251
|
Osuchowski MF, Winkler MS, Skirecki T, Cajander S, Shankar-Hari M, Lachmann G, Monneret G, Venet F, Bauer M, Brunkhorst FM, Weis S, Garcia-Salido A, Kox M, Cavaillon JM, Uhle F, Weigand MA, Flohé SB, Wiersinga WJ, Almansa R, de la Fuente A, Martin-Loeches I, Meisel C, Spinetti T, Schefold JC, Cilloniz C, Torres A, Giamarellos-Bourboulis EJ, Ferrer R, Girardis M, Cossarizza A, Netea MG, van der Poll T, Bermejo-Martín JF, Rubio I. The COVID-19 puzzle: deciphering pathophysiology and phenotypes of a new disease entity. THE LANCET RESPIRATORY MEDICINE 2021; 9:622-642. [PMID: 33965003 PMCID: PMC8102044 DOI: 10.1016/s2213-2600(21)00218-6] [Citation(s) in RCA: 340] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/06/2021] [Accepted: 03/11/2021] [Indexed: 01/08/2023]
Abstract
The zoonotic SARS-CoV-2 virus that causes COVID-19 continues to spread worldwide, with devastating consequences. While the medical community has gained insight into the epidemiology of COVID-19, important questions remain about the clinical complexities and underlying mechanisms of disease phenotypes. Severe COVID-19 most commonly involves respiratory manifestations, although other systems are also affected, and acute disease is often followed by protracted complications. Such complex manifestations suggest that SARS-CoV-2 dysregulates the host response, triggering wide-ranging immuno-inflammatory, thrombotic, and parenchymal derangements. We review the intricacies of COVID-19 pathophysiology, its various phenotypes, and the anti-SARS-CoV-2 host response at the humoral and cellular levels. Some similarities exist between COVID-19 and respiratory failure of other origins, but evidence for many distinctive mechanistic features indicates that COVID-19 constitutes a new disease entity, with emerging data suggesting involvement of an endotheliopathy-centred pathophysiology. Further research, combining basic and clinical studies, is needed to advance understanding of pathophysiological mechanisms and to characterise immuno-inflammatory derangements across the range of phenotypes to enable optimum care for patients with COVID-19.
Collapse
Affiliation(s)
- Marcin F Osuchowski
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Research Center, Vienna, Austria
| | - Martin S Winkler
- Department of Anaesthesiology, University of Göttingen Medical Center, Göttingen, Georg-August University of Göttingen, Göttingen, Germany
| | - Tomasz Skirecki
- Laboratory of Flow Cytometry, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Sara Cajander
- Department of Infectious Diseases, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Manu Shankar-Hari
- Guy's and St Thomas' NHS Foundation Trust, ICU support offices, St Thomas' Hospital, London, UK; School of Immunology & Microbial Sciences, Kings College London, London, UK
| | - Gunnar Lachmann
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité-Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute of Health, Berlin, Germany
| | - Guillaume Monneret
- Hospices Civils de Lyon, Immunology Laboratory, Edouard Herriot Hospital, Lyon, France; Pathophysiology of Injury-Induced Immunosuppression, Equipe d'Accueil 7426, Université Claude Bernard Lyon 1 - bioMérieux - Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Fabienne Venet
- Hospices Civils de Lyon, Immunology Laboratory, Edouard Herriot Hospital, Lyon, France; Pathophysiology of Injury-Induced Immunosuppression, Equipe d'Accueil 7426, Université Claude Bernard Lyon 1 - bioMérieux - Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine and Center for Sepsis Control and Care, Jena University Hospital-Friedrich Schiller University, Jena, Germany
| | - Frank M Brunkhorst
- Department of Anesthesiology and Intensive Care Medicine and Center for Sepsis Control and Care, Jena University Hospital-Friedrich Schiller University, Jena, Germany; Center for Clinical Studies, Jena University Hospital-Friedrich Schiller University, Jena, Germany
| | - Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine and Center for Sepsis Control and Care, Jena University Hospital-Friedrich Schiller University, Jena, Germany; Institute for Infectious Disease and Infection Control, Jena University Hospital-Friedrich Schiller University, Jena, Germany
| | - Alberto Garcia-Salido
- Pediatric Critical Care Unit, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Matthijs Kox
- Department of Intensive Care Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefanie B Flohé
