1
|
Xiao X, Li JX, Li HH, Teng F. ACE2 alleviates sepsis-induced cardiomyopathy through inhibiting M1 macrophage via NF-κB/STAT1 signals. Cell Biol Toxicol 2024; 40:82. [PMID: 39320524 PMCID: PMC11424656 DOI: 10.1007/s10565-024-09923-z] [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: 04/16/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Angiotensin-converting enzyme 2 (ACE2), a crucial element of the renin-angiotensin system (RAS), metabolizes angiotensin II into Ang (1-7), which then combines with the Mas receptor (MasR) to fulfill its protective role in various diseases. Nevertheless, the involvement of ACE2 in sepsis-induced cardiomyopathy (SIC) is still unexplored. In this study, our results revealed that CLP surgery dramatically impaired cardiac function accompanied with disruption of the balance between ACE2-Ang (1-7) and ACE-Ang II axis in septic heart tissues. Moreover, ACE2 knockin markedly alleviated sepsis induced RAS disorder, cardiac dysfunction and improved survival rate in mice, while ACE2 knockout significantly exacerbates these outcomes. Adoptive transfer of bone marrow cells and in vitro experiments showed the positive role of myeloid ACE2 by mitigating oxidative stress, inflammatory response, macrophage polarization and cardiomyocyte apoptosis by blocking NF-κB and STAT1 signals. However, the beneficial impacts were nullified by MasR antagonist A779. Collectively, these findings showed that ACE2 alleviated SIC by inhibiting M1 macrophage via activating the Ang (1-7)-MasR axis, highlight that ACE2 might be a promising target for the management of sepsis and SIC patients.
Collapse
Affiliation(s)
- Xue Xiao
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Roud, Beijing, 100020, China
| | - Jia-Xin Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Roud, Beijing, 100020, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Roud, Beijing, 100020, China.
| | - Fei Teng
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Roud, Beijing, 100020, China.
| |
Collapse
|
2
|
Zhang Z, Tang L, Guo Y, Guo X, Pan Z, Ji X, Gao C. Development of Biomarkers and Prognosis Model of Mortality Risk in Patients with COVID-19. J Inflamm Res 2024; 17:2445-2457. [PMID: 38681069 PMCID: PMC11048291 DOI: 10.2147/jir.s449497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/04/2024] [Indexed: 05/01/2024] Open
Abstract
Background As of 30 April 2023, the COVID-19 pandemic has resulted in over 6.9 million deaths worldwide. The virus continues to spread and mutate, leading to continuously evolving pathological and physiological processes. It is imperative to reevaluate predictive factors for identifying the risk of early disease progression. Methods A retrospective study was conducted on a cohort of 1379 COVID-19 patients who were discharged from Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine between 15 December 2022 and 15 February 2023. Patient symptoms, comorbidities, demographics, vital signs, and laboratory test results were systematically documented. The dataset was split into testing and training sets, and 15 different machine learning algorithms were employed to construct prediction models. These models were assessed for accuracy and area under the receiver operating characteristic curve (AUROC), and the best-performing model was selected for further analysis. Results AUROC for models generated by 15 machine learning algorithms all exceeded 90%, and the accuracy of 10 of them also surpassed 90%. Light Gradient Boosting model emerged as the optimal choice, with accuracy of 0.928 ± 0.0006 and an AUROC of 0.976 ± 0.0028. Notably, the factors with the greatest impact on in-hospital mortality were growth stimulation expressed gene 2 (ST2,19.3%), interleukin-8 (IL-8,17.2%), interleukin-6 (IL-6,6.4%), age (6.1%), NT-proBNP (5.1%), interleukin-2 receptor (IL-2R, 5%), troponin I (TNI,4.6%), congestive heart failure (3.3%) in Light Gradient Boosting model. Conclusion ST-2, IL-8, IL-6, NT-proBNP, IL-2R, TNI, age and congestive heart failure were significant predictors of in-hospital mortality among COVID-19 patients.
Collapse
Affiliation(s)
- Zhishuo Zhang
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Lujia Tang
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yiran Guo
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xin Guo
- School of Information Science and Technology, Sanda University, Shanghai, Pudong District, 201209, China
| | - Zhiying Pan
- School of Information Science and Technology, Sanda University, Shanghai, Pudong District, 201209, China
| | - Xiaojing Ji
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Chengjin Gao
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
3
|
Siratavičiūtė V, Pangonytė D, Utkienė L, Jusienė L, Marcinkevičienė J, Stanionienė Z, Radikė R. Myocardial Angiotensin-Converting Enzyme 2 Protein Expression in Ischemic Heart Failure. Int J Mol Sci 2023; 24:17145. [PMID: 38138974 PMCID: PMC10743033 DOI: 10.3390/ijms242417145] [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: 11/06/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
The angiotensin-converting enzyme 2 (ACE2)-angiotensin-(1-7)-Mas receptor axis plays a significant role in regulating myocardial remodeling and the development of heart failure (HF), with ACE2 being the primary focus. However, contemporary understanding of the membrane-bound form of the human ACE2 protein remains insufficient. The purpose of this study was to determine the expression of ACE2 protein in different cells of the left ventricular myocardium in non-diseased hearts and at various stages of ischemic HF. A total of 103 myocardial tissue samples from the left ventricle underwent quantitative and semi-quantitative immunohistochemical analysis. Upon assessing ACE2 immunostaining in all myocardial cells through unselective digital image analysis, there was no change in the stage A HF group. Nevertheless, the expression of ACE2 membrane protein in cardiomyocytes showed a tendency to increase, while non-cardiomyocyte ACE2 expression decreased significantly (p < 0.001). In the stage B HF group, the intensity of ACE2 immunostaining continued to increase with rising cardiomyocyte ACE2 expression (p < 0.001). Non-cardiomyocyte expression, in contrast, remained similar to that observed in the stage A HF group. In the stages C/D HF group, ACE2 expression reached its highest level in cardiomyocytes (p < 0.001), while ACE2 expression in non-cardiomyocytes was the lowest (p < 0.001). These changes in ACE2 protein levels are associated with left ventricular remodeling in ischemic HF.
Collapse
Affiliation(s)
| | - Dalia Pangonytė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.S.); (L.U.); (L.J.); (J.M.); (Z.S.); (R.R.)
| | | | | | | | | | | |
Collapse
|
4
|
Roth L, Dogan S, Tuna BG, Aranyi T, Benitez S, Borrell-Pages M, Bozaykut P, De Meyer GRY, Duca L, Durmus N, Fonseca D, Fraenkel E, Gillery P, Giudici A, Jaisson S, Johansson M, Julve J, Lucas-Herald AK, Martinet W, Maurice P, McDonnell BJ, Ozbek EN, Pucci G, Pugh CJA, Rochfort KD, Roks AJM, Rotllan N, Shadiow J, Sohrabi Y, Spronck B, Szeri F, Terentes-Printzios D, Tunc Aydin E, Tura-Ceide O, Ucar E, Yetik-Anacak G. Pharmacological modulation of vascular ageing: A review from VascAgeNet. Ageing Res Rev 2023; 92:102122. [PMID: 37956927 DOI: 10.1016/j.arr.2023.102122] [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: 07/05/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
Vascular ageing, characterized by structural and functional changes in blood vessels of which arterial stiffness and endothelial dysfunction are key components, is associated with increased risk of cardiovascular and other age-related diseases. As the global population continues to age, understanding the underlying mechanisms and developing effective therapeutic interventions to mitigate vascular ageing becomes crucial for improving cardiovascular health outcomes. Therefore, this review provides an overview of the current knowledge on pharmacological modulation of vascular ageing, highlighting key strategies and promising therapeutic targets. Several molecular pathways have been identified as central players in vascular ageing, including oxidative stress and inflammation, the renin-angiotensin-aldosterone system, cellular senescence, macroautophagy, extracellular matrix remodelling, calcification, and gasotransmitter-related signalling. Pharmacological and dietary interventions targeting these pathways have shown potential in ameliorating age-related vascular changes. Nevertheless, the development and application of drugs targeting vascular ageing is complicated by various inherent challenges and limitations, such as certain preclinical methodological considerations, interactions with exercise training and sex/gender-related differences, which should be taken into account. Overall, pharmacological modulation of endothelial dysfunction and arterial stiffness as hallmarks of vascular ageing, holds great promise for improving cardiovascular health in the ageing population. Nonetheless, further research is needed to fully elucidate the underlying mechanisms and optimize the efficacy and safety of these interventions for clinical translation.
Collapse
Affiliation(s)
- Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Tamas Aranyi
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Sonia Benitez
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular Biochemistry, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Perinur Bozaykut
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkiye
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Nergiz Durmus
- Department of Pharmacology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkiye
| | - Diogo Fonseca
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital, Pavol Jozef Šafárik University of Košice, Košice, Slovakia
| | - Philippe Gillery
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Centre Hospitalier et Universitaire de Reims, Reims, France
| | - Alessandro Giudici
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, the Netherlands
| | - Stéphane Jaisson
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Centre Hospitalier et Universitaire de Reims, Reims, France
| | | | - Josep Julve
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Endocrinology, Diabetes and Nutrition group, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | | | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Barry J McDonnell
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Emine Nur Ozbek
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkiye
| | - Giacomo Pucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Christopher J A Pugh
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Keith D Rochfort
- School of Nursing, Psychotherapy, and Community Health, Dublin City University, Dublin, Ireland
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Disease and Pharmacology, Erasmus Medical Center, Erasmus University, Rotterdam, the Netherlands
| | - Noemi Rotllan
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Pathophysiology of lipid-related diseases, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yahya Sohrabi
- Molecular Cardiology, Dept. of Cardiology I - Coronary and Peripheral Vascular Disease, University Hospital Münster, Westfälische Wilhelms-Universität, 48149 Münster, Germany; Department of Medical Genetics, Third Faculty of Medicine, Charles University, 100 00 Prague, Czechia
| | - Bart Spronck
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia
| | - Flora Szeri
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Elif Tunc Aydin
- Department of Cardiology, Hospital of Ataturk Training and Research Hospital, Katip Celebi University, Izmir, Turkiye
| | - Olga Tura-Ceide
- Biomedical Research Institute-IDIBGI, Girona, Spain; Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Madrid, Spain
| | - Eda Ucar
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Gunay Yetik-Anacak
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkiye; Department of Pharmacology, Faculty of Pharmacy, Acıbadem Mehmet Aydinlar University, Istanbul, Turkiye.
| |
Collapse
|
5
|
Kacsándi D, Fagyas M, Horváth Á, Végh E, Pusztai A, Czókolyová M, Soós B, Szabó AÁ, Hamar A, Pethő Z, Bodnár N, Kerekes G, Hodosi K, Szamosi S, Szűcs G, Papp Z, Szekanecz Z. Effect of tofacitinib therapy on angiotensin converting enzyme activity in rheumatoid arthritis. Front Med (Lausanne) 2023; 10:1226760. [PMID: 37877017 PMCID: PMC10591318 DOI: 10.3389/fmed.2023.1226760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
Introduction The Renin-Angiotensin-Aldosterone system (RAAS) has been implicated in the regulation of the cardiovascular system and linked to rheumatoid arthritis (RA). Little information has become available on the effects of Janus kinase (JAK) inhibition on RAAS. Here we studied the effects of 12-month tofacitinib treatment on angiotensin converting enzyme (ACE), ACE2 production and ACE/ACE2 ratios in RA along with numerous other biomarkers. Patients and methods Thirty RA patients were treated with tofacitinib in this prospective study. Serum ACE concentrations were assessed by ELISA. ACE2 activity was determined by a specific quenched fluorescent substrate. ACE/ACE2 ratios were calculated. We also determined common carotid intima-media thickness (ccIMT), brachial artery flow-mediated vasodilation (FMD) and carotid-femoral pulse-wave velocity (cfPWV) by ultrasound. C-reactive protein (CRP), rheumatoid factor (RF) and anti-citrullinated protein autoantibodies (ACPA) were also determined. All measurements were performed at baseline, as well as after 6 and 12 months of tofacitinib treatment. Results After the dropout of 4 patients, 26 completed the study. Tofacitinib treatment increased ACE levels after 6 and 12 months, while ACE2 activity only transiently increased at 6 months. The ACE/ACE2 ratio increased after 1 year of therapy (p < 0.05). Logistic regression analyses identified correlations between ACE, ACE2 or ACE/ACE2 ratios and RF at various time points. Baseline disease duration also correlated with erythrocyte sedimentation rate (ESR) (p < 0.05). One-year changes of ACE or ACE2 were determined by tofacitinib treatment plus ACPA or RF, respectively (p < 0.05). Conclusion JAK inhibition increases serum ACE and ACE/ACE2 ratio in RA. Baseline inflammation (ESR), disease duration and ACPA, as well as RF levels at various time points can be coupled to the regulation of ACE/ACE2 ratio. The effect of tofacitinib on RAAS provides a plausible explanation for the cardiovascular effects of JAK inhibition in RA.
