the PLB to SERCA2a ratio in female rats, but did not change in male rats. Conclusion Ovariectomy leads to increased cardiac hypertrophy, ACE2 activity, PLB expression and PLB to SERCA2a ratio, and worsening of hemodynamic variables, whereas in males the removal of testosterone has the opposite effects on RAS components.
Collapse
Affiliation(s)
- P. L. M. Dalpiaz
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
- * E-mail:
| | - A. Z. Lamas
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - I. F. Caliman
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - R. F. Ribeiro
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - G. R. Abreu
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - M. R. Moyses
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - T. U. Andrade
- Department of Pharmacy, University Vila Velha, Vila Velha, Espirito Santo, Brazil
| | - S. A. Gouvea
- Nucleus of Biotechnology, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - M. F. Alves
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - A. K. Carmona
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - N. S. Bissoli
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| |
Collapse
|
38
|
Wang J, Li N, Gao F, Song R, Zhu S, Geng Z. Balance between angiotensin converting enzyme and angiotensin converting enzyme 2 in patients with chronic heart failure. J Renin Angiotensin Aldosterone Syst 2015; 16:553-8. [PMID: 25869724 DOI: 10.1177/1470320315576257] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/10/2015] [Indexed: 11/16/2022] Open
Abstract
AIM It has been reported that angiotensin converting enzyme 2 (ACE2) is an endogenous counter-regulator of the renin-angiotensin-aldosterone system. However, angiotensin converting enzyme (ACE)/ACE2 balance in the development of human heart failure is not well established. METHODS Here we evaluated the expression of ACE and ACE2 at the mRNA and protein levels in the myocardium of 78 patients with mild or moderate to severe heart failure and in 13 cases with normal myocardium. RESULTS In the myocardium of patients with dilated or ischemic cardiomyopathy, ACE and ACE2 expression at the mRNA and protein levels was significantly increased compared with those in normal myocardium (P<0.01, P<0.01, respectively). The ratios of ACE/ACE2 mRNA and ACE/ACE2 were lower in the myocardium of patients with mild heart failure than those in normal myocardium but higher than those in patients with moderate to severe heart failure. CONCLUSIONS ACE and ACE2 expression at the mRNA and protein levels are significantly increased in the myocardium of patients with heart failure. The compensatory mechanism of patients with mild heart may cause the decreased ACE/ACE2 ratio. However, increased ACE/ACE2 ratios may induce angiotensin II over-activation and accelerate cardiac remodeling in patients with moderate to severe heart failure.
Collapse
Affiliation(s)
- Jiang Wang
- Department of Cardiology, Xinqiao Hospital, the Third Military Medical University, China
| | - Nan Li
- Department of Cardiology, Xinqiao Hospital, the Third Military Medical University, China
| | - Feng Gao
- Department of Cardiology, Xinqiao Hospital, the Third Military Medical University, China
| | - Rong Song
- Department of Cardiology, Xinqiao Hospital, the Third Military Medical University, China
| | - Shanjun Zhu
- Department of Cardiology, Xinqiao Hospital, the Third Military Medical University, China
| | - Zhaohua Geng
- Department of Cardiology, Xinqiao Hospital, the Third Military Medical University, China
| |
Collapse
|
39
|
Combination of angiotensin-(1-7) with perindopril is better than single therapy in ameliorating diabetic cardiomyopathy. Sci Rep 2015; 5:8794. [PMID: 25740572 DOI: 10.1038/srep08794] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/27/2015] [Indexed: 01/06/2023] Open
Abstract
We recently found that overexpression of angiotensin (Ang)-converting enzyme 2, which metabolizes Ang-II to Ang-(1-7) and Ang-I to Ang-(1-9), may improve left ventricular remodeling in diabetic cardiomyopathy. Here we aimed to test whether chronic infusion of Ang-(1-7) can dose-dependently ameliorate left ventricular remodeling and function in a rat model of diabetic cardiomyopathy and whether the combination of Ang-(1-7) and Ang-converting enzyme inhibition may be superior to single therapy. Our results showed that Ang-(1-7) treatment dose-dependently ameliorated left ventricular remodeling and dysfunction in diabetic rats by attenuating myocardial fibrosis, myocardial hypertrophy and myocyte apoptosis via both the Mas receptor and angiotensin II type 2 receptor. Furthermore, combining Ang-(1-7) with perindopril provided additional cardioprotection relative to single therapy. Ang-(1-7) administration provides a novel and promising approach for treatment of diabetic cardiomyopathy.
