1
|
Roytenberg R, Yue H, DeHart A, Kim E, Bai F, Kim Y, Denning K, Kwei A, Zhang Q, Liu J, Zheng XL, Li W. Thymidine phosphorylase mediates SARS-CoV-2 spike protein enhanced thrombosis in K18-hACE2 TG mice. Thromb Res 2024; 244:109195. [PMID: 39442286 DOI: 10.1016/j.thromres.2024.109195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/09/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Thymidine phosphorylase (TYMP), which facilitates platelet activation and thrombosis, is significantly increased in COVID-19 patients. We hypothesize that TYMP mediates SARS-CoV-2 spike protein (SP)-induced thrombosis. MATERIALS AND METHODS Plasmids encoding wildtype SP or empty vector (p3.1) were transfected into COS-7 cells, and cell lysates were prepared as a reservoir for SP or p3.1 (control), respectively. K18-hACE2TG and K18-hACE2TG/Tymp-/- mice were treated with a single dose of SP or p3.1 by intraperitoneal injection and then subjected to thrombosis studies three days later. The role of SP on inflammatory signaling activation was assessed in BEAS-2B cells. RESULTS SARS-CoV-2 SP increased the expression of TYMP, resulting in the activation of STAT3 and NF-κB in BEAS-2B cells. A siRNA-mediated knockdown of TYMP attenuated SP-enhanced activation of STAT3. SP significantly promoted arterial thrombosis in K18-hACE2TG mice. SP-accelerated thrombosis was attenuated by inhibition or genetic ablation of TYMP. SP treatment did not influence ADP- or collagen-induced platelet aggregation but significantly increased platelet adhesion to fibrinogen. SP treatment also significantly shortened activated partial thromboplastin time, which was reversed and even prolonged by TYMP deficiency. Additionally, SP binds to platelet factor 4 (PF4) and TYMP. TYMP does not bind PF4 but enhances the formation of the SP/PF4 complex, which may augment the procoagulant and prothrombotic effect of PF4. CONCLUSIONS We conclude that SP is prothrombotic and upregulates TYMP expression, and TYMP inhibition or knockout mitigates SP-enhanced thrombosis. These findings suggest that inhibition of TYMP may be a novel therapeutic strategy for COVID-19-associated thrombosis.
Collapse
Affiliation(s)
- Renat Roytenberg
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Hong Yue
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Autumn DeHart
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Eugene Kim
- Department of Chemistry, College of Sciences, Marshall University, Huntington, WV 25755, USA
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Yongick Kim
- Department of Chemistry, College of Sciences, Marshall University, Huntington, WV 25755, USA
| | - Krista Denning
- Department of Pathology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Alec Kwei
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Quan Zhang
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - X Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
2
|
Jasiczek J, Doroszko A, Trocha T, Trocha M. Role of the RAAS in mediating the pathophysiology of COVID-19. Pharmacol Rep 2024; 76:475-486. [PMID: 38652364 PMCID: PMC11126499 DOI: 10.1007/s43440-024-00596-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
The renin-angiotensin-aldosterone system (RAAS) holds a position of paramount importance as enzymatic and endocrine homeostatic regulator concerning the water-electrolyte and acid-base balance. Nevertheless, its intricacy is influenced by the presence of various complementary angiotensins and their specific receptors, thereby modifying the primary RAAS actions. Angiotensin-converting enzyme 2 (ACE2) acts as a surface receptor for SARS-CoV-2, establishing an essential connection between RAAS and COVID-19 infection. Despite the recurring exploration of the RAAS impact on the trajectory of COVID-19 along with the successful resolution of many inquiries, its complete role in the genesis of delayed consequences encompassing long COVID and cardiovascular thrombotic outcomes during the post-COVID phase as well as post-vaccination, remains not fully comprehended. Particularly noteworthy is the involvement of the RAAS in the molecular mechanisms underpinning procoagulant processes throughout COVID-19. These processes significantly contribute to the pathogenesis of organ complications as well as determine clinical outcomes and are discussed in this manuscript.
Collapse
Affiliation(s)
- Jakub Jasiczek
- Department of Cardiology, Regional Specialist Hospital in Wrocław, Kamieńskiego 73a, Wrocław, 51-124, Poland
| | - Adrian Doroszko
- Department of Cardiology, 4th Military Hospital, Faculty of Medicine, Wroclaw University of Science and Technology, Weigla 5, Wrocław, 50-981, Poland
| | - Tymoteusz Trocha
- Faculty of Medicine, Wroclaw Medical University, Borowska 213, Wrocław, 50-556, Poland.
| | - Małgorzata Trocha
- Clinical Department of Diabetology and Internal Disease, Wroclaw Medical University, Borowska 213, Wrocław, 50-556, Poland
| |
Collapse
|
3
|
Luo D, Bai M, Zhang W, Wang J. The possible mechanism and research progress of ACE2 involved in cardiovascular injury caused by COVID-19: a review. Front Cardiovasc Med 2024; 11:1409723. [PMID: 38863899 PMCID: PMC11165996 DOI: 10.3389/fcvm.2024.1409723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/09/2024] [Indexed: 06/13/2024] Open
Abstract
ACE2 is the earliest receptor discovered to mediate the entry of SARS-CoV-2. In addition to the receptor, it also participates in complex pathological and physiological processes, including regulating the RAS system, apelin, KKS system, and immune system. In addition to affecting the respiratory system, viral infections also interact with cardiovascular diseases. SARS-CoV-2 can directly invade the cardiovascular system through ACE2; Similarly, cardiovascular diseases such as hypertension and coronary heart disease can affect ACE2 levels and exacerbate the disease, and ACE2 dysregulation may also be a potential mechanism for long-term acute sequelae of COVID-19. Since the SARS CoV-2 epidemic, many large population studies have tried to clarify the current focus of debate, that is, whether we should give COVID-19 patients ACEI and ARB drug treatment, but there is still no conclusive conclusion. We also discussed potential disease treatment options for ACE2 at present. Finally, we discussed the researchers' latest findings on ACE2 and their prospects for future research.
Collapse
Affiliation(s)
| | | | | | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
4
|
Nisa A, Kumar R, Ramasamy S, Kolloli A, Olejnik J, Jalloh S, Gummuluru S, Subbian S, Bushkin Y. Modulations of Homeostatic ACE2, CD147, GRP78 Pathways Correlate with Vascular and Endothelial Performance Markers during Pulmonary SARS-CoV-2 Infection. Cells 2024; 13:432. [PMID: 38474396 PMCID: PMC10930588 DOI: 10.3390/cells13050432] [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: 02/05/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The pathologic consequences of Coronavirus Disease-2019 (COVID-19) include elevated inflammation and dysregulated vascular functions associated with thrombosis. In general, disruption of vascular homeostasis and ensuing prothrombotic events are driven by activated platelets, monocytes, and macrophages, which form aggregates (thrombi) attached to the endothelium lining of vessel walls. However, molecular pathways underpinning the pathological interactions between myeloid cells and endothelium during COVID-19 remain undefined. Here, we tested the hypothesis that modulations in the expression of cellular receptors angiotensin-converting enzyme 2 (ACE2), CD147, and glucose-regulated protein 78 (GRP78), which are involved in homeostasis and endothelial performance, are the hallmark responses induced by SARS-CoV-2 infection. Cultured macrophages and lungs of hamster model systems were used to test this hypothesis. The results indicate that while macrophages and endothelial cells are less likely to support SARS-CoV-2 proliferation, these cells may readily respond to inflammatory stimuli generated by the infected lung epithelium. SARS-CoV-2 induced modulations of tested cellular receptors correlated with corresponding changes in the mRNA expression of coagulation cascade regulators and endothelial integrity components in infected hamster lungs. Among these markers, tissue factor (TF) had the best correlation for prothrombotic events during SARS-CoV-2 infection. Furthermore, the single-molecule fluorescence in situ hybridization (smFISH) method alone was sufficient to determine the peak and resolution phases of SARS-CoV-2 infection and enabled screening for cellular markers co-expressed with the virus. These findings suggest possible molecular pathways for exploration of novel drugs capable of blocking the prothrombotic shift events that exacerbate COVID-19 pathophysiology and control the disease.
Collapse
Affiliation(s)
- Annuurun Nisa
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.N.); (R.K.); (S.R.); (A.K.)
| | - Ranjeet Kumar
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.N.); (R.K.); (S.R.); (A.K.)
| | - Santhamani Ramasamy
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.N.); (R.K.); (S.R.); (A.K.)
| | - Afsal Kolloli
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.N.); (R.K.); (S.R.); (A.K.)
| | - Judith Olejnik
- Department of Virology, Immunology & Microbiology, Boston University School of Medicine, Boston, MA 02130, USA; (J.O.); (S.J.); (S.G.)
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02218, USA
| | - Sallieu Jalloh
- Department of Virology, Immunology & Microbiology, Boston University School of Medicine, Boston, MA 02130, USA; (J.O.); (S.J.); (S.G.)
| | - Suryaram Gummuluru
- Department of Virology, Immunology & Microbiology, Boston University School of Medicine, Boston, MA 02130, USA; (J.O.); (S.J.); (S.G.)
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.N.); (R.K.); (S.R.); (A.K.)
| | - Yuri Bushkin
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA; (A.N.); (R.K.); (S.R.); (A.K.)
| |
Collapse
|
5
|
Kusudo E, Murata Y, Kawamoto S, Egi M. Variant-derived SARS-CoV-2 spike protein does not directly cause platelet activation or hypercoagulability. Clin Exp Med 2023; 23:3701-3708. [PMID: 37208552 PMCID: PMC10198021 DOI: 10.1007/s10238-023-01091-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/11/2023] [Indexed: 05/21/2023]
Abstract
Thrombosis has been associated with severity and mortality in COVID-19. SARS-CoV-2 infects the host via its spike protein. However, direct effects of spike proteins from SARS-CoV-2 variants on platelet activity and coagulability have not been examined. An ethically approved ex vivo study was performed under a preplanned power analysis. Venous blood was collected from 6 healthy subjects who gave prior written consent. The samples were divided into 5 groups: without spike proteins (group N) and with spike proteins derived from alpha, beta, gamma, and delta SARS-CoV-2 variants (groups A, B, C, and D, respectively). Platelet aggregability, P-selectin expression, platelet-associated complement-1 (PAC-1) binding, platelet count, and mean platelet volume (MPV) were measured in all 5 groups, and thromboelastography (TEG) parameters were measured in groups N and D. The % change in each parameter in groups A to D was calculated relative to the value in group N. Data were analyzed by Friedman test, except for TEG parameters, which were evaluated by Wilcoxon matched pairs test. P < 0.05 was considered significant. This study included 6 participants based on a power analysis. There were no significant differences in platelet aggregability under stimulation with adenosine diphosphate 5 µg/ml, collagen 0.2 or 0.5 µg/ml, and Ser-Phe-Leu-Leu-Arg-Asn-amide trifluoroacetate salt (SFLLRN) 0.5 or 1 µM in groups A-D compared to group N. There were also no significant differences in P-selectin expression and PAC-1 binding under basal conditions or SFLLRN stimulation, and no significant differences in platelet count, MPV and TEG parameters. Platelet hyperactivity and blood hypercoagulability have been reported in COVID-19 patients, but spike proteins at 5 µg/ml from SARS-CoV-2 variants (alpha, beta, gamma, delta) did not directly cause these effects in an ex vivo study. This study was approved by the Ethics Committee of Kyoto University Hospital (R0978-1) on March 06, 2020.