- Department of Trauma, Hand, and Reconstructive Surgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - W Joost Wiersinga
- Division of Infectious Diseases and Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Raquel Almansa
- Group for Biomedical Research in Sepsis, Hospital Universitario Río Hortega de Valladolid, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Centro de Investigación Biomedica En Red-Enfermedades Respiratorias, Instituto de salud Carlos III, Madrid, Spain
| | - Amanda de la Fuente
- Group for Biomedical Research in Sepsis, Hospital Universitario Río Hortega de Valladolid, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Ignacio Martin-Loeches
- Multidisciplinary Intensive Care Research Organization, St James's Hospital, Dublin, Ireland
| | - Christian Meisel
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology, Labor Berlin-Charité Vivantes, Berlin, Germany
| | - Thibaud Spinetti
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Catia Cilloniz
- Pneumology Department, Respiratory Institute, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, ICREA, CIBERESUCICOVID, Spain
| | - Antoni Torres
- Division of Infectious Diseases and Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands; Pneumology Department, Respiratory Institute, Hospital Clinic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, ICREA, CIBERESUCICOVID, Spain; SGR 911-ICREA Academia, Barcelona, Spain
| | | | - Ricard Ferrer
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias, Instituto de salud Carlos III, Madrid, Spain; Intensive Care Department and Shock, Organ Dysfunction and Resuscitation Research Group, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Massimo Girardis
- Department of Anesthesia and Intensive Care, University Hospital of Modena, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Mihai G Netea
- Department of Intensive Care Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands; Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania; Department for Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Tom van der Poll
- Division of Infectious Diseases and Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jesús F Bermejo-Martín
- Group for Biomedical Research in Sepsis, Hospital Universitario Río Hortega de Valladolid, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Centro de Investigación Biomedica En Red-Enfermedades Respiratorias, Instituto de salud Carlos III, Madrid, Spain
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine and Center for Sepsis Control and Care, Jena University Hospital-Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
252
|
Wiese O, Zemlin AE, Pillay TS. Molecules in pathogenesis: angiotensin converting enzyme 2 (ACE2). J Clin Pathol 2021; 74:285-290. [PMID: 32759311 PMCID: PMC7409947 DOI: 10.1136/jclinpath-2020-206954] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
The renin-angiotensin system is mainly associated with the regulation of blood pressure, but recently many other functions of this system have been described. ACE2, an 805-amino acid monocarboxypeptidase type I transmembrane glycoprotein, was discovered in 2000 and has sequence similarity to two other proteins, namely ACE and collectrin. The ACE2 gene is located on Xp22 and is highly polymorphic. ACE2 is expressed in numerous tissues especially the lung alveolar epithelial cells, heart, kidney and gastrointestinal tract. Animal studies have found that ACE2 is central in diseases affecting almost all organ systems, among other cardiac, respiratory, renal and endocrine functions. ACE2 was identified as the cellular contact point for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of the global pandemic (COVID-19), and is a potential drug target. SARS-CoV-2 infection has several effects on the renin-angiotensin system and conversely, regulation of this receptor may affect the progress of infection. We describe the genetics and functions of ACE2, explore its various physiological functions in the renin-angiotensin system and discuss its role in the pathophysiology of disease. ACE2 opposes the vasopressor ACE pathway of the renin-angiotensin system by converting angiotensin (Ang) I to Ang (1-9) and Ang II to Ang (1-7) which initiates the vasodilatory pathway. ACE2 may have a protective effect in the lung and kidney as knockout mice display susceptibility to acute respiratory distress and hypertensive nephropathy. Binding of SARS-CoV-2 and the subsequent fusion and downregulation of this pathway during SARS-CoV-2 infection may explain some of the unusual sequelae seen in COVID-19.