Collapse
|
40
|
Hao PP, Yang JM, Zhang MX, Zhang K, Chen YG, Zhang C, Zhang Y. Angiotensin-(1-7) treatment mitigates right ventricular fibrosis as a distinctive feature of diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol 2015; 308:H1007-19. [PMID: 25724492 DOI: 10.1152/ajpheart.00563.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/24/2015] [Indexed: 02/02/2023]
Abstract
In diabetic patients, left ventricular (LV) remodeling is highly prevalent; however, little is known about the impact of diabetes on right ventricular (RV) structure and function. We recently found that overexpression of angiotensin (ANG)-converting enzyme 2 (ACE2), which metabolizes ANG-II to ANG-(1-7) and ANG-I to ANG-(1-9), may improve LV remodeling in diabetic cardiomyopathy (DCM). Here, we aimed to assess whether LV remodeling and dysfunction are paralleled by RV alterations and the effects of ANG-(1-7) on RV remodeling in DCM. After 12 wk of diabetes induced by a single intraperitoneal injection of streptozotocin, rats were treated with saline, ANG-(1-7), perindopril, ANG-(1-7) plus perindopril, ANG-(1-7) plus Mas receptor antagonist A779, or ANG-(1-7) plus ANG-II type 2 receptor antagonist PD123319 for 4 wk. RV remodeling in diabetic rats was indicated by fibrosis of the RV free wall in the absence of hypertrophy and apoptosis. Treatment with ANG-(1-7) prevented diabetes-induced RV fibrosis and dysfunction. ANG-(1-7) (800 ng·kg(-1)·min(-1)) was superior to perindopril in improving RV fibrosis. The major mechanisms involved a complex interaction of ANG-II type 2 and Mas receptors for subsequent downregulation of ACE expression and activity and ANG-II type 1 receptor expression, as well as upregulation of ACE2 expression and activity and the expression of ANG-II type 2 receptor and sarco(endo)plasmic reticulum Ca(2+)-ATPase. Thus RV fibrosis and dysfunction plays a central role in DCM, and ANG-(1-7) mitigates diabetes-induced RV alterations.
Collapse
Affiliation(s)
- Pan-Pan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Jian-Min Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Ming-Xiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Kai Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yu-Guo Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
41
|
Sevá Pessôa B, Becher PM, Van Veghel R, De Vries R, Tempel D, Sneep S, Van Beusekom H, Van Der Velden VHJ, Westermann D, Danser AHJ, Roks AJM. Effect of a stable Angiotensin-(1-7) analogue on progenitor cell recruitment and cardiovascular function post myocardial infarction. J Am Heart Assoc 2015; 4:jah3823. [PMID: 25655571 PMCID: PMC4345874 DOI: 10.1161/jaha.114.001510] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Angiotensin‐(1–7) improves cardiac function and remodeling after myocardial infarction (MI). This may involve recruitment of hematopoietic progenitor cells that support angiogenesis. However, angiotensin‐(1–7) is rapidly metabolized in plasma and tissue. The authors investigated in mice the effect of a metabolically stable angiotensin‐(1–7) analogue, cyclic angiotensin‐(1–7), on progenitor cell recruitment and on the heart post MI, when given in the angiogenesis phase of remodeling. Methods and Results Angiogenic progenitor cell recruitment was measured by using flow cytometry 24 and 72 hours after a daily bolus injection of cyclic angiotensin‐(1–7) in healthy C57BL/6 mice. Further, mice underwent MI or sham surgery and subsequently received saline or 2 different doses of cyclic angiotensin‐(1–7) for 3 or 9 weeks. Cyclic angiotensin‐(1–7) increased circulating hematopoietic progenitor cells at 24 hours but not 72 hours. Post MI, cyclic angiotensin‐(1–7) diminished cardiomyocyte hypertrophy and reduced myogenic tone, without altering cardiovascular function or cardiac histology at 9 weeks. Importantly, cyclic angiotensin‐(1–7)–treated mice had reduced cardiac capillary density at 3 weeks after MI but not after 9 weeks. Finally, cyclic angiotensin‐(1–7) decreased tube formation by cultured human umbilical vein endothelial cells. Conclusions Our results suggest that cyclic angiotensin‐(1–7), when given early after MI, recruits progenitor cells but does not lead to improved angiogenesis, most likely because it simultaneously exerts antiangiogenic effect in adult endothelial cells. Apparently, optimal treatment with cyclic angiotensin‐(1–7) depends on the time point of onset of application after MI.