Collapse
Affiliation(s)
- Dorottya Kacsándi
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Fagyas
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Horváth
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Edit Végh
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Pusztai
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Monika Czókolyová
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Boglárka Soós
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Ádám Szabó
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Kálmán Laki Doctoral School of Biomedical and Clinical Sciences, University of Debrecen, Debrecen, Hungary
| | - Attila Hamar
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pethő
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nóra Bodnár
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Kerekes
- Intensive Care Unit, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katalin Hodosi
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Szamosi
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Szűcs
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szekanecz
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
6
|
Karadağ AE, Biltekin SN, Demirci B, Demirci F, Ghani U. Comparative In Vitro and In Silico Enzyme Inhibitory Screening of Rosa x damascena and Pelargonium graveolens Essential Oils and Geraniol. PLANTS (BASEL, SWITZERLAND) 2023; 12:3296. [PMID: 37765459 PMCID: PMC10537268 DOI: 10.3390/plants12183296] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
The present work aims to evaluate Rosa x damascena Herrm. and Pelargonium graveolens L'Hér. essential oils, and the major constituent geraniol for their in vitro and in silico inhibitory activities against 5-lipoxygenase (5-LOX), cyclooxygenase (COX), acetyl cholinesterase (AChE), butyryl cholinesterase (BuChE), and angiotensin converting enzyme (ACE2) enzymes. Geraniol most potently inhibited the ACE2 relative to other enzymes. R. damascena essential oil moderately inhibited the cancer cell lines with no toxic effects on healthy HEK 293 cells. P. graveolens essential oil inhibited a number of cancer cell lines including A549, MCF7, PC3, and HEK 293 that are reported here for the first time. The molecular docking of geraniol with the target enzymes revealed that it binds to the active sites similar to that of known drugs. Geraniol carries the potential for further drug development due to its drug-like binding mode for the target enzymes. Our work confirms that these essential oils possess similar biological activities due to their similar phytochemistry in terms of the major constituents of the plants. The promising biological activities reported in this work further warrant the inclusion of in vivo studies to establish safe use of the target essential oils and their constituents.
Collapse
Affiliation(s)
- Ayşe Esra Karadağ
- Department of Pharmacognosy, School of Pharmacy, Istanbul Medipol University, 34815 Istanbul, Türkiye
| | - Sevde Nur Biltekin
- Department of Pharmaceutical Microbiology, School of Pharmacy, Istanbul Medipol University, 34815 Istanbul, Türkiye;
| | - Betül Demirci
- Department of Pharmacognosy, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Türkiye; (B.D.); (F.D.)
| | - Fatih Demirci
- Department of Pharmacognosy, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Türkiye; (B.D.); (F.D.)
- Faculty of Pharmacy, Eastern Mediterranean University, North Cyprus, 99450 Famagusta, Türkiye
| | - Usman Ghani
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh 12372, Saudi Arabia
| |
Collapse
|
7
|
Bosch A, Poglitsch M, Kannenkeril D, Kolwelter J, Striepe K, Ott C, Rauh M, Schiffer M, Achenbach S, Schmieder RE. Angiotensin pathways under therapy with empagliflozin in patients with chronic heart failure. ESC Heart Fail 2023; 10:1635-1642. [PMID: 36782339 DOI: 10.1002/ehf2.14313] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 09/05/2022] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
AIMS Large outcome studies demonstrated a reduction of heart failure hospitalization or cardiovascular death in patients with chronic heart failure (CHF). The renin-angiotensin system (RAS) is a key player in fluid and sodium regulation. The classic angiotensin-converting enzyme-angiotensin II-angiotensin-1 receptor axis (Ang I-ACE-Ang II receptor axis) is predominantly angiotensin II (Ang-II) induced and promotes vasoconstriction. In contrast, the angiotensin-converting-enzyme-2-angiotensin-(1-7)-Mas axis (Mas-axis) is mediated by the metabolites angiotensin-1-7 (Ang-(1-7)) and angtiotensin-1-5 (Ang-(1-5)) and exerts cardioprotective effects. METHODS We previously investigated the effect of empagliflozin on the systemic haemodynamic in patients with stable CHF (NYHA II-III) in a randomized placebo-controlled clinical trial 'Analysing the Effect of Empagliflozin on Reduction of Tissue Sodium Content in Patients With Chronic Heart Failure (ELSI)'. In a post hoc analysis, we now analysed whether empagliflozin has an effect on the RAS by measuring detailed RAS profiles (LC-MS/MS-based approach) in 72 patients from ELSI. We compared RAS parameters after 1-month and 3-months treatment with empagliflozin or placebo to baseline. The secondary goal was to analyse whether the effect of empagliflozin on RAS parameters was dependent on angiotensin-receptor-blocking (ARB) or angiotensin-converting-enzyme-inhibitor (ACEI) co-medication. RESULTS Empagliflozin medication induced a significant rise in Ang-II [68.5 pmol/L (21.3-324.2) vs. 131.5 pmol/L (34.9-564.0), P = 0.001], angiotensin-I (Ang-I) [78.7 pmol/L (21.5-236.6) vs. 125.9 pmol/L (52.6-512.9), P < 0.001], Ang-(1-7) [3.0 pmol/L (3.0-15.0) vs. 10.1 pmol/L (3.0-31.3), P = 0.006], and Ang-(1-5) [5.4 pmol/L (2.0-22.9) vs. 9.9 pmol/L (2.8-36.4), P = 0.004], which was not observed in the placebo group (baseline to 3-months treatment). A significant rise in Ang-II (206.4 pmol/L (64.2-750.6) vs. 568.2 pmol/L (164.7-1616.4), P = 0.001), Ang-(1-7) (3.0 pmol/L (3.0-14.1) vs. 15.0 pmol/L (3.0-31.3), P = 0.017), and Ang-(1-5) [12.2 pmol/L (3.8-46.6) vs. 36.4 pmol/L (11.1-90.7), P = 0.001] under empagliflozin treatment was only seen in the subgroup of patients with ARB co-medication, whereas no change of Ang-II (16.7 pmol/L (2.0-60.8) vs. 26.4 pmol/L (10.7-63.4), P = 0.469), Ang-(1-7) (6.6 pmol/L (3.0-20.7) vs. 10.5 pmol/L (3.0-50.5), P = 0.221), and Ang-(1-5) (2.7 pmol/L (2.0-8.4) vs. 2.8 pmol/L (2.0-6.9), P = 0.851) was observed in patients with empagliflozin that were on ACEI co-medication (baseline to 3-months treatment). CONCLUSIONS Our data indicate that empagliflozin might lead to an activation of both the Ang I-ACE-Ang II receptor axis and the Mas-axis pathway. Activation of the Ang I-ACE-Ang II receptor axis and the protective Mas-axis pathway after initiating treatment with empagliflozin was only seen in patients with ARB co-medication, in contrast to co-medication with ACEI.
Collapse
Affiliation(s)
- Agnes Bosch
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Dennis Kannenkeril
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Julie Kolwelter
- Department of Cardiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kristina Striepe
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christian Ott
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Nephrology and Hypertension, Paracelsus Medical University, Nürnberg, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stephan Achenbach
- Department of Cardiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Roland E Schmieder
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
8
|
Biltekin S, Karadaǧ AE, Demirci B, Demirci F. ACE2 and LOX Enzyme Inhibitions of Different Lavender Essential Oils and Major Components Linalool and Camphor. ACS OMEGA 2022; 7:36561-36566. [PMID: 36278093 PMCID: PMC9583641 DOI: 10.1021/acsomega.2c04518] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/21/2022] [Indexed: 05/30/2023]
Abstract
In this present study, Lavandula angustifolia, Lavandula stoechas, and Lavandula × heterophylla essential oils and their main compounds linalool and camphor were evaluated in vitro for lipoxygenase enzyme (LOX) and for angiotensin converting enzyme 2 (ACE2) inhibition potential. The chemical compositions of L. angustifolia, L. stoechas, and L. heterophylla essential oils were confirmed both by gas chromatography-mass spectrometry and gas chromatography-flame ionization detection, where 22.4, 0.9, and 30.6% linalool and 17.8, 54.7, and 15% camphor were identified for each oil among other components, respectively. Enzyme inhibitory activity studies were performed at 20 μg/mL for the tested essential oils, whereas for linalool and camphor concentrations, 5 μg/mL was used. The ACE2 inhibitions of L. angustifolia, L. stoechas, and L. heterophylla essential oils were 25.4, 34.1, and 27.1%, while the LOX inhibitions were observed as 79, 49.1, and 86.7%, respectively. In addition, linalool and camphor showed remarkable ACE2 inhibition with 77.1 and 85.1%, whereas the LOX inhibition was observed at 92 and 67.2%, respectively. In conclusion of the initial findings, further detailed in vivo studies are needed to confirm the safe use.
Collapse
Affiliation(s)
- Sevde
Nur Biltekin
- Department
of Pharmaceutical Microbiology, School of Pharmacy, Istanbul Medipol University, Beykoz, Istanbul 34810, Turkey
- Institute
of Sciences, Istanbul University, Istanbul 34116, Turkey
| | - Ayşe Esra Karadaǧ
- Department
of Pharmacognosy, School of Pharmacy, Istanbul
Medipol University, Beykoz, Istanbul 34810, Turkey
- Graduate
School of Health Sciences, Anadolu University, Eskişehir 26470, Turkey
| | - Betül Demirci
- Department
of Pharmacognosy, Faculty of Pharmacy, Anadolu
University, Eskişehir 26470, Turkey
| | - Fatih Demirci
- Department
of Pharmacognosy, Faculty of Pharmacy, Anadolu
University, Eskişehir 26470, Turkey
- Faculty of
Pharmacy, Eastern Mediterranean University, N. Cyprus, Mersin 10, Famagusta 99450, Turkey
| |
Collapse
|
9
|
Hedges JF, Snyder DT, Robison A, Grifka-Walk HM, Blackwell K, Shepardson K, Kominsky D, Rynda-Apple A, Walcheck B, Jutila MA. An ADAM17-Neutralizing Antibody Reduces Inflammation and Mortality While Increasing Viral Burden in a COVID-19 Mouse Model. Front Immunol 2022; 13:918881. [PMID: 35757773 PMCID: PMC9226444 DOI: 10.3389/fimmu.2022.918881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 12/12/2022] Open
Abstract
Angiotensin Converting Enzyme 2 (ACE2) is the primary cell entry receptor for SARS-CoV and SARS-CoV-2 viruses. A disintegrin and metalloproteinase 17 (ADAM17) is a protease that cleaves ectodomains of transmembrane proteins, including that of ACE2 and the proinflammatory cytokine TNF-α, from cell surfaces upon cellular activation. We hypothesized that blockade of ADAM17 activity would alter COVID-19 pathogenesis. To assess this pathway, we blocked the function of ADAM17 using the monoclonal antibody MEDI3622 in the K18-hACE2 transgenic mouse model of COVID-19. Antibody-treated mice were healthier, less moribund, and had significantly lower lung pathology than saline-treated mice. However, the viral burden in the lungs of MEDI3622-treated mice was significantly increased. Thus, ADAM17 appears to have a critical anti-viral role, but also may promote inflammatory damage. Since the inflammatory cascade is ultimately the reason for adverse outcomes in COVID-19 patients, there may be a therapeutic application for the MEDI3622 antibody.
