1
|
Luo W, Li C, Yan G, Huang Z, Yue Y, Yang D, Zhang S. Predictive model development combining CT-FFR and SYNTAX score for major adverse cardiovascular events in complex coronary artery disease. Sci Rep 2025; 15:7152. [PMID: 40021816 PMCID: PMC11871039 DOI: 10.1038/s41598-025-91708-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/23/2025] [Indexed: 03/03/2025] Open
Abstract
Patients with complex coronary artery disease (CAD) often have poor clinical outcomes. This study aimed to develop a predictive model for assessing the 1-year risk of major adverse cardiovascular events (MACE) in patients with stable complex CAD, using retrospective data collected from January 2020 to September 2023 at Guangzhou Red Cross Hospital. The goal was to enable early risk stratification and intervention to improve clinical outcomes. A total of 369 patients were included and randomly divided into a training set (70%) for model development and a validation set (30%) for performance evaluation. Predictive factors were selected using least absolute shrinkage and selection operator (LASSO) regression, followed by logistic regression to construct the model and create a nomogram. Seven independent predictors were identified: functional SYNTAX score (OR 1.257, 95% CI 1.159-1.375), low-density lipoprotein cholesterol (LDL-C, OR 1.487, 95% CI 1.147-1.963, /1mmol/L), left ventricular ejection fraction (LVEF, OR 0.934, 95% CI 0.882-0.985, /1%), albumin (OR 0.889, 95% CI 0.809-0.974, /1g/L), pulse pressure ≥ 72 mmHg (OR 3.358, 95% CI 1.621-7.118), angiotensin-converting enzyme 2 (ACE2) ≥ 27.5 U/L (OR 2.503, 95% CI 1.290-5.014), and diabetes (OR 2.261, 95% CI 1.186-4.397). Among these, the functional SYNTAX score was the strongest predictor. The area under the receiver operating characteristic curve (AUC) was 0.843 for the training set and 0.844 for the validation set, with Youden indices of 0.561 and 0.601, respectively. Calibration curves and decision curve analysis demonstrated good predictive accuracy and clinical utility of the model. These findings suggest that the developed model has strong predictive performance for 1-year MACE risk in patients with complex CAD, and early risk stratification and intervention based on this model may improve clinical outcomes.
Collapse
Affiliation(s)
- Weiqing Luo
- Department of Cardiology, Guangzhou Red Cross Hospital of Jinan University, 396 Tongfu Middle Road, Haizhu District, Guangzhou, 510220, China
- Jinan University, Guangzhou, 510632, China
| | - Chen Li
- Department of Cardiology, Guangzhou Red Cross Hospital of Jinan University, 396 Tongfu Middle Road, Haizhu District, Guangzhou, 510220, China
- Jinan University, Guangzhou, 510632, China
| | - Guangdong Yan
- Department of Cardiology, Guangzhou Red Cross Hospital of Jinan University, 396 Tongfu Middle Road, Haizhu District, Guangzhou, 510220, China
| | - Zhichuan Huang
- Department of Cardiology, Guangzhou Red Cross Hospital of Jinan University, 396 Tongfu Middle Road, Haizhu District, Guangzhou, 510220, China
- Jinan University, Guangzhou, 510632, China
| | - Yilin Yue
- Jinan University, Guangzhou, 510632, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Deguang Yang
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Shaoheng Zhang
- Department of Cardiology, Guangzhou Red Cross Hospital of Jinan University, 396 Tongfu Middle Road, Haizhu District, Guangzhou, 510220, China.
| |
Collapse
|
2
|
Lebedin M, Ratswohl C, Garg A, Schips M, García CV, Spatt L, Thibeault C, Obermayer B, Weiner J, Velásquez IM, Gerhard C, Stubbemann P, Hanitsch LG, Pischon T, Witzenrath M, Sander LE, Kurth F, Meyer-Hermann M, de la Rosa K. Soluble ACE2 correlates with severe COVID-19 and can impair antibody responses. iScience 2024; 27:109330. [PMID: 38496296 PMCID: PMC10940809 DOI: 10.1016/j.isci.2024.109330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/25/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Identifying immune modulators that impact neutralizing antibody responses against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is of great relevance. We postulated that high serum concentrations of soluble angiotensin-converting enzyme 2 (sACE2) might mask the spike and interfere with antibody maturation toward the SARS-CoV-2-receptor-binding motif (RBM). We tested 717 longitudinal samples from 295 COVID-19 patients and showed a 2- to 10-fold increase of enzymatically active sACE2 (a-sACE2), with up to 1 μg/mL total sACE2 in moderate and severe patients. Fifty percent of COVID-19 sera inhibited ACE2 activity, in contrast to 1.3% of healthy donors and 4% of non-COVID-19 pneumonia patients. A mild inverse correlation of a-sACE2 with RBM-directed serum antibodies was observed. In silico, we show that sACE2 concentrations measured in COVID-19 sera can disrupt germinal center formation and inhibit timely production of high-affinity antibodies. We suggest that sACE2 is a biomarker for COVID-19 and that soluble receptors may contribute to immune suppression informing vaccine design.
Collapse
Affiliation(s)
- Mikhail Lebedin
- Max-Delbück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Ratswohl
- Max-Delbück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Free University of Berlin, Department of Biology, Chemistry and Pharmacy, 14195 Berlin, Berlin, Germany
| | - Amar Garg
- Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Marta Schips
- Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Clara Vázquez García
- Max-Delbück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lisa Spatt
- Max-Delbück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Charlotte Thibeault
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Benedikt Obermayer
- Core Unit Bioinformatics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - January Weiner
- Core Unit Bioinformatics, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ilais Moreno Velásquez
- Molecular Epidemiology Research Group, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Cathrin Gerhard
- Max-Delbück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Paula Stubbemann
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Leif-Gunnar Hanitsch
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Tobias Pischon
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- Molecular Epidemiology Research Group, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
- CAPNETZ STIFTUNG, 30625 Hannover, Germany
| | - Leif Erik Sander
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
| | - Michael Meyer-Hermann
- Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kathrin de la Rosa
- Max-Delbück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
3
|
Hasan MM, Saha CK, Hamidullah Mehedi HM, Chakma K, Salauddin A, Hossain MS, Sharmen F, Rafiqul Islam SM, Tanni AA, Yasmin F, Akash A, Hossain ME, Miah M, Biswas SK, Sultana N, Mannan A. Genetic determinants of SARS-CoV-2 and the clinical outcome of COVID-19 in Southern Bangladesh. Immun Inflamm Dis 2024; 12:e1171. [PMID: 38415978 PMCID: PMC10845815 DOI: 10.1002/iid3.1171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/13/2023] [Accepted: 01/21/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has had a severe impact on population health. The genetic determinants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in southern Bangladesh are not well understood. METHODS This study aimed to determine the genomic variation in SARS-CoV-2 genomes that have evolved over 2 years of the pandemic in southern Bangladesh and their association with disease outcomes and virulence of this virus. We investigated demographic variables, disease outcomes of COVID-19 patients and genomic features of SARS-CoV-2. RESULTS We observed that the disease severity was significantly higher in adults (85.3%) than in children (14.7%), because the expression of angiotensin-converting enzyme-2 (ACE-2) diminishes with ageing that causes differences in innate and adaptive immunity. The clade GK (n = 66) was remarkable between June 2021 and January 2022. Because of the mutation burden, another clade, GRA started a newly separated clustering in December 2021. The burden was significantly higher in GRA (1.5-fold) highlighted in mild symptoms of COVID-19 patients than in other clades (GH, GK, and GR). Mutations were accumulated mainly in S (22.15 mutations per segment) and ORF1ab segments. Missense (67.5%) and synonymous (18.31%) mutations were highly noticed in adult patients with mild cases rather than severe cases, especially in ORF1ab segments. Moreover, we observed many unique mutations in S protein in mild cases compared to severe, and homology modeling revealed that those might cause more folding in the protein's alpha helix and beta sheets. CONCLUSION Our study identifies some risk factors such as age comorbidities (diabetes, hypertension, and renal disease) that are associated with severe COVID-19, providing valuable insight regarding prioritizing vaccination for high-risk individuals and allocating health care and resources. The findings of this work outlined the knowledge and mutational basis of SARS-CoV-2 for the next treatment steps. Further studies are needed to confirm the effects of structural and functional proteins of SARS-CoV-2 in detail for monitoring the emergence of new variants in future.
Collapse
Affiliation(s)
- Md. Mahbub Hasan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| | | | | | - Kallyan Chakma
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| | - Asma Salauddin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
- International Centre for Diarrhoeal Disease ResearchBangladesh (icddr,b)DhakaBangladesh
| | - Md. Shakhawat Hossain
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| | - Farjana Sharmen
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| | - S. M. Rafiqul Islam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| | - Afroza Akter Tanni
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| | - Farhana Yasmin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| | - Al‐Shahriar Akash
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| | | | - Mojnu Miah
- International Centre for Diarrhoeal Disease ResearchBangladesh (icddr,b)DhakaBangladesh
| | - Sanjoy Kanti Biswas
- Department of MicrobiologyChattogram Maa‐O‐Shishu HospitalChattogramBangladesh
| | - Nahid Sultana
- Department of MicrobiologyChattogram Maa‐O‐Shishu HospitalChattogramBangladesh
| | - Adnan Mannan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological SciencesUniversity of ChittagongChattogramBangladesh
- Next Generation Sequencing, Research and Innovation Laboratory Chittagong (NRICh), Biotechnology Research and Innovation Center (BRIC)University of ChittagongChattogramBangladesh
| |
Collapse
|
4
|
Neves RL, Branquinho J, Arata JG, Bittencourt CA, Gomes CP, Riguetti M, da Mata GF, Fernandes DE, Icimoto MY, Kirsztajn GM, Pesquero JB. ACE2, ACE, DPPIV, PREP and CAT L enzymatic activities in COVID-19: imbalance of ACE2/ACE ratio and potential RAAS dysregulation in severe cases. Inflamm Res 2023; 72:1719-1731. [PMID: 37537367 DOI: 10.1007/s00011-023-01775-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/07/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023] Open
Abstract
OBJECTIVE AND DESIGN Circulating enzymatic activity and RAAS regulation in severe cases of COVID-19 remains unclear, therefore we measured the serum activity of several proteases as potential targets to control the SARS-CoV-2 infection. MATERIAL OR SUBJECTS 152 patients with COVID-19-like symptoms were grouped according to the severity of symptoms (COVID-19 negative, mild, moderate and severe). METHODS Serum samples of COVID-19 patients and controls were subjected to biochemical analysis and enzymatic assays of ACE2, ACE, DPPIV, PREP and CAT L. One-way ANOVA and multivariate logistic regression analysis were used. Statistical significance was accepted at p < 0.05. RESULTS We detected a positive correlation among comorbidities, higher C-reactive protein (CRP) and D-dimer levels with disease severity. Enzymatic assays revealed an increase in serum ACE2 and CAT L activities in severe COVID-19 patients, while ACE, DPPIV and PREP activities were significantly reduced. Notably, analysis of ACE2/ACE activity ratio suggests a possible imbalance of ANG II/ANG(1-7) ratio, in a positive association with the disease severity. CONCLUSION Our findings reveal a correlation between proteases activity and the severity of COVID-19. These enzymes together contribute to the activation of pro-inflammatory pathways, trigger a systemic activation of inflammatory mediators, leading to a RAAS dysregulation and generating a significant damage in several organs, contributing to poor outcomes of severe cases.
Collapse
Affiliation(s)
- Raquel Leão Neves
- Department of Biophysics, Center for Research and Molecular Diagnostic of Genetic Diseases, Federal University of São Paulo, São Paulo, Brazil
| | - Jéssica Branquinho
- Department of Biophysics, Center for Research and Molecular Diagnostic of Genetic Diseases, Federal University of São Paulo, São Paulo, Brazil
| | - Júlia Galanakis Arata
- Department of Biophysics, Center for Research and Molecular Diagnostic of Genetic Diseases, Federal University of São Paulo, São Paulo, Brazil
| | - Clarissa Azevedo Bittencourt
- Department of Biophysics, Center for Research and Molecular Diagnostic of Genetic Diseases, Federal University of São Paulo, São Paulo, Brazil
| | - Caio Perez Gomes
- Department of Biophysics, Center for Research and Molecular Diagnostic of Genetic Diseases, Federal University of São Paulo, São Paulo, Brazil
| | - Michelle Riguetti
- Division of Nephrology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Gustavo Ferreira da Mata
- Division of Nephrology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | - João Bosco Pesquero
- Department of Biophysics, Center for Research and Molecular Diagnostic of Genetic Diseases, Federal University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
5
|
Farid Fahmy S, El Derany MO, Khorshid H, Saleh A, El-Demerdash E. Effect of renin angiotensin blockers on angiotensin converting enzyme 2 level in cardiovascular patients. BMC Pharmacol Toxicol 2023; 24:24. [PMID: 37060024 PMCID: PMC10103030 DOI: 10.1186/s40360-023-00667-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/03/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Renin-angiotensin-aldosterone system (RAAS) is hypothesized to be in the center of COVID pathophysiology as the angiotensin converting enzyme 2 (ACE2) represents the main entrance of the virus, thus there is a need to address the effect of chronic use of RAAS blockers, as in case of treatment of cardiovascular diseases, on the expression of ACE2. Accordingly, this study aimed to clarify the effect of ACE inhibitors (ACEIs) and angiotensin-receptor blockers (ARBs) on ACE2 and to assess the correlation between ACE2 and several anthropometric and clinic-pathological factors. METHODS A total of 40 healthy controls and 60 Egyptian patients suffering from chronic cardiovascular diseases were enrolled in this study. Patients were divided into 40 patients treated with ACEIs and 20 patients treated with ARBs. Serum ACE2 levels were assessed by ELISA. RESULTS Assessment of serum ACE2 level in different groups showed a significant difference between ACEIs and healthy groups and ACEIs and ARBs group, while there was no difference between ARBs and healthy. Multivariate analysis using ACE2 level as constant and age, female sex, ACEIs use and myocardial infarction (MI) showed that there was a significant effect of female sex and ACEIs use on ACE2 level with no effect of age, MI and diabetes. CONCLUSION ACE2 levels varied between ACEIs and ARBs. It tends to be lower in ACEIs group and there is a strong positive association between ACE2 level and the female sex. This needs to be considered in Future studies to further understand the relationship between gender, sex hormones and ACE2 level. TRIAL REGISTRATION Retrospectively registered ClinicalTrials.gov ID: NCT05418361 (June 2022).