Collapse
Affiliation(s)
- Bruno Sevá Pessôa
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| | - Peter Moritz Becher
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Germany (P.M.B., D.W.)
| | - Richard Van Veghel
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| | - René De Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| | - Dennie Tempel
- Division of Cardiology and Pulmonology, Department of Interventional Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands (D.T.)
| | - Stefan Sneep
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (S.S., H.V.B.)
| | - Heleen Van Beusekom
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (S.S., H.V.B.)
| | - Vincent H J Van Der Velden
- Department of Immunology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (V.J.V.D.V.)
| | - Dirk Westermann
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Germany (P.M.B., D.W.)
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| |
Collapse
|
42
|
Abstract
The renin-angiotensin system (RAS) plays a major role in the pathophysiology of cardiovascular disorders. Angiotensin II (Ang-II), the final product of this pathway, is known for its vasoconstrictive and proliferative effects. Angiotensin-converting enzyme 2 (ACE2), a newly discovered homolog of ACE, plays a key role as the central negative regulator of the RAS. It diverts the generation of vasoactive Ang-II into the vasodilatory and growth inhibiting peptide angiotensin(1-7) [Ang(1-7)], thereby providing counter-regulatory responses to neurohormonal activation. There is substantial experimental evidence evaluating the role of ACE2/Ang(1-7) in hypertension, heart failure, and atherosclerosis. In this review, we aim to focus on the conceptual facts of the ACE2-Ang(1-7) axis with regards to clinical implications and therapeutic targets in cardiovascular disorders, with emphasis on the potential therapeutic role in cardiovascular diseases.
Collapse
|
43
|
Schuetze KB, McKinsey TA, Long CS. Targeting cardiac fibroblasts to treat fibrosis of the heart: focus on HDACs. J Mol Cell Cardiol 2014; 70:100-7. [PMID: 24631770 PMCID: PMC4080911 DOI: 10.1016/j.yjmcc.2014.02.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/24/2014] [Accepted: 02/28/2014] [Indexed: 12/27/2022]
Abstract
Cardiac fibrosis is implicated in numerous physiologic and pathologic conditions, including scar formation, heart failure and cardiac arrhythmias. However the specific cells and signaling pathways mediating this process are poorly understood. Lysine acetylation of nucleosomal histone tails is an important mechanism for the regulation of gene expression. Additionally, proteomic studies have revealed that thousands of proteins in all cellular compartments are subject to reversible lysine acetylation, and thus it is becoming clear that this post-translational modification will rival phosphorylation in terms of biological import. Acetyl groups are conjugated to lysine by histone acetyltransferases (HATs) and removed from lysine by histone deacetylases (HDACs). Recent studies have shown that pharmacologic agents that alter lysine acetylation by targeting HDACs have the remarkable ability to block pathological fibrosis. Here, we review the current understanding of cardiac fibroblasts and the fibrogenic process with respect to the roles of lysine acetylation in the control of disease-related cardiac fibrosis. Potential for small molecule HDAC inhibitors as anti-fibrotic therapeutics that target cardiac fibroblasts is highlighted. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium."
Collapse
Affiliation(s)
- Katherine B Schuetze
- Department of Medicine, Division of Cardiology, University of Colorado Denver, 12700 E. 19th Ave., Aurora, CO 80045-0508, USA
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology, University of Colorado Denver, 12700 E. 19th Ave., Aurora, CO 80045-0508, USA.
| | - Carlin S Long
- Department of Medicine, Division of Cardiology, University of Colorado Denver, 12700 E. 19th Ave., Aurora, CO 80045-0508, USA.