Collapse
Affiliation(s)
- Eriko Kusudo
- Department of Anesthesia, Kyoto University Hospital, 54 Shogoin-kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yutaka Murata
- Department of Anesthesia, Kyoto University Hospital, 54 Shogoin-kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Anesthesia, Kitano Hospital, 2-4-20 Ohgimachi, Kita-ku, Osaka, 530-8480, Japan
| | - Shuji Kawamoto
- Department of Anesthesia, Kyoto University Hospital, 54 Shogoin-kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Moritoki Egi
- Department of Anesthesia, Kyoto University Hospital, 54 Shogoin-kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
6
|
Kim D, Jeong W, Kim Y, Lee J, Cho SW, Oh CM, Park R. Pharmacologic Activation of Angiotensin-Converting Enzyme II Alleviates Diabetic Cardiomyopathy in db/db Mice by Reducing Reactive Oxidative Stress. Diabetes Metab J 2023; 47:487-499. [PMID: 37096378 PMCID: PMC10404524 DOI: 10.4093/dmj.2022.0125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/29/2022] [Indexed: 04/26/2023] Open
Abstract
BACKGRUOUND Diabetes mellitus is one of the most common chronic diseases worldwide, and cardiovascular disease is the leading cause of morbidity and mortality in diabetic patients. Diabetic cardiomyopathy (DCM) is a phenomenon characterized by a deterioration in cardiac function and structure, independent of vascular complications. Among many possible causes, the renin-angiotensin-aldosterone system and angiotensin II have been proposed as major drivers of DCM development. In the current study, we aimed to investigate the effects of pharmacological activation of angiotensin-converting enzyme 2 (ACE2) on DCM. METHODS The ACE2 activator diminazene aceturate (DIZE) was administered intraperitoneally to male db/db mice (8 weeks old) for 8 weeks. Transthoracic echocardiography was used to assess cardiac mass and function in mice. Cardiac structure and fibrotic changes were examined using histology and immunohistochemistry. Gene and protein expression levels were examined using quantitative reverse transcription polymerase chain reaction and Western blotting, respectively. Additionally, RNA sequencing was performed to investigate the underlying mechanisms of the effects of DIZE and identify novel potential therapeutic targets for DCM. RESULTS Echocardiography revealed that in DCM, the administration of DIZE significantly improved cardiac function as well as reduced cardiac hypertrophy and fibrosis. Transcriptome analysis revealed that DIZE treatment suppresses oxidative stress and several pathways related to cardiac hypertrophy. CONCLUSION DIZE prevented the diabetes mellitus-mediated structural and functional deterioration of mouse hearts. Our findings suggest that the pharmacological activation of ACE2 could be a novel treatment strategy for DCM.
Collapse
Affiliation(s)
- Donghyun Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Wooju Jeong
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jibeom Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, College of Medicine, Inje University, Goyang, Korea
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University, Busan, Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
7
|
Salehi Z, Motlagh Ghoochani BFN, Hasani Nourian Y, Jamalkandi SA, Ghanei M. The controversial effect of smoking and nicotine in SARS-CoV-2 infection. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2023; 19:49. [PMID: 37264452 PMCID: PMC10234254 DOI: 10.1186/s13223-023-00797-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 04/18/2023] [Indexed: 06/03/2023]
Abstract
The effects of nicotine and cigarette smoke in many diseases, notably COVID-19 infection, are being debated more frequently. The current basic data for COVID-19 is increasing and indicating the higher risk of COVID-19 infections in smokers due to the overexpression of corresponding host receptors to viral entry. However, current multi-national epidemiological reports indicate a lower incidence of COVID-19 disease in smokers. Current data indicates that smokers are more susceptible to some diseases and more protective of some other. Interestingly, nicotine is also reported to play a dual role, being both inflammatory and anti-inflammatory. In the present study, we tried to investigate the effect of pure nicotine on various cells involved in COVID-19 infection. We followed an organ-based systematic approach to decipher the effect of nicotine in damaged organs corresponding to COVID-19 pathogenesis (12 related diseases). Considering that the effects of nicotine and cigarette smoke are different from each other, it is necessary to be careful in generalizing the effects of nicotine and cigarette to each other in the conducted researches. The generalization and the undifferentiation of nicotine from smoke is a significant bias. Moreover, different doses of nicotine stimulate different effects (dose-dependent response). In addition to further assessing the role of nicotine in COVID-19 infection and any other cases, a clever assessment of underlying diseases should also be considered to achieve a guideline for health providers and a personalized approach to treatment.
Collapse
Affiliation(s)
- Zahra Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Yazdan Hasani Nourian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Chen H, Peng J, Wang T, Wen J, Chen S, Huang Y, Zhang Y. Counter-regulatory renin-angiotensin system in hypertension: Review and update in the era of COVID-19 pandemic. Biochem Pharmacol 2023; 208:115370. [PMID: 36481346 PMCID: PMC9721294 DOI: 10.1016/j.bcp.2022.115370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is the major cause of mortality and disability, with hypertension being the most prevalent risk factor. Excessive activation of the renin-angiotensin system (RAS) under pathological conditions, leading to vascular remodeling and inflammation, is closely related to cardiovascular dysfunction. The counter-regulatory axis of the RAS consists of angiotensin-converting enzyme 2 (ACE2), angiotensin (1-7), angiotensin (1-9), alamandine, proto-oncogene Mas receptor, angiotensin II type-2 receptor and Mas-related G protein-coupled receptor member D. Each of these components has been shown to counteract the effects of the overactivated RAS. In this review, we summarize the latest insights into the complexity and interplay of the counter-regulatory RAS axis in hypertension, highlight the pathophysiological functions of ACE2, a multifunctional molecule linking hypertension and COVID-19, and discuss the function and therapeutic potential of targeting this counter-regulatory RAS axis to prevent and treat hypertension in the context of the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Hongyin Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518000, Guangdong, China
| | - Jiangyun Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Jielu Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China,Corresponding authors
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518000, Guangdong, China,Corresponding authors
| |
Collapse
|
9
|
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: 1.0] [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
|
10
|
Association between COVID-19 Diagnosis and Coronary Artery Thrombosis: A Narrative Review. Biomedicines 2022; 10:biomedicines10030702. [PMID: 35327504 PMCID: PMC8945192 DOI: 10.3390/biomedicines10030702] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/13/2022] [Accepted: 03/16/2022] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 is characterized by its severe respiratory effects. Data early on indicated an increased risk of mortality in patients with cardiovascular comorbidities. Early reports highlighted the multisystem inflammatory syndrome, cytokine storm, and thromboembolic events as part of the disease processes. The aim of this review is to assess the association between COVID-19 and its thrombotic complications, specifically related to the cardiovascular system. The role of neutrophil extracellular traps (NETs) is explored in the pathogenesis of the disease. The structure and anatomy of the virus are pivotal to its virulence in comparison to other α and β Coronaviridae (HCoV-229E, HCoV-OC43, HCoV-229E, HCoV-NL63, HCoV-OC43, and HCoV-HKU1). In particular, the host interaction and response may explain the variability of severity in patients. Angio tensin-converting enzyme 2 (ACE2) activation may be implicated in the cardiovascular and throm bogenic potential of the disease. The virus may also have direct effects on the endothelial lining affecting hemostasis and resulting in thrombosis through several mechanisms. Dipyridamole may have a therapeutic benefit in NET suppression. Therapeutic avenues should be concentrated on the different pathophysiological steps involving the virus and the host.
Collapse
|
11
|
García-Escobar A, Vera-Vera S, Jurado-Román A, Jiménez-Valero S, Galeote G, Moreno R. Calcium Signaling Pathway Is Involved in the Shedding of ACE2 Catalytic Ectodomain: New Insights for Clinical and Therapeutic Applications of ACE2 for COVID-19. Biomolecules 2022; 12:biom12010076. [PMID: 35053224 PMCID: PMC8774087 DOI: 10.3390/biom12010076] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 02/04/2023] Open
Abstract
The angiotensin-converting enzyme 2 (ACE2) is a type I integral membrane that exists in two forms: the first is a transmembrane protein; the second is a soluble catalytic ectodomain of ACE2. The catalytic ectodomain of ACE2 undergoes shedding by a disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), in which calmodulin mediates the calcium signaling pathway that is involved in ACE2 release, resulting in a soluble catalytic ectodomain of ACE2 that can be measured as soluble ACE2 plasma activity. The shedding of the ACE2 catalytic ectodomain plays a role in cardiac remodeling and endothelial dysfunction and is a predictor of all-cause mortality, including cardiovascular mortality. Moreover, considerable evidence supports that the ACE2 catalytic ectodomain is an essential entry receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Additionally, endotoxins and the pro-inflammatory cytokines interleukin (IL)-1β and tumor necrosis factor-alpha (TNFα) all enhanced soluble catalytic ectodomain ACE2 shedding from the airway epithelia, suggesting that the shedding of ACE2 may represent a mechanism by which viral entry and infection may be controlled such as some types of betacoronavirus. In this regard, ACE2 plays an important role in inflammation and thrombotic response, and its down-regulation may aggravate COVID-19 via the renin-angiotensin system, including by promoting pathological changes in lung injury. Soluble forms of ACE2 have recently been shown to inhibit SARS-CoV-2 infection. Furthermore, given that vitamin D enhanced the shedding of ACE2, some studies reported that vitamin D treatment is associated with prognosis improvement in COVID-19. This is an updated review on the evidence, clinical, and therapeutic applications of ACE2 for COVID-19.
Collapse
Affiliation(s)
- Artemio García-Escobar
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, 28046 Madrid, Spain; (S.V.-V.); (A.J.-R.); (S.J.-V.); (G.G.); (R.M.)
- Instituto de Investigación Hospital La Paz (IDIPAZ), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-917-27-70-00
| | - Silvio Vera-Vera
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, 28046 Madrid, Spain; (S.V.-V.); (A.J.-R.); (S.J.-V.); (G.G.); (R.M.)
- Instituto de Investigación Hospital La Paz (IDIPAZ), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alfonso Jurado-Román
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, 28046 Madrid, Spain; (S.V.-V.); (A.J.-R.); (S.J.-V.); (G.G.); (R.M.)
- Instituto de Investigación Hospital La Paz (IDIPAZ), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Santiago Jiménez-Valero
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, 28046 Madrid, Spain; (S.V.-V.); (A.J.-R.); (S.J.-V.); (G.G.); (R.M.)
- Instituto de Investigación Hospital La Paz (IDIPAZ), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Guillermo Galeote
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, 28046 Madrid, Spain; (S.V.-V.); (A.J.-R.); (S.J.-V.); (G.G.); (R.M.)
- Instituto de Investigación Hospital La Paz (IDIPAZ), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Raúl Moreno
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, 28046 Madrid, Spain; (S.V.-V.); (A.J.-R.); (S.J.-V.); (G.G.); (R.M.)
- Instituto de Investigación Hospital La Paz (IDIPAZ), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
12
|
Augustine R, S A, Nayeem A, Salam SA, Augustine P, Dan P, Maureira P, Mraiche F, Gentile C, Hansbro PM, McClements L, Hasan A. Increased complications of COVID-19 in people with cardiovascular disease: Role of the renin-angiotensin-aldosterone system (RAAS) dysregulation. Chem Biol Interact 2022; 351:109738. [PMID: 34740598 PMCID: PMC8563522 DOI: 10.1016/j.cbi.2021.109738] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 01/28/2023]
Abstract
The rapid spread of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) that causes coronavirus disease 2019 (COVID-19), has had a dramatic negative impact on public health and economies worldwide. Recent studies on COVID-19 complications and mortality rates suggest that there is a higher prevalence in cardiovascular diseases (CVD) patients. Past investigations on the associations between pre-existing CVDs and susceptibility to coronavirus infections including SARS-CoV and the Middle East Respiratory Syndrome coronavirus (MERS-CoV), have demonstrated similar results. However, the underlying mechanisms are poorly understood. This has impeded adequate risk stratification and treatment strategies for CVD patients with SARS-CoV-2 infections. Generally, dysregulation of the expression of angiotensin-converting enzyme (ACE) and the counter regulator, angiotensin-converting enzyme 2 (ACE2) is a hallmark of cardiovascular risk and CVD. ACE2 is the main host receptor for SARS-CoV-2. Although further studies are required, dysfunction of ACE2 after virus binding and dysregulation of the renin-angiotensin-aldosterone system (RAAS) signaling may worsen the outcomes of people affected by COVID-19 and with preexisting CVD. Here, we review the current knowledge and outline the gaps related to the relationship between CVD and COVID-19 with a focus on the RAAS. Improved understanding of the mechanisms regulating viral entry and the role of RAAS may direct future research with the potential to improve the prevention and management of COVID-19.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| | - Abhilash S
- Department of Microbiology, Majlis Arts and Science College, Puramannur, Malappuram, Kerala, 676552, India
| | - Ajisha Nayeem
- Department of Biotechnology, St. Mary's College, Thrissur, 680020, Kerala, India
| | - Shaheen Abdul Salam
- Department of Biosciences, MES College Marampally, Aluva, Ernakulam, 683107, Kerala, India
| | - Priya Augustine
- Department of Zoology, Kongunadu Arts and Science College, Coimbatore, Tamil Nadu, 641029, India
| | - Pan Dan
- Department of Cardiovascular and Transplantation Surgery, Regional Central Hospital of Nancy, Lorraine University, France; Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Pablo Maureira
- Department of Cardiovascular and Transplantation Surgery, Regional Central Hospital of Nancy, Lorraine University, France
| | - Fatima Mraiche
- College of Pharmacy, QU-Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW, Australia; School of Medicine, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| |
Collapse
|
13
|
Research Progress on Pulmonary Arterial Hypertension and the Role of the Angiotensin Converting Enzyme 2-Angiotensin-(1-7)-Mas Axis in Pulmonary Arterial Hypertension. Cardiovasc Drugs Ther 2022; 36:363-370. [PMID: 33394361 PMCID: PMC7779643 DOI: 10.1007/s10557-020-07114-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2020] [Indexed: 01/31/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease with a complex aetiology and high mortality. Functional and structural changes in the small pulmonary arteries lead to elevated pulmonary arterial pressure, resulting in right heart failure. The pathobiology of PAH is not fully understood, and novel treatment targets in PAH are desperately needed. The renin-angiotensin system is critical for maintaining homeostasis of the cardiovascular system. The system consists of the angiotensin converting enzyme (ACE)-angiotensin (Ang) II-angiotensin type 1 receptor (AT1R) axis and the ACE2-Ang-(1-7)-Mas receptor axis. The former, the ACE-Ang II-AT1R axis, is involved in vasoconstrictive and hypertensive actions along with cardiac and vascular remodelling. The latter, the ACE2-Ang-(1-7)-Mas axis, generally mediates counterbalancing effects against those mediated by the ACE-Ang II-AT1R axis. Based on established functions, the ACE2-Ang-(1-7)-Mas axis may represent a novel target for the treatment of PAH. This review focuses on recent advances in pulmonary circulation science and the role of the ACE2-Ang-(1-7)-Mas axis in PAH.