Collapse
Affiliation(s)
- Sarah Farid Fahmy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Marwa Omar El Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Hazem Khorshid
- Department of Cardiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ayman Saleh
- Department of Cardiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
- Preclinical and Translational Research Center, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
6
|
Atiku SM, Kasozi D, Campbell K. Single Nucleotide Variants (SNVs) of Angiotensin-Converting Enzymes (ACE1 and ACE2): A Plausible Explanation for the Global Variation in COVID-19 Prevalence. J Renin Angiotensin Aldosterone Syst 2023; 2023:9668008. [PMID: 37051471 PMCID: PMC10085651 DOI: 10.1155/2023/9668008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/02/2023] [Accepted: 03/10/2023] [Indexed: 04/07/2023] Open
Abstract
Background. Although it is common knowledge that the coronavirus disease of 2019 (COVID-19) and other viral infections have an uneven impact globally, the reasons for this are still indistinct. The absence of equivalent capacities worldwide in screening, testing, and reporting of cases is one of the ideas put forward to explain this discrepancy. The molecular developments are noteworthy, particularly the role played by single nucleotide polymorphisms (SNPs) in ACEs (ACE1 and ACE2). The virus can enter the host cell thanks to the transmembrane protein ACE2, which is a homolog of ACE1. Objectives. With a focus on the I/D genotype of ACE1 and the rs2285666 SNV of ACE2, we elucidated the prevalence of SNPs in ACE1 and ACE2 in various geographic locations. We examined the relationship between these SNPs and the global patterns of COVID-19 prevalence. Methods. 66 of the 127 articles obtained using PubMed, Google Scholar, and Google directly conformed to the search terms; geographical distribution of viral infections, the prevalence of COVID-19, ACE1, ACE2, SNPs, and prevalence of the DD genotype, and rs2285666. Results. The DD genotype of ACE1 and the rs2285666 SNV of ACE2 are vital in their gene expression and contribute greatly to viral disease susceptibility, development, and severity. There was generally a high prevalence of the DD genotype in Europe and America, where COVID-19 had a more devastating effect than in Asia and Africa. The prevalence of the SNV rs2285666 varied in the following order: East Asia> South Asia >America>Europe >Africa. However, there were conflicting agreements in the association of rs2285666 with COVID-19 susceptibility and prevalence. Conclusion. The ACE1 DD genotype and COVID-19 prevalence have been positively linked in a number of studies. The ACE2 rs2285666 SNV, however, has yielded no definitive results. To determine the relationship between these SNVs and COVID-19 incidence, more research is required.
Collapse
Affiliation(s)
- Saad Mahjub Atiku
- Department of Medical Laboratory Sciences, Department of Nursing and Midwifery, Faculty of Health Sciences, Muni University, P. O. Box 725 Arua, Uganda
- Department of Biochemistry, Habib Medical School Faculty of Health Sciences, Islamic University in Uganda, P. O. Box 7689 Kampala, Uganda
| | - Dennis Kasozi
- Department of Biochemistry and Sports Science, College of Natural Sciences, Makerere University Kampala, P. O. Box 7062 Kampala, Uganda
| | - Katrina Campbell
- Institute for Global Food Security, School of Biological Sciences, OG.076 / O2.037, 19 Chlorine Gardens, Queen’s University Belfast, BT9 5DL, UK
| |
Collapse
|
7
|
Kim K, Moon JH, Ahn CH, Lim S. Effect of olmesartan and amlodipine on serum angiotensin-(1-7) levels and kidney and vascular function in patients with type 2 diabetes and hypertension. Diabetol Metab Syndr 2023; 15:43. [PMID: 36899369 PMCID: PMC10005920 DOI: 10.1186/s13098-023-00987-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/27/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Recent studies suggest that angiotensin-converting enzyme 2 (ACE2) and angiotensin-(1-7) [Ang-(1-7)] might have beneficial effects on the cardiovascular system. We investigated the effects of olmesartan on the changes in serum ACE2 and Ang-(1-7) levels as well as kidney and vascular function in patients with type 2 diabetes and hypertension. METHODS This was a prospective, randomized, active comparator-controlled trial. Eighty participants with type 2 diabetes and hypertension were randomized to receive 20 mg of olmesartan (N = 40) or 5 mg of amlodipine (N = 40) once daily. The primary endpoint was changes of serum Ang-(1-7) from baseline to week 24. RESULTS Both olmesartan and amlodipine treatment for 24 weeks decreased systolic and diastolic blood pressures significantly by > 18 mmHg and > 8 mmHg, respectively. Serum Ang-(1-7) levels were more significantly increased by olmesartan treatment (25.8 ± 34.5 pg/mL → 46.2 ± 59.4 pg/mL) than by amlodipine treatment (29.2 ± 38.9 pg/mL → 31.7 ± 26.0 pg/mL), resulting in significant between-group differences (P = 0.01). Serum ACE2 levels showed a similar pattern (6.31 ± 0.42 ng/mL → 6.74 ± 0.39 ng/mL by olmesartan treatment vs. 6.43 ± 0.23 ng/mL → 6.61 ± 0.42 ng/mL by amlodipine treatment; P < 0.05). The reduction in albuminuria was significantly associated with the increases in ACE2 and Ang-(1-7) levels (r = - 0.252 and r = - 0.299, respectively). The change in Ang-(1-7) levels was positively associated with improved microvascular function (r = 0.241, P < 0.05). Multivariate regression analyses showed that increases in serum Ang-(1-7) levels were an independent predictor of a reduction in albuminuria. CONCLUSIONS These findings suggest that the beneficial effects of olmesartan on albuminuria may be mediated by increased ACE2 and Ang-(1-7) levels. These novel biomarkers may be therapeutic targets for the prevention and treatment of diabetic kidney disease. TRIAL REGISTRATION ClinicalTrials.gov NCT05189015.
Collapse
Affiliation(s)
- Kyuho Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-707, South Korea
| | - Ji Hye Moon
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-707, South Korea
| | - Chang Ho Ahn
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-707, South Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam, 463-707, South Korea.
| |
Collapse
|
8
|
Zhou G, Liu J. Prognostic value of elevated plasma angiotensin-converting enzyme 2 in cardiometabolic diseases: A review. Medicine (Baltimore) 2023; 102:e33251. [PMID: 36897667 PMCID: PMC9997766 DOI: 10.1097/md.0000000000033251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Angiotensin-converting enzyme 2, as an internal anti regulator of the renin-angiotensin hormone cascade reaction, plays a protective role in vasodilation, inhibition of fibrosis, and initiation of anti-inflammatory and antioxidative stress by degrading angiotensin II and generating angiotensin (1-7). Multiple studies have shown that plasma angiotensin-converting enzyme 2 activity is low in healthy populations without significant cardiometabolic disease, and elevated plasma angiotensin-converting enzyme 2 levels can be used as a novel biomarker of abnormal myocardial structure and/or adverse events in cardiometabolic diseases. This article aims to elaborate the determinants of plasma angiotensin-converting enzyme 2 concentration, the relevance between angiotensin-converting enzyme 2 and cardiometabolic disease risk markers, and its relative importance compared with known cardiovascular disease risk factors. Confronted with the known cardiovascular risk factors, plasma angiotensin-converting enzyme 2 (ACE2) concentration uniformly emerged as a firm predictor of abnormal myocardial structure and/or adverse events in cardiometabolic diseases and may improve the risk prediction of cardiometabolic diseases when combined with other conventional risk factors. Cardiovascular disease is the leading cause of death worldwide, while the renin-angiotensin system is the main hormone cascade system involved in the pathophysiology of cardiovascular disease. A multi-ancestry global cohort study from the general population by Narula et al revealed that plasma ACE2 concentration was strongly associated with cardiometabolic disease and might be an easily measurable indicator of renin-angiotensin system disorder. The association between this atypical hormone disorder marker and cardiometabolic disease is isolated from conventional cardiac risk factors and brain natriuretic peptide, suggesting that a clearer comprehending of the changes in plasma ACE2 concentration and activity may help us to improve the risk prediction of cardiometabolic disease, guide early diagnosis and feasible therapies, and develop and test new therapeutic targets.
Collapse
Affiliation(s)
- Gang Zhou
- Department of First Clinical Medical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Jingchen Liu
- Department of Anesthesiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
9
|
Zamai L. Hypothesis: Efficacy of early treatments with some NSAIDs in COVID-19: Might it also depend on their direct and/or indirect zinc chelating ability? Br J Pharmacol 2023; 180:279-286. [PMID: 36482040 PMCID: PMC9877557 DOI: 10.1111/bph.15989] [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: 09/10/2022] [Accepted: 11/10/2022] [Indexed: 12/13/2022] Open
Abstract
The present work argues for the involvement of the zinc chelating ability of some non-steroidal anti-inflammatory drugs as an additive mechanism able to increase their efficacy against COVID-19.
Collapse
Affiliation(s)
- Loris Zamai
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly,National Institute for Nuclear Physics (INFN)—Gran Sasso National Laboratory (LNGS)L'AquilaItaly
| |
Collapse
|
10
|
Bani Hani A, Abu Tarboush N, Bani Ali M, Alabhoul F, Alansari F, Abuhani A, Al-Kawak M, Shamoun B, Albdour S, Abu Abeeleh M, Ahram M. Serum ACE2 Level is Associated With Severe SARS-CoV-2 Infection: A Cross-Sectional Observational Study. Biomark Insights 2022; 17:11772719221125123. [PMID: 36156891 PMCID: PMC9500304 DOI: 10.1177/11772719221125123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/24/2022] [Indexed: 01/08/2023] Open
Abstract
Objectives: Angiotensin-converting enzyme 2 (ACE2) represents the primary receptor for SARS-CoV-2 to enter endothelial cells, causing coronavirus disease of 2019 (COVID-19). In this study, we investigate the association between circulating ACE2 levels with the severity of COVID-19. Methods: Serum ACE2 levels were measured in 144 COVID-19-positive subjects at hospital admission, and 123 COVID-19-negative control subjects. The association between ACE2 and clinical outcomes was analyzed. Results: About 144 COVID-19 patients and 123 healthy controls data were analyzed, the mean age of patients was 62 years and 50% of them were males. The mean age of the control group was 55 years and 63% were males. ACE-II level was measured and compared between COVID-19 patients and controls and revealed no significant differences (P > .05). ACE-II level was measured in COVID-19 patients and compared between different patient’s subgroups, ACE II level was not dependent on gender, smoking, ACE intake, or comorbidities (P > .05), and was significantly correlated with cardiovascular diseases (CVS) (P-value = .046) ICU admission (P-value = .0007) and Death (P-value = .0082). Conclusion: There was no significant difference between the COVID-19 and Control group, however, ACE2 serum level was significantly higher in patients with COVID-19 who were critically ill or non-survivors, its increased level is also associated with length of stay. Elevated ACE2 level is associated with the severity of COVID-19 disease, and it has the potential to be a predictor of the severity of the disease.
Collapse
Affiliation(s)
- Amjad Bani Hani
- Department of General Surgery, School of Medicine, The University of Jordan, Amman, Jordan
| | - Nafez Abu Tarboush
- Department of Biochemistry and Physiology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mo'ath Bani Ali
- Department of Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Fahad Alabhoul
- Department of Biochemistry and Physiology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Fahad Alansari
- Department of Biochemistry and Physiology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Ahmad Abuhani
- Department of Biochemistry and Physiology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mustafa Al-Kawak
- Department of Biochemistry and Physiology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Badea'a Shamoun
- Department of Anesthesia and Critical Care, Prince Hamza Hospital, Amman, Jordan
| | - Suzan Albdour
- Department of Biochemistry and Physiology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mahmoud Abu Abeeleh
- Department of General Surgery, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mamoun Ahram
- Department of General Surgery, School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
11
|
Plasma Angiotensin Converting Enzyme 2 (ACE2) Activity in Healthy Controls and Patients with Cardiovascular Risk Factors and/or Disease. J Pers Med 2022; 12:jpm12091495. [PMID: 36143280 PMCID: PMC9501250 DOI: 10.3390/jpm12091495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is an endogenous negative regulator of the renin-angiotensin system, a key factor in the development of cardiovascular disease (CVD). ACE2 is also used by SARS-CoV-2 for host cell entry. Given that COVID-19 is associated with hypercoagulability, it is timely to explore the potential relationship between plasma ACE2 activity and the coagulation profile. In this cross-sectional study, ACE2 activity and global coagulation assays (GCA) including thromboelastography, thrombin, and fibrin generation were measured in adult healthy controls (n = 123; mean age 41 ± 17 years; 35% male) and in patients with cardiovascular risk factors and/or disease (n = 258; mean age 65 ± 14 years; 55% male). ACE2 activity was significantly lower in controls compared to patients with cardiovascular risk factors and/or disease (median 0.10 (0.02, 3.33) vs. 5.99 (1.95, 10.37) pmol/mL/min, p < 0.001). Of the healthy controls, 48% had undetectable ACE2 activity. Controls with detectable ACE2 had lower maximum amplitude (p < 0.001). In patients with cardiovascular risk factors and/or disease, those in the 3rd tertile were older and male (p = 0.002), with a higher Framingham grade and increased number of cardiovascular risk factors (p < 0.001). In conclusion, plasma ACE2 activity is undetectable to very low in young healthy controls with minimal clinically relevant associations to GCA. Patients with cardiovascular risk factors and/or disease have increased plasma ACE2 activity, suggesting that it may be an important biomarker of endothelial dysfunction and atherosclerosis.
Collapse
|
12
|
Clerbaux LA, Albertini MC, Amigó N, Beronius A, Bezemer GFG, Coecke S, Daskalopoulos EP, del Giudice G, Greco D, Grenga L, Mantovani A, Muñoz A, Omeragic E, Parissis N, Petrillo M, Saarimäki LA, Soares H, Sullivan K, Landesmann B. Factors Modulating COVID-19: A Mechanistic Understanding Based on the Adverse Outcome Pathway Framework. J Clin Med 2022; 11:4464. [PMID: 35956081 PMCID: PMC9369763 DOI: 10.3390/jcm11154464] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/10/2022] Open
Abstract
Addressing factors modulating COVID-19 is crucial since abundant clinical evidence shows that outcomes are markedly heterogeneous between patients. This requires identifying the factors and understanding how they mechanistically influence COVID-19. Here, we describe how eleven selected factors (age, sex, genetic factors, lipid disorders, heart failure, gut dysbiosis, diet, vitamin D deficiency, air pollution and exposure to chemicals) influence COVID-19 by applying the Adverse Outcome Pathway (AOP), which is well-established in regulatory toxicology. This framework aims to model the sequence of events leading to an adverse health outcome. Several linear AOPs depicting pathways from the binding of the virus to ACE2 up to clinical outcomes observed in COVID-19 have been developed and integrated into a network offering a unique overview of the mechanisms underlying the disease. As SARS-CoV-2 infectibility and ACE2 activity are the major starting points and inflammatory response is central in the development of COVID-19, we evaluated how those eleven intrinsic and extrinsic factors modulate those processes impacting clinical outcomes. Applying this AOP-aligned approach enables the identification of current knowledge gaps orientating for further research and allows to propose biomarkers to identify of high-risk patients. This approach also facilitates expertise synergy from different disciplines to address public health issues.
Collapse
Affiliation(s)
- Laure-Alix Clerbaux
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | | | - Núria Amigó
- Biosfer Teslab SL., 43204 Reus, Spain;
- Department of Basic Medical Sciences, Universitat Rovira i Virgili (URV), 23204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Anna Beronius
- Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Gillina F. G. Bezemer
- Impact Station, 1223 JR Hilversum, The Netherlands;
- Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Sandra Coecke
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | - Evangelos P. Daskalopoulos
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | - Giusy del Giudice
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland; (G.d.G.); (D.G.); (L.A.S.)