Collapse
Affiliation(s)
- Jodi F Hedges
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Deann T Snyder
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Amanda Robison
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Heather M Grifka-Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Karlin Blackwell
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Kelly Shepardson
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Douglas Kominsky
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Agnieszka Rynda-Apple
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Mark A Jutila
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
10
|
Sarkar S, Sen R. Insights into Cardiovascular Defects and Cardiac Epigenome in the Context of COVID-19. EPIGENOMES 2022; 6:epigenomes6020013. [PMID: 35645252 PMCID: PMC9150012 DOI: 10.3390/epigenomes6020013] [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: 02/08/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Although few in number, studies on epigenome of the heart of COVID-19 patients show that epigenetic signatures such as DNA methylation are significantly altered, leading to changes in expression of several genes. It contributes to pathogenic cardiac phenotypes of COVID-19, e.g., low heart rate, myocardial edema, and myofibrillar disarray. DNA methylation studies reveal changes which likely contribute to cardiac disease through unknown mechanisms. The incidence of severe COVID-19 disease, including hospitalization, requiring respiratory support, morbidity, and mortality, is disproportionately higher in individuals with co-morbidities. This poses unprecedented strains on the global healthcare system. While their underlying conditions make patients more susceptible to severe COVID-19 disease, strained healthcare systems, lack of adequate support, or sedentary lifestyles from ongoing lockdowns have proved detrimental to their underlying health conditions, thus pushing them to severe risk of congenital heart disease (CHD) itself. Prophylactic vaccines against COVID-19 have ushered new hope for CHD. A common connection between COVID-19 and CHD is SARS-CoV-2’s host receptor ACE2, because ACE2 regulates and protects organs, including the heart, in various ways. ACE2 is a common therapeutic target against cardiovascular disease and COVID-19 which damages organs. Hence, this review explores the above regarding CHDs, cardiovascular damage, and cardiac epigenetics, in COVID-19 patients.
Collapse
Affiliation(s)
- Shreya Sarkar
- New Brunswick Heart Centre, Saint John Regional Hospital, Saint John, NB E2L 4L2, Canada;
| | - Rwik Sen
- Active Motif, Inc., 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
- Correspondence:
| |
Collapse
|
11
|
Boby N, Cao X, Williams K, Gadila SKG, Shroyer MN, Didier PJ, Srivastav SK, Das A, Baker K, Sha Q, Pahar B. Simian Immunodeficiency Virus Infection Mediated Changes in Jejunum and Peripheral SARS-CoV-2 Receptor ACE2 and Associated Proteins or Genes in Rhesus Macaques. Front Immunol 2022; 13:835686. [PMID: 35281029 PMCID: PMC8914048 DOI: 10.3389/fimmu.2022.835686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Angiotensin converting enzyme-2 (ACE2) and associated proteins play a pivotal role in various physiological and pathological events, such as immune activation, inflammation, gut barrier maintenance, intestinal stem cell proliferation, and apoptosis. Although many of these clinical events are quite significant in SIV/HIV infection, expression profiling of these proteins has not been well reported. Considering the different pathological consequences in the gut after HIV infection, we hypothesized that the expression of ACE2 and associated proteins of the Renin-angiotensin system (RAS) could be compromised after SIV/HIV infection. We quantified the gene expression of ACE2 as well as AGTR1/2, ADAM17, and TMPRSS2, and compared between SIV infected and uninfected rhesus macaques (Macaca mulatta; hereafter abbreviated RMs). The gene expression analysis revealed significant downregulation of ACE2 and upregulation of AGTR2 and inflammatory cytokine IL-6 in the gut of infected RMs. Protein expression profiling also revealed significant upregulation of AGTR2 after infection. The expression of ACE2 in protein level was also decreased, but not significantly, after infection. To understand the entirety of the process in newly regenerated epithelial cells, a global transcriptomic study of enteroids raised from intestinal stem cells was performed. Interestingly, most of the genes associated with the RAS, such as DPP4, MME, ANPEP, ACE2, ENPEP, were found to be downregulated in SIV infection. HNFA1 was found to be a key regulator of ACE2 and related protein expression. Jejunum CD4+ T cell depletion and increased IL-6 mRNA, MCP-1 and AGTR2 expression may signal inflammation, monocyte/macrophage accumulation and epithelial apoptosis in accelerating SIV pathogenesis. Overall, the findings in the study suggested a possible impact of SIV/HIV infection on expression of ACE2 and RAS-associated proteins resulting in the loss of gut homeostasis. In the context of the current COVID-19 pandemic, the outcome of SARS-CoV-2 and HIV co-infection remains uncertain and needs further investigation as the significance profile of ACE2, a viral entry receptor for SARS-CoV-2, and its expression in mRNA and protein varied in the current study. There is a concern of aggravated SARS-CoV-2 outcomes due to possible serious pathological events in the gut resulting from compromised expression of RAS- associated proteins in SIV/HIV infection.
Collapse
Affiliation(s)
- Nongthombam Boby
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, United States
| | - Kelsey Williams
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Shiva Kumar Goud Gadila
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, United States
| | - Monica N. Shroyer
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Peter J. Didier
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Sudesh K. Srivastav
- Department of Biostatistics, Tulane University, New Orleans, LA, United States
| | - Arpita Das
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Kate Baker
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, Houghton, MI, United States
| | - Bapi Pahar
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
- *Correspondence: Bapi Pahar,
| |
Collapse
|
12
|
Soós B, Fagyas M, Horváth Á, Végh E, Pusztai A, Czókolyová M, Csongrádi A, Hamar A, Pethő Z, Bodnár N, Kerekes G, Hodosi K, Szekanecz É, Szamosi S, Szántó S, Szűcs G, Papp Z, Szekanecz Z. Angiotensin Converting Enzyme Activity in Anti-TNF-Treated Rheumatoid Arthritis and Ankylosing Spondylitis Patients. Front Med (Lausanne) 2022; 8:785744. [PMID: 35155468 PMCID: PMC8828652 DOI: 10.3389/fmed.2021.785744] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022] Open
Abstract
Introduction Angiotensin-converting enzyme (ACE) and ACE2 have been implicated in the regulation of vascular physiology. Elevated synovial and decreased or normal ACE or ACE2 levels have been found in rheumatoid arthritis (RA). Very little is known about the effects of tumor necrosis factor α (TNF-α) inhibition on ACE or ACE2 homeostasis. In this study, we assessed the effects of one-year anti-TNF therapy on ACE and ACE2 production in RA and ankylosing spondylitis (AS) in association with other biomarkers. Patients and Methods Forty patients including 24 RA patients treated with either etanercept (ETN) or certolizumab pegol (CZP) and 16 AS patients treated with ETN were included in a 12-month follow-up study. Serum ACE levels were determined by commercial ELISA, while serum ACE2 activity was assessed using a specific quenched fluorescent substrate. Ultrasonography was performed to determine flow-mediated vasodilation (FMD), common carotid intima-media thickness (ccIMT) and arterial pulse-wave velocity (PWV) in all patients. In addition, CRP, rheumatoid factor (RF) and ACPA were also measured. All assessments were performed at baseline and 6 and 12 months after treatment initiation. Results Anti-TNF therapy increased ACE levels in the full cohort, as well as in the RA and AS subsets. ACE2 activity increased in the full cohort, while the ACE/ACE2 ratio increased in the full cohort and in the RA subset (p < 0.05). Uni- and multivariable regression analyses determined associations between ACE or ACE/ACE2 ratios at different time points and disease duration, CRP, RF, FMD and IMT (p < 0.05). ACE2 activity correlated with CRP. The changes of ACE or ACE2 over 12 months were determined by treatment together with either RF or FMD (p < 0.05). Conclusions Anti-TNF treatment may increase ACE and ACE2 in the sera of RA and AS patients. ACE and ACE2 may be associated with disease duration, markers of inflammation and vascular pathophysiology. The effects of TNF inhibition on ACE and ACE2 may reflect, in part, the effects of these biologics on the cardiovascular system.
Collapse
Affiliation(s)
- Boglárka Soós
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Fagyas
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Horváth
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Edit Végh
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Pusztai
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Monika Czókolyová
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Alexandra Csongrádi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Hamar
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pethő
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nóra Bodnár
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Kerekes
- Intensive Care Unit, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katalin Hodosi
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Szekanecz
- Department of Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Szamosi
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sándor Szántó
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Sports Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Szűcs
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szekanecz
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
13
|
Oliveira LCG, Cruz NAN, Ricelli B, Tedesco-Silva H, Medina-Pestana JO, Casarini DE. Interactions amongst inflammation, renin-angiotensin-aldosterone and kallikrein-kinin systems: suggestive approaches for COVID-19 therapy. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200181. [PMID: 34925477 PMCID: PMC8651214 DOI: 10.1590/1678-9199-jvatitd-2020-0181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a rapid-spread infectious disease caused by the SARS-CoV-2 virus, which can culminate in the renin-angiotensin-aldosterone (RAAS) and kallikrein-kinin (KKS) systems imbalance, and in serious consequences for infected patients. This scoping review of published research exploring the RAAS and KKS was undertaken in order to trace the history of the discovery of both systems and their multiple interactions, discuss some aspects of the viral-cell interaction, including inflammation and the system imbalance triggered by SARS-CoV-2 infection, and their consequent disorders. Furthermore, we correlate the effects of continued use of the RAAS blockers in chronic diseases therapies with the virulence and physiopathology of COVID-19. We also approach the RAAS and KKS-related proposed potential therapies for treatment of COVID-19. In this way, we reinforce the importance of exploring both systems and the application of their components or their blockers in the treatment of coronavirus disease.
Collapse
Affiliation(s)
| | | | - Bruna Ricelli
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Helio Tedesco-Silva
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - José Osmar Medina-Pestana
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Dulce Elena Casarini
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| |
Collapse
|
14
|
Protocol-based cardiotoxicity monitoring in hydroxychloroquine medicated COVID-19 pediatric patients. Rev Port Cardiol 2021; 41:155-163. [PMID: 34566257 PMCID: PMC8455282 DOI: 10.1016/j.repc.2021.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/05/2021] [Indexed: 11/22/2022] Open
Abstract
Introduction/objectives By May 2020, SARS-CoV-2 had caused more than 400 000 deaths worldwide. Initially, hydroxychloroquine was a treatment option for COVID-19. More recent studies have questioned its safety and efficacy and, until stronger evidence is available, it was suspended from therapy protocols. We describe our experience treating COVID-19 Portuguese pediatric patients with hydroxychloroquine, having applied a protocol for monitoring cardiac toxicity. Methods An observational retrospective study of COVID-19 pediatric patients, admitted from March to April 2020 and treated with hydroxychloroquine. Cardiotoxicity was assessed using ECG recordings and corrected QT-time (QTc). Patients were classified into risk-groups depending on QTc value: normal, slightly elevated or severely elevated (>500 ms). Results Total of 14 patients, with a median age of 10 years [four months; 17 years], treated with hydroxychloroquine for a median of five days. Hydroxychloroquine was used in monotherapy in six patients (mainly mild disease with comorbidities), and in association with lopinavir/ritonavir (3) and azithromycin (5) in moderate to severe disease. Other QT-prolonging therapies were used in five patients: oseltamivir (3), omeprazole (1), morphine (1) and ketamine (1). At 48 hours of treatment, two patients temporarily suspended hydroxychloroquine due to QTc prolongation (>500 ms). All patients completed the whole treatment. No other side effects or deaths occurred. Conclusion Clinical trials are evolving to define hydroxychloroquine effectivity and safety. Our considerable pediatric population supports the need for cardiotoxicity monitoring during therapy but suggest its use seems to be safe in COVID-19 pediatric patients, even in association with other QT-prolonging therapies.