Collapse
|
14
|
Abstract
It is hypothesized that several comorbidities increase the severity of COVID-19 symptoms. Cardiovascular disease including hypertension was shown to play a critical role in the severity of COVID-19 infection by affecting the survival of patients with COVID-19. Hypertension and the renin-angiotensin-aldosterone system are involved in increasing vascular inflammation and endothelial dysfunction (ED), and both processes are instrumental in COVID-19. Angiotensin-converting enzyme 2 is an essential component of the renin-angiotensin-aldosterone system and the target receptor that mediates SARS-CoV-2 entry to the cell. This led to speculations that major renin-angiotensin-aldosterone system inhibitors, such as angiotensin receptor blockers and angiotensin-converting enzyme inhibitors might affect the course of the disease, since their administration enhances angiotensin-converting enzyme (ACE)2 expression. An increase in ACE2 activity could reduce angiotensin II concentration in the lungs and mitigate virus-driven lung injury. This could also be associated with a reduction in blood coagulation, which plays a critical role in the pathogenesis of SARS-CoV-2; of note, COVID-19 is now regarded as a disorder of blood clotting. Therefore, there is an urgent need to better understand the effect of targeting ACE2 as a potential treatment for SARS-CoV-2 driven injury, and in alleviating COVID-19 symptoms by reversing SARS-CoV-2-induced excessive coagulation and fatalities. Ongoing therapeutic strategies that include recombinant human ACE2 and anti-spike monoclonal antibodies are essential for future clinical practice in order to better understand the effect of targeting ED in COVID-19.
Collapse
Affiliation(s)
- Jalil Daher
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli PO Box 100, Lebanon
| |
Collapse
|
15
|
Gallo CG, Fiorino S, Posabella G, Antonacci D, Tropeano A, Pausini E, Pausini C, Guarniero T, Hong W, Giampieri E, Corazza I, Federico L, de Biase D, Zippi M, Zancanaro M. COVID-19, what could sepsis, severe acute pancreatitis, gender differences, and aging teach us? Cytokine 2021; 148:155628. [PMID: 34411989 PMCID: PMC8343368 DOI: 10.1016/j.cyto.2021.155628] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/02/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a potentially life-threatening disease, defined as Coronavirus Disease 19 (COVID-19). The most common signs and symptoms of this pathological condition include cough, fever, shortness of breath, and sudden onset of anosmia, ageusia, or dysgeusia. The course of COVID-19 is mild or moderate in more than 80% of cases, but it is severe or critical in about 14% and 5% of infected subjects respectively, with a significant risk of mortality. SARS-CoV-2 related infection is characterized by some pathogenetic events, resembling those detectable in other pathological conditions, such as sepsis and severe acute pancreatitis. All these syndromes are characterized by some similar features, including the coexistence of an exuberant inflammatory- as well as an anti-inflammatory-response with immune depression. Based on current knowledge concerning the onset and the development of acute pancreatitis and sepsis, we have considered these syndromes as a very interesting paradigm for improving our understanding of pathogenetic events detectable in patients with COVID-19. The aim of our review is: 1)to examine the pathogenetic mechanisms acting during the emergence of inflammatory and anti-inflammatory processes in human pathology; 2)to examine inflammatory and anti-inflammatory events in sepsis, acute pancreatitis, and SARS-CoV-2 infection and clinical manifestations detectable in patients suffering from these syndromes also according to the age and gender of these individuals; as well as to analyze the possible common and different features among these pathological conditions; 3)to obtain insights into our knowledge concerning COVID-19 pathogenesis. This approach may improve the management of patients suffering from this disease and it may suggest more effective diagnostic approaches and schedules of therapy, depending on the different phases and/or on the severity of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Claudio G Gallo
- Emilian Physiolaser Therapy Center, Castel S. Pietro Terme, Bologna, Italy.
| | - Sirio Fiorino
- Internal Medicine Unit, Budrio Hospital Azienda USL, Bologna, Italy
| | | | - Donato Antonacci
- Medical Science Department, "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo (FG), Italy
| | | | | | | | | | - Wandong Hong
- Department of Gastroenterology and Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang, The People's Republic of China
| | - Enrico Giampieri
- Experimental, Diagnostic and Specialty Medicine Department, University of Bologna, Bologna, Italy
| | - Ivan Corazza
- Experimental, Diagnostic and Specialty Medicine Department, University of Bologna, Bologna, Italy
| | - Lari Federico
- Internal Medicine Unit, Budrio Hospital Azienda USL, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Maddalena Zippi
- Unit of Gastroenterology and Digestive Endoscopy, Sandro Pertini Hospital, Rome, Italy
| | | |
Collapse
|
16
|
Satta S, Lai A, Cavallero S, Williamson C, Chen J, Blázquez‐Medela AM, Roustaei M, Dillon BJ, Ashammakhi N, Carlo DD, Li Z, Sun R, Hsiai TK. Rapid Detection and Inhibition of SARS-CoV-2-Spike Mutation-Mediated Microthrombosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2103266. [PMID: 34687279 PMCID: PMC8646611 DOI: 10.1002/advs.202103266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/21/2021] [Indexed: 05/26/2023]
Abstract
Activation of endothelial cells following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is thought to be the primary driver for the increasingly recognized thrombotic complications in coronavirus disease 2019 patients, potentially due to the SARS-CoV-2 Spike protein binding to the human angiotensin-converting enzyme 2 (hACE2). Vaccination therapies use the same Spike sequence or protein to boost host immune response as a protective mechanism against SARS-CoV-2 infection. As a result, cases of thrombotic events are reported following vaccination. Although vaccines are generally considered safe, due to genetic heterogeneity, age, or the presence of comorbidities in the population worldwide, the prediction of severe adverse outcome in patients remains a challenge. To elucidate Spike proteins underlying patient-specific-vascular thrombosis, the human microcirculation environment is recapitulated using a novel microfluidic platform coated with human endothelial cells and exposed to patient specific whole blood. Here, the blood coagulation effect is tested after exposure to Spike protein in nanoparticles and Spike variant D614G in viral vectors and the results are corroborated using live SARS-CoV-2. Of note, two potential strategies are also examined to reduce blood clot formation, by using nanoliposome-hACE2 and anti-Interleukin (IL) 6 antibodies.
Collapse
Affiliation(s)
- Sandro Satta
- Division of CardiologyDepartment of MedicineDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA90095USA
| | - Angela Lai
- Division of CardiologyDepartment of MedicineDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA90095USA
| | - Susana Cavallero
- Division of CardiologyDepartment of MedicineDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA90095USA
- Department of MedicineVeterans Affairs Greater Los Angeles Healthcare SystemLos AngelesCA90073USA
| | - Cayden Williamson
- Department of BioengineeringHenry Samueli School of Engineering & Applied ScienceUniversity of CaliforniaLos AngelesCA90095USA
| | - Justin Chen
- Department of BioengineeringHenry Samueli School of Engineering & Applied ScienceUniversity of CaliforniaLos AngelesCA90095USA
| | - Ana M. Blázquez‐Medela
- Division of CardiologyDepartment of MedicineDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA90095USA
| | - Mehrdad Roustaei
- Department of BioengineeringHenry Samueli School of Engineering & Applied ScienceUniversity of CaliforniaLos AngelesCA90095USA
| | - Barbara J. Dillon
- Division of CardiologyDepartment of MedicineDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA90095USA
| | - Nureddin Ashammakhi
- Department of BioengineeringHenry Samueli School of Engineering & Applied ScienceUniversity of CaliforniaLos AngelesCA90095USA
| | - Dino Di Carlo
- Department of BioengineeringHenry Samueli School of Engineering & Applied ScienceUniversity of CaliforniaLos AngelesCA90095USA
| | - Zhaoping Li
- Department of MedicineVeterans Affairs Greater Los Angeles Healthcare SystemLos AngelesCA90073USA
- Division of Clinical NutritionDepartment of MedicineDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA90095USA
| | - Ren Sun
- Department of Molecular and Medical PharmacologyDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA90095USA
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Tzung K. Hsiai
- Division of CardiologyDepartment of MedicineDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA90095USA
- Department of MedicineVeterans Affairs Greater Los Angeles Healthcare SystemLos AngelesCA90073USA
- Department of BioengineeringHenry Samueli School of Engineering & Applied ScienceUniversity of CaliforniaLos AngelesCA90095USA
| |
Collapse
|
17
|
Rajtik T, Galis P, Bartosova L, Paulis L, Goncalvesova E, Klimas J. Alternative RAS in Various Hypoxic Conditions: From Myocardial Infarction to COVID-19. Int J Mol Sci 2021; 22:ijms222312800. [PMID: 34884604 PMCID: PMC8657827 DOI: 10.3390/ijms222312800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 12/28/2022] Open
Abstract
Alternative branches of the classical renin–angiotensin–aldosterone system (RAS) represent an important cascade in which angiotensin 2 (AngII) undergoes cleavage via the action of the angiotensin-converting enzyme 2 (ACE2) with subsequent production of Ang(1-7) and other related metabolites eliciting its effects via Mas receptor activation. Generally, this branch of the RAS system is described as its non-canonical alternative arm with counterbalancing actions to the classical RAS, conveying vasodilation, anti-inflammatory, anti-remodeling and anti-proliferative effects. The implication of this branch was proposed for many different diseases, ranging from acute cardiovascular conditions, through chronic respiratory diseases to cancer, nonetheless, hypoxia is one of the most prominent common factors discussed in conjugation with the changes in the activity of alternative RAS branches. The aim of this review is to bring complex insights into the mechanisms behind the various forms of hypoxic insults on the activity of alternative RAS branches based on the different duration of stimuli and causes (acute vs. intermittent vs. chronic), localization and tissue (heart vs. vessels vs. lungs) and clinical relevance of studied phenomenon (experimental vs. clinical condition). Moreover, we provide novel insights into the future strategies utilizing the alternative RAS as a diagnostic tool as well as a promising pharmacological target in serious hypoxia-associated cardiovascular and cardiopulmonary diseases.