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland; (G.d.G.); (D.G.); (L.A.S.)
| | - Lucia Grenga
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, F-30200 Bagnols-sur-Ceze, France;
| | - Alberto Mantovani
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Amalia Muñoz
- European Commission, Joint Research Centre (JRC), 2440 Geel, Belgium;
| | - Elma Omeragic
- Faculty of Pharmacy, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Nikolaos Parissis
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | - Mauro Petrillo
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | - Laura A. Saarimäki
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland; (G.d.G.); (D.G.); (L.A.S.)
| | - Helena Soares
- Laboratory of Immunobiology and Pathogenesis, Chronic Diseases Research Centre, Faculdade de Ciências Médicas Medical School, University of Lisbon, 1649-004 Lisbon, Portugal;
| | - Kristie Sullivan
- Physicians Committee for Responsible Medicine, Washington, DC 20016, USA;
| | - Brigitte Landesmann
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| |
Collapse
|
13
|
Lim HY, Burrell LM, Brook R, Nandurkar HH, Donnan G, Ho P. The Need for Individualized Risk Assessment in Cardiovascular Disease. J Pers Med 2022; 12:jpm12071140. [PMID: 35887637 PMCID: PMC9323107 DOI: 10.3390/jpm12071140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease remains the leading cause of death in the era of modern medicine despite major advancements in this field. Current available clinical surrogate markers and blood tests do not adequately predict individual risk of cardiovascular disease. A more precise and sophisticated tool that can reliably predict the thrombosis and bleeding risks at an individual level is required in order for clinicians to confidently recommend early interventions with a favorable risk–benefit profile. Critical to the development of this tool is the assessment and understanding of Virchow’s triad and its complex interactions between hypercoagulability, endothelial dysfunction and vessel flow, a fundamental concept to the development of thrombosis. This review explores the pathophysiology of cardiovascular disease stemming from the triad of factors and how individualized risk assessment can be improved through the multimodal use of tools such as global coagulation assays, endothelial biomarkers and vessel flow assessment.
Collapse
Affiliation(s)
- Hui Yin Lim
- Northern Pathology Victoria, Northern Health, Epping, Melbourne, VIC 3076, Australia; (H.Y.L.); (R.B.)
- Department of Hematology, Northern Health, Epping, Melbourne, VIC 3076, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia;
- Department of Medicine, Northern Health, University of Melbourne, Epping, Melbourne, VIC 3076, Australia
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Melbourne, VIC 3084, Australia;
| | - Louise M. Burrell
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Melbourne, VIC 3084, Australia;
| | - Rowena Brook
- Northern Pathology Victoria, Northern Health, Epping, Melbourne, VIC 3076, Australia; (H.Y.L.); (R.B.)
- Department of Hematology, Northern Health, Epping, Melbourne, VIC 3076, Australia
| | - Harshal H. Nandurkar
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia;
| | - Geoffrey Donnan
- The Melbourne Brain Centre, Royal Melbourne Hospital, University of Melbourne, Parkville, Melbourne, VIC 3010, Australia;
| | - Prahlad Ho
- Northern Pathology Victoria, Northern Health, Epping, Melbourne, VIC 3076, Australia; (H.Y.L.); (R.B.)
- Department of Hematology, Northern Health, Epping, Melbourne, VIC 3076, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia;
- Department of Medicine, Northern Health, University of Melbourne, Epping, Melbourne, VIC 3076, Australia
- Correspondence: ; Tel.: +613-8405-8480
| |
Collapse
|
14
|
Ashktorab H, Pizuorno A, Adeleye F, Laiyemo A, Dalivand MM, Aduli F, Sherif ZA, Oskrochi G, Angesom K, Oppong-Twene P, Challa SR, Okorie N, Moon ES, Romos E, Jones-Wonni B, Kone AM, Rankine S, Thrift C, Scholes D, Ekwunazu C, Banson A, Mitchell B, Maskalo G, Ross J, Curtis J, Kim R, Gilliard C, Ahuja G, Mathew J, Gavin W, Kara A, Hache-Marliere M, Palaiodimos L, Mani VR, Kalabin A, Gayam VR, Garlapati PR, Miller J, Chirumamilla LG, Jackson F, Carethers JM, Kamangar F, Brim H. Symptomatic, clinical and biomarker associations for mortality in hospitalized COVID-19 patients enriched for African Americans. BMC Infect Dis 2022; 22:552. [PMID: 35715729 PMCID: PMC9204073 DOI: 10.1186/s12879-022-07520-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 05/18/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND AIMS Initial reports on US COVID-19 showed different outcomes in different races. In this study we use a diverse large cohort of hospitalized COVID-19 patients to determine predictors of mortality. METHODS We analyzed data from hospitalized COVID-19 patients (n = 5852) between March 2020- August 2020 from 8 hospitals across the US. Demographics, comorbidities, symptoms and laboratory data were collected. RESULTS The cohort contained 3,662 (61.7%) African Americans (AA), 286 (5%) American Latinx (LAT), 1,407 (23.9%), European Americans (EA), and 93 (1.5%) American Asians (AS). Survivors and non-survivors mean ages in years were 58 and 68 for AA, 58 and 77 for EA, 44 and 61 for LAT, and 51 and 63 for AS. Mortality rates for AA, LAT, EA and AS were 14.8, 7.3, 16.3 and 2.2%. Mortality increased among patients with the following characteristics: age, male gender, New York region, cardiac disease, COPD, diabetes mellitus, hypertension, history of cancer, immunosuppression, elevated lymphocytes, CRP, ferritin, D-Dimer, creatinine, troponin, and procalcitonin. Use of mechanical ventilation (p = 0.001), shortness of breath (SOB) (p < 0.01), fatigue (p = 0.04), diarrhea (p = 0.02), and increased AST (p < 0.01), significantly correlated with death in multivariate analysis. Male sex and EA and AA race/ethnicity had higher frequency of death. Diarrhea was among the most common GI symptom amongst AAs (6.8%). When adjusting for comorbidities, significant variables among the demographics of study population were age (over 45 years old), male sex, EA, and patients hospitalized in New York. When adjusting for disease severity, significant variables were age over 65 years old, male sex, EA as well as having SOB, elevated CRP and D-dimer. Glucocorticoid usage was associated with an increased risk of COVID-19 death in our cohort. CONCLUSION Among this large cohort of hospitalized COVID-19 patients enriched for African Americans, our study findings may reflect the extent of systemic organ involvement by SARS-CoV-2 and subsequent progression to multi-system organ failure. High mortality in AA in comparison with LAT is likely related to high frequency of comorbidities and older age among AA. Glucocorticoids should be used carefully considering the poor outcomes associated with it. Special focus in treating patients with elevated liver enzymes and other inflammatory biomarkers such as CRP, troponin, ferritin, procalcitonin, and D-dimer are required to prevent poor outcomes.
Collapse
Affiliation(s)
- Hassan Ashktorab
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA.
| | - Antonio Pizuorno
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Folake Adeleye
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Adeyinka Laiyemo
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Maryam Mehdipour Dalivand
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Farshad Aduli
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Zaki A Sherif
- Department of Pathology and Cancer Center, Department of Biochemistry & Molecular Biology, Howard University College of Medicine, Washington, DC, USA
| | - Gholamreza Oskrochi
- College of Engineering and Technology, American University of the Middle East, Salmiya, Kuwait
| | - Kibreab Angesom
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Philip Oppong-Twene
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Suryanarayana Reddy Challa
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Nnaemeka Okorie
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Esther S Moon
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Edward Romos
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Boubini Jones-Wonni
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Abdoul Madjid Kone
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Sheldon Rankine
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Camelita Thrift
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Derek Scholes
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Chiamaka Ekwunazu
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Abigail Banson
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Brianna Mitchell
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Guttu Maskalo
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Jillian Ross
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Julencia Curtis
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Rachel Kim
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Chandler Gilliard
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Geeta Ahuja
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Joseph Mathew
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Warren Gavin
- Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Areeba Kara
- Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Vishnu R Mani
- Department of Trauma, Acute and Critical Care Surgery, Duke University Medical Center, Durham, NC, USA
| | - Aleksandr Kalabin
- Dartment of Surgery, Columbia University College of Physicians and Surgeons at Harlem Hospital, New York, NY, USA
| | - Vijay Reddy Gayam
- Department of Medicine, Interfaith Medical Center, New York, NY, USA
| | | | - Joseph Miller
- Departments of Emergency Medicine and Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Lakshmi Gayathri Chirumamilla
- Department of Medicine, GI Division, Cancer Center, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, DC, USA
| | - Fatimah Jackson
- Department of Pathology and Cancer Center, Department of Biochemistry & Molecular Biology, Howard University College of Medicine, Washington, DC, USA
| | - John M Carethers
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Department of Human Genetics and Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Farin Kamangar
- Department of Biology, School of Computer, Mathematical, and Natural Sciences, Morgan State University, Baltimore, MD, USA
| | - Hassan Brim
- Department of Pathology and Cancer Center, Department of Biochemistry & Molecular Biology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
15
|
Su S, Chen R, Zhang S, Shu H, Luo J. Immune system changes in those with hypertension when infected with SARS-CoV-2. Cell Immunol 2022; 378:104562. [PMID: 35901625 PMCID: PMC9183242 DOI: 10.1016/j.cellimm.2022.104562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) outbreak has become an evolving global health crisis. With an increasing incidence of primary hypertension, there is greater awareness of the relationship between primary hypertension and the immune system [including CD4+, CD8+ T cells, interleukin-17 (IL-17)/T regulatory cells (Treg) balance, macrophages, natural killer (NK) cells, neutrophils, B cells, and cytokines]. Hypertension is associated with an increased risk of various infections, post-infection complications, and increased mortality from severe infections. Despite ongoing reports on the epidemiological and clinical features of COVID-19, no articles have systematically addressed the role of primary hypertension in COVID-19 or how COVID-19 affects hypertension or specific treatment in these high-risk groups. Here, we synthesize recent advances in understanding the relationship between primary hypertension and COVID-19 and its underlying mechanisms and provide specific treatment guidelines for these high-risk groups.
Collapse
|
16
|
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
|
17
|
Quarleri J, Delpino MV. SARS-CoV-2 interacts with renin-angiotensin system: impact on the central nervous system in elderly patients. GeroScience 2022; 44:547-565. [PMID: 35157210 PMCID: PMC8853071 DOI: 10.1007/s11357-022-00528-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/08/2022] [Indexed: 01/18/2023] Open
Abstract
SARS-CoV-2 is a recently identified coronavirus that causes the current pandemic disease known as COVID-19. SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) as a receptor, suggesting that the initial steps of SARS-CoV-2 infection may have an impact on the renin-angiotensin system (RAS). Several processes are influenced by RAS in the brain. The neurological symptoms observed in COVID-19 patients, including reduced olfaction, meningitis, ischemic stroke, cerebral thrombosis, and delirium, could be associated with RAS imbalance. In this review, we focus on the potential role of disturbances in the RAS as a cause for central nervous system sequelae of SARS-CoV-2 infection in elderly patients.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus Y Sida (INBIRS), Universidad de Buenos Aires-CONICET, Paraguay 2155-Piso 11 (1121), Buenos Aires, Argentina.
| | - M Victoria Delpino
- Instituto de Investigaciones Biomédicas en Retrovirus Y Sida (INBIRS), Universidad de Buenos Aires-CONICET, Paraguay 2155-Piso 11 (1121), Buenos Aires, Argentina.
| |
Collapse
|
18
|
Fagyas M, Fejes Z, Sütő R, Nagy Z, Székely B, Pócsi M, Ivády G, Bíró E, Bekő G, Nagy A, Kerekes G, Szentkereszty Z, Papp Z, Tóth A, Kappelmayer J, Nagy B. Circulating ACE2 activity predicts mortality and disease severity in hospitalized COVID-19 patients. Int J Infect Dis 2021; 115:8-16. [PMID: 34838959 PMCID: PMC8613979 DOI: 10.1016/j.ijid.2021.11.028] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 02/08/2023] Open
Abstract
Objectives Angiotensin-converting enzyme 2 (ACE2) represents the primary receptor for SARS-CoV-2 to enter endothelial cells. Here we investigated circulating ACE2 activity to predict the severity and mortality of COVID-19. Methods Serum ACE2 activity was measured in COVID-19 (110 critically ill and 66 severely ill subjects at hospital admission and 106 follow-up samples) and in 32 non-COVID-19 severe sepsis patients. Associations between ACE2, inflammation-dependent biomarkers, pre-existing comorbidities, and clinical outcomes were studied. Results Initial ACE2 activity was significantly higher in critically ill COVID-19 patients (54.4 [36.7-90.8] mU/L) than in severe COVID-19 (34.5 [25.2-48.7] mU/L; P<0.0001) and non-COVID-19 sepsis patients (40.9 [21.4-65.7] mU/L; P=0.0260) regardless of comorbidities. Circulating ACE2 activity correlated with inflammatory biomarkers and was further elevated during the hospital stay in critically ill patients. Based on ROC-curve analysis and logistic regression test, baseline ACE2 independently indicated the severity of COVID-19 with an AUC value of 0.701 (95% CI [0.621-0.781], P<0.0001). Furthermore, non-survivors showed higher serum ACE2 activity vs. survivors at hospital admission (P<0.0001). Finally, high ACE2 activity (≥45.4 mU/L) predicted a higher risk (65 vs. 37%) for 30-day mortality (Log-Rank P<0.0001). Conclusions Serum ACE2 activity correlates with COVID-19 severity and predicts mortality.
Collapse
Affiliation(s)
- Miklós Fagyas
- Department of Cardiology, Division of Clinical Physiology
| | | | - Renáta Sütő
- Doctoral School of Kálmán Laki, University of Debrecen; Gyula Kenézy Campus, Intensive Care Unit
| | | | - Borbála Székely
- Department of Internal Medicine, Intensive Care Unit, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Marianna Pócsi
- Department of Laboratory Medicine; Doctoral School of Kálmán Laki, University of Debrecen
| | | | - Edina Bíró
- South-Pest Central Hospital, National Institute of Hematology and Infectiology, Budapest, Hungary
| | - Gabriella Bekő
- South-Pest Central Hospital, National Institute of Hematology and Infectiology, Budapest, Hungary
| | - Attila Nagy
- Department of Preventive Medicine, Faculty of Public Health, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Kerekes
- Department of Internal Medicine, Intensive Care Unit, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Zoltán Papp
- Department of Cardiology, Division of Clinical Physiology; HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Attila Tóth
- Department of Cardiology, Division of Clinical Physiology; HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | |
Collapse
|
19
|
Pucci F, Annoni F, dos Santos RAS, Taccone FS, Rooman M. Quantifying Renin-Angiotensin-System Alterations in COVID-19. Cells 2021; 10:2755. [PMID: 34685735 PMCID: PMC8535134 DOI: 10.3390/cells10102755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in a wide series of physiological processes, among which inflammation and blood pressure regulation. One of its key components, the angiotensin-converting enzyme 2, has been identified as the entry point of the SARS-CoV-2 virus into the host cells, and therefore a lot of research has been devoted to study RAS dysregulation in COVID-19. Here we discuss the alterations of the regulatory RAS axes due to SARS-CoV-2 infection on the basis of a series of recent clinical investigations and experimental analyzes quantifying, e.g., the levels and activity of RAS components. We performed a comprehensive meta-analysis of these data in view of disentangling the links between the impaired RAS functioning and the pathophysiological characteristics of COVID-19. We also review the effects of several RAS-targeting drugs and how they could potentially help restore the normal RAS functionality and minimize the COVID-19 severity. Finally, we discuss the conflicting evidence found in the literature and the open questions on RAS dysregulation in SARS-CoV-2 infection whose resolution would improve our understanding of COVID-19.