Collapse
|
15
|
Razeghian-Jahromi I, Zibaeenezhad MJ, Lu Z, Zahra E, Mahboobeh R, Lionetti V. Angiotensin-converting enzyme 2: a double-edged sword in COVID-19 patients with an increased risk of heart failure. Heart Fail Rev 2021; 26:371-380. [PMID: 32844337 PMCID: PMC7447089 DOI: 10.1007/s10741-020-10016-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The coronavirus disease (COVID-19) pandemic is a global health priority. Given that cardiovascular diseases (CVD) are the leading cause of morbidity around the world and that several trials have reported severe cardiovascular damage in patients infected with SARS-CoV-2, a substantial number of COVID-19 patients with underlying cardiovascular diseases need to continue their medications in order to improve myocardial contractility and to prevent the onset of major adverse cardiovascular events (MACEs), including heart failure. Some of the current life-saving medications may actually simultaneously expose patients to a higher risk of severe COVID-19. Angiotensin-converting enzyme 2 (ACE2), a key counter regulator of the renin-angiotensin system (RAS), is the main entry gate of SARS-CoV-2 into human host cells and an established drug target to prevent heart failure. In fact, ACE inhibitors, angiotensin II receptor blockers, and mineralocorticoid antagonists may augment ACE2 levels to protect organs from angiotensin II overload. Elevated ACE2 expression on the host cell surface might facilitate viral entrance, at the same time sudden nonadherence to these medications triggers MACEs. Hence, safety issues in the use of RAS inhibitors in COVID-19 patients with cardiac dysfunction remain an unsolved dilemma and need paramount attention. Although ACE2 generally plays an adaptive role in both healthy subjects and patients with systolic and/or diastolic dysfunction, we conducted a literature appraisal on its maladaptive role. Understanding the exact role of ACE2 in COVID-19 patients at risk of heart failure is needed to safely manage RAS inhibitors in frail and non-frail critically ill patients.
Collapse
Affiliation(s)
| | | | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Elyaspour Zahra
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razmkhah Mahboobeh
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vicenzo Lionetti
- Unit of Translational Critical Care Medicine, Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
- UOS Anesthesiology and Intensive Care Medicine, Fondazione Toscana G. Monasterio, Pisa, Italy.
| |
Collapse
|
16
|
The Angiotensin II Type 1(AT1) Receptor and Cardiac Hypertrophy: Did We Have It Wrong All Along? J Cardiovasc Pharmacol 2021; 77:531-535. [PMID: 33657051 DOI: 10.1097/fjc.0000000000000999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/08/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT An ongoing issue in cardiac pharmacology is whether angiotensin II has direct growth promoting effects on the heart via the angiotensin II type 1 (AT1) receptor. This question has relevance for whether angiotensin-converting enzyme inhibitors and AT1 receptor blockers offer additional benefit in preventing adverse cardiac remodeling in hypertension. In a recent study, 2 strains of mice were infused with angiotensin II. In both, AT1 receptors were deleted in the heart and conduit vessels, but in one, AT1 receptors were also deleted in resistance vessels. Angiotensin II caused hypertrophy and hypertension in the strain lacking AT1 receptors in the heart and conduit vessels, but not in the strain without AT1 receptors in resistance vessels. This finding supports the conclusion that blood pressure is more important in determining cardiac hypertrophy than direct AT1 activation by angiotensin II, when the two are rapidly and simultaneously introduced. Surprisingly, mice with no cardiac AT1 receptor expression developed ventricular dilation and eccentric hypertrophy with pressure overload, in contrast to wild type mice that exhibited concentric hypertrophy, suggesting that cardiac AT1 receptors protect against high blood pressure. This interpretation revives issues related to β-arrestin-biased signaling and mechanosensitivity of AT1 receptors. Synthetic nanobodies, which are based on the variable regions of camelid-derived heavy chain-only antibodies, could be applied to explore the therapeutic potential of exploiting different activation states of AT1 under stress conditions, such as hypertension and heart failure. At the very least, this experimental approach is likely to reveal new facets of AT1 receptor signaling in the heart.
Collapse
|
17
|
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
|
18
|
Thankam FG, Agrawal DK. Molecular chronicles of cytokine burst in patients with coronavirus disease 2019 (COVID-19) with cardiovascular diseases. J Thorac Cardiovasc Surg 2021; 161:e217-e226. [PMID: 32631657 PMCID: PMC7834736 DOI: 10.1016/j.jtcvs.2020.05.083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/12/2020] [Accepted: 05/16/2020] [Indexed: 02/02/2023]
Affiliation(s)
| | - Devendra K. Agrawal
- Address for reprints: Devendra K. Agrawal, PhD (Biochem), PhD (Med Sci), MBA, Department of Translational Research, Western University of Health Sciences, 309 E Second St, Pomona, CA 91766
| |
Collapse
|
19
|
Sharma R, Li J, Krishnan S, Richards E, Raizada M, Mohandas R. Angiotensin-converting enzyme 2 and COVID-19 in cardiorenal diseases. Clin Sci (Lond) 2021; 135:1-17. [PMID: 33399851 PMCID: PMC7796300 DOI: 10.1042/cs20200482] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 01/08/2023]
Abstract
The rapid spread of the novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has brought into focus the key role of angiotensin-converting enzyme 2 (ACE2), which serves as a cell surface receptor required for the virus to enter cells. SARS-CoV-2 can decrease cell surface ACE2 directly by internalization of ACE2 bound to the virus and indirectly by increased ADAM17 (a disintegrin and metalloproteinase 17)-mediated shedding of ACE2. ACE2 is widely expressed in the heart, lungs, vasculature, kidney and the gastrointestinal (GI) tract, where it counteracts the deleterious effects of angiotensin II (AngII) by catalyzing the conversion of AngII into the vasodilator peptide angiotensin-(1-7) (Ang-(1-7)). The down-regulation of ACE2 by SARS-CoV-2 can be detrimental to the cardiovascular system and kidneys. Further, decreased ACE2 can cause gut dysbiosis, inflammation and potentially worsen the systemic inflammatory response and coagulopathy associated with SARS-CoV-2. This review aims to elucidate the crucial role of ACE2 both as a regulator of the renin-angiotensin system and a receptor for SARS-CoV-2 as well as the implications for Coronavirus disease 19 and its associated cardiovascular and renal complications.
Collapse
Affiliation(s)
- Ravindra K. Sharma
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Jing Li
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Suraj Krishnan
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Elaine M. Richards
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Mohan K. Raizada
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Rajesh Mohandas
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| |
Collapse
|
20
|
Amirfakhryan H, Safari F. Outbreak of SARS-CoV2: Pathogenesis of infection and cardiovascular involvement. Hellenic J Cardiol 2021; 62:13-23. [PMID: 32522617 PMCID: PMC7275139 DOI: 10.1016/j.hjc.2020.05.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Since the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has emerged from China, the infection (novel corona virus disease-2019, COVID-19) has affected many countries and led to many deaths worldwide. Like SARS-CoV, angiotencin converting enzyme (ACE)2 as a functional receptor for SARS-CoV2 is essential for the virus to make an entry into the cell. ACE2 is a part of Renin-Angiotensin-Aldosterone System, which is expressed in several organs that opposes the angiotensin (Ang) II functions by converting Ang II to Ang (1-7), the one with vasodilation effects. The death rate of COVID-19 is estimated to be approximately 3.4%; however, some comorbid conditions like underlying cardiovascular disease, hypertension, and diabetes increase the risk of mortality. In addition, cardiovascular involvement as a complication of SARS-CoV2 could be direct through either ACE2 receptors that are expressed tremendously in the heart, or by the surge of different cytokines or by acute respiratory distress syndrome-induced hypoxia. Traditional risk factors could aggravate the process of COVID-19 infection that urges the triage of these high-risk patients for SARS-CoV2. Currently, there is no effective, proven treatment or vaccination for COVID-19, but many investigators are struggling to find a treatment strategy as soon as possible. Some potential medications like chloroquine by itself or in combination with azithromycin and some protease inhibitors used for the treatment of COVID-19 have cardiovascular adverse effects, which should be kept in mind while the patients taking these medications are being closely monitored.
Collapse
Affiliation(s)
- Hamideh Amirfakhryan
- University of South Wales, Faculty of Health Science, Preventative Cardiovascular Medicine, UK.