Collapse
Affiliation(s)
- Tomas Rajtik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
- Correspondence: ; Tel.: +42-12-501-17-391
| | - Peter Galis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| | - Linda Bartosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| | - Ludovit Paulis
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia;
| | - Eva Goncalvesova
- Department of Heart Failure, Clinic of Cardiology, National Institute of Cardiovascular Diseases, 831 01 Bratislava, Slovakia;
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia; (P.G.); (L.B.); (J.K.)
| |
Collapse
|
18
|
Ionescu M, Stoian AP, Rizzo M, Serban D, Nuzzo D, Mazilu L, Suceveanu AI, Dascalu AM, Parepa IR. The Role of Endothelium in COVID-19. Int J Mol Sci 2021; 22:11920. [PMID: 34769350 PMCID: PMC8584762 DOI: 10.3390/ijms222111920] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 01/08/2023] Open
Abstract
The 2019 novel coronavirus, known as severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) or coronavirus disease 2019 (COVID-19), is causing a global pandemic. The virus primarily affects the upper and lower respiratory tracts and raises the risk of a variety of non-pulmonary consequences, the most severe and possibly fatal of which are cardiovascular problems. Data show that almost one-third of the patients with a moderate or severe form of COVID-19 had preexisting cardiovascular comorbidities such as diabetes mellitus, obesity, hypertension, heart failure, or coronary artery disease. SARS-CoV2 causes hyper inflammation, hypoxia, apoptosis, and a renin-angiotensin system imbalance in a variety of cell types, primarily endothelial cells. Profound endothelial dysfunction associated with COVID-19 can be the cause of impaired organ perfusion that may generate acute myocardial injury, renal failure, and a procoagulant state resulting in thromboembolic events. We discuss the most recent results on the involvement of endothelial dysfunction in the pathogenesis of COVID-19 in patients with cardiometabolic diseases in this review. We also provide insights on treatments that may reduce the severity of this viral infection.
Collapse
Affiliation(s)
- Mihaela Ionescu
- Cardiology Department, Faculty of Medicine, Ovidius University of Constanţa, 900527 Constanţa, Romania; (M.I.); (I.R.P.)
| | - Anca Pantea Stoian
- Diabetes, Nutrition, and Metabolic Diseases Department, Faculty of Medicine, Carol Davila University, 050474 Bucharest, Romania; (A.P.S.); (M.R.)
| | - Manfredi Rizzo
- Diabetes, Nutrition, and Metabolic Diseases Department, Faculty of Medicine, Carol Davila University, 050474 Bucharest, Romania; (A.P.S.); (M.R.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90100 Palermo, Italy
| | - Dragos Serban
- Forth Surgery Department, Emergency University Hospital Bucharest and Faculty of Medicine, Carol Davila University, 050474 Bucharest, Romania;
| | - Domenico Nuzzo
- Italian National Research Council, Institute for Research and Biomedical Innovation (CNR-IRIB), 90100 Palermo, Italy
| | - Laura Mazilu
- Oncology Department, Faculty of Medicine, Ovidius University of Constanţa, 900527 Constanţa, Romania;
| | - Andra Iulia Suceveanu
- Internal Medicine Department, Faculty of Medicine, Ovidius University of Constanţa, 900527 Constanţa, Romania;
| | - Ana Maria Dascalu
- Department of Ophthalmology, Emergency University Hospital Bucharest and Faculty of Medicine, Carol Davila University, 050474 Bucharest, Romania;
| | - Irinel Raluca Parepa
- Cardiology Department, Faculty of Medicine, Ovidius University of Constanţa, 900527 Constanţa, Romania; (M.I.); (I.R.P.)
| |
Collapse
|
19
|
Ahmad I, Pawara R, Surana S, Patel H. The Repurposed ACE2 Inhibitors: SARS-CoV-2 Entry Blockers of Covid-19. Top Curr Chem (Cham) 2021; 379:40. [PMID: 34623536 PMCID: PMC8498772 DOI: 10.1007/s41061-021-00353-7] [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] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 09/14/2021] [Indexed: 12/17/2022]
Abstract
The highly infectious disease COVID-19 is induced by SARS-coronavirus 2 (SARS-CoV-2), which has spread rapidly around the globe and was announced as a pandemic by the World Health Organization (WHO) in March 2020. SARS-CoV-2 binds to the host cell's angiotensin converting enzyme 2 (ACE2) receptor through the viral surface spike glycoprotein (S-protein). ACE2 is expressed in the oral mucosa and can therefore constitute an essential route for entry of SARS-CoV-2 into hosts through the tongue and lung epithelial cells. At present, no effective treatments for SARS-CoV-2 are yet in place. Blocking entry of the virus by inhibiting ACE2 is more advantageous than inhibiting the subsequent stages of the SARS-CoV-2 life cycle. Based on current published evidence, we have summarized the different in silico based studies and repurposing of anti-viral drugs to target ACE2, SARS-CoV-2 S-Protein: ACE2 and SARS-CoV-2 S-RBD: ACE2. This review will be useful to researchers looking to effectively recognize and deal with SARS-CoV-2, and in the development of repurposed ACE2 inhibitors against COVID-19.
Collapse
Affiliation(s)
- Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur (Dhule), Maharashtra, 425405, India
| | - Rahul Pawara
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur (Dhule), Maharashtra, 425405, India
| | - Sanjay Surana
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur (Dhule), Maharashtra, 425405, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur (Dhule), Maharashtra, 425405, India.
| |
Collapse
|
20
|
Wang R, Xu J, Wu J, Gao S, Wang Z. Angiotensin-converting enzyme 2 alleviates pulmonary artery hypertension through inhibition of focal adhesion kinase expression. Exp Ther Med 2021; 22:1165. [PMID: 34504610 PMCID: PMC8393266 DOI: 10.3892/etm.2021.10599] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
Focal adhesion kinase (FAK) is an important therapeutic target in pulmonary artery hypertension (PAH); however, the mechanism of its activation remains unknown. The present study aimed to investigate whether angiotensin-converting enzyme 2 (ACE2) could regulate FAK and alleviate PAH in a rat model of PAH established with a single administration of monocrotaline followed by continuous hypoxia treatment. In the current study, right ventricular pressure, body weight and the right ventricular hypertrophy index were measured, and hematoxylin-eosin staining was performed on lung tissues to determine whether the modeling was successful. Changes in the serum levels of FAK were measured using an ELISA kit to evaluate the association between ACE2 and FAK. The mRNA expression levels of ACE2, FAK, caspase-3 and survivin were determined using reverse transcription-quantitative PCR (RT-qPCR). The protein expression levels of ACE2, phosphorylated FAK/FAK, cleaved caspase-3/pro-caspase-3 and survivin were determined via western blotting. Immunohistochemistry was applied to detect the expression of FAK around the pulmonary arterioles. Apoptosis of smooth muscle cells around pulmonary arterioles was observed by TUNEL staining. After treatment with the ACE2 activator DIZE or inhibitor DX-600, the results demonstrated that ACE2 reduced PAH-induced changes in arteriole morphology compared with the control. It also inhibited FAK expression in serum. WB and RT-qPCR results suggested that ACE2 inhibited the expression of FAK and pathway-related proteins, and promoted caspase-3 expression. Additionally, ACE2 reduced FAK expression around the pulmonary arterioles and promoted smooth muscle cell apoptosis. The results indicated that ACE2 activation inhibited FAK expression, leading to alleviation of the symptoms of PAH.
Collapse
Affiliation(s)
- Rui Wang
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China.,Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jingjing Xu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jinbo Wu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Shunheng Gao
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Zhiping Wang
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China.,Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| |
Collapse
|
21
|
Which ones, when and why should renin-angiotensin system inhibitors work against COVID-19? Adv Biol Regul 2021; 81:100820. [PMID: 34419773 PMCID: PMC8359569 DOI: 10.1016/j.jbior.2021.100820] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 12/15/2022]
Abstract
The article describes the possible pathophysiological origin of COVID-19 and the crucial role of renin-angiotensin system (RAS), providing several “converging” evidence in support of this hypothesis. SARS-CoV-2 has been shown to initially upregulate ACE2 systemic activity (early phase), which can subsequently induce compensatory responses leading to upregulation of both arms of the RAS (late phase) and consequently to critical, advanced and untreatable stages of COVID-19 disease. The main and initial actors of the process are ACE2 and ADAM17 zinc-metalloproteases, which, initially triggered by SARS-CoV-2 spike proteins, work together in increasing circulating Ang 1–7 and Ang 1–9 peptides and downstream (Mas and Angiotensin type 2 receptors) pathways with anti-inflammatory, hypotensive and antithrombotic activities. During the late phase of severe COVID-19, compensatory secretion of renin and ACE enzymes are subsequently upregulated, leading to inflammation, hypertension and thrombosis, which further sustain ACE2 and ADAM17 upregulation. Based on this hypothesis, COVID-19-phase-specific inhibition of different RAS enzymes is proposed as a pharmacological strategy against COVID-19 and vaccine-induced adverse effects. The aim is to prevent the establishment of positive feedback-loops, which can sustain hyperactivity of both arms of the RAS independently of viral trigger and, in some cases, may lead to Long-COVID syndrome.
Collapse
|
22
|
Ghasemi M, Umeton RP, Keyhanian K, Mohit B, Rahimian N, Eshaghhosseiny N, Davoudi V. SARS-CoV-2 and Acute Cerebrovascular Events: An Overview. J Clin Med 2021; 10:3349. [PMID: 34362133 PMCID: PMC8348889 DOI: 10.3390/jcm10153349] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Since the coronavirus disease 2019 (COVID-19) pandemic, due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, accumulating evidence indicates that SARS-CoV-2 infection may be associated with various neurological manifestations, including acute cerebrovascular events (i.e., stroke and cerebral venous thrombosis). These events can occur prior to, during and even after the onset of COVID-19's general symptoms. Although the mechanisms underlying the cerebrovascular complications in patients with COVID-19 are yet to be fully elucidated, the hypercoagulability state, inflammation and altered angiotensin-converting enzyme 2 (ACE-2) signaling in association with SARS-CoV-2 may play key roles. ACE-2 plays a critical role in preserving heart and brain homeostasis. In this review, we discuss the current state of knowledge of the possible mechanisms underlying the acute cerebrovascular events in patients with COVID-19, and we review the current epidemiological studies and case reports of neurovascular complications in association with SARS-CoV-2, as well as the relevant therapeutic approaches that have been considered worldwide. As the number of published COVID-19 cases with cerebrovascular events is growing, prospective studies would help gather more valuable insights into the pathophysiology of cerebrovascular events, effective therapies, and the factors predicting poor functional outcomes related to such events in COVID-19 patients.
Collapse
Affiliation(s)
- Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA; (R.P.U.); (K.K.)
| | - Raffaella Pizzolato Umeton
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA; (R.P.U.); (K.K.)
| | - Kiandokht Keyhanian
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA; (R.P.U.); (K.K.)
| | - Babak Mohit
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Sleep Disorders Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Nasrin Rahimian
- Department of Neurology, Tehran University of Medical Sciences, Tehran 1417613151, Iran;
| | - Niloofarsadaat Eshaghhosseiny
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Vahid Davoudi
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
23
|
Marwah V, Peter DK, Malik V, Mishra SC, Kumar TA, Kumar A, Bhati G, Kumar N, Singh S, Choudhary R. Pulmonary embolism in coronavirus disease 2019: the silent killer. Med J Armed Forces India 2021; 77:S312-S318. [PMID: 34334899 PMCID: PMC8313079 DOI: 10.1016/j.mjafi.2021.03.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/27/2021] [Indexed: 01/08/2023] Open
Abstract
Background Pulmonary embolism (PE) has been identified as one of the deadliest complications of coronavirus disease 2019 (COVID-19), especially in patients admitted to the intensive care unit (ICU). Western literature reminds us of the high prevalence of PE in COVID. Here, we report a series of 13 cases of PE diagnosed and managed at our hospital. Methods Retrospective analysis of medical records of 13 cases of PE admitted at our hospital from February 1, 2020, to September 31, 2020, were done. Their clinical, laboratory, and radiologic data were assessed in detail. Results Computed tomography pulmonary arteriography was used to make the diagnosis in eight patients (61.53%), and clinical findings with corroborative ultrasound and laboratory parameters were used to label PE in five patients (38.46%). Five patients were hemodynamically unstable, requiring thrombolysis with recombinant tissue plasminogen activator, and four patients (30.76%) suffered a fatal outcome. Conclusion COVID-19 is a highly prothrombotic state, and all physicians should keep a high vigilance for PE. All hospitalized patients with COVID-19, especially those admitted in ICU, should be on prophylactic anticoagulation and, if there is any worsening, should be started on therapeutic regimen. Patients at the time of discharge should be switched to oral anticoagulation, which should be continued for at least 3–6 months.