Collapse
Affiliation(s)
- Fabrizio Pucci
- 3BIO—Computational Biology and Bioinformatics, Université Libre de Bruxelles, 1050 Brussels, Belgium;
- (IB)—Interuniversity Institute of Bioinformatics in Brussels, 1050 Brussels, Belgium
| | - Filippo Annoni
- Department of Intensive Care, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium; (F.A.); (F.S.T.)
| | | | - Fabio Silvio Taccone
- Department of Intensive Care, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium; (F.A.); (F.S.T.)
| | - Marianne Rooman
- 3BIO—Computational Biology and Bioinformatics, Université Libre de Bruxelles, 1050 Brussels, Belgium;
- (IB)—Interuniversity Institute of Bioinformatics in Brussels, 1050 Brussels, Belgium
| |
Collapse
|
20
|
Alterations in ACE and ACE2 Activities and Cardiomyocyte Signaling Underlie Improved Myocardial Function in a Rat Model of Repeated Remote Ischemic Conditioning. Int J Mol Sci 2021; 22:ijms222011064. [PMID: 34681724 PMCID: PMC8537248 DOI: 10.3390/ijms222011064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
Post-ischemic left ventricular (LV) remodeling and its hypothetical prevention by repeated remote ischemic conditioning (rRIC) in male Sprague–Dawley rats were studied. Myocardial infarction (MI) was evoked by permanent ligation of the left anterior descending coronary artery (LAD), and myocardial characteristics were tested in the infarcted anterior and non-infarcted inferior LV regions four and/or six weeks later. rRIC was induced by three cycles of five-minute-long unilateral hind limb ischemia and five minutes of reperfusion on a daily basis for a period of two weeks starting four weeks after LAD occlusion. Sham operated animals served as controls. Echocardiographic examinations and invasive hemodynamic measurements revealed distinct changes in LV systolic function between four and six weeks after MI induction in the absence of rRIC (i.e., LV ejection fraction (LVEF) decreased from 52.8 ± 2.1% to 50 ± 1.6%, mean ± SEM, p < 0.05) and in the presence of rRIC (i.e., LVEF increased from 48.2 ± 4.8% to 55.2 ± 4.1%, p < 0.05). Angiotensin-converting enzyme (ACE) activity was about five times higher in the anterior LV wall at six weeks than that in sham animals. Angiotensin-converting enzyme 2 (ACE2) activity roughly doubled in post-ischemic LVs. These increases in ACE and ACE2 activities were effectively mitigated by rRIC. Ca2+-sensitivities of force production (pCa50) of LV permeabilized cardiomyocytes were increased at six weeks after MI induction together with hypophosphorylation of 1) cardiac troponin I (cTnI) in both LV regions, and 2) cardiac myosin-binding protein C (cMyBP-C) in the anterior wall. rRIC normalized pCa50, cTnI and cMyBP-C phosphorylations. Taken together, post-ischemic LV remodeling involves region-specific alterations in ACE and ACE2 activities together with changes in cardiomyocyte myofilament protein phosphorylation and function. rRIC has the potential to prevent these alterations and to improve LV performance following MI.
Collapse
|
21
|
Fagyas M, Bánhegyi V, Úri K, Enyedi A, Lizanecz E, Mányiné IS, Mártha L, Fülöp GÁ, Radovits T, Pólos M, Merkely B, Kovács Á, Szilvássy Z, Ungvári Z, Édes I, Csanádi Z, Boczán J, Takács I, Szabó G, Balla J, Balla G, Seferovic P, Papp Z, Tóth A. Changes in the SARS-CoV-2 cellular receptor ACE2 levels in cardiovascular patients: a potential biomarker for the stratification of COVID-19 patients. GeroScience 2021; 43:2289-2304. [PMID: 34674152 PMCID: PMC8529378 DOI: 10.1007/s11357-021-00467-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/22/2021] [Indexed: 01/25/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is essential for SARS-CoV-2 cellular entry. Here we studied the effects of common comorbidities in severe COVID-19 on ACE2 expression. ACE2 levels (by enzyme activity and ELISA measurements) were determined in human serum, heart and lung samples from patients with hypertension (n = 540), heart transplantation (289) and thoracic surgery (n = 49). Healthy individuals (n = 46) represented the controls. Serum ACE2 activity was increased in hypertensive subjects (132%) and substantially elevated in end-stage heart failure patients (689%) and showed a strong negative correlation with the left ventricular ejection fraction. Serum ACE2 activity was higher in male (147%), overweight (122%), obese (126%) and elderly (115%) hypertensive patients. Primary lung cancer resulted in higher circulating ACE2 activity, without affecting ACE2 levels in the surrounding lung tissue. Male sex resulted in elevated serum ACE2 activities in patients with heart transplantation or thoracic surgery (146% and 150%, respectively). Left ventricular (tissular) ACE2 activity was unaffected by sex and was lower in overweight (67%), obese (62%) and older (73%) patients with end-stage heart failure. There was no correlation between serum and tissular (left ventricular or lung) ACE2 activities. Neither serum nor tissue (left ventricle or lung) ACE2 levels were affected by RAS inhibitory medications. Abandoning of ACEi treatment (non-compliance) resulted in elevated blood pressure without effects on circulating ACE2 activities. ACE2 levels associate with the severity of cardiovascular diseases, suggestive for a role of ACE2 in the pathomechanisms of cardiovascular diseases and providing a potential explanation for the higher mortality of COVID-19 among cardiovascular patients. Abandoning RAS inhibitory medication worsens the cardiovascular status without affecting circulating or tissue ACE2 levels.
Collapse
Affiliation(s)
- Miklós Fagyas
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Viktor Bánhegyi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
- Doctoral School of Kálmán Laki, University of Debrecen, Debrecen, Hungary
- Department of Cardiac Surgery, University of Halle, Halle (Saale), Germany
| | - Katalin Úri
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
| | - Attila Enyedi
- Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Erzsébet Lizanecz
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ivetta Siket Mányiné
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
| | - Lilla Mártha
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
| | - Gábor Áron Fülöp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
- Doctoral School of Kálmán Laki, University of Debrecen, Debrecen, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Miklós Pólos
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Árpád Kovács
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Ungvári
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - István Édes
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Csanádi
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Boczán
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Takács
- Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Szabó
- Department of Cardiac Surgery, University of Halle, Halle (Saale), Germany
| | - József Balla
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - György Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Petar Seferovic
- Heart Failure Center, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond street, Debrecen, 4032, Hungary.
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
22
|
Azinheira Nobrega Cruz N, Stoll D, Casarini D, Bertagnolli M. Role of ACE2 in pregnancy and potential implications for COVID-19 susceptibility. Clin Sci (Lond) 2021; 135:1805-1824. [PMID: 34338772 PMCID: PMC8329853 DOI: 10.1042/cs20210284] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023]
Abstract
In times of coronavirus disease 2019 (COVID-19), the impact of severe acute respiratory syndrome (SARS)-coronavirus (CoV)-2 infection on pregnancy is still unclear. The presence of angiotensin-converting enzyme (ACE) 2 (ACE2), the main receptor for SARS-CoV-2, in human placentas indicates that this organ can be vulnerable for viral infection during pregnancy. However, for this to happen, additional molecular processes are critical to allow viral entry in cells, its replication and disease manifestation, particularly in the placenta and/or feto-maternal circulation. Beyond the risk of vertical transmission, COVID-19 is also proposed to deplete ACE2 protein and its biological actions in the placenta. It is postulated that such effects may impair essential processes during placentation and maternal hemodynamic adaptations in COVID-19 pregnancy, features also observed in several disorders of pregnancy. This review gathers information indicating risks and protective features related to ACE2 changes in COVID-19 pregnancies. First, we describe the mechanisms of SARS-CoV-2 infection having ACE2 as a main entry door and current evidence of viral infection in the placenta. Further, we discuss the central role of ACE2 in physiological systems such as the renin-angiotensin system (RAS) and the kallikrein-kinin system (KKS), both active during placentation and hemodynamic adaptations of pregnancy. Significant knowledge gaps are also identified and should be urgently filled to better understand the fate of ACE2 in COVID-19 pregnancies and the potential associated risks. Emerging knowledge will be able to improve the early stratification of high-risk pregnancies with COVID-19 exposure as well as to guide better management and follow-up of these mothers and their children.
Collapse
Affiliation(s)
- Nayara Azinheira Nobrega Cruz
- Department of Medicine, Discipline of Nephrology, Federal University of Sao Paulo, São Paulo, Brazil
- Research Center of the Hospital Sacré-Coeur, CIUSSS Nord-de-l’Île-de-Montréal, Montréal, Canada
| | - Danielle Stoll
- Department of Medicine, Discipline of Nephrology, Federal University of Sao Paulo, São Paulo, Brazil
| | - Dulce Elena Casarini
- Department of Medicine, Discipline of Nephrology, Federal University of Sao Paulo, São Paulo, Brazil
| | - Mariane Bertagnolli
- Research Center of the Hospital Sacré-Coeur, CIUSSS Nord-de-l’Île-de-Montréal, Montréal, Canada
- School of Physical and Occupational Therapy, Faculty of Medicine, McGill University, Montréal, Canada
| |
Collapse
|
23
|
Fusco-Allison G, Li DK, Hunter B, Jackson D, Bannon PG, Lal S, O'Sullivan JF. Optimizing the discovery and assessment of therapeutic targets in heart failure with preserved ejection fraction. ESC Heart Fail 2021; 8:3643-3655. [PMID: 34342166 PMCID: PMC8497375 DOI: 10.1002/ehf2.13504] [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: 12/01/2020] [Revised: 06/02/2021] [Accepted: 06/21/2021] [Indexed: 01/09/2023] Open
Abstract
There is an urgent need for models that faithfully replicate heart failure with preserved ejection fraction (HFpEF), now recognized as the most common form of heart failure in the world. In vitro approaches have several shortcomings, most notably the immature nature of stem cell‐derived human cardiomyocytes [induced pluripotent stem cells (iPSC)] and the relatively short lifespan of primary cardiomyocytes. Three‐dimensional ‘organoids’ incorporating mature iPSCs with other cell types such as endothelial cells and fibroblasts are a significant advance, but lack the complexity of true myocardium. Animal models can replicate many features of human HFpEF, and rodent models are the most common, and recent attempts to incorporate haemodynamic, metabolic, and ageing contributions are encouraging. Differences relating to species, physiology, heart rate, and heart size are major limitations for rodent models. Porcine models mitigate many of these shortcomings and approximate human physiology more closely, but cost and time considerations limit their potential for widespread use. Ex vivo analysis of failing hearts from animal models offer intriguing possibilities regarding cardiac substrate utilisation, but are ultimately subject to the same constrains as the animal models from which the hearts are obtained. Ex vivo approaches using human myocardial biopsies can uncover new insights into pathobiology leveraging myocardial energetics, substrate turnover, molecular changes, and systolic/diastolic function. In collaboration with a skilled cardiothoracic surgeon, left ventricular endomyocardial biopsies can be obtained at the time of valvular surgery in HFpEF patients. Critically, these tissues maintain their disease phenotype, preserving inter‐relationship of myocardial cells and extracellular matrix. This review highlights a novel approach, where ultra‐thin myocardial tissue slices from human HFpEF hearts can be used to assess changes in myocardial structure and function. We discuss current approaches to modelling HFpEF, describe in detail the novel tissue slice model, expand on exciting opportunities this model provides, and outline ways to improve this model further.
Collapse
Affiliation(s)
- Gabrielle Fusco-Allison
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Desmond K Li
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Hunter
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Dan Jackson
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Surgery, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul G Bannon
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Surgery, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F O'Sullivan
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,Faculty of Medicine, TU Dresden, Dresden, Germany
| |
Collapse
|
24
|
Bisceglia I, Gabrielli D, Canale ML, Gallucci G, Parrini I, Turazza FM, Russo G, Maurea N, Quagliariello V, Lestuzzi C, Oliva S, Di Fusco SA, Lucà F, Tarantini L, Trambaiolo P, Gulizia MM, Colivicchi F. ANMCO POSITION PAPER: cardio-oncology in the COVID era (CO and CO). Eur Heart J Suppl 2021; 23:C128-C153. [PMID: 34456641 PMCID: PMC8388610 DOI: 10.1093/eurheartj/suab067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The COVID-19 pandemic and its impact on patients with cancer and cardiovascular disease have confirmed the particular vulnerability of these populations. Indeed, not only a higher risk of contracting the infection has been reported but also an increased occurrence of a more severe course and unfavourable outcome. Beyond the direct consequences of COVID-19 infection, the pandemic has an enormous impact on global health systems. Screening programmes and non-urgent tests have been postponed; clinical trials have suffered a setback. Similarly, in the area of cardiology care, a significant decline in STEMI accesses and an increase in cases of late presenting heart attacks with increased mortality and complication rates have been reported. Health care systems must therefore get ready to tackle the 'rebound effect' that will likely show a relative increase in the short- and medium-term incidence of diseases such as heart failure, myocardial infarction, arrhythmias, and cardio- and cerebrovascular complications. Scientific societies are taking action to provide general guidance and recommendations aimed at mitigating the unfavourable outcomes of this pandemic emergency. Cardio-oncology, as an emerging discipline, is more flexible in modulating care pathways and represents a beacon of innovation in the development of multi-specialty patient management. In the era of the COVID-19 pandemic, cardio-oncology has rapidly modified its clinical care pathways and implemented flexible monitoring protocols that include targeted use of cardiac imaging, increased use of biomarkers, and telemedicine systems. The goal of these strategic adjustments is to minimize the risk of infection for providers and patients while maintaining standards of care for the treatment of oncologic and cardiovascular diseases. The aim of this document is to evaluate the impact of the pandemic on the management of cardio-oncologic patients with the-state-of-the-art knowledge about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease (COVID-19) in order to optimize medical strategies during and after the pandemic.