| | - Fatemeh Safari
- University of Alberta, Edmonton, Faculty of Medicine, AB, Canada
| |
Collapse
|
21
|
Zhang H, Viveiros A, Nikhanj A, Nguyen Q, Wang K, Wang W, Freed DH, Mullen JC, MacArthur R, Kim DH, Tymchak W, Sergi CM, Kassiri Z, Wang S, Oudit GY. The Human Explanted Heart Program: A translational bridge for cardiovascular medicine. Biochim Biophys Acta Mol Basis Dis 2021; 1867:165995. [PMID: 33141063 PMCID: PMC7581399 DOI: 10.1016/j.bbadis.2020.165995] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022]
Abstract
The progression of cardiovascular research is often impeded by the lack of reliable disease models that fully recapitulate the pathogenesis in humans. These limitations apply to both in vitro models such as cell-based cultures and in vivo animal models which invariably are limited to simulate the complexity of cardiovascular disease in humans. Implementing human heart tissue in cardiovascular research complements our research strategy using preclinical models. We established the Human Explanted Heart Program (HELP) which integrates clinical, tissue and molecular phenotyping thereby providing a comprehensive evaluation into human heart disease. Our collection and storage of biospecimens allow them to retain key pathogenic findings while providing novel insights into human heart failure. The use of human non-failing control explanted hearts provides a valuable comparison group for the diseased explanted hearts. Using HELP we have been able to create a tissue repository which have been used for genetic, molecular, cellular, and histological studies. This review describes the process of collection and use of explanted human heart specimens encompassing a spectrum of pediatric and adult heart diseases, while highlighting the role of these invaluable specimens in translational research. Furthermore, we highlight the efficient procurement and bio-preservation approaches ensuring analytical quality of heart specimens acquired in the context of heart donation and transplantation.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Anissa Viveiros
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anish Nikhanj
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Quynh Nguyen
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kaiming Wang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Wei Wang
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Darren H Freed
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John C Mullen
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Roderick MacArthur
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel H Kim
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Wayne Tymchak
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Consolato M Sergi
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Anatomical Pathology, Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shaohua Wang
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
22
|
Colombier S, Mahendiran T, Niclauss L, Kirsch M. Cardiac arrest and COVID-19: inflammation, angiotensin-converting enzyme 2, and the destabilization of non-significant coronary artery disease-a case report. EUROPEAN HEART JOURNAL-CASE REPORTS 2020; 5:ytaa475. [PMID: 33542974 PMCID: PMC7799244 DOI: 10.1093/ehjcr/ytaa475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/09/2020] [Accepted: 11/12/2020] [Indexed: 01/08/2023]
Abstract
Background The new β-coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) appears to exhibit cardiovascular pathogenicity through use of angiotensin-converting enzyme 2 (ACE2) for cell entry and the development of a major systemic inflammation. Furthermore, cardiovascular comorbidities increase susceptibility to SARS-CoV-2 infection and the development of a severe form of COronaVIrus Disease 2019 (COVID-19). Case summary We describe the case of a COVID-19 patient whose inaugural presentation was a refractory cardiac arrest secondary to the destabilization of known, non-significant coronary artery disease. Patient was supported by venoarterial extracorporeal life support. After 12 h of support, cardiac function remained stable on low vasopressor support but the patient remained in a coma and brainstem death was diagnosed. Discussion Myocardial injury is frequently seen among critically unwell COVID-19 patients and increases the risk of mortality. This case illustrates several potential mechanisms that are thought to drive the cardiac complications seen in COVID-19. We present the potential role of inflammation and ACE2 in the pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Sébastien Colombier
- Department of Cardiovascular Surgery, Lausanne University Hospital CHUV, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Thabodhan Mahendiran
- Department of Cardiology, Lausanne University Hospital CHUV, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Lars Niclauss
- Department of Cardiovascular Surgery, Lausanne University Hospital CHUV, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | - Matthias Kirsch
- Department of Cardiovascular Surgery, Lausanne University Hospital CHUV, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| |
Collapse
|
23
|
Bártová E, Legartová S, Krejčí J, Arcidiacono OA. Cell differentiation and aging accompanied by depletion of the ACE2 protein. Aging (Albany NY) 2020; 12:22495-22508. [PMID: 33203793 PMCID: PMC7746349 DOI: 10.18632/aging.202221] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022]
Abstract
ACE2 was observed as the cell surface receptor of the SARS-CoV-2 virus. Interestingly, we also found ACE2 positivity inside the cell nucleus. The ACE2 levels changed during cell differentiation and aging and varied in distinct cell types. We observed ACE2 depletion in the aortas of aging female mice, similarly, the aging caused ACE2 decrease in the kidneys. Compared with that in the heart, brain and kidneys, the ACE2 level was the lowest in the mouse lungs. In mice exposed to nicotine, ACE2 was not changed in olfactory bulbs but in the lungs, ACE2 was upregulated in females and downregulated in males. These observations indicate the distinct gender-dependent properties of ACE2. Differentiation into enterocytes, and cardiomyocytes, caused ACE2 depletion. The cardiomyogenesis was accompanied by renin upregulation, delayed in HDAC1-depleted cells. In contrast, vitamin D2 decreased the renin level while ACE2 was upregulated. Together, the ACE2 level is high in non-differentiated cells. This protein is more abundant in the tissues of mouse embryos and young mice in comparison with older animals. Mostly, downregulation of ACE2 is accompanied by renin upregulation. Thus, the pathophysiology of COVID-19 disease should be further studied not only by considering the ACE2 level but also the whole renin-angiotensin system.
Collapse
Affiliation(s)
- Eva Bártová
- Institute of Biophysics, Academy of Sciences of the Czech Republic, 612 65, Brno, Czech Republic
| | - Soňa Legartová
- Institute of Biophysics, Academy of Sciences of the Czech Republic, 612 65, Brno, Czech Republic
| | - Jana Krejčí
- Institute of Biophysics, Academy of Sciences of the Czech Republic, 612 65, Brno, Czech Republic
| | | |
Collapse
|
24
|
Stein RA, Young LM. From ACE2 to COVID-19: A multiorgan endothelial disease. Int J Infect Dis 2020; 100:425-430. [PMID: 32896660 PMCID: PMC7832810 DOI: 10.1016/j.ijid.2020.08.083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Richard A Stein
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, USA; LaGuardia Community College, Department of Natural Sciences, City University of New York, New York, NY 11101, USA.
| | - Lauren M Young
- University of Chicago, Department of Internal Medicine, 5841 S Maryland Ave, Chicago, IL 60637, USA.
| |
Collapse
|
25
|
Yu W, Ou X, Liu X, Zhang S, Gao X, Cheng H, Zhu B, Yan J. ACE2 contributes to the maintenance of mouse epithelial barrier function. Biochem Biophys Res Commun 2020; 533:1276-1282. [PMID: 33097186 PMCID: PMC7576438 DOI: 10.1016/j.bbrc.2020.10.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 12/19/2022]
Abstract
The whole world was hit hard by the coronavirus disease-19 (COVID-19). Given that angiotensin I converting enzyme 2 (ACE2) is the viral entry molecule, understanding ACE2 has become a major focus of current COVID-19 research. ACE2 is highly expressed in the gut, but its role has not been fully understood and thus COVID-19 treatments intending to downregulate ACE2 level may cause untoward side effects. Gaining insight into the functions of ACE2 in gut homeostasis therefore merits closer examination, and is beneficial to find potential therapeutic alternatives for COVID-19. Methods: We took advantage of Ace2 knockout out mice and isolated intestinal organoids to examine the role of ACE2 in intestinal stemness. Inflammatory bowel disease (IBD) mouse model was established by 4% dextran sodium sulfate. LGR5 and KI67 levels were quantitated to reflect the virtue of intestinal stem cells (ISCs). FITC-dextran 4 (FD-4) assay was used to assess intestinal barrier function. Results: Western blotting identified the expression of ACE2 in colon, which was consistent with the results of immunofluorescence and RT-PCR. Moreover, Ace2−/− organoids showed decreased LRG5 and KI67 levels, and elevated calcium concentration. Furthermore, the permeability of ace2−/− organoids was markedly increased compared with ace2+/+ organoids. Collectively, ace2−/− mice were more susceptible than ace2+/+ mice to IBD, including earlier bloody stool, undermined intestinal architecture and more pronounced weight loss. Conclusions: Our data reveal that ACE2 contributes to the proliferation of intestinal stem cells and hence orchestrates the mucosal homeostasis.
Collapse
Affiliation(s)
- Wei Yu
- Department of Physiology, Jining Medical University, Jining, Shandong Province, 272067, PR China
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, PR China
| | - Xiaofan Liu
- Department of Physiology, Jining Medical University, Jining, Shandong Province, 272067, PR China
| | - Shuaixu Zhang
- Department of Physiology, Jining Medical University, Jining, Shandong Province, 272067, PR China
| | - Xinxin Gao
- Department of Physiology, Jining Medical University, Jining, Shandong Province, 272067, PR China
| | - Hongju Cheng
- Department of Physiology, Jining Medical University, Jining, Shandong Province, 272067, PR China
| | - Baoliang Zhu
- Department of Physiology, Jining Medical University, Jining, Shandong Province, 272067, PR China
| | - Jing Yan
- Department of Physiology, Jining Medical University, Jining, Shandong Province, 272067, PR China.
| |
Collapse
|
26
|
Aleksova A, Ferro F, Gagno G, Cappelletto C, Santon D, Rossi M, Ippolito G, Zumla A, Beltrami AP, Sinagra G. COVID-19 and renin-angiotensin system inhibition: role of angiotensin converting enzyme 2 (ACE2) - Is there any scientific evidence for controversy? J Intern Med 2020; 288:410-421. [PMID: 32459372 PMCID: PMC7283873 DOI: 10.1111/joim.13101] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022]
Abstract
Renin-angiotensin system (RAS) blockers are extensively used worldwide to treat many cardiovascular disorders, where they are effective in reducing both mortality and morbidity. These drugs are known to induce an increased expression of angiotensin-converting enzyme 2 (ACE2). ACE2 acts as receptor for the novel SARS coronavirus-2 (SARS-CoV-2) which raising the important issue of possible detrimental effects that RAS blockers could exert on the natural history and pathogenesis of the coronavirus disease-19 (COVID-19) and associated excessive inflammation, myocarditis and cardiac arrhythmias. We review the current knowledge on the interaction between SARS-CoV-2 infection and RAS blockers and suggest a scientific rationale for continuing RAS blockers therapy in patients with COVID-19 infection.
Collapse
Affiliation(s)
- A Aleksova
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - F Ferro
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - G Gagno
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - C Cappelletto
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - D Santon
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - M Rossi
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - G Ippolito
- National Institute for Infectious Diseases Lazzaro Spallanzani - IRCCS, Rome, Italy
| | - A Zumla
- Division of Infection and Immunity, University College London, London, UK.,National Institute of Health Research, Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | | | - G Sinagra
- From the, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Trieste, Italy
| |
Collapse
|
27
|
McGowan EM, Haddadi N, Nassif NT, Lin Y. Targeting the SphK-S1P-SIPR Pathway as a Potential Therapeutic Approach for COVID-19. Int J Mol Sci 2020; 21:ijms21197189. [PMID: 33003377 PMCID: PMC7583882 DOI: 10.3390/ijms21197189] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
The world is currently experiencing the worst health pandemic since the Spanish flu in 1918-the COVID-19 pandemic-caused by the coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This pandemic is the world's third wake-up call this century. In 2003 and 2012, the world experienced two major coronavirus outbreaks, SARS-CoV-1 and Middle East Respiratory syndrome coronavirus (MERS-CoV), causing major respiratory tract infections. At present, there is neither a vaccine nor a cure for COVID-19. The severe COVID-19 symptoms of hyperinflammation, catastrophic damage to the vascular endothelium, thrombotic complications, septic shock, brain damage, acute disseminated encephalomyelitis (ADEM), and acute neurological and psychiatric complications are unprecedented. Many COVID-19 deaths result from the aftermath of hyperinflammatory complications, also referred to as the "cytokine storm syndrome", endotheliitus and blood clotting, all with the potential to cause multiorgan dysfunction. The sphingolipid rheostat plays integral roles in viral replication, activation/modulation of the immune response, and importantly in maintaining vasculature integrity, with sphingosine 1 phosphate (S1P) and its cognate receptors (SIPRs: G-protein-coupled receptors) being key factors in vascular protection against endotheliitus. Hence, modulation of sphingosine kinase (SphK), S1P, and the S1P receptor pathway may provide significant beneficial effects towards counteracting the life-threatening, acute, and chronic complications associated with SARS-CoV-2 infection. This review provides a comprehensive overview of SARS-CoV-2 infection and disease, prospective vaccines, and current treatments. We then discuss the evidence supporting the targeting of SphK/S1P and S1P receptors in the repertoire of COVID-19 therapies to control viral replication and alleviate the known and emerging acute and chronic symptoms of COVID-19. Three clinical trials using FDA-approved sphingolipid-based drugs being repurposed and evaluated to help in alleviating COVID-19 symptoms are discussed.
Collapse
Affiliation(s)
- Eileen M McGowan
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China;
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
- Correspondence: ; Tel.: +61-405814048
| | - Nahal Haddadi
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| | - Yiguang Lin
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, Guangdong Pharmaceutical University, Guangzhou 510080, China;
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia; (N.H.); (N.T.N.)
| |
Collapse
|
28
|
Viana SD, Nunes S, Reis F. ACE2 imbalance as a key player for the poor outcomes in COVID-19 patients with age-related comorbidities - Role of gut microbiota dysbiosis. Ageing Res Rev 2020; 62:101123. [PMID: 32683039 PMCID: PMC7365123 DOI: 10.1016/j.arr.2020.101123] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 patients with pre-existing age-related comorbidities have poor outcomes. Gut microbiota dysbiosis is associated with ageing and age-related diseases. Viral-mediated ACE2 shedding favors poor outcomes by RAS-dependent mechanisms. Viral-mediated ACE2 shedding favors poor outcomes by RAS-independent gut dysbiosis. Potential of ACE2 and gut microbiota-based therapeutic opportunities for COVID-19.