| |
Collapse
|
44
|
Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, Zhang K, Zhang C. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol 2014; 11:413-26. [PMID: 24776703 PMCID: PMC7097196 DOI: 10.1038/nrcardio.2014.59] [Citation(s) in RCA: 300] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) 2 and its product angiotensin 1–7 are thought to have effects that counteract the adverse actions of other, better-known renin–angiotensin system (RAS) components Numerous experimental studies have suggested that ACE2 and angiotensin 1–7 have notable protective effects in the heart and blood vessels ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the functional importance and signalling mechanisms of angiotensin-1–7-induced actions remain unclear New pharmacological interventions targeting ACE2 are expected to be useful in clinical treatment of cardiovascular disease, especially those associated with overactivation of the conventional RAS More studies, especially randomized controlled clinical trials, are needed to clearly delineate the benefits of therapies targeting angiotensin 1–7 actions
Angiotensin-converting enzyme 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of the better-known members of the renin–angiotensin system and might, therefore, be useful therapeutic targets in patients with cardiovascular disease. Professor Jiang and colleagues review the evidence for the potential roles of these proteins in various cardiovascular conditions, including hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes. The renin–angiotensin system (RAS) has pivotal roles in the regulation of normal physiology and the pathogenesis of cardiovascular disease. Angiotensin-converting enzyme (ACE) 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of other, better known and understood, members of the RAS. The physiological and pathological importance of ACE2 and angiotensin 1–7 in the cardiovascular system are not completely understood, but numerous experimental studies have indicated that these components have protective effects in the heart and blood vessels. Here, we provide an overview on the basic properties of ACE2 and angiotensin 1–7 and a summary of the evidence from experimental and clinical studies of various pathological conditions, such as hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes mellitus. ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the relevant functions and signalling mechanisms of actions induced by angiotensin 1–7 have not been conclusively determined. The ACE2–angiotensin 1–7 pathway, however, might provide a useful therapeutic target for the treatment of cardiovascular disease, especially in patients with overactive RAS.
Collapse
Affiliation(s)
- Fan Jiang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Jianmin Yang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Yongtao Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Mei Dong
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Shuangxi Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Fang Fang Liu
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Kai Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| |
Collapse
|
45
|
Wang M, Zhang W, Zhou Y, Zhou X. Association between serum angiotensin-converting enzyme 2 levels and postoperative myocardial infarction following coronary artery bypass grafting. Exp Ther Med 2014; 7:1721-1727. [PMID: 24926374 PMCID: PMC4043627 DOI: 10.3892/etm.2014.1640] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/07/2014] [Indexed: 01/22/2023] Open
Abstract
Myocardial infarction (MI) is a predominant and severe complication in patients that undergo aortocoronary bypass surgery. Angiotensin-converting enzyme 2 (ACE2) activation is reportedly a protective mechanism in MI; therefore, in the present study, the association between serum ACE2 levels and postoperative MI following coronary artery bypass grafting (CABG) was investigated. Preoperative and postoperative serum ACE2 levels in 136 subjects undergoing CABG were observed and the serum ACE2 levels, 1 h post surgery, were divided into quartile categories. Following adjustment for age, gender, body mass index, hypertension, previous MI, current smoking status, hyperlipidemia, diabetes mellitus, Gensini score, aortic clamp time, number of grafts and pre-CABG medications; the risk of developing postoperative MI following CABG was observed to be significantly higher in the lowest serum ACE2 level quartile than when compared with the highest quartile (hazard ratio, 2.94; 95% confidence interval, 1.85-4.16; P=0.009). The subjects that exhibited a serum ACE2 level ≤1.06 ng/ml showed significantly higher rates of postoperative MI, arrhythmia and reduced cardiac output in addition to increased instances of in-hospital mortality post CABG, compared with those exhibiting a serum ACE2 level >1.06 ng/ml. A significant negative correlation was observed between serum ACE2 and serum cardiac troponin I levels, however, no significant association was identified between the serum ACE2 level quartiles and the ACE2 gene polymorphisms. The present study indicated that a low serum ACE2 level, 1 h post CABG was independently associated with an increased risk of postoperative MI. Thus, the serum ACE2 level may be a potential novel prognostic factor for postoperative MI following CABG.
Collapse
Affiliation(s)
- Min Wang
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Wei Zhang
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yu Zhou
- The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xinmin Zhou
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
46
|
Varagic J, Ahmad S, Nagata S, Ferrario CM. ACE2: angiotensin II/angiotensin-(1-7) balance in cardiac and renal injury. Curr Hypertens Rep 2014; 16:420. [PMID: 24510672 PMCID: PMC4286874 DOI: 10.1007/s11906-014-0420-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Our current recognition of the renin-angiotensin system is more convoluted than originally thought due to the discovery of multiple novel enzymes, peptides, and receptors inherent in this interactive biochemical cascade. Over the last decade, angiotensin-converting enzyme 2 (ACE2) has emerged as a key player in the pathophysiology of hypertension and cardiovascular and renal disease due to its pivotal role in metabolizing vasoconstrictive/hypertrophic/proliferative angiotensin II into favorable angiotensin-(1-7). This review addresses the considerable advancement in research on the role of tissue ACE2 in the development and progression of hypertension and cardiac and renal injury. We summarize the results from recent clinical and experimental studies suggesting that serum or urine soluble ACE2 may serve as a novel biomarker or independent risk factor relevant for diagnosis and prognosis of cardiorenal disease. We also review recent proceedings on novel therapeutic approaches to enhance ACE2/angiotensin-(1-7) axis.