Collapse
Affiliation(s)
- Irma Bisceglia
- Integrated Cardiology Services, Cardio-Thoracic-Vascular Department, Azienda Ospedaliera San Camillo Forlanini, Roma, Italy
| | - Domenico Gabrielli
- Cardiology Unit, Cardio-Thoracic-Vascular Department, Azienda Ospedaliera San Camillo Forlanini, Roma, Italy
| | - Maria Laura Canale
- Cardiology Department, Nuovo Ospedale Versilia Lido Di Camaiore, LU, Italy
| | | | - Iris Parrini
- Cardiology Department, Ospedale Mauriziano Umberto I, Torino, Italy
| | | | - Giulia Russo
- Cardiovascular and Sports Medicine Department, ASUGI Trieste, Trieste, Italy
| | - Nicola Maurea
- Cardiology Department, Fondazione Pascale, Napoli, Italy
| | | | - Chiara Lestuzzi
- Cardiology Department, Centro di Riferimento Oncologico (CRO), Aviano, PN, Italy
| | - Stefano Oliva
- Cardio-Oncology Department, Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Stefania Angela Di Fusco
- Clinical and Rehabilitation Cardiology Department, Presidio Ospedaliero San Filippo Neri, ASL Roma 1, Roma, Italy
| | - Fabiana Lucà
- Cardiology Department, Grande Osp. Metropol-Bianchi Melacrino-Morelli, Reggio Calabria, Italy
| | - Luigi Tarantini
- Cardiology Department, Presidio Ospedaliero. Santa Maria Nuova—AUSL RE IRCCS, Reggio Emilia, Italy
| | | | - Michele Massimo Gulizia
- Cardiology Department, Azienda di Rilievo Nazionale e Alta Specializzazione “Garibaldi”, Catania, Italy
- Fondazione per il Tuo cuore—Heart Care Foundation, Firenze, Italy
| | - Furio Colivicchi
- Clinical and Rehabilitation Cardiology Department, Presidio Ospedaliero San Filippo Neri, ASL Roma 1, Roma, Italy
| |
Collapse
|
25
|
Human Tissue Angiotensin Converting Enzyme (ACE) Activity Is Regulated by Genetic Polymorphisms, Posttranslational Modifications, Endogenous Inhibitors and Secretion in the Serum, Lungs and Heart. Cells 2021; 10:cells10071708. [PMID: 34359878 PMCID: PMC8305685 DOI: 10.3390/cells10071708] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/22/2022] Open
Abstract
Objective: Inhibitors of the angiotensin converting enzyme (ACE) are the primarily chosen drugs to treat heart failure and hypertension. Moreover, an imbalance in tissue ACE/ACE2 activity is implicated in COVID-19. In the present study, we tested the relationships between circulating and tissue (lung and heart) ACE levels in men. Methods: Serum, lung (n = 91) and heart (n = 72) tissue samples were collected from Caucasian patients undergoing lung surgery or heart transplantation. ACE I/D genotype, ACE concentration and ACE activity were determined from serum and tissue samples. Clinical parameters were also recorded. Results: A protocol for ACE extraction was developed for tissue ACE measurements. Extraction of tissue-localized ACE was optimal in a 0.3% Triton-X-100 containing buffer, resulting in 260 ± 12% higher ACE activity over detergent-free conditions. SDS or higher Triton-X-100 concentrations inhibited the ACE activity. Serum ACE concentration correlated with ACE I/D genotype (II: 166 ± 143 ng/mL, n = 19, ID: 198 ± 113 ng/mL, n = 44 and DD: 258 ± 109 ng/mL, n = 28, p < 0.05) as expected. In contrast, ACE expression levels in the lung tissue were approximately the same irrespective of the ACE I/D genotype (II: 1423 ± 1276 ng/mg, ID: 1040 ± 712 ng/mg and DD: 930 ± 1273 ng/mg, p > 0.05) in the same patients (values are in median ± IQR). Moreover, no correlations were found between circulating and lung tissue ACE concentrations and activities (Spearman’s p > 0.05). In contrast, a significant correlation was identified between ACE activities in serum and heart tissues (Spearman’s Rho = 0.32, p < 0.01). Finally, ACE activities in lung and the serum were endogenously inhibited to similar degrees (i.e., to 69 ± 1% and 53 ± 2%, respectively). Conclusion: Our data suggest that circulating ACE activity correlates with left ventricular ACE, but not with lung ACE in human. More specifically, ACE activity is tightly coordinated by genotype-dependent expression, endogenous inhibition and secretion mechanisms.
Collapse
|
26
|
ACE2 and Apelin-13: Biomarkers with a Prognostic Value in Congestive Heart Failure. DISEASE MARKERS 2021; 2021:5569410. [PMID: 34257745 PMCID: PMC8245235 DOI: 10.1155/2021/5569410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/26/2021] [Accepted: 04/19/2021] [Indexed: 01/19/2023]
Abstract
The progression of heart failure is the result of the interaction of several pathogenetic processes that involve the activation of biomarkers belonging to the renin angiotensin aldosterone system (RAAS), to its counterregulatory mechanisms, to the sympathetic nervous system and inflammation, and to oxidative stress. This study is aimed at determining the prognostic role of biomarkers in the evolution of patients with heart failure. These biomarkers are representative of different pathogenetic pathways involved in the progression of heart failure and the possible interrelationships between them and heart remodelling. Method. This is a progressive observational study on 53 hospitalized patients with low ejection fraction heart failure, who were followed up for 12 months. The aetiology of heart failure was ischemic heart disease and dilated cardiomyopathy. The patients were clinically and biochemically evaluated by EKG (echocardiography) on admission and at 6 and 12 months. The biomarkers included in the present study were angiotensin-converting enzyme type 2 (ACE2), apelin-13, NT-proBNP (biomarkers involved in the counterregulation of RAAS), interleukin 17 (IL-17), hsCRP (inflammatory biomarkers), and urinary 8-iso-PGF2α (oxidative stress biomarker). The evolution was considered unfavourable if the patients presented complications during hospitalization, were readmitted for decompensated heart failure, or died. Results. From the study group, 14 patients (24.52%) presented an unfavourable clinical evolution. The biomarkers that were associated with the evolution of patients during hospitalization were ACE2, apelin-13, NT-proBNP, and hsCRP. Multivariate logistic regression analysis identified ACE2 and apelin-13 as independent, predictive biomarkers for the unfavourable evolution of patients over the study period. Values of ACE2 above 4000.75 pg/mL and of apelin-13 less than 402.5 pg/mL were associated with an unfavourable evolution (poor clinical outcomes). Conclusion. The serum values of ACE2 and apelin-13 correlate with the unfavourable evolution of patients with reduced ejection fraction heart failure.
Collapse
|
27
|
Lanser L, Burkert FR, Thommes L, Egger A, Hoermann G, Kaser S, Pinggera GM, Anliker M, Griesmacher A, Weiss G, Bellmann-Weiler R. Testosterone Deficiency Is a Risk Factor for Severe COVID-19. Front Endocrinol (Lausanne) 2021; 12:694083. [PMID: 34226825 PMCID: PMC8253686 DOI: 10.3389/fendo.2021.694083] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022] Open
Abstract
Background Male sex is related to increased COVID-19 severity and fatality although confirmed infections are similarly distributed between men and women. The aim of this retrospective analysis was to investigate the impact of sex hormones on disease progression and immune activation in men with COVID-19. Patients and Methods We studied for effects of sex hormones on disease severity and immune activation in 377 patients (230 men, 147 women) with PCR-confirmed SARS-CoV-2 infections hospitalized at the Innsbruck University Hospital between February and December 2020. Results Men had more severe COVID-19 with concomitant higher immune system activation upon hospital admission when compared to women. Men with a severe course of infection had lower serum total testosterone (tT) levels whereas luteinizing hormone (LH) and estradiol (E2) levels were within the normal range. tT deficiency was associated with elevated CRP (rs = - 0.567, p < 0.001), IL-6 levels (rs = - 0.563, p < 0.001), lower cholesterol levels (rs = 0.407, p < 0.001) and an increased morbidity and mortality. Men with tT levels < 100 ng/dL had a more than eighteen-fold higher in-hospital mortality risk (OR 18.243 [95%CI 2.301 - 144.639], p = 0.006) compared to men with tT levels > 230 ng/dL. Moreover, while morbidity and mortality showed a positive correlation with E2 levels at admission, we detected a negative correlation with the tT/E2 ratio upon hospital admission. Conclusion Hospitalized men with COVID-19 present with rather low testosterone levels linked to more advanced immune activation, severe clinical manifestations translating into an increased risk for ICU admission or death. The underlying mechanisms remain elusive but may include infection driven hypogonadism as well as inflammation mediated cholesterol reduction causing gonadotropin suppression and impaired androgen formation. Finally, in elderly late onset hypogonadism might also contribute to lower testosterone levels.
Collapse
Affiliation(s)
- Lukas Lanser
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| | | | - Lis Thommes
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| | - Alexander Egger
- Central Institute for Medical and Chemical Laboratory Diagnosis, Innsbruck University Hospital, Innsbruck, Austria
| | - Gregor Hoermann
- Central Institute for Medical and Chemical Laboratory Diagnosis, Innsbruck University Hospital, Innsbruck, Austria
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - Susanne Kaser
- Department of Internal Medicine I, Innsbruck Medical University, Innsbruck, Austria
| | | | - Markus Anliker
- Central Institute for Medical and Chemical Laboratory Diagnosis, Innsbruck University Hospital, Innsbruck, Austria
| | - Andrea Griesmacher
- Central Institute for Medical and Chemical Laboratory Diagnosis, Innsbruck University Hospital, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| | - Rosa Bellmann-Weiler
- Department of Internal Medicine II, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
28
|
Liu LP, Zhang XL, Li J. New perspectives on angiotensin-converting enzyme 2 and its related diseases. World J Diabetes 2021; 12:839-854. [PMID: 34168732 PMCID: PMC8192247 DOI: 10.4239/wjd.v12.i6.839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/30/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Since the worldwide outbreak of coronavirus disease 2019, angiotensin-converting enzyme 2 (ACE2) has received widespread attention as the cell receptor of the severe acute respiratory syndrome coronavirus 2 virus. At the same time, as a key enzyme in the renin-angiotensin-system, ACE2 is considered to be an endogenous negative regulator of vasoconstriction, proliferation, fibrosis, and proinflammation caused by the ACE-angiotensin II-angiotensin type 1 receptor axis. ACE2 is now implicated as being closely connected to diabetes, cardiovascular, kidney, and lung diseases, and so on. This review covers the available information on the host factors regulating ACE2 and discusses its role in a variety of pathophysiological conditions in animal models and humans.
Collapse
Affiliation(s)
- Li-Ping Liu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Xiao-Li Zhang
- TheFifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg 68135, Baden-Württemberg, Germany
| | - Jian Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, Hunan Province, China
| |
Collapse
|
29
|
Chashchin MG, Gorshkov AY, Drapkina OM. Acute coronary syndrome in COVID-19 patients. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2021. [DOI: 10.15829/1728-8800-2021-2806] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Acute coronary syndrome (ACS) is caused by an acute mismatch between myocardial oxygen demand and its supply. This mechanism is largely associated with the progression of coronary atherosclerosis in combination with an inflammatory response, hypoxemia, and blood procoagulation. Patients with the coronavirus disease 2019 (COVID-19), aggravated by cardiovascular diseases and comorbidities, are at high risk of ACS.Aim. To analyze the publications, which reflects the development of ACS in patients with COVID-19, its pathogenesis, and clinical course. Material and methods. Literature data were searched using Google Scholar, PubMed, ScienceDirect, and Cyberleninka services. The analysis included data from clinical guidelines on COVID-19, data from clinical studies, reports, and systematic reviews.Results. This literature review summarizes and systematizes the data presented in modern publications, highlights the aspects of the clinical course and pathogenetic mechanisms underlying ACS in patients with COVID-19.Conclusion. The pathogenesis of COVID-19 is inextricably associated with the widespread cytopathic effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), uncontrolled immune response that causes systemic inflammation, as well as the coagulation system activation. In patients with COVID-19, along with the atherosclerosis, these mechanisms significantly increase the risk of ACS and can worsen its in-hospital course.
Collapse
Affiliation(s)
| | - A. Yu. Gorshkov
- National Medical Research Center for Therapy and Preventive Medicine
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
30
|
Shakaib B, Zohra T, Ikram A, Shakaib MB, Ali A, Bashir A, Salman M, Khan MA, Ansari J. A comprehensive review on clinical and mechanistic pathophysiological aspects of COVID-19 Malady: How far have we come? Virol J 2021; 18:120. [PMID: 34098986 PMCID: PMC8182739 DOI: 10.1186/s12985-021-01578-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/17/2021] [Indexed: 01/08/2023] Open
Abstract
Since its outbreak in 2019, the coronavirus disease (COVID-19) has become a pandemic, affecting more than 52 million people and causing more than 1 million mortalities globally till date. Current research reveals a wide array of disease manifestations and behaviors encompassing multiple organ systems in body and immense systemic inflammation, which have been summarized in this review. Data from a number of scientific reviews, research articles, case series, observational studies, and case reports were retrieved by utilizing online search engines such as Cochrane, PubMed, and Scopus from December 2019 to November 2020. The data for prevalence of signs and symptoms, underlying disease mechanisms and comorbidities were analyzed using SPSS version 25. This review will discuss a wide range of COVID-19 clinical presentations recorded till date, and the current understanding of both the underlying general as well as system specific pathophysiologic, and pathogenetic pathways. These include direct viral penetration into host cells through ACE2 receptors, induction of inflammosomes and immune response through viral proteins, and the initiation of system-wide inflammation and cytokine production. Moreover, peripheral organ damage and underlying comorbid diseases which can lead to short term and long term, reversible and irreversible damage to the body have also been studied. We concluded that underlying comorbidities and their pathological effects on the body contributed immensely and determine the resultant disease severity and mortality of the patients. Presently there is no drug approved for treatment of COVID-19, however multiple vaccines are now in use and research for more is underway.