Collapse
Affiliation(s)
- Vikas Marwah
- Professor & Head (Pulmonary, Critical Care & Sleep Medicine), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Deepu K Peter
- Graded Specialist (Pulmonary, Critical Care & Sleep Medicine), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Virender Malik
- Associate Professor (Radiology), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Satish Chandra Mishra
- Consultant & Head (Cardiology), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Tentu Ajai Kumar
- Associate Professor (Pulmonary, Critical Care & Sleep Medicine), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Arvind Kumar
- Resident (Paediatric Pulmonology & Intensive Care), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Gaurav Bhati
- Assistant Professor (Pulmonary, Critical Care & Sleep Medicine), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Nikhil Kumar
- Resident (Pulmonary, Critical Care & Sleep Medicine), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| | - Shalendra Singh
- Assistant Professor & Neuroanaesthesiologist, Department of Anaesthesia, Armed Forces Medical College, Pune, India
| | - Robin Choudhary
- Resident (Pulmonary, Critical Care & Sleep Medicine), Army Institute of Cardiothoracic Sciences (AICTS), Pune, India
| |
Collapse
|
24
|
Cao Y, Geng C, Li Y, Zhang Y. In situ Pulmonary Artery Thrombosis: A Previously Overlooked Disease. Front Pharmacol 2021; 12:671589. [PMID: 34305592 PMCID: PMC8296465 DOI: 10.3389/fphar.2021.671589] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/28/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary thromboembolism (PTE) is the third leading cause of death in cardiovascular diseases. PTE is believed to be caused by thrombi detached from deep veins of lower extremities. The thrombi travel with systemic circulation to the lung and block pulmonary arteries, leading to sudden disruption of hemodynamics and blood gas exchange. However, this concept has recently been challenged by accumulating evidence demonstrating that de novo thrombosis may be formed in pulmonary arteries without deep venous thrombosis. On the other hand, chronic thromboembolic pulmonary hypertension (CTEPH), a subtype of pulmonary hypertension, could have different pathogenesis than traditional PTE. Therefore, this article summarized and compared the risk factors, the common and specific pathogenic mechanisms underlying PTE, in situ pulmonary artery thrombosis, and CTEPH at molecular and cellular levels, and suggested the therapeutic strategies to these diseases, aiming to facilitate understanding of pathogenesis, differential diagnosis, and precision therapeutics of the three pulmonary artery thrombotic diseases.
Collapse
Affiliation(s)
- Yunshan Cao
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, China
| | - Chao Geng
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Yahong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Yan Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| |
Collapse
|
25
|
de Leeuw AJM, Oude Luttikhuis MAM, Wellen AC, Müller C, Calkhoven CF. Obesity and its impact on COVID-19. J Mol Med (Berl) 2021; 99:899-915. [PMID: 33824998 PMCID: PMC8023779 DOI: 10.1007/s00109-021-02072-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/22/2021] [Accepted: 03/30/2021] [Indexed: 01/08/2023]
Abstract
The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic has proven a challenge to healthcare systems since its first appearance in late 2019. The global spread and devastating effects of coronavirus disease 2019 (COVID-19) on patients have resulted in countless studies on risk factors and disease progression. Overweight and obesity emerged as one of the major risk factors for developing severe COVID-19. Here we review the biology of coronavirus infections in relation to obesity. In particular, we review literature about the impact of adiposity-related systemic inflammation on the COVID-19 disease severity, involving cytokine, chemokine, leptin, and growth hormone signaling, and we discuss the involvement of hyperactivation of the renin-angiotensin-aldosterone system (RAAS). Due to the sheer number of publications on COVID-19, we cannot be completed, and therefore, we apologize for all the publications that we do not cite.
Collapse
Affiliation(s)
- Angélica J M de Leeuw
- University Medical Center Groningen (UMCG), University of Groningen, 9700, AD, Groningen, The Netherlands
| | | | - Annemarijn C Wellen
- University Medical Center Groningen (UMCG), University of Groningen, 9700, AD, Groningen, The Netherlands
| | - Christine Müller
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700, AD, Groningen, The Netherlands
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700, AD, Groningen, The Netherlands.
| |
Collapse
|
26
|
Gando S, Wada T. Thromboplasminflammation in COVID-19 Coagulopathy: Three Viewpoints for Diagnostic and Therapeutic Strategies. Front Immunol 2021; 12:649122. [PMID: 34177896 PMCID: PMC8226122 DOI: 10.3389/fimmu.2021.649122] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 05/28/2021] [Indexed: 01/08/2023] Open
Abstract
Thromboplasminflammation in coronavirus disease 2019 (COVID-19) coagulopathy consists of angiotensin II (Ang II)-induced coagulopathy, activated factor XII (FXIIa)- and kallikrein, kinin system-enhanced fibrinolysis, and disseminated intravascular coagulation (DIC). All three conditions induce systemic inflammation via each pathomechanism-developed production of inflammatory cytokines. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) downregulates angiotensin-converting enzyme 2, leading to an increase in Ang II levels. Ang II-induced coagulopathy comprising platelet activation, thrombin generation, plasminogen activator inhibitor-1 expression and endothelial injury causes thrombosis via the angiotensin II type 1 receptor. SARS-CoV-2 RNA and neutrophil extracellular trap (NET) DNA activate FXII, resulting in plasmin generation through FXIIa- and kallikrein-mediated plasminogen conversion to plasmin and bradykinin-induced tissue-type plasminogen activator release from the endothelium via the kinin B2 receptor. NETs induce immunothrombosis at the site of infection (lungs), through histone- and DNA-mediated thrombin generation, insufficient anticoagulation control, and inhibition of fibrinolysis. However, if the infection is sufficiently severe, immunothrombosis disseminates into the systemic circulation, and DIC, which is associated with the endothelial injury, occurs. Inflammation, and serine protease networks of coagulation and fibrinolysis, militate each other through complement pathways, which exacerbates three pathologies of COVID-19 coagulopathy. COVID-19 coagulopathy causes microvascular thrombosis and bleeding, resulting in multiple organ dysfunction and death in critically ill patients. Treatment targets for improving the prognosis of COVID-19 coagulopathy include thrombin, plasmin, and inflammation, and SARS-CoV-2 infection. Several drugs are candidates for controlling these conditions; however, further advances are required to establish robust treatments based on a clear understanding of molecular mechanisms of COVID-19 coagulopathy.
Collapse
Affiliation(s)
- Satoshi Gando
- Acute and Critical Center, Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan.,Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|
27
|
Beura SK, Panigrahi AR, Yadav P, Singh SK. Phytochemicals as Potential Therapeutics for SARS-CoV-2-Induced Cardiovascular Complications: Thrombosis and Platelet Perspective. Front Pharmacol 2021; 12:658273. [PMID: 33981235 PMCID: PMC8107428 DOI: 10.3389/fphar.2021.658273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
After gaining entry through ACE2 aided by TMPRSS2, the SARS-CoV-2 causes serious complications of the cardiovascular system leading to myocarditis and other myocardial injuries apart from causing lung, kidney and brain dysfunctions. Here in this review, we are going to divulge the cellular and immunological mechanisms behind the cardiovascular, thrombotic and platelet impairments that are caused in COVID-19. In addition, we also propose the significance of various anti-platelet and anti-thrombotic phytochemicals in the treatment of COVID-19. The virus induces many immune-modulatory cytokines and chemokines which help in the intravascular coagulation and create a pro-thrombotic environment along with pulmonary embolism and thrombocytopenia. Different types of innate and adaptive immune cells and their granular contents regulate the pathophysiology of SARS-CoV-2 induced endothelial and platelet dysfunctions which correlate the involvement of platelets with myocardial injury and intravascular thrombi directly or indirectly. Hence, by exploiting the natural bioactive compounds from medicinal plants and inhibiting the platelet mediated thrombus formation can be beneficial for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Samir K Beura
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Abhishek R Panigrahi
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Pooja Yadav
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Sunil K Singh
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
28
|
Kincaid KJ, Simpkins AN. Failure of Anticoagulation to Prevent Stroke in Context of Lupus-Associated Anti-Phospholipid Syndrome and Mild COVID-19. J Stroke Cerebrovasc Dis 2021; 30:105817. [PMID: 33933349 PMCID: PMC8041145 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105817] [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: 01/31/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 10/29/2022] Open
Abstract
Hypercoagulability and virally-mediated vascular inflammation have become well-recognized features of the SARS-CoV-2 virus infection, COVID-19. Of growing concern is the apparent ineffectiveness of therapeutic anticoagulation in preventing thromboembolic events among some at-risk patient subtypes with COVID-19. We present a 43-year-old female with a history of seropositive-antiphospholipid syndrome and systemic lupus erythematosus who developed an acute ischemic stroke in the setting of mild COVID-19 infection despite adherence to chronic systemic anticoagulation. The clinical significance of SARS-CoV-2-mediated endothelial cell dysfunction and its potential to cause macrovascular events in spite of full anticoagulation warrants further investigation and likely represents another disease-defining pathology of COVID-19.
Collapse
Affiliation(s)
- Keith J Kincaid
- Department of Neurology, University of Florida, Gainesville, USA.
| | | |
Collapse
|
29
|
Rodríguez C, Luque N, Blanco I, Sebastian L, Barberà JA, Peinado VI, Tura-Ceide O. Pulmonary Endothelial Dysfunction and Thrombotic Complications in Patients with COVID-19. Am J Respir Cell Mol Biol 2021; 64:407-415. [PMID: 33180562 PMCID: PMC8008805 DOI: 10.1165/rcmb.2020-0359ps] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a new strain of a Coronaviridae virus that presents 79% genetic similarity to the severe acute respiratory syndrome coronavirus, has been recently recognized as the cause of a global pandemic by the World Health Organization, implying a major threat to world public health. SARS-CoV-2 infects host human cells by binding through the viral spike proteins to the ACE-2 (angiotensin-converting enzyme 2) receptor, fuses with the cell membrane, enters, and starts its replication process to multiply its viral load. Coronavirus disease (COVID-19) was initially considered a respiratory infection that could cause pneumonia. However, in severe cases, it extends beyond the respiratory system and becomes a multiorgan disease. This transition from localized respiratory infection to multiorgan disease is due to two main complications of COVID-19. On the one hand, it is due to the so-called cytokine storm: an uncontrolled inflammatory reaction of the immune system in which defensive molecules become aggressive for the body itself. On the other hand, it is due to the formation of a large number of thrombi that can cause myocardial infarction, stroke, and pulmonary embolism. The pulmonary endothelium actively participates in these two processes, becoming the last barrier before the virus spreads throughout the body. In this review, we examine the role of the pulmonary endothelium in response to COVID-19, the existence of potential biomarkers, and the development of novel therapies to restore vascular homeostasis and to protect and/or treat coagulation, thrombosis patients. In addition, we review the thrombotic complications recently observed in patients with COVID-19 and its potential threatening sequelae.
Collapse
Affiliation(s)
- Cristina Rodríguez
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute, Girona, Spain.,Department of Pulmonary Medicine, Hospital Clínic-Biomedical Research Institute August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; and
| | - Neus Luque
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute, Girona, Spain
| | - Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic-Biomedical Research Institute August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; and.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Laura Sebastian
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute, Girona, Spain
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic-Biomedical Research Institute August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; and.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Víctor I Peinado
- Department of Pulmonary Medicine, Hospital Clínic-Biomedical Research Institute August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; and.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute, Girona, Spain.,Department of Pulmonary Medicine, Hospital Clínic-Biomedical Research Institute August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; and.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| |
Collapse
|
30
|
Wang H, Saha AK, Sun X, Kon ND, Ferrario CM, Groban L. Atrial appendage angiotensin-converting enzyme-2, aging and cardiac surgical patients: a platform for understanding aging-related coronavirus disease-2019 vulnerabilities. Curr Opin Anaesthesiol 2021; 34:187-198. [PMID: 33606395 PMCID: PMC8249166 DOI: 10.1097/aco.0000000000000965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Hospitalizations for COVID-19 dramatically increase with age. This is likely because of increases in fragility across biological repair systems and a weakened immune system, including loss of the cardiorenal protective arm of the renin--angiotensin system (RAS), composed of angiotensin-converting enzyme-2 (ACE2)/angiotensin-(1--7) [Ang-(1--7)] and its actions through the Mas receptor. The purpose of this review is to explore how cardiac ACE2 changes with age, cardiac diseases, comorbid conditions and pharmaceutical regimens in order to shed light on a potential hormonal unbalance facilitating SARs-CoV-2 vulnerabilities in older adults. RECENT FINDINGS Increased ACE2 gene expression has been reported in human hearts with myocardial infarction, cardiac remodeling and heart failure. We also found ACE2 mRNA in atrial appendage tissue from cardiac surgical patients to be positively associated with age, elevated by certain comorbid conditions (e.g. COPD and previous stroke) and increased in conjunction with patients' chronic use of antithrombotic agents and thiazide diuretics but not drugs that block the renin--angiotensin system. SUMMARY Cardiac ACE2 may have bifunctional roles in COVID-19 as ACE2 not only mediates cellular susceptibility to SARS-CoV-2 infection but also protects the heart via the ACE2/Ang-(1--7) pathway. Linking tissue ACE2 from cardiac surgery patients to their comorbid conditions and medical regimens provides a unique latform to address the influence that altered expression of the ACE2/Ang-(1-7)/Mas receptor axis might have on SARs-CoV-2 vulnerability in older adults.