Collapse
Affiliation(s)
- Owen Wiese
- Division of Chemical Pathology, Faculty of Health Sciences, Stellenbosch University & National Health Laboratory Service (NHLS), Cape Town, South Africa
| | - Annalise E Zemlin
- Division of Chemical Pathology, Faculty of Health Sciences, Stellenbosch University & National Health Laboratory Service (NHLS), Cape Town, South Africa
| | - Tahir S Pillay
- Department of Chemical Pathology, University of Pretoria & National Health Laboratory Service (NHLS), Pretoria, South Africa
- Division of Chemical Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
253
|
Eun LY. Is multisystem inflammatory syndrome related with coronavirus disease 2019, Kawasaki disease, and angiotensin-converting enzyme 2 in children? Clin Exp Pediatr 2021; 64:225-226. [PMID: 33677852 PMCID: PMC8103036 DOI: 10.3345/cep.2021.00031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/01/2021] [Indexed: 01/05/2023] Open
Affiliation(s)
- Lucy Youngmin Eun
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
254
|
Bae JH, Choi SK, Kim NH, Lee J, Kim SG. Use of Renin-Angiotensin-Aldosterone System Inhibitors and Severe COVID-19 Outcomes in Patients with Hypertension: A Nationwide Cohort Study. Diabetes Metab J 2021; 45:430-438. [PMID: 33611884 PMCID: PMC8164940 DOI: 10.4093/dmj.2020.0279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/10/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Angiotensin-converting enzyme 2 facilitates the entry of severe acute respiratory syndrome coronavirus 2 into the human body. We investigated the association of renin-angiotensin-aldosterone system (RAAS) inhibitor use with severe coronavirus disease 2019 (COVID-19) outcomes in hypertensive patients. METHODS We identified hypertensive patients with confirmed COVID-19 from the Korean Health Insurance Review and Assessment Service from inception to May 15, 2020. The primary outcome was the composite of intensive care unit (ICU) admission, invasive mechanical ventilation (IMV), continuous renal replacement therapy (CRRT), extracorporeal membrane oxygenation (ECMO), and death from COVID-19. The individual components were evaluated as secondary outcomes. RESULTS Of 1,374 hypertensive patients with COVID-19, 1,076 (78.3%) and 298 (21.7%) were users and never-users of RAAS inhibitors, respectively. The RAAS inhibitor users were not associated with the risk of the primary outcome (adjusted odds ratio [aOR], 0.72; 95% confidence interval [CI], 0.46 to 1.10). The risk of ICU admission was significantly lower in the users than the never-users (aOR, 0.44; 95% CI, 0.24 to 0.84). The RAAS inhibitors were beneficial only in ICU admissions that did not require IMV (aOR, 0.28; 95% CI, 0.14 to 0.58). The risk of death from COVID-19 was comparable between the groups (aOR, 1.09; 95% CI, 0.64 to 1.85). We could not evaluate the risks of CRRT and ECMO owing to the small number of events. CONCLUSION RAAS inhibitor use was not associated with the composite of severe outcomes in the hypertensive patients with COVID-19 but significantly lowered the risk of ICU admission, particularly in patients who did not require IMV.
Collapse
Affiliation(s)
- Jae Hyun Bae
- Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Sun Kyu Choi
- Department of Biostatistics, Korea University College of Medicine, Seoul, Korea
| | - Nam Hoon Kim
- Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Juneyoung Lee
- Department of Biostatistics, Korea University College of Medicine, Seoul, Korea
| | - Sin Gon Kim
- Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
255
|
COVID-19 and diabetes: A bidirectional relationship. CLÍNICA E INVESTIGACIÓN EN ARTERIOSCLEROSIS (ENGLISH EDITION) 2021. [PMCID: PMC8064838 DOI: 10.1016/j.artere.2021.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causal agent of coronavirus disease 2019 (COVID-19). Diabetes is one of the most frequent comorbidities in people with COVID-19 with a prevalence that varies between 7 and 30%. Diabetics infected with SARS-CoV-2 have a higher rate of hospital admission, severe pneumonia, and higher mortality compared to non-diabetic subjects. Chronic hyperglycemia can compromise innate and humoral immunity. Furthermore, diabetes is associated with a low-grade chronic inflammatory state that favours the development of an exaggerated inflammatory response and therefore the appearance of acute respiratory distress syndrome. Recent evidence has shown that SARS-CoV-2 is also capable of causing direct damage to the pancreas that could worsen hyperglycemia and even induce the onset of diabetes in previously non-diabetic subjects. Therapeutic strategies should be aimed at facilitating patient access to the healthcare system. Control of blood glucose and comorbidities must be individualised in order to reduce the incidence of complications and decrease the burden on health systems. In this article we will review the pathophysiological mechanisms that explain the bidirectional relationship between COVID-19 and diabetes mellitus, its implication in the prognosis and management of hyperglycemia in this group of patients.