Coronavirus disease 19 (COVID-19) is a pandemic condition caused by the new coronavirus SARS-CoV-2. The typical symptoms are fever, cough, shortness of breath, evolving to a clinical picture of pneumonia and, ultimately, death. Nausea and diarrhea are equally frequent, suggesting viral infection or transmission via the gastrointestinal-enteric system. SARS-CoV-2 infects human cells by using angiotensin converting enzyme 2 (ACE2) as a receptor, which is cleaved by transmembrane proteases during host cells infection, thus reducing its activities. ACE2 is a relevant player in the renin-angiotensin system (RAS), counterbalancing the deleterious effects of angiotensin II. Furthermore, intestinal ACE2 functions as a chaperone for the aminoacid transporter B0AT1. It has been suggested that B0AT1/ACE2 complex in the intestinal epithelium regulates gut microbiota (GM) composition and function, with important repercussions on local and systemic immune responses against pathogenic agents, namely virus. Notably, productive infection of SARS-CoV-2 in ACE2+ mature human enterocytes and patients’ GM dysbiosis was recently demonstrated. This review outlines the evidence linking abnormal ACE2 functions with the poor outcomes (higher disease severity and mortality rate) in COVID-19 patients with pre-existing age-related comorbidities and addresses a possible role for GM dysbiosis. The article culminates with the therapeutics opportunities based on these pathways.
Collapse
|
29
|
Pahan P, Pahan K. Smooth or Risky Revisit of an Old Malaria Drug for COVID-19? J Neuroimmune Pharmacol 2020; 15:174-180. [PMID: 32415419 PMCID: PMC7225253 DOI: 10.1007/s11481-020-09923-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Hydroxychloroquine (HCQ) is an old medication for malaria. In addition to handling this parasitic disease, HCQ is also used to treat a number of autoimmune disorders including rheumatoid arthritis and systemic lupus erythematosus when other medications are not effective. Recently a new viral infection (COVID-19) is rocking the entire world so much that it has already taken more than 200,000 lives throughout the world within the last two months and the World Health Organization was forced to declare it as a pandemic on March 11, 2020. Interestingly, some reports indicate that this wonder drug may be also beneficial for COVID-19 and accordingly, many clinical trials have begun. Here, we discuss different modes of action (anti-inflammatory, antioxidant, inhibition of endosomal acidification, suppression of angiotensin-converting enzyme 2 or ACE2 glycosylation, etc.) of HCQ that might be responsible for its possible anti-COVID-19 effect. On the other hand, this review also makes an honest attempt to delineate mechanisms (increase in vasoconstriction, inhibition of autophagy, depletion of T cells, etc.) indicating how it may aggravate certain conditions and why caution should be taken before granting widespread repurposing of HCQ for COVID-19. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Priyanka Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St Suite Cohn 310, 60612, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St Suite Cohn 310, 60612, Chicago, IL, USA. .,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA.
| |
Collapse
|
30
|
Greenwell AA, Chahade JJ, Ussher JR. Cardiovascular biology of the GIP receptor. Peptides 2020; 125:170228. [PMID: 31812593 DOI: 10.1016/j.peptides.2019.170228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is a gut hormone secreted primarily from enteroendocrine K cells in the duodenum and proximal jejunum following nutrient ingestion, primarily acting on islet β-cells to potentiate insulin secretion in a glucose-dependent manner. New discoveries of GIP receptor (GIPR) biology in adipose tissue, as well as findings that co-agonists for the glucagon-like peptide-1 receptor (GLP-1R) and GIPR induce greater weight loss than that seen with GLP-1R agonists alone, has led to continued interest in manipulating GIPR activity for the treatment of obesity/type 2 diabetes mellitus (T2DM). As cardiovascular diseases represent the number one cause of death in people with T2DM, there has also been growing interest in understanding the cardiovascular actions of the GIP/GIPR axis. Although controversy surrounds whether GIPR agonism or antagonism will induce weight loss in obesity/T2DM, such actions undoubtedly will impact cardiovascular function and outcomes since obesity is a major risk factor for cardiovascular disease. Furthermore, GIPR agonism is associated with reduced progression of atherosclerotic lesions in preclinical studies. Conversely, genetic deletion of GIPR activity within cardiac myocytes of the heart results in robust protection against experimental myocardial infarction. Nonetheless, interrogation of the GIP/GIPR axis on cardiac function in humans will involve the systemic actions of the GIPR within the myocardium and other systems (e.g. adipose tissue, vasculature), which will influence the long-term future of GIPR modification for the treatment of obesity/T2DM.
Collapse
Affiliation(s)
- Amanda A Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Jadin J Chahade
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
31
|
Wang T, Liu C, Jia L, Ding J. The association between apelin polymorphisms and hypertension in China: A meta-analysis. J Renin Angiotensin Aldosterone Syst 2019; 20:1470320319827204. [PMID: 30755060 PMCID: PMC6376513 DOI: 10.1177/1470320319827204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION: Apelin plays an important part in regulating blood pressure, metabolism, and the development of cancer. Recent studies have investigated the association of apelin polymorphisms and hypertension risk, but no meta-analysis has been conducted. MATERIALS AND METHODS: Five studies were included in this meta-analysis in total. The pooled odds ratio and its corresponding 95% confidence interval were calculated by the random-effect model. RESULTS: The overall pooled odds ratio of the distribution of rs3761581 G allelic frequency was 0.90 (95% confidence interval: 0.82-1.00). In female participants, the pooled odds ratio of the frequency of G allele was 1.01 (95% confidence interval: 0.89-1.14). For males, the pooled odds ratio of the frequency of G allele was 0.69 (95% confidence interval: 0.46-1.03). As for rs56204867, the overall pooled odds ratio of the frequency of G allele was 1.09 (95% confidence interval: 0.86-1.37). In females, the pooled odds ratio of the frequencies of the G allele was 1.05 (95% confidence interval: 0.86-1.29). In male participants, the frequency of G allele did not show significant correlation with hypertension (pooled odds ratio=1.21 95% confidence interval: 0.81-1.79). CONCLUSION: This meta-analysis revealed that there was no correlation between apelin polymorphisms, rs3761581 and rs56204867, and the prevalence of hypertension.
Collapse
Affiliation(s)
- Tianyi Wang
- 1 NHC Key Laboratory of Radiobiology (Ministry of Health), Jilin University, P.R. China
| | - Conghe Liu
- 1 NHC Key Laboratory of Radiobiology (Ministry of Health), Jilin University, P.R. China
| | - Lili Jia
- 1 NHC Key Laboratory of Radiobiology (Ministry of Health), Jilin University, P.R. China
| | - Jun Ding
- 2 China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| |
Collapse
|
32
|
Wu CH, Mohammadmoradi S, Chen JZ, Sawada H, Daugherty A, Lu HS. Renin-Angiotensin System and Cardiovascular Functions. Arterioscler Thromb Vasc Biol 2018; 38:e108-e116. [PMID: 29950386 PMCID: PMC6039412 DOI: 10.1161/atvbaha.118.311282] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Chia-Hua Wu
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
| | - Shayan Mohammadmoradi
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
| | - Jeff Z Chen
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| | - Hisashi Sawada
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| | - Hong S Lu
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| |
Collapse
|
33
|
Santos RAS, Sampaio WO, Alzamora AC, Motta-Santos D, Alenina N, Bader M, Campagnole-Santos MJ. The ACE2/Angiotensin-(1-7)/MAS Axis of the Renin-Angiotensin System: Focus on Angiotensin-(1-7). Physiol Rev 2018; 98:505-553. [PMID: 29351514 PMCID: PMC7203574 DOI: 10.1152/physrev.00023.2016] [Citation(s) in RCA: 768] [Impact Index Per Article: 109.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 05/09/2017] [Accepted: 06/18/2017] [Indexed: 12/16/2022] Open
Abstract
The renin-angiotensin system (RAS) is a key player in the control of the cardiovascular system and hydroelectrolyte balance, with an influence on organs and functions throughout the body. The classical view of this system saw it as a sequence of many enzymatic steps that culminate in the production of a single biologically active metabolite, the octapeptide angiotensin (ANG) II, by the angiotensin converting enzyme (ACE). The past two decades have revealed new functions for some of the intermediate products, beyond their roles as substrates along the classical route. They may be processed in alternative ways by enzymes such as the ACE homolog ACE2. One effect is to establish a second axis through ACE2/ANG-(1-7)/MAS, whose end point is the metabolite ANG-(1-7). ACE2 and other enzymes can form ANG-(1-7) directly or indirectly from either the decapeptide ANG I or from ANG II. In many cases, this second axis appears to counteract or modulate the effects of the classical axis. ANG-(1-7) itself acts on the receptor MAS to influence a range of mechanisms in the heart, kidney, brain, and other tissues. This review highlights the current knowledge about the roles of ANG-(1-7) in physiology and disease, with particular emphasis on the brain.
Collapse
Affiliation(s)
- Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Walkyria Oliveira Sampaio
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Andreia C Alzamora
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Daisy Motta-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Natalia Alenina
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Michael Bader
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Maria Jose Campagnole-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
34
|
Patel S, Rauf A, Khan H, Abu-Izneid T. Renin-angiotensin-aldosterone (RAAS): The ubiquitous system for homeostasis and pathologies. Biomed Pharmacother 2017; 94:317-325. [PMID: 28772209 DOI: 10.1016/j.biopha.2017.07.091] [Citation(s) in RCA: 382] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/10/2017] [Accepted: 07/19/2017] [Indexed: 12/22/2022] Open
Abstract
Renin-angiotensin-aldosterone system (RAAS) is a vital system of human body, as it maintains plasma sodium concentration, arterial blood pressure and extracellular volume. Kidney-secreted renin enzyme acts on its substrate to form angiotensin II, a versatile effector peptide hormone. Every organ is affected by RAAS activation and the resultant hypertension, cell proliferation, inflammation, and fibrosis. The imbalance of renin and angiotensin II can result in an overwhelming number of chronic and acute diseases. RAAS is influenced by other enzymes, hormones, pumps and signaling pathways, hence, this review discusses important facets of this system, its crosstalk with other crucial factors like estrogen, thyroid, cortisol, kallikrein-kinin system, Wnt/β-catenin signaling, and sodium-potassium pump. The nexus of RAAS with the above-discussed systems was scantily explored before. So, this review furnishes a new perspective in comprehension of inflammation diseases. It is followed by the formulation of hypotheses, which can contribute to better management of an array of pathologies plaguing mankind. Manipulation of RAAS, by bending it towards ACE2 expression can regulate endocrine functions, which can be critical for a number of pathological management. Dietary intervention can restore RAAS to normalcy.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, San Diego, 92182, USA.