Collapse
Affiliation(s)
- Jasmina Varagic
- Hypertension & Vascular Research Center, Division of Surgical Sciences, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA,
| | | | | | | |
Collapse
|
47
|
Manipulating angiotensin metabolism with angiotensin converting enzyme 2 (ACE2) in heart failure. ACTA ACUST UNITED AC 2014; 9:e141-e148. [PMID: 32362932 PMCID: PMC7185729 DOI: 10.1016/j.ddstr.2013.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Heart failure is increasing in prevalence associated with a huge economic burden. ACE2 is a negative regulator of the renin–angiotensin system. Elevated ACE2 activity is a biomarker in heart failure. Enhancing ACE2 action may have unique therapeutic effects in patients with heart failure.
Angiotensin converting enzyme 2 (ACE2), is a monocarboxypeptidase which metabolizes several peptides including the degradation of Ang II, a peptide with vasoconstrictive/proliferative/effects, to generate Ang 1–7, which acting through its receptor Mas exerts vasodilatory/anti-proliferative actions. The classical pathway of the RAS involving the ACE-Ang II-AT1 receptor axis is antagonized by the second arm constituted by the ACE2-Ang 1–7/Mas receptor axis. Loss of ACE2 enhances the adverse pathological remodeling susceptibility to pressure-overload and myocardial infarction. Human recombinant ACE2 is also a negative regulator of Ang II-induced myocardial hypertrophy, fibrosis and diastolic dysfunction and suppresses pressure-overload induced heart failure. Due to its characteristics, the ACE2-Ang 1–7/Mas axis may represent new possibilities for developing novel therapeutic strategies for the treatment of heart failure. Human recombinant ACE2 has been safely administered to healthy human volunteers intravenously resulting in sustained lowering of plasma Ang II levels. In this review, we will summarize the beneficial effects of ACE2 in heart disease and the potential use of human recombinant ACE2 as a novel therapy for heart failure.
Collapse
|
48
|
Liu W, Zhou X, Yu F, Hu J, Hu W. Arg972 Insulin receptor substrate-1 is associated with decreased serum angiotensin-converting enzyme 2 levels in acute myocardial infarction patients: in vivo and in vitro evidence. Cardiovasc Diabetol 2013; 12:151. [PMID: 24134599 PMCID: PMC4015180 DOI: 10.1186/1475-2840-12-151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/02/2013] [Indexed: 12/17/2022] Open
Abstract
Background Activation of the renin-angiotensin system (RAS) plays a critical role in the pathophysiology of myocardial infarction (MI) and the development of heart failure. Both angiotensin-converting enzyme 2 (ACE2) and insulin/insulin receptor substrate-1 (IRS-1) show cardioprotective effects after acute MI. The Arg972 IRS-1 polymorphism is associated with diminished activity of insulin. In the present study, we explored the association among Arg972 IRS-1, acute MI, and serum levels of ACE2. Methods A total of 711 subjects, including 351 subjects with first-time acute MI and 360 subjects without a history of MI were genotyped for Arg972 IRS-1 polymorphism. Serum levels of ACE2 and MI severity scores were determined. Primary human cardiomyocytes with overexpression of wild type IRS-1 or Arg972 IRS-1 or knockdown of endogenous IRS-1 were exposed to normoxia and hypoxia, and the expression levels of ACE2 were determined. Results The serum ACE2 level was significantly increased in acute MI patients compared with that of non-MI controls. Compared with wild type IRS-1 carriers, Arg972 IRS-1 carriers exhibited decreased serum ACE2 levels and increased MI severity scores after MI. Our in vitro data demonstrate that impairment of insulin/IRS-1/PI3K signaling by overexpression of Arg972-IRS-1, knockdown of endogenous IRS-1, or PI3K inhibitor can abolish hypoxia-induced IRS-1-associated PI3K activity and ACE2 expression in human cardiomyocytes, which suggests a causal relationship between Arg972-IRS-1 and decreased serum ACE2 levels in acute MI patients. Our in vitro data also indicate that insulin/IRS-1/PI3K signaling is required for ACE2 expression in cardiomyocytes, and that hypoxia can enhance the induction effect of insulin/IRS-1/PI3K signaling on ACE2 expression in cardiomyocytes. Conclusions This study provides the first evidence of crosstalk between insulin/IRS-1/PI3K signaling and RAS after acute MI, thereby adding fresh insights into the pathophysiology and treatment of acute MI.