Collapse
|
256
|
Sarahian N, Saei Ghare Naz M, Ramezani Tehrani F. Following SARS-CoV-2 in the first trimester of pregnancy, what should we do in the 2nd, 3rd trimesters, and postpartum in terms of thyroid assessment? Endocrine 2021; 72:356-357. [PMID: 33721205 PMCID: PMC7958590 DOI: 10.1007/s12020-021-02678-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/27/2021] [Indexed: 12/03/2022]
Affiliation(s)
- Nahid Sarahian
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Saei Ghare Naz
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
257
|
Patil M, Singh S, Henderson J, Krishnamurthy P. Mechanisms of COVID-19-induced cardiovascular disease: Is sepsis or exosome the missing link? J Cell Physiol 2021; 236:3366-3382. [PMID: 33078408 PMCID: PMC7920909 DOI: 10.1002/jcp.30109] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has reached a pandemic level, spreading across the globe by affecting over 33 million people and causing over 1,009,270 deaths. SARS-CoV-2 is highly infectious with a high basic reproduction number (R0 ) of 2.2-5.7 that has led to its exponential spread. Besides, very little is known about it in terms of immunogenicity and its molecular targets. SARS-CoV-2 causes acute respiratory distress syndrome, followed by multiple organ failure and death in a small percentage of individuals. Cardiac injury has emerged as another dreaded outcome of COVID-19 complications. However, a thorough understanding of the pathogenesis of SARS-CoV-2 is lacking. In this review, we discuss the virus, possible mechanisms of COVID-19-induced cardiac injury, and potential therapeutic strategies, and we explore if exosomes could be targeted to treat symptoms of COVID-19. Furthermore, we discussed the virus-induced sepsis, which may be the cause of multiple organ failure, including myocardial injury.
Collapse
Affiliation(s)
- Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Sarojini Singh
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - John Henderson
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, AL 35294, USA
| |
Collapse
|
258
|
Yim J, Lim HH, Kwon Y. COVID-19 and pulmonary fibrosis: therapeutics in clinical trials, repurposing, and potential development. Arch Pharm Res 2021; 44:499-513. [PMID: 34047940 PMCID: PMC8161353 DOI: 10.1007/s12272-021-01331-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
In 2019, an unprecedented disease named coronavirus disease 2019 (COVID-19) emerged and spread across the globe. Although the rapid transmission of COVID-19 has resulted in thousands of deaths and severe lung damage, conclusive treatment is not available. However, three COVID-19 vaccines have been authorized, and two more will be approved soon, according to a World Health Organization report on December 12, 2020. Many COVID-19 patients show symptoms of acute lung injury that eventually leads to pulmonary fibrosis. Our aim in this article is to present the relationship between pulmonary fibrosis and COVID-19, with a focus on angiotensin converting enzyme-2. We also evaluate the radiological imaging methods computed tomography (CT) and chest X-ray (CXR) for visualization of patient lung condition. Moreover, we review possible therapeutics for COVID-19 using four categories: treatments related and unrelated to lung disease and treatments that have and have not entered clinical trials. Although many treatments have started clinical trials, they have some drawbacks, such as short-term and small-group testing, that need to be addressed as soon as possible.