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar-23561, Khyber Pakhtunkhwa, Pakistan.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Tareq Abu-Izneid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah, P.O. Box 42, Saudi Arabia
| |
Collapse
|
35
|
Chen J, Cui L, Yuan J, Zhang S, Ma R, Sang H, Liu Q, Shan L. Protective effect of diminazene attenuates myocardial infarction in rats via increased inflammation and ACE2 activity. Mol Med Rep 2017; 16:4791-4796. [DOI: 10.3892/mmr.2017.7152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
|
36
|
Schreiber R, Paim LR, de Rossi G, Matos-Souza JR, Costa e Silva ADA, Nogueira CD, Azevedo ER, Alonso KC, Palomino Z, Sposito AC, Casarini DE, Gorla JI, Cliquet A, Nadruz W. Reduced Sympathetic Stimulus and Angiotensin 1–7 Are Related to Diastolic Dysfunction in Spinal Cord–Injured Subjects. J Neurotrauma 2017; 34:2323-2328. [DOI: 10.1089/neu.2016.4902] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Roberto Schreiber
- Department of Internal Medicine, University of Campinas, Campinas, Brazil
| | - Layde R. Paim
- Department of Internal Medicine, University of Campinas, Campinas, Brazil
| | - Guilherme de Rossi
- Department of Internal Medicine, University of Campinas, Campinas, Brazil
| | | | | | | | - Eliza R. Azevedo
- Department of Orthopedics, University of Campinas, Campinas, Brazil
| | - Karina C. Alonso
- Department of Orthopedics, University of Campinas, Campinas, Brazil
| | - Zaira Palomino
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Brazil
| | - Andrei C. Sposito
- Department of Internal Medicine, University of Campinas, Campinas, Brazil
| | - Dulce E. Casarini
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, Brazil
| | - José I. Gorla
- School of Physical Education, University of Campinas, Campinas, Brazil
| | - Alberto Cliquet
- Department of Orthopedics, University of Campinas, Campinas, Brazil
- Department of Electrical Engineering, University of São Paulo (USP), São Carlos, Brazil
| | - Wilson Nadruz
- Department of Internal Medicine, University of Campinas, Campinas, Brazil
| |
Collapse
|
37
|
Hoffmann BR, Stodola TJ, Wagner JR, Didier DN, Exner EC, Lombard JH, Greene AS. Mechanisms of Mas1 Receptor-Mediated Signaling in the Vascular Endothelium. Arterioscler Thromb Vasc Biol 2017; 37:433-445. [PMID: 28082260 DOI: 10.1161/atvbaha.116.307787] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/02/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Angiotensin II (AngII) has been shown to regulate angiogenesis and at high pathophysiological doses to cause vasoconstriction through the AngII receptor type 1. Angiotensin 1 to 7 (Ang-(1-7)) acting through the Mas1 receptor can act antagonistically to high pathophysiological levels of AngII by inducing vasodilation, whereas the effects of Ang-(1-7) signaling on angiogenesis are less defined. To complicate the matter, there is growing evidence that a subpressor dose of AngII produces phenotypes similar to Ang-(1-7). APPROACH AND RESULTS This study shows that low-dose Ang-(1-7), acting through the Mas1 receptor, promotes angiogenesis and vasodilation similar to a low, subpressor dose of AngII acting through AngII receptor type 1. In addition, we show through in vitro tube formation that Ang-(1-7) augments the angiogenic response in rat microvascular endothelial cells. Using proteomic and genomic analyses, downstream components of Mas1 receptor signaling were identified, including Rho family of GTPases, phosphatidylinositol 3-kinase, protein kinase D1, mitogen-activated protein kinase, and extracellular signal-related kinase signaling. Further experimental antagonism of extracellular signal-related kinases 1/2 and p38 mitogen-activated protein kinase signaling inhibited endothelial tube formation and vasodilation when stimulated with equimolar, low doses of either AngII or Ang-(1-7). CONCLUSIONS These results significantly expand the known Ang-(1-7)/Mas1 receptor signaling pathway and demonstrate an important distinction between the pathological effects of elevated and suppressed AngII compared with the beneficial effects of AngII normalization and Ang-(1-7) administration. The observed convergence of Ang-(1-7)/Mas1 and AngII/AngII receptor type 1 signaling at low ligand concentrations suggests a nuanced regulation in vasculature. These data also reinforce the importance of mitogen-activated protein kinase/extracellular signal-related kinase signaling in maintaining vascular function.
Collapse
Affiliation(s)
- Brian R Hoffmann
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Timothy J Stodola
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Jordan R Wagner
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Daniela N Didier
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Eric C Exner
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Julian H Lombard
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Andrew S Greene
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee.
| |
Collapse
|
38
|
Pan WQ, He YH, Su Q, Yang J, Fang YH, Ding FH, Yan XX, Liu ZH, Wang XQ, Yang K, Zhang RY, Shen WF, Zhang FR, Lu L. Association of decreased serum vasostatin-2 level with ischemic chronic heart failure and with MACE in 3-year follow-up: Vasostatin-2 prevents heart failure in myocardial infarction rats. Int J Cardiol 2016; 221:1-11. [PMID: 27395818 DOI: 10.1016/j.ijcard.2016.06.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/19/2016] [Indexed: 11/17/2022]
Abstract
BACKGROUND We investigated whether serum vasostatin-2 level is related to chronic heart failure (CHF) in patients with previous myocardial infarction (MI) and MACE in 3-year follow-up. The biological effect of vasostatin-2 on ischemic HF was evaluated in animal experiments. METHODS After exclusion of the subjects not eligible, this study included 450 patients with CHF and previous MI, and 149 healthy controls. Serum vasostatin-2 level was analyzed. CHF patients were followed up for three years and major adverse cardiac events (MACE) were recorded, defined as reinfarction, target-vessel revascularization, cardiovascular death and refractory HF requiring hospitalizations. RESULTS Notably, serum vasostatin-2 level was decreased in CHF patients than in controls, and significant difference was observed between CHF patients with MACE and those without (both P<0.05). Vasostatin-2 level was correlated with HF stages (Spearman's r=-0.288, P<0.05), LVEF (r=0.377, P<0.05) and pro-BNP level (r=-0.294, P<0.05). Multivariable logistic regression analysis suggested that vasostatin-2, conventional risk factors, severity of HF stages and LVEF were independently associated with MACE in CHF patients. Vasostatin-2 (100μg) or PBS was injected intraperitoneally every other day in MI rats, follow by echocardiography, hemodynamic analysis after 2months. Compared with PBS, vasostatin-2 treatment prevented ischemic HF in MI rats, accompanied with reduction of infarct size, remodeling, fibrosis and inflammation, mainly through inhibition of Rho, Wnt and TLR-4 pathways and modulation of renin-angiotensin system. CONCLUSION Decreased serum vasostatin-2 level is associated with ischemic CHF and with MACE in three-year follow-up. Intraperitoneal injection of vasostatin-2 protects against ischemic HF in MI rats.
Collapse
Affiliation(s)
- Wen Qi Pan
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Yu Hu He
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Qian Su
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Jie Yang
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Yue Hua Fang
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Feng Hua Ding
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Xiao Xiang Yan
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China; Institute of Cardiovascular Diseases, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Zhu Hui Liu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Xiao Qun Wang
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China; Institute of Cardiovascular Diseases, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Ke Yang
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Rui Yan Zhang
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Wei Feng Shen
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China; Institute of Cardiovascular Diseases, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Feng Ru Zhang
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China
| | - Lin Lu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, People's Republic of China; Institute of Cardiovascular Diseases, Shanghai Jiaotong University School of Medicine, Shanghai 200025, People's Republic of China.
| |
Collapse
|
39
|
Abdel-Rahman O, Alorabi M. Use of angiotensin-converting enzyme inhibitors in the prophylaxis of anthracycline or trastuzumab-related cardiac dysfunction: preclinical and clinical considerations. Expert Rev Anticancer Ther 2015; 15:829-837. [PMID: 26013380 DOI: 10.1586/14737140.2015.1047766] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Cardiac toxicity is one of the most important long-term toxicities experienced by cancer survivors so that survival rate due to anticancer therapy may be negatively affected due to cardiac complications. Thus, the search for novel methods to lower the incidence of treatment-related cardiotoxicity is a priority for all cancer-related disciplines. In this review, we provide an overview of the available preclinical and clinical data evaluating the role of angiotensin-converting enzyme inhibitors in the prophylaxis of cardiac dysfunction secondary to anthracyclines and trastuzumab use. The available clinical data suggest some degree of protective utility for the use of some angiotensin-converting enzyme inhibitors against anthracycline-related cardiotoxicity while the data are scarce with regard to a similar role for patients receiving trastuzumab.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Ain Shams University, Clinical oncology department, Faculty of medicine, Cairo, Egypt
| | | |
Collapse
|
40
|
Antagonism of angiotensin 1-7 prevents the therapeutic effects of recombinant human ACE2. J Mol Med (Berl) 2015; 93:1003-13. [PMID: 25874965 DOI: 10.1007/s00109-015-1285-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/04/2015] [Accepted: 03/27/2015] [Indexed: 01/24/2023]
Abstract
UNLABELLED Activation of the angiotensin 1-7/Mas receptor (MasR) axis counteracts angiotensin II (Ang II)-mediated cardiovascular disease. Recombinant human angiotensin-converting enzyme 2 (rhACE2) generates Ang 1-7 from Ang II. We hypothesized that the therapeutic effects of rhACE2 are dependent on Ang 1-7 action. Wild type male C57BL/6 mice (10-12 weeks old) were infused with Ang II (1.5 mg/kg/d) and treated with rhACE2 (2 mg/kg/d). The Ang 1-7 antagonist, A779 (200 ng/kg/min), was administered to a parallel group of mice. rhACE2 prevented Ang II-induced hypertrophy and diastolic dysfunction while A779 prevented these beneficial effects and precipitated systolic dysfunction. rhACE2 effectively antagonized Ang II-mediated myocardial fibrosis which was dependent on the action of Ang 1-7. Myocardial oxidative stress and matrix metalloproteinase 2 activity was further increased by Ang 1-7 inhibition even in the presence of rhACE2. Activation of Akt and endothelial nitric oxide synthase (eNOS) by rhACE2 were suppressed by the antagonism of Ang 1-7 while the activation of pathological signaling pathways was maintained. Blocking Ang 1-7 action prevents the therapeutic effects of rhACE2 in the setting of elevated Ang II culminating in systolic dysfunction. These results highlight a key cardioprotective role of Ang 1-7, and increased Ang 1-7 action represents a potential therapeutic strategy for cardiovascular diseases. KEY MESSAGES Activation of the renin-angiotensin system (RAS) plays a key pathogenic role in cardiovascular disease. ACE2, a monocarboxypeptidase, negatively regulates pathological effects of Ang II. Antagonizing Ang 1-7 prevents the therapeutic effects of recombinant human ACE2. Our results highlight a key protective role of Ang 1-7 in cardiovascular disease.
Collapse
|
41
|
Farag E, Maheshwari K, Morgan J, Sakr Esa WA, Doyle DJ. An update of the role of renin angiotensin in cardiovascular homeostasis. Anesth Analg 2015; 120:275-92. [PMID: 25602448 DOI: 10.1213/ane.0000000000000528] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The renin angiotensin system (RAS) is thought to be the body's main vasoconstrictor system, with physiological effects mediated via the interaction of angiotensin II with angiotensin I receptors (the "classic" RAS model). However, since the discovery of the heptapeptide angiotensin 1-7 and the development of the concept of the "alternate" RAS system, with its ability to reduce arterial blood pressure, our understanding of this physiologic system has changed dramatically. In this review, we focus on the newly discovered functions of the RAS, particularly the potential clinical significance of these developments, especially in the realm of new pharmacologic interventions for treating cardiovascular disease.