Collapse
Affiliation(s)
- Baila Shakaib
- Rawal Institute of Health Sciences, Islamabad, Pakistan
| | | | - Aamer Ikram
- National Institute of Health, Islamabad, Pakistan
| | | | - Amna Ali
- National Institute of Health, Islamabad, Pakistan
| | - Adnan Bashir
- National Institute of Health, Islamabad, Pakistan
| | | | | | - Jamil Ansari
- National Institute of Health, Islamabad, Pakistan
| |
Collapse
|
31
|
Kaur G, Yogeswaran S, Muthumalage T, Rahman I. Persistently Increased Systemic ACE2 Activity Is Associated With an Increased Inflammatory Response in Smokers With COVID-19. Front Physiol 2021; 12:653045. [PMID: 34122129 PMCID: PMC8194708 DOI: 10.3389/fphys.2021.653045] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Tobacco smoking is known to be involved in the pathogenesis of several cardiopulmonary diseases. Additionally, smokers are highly susceptible to infectious agents due to weakened immunity. However, the progression of lung injury based on SARS-CoV-2-mediated COVID-19 pathogenesis amongst smokers and those with pre-existing pulmonary diseases is not known. We determined the systemic levels and activity of COVID-19 associated proteins, cytokine/chemokines, and lipid mediators (lipidomics) amongst COVID-19 patients with and without a history of smoking to understand the underlying susceptible factor in the pathogenesis of COVID-19. Methods: We obtained serum from healthy (CoV−), COVID-19 positive (CoV+), and COVID-19 recovered (CoV Rec) subjects with and without a history of smoking. We conducted a Luminex multiplex assay (cytokine levels), LC/MS (eicosanoids or oxylipin panel), and ACE2 enzymatic activity assays on the serum samples to determine the systemic changes in COVID-19 patients. Results: On comparing the levels of serum ACE2 amongst COVID-19 (positive and recovered) patients and healthy controls, we found a pronounced increase in serum ACE2 levels in patients with COVID-19 infection. Furthermore, ACE2 enzyme activity was significantly increased amongst COVID-19 patients with a smoking history. Also, we analyzed the levels of Angiotensin 1–7 (Ang1–7) peptide, the product of enzymatic action of ACE2, in the serum samples. We found significantly high levels of Ang1–7 in the serum of both CoV+ and CoV Rec patients. Our data further demonstrated a smoking-induced increase in serum furin and inflammatory cytokine [IFNγ(p = 0.0836), Eotaxin (p < 0.05), MCP-1 (p < 0.05), and IL-9 (p = 0.0991)] levels in COVID-19 patients as compared to non-smoking controls. Overall, our results show that smoking adversely affects the levels of systemic inflammatory markers and COVID-19 associated proteins, thus suggesting that COVID-19 infection may have severe outcomes amongst smokers.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Shaiesh Yogeswaran
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Thivanka Muthumalage
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
32
|
Ren L, Yu S, Xu W, Overton JL, Chiamvimonvat N, Thai PN. Lack of association of antihypertensive drugs with the risk and severity of COVID-19: A meta-analysis. J Cardiol 2021; 77:482-491. [PMID: 33168337 PMCID: PMC7605745 DOI: 10.1016/j.jjcc.2020.10.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/08/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The association of antihypertensive drugs with the risk and severity of COVID-19 remains unknown. METHODS AND RESULTS We systematically searched PubMed, MEDLINE, The Cochrane Library, Cochrane Central Register of Controlled Trials (CENTRAL), ClinicalTrials.gov, and medRxiv for publications before July 13, 2020. Cohort studies and case-control studies that contain information on the association of antihypertensive agents including angiotensin-converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), calcium-channel blockers (CCBs), β-blockers, and diuretics with the risk and severity of COVID-19 were selected. The random or fixed-effects models were used to pool the odds ratio (OR) with 95% confidence interval (CI) for the outcomes. The literature search yielded 53 studies that satisfied our inclusion criteria, which comprised 39 cohort studies and 14 case-control studies. These studies included a total of 2,100,587 participants. We observed no association between prior usage of antihypertensive medications including ACEIs/ARBs, CCBs, β-blockers, or diuretics and the risk and severity of COVID-19. Additionally, when only hypertensive patients were included, the severity and mortality were lower with prior usage of ACEIs/ARBs (overall OR of 0.81, 95% CI 0.66-0.99, p < 0.05 and overall OR of 0.77, 95% CI 0.66-0.91, p < 0.01). CONCLUSIONS Taken together, usage of antihypertensive drugs is not associated with the risk and severity of COVID-19. Based on the current available literature, it is not recommended to abstain from the usage of these drugs in COVID-19 patients. REGISTRATION The meta-analysis was registered on OSF (https://osf.io/ynd5g).
Collapse
Affiliation(s)
- Lu Ren
- Department of Internal Medicine, Cardiology, UC Davis, Davis, CA, USA
| | - Shandong Yu
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Wilson Xu
- Department of Internal Medicine, Cardiology, UC Davis, Davis, CA, USA
| | - James L Overton
- Department of Internal Medicine, Cardiology, UC Davis, Davis, CA, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Cardiology, UC Davis, Davis, CA, USA; Department of Veteran Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA.
| | - Phung N Thai
- Department of Internal Medicine, Cardiology, UC Davis, Davis, CA, USA.
| |
Collapse
|
33
|
Flinn B, Royce N, Gress T, Chowdhury N, Santanam N. Dual role for angiotensin-converting enzyme 2 in Severe Acute Respiratory Syndrome Coronavirus 2 infection and cardiac fat. Obes Rev 2021; 22:e13225. [PMID: 33660398 PMCID: PMC8013367 DOI: 10.1111/obr.13225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/25/2021] [Accepted: 02/04/2021] [Indexed: 12/15/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been an increasingly prevalent target for investigation since its discovery 20 years ago. The finding that it serves a counterregulatory function within the traditional renin-angiotensin system, implicating it in cardiometabolic health, has increased its clinical relevance. Focus on ACE2's role in cardiometabolic health has largely centered on its apparent functions in the context of obesity. Interest in ACE2 has become even greater with the discovery that it serves as the cell receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), opening up numerous mechanisms for deleterious effects of infection. The proliferation of ACE2 within the literature coupled with its dual role in SARS-CoV-2 infection and obesity necessitates review of the current understanding of ACE2's physiological, pathophysiological, and potential therapeutic functions. This review highlights the roles of ACE2 in cardiac dysfunction and obesity, with focus on epicardial adipose tissue, to reconcile the data in the context of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Brendin Flinn
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Huntington, West Virginia, USA
| | - Nicholas Royce
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Huntington, West Virginia, USA
| | - Todd Gress
- Research Service, Hershel "Woody" Williams VA Medical Center, Huntington, West Virginia, USA
| | - Nepal Chowdhury
- Department of Cardiovascular and Thoracic Surgery, St. Mary's Heart Center, Huntington, WV, USA
| | - Nalini Santanam
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Huntington, West Virginia, USA
| |
Collapse
|
34
|
Hussain A, Tang O, Sun C, Jia X, Selvin E, Nambi V, Folsom A, Heiss G, Zannad F, Mosley T, Virani SS, Coresh J, Boerwinkle E, Yu B, Cunningham JW, Shah AM, Solomon SD, de Lemos JA, Hoogeveen RC, Ballantyne CM. Soluble Angiotensin-Converting Enzyme 2, Cardiac Biomarkers, Structure, and Function, and Cardiovascular Events (from the Atherosclerosis Risk in Communities Study). Am J Cardiol 2021; 146:15-21. [PMID: 33539861 PMCID: PMC8038970 DOI: 10.1016/j.amjcard.2021.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
Membrane-bound angiotensin-converting enzyme 2 is important in regulation of the renin-angiotensin-aldosterone system, but the association of cleaved soluble ACE2 (sACE2) with cardiovascular disease (CVD) is unclear. We evaluated the association of sACE2 with cardiac biomarkers, structure, and function and cardiovascular events in the Atherosclerosis Risk in Communities Study. sACE2 was measured in a subset of 497 participants (mean age 78±5.4 years, 53% men, 27% black); Cox regression analyses assessed prospective associations of sACE2 with time to first CVD event at median 6.1-year follow-up. sACE2 was higher in men, blacks, and participants with prevalent CVD, diabetes, or hypertension. Higher sACE2 levels were associated with significantly higher biomarkers of cardiac injury (high-sensitivity cardiac troponin I and T, N-terminal pro-B-type natriuretic peptide), greater left ventricular mass index, and impaired diastolic function in linear regression analyses, and with increased risk for heart failure hospitalization (adjusted hazard ratio per natural log unit increase [HR] 1.32, 95% confidence interval [CI] 1.10 to 1.58), CVD events (HR 1.34, 95% CI 1.13 to 1.60), and all-cause death (HR 1.26, 95% CI 1.01 to 1.57). In an elderly biracial cohort, sACE2 was positively associated with biomarkers reflecting myocardial injury and neurohormonal activation, left ventricular mass index, impaired diastolic function, CVD, events and all-cause death.
Collapse
Affiliation(s)
- Aliza Hussain
- Department of Medicine, Baylor College of Medicine, Houston, Texax; Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, Texas
| | - Olive Tang
- Department of Epidemiology and Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Caroline Sun
- Department of Medicine, Baylor College of Medicine, Houston, Texax
| | - Xiaoming Jia
- Department of Medicine, Baylor College of Medicine, Houston, Texax
| | - Elizabeth Selvin
- Department of Epidemiology and Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Vijay Nambi
- Department of Medicine, Baylor College of Medicine, Houston, Texax; Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, Texas; Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Aaron Folsom
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota
| | - Gerardo Heiss
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Faiez Zannad
- French Clinical Research Infrastructure Network Investigation Network Initiative-Cardiovascular and Renal Clinical Trialists, Université de Lorraine, Nancy, France
| | - Thomas Mosley
- Memory Impairment and Neurodegenerative Dementia Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Salim S Virani
- Department of Medicine, Baylor College of Medicine, Houston, Texax; Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, Texas; Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Josef Coresh
- Department of Epidemiology and Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Eric Boerwinkle
- School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| | - Bing Yu
- School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| | - Jonathan W Cunningham
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Amil M Shah
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Scott D Solomon
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - James A de Lemos
- Division of Cardiology, University of Texas-Southwestern Medical Center, Dallas, Texas
| | - Ron C Hoogeveen
- Department of Medicine, Baylor College of Medicine, Houston, Texax; Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, Texas
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, Texax; Center for Cardiometabolic Disease Prevention, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
35
|
Oz M, Lorke DE, Kabbani N. A comprehensive guide to the pharmacologic regulation of angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 entry receptor. Pharmacol Ther 2021; 221:107750. [PMID: 33275999 PMCID: PMC7854082 DOI: 10.1016/j.pharmthera.2020.107750] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023]
Abstract
The recent emergence of coronavirus disease-2019 (COVID-19) as a global pandemic has prompted scientists to address an urgent need for defining mechanisms of disease pathology and treatment. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent for COVID-19, employs angiotensin converting enzyme 2 (ACE2) as its primary target for cell surface attachment and likely entry into the host cell. Thus, understanding factors that may regulate the expression and function of ACE2 in the healthy and diseased body is critical for clinical intervention. Over 66% of all adults in the United States are currently using a prescription drug and while earlier findings have focused on possible upregulation of ACE2 expression through the use of renin angiotensin system (RAS) inhibitors, mounting evidence suggests that various other widely administered drugs used in the treatment of hypertension, heart failure, diabetes mellitus, hyperlipidemias, coagulation disorders, and pulmonary disease may also present a varied risk for COVID-19. Specifically, we summarize mechanisms on how heparin, statins, steroids and phytochemicals, besides their established therapeutic effects, may also interfere with SARS-CoV-2 viral entry into cells. We also describe evidence on the effect of several vitamins, phytochemicals, and naturally occurring compounds on ACE2 expression and activity in various tissues and disease models. This comprehensive review aims to provide a timely compendium on the potential impact of commonly prescribed drugs and pharmacologically active compounds on COVID-19 pathology and risk through regulation of ACE2 and RAS signaling.
Collapse
Key Words
- adam17, a disintegrin and metalloprotease 17
- ace, angiotensin i converting enzyme
- ace-inh., angiotensin i converting enzyme inhibitor
- ampk, amp-activated protein kinase
- ang-ii, angiotensin ii
- arb, angiotensin ii type 1-receptor blocker
- ards, acute respiratory distress syndrome
- at1-r, angiotensin ii type 1-receptor
- βarb, β-adrenergic receptor blockers
- bk, bradykinin
- ccb, calcium channel blockers
- ch25h, cholesterol-25-hydroxylase
- copd, chronic obstructive lung disease
- cox, cyclooxygenase
- covid-19, coronavirus disease-2019
- dabk, [des-arg9]-bradykinin
- erk, extracellular signal-regulated kinase
- 25hc, 25-hydroxycholesterol
- hs, heparan sulfate
- hspg, heparan sulfate proteoglycan
- ibd, inflammatory bowel disease
- map, mitogen-activated protein
- mers, middle east respiratory syndrome
- mrb, mineralocorticoid receptor blocker
- nos, nitric oxide synthase
- nsaid, non-steroid anti-inflammatory drug
- ras, renin-angiotensin system
- sars-cov, severe acute respiratory syndrome coronavirus
- sh, spontaneously hypertensive
- s protein, spike protein
- sirt1, sirtuin 1
- t2dm, type 2 diabetes mellitus
- tcm, traditional chinese medicine
- tmprss2, transmembrane protease, serine 2
- tnf, tumor necrosis factor
- ufh, unfractionated heparin
Collapse
Affiliation(s)
- Murat Oz
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Safat 13110, Kuwait.
| | - Dietrich Ernst Lorke
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nadine Kabbani
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
36
|
Herman-Edelstein M, Guetta T, Barnea A, Waldman M, Ben-Dor N, Barak Y, Kornowski R, Arad M, Hochhauser E, Aravot D. Expression of the SARS-CoV-2 receptorACE2 in human heart is associated with uncontrolled diabetes, obesity, and activation of the renin angiotensin system. Cardiovasc Diabetol 2021; 20:90. [PMID: 33906662 PMCID: PMC8078096 DOI: 10.1186/s12933-021-01275-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
Background Diabetic and obese patients are at higher risk of severe disease and cardiac injury in corona virus 2 (SARS-CoV-2) infections. Cellular entry of SARS-CoV-2 is mainly via the angiotensin-converting enzyme 2 (ACE2) receptor, which is highly expressed in normal hearts. There is a disagreement regarding the effect of factors such as obesity and diabetes on ACE2 expression in the human heart and whether treatment with renin–angiotensin system inhibitors or anti-diabetic medications increases ACE2 expression and subsequently the susceptibility to infection. We designed this study to elucidate factors that control ACE2 expression in human serum, human heart biopsies, and mice. Methods Right atrial appendage biopsies were collected from 79 patients that underwent coronary artery bypass graft (CABG) surgery. We investigated the alteration in ACE2 mRNA and protein expression in heart tissue and serum. ACE2 expression was compared with clinical risk factors: diabetes, obesity and different anti-hypertensive or anti-diabetic therapies. WT or db/db mice were infused with Angiotensin II (ATII), treated with different anti-diabetic drugs (Metformin, GLP1A and SGLT2i) were also tested. Results ACE2 gene expression was increased in diabetic hearts compared to non-diabetic hearts and was positively correlated with glycosylated hemoglobin (HbA1c), body mass index (BMI), and activation of the renin angiotensin system (RAS), and negatively correlated with ejection fraction. ACE2 was not differentially expressed in patients who were on angiotensin converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARBs) prior to the operation. We found no correlation between plasma free ACE2 and cardiac tissue ACE2 expression. Transmembrane serine protease 2 (TMPRSS2), metalloprotease ADAM10 and ADAM17 that facilitate viral-ACE2 complex entry and degradation were increased in diabetic hearts. ACE2 expression in mice was increased with ATII infusion and attenuated following anti-diabetic drugs treatment. Conclusion Patients with uncontrolled diabetes or obesity with RAS activation have higher ACE2 expressions therefore are at higher risk for severe infection. Since ACEi or ARBs show no effect on ACE2 expression in the heart further support their safety.