Collapse
Affiliation(s)
- Hao Wang
- Department of Anesthesiology of Wake Forest School of Medicine, Winston Salem, North Carolina
- Department of Internal Medicine-Section of Molecular Medicine of Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Amit K. Saha
- Department of Anesthesiology of Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Xuming Sun
- Department of Anesthesiology of Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Neal D. Kon
- Department of Cardiothoracic Surgery of Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Carlos M. Ferrario
- Department of General Surgery of Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Leanne Groban
- Department of Anesthesiology of Wake Forest School of Medicine, Winston Salem, North Carolina
- Department of Internal Medicine-Section of Molecular Medicine of Wake Forest School of Medicine, Winston Salem, North Carolina
| |
Collapse
|
31
|
Rivas-Fuentes S, Valdés VJ, Espinosa B, Gorocica-Rosete P, Salgado-Aguayo A. Could SARS-CoV-2 blocking of ACE2 in endothelial cells result in upregulation of CX3CL1, promoting thrombosis in COVID-19 patients? Med Hypotheses 2021; 151:110570. [PMID: 33836338 PMCID: PMC7992310 DOI: 10.1016/j.mehy.2021.110570] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/03/2021] [Accepted: 03/14/2021] [Indexed: 12/22/2022]
Abstract
SARS-CoV-2 is the causal agent of COVID-19 disease. Currently, infection with SARS-CoV-2 has been the cause of death of over 2.5 million people globally, and there is still no effective curative treatment. Clinically, the severe symptoms caused by COVID-19, in addition to pneumonia, are associated with the development of hyperinflammatory syndrome and thrombosis. It is urgent to expand our understanding of the molecular mechanisms involved in the pathophysiology of COVID-19. This article discusses the potential role that the chemokine CX3CL1 could have in the development of COVID-19-associated thrombosis. CX3CL1 is abundantly expressed by activated endothelium and is an important regulator of many aspects of endothelial function and dysfunction, including thrombosis. The generation of hypotheses about molecules that could be relevant in well-defined aspects of the pathophysiology of COVID-19 encourages the development of basic and clinical studies, that could help find effective and much needed treatments.
Collapse
Affiliation(s)
- Selma Rivas-Fuentes
- Department of Research on Biochemistry, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico.
| | - Víctor Julián Valdés
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Blanca Espinosa
- Department of Research on Biochemistry, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Patricia Gorocica-Rosete
- Department of Research on Biochemistry, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Alfonso Salgado-Aguayo
- Laboratory of Research on Rheumatic Diseases, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico.
| |
Collapse
|
32
|
ACE2 as therapeutic agent. Clin Sci (Lond) 2021; 134:2581-2595. [PMID: 33063820 DOI: 10.1042/cs20200570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
The angiotensin-converting enzyme 2 (ACE2) has emerged as a critical regulator of the renin-angiotensin system (RAS), which plays important roles in cardiovascular homeostasis by regulating vascular tone, fluid and electrolyte balance. ACE2 functions as a carboxymonopeptidase hydrolyzing the cleavage of a single C-terminal residue from Angiotensin-II (Ang-II), the key peptide hormone of RAS, to form Angiotensin-(1-7) (Ang-(1-7)), which binds to the G-protein-coupled Mas receptor and activates signaling pathways that counteract the pathways activated by Ang-II. ACE2 is expressed in a variety of tissues and overwhelming evidence substantiates the beneficial effects of enhancing ACE2/Ang-(1-7)/Mas axis under many pathological conditions in these tissues in experimental models. This review will provide a succinct overview on current strategies to enhance ACE2 as therapeutic agent, and discuss limitations and future challenges. ACE2 also has other functions, such as acting as a co-factor for amino acid transport and being exploited by the severe acute respiratory syndrome coronaviruses (SARS-CoVs) as cellular entry receptor, the implications of these functions in development of ACE2-based therapeutics will also be discussed.
Collapse
|
33
|
Wang J, Chen S, Meghana Yerrapragada S, Zhang W, Bihl JC. Therapeutic effects of exosomes from angiotensin-converting enzyme 2 -overexpressed endothelial progenitor cells on intracerebral hemorrhagic stroke. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2020.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
34
|
Price S, Targoński R, Sadowski J, Targoński R. To Curb the Progression of Fatal COVID-19 Course-Dream or Reality. Curr Hypertens Rep 2021; 23:12. [PMID: 33638064 PMCID: PMC7910199 DOI: 10.1007/s11906-021-01130-z] [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] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW To analyze the impact of sodium retention states on the course of COVID-19 and propose possible interventions to curb disease progression. RECENT FINDINGS Numerous data confirm a positive association of non-communicable diseases, aging, and other sodium-retaining states, including iatrogenic ones, with more severe sometimes fatal clinical course of COVID-19. Reasons for this effect could include increased angiotensin signaling via the AT1R receptor. The endothelial glycocalyx also plays an important role in infection, leading to a vicious cycle of inflammation and tissue sodium retention when damaged. RAS inhibitors may help restore glycocalyx function and prevent severe organ damage. Anticoagulants, especially heparin, may also have therapeutic applications due to antithrombotic, anti-inflammatory, glycocalyx-repairing, and antialdosteronic properties. The ambiguous influence of some diuretics on sodium balance was also discussed. Abnormal sodium storage and increased angiotensin-converting enzyme activity are related to the severity of COVID-19. Inducing sodium removal and reducing intake might improve outcomes.
Collapse
Affiliation(s)
- Szymon Price
- Miejski Szpital Zespolony w Olsztynie, Klinika Kardiologii i Chorób Wewnętrznych, Clinic of Cardiology and Internal Medicine, Metropolitan Hospital of Nicolaus Copernicus, University of Warmia and Mazury, Niepodległości 44, 10-045 Olsztyn, Poland
| | - Radosław Targoński
- Department of Cardiac & Vascular Surgery, University Hospital of Gdańsk, M. Skłodowskiej-Curie 3a street, 80-210 Gdańsk, Poland
| | - Janusz Sadowski
- Miejski Szpital Zespolony w Olsztynie, Klinika Kardiologii i Chorób Wewnętrznych, Clinic of Cardiology and Internal Medicine, Metropolitan Hospital of Nicolaus Copernicus, University of Warmia and Mazury, Niepodległości 44, 10-045 Olsztyn, Poland
| | - Ryszard Targoński
- Department of Cardiac & Vascular Surgery, University Hospital of Gdańsk, M. Skłodowskiej-Curie 3a street, 80-210 Gdańsk, Poland
| |
Collapse
|
35
|
Du F, Liu B, Zhang S. COVID-19: the role of excessive cytokine release and potential ACE2 down-regulation in promoting hypercoagulable state associated with severe illness. J Thromb Thrombolysis 2021; 51:313-329. [PMID: 32676883 PMCID: PMC7365308 DOI: 10.1007/s11239-020-02224-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The novel coronavirus disease (COVID-19) has become a universally prevalent infectious disease. The causative virus of COVID-19 is severe acute respiratory syndrome coronavirus type 2. Recent retrospective clinical studies have established a significant association between the incidence of vascular thrombotic events and the severity of COVID-19. The enhancement in serum levels of markers that reflect a hypercoagulable state has been suggested to indicate a poor prognosis. Therefore, at present, it is crucial to understand the mechanisms that foster the hypercoagulable state in COVID-19. Over-activated inflammatory response, which is manifested as excessive cytokine release in COVID-19 patients, is also associated with COVID-19 severity. This review discusses the immuno-pathological basis of the excessive cytokine release in COVID-19. Besides, this article reviews the role of pro-inflammatory or anti-inflammatory cytokines, whose significant elevations in their serum levels have been consistently detected in multiple different clinical studies, in promoting the hypercoagulable state. Since the expression of angiotensin-converting enzyme 2 (ACE2) is potentially down-regulated in COVID-19, as proposed by a recent bio-informatic analysis, mechanisms through which reduced ACE2 expressions promote vascular thrombosis are summarized. In addition, the reciprocal-enhancing effects of the excessive cytokine release and the downregulated ACE2 expression on their pro-thrombotic activities are further discussed. Here, based on currently available evidence, we review the pathogenic mechanisms of the hypercoagulable state associated with severe cases of COVID-19 to give insights into prevention and treatment of the vascular thrombotic events in COVID-19.
Collapse
Affiliation(s)
- Fenghe Du
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1st, Dongcheng District, Beijing, 100730, China.,Four-Year Program of Clinical Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Bao Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1st, Dongcheng District, Beijing, 100730, China.
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1st, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
36
|
Malinowska B, Baranowska-Kuczko M, Kicman A, Schlicker E. Opportunities, Challenges and Pitfalls of Using Cannabidiol as an Adjuvant Drug in COVID-19. Int J Mol Sci 2021; 22:1986. [PMID: 33671463 PMCID: PMC7922403 DOI: 10.3390/ijms22041986] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may lead to coronavirus disease 2019 (COVID-19) which, in turn, may be associated with multiple organ dysfunction. In this review, we present advantages and disadvantages of cannabidiol (CBD), a non-intoxicating phytocannabinoid from the cannabis plant, as a potential agent for the treatment of COVID-19. CBD has been shown to downregulate proteins responsible for viral entry and to inhibit SARS-CoV-2 replication. Preclinical studies have demonstrated its effectiveness against diseases of the respiratory system as well as its cardioprotective, nephroprotective, hepatoprotective, neuroprotective and anti-convulsant properties, that is, effects that may be beneficial for COVID-19. Only the latter two properties have been demonstrated in clinical studies, which also revealed anxiolytic and antinociceptive effects of CBD (given alone or together with Δ9-tetrahydrocannabinol), which may be important for an adjuvant treatment to improve the quality of life in patients with COVID-19 and to limit post-traumatic stress symptoms. However, one should be aware of side effects of CBD (which are rarely serious), drug interactions (also extending to drugs acting against COVID-19) and the proper route of its administration (vaping may be dangerous). Clearly, further clinical studies are necessary to prove the suitability of CBD for the treatment of COVID-19.
Collapse
Affiliation(s)
- Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, 15-222 Białystok, Poland; (M.B.-K.); (A.K.)
| | - Marta Baranowska-Kuczko
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, 15-222 Białystok, Poland; (M.B.-K.); (A.K.)
- Department of Clinical Pharmacy, Medical University of Białystok, 15-222 Białystok, Poland
| | - Aleksandra Kicman
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, 15-222 Białystok, Poland; (M.B.-K.); (A.K.)
| | - Eberhard Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
37
|
ACE2 and energy metabolism: the connection between COVID-19 and chronic metabolic disorders. Clin Sci (Lond) 2021; 135:535-554. [PMID: 33533405 DOI: 10.1042/cs20200752] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022]
Abstract
The renin-angiotensin system (RAS) has currently attracted increasing attention due to its potential function in regulating energy homeostasis, other than the actions on cellular growth, blood pressure, fluid, and electrolyte balance. The existence of RAS is well established in metabolic organs, including pancreas, liver, skeletal muscle, and adipose tissue, where activation of angiotensin-converting enzyme (ACE) - angiotensin II pathway contributes to the impairment of insulin secretion, glucose transport, fat distribution, and adipokines production. However, the activation of angiotensin-converting enzyme 2 (ACE2) - angiotensin (1-7) pathway, a novel branch of the RAS, plays an opposite role in the ACE pathway, which could reverse these consequences by improving local microcirculation, inflammation, stress state, structure remolding, and insulin signaling pathway. In addition, new studies indicate the protective RAS arm possesses extraordinary ability to enhance brown adipose tissue (BAT) activity and induces browning of white adipose tissue, and consequently, it leads to increased energy expenditure in the form of heat instead of ATP synthesis. Interestingly, ACE2 is the receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is threating public health worldwide. The main complications of SARS-CoV-2 infected death patients include many energy metabolism-related chronic diseases, such as diabetes. The specific mechanism leading to this phenomenon is largely unknown. Here, we summarize the latest pharmacological and genetic tools on regulating ACE/ACE2 balance and highlight the beneficial effects of the ACE2 pathway axis hyperactivity on glycolipid metabolism, as well as the thermogenic modulation.