Collapse
Affiliation(s)
- Ehab Farag
- From the Departments of *General Anesthesia and †Outcomes Research, Cleveland Clinic, Cleveland, Ohio; ‡Anesthesiology Institute, Cleveland Clinic, Cleveland, Ohio; and §Cleveland Clinic Lerner College of Medicine of Case Western Reserve University/Department of General Anesthesia, Cleveland Clinic, Cleveland, Ohio
| | | | | | | | | |
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
|
Wu D, He L, Chen L. Apelin/APJ system: a promising therapy target for hypertension. Mol Biol Rep 2014; 41:6691-703. [DOI: 10.1007/s11033-014-3552-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/20/2014] [Indexed: 10/25/2022]
|
44
|
Time-Course of the Effects of QSYQ in Promoting Heart Function in Ameroid Constrictor-Induced Myocardial Ischemia Pigs. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:571076. [PMID: 24817898 PMCID: PMC4003740 DOI: 10.1155/2014/571076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 03/09/2014] [Accepted: 03/09/2014] [Indexed: 11/17/2022]
Abstract
We aim to investigate the therapeutic effects of QSYQ on a pig myocardial ischemia (MI) model and to determine its mechanism of action. The MI model was induced by Ameroid constriction of the left anterior descending coronary (LAD) in Ba-Ma miniature pigs. Four groups were created: model group, digoxin group, QSYQ group, and sham-operated group. Heart function, Ang II, CGMP, TXB2, BNP, and cTnT were evaluated before (3 weeks after operation: 0 weeks) and at 2, 4, and 8 weeks after drug administration. After 8 weeks of administration, the pigs were sacrificed for cardiac injury measurements. Pigs with MI showed obvious histological changes, including BNP, cTnT, Ang II, CGRP, TXB2, and ET, deregulated heart function, and increased levels of apoptotic cells in myocardial tissue. Treatment with QSYQ improved cardiac remodeling by counteracting those events. The administration of QSYQ was accompanied by a restoration of heart function and of the levels of Ang II, CGRP, TXB2, ET BNP, and cTnT. In addition, QSYQ attenuated administration, reduced the apoptosis, and decreased the level of TNF- α and active caspase-3. In conclusion, administration of QSYQ could attenuate Ameroid constrictor induced myocardial ischemia, and TNF- α and active caspase-3 seemed to be the critical potential target of QSYQ.
Collapse
|
45
|
Parajuli N, Ramprasath T, Patel VB, Wang W, Putko B, Mori J, Oudit GY. Targeting angiotensin-converting enzyme 2 as a new therapeutic target for cardiovascular diseases. Can J Physiol Pharmacol 2014; 92:558-65. [PMID: 24861775 DOI: 10.1139/cjpp-2013-0488] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a monocarboxypeptidase that metabolizes several vasoactive peptides, including angiotensin II (Ang-II; a vasoconstrictive/proliferative peptide), which it converts to Ang-(1-7). Ang-(1-7) acts through the Mas receptor to mediate vasodilatory/antiproliferative actions. The renin-angiotensin system involving the ACE-Ang-II-Ang-II type-1 receptor (AT1R) axis is antagonized by the ACE2-Ang-(1-7)-Mas receptor axis. Loss of ACE2 enhances adverse remodeling and susceptibility to pressure and volume overload. Human recombinant ACE2 may act to suppress myocardial hypertrophy, fibrosis, inflammation, and diastolic dysfunction in heart failure patients. The ACE2-Ang-(1-7)-Mas axis may present a new therapeutic target for the treatment of heart failure patients. This review is mainly focused on the analysis of ACE2, including its influence and potentially positive effects, as well as the potential use of human recombinant ACE2 as a novel therapy for the treatment cardiovascular diseases, such as hypertension and heart failure.
Collapse
Affiliation(s)
- Nirmal Parajuli
- a Division of Cardiology, Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | | | | | | | | | | | | |
Collapse
|
46
|
New perspectives in the renin-angiotensin-aldosterone system (RAAS) IV: circulating ACE2 as a biomarker of systolic dysfunction in human hypertension and heart failure. PLoS One 2014; 9:e87845. [PMID: 24691269 PMCID: PMC3972189 DOI: 10.1371/journal.pone.0087845] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 12/30/2013] [Indexed: 12/11/2022] Open
Abstract
Background Growing evidence exists for soluble Angiotensin Converting Enzyme-2 (sACE2) as a biomarker in definitive heart failure (HF), but there is little information about changes in sACE2 activity in hypertension with imminent heart failure and in reverse remodeling. Methods, Findings Patients with systolic HF (NYHAII-IV, enrolled for cardiac resynchronisation therapy, CRT, n = 100) were compared to hypertensive patients (n = 239) and to a healthy cohort (n = 45) with preserved ejection fraction (EF>50%) in a single center prospective clinical study. The status of the heart failure patients were checked before and after CRT. Biochemical (ACE and sACE2 activity, ACE concentration) and echocardiographic parameters (EF, left ventricular end-diastolic (EDD) and end-systolic diameter (ESD) and dP/dt) were measured. sACE2 activity negatively correlated with EF and positively with ESD and EDD in all patient's populations, while it was independent in the healthy cohort. sACE2 activity was already increased in the hypertensive group, where signs for imminent heart failure (slightly decreased EF and barely increased NT-proBNP levels) were detected. sACE2 activities further increased in patients with definitive heart failure (EF<50%), while sACE2 activities decreased with the improvement of the heart failure after CRT (reverse remodeling). Serum angiotensin converting enzyme (ACE) concentrations were lower in the diseased populations, but did not show a strong correlation with the echocardiographic parameters. Conclusions Soluble ACE2 activity appears to be biomarker in heart failure, and in hypertension, where heart failure may be imminent. Our data suggest that sACE2 is involved in the pathomechanism of hypertension and HF.
Collapse
|
47
|
Huynh K, Bernardo BC, McMullen JR, Ritchie RH. Diabetic cardiomyopathy: mechanisms and new treatment strategies targeting antioxidant signaling pathways. Pharmacol Ther 2014; 142:375-415. [PMID: 24462787 DOI: 10.1016/j.pharmthera.2014.01.003] [Citation(s) in RCA: 425] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 01/08/2014] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease is the primary cause of morbidity and mortality among the diabetic population. Both experimental and clinical evidence suggest that diabetic subjects are predisposed to a distinct cardiomyopathy, independent of concomitant macro- and microvascular disorders. 'Diabetic cardiomyopathy' is characterized by early impairments in diastolic function, accompanied by the development of cardiomyocyte hypertrophy, myocardial fibrosis and cardiomyocyte apoptosis. The pathophysiology underlying diabetes-induced cardiac damage is complex and multifactorial, with elevated oxidative stress as a key contributor. We now review the current evidence of molecular disturbances present in the diabetic heart, and their role in the development of diabetes-induced impairments in myocardial function and structure. Our focus incorporates both the contribution of increased reactive oxygen species production and reduced antioxidant defenses to diabetic cardiomyopathy, together with modulation of protein signaling pathways and the emerging role of protein O-GlcNAcylation and miRNA dysregulation in the progression of diabetic heart disease. Lastly, we discuss both conventional and novel therapeutic approaches for the treatment of left ventricular dysfunction in diabetic patients, from inhibition of the renin-angiotensin-aldosterone-system, through recent evidence favoring supplementation of endogenous antioxidants for the treatment of diabetic cardiomyopathy. Novel therapeutic strategies, such as gene therapy targeting the phosphoinositide 3-kinase PI3K(p110α) signaling pathway, and miRNA dysregulation, are also reviewed. Targeting redox stress and protective protein signaling pathways may represent a future strategy for combating the ever-increasing incidence of heart failure in the diabetic population.
Collapse
Affiliation(s)
- Karina Huynh
- Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia
| | | | - Julie R McMullen
- Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia; Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | - Rebecca H Ritchie
- Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
48
|
Patel VB, Clarke N, Wang Z, Fan D, Parajuli N, Basu R, Putko B, Kassiri Z, Turner AJ, Oudit GY. Angiotensin II induced proteolytic cleavage of myocardial ACE2 is mediated by TACE/ADAM-17: a positive feedback mechanism in the RAS. J Mol Cell Cardiol 2013; 66:167-76. [PMID: 24332999 DOI: 10.1016/j.yjmcc.2013.11.017] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/22/2013] [Accepted: 11/25/2013] [Indexed: 02/08/2023]
Abstract
Angiotensin converting enzyme (ACE) 2 is a key negative regulator of the renin-angiotensin system where it metabolizes angiotensin (Ang) II into Ang 1-7. We hypothesize that Ang II suppresses ACE2 by increasing TNF-α converting enzyme (TACE) activity and ACE2 cleavage. Ang II infusion (1.5 mg/kg/day) in wild-type mice for 2 weeks resulted in substantial decrease in myocardial ACE2 protein levels and activity with corresponding increase in plasma ACE2 activity, prevented by AT1R blockade. Ang II resulted in AT1R-mediated increase in myocardial TACE expression and activity, and membrane translocation of TACE. Ang II treatment in Huh7 cells exhibited AT1R-dependent metalloproteinase mediated shedding of ACE2 while transfection with siTACE prevented shedding of ACE2; cardiomyocyte-specific deletion of TACE also prevented shedding of ACE2. Reactive oxygen species played a key role since p47(phox)KO mice were resistant to Ang II-induced TACE phosphorylation and activation with preservation of myocardial ACE2 which dampened Ang II-induced cardiac dysfunction and hypertrophy. In conclusion, Ang II induces ACE2 shedding by promoting TACE activity as a positive feedback mechanism whereby Ang II facilitates the loss of its negative regulator, ACE2. In HF, elevated plasma ACE2 activity likely represents loss of the protective effects of ACE2 in the heart.
Collapse
Affiliation(s)
- Vaibhav B Patel
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Nicola Clarke
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Zuocheng Wang
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Dong Fan
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada; Department of Physiology, University of Alberta, Edmonton, Canada
| | - Nirmal Parajuli
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Ratnadeep Basu
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Brendan Putko
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Zamaneh Kassiri
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada; Department of Physiology, University of Alberta, Edmonton, Canada
| | - Anthony J Turner
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|
49
|
Role of angiotensin-converting enzyme 2 (ACE2) in diabetic cardiovascular complications. Clin Sci (Lond) 2013; 126:471-82. [DOI: 10.1042/cs20130344] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Diabetes mellitus results in severe cardiovascular complications, and heart disease and failure remain the major causes of death in patients with diabetes. Given the increasing global tide of obesity and diabetes, the clinical burden of diabetes-induced cardiovascular disease is reaching epidemic proportions. Therefore urgent actions are needed to stem the tide of diabetes which entails new prevention and treatment tools. Clinical and pharmacological studies have demonstrated that AngII (angiotensin II), the major effector peptide of the RAS (renin–angiotensin system), is a critical promoter of insulin resistance and diabetes mellitus. The role of RAS and AngII has been implicated in the progression of diabetic cardiovascular complications and AT1R (AngII type 1 receptor) blockers and ACE (angiotensin-converting enzyme) inhibitors have shown clinical benefits. ACE2, the recently discovered homologue of ACE, is a monocarboxypeptidase which converts AngII into Ang-(1–7) [angiotensin-(1–7)] which, by virtue of its actions on the MasR (Mas receptor), opposes the effects of AngII. In animal models of diabetes, an early increase in ACE2 expression and activity occurs, whereas ACE2 mRNA and protein levels have been found to decrease in older STZ (streptozotocin)-induced diabetic rats. Using the Akita mouse model of Type 1 diabetes, we have recently shown that loss of ACE2 disrupts the balance of the RAS in a diabetic state and leads to AngII/AT1R-dependent systolic dysfunction and impaired vascular function. In the present review, we will discuss the role of the RAS in the pathophysiology and treatment of diabetes and its complications with particular emphasis on potential benefits of the ACE2/Ang-(1–7)/MasR axis activation.
Collapse
|
50
|
Mori J, Zhang L, Oudit GY, Lopaschuk GD. Impact of the renin–angiotensin system on cardiac energy metabolism in heart failure. J Mol Cell Cardiol 2013; 63:98-106. [DOI: 10.1016/j.yjmcc.2013.07.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 07/12/2013] [Accepted: 07/14/2013] [Indexed: 01/12/2023]
|