Collapse
Affiliation(s)
- Michal Herman-Edelstein
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel.,Nephrology Department, Rabin Medical Center, Petach Tikva, Israel
| | - Tali Guetta
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel.,Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Amir Barnea
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel.,Department of Cardiothoracic Surgery, Rabin Medical Center, Petach Tikva, Israel
| | - Maayan Waldman
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel.,Department of Cardiothoracic Surgery, Rabin Medical Center, Petach Tikva, Israel
| | - Naomi Ben-Dor
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel.,Nephrology Department, Rabin Medical Center, Petach Tikva, Israel
| | - Yaron Barak
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel.,Department of Cardiothoracic Surgery, Rabin Medical Center, Petach Tikva, Israel
| | - Ran Kornowski
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel.,Department of Cardiology, Rabin Medical Center, 49100, Petach Tikva, Israel
| | - Michael Arad
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel. .,Department of Cardiothoracic Surgery, Rabin Medical Center, Petach Tikva, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Dan Aravot
- Cardiac Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine Tel-Aviv University, Tel Aviv, Israel.,Department of Cardiothoracic Surgery, Rabin Medical Center, Petach Tikva, Israel
| |
Collapse
|
37
|
AlGhatrif M, Tanaka T, Moore AZ, Bandinelli S, Lakatta EG, Ferrucci L. Age-associated difference in circulating ACE2, the gateway for SARS-COV-2, in humans: results from the InCHIANTI study. GeroScience 2021; 43:619-627. [PMID: 33462706 PMCID: PMC7813532 DOI: 10.1007/s11357-020-00314-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
Levels of angiotensin-converting enzyme 2 (ACE2), the gateway for COVID-19 virus into the cells, have been implicated in worse COVID-19 outcomes associated with aging and cardiovascular disease (CVD). Data on age-associated differences in circulating ACE2 levels in humans and the role of CVD and medications is limited. We analyzed data from 967 participants of the InCHIANTI study, a community-dwelling cohort in the Chianti region, Italy. Relative abundance of ACE2 in plasma was assessed using a proteomics platform. CVD diagnoses, use of renin-angiotensin-aldosterone system (RAAS) antagonists: ACEi, ARBs, and aldosterone antagonists, were ascertained. Multiple linear analyses were performed to examine the independent association of ACE2 with age, CVD, and RAAS antagonist use. Age was independently associated with lower log (ACE2) in persons aged ≥ 55 years (STD β = - 0.12, p = 0.0002). ACEi treatment was also independently associated with significantly lower ACE2 levels, and ACE2 was inversely associated with weight, and positively associated with peripheral artery disease (PAD) status. There was a trend toward higher circulating ACE2 levels in hypertensive individuals, but it did not reach statistical significance. In a stratified analysis, the association between log (ACE2) and log (IL-6) was more evidenced in participants with PAD. Circulating ACE2 levels demonstrate curvilinear association with age, with older individuals beyond the sixth decade age having lower levels. ACEi was associated with greater circulating ACE2 levels. Interestingly, ACE2 was elevated in PAD and positively associated with inflammatory markers, suggesting compensatory upregulation in the setting of chronic inflammation. Further studies are needed to comprehensively characterize RAAS components with aging and disease, and assess its prognostic role in predicting COVID-19 outcomes.
Collapse
Affiliation(s)
- Majd AlGhatrif
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA.
- Longitudinal Study Section, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
- Divisions of Cardiology and Hospital Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Toshiko Tanaka
- Longitudinal Study Section, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ann Zenobia Moore
- Longitudinal Study Section, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | | | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Luigi Ferrucci
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| |
Collapse
|
38
|
Interaction between the apelinergic system and ACE2 in the cardiovascular system: therapeutic implications. Clin Sci (Lond) 2021; 134:2319-2336. [PMID: 32901821 DOI: 10.1042/cs20200479] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
The apelinergic system is widely expressed and acts through autocrine and paracrine signaling to exert protective effects, including vasodilatory, metabolic, and inotropic effects on the cardiovascular (CV) system. The apelin pathway's dominant physiological role has delineated therapeutic implications for coronary artery disease, heart failure (HF), aortic aneurysm, pulmonary arterial hypertension (PAH), and transplant vasculopathy. Apelin peptides interact with the renin-angiotensin system (RAS) by promoting angiotensin converting enzyme 2 (ACE2) transcription leading to increased ACE2 protein and activity while also antagonizing the effects of angiotensin II (Ang II). Apelin modulation of the RAS by increasing ACE2 action is limited due to its rapid degradation by proteases, including ACE2, neprilysin (NEP), and kallikrein. Apelin peptides are hence tightly regulated in a negative feedback manner by ACE2. Plasma apelin levels are suppressed in pathological conditions, but its diagnostic and prognostic utility requires further clinical exploration. Enhancing the beneficial actions of apelin peptides and ACE2 axes while complementing existing pharmacological blockade of detrimental pathways is an exciting pathway for developing new therapies. In this review, we highlight the interaction between the apelin and ACE2 systems, discuss their pathophysiological roles and potential for treating a wide array of CV diseases (CVDs).
Collapse
|
39
|
Pathangey G, Fadadu PP, Hospodar AR, Abbas AE. Angiotensin-converting enzyme 2 and COVID-19: patients, comorbidities, and therapies. Am J Physiol Lung Cell Mol Physiol 2021; 320:L301-L330. [PMID: 33237815 PMCID: PMC7938645 DOI: 10.1152/ajplung.00259.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
On March 11, 2020, the World Health Organization declared coronavirus disease 2019 (COVID-19) a pandemic, and the reality of the situation has finally caught up to the widespread reach of the disease. The presentation of the disease is highly variable, ranging from asymptomatic carriers to critical COVID-19. The availability of angiotensin-converting enzyme 2 (ACE2) receptors may reportedly increase the susceptibility and/or disease progression of COVID-19. Comorbidities and risk factors have also been noted to increase COVID-19 susceptibility. In this paper, we hereby review the evidence pertaining to ACE2's relationship to common comorbidities, risk factors, and therapies associated with the susceptibility and severity of COVID-19. We also highlight gaps of knowledge that require further investigation. The primary comorbidities of respiratory disease, cardiovascular disease, renal disease, diabetes, obesity, and hypertension had strong evidence. The secondary risk factors of age, sex, and race/genetics had limited-to-moderate evidence. The tertiary factors of ACE inhibitors and angiotensin II receptor blockers had limited-to-moderate evidence. Ibuprofen and thiazolidinediones had limited evidence.
Collapse
Affiliation(s)
- Girish Pathangey
- William Beaumont School of Medicine, Oakland University, Rochester, Michigan
| | | | | | - Amr E Abbas
- William Beaumont School of Medicine, Oakland University, Rochester, Michigan
- Department of Cardiovascular Medicine, Beaumont Hospital Royal Oak, Royal Oak, Michigan
| |
Collapse
|
40
|
Zamai L. Upregulation of the Renin-Angiotensin System Pathways and SARS-CoV-2 Infection: The Rationale for the Administration of Zinc-Chelating Agents in COVID-19 Patients. Cells 2021; 10:506. [PMID: 33673459 PMCID: PMC7997276 DOI: 10.3390/cells10030506] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
The article describes the rationale for the administration of zinc-chelating agents in COVID-19 patients. In a previous work I have highlighted that the binding of the SARS-CoV spike proteins to the zinc-metalloprotease ACE2 has been shown to induce ACE2 shedding by activating the zinc-metalloprotease ADAM17, which ultimately leads to systemic upregulation of ACE2 activity. Moreover, based on experimental models, it was also shown the detrimental effect of the excessive systemic activity of ACE2 through its downstream pathways, which leads to "clinical" manifestations resembling COVID-19. In this regard, strong upregulation of circulating ACE2 activity was recently reported in COVID-19 patients, thus supporting the previous hypothesis that COVID-19 may derive from upregulation of ACE2 activity. Based on this, a reasonable hypothesis of using inhibitors that curb the upregulation of both ACE2 and ADAM17 zinc-metalloprotease activities and consequent positive feedback-loops (initially triggered by SARS-CoV-2 and subsequently sustained independently on viral trigger) is proposed as therapy for COVID-19. In particular, zinc-chelating agents such as citrate and ethylenediaminetetraacetic acid (EDTA) alone or in combination are expected to act in protecting from COVID-19 at different levels thanks to their both anticoagulant properties and inhibitory activity on zinc-metalloproteases. Several arguments are presented in support of this hypothesis and based on the current knowledge of both beneficial/harmful effects and cost/effectiveness, the use of chelating agents in the prevention and therapy of COVID-19 is proposed. In this regard, clinical trials (currently absent) employing citrate/EDTA in COVID-19 are urgently needed in order to shed more light on the efficacy of zinc chelators against SARS-CoV-2 infection in vivo.
Collapse
Affiliation(s)
- Loris Zamai
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
- National Institute for Nuclear Physics (INFN)-Gran Sasso National Laboratory (LNGS), Assergi, 67100 L’Aquila, Italy
| |
Collapse
|
41
|
Gressens SB, Leftheriotis G, Dussaule JC, Flamant M, Levy BI, Vidal-Petiot E. Controversial Roles of the Renin Angiotensin System and Its Modulators During the COVID-19 Pandemic. Front Physiol 2021; 12:624052. [PMID: 33692701 PMCID: PMC7937723 DOI: 10.3389/fphys.2021.624052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/12/2021] [Indexed: 12/15/2022] Open
Abstract
Since December 2019, the coronavirus 2019 (COVID-19) pandemic has rapidly spread and overwhelmed healthcare systems worldwide, urging physicians to understand how to manage this novel infection. Early in the pandemic, more severe forms of COVID-19 have been observed in patients with cardiovascular comorbidities, who are often treated with renin-angiotensin aldosterone system (RAAS)-blockers, such as angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs), but whether these are indeed independent risk factors is unknown. The cellular receptor for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the membrane-bound angiotensin converting enzyme 2 (ACE2), as for SARS-CoV(-1). Experimental data suggest that expression of ACE2 may be increased by RAAS-blockers, raising concerns that these drugs may facilitate viral cell entry. On the other hand, ACE2 is a key counter-regulator of the RAAS, by degrading angiotensin II into angiotensin (1-7), and may thereby mediate beneficial effects in COVID-19. These considerations have raised concerns about the management of these drugs, and early comments shed vivid controversy among physicians. This review will describe the homeostatic balance between ACE-angiotensin II and ACE2-angiotensin (1-7) and summarize the pathophysiological rationale underlying the debated role of the RAAS and its modulators in the context of the pandemic. In addition, we will review available evidence investigating the impact of RAAS blockers on the course and prognosis of COVID-19 and discuss why retrospective observational studies should be interpreted with caution. These considerations highlight the importance of solid evidence-based data in order to guide physicians in the management of RAAS-interfering drugs in the general population as well as in patients with more or less severe forms of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Simon B Gressens
- Department of Infectious and Tropical Diseases, Assistance Publique-Hôpitaux de Paris, Bichat-Claude Bernard University Hospital, Paris, France
| | - Georges Leftheriotis
- Laboratory of Molecular Physiology and Medicine, Université Cote d'Azur, Nice, France
| | - Jean-Claude Dussaule
- Sorbonne Université, INSERM, Unité des Maladies Rénales Fréquentes et Rares: des Mécanismes Moléculaires à la Médecine Personnalisée, AP-HP, Hôpital Tenon, Paris, France.,Faculty of Medicine, Sorbonne University, Paris, France
| | - Martin Flamant
- Department of Physiology, Assistance Publique-Hôpitaux de Paris, Bichat-Claude Bernard University Hospital, Paris, France.,Inserm U1149, Centre for Research on Inflammation, Université de Paris, Paris, France
| | | | - Emmanuelle Vidal-Petiot
- Department of Physiology, Assistance Publique-Hôpitaux de Paris, Bichat-Claude Bernard University Hospital, Paris, France.,Inserm U1149, Centre for Research on Inflammation, Université de Paris, Paris, France
| |
Collapse
|
42
|
Seeherman S, Suzuki YJ. Viral Infection and Cardiovascular Disease: Implications for the Molecular Basis of COVID-19 Pathogenesis. Int J Mol Sci 2021; 22:ijms22041659. [PMID: 33562193 PMCID: PMC7914972 DOI: 10.3390/ijms22041659] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
The current pandemic of coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While this respiratory virus only causes mild symptoms in younger healthy individuals, elderly people and those with cardiovascular diseases such as systemic hypertension are susceptible to developing severe conditions that can be fatal. SARS-CoV-2 infection is also associated with an increased incidence of cardiovascular diseases such as myocardial injury, acute coronary syndrome, and thromboembolism. Understanding the mechanisms of the effects of this virus on the cardiovascular system should thus help develop therapeutic strategies to reduce the mortality and morbidity associated with SARS-CoV-2 infection. Since this virus causes severe and fatal conditions in older individuals with cardiovascular comorbidities, effective therapies targeting specific populations will likely contribute to ending this pandemic. In this review article, the effects of various viruses—including other coronaviruses, influenza, dengue, and human immunodeficiency virus—on the cardiovascular system are described to help provide molecular mechanisms of pathologies associated with SARS-CoV-2 infection and COVID-19. The goal is to provide mechanistic information from the biology of other viral infections in relation to cardiovascular pathologies for the purpose of developing improved vaccines and therapeutic agents effective in preventing and/or treating the acute and long-term consequences of SARS-CoV-2 and COVID-19.
Collapse
Affiliation(s)
- Sarah Seeherman
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA;
| | - Yuichiro J. Suzuki
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20007, USA
- Correspondence:
| |
Collapse
|
43
|
Lopes-Pacheco M, Silva PL, Cruz FF, Battaglini D, Robba C, Pelosi P, Morales MM, Caruso Neves C, Rocco PRM. Pathogenesis of Multiple Organ Injury in COVID-19 and Potential Therapeutic Strategies. Front Physiol 2021; 12:593223. [PMID: 33584343 PMCID: PMC7876335 DOI: 10.3389/fphys.2021.593223] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/08/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory disease coronavirus 2 (SARS-CoV-2, formerly 2019-nCoV) is a novel coronavirus that has rapidly disseminated worldwide, causing the coronavirus disease 2019 (COVID-19) pandemic. As of January 6th, 2021, there were over 86 million global confirmed cases, and the disease has claimed over 1.87 million lives (a ∼2.2% case fatality rate). SARS-CoV-2 is able to infect human cells by binding its spike (S) protein to angiotensin-conversing enzyme 2 (ACE2), which is expressed abundantly in several cell types and tissues. ACE2 has extensive biological activities as a component of the renin-angiotensin-aldosterone system (RAAS) and plays a pivotal role as counter-regulator of angiotensin II (Ang II) activity by converting the latter to Ang (1-7). Virion binding to ACE2 for host cell entry leads to internalization of both via endocytosis, as well as activation of ADAM17/TACE, resulting in downregulation of ACE2 and loss of its protective actions in the lungs and other organs. Although COVID-19 was initially described as a purely respiratory disease, it is now known that infected individuals can rapidly progress to a multiple organ dysfunction syndrome. In fact, all human structures that express ACE2 are susceptible to SARS-CoV-2 infection and/or to the downstream effects of reduced ACE2 levels, namely systemic inflammation and injury. In this review, we aim to summarize the major features of SARS-CoV-2 biology and the current understanding of COVID-19 pathogenesis, as well as its clinical repercussions in the lung, heart, kidney, bowel, liver, and brain. We also highlight potential therapeutic targets and current global efforts to identify safe and effective therapies against this life-threatening condition.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science, Technology and Innovation, Brasília, Brazil
- COVID-19 Virus Network, Brazilian Council for Scientific and Technological Development, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science, Technology and Innovation, Brasília, Brazil
- COVID-19 Virus Network, Brazilian Council for Scientific and Technological Development, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
| | - Denise Battaglini
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Chiara Robba
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostic, University of Genoa, Genoa, Italy
| | - Marcelo Marcos Morales
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science, Technology and Innovation, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Celso Caruso Neves
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Brazilian Council for Scientific and Technological Development, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
- Laboratory of Biochemistry and Cell Signaling, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science, Technology and Innovation, Brasília, Brazil
- COVID-19 Virus Network, Brazilian Council for Scientific and Technological Development, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Level of the SARS-CoV-2 receptor ACE2 activity is highly elevated in old-aged patients with aortic stenosis: implications for ACE2 as a biomarker for the severity of COVID-19. GeroScience 2021; 43:19-29. [PMID: 33469835 PMCID: PMC7815502 DOI: 10.1007/s11357-020-00300-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has a high mortality in elderly patients with pre-existing cardiovascular diseases. The cellular receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the angiotensin-converting enzyme 2 (ACE2), thereby implicating a link between cardiovascular diseases and SARS-CoV-2 susceptibility. Aortic stenosis (AS) represents a chronic inflammatory state with severe cardiovascular complications in the elderly, a prime condition for COVID-19 mortality. The circulating ACE2 levels were measured in 111 patients with severe AS and compared to patients with hypertension and healthy individuals. About 4 times higher circulating ACE2 activity was found in patients with severe AS than in hypertensives or healthy individuals (88.3 ± 61.6., n = 111, 20.6 ± 13.4, n = 540, and 16.1 ± 7.4 mU/L, n = 46, respectively). Patients with severe AS were older than patients with hypertension (80 ± 6 years vs. 60 ± 15 years, P < 0.05). Serum ACE2 activity correlated negatively with the left ventricular ejection fraction, aortic root area, TAPSE, and positively with the right ventricular systolic pressure, cardiac diameters in patients with AS. In contrast, circulating ACE2 activity was independent of the blood pressure, peak flow velocity at the aortic root, kidney function (GFR), and inflammatory state (CRP). We found no effect of RAAS inhibitory drugs on the serum ACE2 activity in this group of patients. Our results illustrate circulating ACE2 as a potential interface between chronic inflammation, cardiovascular disease, and COVID-19 susceptibility. Elderly patients with AS have markedly elevated ACE2 levels together with altered left and right ventricular functions, which may pose higher risks during COVID-19. Our clinical data do not support a role for RAAS inhibitors in regulating circulating ACE2 levels.