Collapse
|
38
|
Update on New Aspects of the Renin-Angiotensin System in Hepatic Fibrosis and Portal Hypertension: Implications for Novel Therapeutic Options. J Clin Med 2021; 10:jcm10040702. [PMID: 33670126 PMCID: PMC7916881 DOI: 10.3390/jcm10040702] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/29/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
There is considerable experimental evidence that the renin angiotensin system (RAS) plays a central role in both hepatic fibrogenesis and portal hypertension. Angiotensin converting enzyme (ACE), a key enzyme of the classical RAS, converts angiotensin I (Ang I) to angiotensin II (Ang II), which acts via the Ang II type 1 receptor (AT1R) to stimulate hepatic fibrosis and increase intrahepatic vascular tone and portal pressure. Inhibitors of the classical RAS, drugs which are widely used in clinical practice in patients with hypertension, have been shown to inhibit liver fibrosis in animal models but their efficacy in human liver disease is yet to be tested in adequately powered clinical trials. Small trials in cirrhotic patients have demonstrated that these drugs may lower portal pressure but produce off-target complications such as systemic hypotension and renal failure. More recently, the alternate RAS, comprising its key enzyme, ACE2, the effector peptide angiotensin-(1–7) (Ang-(1–7)) which mediates its effects via the putative receptor Mas (MasR), has also been implicated in the pathogenesis of liver fibrosis and portal hypertension. This system is activated in both preclinical animal models and human chronic liver disease and it is now well established that the alternate RAS counter-regulates many of the deleterious effects of the ACE-dependent classical RAS. Work from our laboratory has demonstrated that liver-specific ACE2 overexpression reduces hepatic fibrosis and liver perfusion pressure without producing off-target effects. In addition, recent studies suggest that the blockers of the receptors of alternate RAS, such as the MasR and Mas related G protein-coupled receptor type-D (MrgD), increase splanchnic vascular resistance in cirrhotic animals, and thus drugs targeting the alternate RAS may be useful in the treatment of portal hypertension. This review outlines the role of the RAS in liver fibrosis and portal hypertension with a special emphasis on the possible new therapeutic approaches targeting the ACE2-driven alternate RAS.
Collapse
|
39
|
Su H, Ma C, Li H. Anti-infection mechanism of phosphodiesterase-5 inhibitors and their roles in coronavirus disease 2019 (Review). Exp Ther Med 2021; 21:320. [PMID: 33732293 PMCID: PMC7903479 DOI: 10.3892/etm.2021.9751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/19/2021] [Indexed: 11/11/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has a variety of impacts on the human body. Severe acute respiratory syndrome coronavirus 2 is the pathogen that causes COVID-19. It invades human tissues through the receptor angiotensin-converting enzyme 2, resulting in an imbalance in the angiotensin II (AngII) level and upregulation of renin-angiotensin system/AngII pathway activity. Furthermore, the binding of AngII to its receptor leads to vasoconstriction, endothelial injury and intravascular thrombosis. In addition, COVID-19 may have adverse effects on male reproductive organs and a marked impact on male reproductive health. Phosphodiesterase-5 inhibitors (PDE5Is) may improve vascular endothelial function, promote testicular and systemic blood circulation and testosterone secretion and enhance epididymal function, as well as sperm maturation and capacitation. PDE5Is may also be of use in the treatment of infectious diseases by enhancing immunity and anti-inflammatory responses and improving vascular endothelial function. Based on the pharmacological mechanism of PDE5Is, they are of unique value in the fight against infectious diseases and may be effective in combination with direct antiviral drugs. The anti-infection mechanisms of PDE5Is and their roles in COVID-19 were reviewed in the present study.
Collapse
Affiliation(s)
- Hao Su
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Chengquan Ma
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Hongjun Li
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| |
Collapse
|
40
|
Cohen JB, South AM, Shaltout HA, Sinclair MR, Sparks MA. Renin-angiotensin system blockade in the COVID-19 pandemic. Clin Kidney J 2021; 14:i48-i59. [PMID: 33796285 PMCID: PMC7929063 DOI: 10.1093/ckj/sfab026] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/19/2021] [Indexed: 01/08/2023] Open
Abstract
In the early months of the coronavirus disease 2019 (COVID-19) pandemic, a hypothesis emerged suggesting that pharmacologic inhibitors of the renin–angiotensin system (RAS) may increase COVID-19 severity. This hypothesis was based on the role of angiotensin-converting enzyme 2 (ACE2), a counterregulatory component of the RAS, as the binding site for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), allowing viral entry into host cells. Extrapolations from prior evidence led to speculation that upregulation of ACE2 by RAS blockade may increase the risk of adverse outcomes from COVID-19. However, counterarguments pointed to evidence of potential protective effects of ACE2 and RAS blockade with regard to acute lung injury, as well as substantial risks from discontinuing these commonly used and important medications. Here we provide an overview of classic RAS physiology and the crucial role of ACE2 in systemic pathways affected by COVID-19. Additionally, we critically review the physiologic and epidemiologic evidence surrounding the interactions between RAS blockade and COVID-19. We review recently published trial evidence and propose important future directions to improve upon our understanding of these relationships.
Collapse
Affiliation(s)
- Jordana B Cohen
- Renal-Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew M South
- Section of Nephrology, Department of Pediatrics, Brenner Children's Hospital, Wake Forest School of Medicine, Winston Salem, NC, USA.,Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston Salem, NC, USA.,Department of Surgery, Hypertension and Vascular Research, Wake Forest School of Medicine, Winston Salem, NC, USA.,Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Hossam A Shaltout
- Department of Surgery, Hypertension and Vascular Research, Wake Forest School of Medicine, Winston Salem, NC, USA.,Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston Salem, NC, USA.,Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston Salem, NC, USA.,Department of Pharmacology and Toxicology, School of Pharmacy, University of Alexandria, Alexandria, Egypt
| | - Matthew R Sinclair
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.,Renal Section, Durham VA Health Care System, Durham, NC, USA
| |
Collapse
|
41
|
Zhang J, Tecson KM, McCullough PA. Role of endothelial cell receptors in the context of SARS-CoV-2 infection (COVID-19). Proc (Bayl Univ Med Cent) 2021; 34:262-268. [PMID: 33664552 PMCID: PMC7852287 DOI: 10.1080/08998280.2021.1874231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Endothelial cell (EC) dysfunction contributes to COVID-19-associated vascular inflammation and coagulopathy, and the angiotensin-converting enzyme 2 (ACE2) receptor plays a role in EC dysfunction in COVID-19. To expand the understanding of the role of the ACE2 receptor relative to EC dysfunction, this review addresses (1) tissue distribution of the ACE2 protein and its mRNA expression in humans, (2) susceptibility of the capillary ECs to SARS-CoV-2 infection, and (3) the role of EC dysfunction relevant to ACE2 and nuclear factor-κB in COVID-19.
Collapse
Affiliation(s)
- Jun Zhang
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas
| | - Kristen M Tecson
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas
| | - Peter A McCullough
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas.,Division of Cardiology, Department of Internal Medicine, Baylor University Medical Center, Dallas, Texas.,Division of Cardiology, Department of Internal Medicine, Baylor Jack and Jane Hamilton Heart and Vascular Hospital, Dallas, Texas
| |
Collapse
|
42
|
García-Escobar A, Jiménez-Valero S, Galeote G, Jurado-Román A, García-Rodríguez J, Moreno R. The soluble catalytic ectodomain of ACE2 a biomarker of cardiac remodelling: new insights for heart failure and COVID19. Heart Fail Rev 2021; 26:961-971. [PMID: 33404999 PMCID: PMC7786157 DOI: 10.1007/s10741-020-10066-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/08/2020] [Indexed: 01/06/2023]
Abstract
The angiotensin-converting enzyme 2 (ACE2) is a type I integral membrane that was discovered two decades ago. The ACE2 exists as a transmembrane protein and as a soluble catalytic ectodomain of ACE2, also known as the soluble ACE2 that can be found in plasma and other body fluids. ACE2 regulates the local actions of the renin-angiotensin system in cardiovascular tissues, and the ACE2/Angiotensin 1–7 axis exerts protective actions in cardiovascular disease. Increasing soluble ACE2 has been associated with heart failure, cardiovascular disease, and cardiac remodelling. This is a review of the molecular structure and biochemical functions of the ACE2, as well we provided an updated on the evidence, clinical applications, and emerging potential therapies with the ACE2 in heart failure, cardiovascular disease, lung injury, and COVID-19 infection.
Collapse
Affiliation(s)
- Artemio García-Escobar
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, Madrid, Spain.
| | - Santiago Jiménez-Valero
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, Madrid, Spain
| | - Guillermo Galeote
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, Madrid, Spain
| | - Alfonso Jurado-Román
- Cardiology Department, Interventional Cardiology Section, University Hospital La Paz, Madrid, Spain
| | | | - Raúl Moreno
- Cardiology Department, Head of Interventional Cardiology Section, University Hospital La Paz, Madrid, Spain
| |
Collapse
|
43
|
Covid-19 and in situ pulmonary artery thrombosis. Respir Med 2021; 176:106176. [PMID: 33092932 PMCID: PMC7529605 DOI: 10.1016/j.rmed.2020.106176] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 01/04/2023]
|
44
|
Marquez A, Wysocki J, Pandit J, Batlle D. An update on ACE2 amplification and its therapeutic potential. Acta Physiol (Oxf) 2021; 231:e13513. [PMID: 32469114 PMCID: PMC7267104 DOI: 10.1111/apha.13513] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/11/2022]
Abstract
The renin angiotensin system (RAS) plays an important role in the pathogenesis of variety of diseases. Targeting the formation and action of angiotensin II (Ang II), the main RAS peptide, has been the key therapeutic target for last three decades. ACE‐related carboxypeptidase (ACE2), a monocarboxypeptidase that had been discovered 20 years ago, is one of the catalytically most potent enzymes known to degrade Ang II to Ang‐(1‐7), a peptide that is increasingly accepted to have organ‐protective properties that oppose and counterbalance those of Ang II. In addition to its role as a RAS enzyme ACE2 is the main receptor for SARS‐CoV‐2. In this review, we discuss various strategies that have been used to achieve amplification of ACE2 activity including the potential therapeutic potential of soluble recombinant ACE2 protein and novel shorter ACE2 variants.
Collapse
Affiliation(s)
- Alonso Marquez
- Feinberg Medical SchoolNorthwestern University Chicago IL USA
- Department of Medicine Division of Nephrology and Hypertension Chicago IL USA
| | - Jan Wysocki
- Feinberg Medical SchoolNorthwestern University Chicago IL USA
- Department of Medicine Division of Nephrology and Hypertension Chicago IL USA
| | - Jay Pandit
- Feinberg Medical SchoolNorthwestern University Chicago IL USA
- Department of Medicine Division of Nephrology and Hypertension Chicago IL USA
| | - Daniel Batlle
- Feinberg Medical SchoolNorthwestern University Chicago IL USA
- Department of Medicine Division of Nephrology and Hypertension Chicago IL USA
| |
Collapse
|
45
|
ACE2 in the renin-angiotensin system. Clin Sci (Lond) 2020; 134:3063-3078. [PMID: 33264412 DOI: 10.1042/cs20200478] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/12/2020] [Accepted: 11/19/2020] [Indexed: 01/01/2023]
Abstract
In 2020 we are celebrating the 20th anniversary of the angiotensin-converting enzyme 2 (ACE2) discovery. This event was a landmark that shaped the way that we see the renin-angiotensin system (RAS) today. ACE2 is an important molecular hub that connects the RAS classical arm, formed mainly by the octapeptide angiotensin II (Ang II) and its receptor AT1, with the RAS alternative or protective arm, formed mainly by the heptapeptides Ang-(1-7) and alamandine, and their receptors, Mas and MrgD, respectively. In this work we reviewed classical and modern literature to describe how ACE2 is a critical component of the protective arm, particularly in the context of the cardiac function, coagulation homeostasis and immune system. We also review recent literature to present a critical view of the role of ACE2 and RAS in the SARS-CoV-2 pandemic.