Collapse
Affiliation(s)
| | | | | | | | - Wen Hu
- Department of Thoracic and Cardiovascular Surgery, Second Xiangya Hospital, Central South University, 138 Renmin Road, Changsha, Hunan 410011, P,R, China.
| |
Collapse
|
49
|
Abstract
SIGNIFICANCE The renin-angiotensin system (RAS) plays an important role in the normal control of cardiovascular and renal function in the healthy state and is a contributing factor in the development and progression of various types of cardiovascular diseases (CVD), including hypertension, diabetes, and heart failure. RECENT ADVANCES Evidence suggests that a balance between activation of the ACE/Ang II/AT1 receptor axis and the ACE2/Ang-(1-7)/Mas receptor axis is important for the function of the heart, kidney, and autonomic nervous system control of the circulation in the normal healthy state. An imbalance in these opposing pathways toward the ACE/Ang II/AT1 receptor axis is associated with CVD. The key component of this imbalance with respect to neural control of the circulation is the negative interaction between oxidative and NO• mechanisms, which leads to enhanced sympathetic tone and activation in disease conditions such as hypertension and heart failure. CRITICAL ISSUES The key mechanisms that disrupt normal regulation of Ang II and Ang-(1-7) signaling and promote pathogenesis of CVD at all organ levels remain poorly understood. The reciprocal relation between ACE and ACE2 expression and function suggests they are controlled interdependently at pre- and post-translational levels. Insights from neural studies suggest that an interaction between oxidative and nitrosative pathways may be key. FUTURE DIRECTIONS The role of redox mechanisms in the control of expression and activity of RAS enzymes and Ang receptors may provide important insight into the function of local tissue RAS in health and disease.
Collapse
Affiliation(s)
- Kaushik P Patel
- 1 Department of Cellular and Integrative Physiology, University of Nebraska Medical Center , Omaha, Nebraska
| | | |
Collapse
|
50
|
Qi Y, Zhang J, Cole-Jeffrey CT, Shenoy V, Espejo A, Hanna M, Song C, Pepine CJ, Katovich MJ, Raizada MK. Diminazene aceturate enhances angiotensin-converting enzyme 2 activity and attenuates ischemia-induced cardiac pathophysiology. Hypertension 2013; 62:746-52. [PMID: 23959549 DOI: 10.1161/hypertensionaha.113.01337] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Angiotensin-converting enzyme 2 (ACE2) plays a critical role against myocardial infarction (MI). We hypothesized that activation of intrinsic ACE2 would be protective against ischemia-induced cardiac pathophysiology. Diminazene aceturate (DIZE), a small molecule ACE2 activator, has been used to evaluate this hypothesis. DIZE (15 mg/kg per day, s.c.) was injected 2 days before MI surgery and continued throughout the study period. MI rats showed a 62% decrease in fractional shortening (%; control, 51.1±3.2; DIZE alone, 52.1±3.2; MI, 19.1±3.0), a 55% decrease in contractility (dP/dtmax mm Hg/s; control, 9480±425.3; DIZE alone, 9585±597.4; MI, 4251±657.7), and a 27% increase in ventricular hypertrophy (mg/mm; control, 26.5±1.5; DIZE alone, 26.9±1.4; MI, 33.4±1.1). DIZE attenuated the MI-induced decrease in fractional shortening by 89%, improved dP/dtmax by 92%, and reversed ventricular hypertrophy by 18%. MI also significantly increased ACE and angiotensin type 1 receptor levels but decreased ACE2 activity by 40% (control, 246.2±25.1; DIZE alone, 254.2±20.6; MI, 148.9±29.2; RFU/min), which was reversed by DIZE treatment. Thus, DIZE treatment decreased the infarct area, attenuated LV remodeling post-MI, and restored normal balance of the cardiac renin-angiotensin system. In addition, DIZE treatment increased circulating endothelial progenitor cells, increased engraftment of cardiac progenitor cells, and decreased inflammatory cells in peri-infarct cardiac regions. All of the beneficial effects associated with DIZE treatment were abolished by C-16, an ACE2 inhibitor. Collectively, DIZE and DIZE-like small molecules may represent promising new therapeutic agents for MI.
Collapse
Affiliation(s)
- Yanfei Qi
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610.
| | | | | | | | | | | | | | | | | | | |
Collapse
|