Collapse
|
45
|
Tariq S, Van Eeden C, Tervaert JWC, Osman MS. COVID-19, rheumatic diseases and immune dysregulation-a perspective. Clin Rheumatol 2021; 40:433-442. [PMID: 33411143 PMCID: PMC7788381 DOI: 10.1007/s10067-020-05529-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/30/2020] [Accepted: 11/26/2020] [Indexed: 12/26/2022]
Abstract
The COVID-19 pandemic has resulted in widespread hospitalisations and deaths around the world. As patients with rheumatic diseases generally have increased risk of infections and complications, understandably, there is significant concern of the impact of SARS-CoV-2 on these patients. However, there is a paucity of data in rheumatic patients. We review mechanisms through which SARS-CoV-2 results in infection, including ACE2 receptor, and complications (including immune dysregulation, thrombosis and complement activation). We assess these pathways in patients with rheumatic disease and those on immune modulating therapy. Although data thus far does not appear to show worse outcomes in rheumatic patients as a whole, given alterations in the underlying immune pathways in certain diseases (such as systemic lupus erythematosus), we posit that the risk is not equal in all rheumatic patients. We also discuss the benefit of underlying disease control with respect to COVID-19 risk reduction and potential increased risk of disease flares following viral infection from an immune standpoint.
Collapse
Affiliation(s)
- Shahna Tariq
- Division of Rheumatology, Department of Medicine, University of Alberta, 8-130 Clinical Sciences Building, Edmonton, Alberta, T6G 2B7, Canada
| | - Charmaine Van Eeden
- Division of Rheumatology, Department of Medicine, University of Alberta, 8-130 Clinical Sciences Building, Edmonton, Alberta, T6G 2B7, Canada
| | - Jan Willem Cohen Tervaert
- Division of Rheumatology, Department of Medicine, University of Alberta, 8-130 Clinical Sciences Building, Edmonton, Alberta, T6G 2B7, Canada
| | - Mohammed S Osman
- Division of Rheumatology, Department of Medicine, University of Alberta, 8-130 Clinical Sciences Building, Edmonton, Alberta, T6G 2B7, Canada.
| |
Collapse
|
46
|
Senapati S, Banerjee P, Bhagavatula S, Kushwaha PP, Kumar S. Contributions of human ACE2 and TMPRSS2 in determining host-pathogen interaction of COVID-19. J Genet 2021; 100:12. [PMID: 33707363 PMCID: PMC7904510 DOI: 10.1007/s12041-021-01262-w] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 02/05/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is at present an emerging global public health crisis. Angiotensin converting enzyme 2 (ACE2) and trans-membrane protease serine 2 (TMPRSS2) are the two major host factors that contribute to the virulence of SARS-CoV-2 and pathogenesis of coronavirus disease-19 (COVID-19). Transmission of SARS-CoV-2 from animal to human is considered a rare event that necessarily requires strong evolutionary adaptations. Till date no other human cellular receptors are identified beside ACE2 for SARS-CoV-2 entry inside the human cell. Proteolytic cleavage of viral spike (S)-protein and ACE2 by TMPRSS2 began the entire host-pathogen interaction initiated with the physical binding of ACE2 to S-protein. SARS-CoV-2 S-protein binds to ACE2 with much higher affinity and stability than that of SARS-CoVs. Molecular interactions between ACE2-S and TMPRSS2-S are crucial and preciously mediated by specific residues. Structural stability, binding affinity and level of expression of these three interacting proteins are key susceptibility factors for COVID-19. Specific protein-protein interactions (PPI) are being identified that explains uniqueness of SARS-CoV-2 infection. Amino acid substitutions due to naturally occurring genetic polymorphisms potentially alter these PPIs and poses further clinical heterogeneity of COVID-19. Repurposing of several phytochemicals and approved drugs against ACE2, TMPRSS2 and S-protein have been proposed that could inhibit PPI between them. We have also identified some novel lead phytochemicals present in Azadirachta indica and Aloe barbadensis which could be utilized for further in vitro and in vivo anti-COVID-19 drug discovery. Uncovering details of ACE2-S and TMPRSS2-S interactions would further contribute to future research on COVID-19.
Collapse
Affiliation(s)
- Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda 151 401, India.
| | | | | | | | | |
Collapse
|
47
|
Larouche‐Lebel É, Loughran KA, Huh T, Oyama MA. Effect of angiotensin receptor blockers and angiotensin converting enzyme 2 on plasma equilibrium angiotensin peptide concentrations in dogs with heart disease. J Vet Intern Med 2021; 35:22-32. [PMID: 33368659 PMCID: PMC7848346 DOI: 10.1111/jvim.16025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The pathophysiology of heart failure involves maladaptive angiotensin peptides (APs) and enzymes, including angiotensin 2 (AT2) and angiotensin converting enzyme (ACE), as well as recently described alternative components, such as angiotensin 1-7 (Ang1-7) and angiotensin converting enzyme 2 (ACE2). The relative effects of different neurohormonal-targeting drugs on balance of APs in dogs with heart disease are unknown. HYPOTHESIS/OBJECTIVES Plasma AP concentrations differ in dogs receiving angiotensin converting enzyme inhibitors (ACEIs) vs angiotensin receptor blockers (ARBs) and recombinant human ACE2 (rhACE2) will further increase these differences. ANIMALS Eight dogs with degenerative mitral valve disease (DMVD). METHODS Prospective open-label trial. Equilibrium concentrations of APs from plasma during PO ACEI treatment and then after 14 days of PO ARB treatment using telmisartan were measured using liquid chromatography-tandem mass spectroscopy before and after in vitro incubation with rhACE2. RESULTS Concentration of Ang1-7 was increased during ARB treatment (Ang1-7: 443 pg/mL; 95% confidence interval [CI] = 247-794 pg/mL) vs ACEI (Ang1-7: 182 pg/mL; 95% CI = 66.2-503 pg/mL; P = .01). Incubation with rhACE2 decreased traditional APs while increasing beneficial alternative APs, and Ang1-7 was significantly higher in the ARB + rhACE2 (880 pg/mL; 95% CI = 560-1383 pg/mL) vs ACEI + rhACE2 (455 pg/mL; 95% CI = 188-1104 pg/mL; P = .03) group. The most favorable theoretical AP profile was achieved in the ARB + rhACE2 group. CONCLUSIONS AND CLINICAL IMPORTANCE The AP profile during telmisartan treatment is associated with higher plasma Ang1-7 as compared with during ACEI. This favorable shift is potentiated in vitro by combination of ARB + rhACE2. These data support potential AP-targeting strategies and drugs in dogs with DMVD.
Collapse
Affiliation(s)
- Éva Larouche‐Lebel
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Kerry A. Loughran
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Terry Huh
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Mark A. Oyama
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Translational Medicine and Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
48
|
Moorthy N, Saligrama Ramegowda K, Jain S, Bharath G, Sinha A, Nanjappa MC, Christopher R. Role of Angiotensin-Converting Enzyme (ACE) gene polymorphism and ACE activity in predicting outcome after acute myocardial infarction. IJC HEART & VASCULATURE 2021; 32:100701. [PMID: 33426268 PMCID: PMC7782316 DOI: 10.1016/j.ijcha.2020.100701] [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: 08/28/2020] [Revised: 12/01/2020] [Accepted: 12/06/2020] [Indexed: 12/06/2022]
Abstract
The role of ACE gene polymorphism and its influence on ischemic heart disease and acute coronary syndrome are studied in last so many years without any concrete conclusion. In this study we investigated role of ACE gene polymorphism and Ace activity in large number of study population. The ACE (I/D) polymorphism showed no association with development of acute STEMI. Neither ACE I/D polymorphism nor ACE activity predicted in-hospital mortality in patients admitted with acute STEMI. Hence knowledge of ACE polymorphism and Ace activity is not useful in predicting STEMI or mortality after STEMI.
Background The Ace polymorphism had shown association with ACE activity, premature atherosclerosis, myocardial infarction, LV dysfunction, LV remodelling, severity and extent of CAD and mortality after MI. Though ACE I/D polymorphism has been reported to be associated with various cardiovascular diseases it remained a controversial risk factor and studies have presented conflicting results. This study was designed to determine the association between ACE) gene insertion/deletion (I/D) polymorphism, ACE activity and acute STEMI in Indian population and to determine its influence on outcome after acute MI. Materials and methods We investigated 934 patients diagnosed with acute STEMI who underwent thrombolysis. ACE I/D polymorphism was detected by polymerase chain reaction and ACE activity was measured in 615 patients. Results The prevalence of DD, ID, and II genotypes in our study group were 41.97%, 34.36%, and 23.66% respectively. The ACE polymorphism was not significantly associated with the type of myocardial infarction, the LV ejection fraction, the number of vessels diseased and patency of the vessel after thrombolysis. The polymorphism had no influence on in hospital mortality (P = 0.453). The ACE activity also showed no influence on in hospital mortality (P = 0.482). The age > 60 years, Male gender, occluded artery and severe LV dysfunction (LVEF < 35%) were predictors of in-hospital mortality on multivariate regression analysis. Conclusion There was no differences among ACE (I/D) polymorphism observed in STEMI population. Neither ACE I/D polymorphism nor ACE activity predicted in-hospital mortality inpatients admitted with acute STEMI.
Collapse
Affiliation(s)
- Nagaraja Moorthy
- Department of Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bangalore 560069, India
| | - Kalpana Saligrama Ramegowda
- Department of Pathology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bangalore 560069, India
| | - Simran Jain
- Department of Pathology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bangalore 560069, India
| | - G Bharath
- Department of Pathology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bangalore 560069, India
| | - Archana Sinha
- Department of Clinical Nutrition, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Manjunath C Nanjappa
- Department of Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bangalore 560069, India
| | - Rita Christopher
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| |
Collapse
|
49
|
Medina-Enríquez MM, Lopez-León S, Carlos-Escalante JA, Aponte-Torres Z, Cuapio A, Wegman-Ostrosky T. ACE2: the molecular doorway to SARS-CoV-2. Cell Biosci 2020; 10:148. [PMID: 33380340 PMCID: PMC7772801 DOI: 10.1186/s13578-020-00519-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022] Open
Abstract
The angiotensin-converting enzyme 2 (ACE2) is the host functional receptor for the new virus SARS-CoV-2 causing Coronavirus Disease 2019. ACE2 is expressed in 72 different cell types. Some factors that can affect the expression of the ACE2 are: sex, environment, comorbidities, medications (e.g. anti-hypertensives) and its interaction with other genes of the renin-angiotensin system and other pathways. Different factors can affect the risk of infection of SARS-CoV-2 and determine the severity of the symptoms. The ACE2 enzyme is a negative regulator of RAS expressed in various organ systems. It is with immunity, inflammation, increased coagulopathy, and cardiovascular disease. In this review, we describe the genetic and molecular functions of the ACE2 receptor and its relation with the physiological and pathological conditions to better understand how this receptor is involved in the pathogenesis of COVID-19. In addition, it reviews the different comorbidities that interact with SARS-CoV-2 in which also ACE2 plays an important role. It also describes the different factors that interact with the virus that have an influence in the expression and functional activities of the receptor. The goal is to provide the reader with an understanding of the complexity and importance of this receptor.
Collapse
Affiliation(s)
| | - Sandra Lopez-León
- Global Drug Development, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | | | | | - Angelica Cuapio
- Center of Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Talia Wegman-Ostrosky
- Department of Basic Research, Instituto Nacional de Cancerología, 22 San Fernando Avenue, Belisario Domínguez Sección XVI, 14080, Mexico City, Mexico.
| |
Collapse
|
50
|
Ramezankhani R, Solhi R, Memarnejadian A, Nami F, Hashemian SMR, Tricot T, Vosough M, Verfaillie C. Therapeutic modalities and novel approaches in regenerative medicine for COVID-19. Int J Antimicrob Agents 2020; 56:106208. [PMID: 33213829 PMCID: PMC7582055 DOI: 10.1016/j.ijantimicag.2020.106208] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/25/2020] [Accepted: 10/17/2020] [Indexed: 02/08/2023]
Abstract
The recent coronavirus disease 2019 outbreak around the world has had an enormous impact on the global health burden, threatening the lives of many individuals, and has had severe socio-economic consequences. Many pharmaceutical and biotechnology companies have commenced intensive research on different therapeutic strategies, from repurposed antiviral drugs to vaccines and monoclonal antibodies to prevent the spread of the disease and treat infected patients. Among the various strategies, advanced therapeutic approaches including cell- and gene-editing-based therapeutics are also being investigated, and initial results in in-vitro and early phase I studies have been promising. However, further assessments are required. This article reviews the underlying mechanisms for the pathogenesis of severe acute respiratory syndrome coronavirus-2, and discusses available therapeutic candidates and advanced modalities that are being evaluated in in-vitro/in-vivo models and are of note in clinical trials.
Collapse
Affiliation(s)
- Roya Ramezankhani
- Department of Applied Cell Sciences, Faculty of Basic Science and Advanced Medical Technologies, Royan Institute, Academic Centre for Education, Culture and Research, Tehran, Iran; Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium; Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, Academic Centre for Education, Culture and Research, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, Academic Centre for Education, Culture and Research, Tehran, Iran
| | - Roya Solhi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, Academic Centre for Education, Culture and Research, Tehran, Iran
| | | | - Fatemeharefeh Nami
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Seyed Mohammad Reza Hashemian
- Chronic Respiratory Diseases Research Centre, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tine Tricot
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, Academic Centre for Education, Culture and Research, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, Academic Centre for Education, Culture and Research, Tehran, Iran.
| | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium.
| |
Collapse
|