Collapse
|
46
|
Maiuolo J, Mollace R, Gliozzi M, Musolino V, Carresi C, Paone S, Scicchitano M, Macrì R, Nucera S, Bosco F, Scarano F, Zito MC, Ruga S, Tavernese A, Mollace V. The Contribution of Endothelial Dysfunction in Systemic Injury Subsequent to SARS-Cov-2 Infection. Int J Mol Sci 2020; 21:E9309. [PMID: 33291346 PMCID: PMC7730352 DOI: 10.3390/ijms21239309] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/28/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) infection is associated, alongside with lung infection and respiratory disease, to cardiovascular dysfunction that occurs at any stage of the disease. This includes ischemic heart disease, arrhythmias, and cardiomyopathies. The common pathophysiological link between SARS-CoV-2 infection and the cardiovascular events is represented by coagulation abnormalities and disruption of factors released by endothelial cells, which contribute in maintaining the blood vessels into an anti-thrombotic state. Thus, early alteration of the functionality of endothelial cells, which may be found soon after SARS-CoV-2 infection, seems to represent the major target of a SARS CoV-2 disease state and accounts for the systemic vascular dysfunction that leads to a detrimental effect in terms of hospitalization and death accompanying the disease. In particular, the molecular interaction of SARS-CoV-2 with the ACE2 receptor located in the endothelial cell surface, either at the pulmonary and systemic level, leads to early impairment of endothelial function, which, in turn, is followed by vascular inflammation and thrombosis of peripheral blood vessels. This highlights systemic hypoxia and further aggravates the vicious circle that compromises the development of the disease, leading to irreversible tissue damage and death of people with SARS CoV-2 infection. The review aims to assess some recent advances to define the crucial role of endothelial dysfunction in the pathogenesis of vascular complications accompanying SARS-CoV-2 infection. In particular, the molecular mechanisms associated with the interaction of SARS CoV-2 with the ACE2 receptor located on the endothelial cells are highlighted to support its role in compromising endothelial cell functionality. Finally, the consequences of endothelial dysfunction in enhancing pro-inflammatory and pro-thrombotic effects of SARS-CoV-2 infection are assessed in order to identify early therapeutic interventions able to reduce the impact of the disease in high-risk patients.
Collapse
Affiliation(s)
- Jessica Maiuolo
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Rocco Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Department of Medicine, Chair of Cardiology, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Micaela Gliozzi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Sara Paone
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
| | - Roberta Macrì
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Saverio Nucera
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Federica Scarano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
| | - Stefano Ruga
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
| | - Annamaria Tavernese
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Department of Medicine, Chair of Cardiology, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele Pisana, 00163 Roma, Italy
| |
Collapse
|
47
|
Kumar A, Narayan RK, Kumari C, Faiq MA, Kulandhasamy M, Kant K, Pareek V. SARS-CoV-2 cell entry receptor ACE2 mediated endothelial dysfunction leads to vascular thrombosis in COVID-19 patients. Med Hypotheses 2020; 145:110320. [PMID: 33032170 PMCID: PMC7525265 DOI: 10.1016/j.mehy.2020.110320] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 01/05/2023]
Abstract
Several studies have described unusually high incidence of vascular thrombosis in coronavirus disease-2019 (COVID-19) patients. Pathogenesis of the vascular thrombosis in COVID-19 is least understood for now and presents a challenge to the treating physicians. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative pathogen for COVID-19, has been shown to bind to angiotensin converting enzyme 2 (ACE2) protein in human epithelial cells which facilitates its entry in the organ and mediate tissue specific pathogenesis. For ACE2 mediated cell entry of the SARS-CoV-2, co-expression of one more protein-Transmembrane protease serine 2 (TMPRSS2) is essential. Existing studies suggested significant expression of ACE2 and TMPRSS2 in human vascular endothelium. Vascular endothelial dysfunction can potentially activate coagulation cascade eventually resulting in thrombosis. ACE2 has proven role in the maintenance of endothelial integrity inside the vessels. Existing in situ evidence for SARS-CoV-1 (the causative agent for SARS pandemic of 2002, which shared ACE2 as cell entry receptor) suggested that virus binding can downregulate ACE2, thus can induce endothelial dysfunction. Recently, in situ evidence has been presented that SARS-CoV-2 can infect cells in engineered human vascular endothelium, which can be effectively blocked by using clinical-grade recombinant human ACE2. Based on the circumstantial evidence present in the literature, we propose a SARS-CoV-2 cell entry receptor ACE2 based mechanism for vascular thrombosis in COVID-19 patients.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India; Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India.
| | - Ravi K Narayan
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India; Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Chiman Kumari
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India; Department of Anatomy, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Muneeb A Faiq
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India; New York University (NYU) Langone Health Center, NYU Robert I Grossman School of Medicine, New York, NY, USA
| | - Maheswari Kulandhasamy
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India; Department of Biochemistry, Maulana Azad Medical College (MAMC), New Delhi, India
| | - Kamla Kant
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India; Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Bathinda, India
| | - Vikas Pareek
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India; National Brain Research Center, Manesar, Haryana, India
| |
Collapse
|
48
|
Medzikovic L, Cunningham CM, Li M, Amjedi M, Hong J, Ruffenach G, Eghbali M. Sex differences underlying preexisting cardiovascular disease and cardiovascular injury in COVID-19. J Mol Cell Cardiol 2020; 148:25-33. [PMID: 32835666 PMCID: PMC7442559 DOI: 10.1016/j.yjmcc.2020.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023]
Abstract
The novel 2019 coronavirus disease (COVID-19), resulting from severe acute respiratory syndrome coronarvirus-2 (SARS-CoV-2) infection, typically leads to respiratory failure in severe cases; however, cardiovascular injury is reported to contribute to a substantial proportion of COVID-19 deaths. Preexisting cardiovascular disease (CVD) is among the most common risk factors for hospitalization and death in COVID-19 patients, and the pathogenic mechanisms of COVID-19 disease progression itself may promote the development of cardiovascular injury, increasing risk of in-hospital death. Sex differences in COVID-19 are becoming more apparent as mounting data indicate that males seem to be disproportionately at risk of severe COVID-19 outcome due to preexisting CVD and COVID-19-related cardiovascular injury. In this review, we will provide a basic science perspective on current clinical observations in this rapidly evolving field and discuss the interplay sex differences, preexisting CVD and COVID-19-related cardiac injury.
Collapse
Affiliation(s)
- Lejla Medzikovic
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Christine M Cunningham
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Min Li
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Marjan Amjedi
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Jason Hong
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Department of Medicine, Division of Pulmonary and Critical Care, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Gregoire Ruffenach
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Mansoureh Eghbali
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
49
|
Sparks MA, South AM, Badley AD, Baker-Smith CM, Batlle D, Bozkurt B, Cattaneo R, Crowley SD, Dell’Italia LJ, Ford AL, Griendling K, Gurley SB, Kasner SE, Murray JA, Nath KA, Pfeffer MA, Rangaswami J, Taylor WR, Garovic VD. Severe Acute Respiratory Syndrome Coronavirus 2, COVID-19, and the Renin-Angiotensin System: Pressing Needs and Best Research Practices. Hypertension 2020; 76:1350-1367. [PMID: 32981369 PMCID: PMC7685174 DOI: 10.1161/hypertensionaha.120.15948] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is associated with significant morbidity and mortality throughout the world, predominantly due to lung and cardiovascular injury. The virus responsible for COVID-19-severe acute respiratory syndrome coronavirus 2-gains entry into host cells via ACE2 (angiotensin-converting enzyme 2). ACE2 is a primary enzyme within the key counter-regulatory pathway of the renin-angiotensin system (RAS), which acts to oppose the actions of Ang (angiotensin) II by generating Ang-(1-7) to reduce inflammation and fibrosis and mitigate end organ damage. As COVID-19 spans multiple organ systems linked to the cardiovascular system, it is imperative to understand clearly how severe acute respiratory syndrome coronavirus 2 may affect the multifaceted RAS. In addition, recognition of the role of ACE2 and the RAS in COVID-19 has renewed interest in its role in the pathophysiology of cardiovascular disease in general. We provide researchers with a framework of best practices in basic and clinical research to interrogate the RAS using appropriate methodology, especially those who are relatively new to the field. This is crucial, as there are many limitations inherent in investigating the RAS in experimental models and in humans. We discuss sound methodological approaches to quantifying enzyme content and activity (ACE, ACE2), peptides (Ang II, Ang-[1-7]), and receptors (types 1 and 2 Ang II receptors, Mas receptor). Our goal is to ensure appropriate research methodology for investigations of the RAS in patients with severe acute respiratory syndrome coronavirus 2 and COVID-19 to ensure optimal rigor and reproducibility and appropriate interpretation of results from these investigations.
Collapse
Affiliation(s)
- Matthew A. Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC
- Renal Section, Durham VA Health Care System, Durham, NC
- American Heart Association, Council on Kidney in Cardiovascular Disease
| | - Andrew M. South
- American Heart Association, Council on Kidney in Cardiovascular Disease
- American Heart Association, Council on Hypertension
- Section of Nephrology, Department of Pediatrics, Brenner Children’s Hospital, Wake Forest School of Medicine, Winston Salem, NC
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston Salem, NC
- Department of Surgery-Hypertension and Vascular Research, Wake Forest School of Medicine, Winston Salem, NC
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston Salem, NC
| | - Andrew D. Badley
- Division of Infectious Diseases, Mayo Clinic College of Medicine, Rochester, MN
| | - Carissa M. Baker-Smith
- Director of Preventive Cardiology, Division of Pediatric Cardiology, Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
- American Heart Association, Council on Lifelong Congenital Heart Disease and Heart Health in the Young
| | - Daniel Batlle
- Division of Nephrology and Hypertension, Northwestern University Feinberg Medical School, Chicago, IL
- American Heart Association, Council on Hypertension
| | - Biykem Bozkurt
- Section of Cardiology, Department of Internal Medicine, Baylor College of Medicine, Houston, TX
- Michael E. DeBakey VA Medical Center, Houston, TX
- American Heart Association, Council on Clinical Cardiology
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN
| | - Steven D. Crowley
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC
- Renal Section, Durham VA Health Care System, Durham, NC
- American Heart Association, Council on Kidney in Cardiovascular Disease
| | - Louis J. Dell’Italia
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
- Department of Veterans Affairs Medical Center, Birmingham, AL
- American Heart Association, Council on Basic Cardiovascular Sciences
| | - Andria L. Ford
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO
- American Heart Association, Stroke Council
| | - Kathy Griendling
- American Heart Association, Council on Basic Cardiovascular Sciences
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA
| | - Susan B. Gurley
- American Heart Association, Council on Kidney in Cardiovascular Disease
- Department of Medicine, Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, OR
| | - Scott E. Kasner
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania
- American Heart Association, Stroke Council
| | - Joseph A. Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | - Karl A. Nath
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, MN
| | - Marc A. Pfeffer
- American Heart Association, Council on Clinical Cardiology
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Janani Rangaswami
- American Heart Association, Council on Kidney in Cardiovascular Disease
- Department of Medicine, Einstein Medical Center Philadelphia, Philadelphia, PA
- Sidney Kimmel College of Thomas Jefferson University, Philadelphia, PA
| | - W. Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA
- Division of Cardiology, Atlanta VA Medical Center, Decatur, GA
- American Heart Association, Council on Arteriosclerosis, Thrombosis and Vascular Biology
| | - Vesna D. Garovic
- American Heart Association, Council on Hypertension
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, MN
- Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
50
|
Stein RA, Young LM. From ACE2 to COVID-19: A multiorgan endothelial disease. Int J Infect Dis 2020; 100:425-430. [PMID: 32896660 PMCID: PMC7832810 DOI: 10.1016/j.ijid.2020.08.083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Richard A Stein
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn, NY 11201, USA; LaGuardia Community College, Department of Natural Sciences, City University of New York, New York, NY 11101, USA.
| | - Lauren M Young
- University of Chicago, Department of Internal Medicine, 5841 S Maryland Ave, Chicago, IL 60637, USA.
| |
Collapse
|