Collapse
Affiliation(s)
- Joowon Yim
- College of Pharmacy, Ewha Womans University, 120-750, Seoul, Republic of Korea
| | - Hee Hyun Lim
- College of Pharmacy, Ewha Womans University, 120-750, Seoul, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Ewha Womans University, 120-750, Seoul, Republic of Korea.
| |
Collapse
|
259
|
Bui ATN, Tyan K, Giobbie-Hurder A, Klein IA, Manos MP, Zubiri L, Reynolds K, Grover S, Weinhouse GL, Ott PA, LeBoeuf NR, Rahma O. Impact of COVID-19 on Patients with Cancer Receiving Immune Checkpoint Inhibitors. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2021; 4:35-44. [PMID: 35663537 PMCID: PMC9153254 DOI: 10.36401/jipo-20-34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/09/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022]
Abstract
Introduction To evaluate the impact of Sars-Cov-2 infection on mortality and immune checkpoint inhibitor (ICI) toxicity in patients with cancer receiving ICIs compared to those not receiving ICIs. Methods We conducted a retrospective matched cohort study of 25 patients receiving ICIs within 1 year of coronavirus disease 2019 (COVID-19) diagnosis between March 20, 2020, and June 3, 2020, at the Dana-Farber Cancer Institute/Mass General Brigham. Cases were matched 1:1 with controls based on age, sex, and anticancer therapy within the prior 6 months. Results Seven of 25 (28%) patients receiving ICIs died from COVID-19 as compared with nine of 25 (36%) controls. Through multivariable analysis adjusting for age, sex, and anticancer therapy, ICI use was not associated with increased risk for COVID-19 death (OR [odds ratio] 0.36, 95% CI 0.07-1.87). Determinants of mortality included age (OR 1.14, 95% CI 1.03-1.27) and chronic obstructive pulmonary disease (OR 12.26, 95% CI 1.76-85.14). Statin use was protective against mortality (OR 0.08, 95% CI 0.01-0.63). Two patients experienced persistent immune-related adverse events (irAEs) (hypophysitis); one had new-onset irAE (hypothyroidism) during their COVID-19 course. Patients with ICIs had significantly higher platelet (p = 0.017) and D-dimer (p = 0.037) levels. Elevated troponin levels (p = 0.01) were associated with COVID-19 death in patients using ICI. Conclusion There is insufficient evidence to conclude COVID-19-related outcomes are associated with ICIs, and we did not observe an increased risk of COVID-19-related death associated with ICIs. The potential protective effect of statin therapy and role of laboratory biomarkers warrant further investigation.
Collapse
Affiliation(s)
| | | | - Anita Giobbie-Hurder
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Isaac A. Klein
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Whitehead Institute of Biomedical Research, Cambridge, MA, USA
| | - Michael P. Manos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Leyre Zubiri
- Department of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Kerry Reynolds
- Department of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Shilpa Grover
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Gerald L. Weinhouse
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Patrick A. Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicole R. LeBoeuf
- Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
| | - Osama Rahma
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
260
|
Pan Y, Gao F, Zhao S, Han J, Chen F. Role of the SphK-S1P-S1PRs pathway in invasion of the nervous system by SARS-CoV-2 infection. Clin Exp Pharmacol Physiol 2021; 48:637-650. [PMID: 33565127 PMCID: PMC8014301 DOI: 10.1111/1440-1681.13483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 01/08/2023]
Abstract
Global spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still ongoing. Before an effective vaccine is available, the development of potential treatments for resultant coronavirus disease 2019 (COVID-19) is crucial. One of the disease hallmarks is hyper-inflammatory responses, which usually leads to a severe lung disease. Patients with COVID-19 also frequently suffer from neurological symptoms such as acute diffuse encephalomyelitis, brain injury and psychiatric complications. The metabolic pathway of sphingosine-1-phosphate (S1P) is a dynamic regulator of various cell types and disease processes, including the nervous system. It has been demonstrated that S1P and its metabolic enzymes, regulating neuroinflammation and neurogenesis, exhibit important functions during viral infection. S1P receptor 1 (S1PR1) analogues including AAL-R and RP-002 inhibit pathophysiological responses at the early stage of H1N1 virus infection and then play a protective role. Fingolimod (FTY720) is an S1P receptor modulator and is being tested for treating COVID-19. Our review provides an overview of SARS-CoV-2 infection and critical role of the SphK-S1P-SIPR pathway in invasion of SARS-CoV-2 infection, particularly in the central nervous system (CNS). This may help design therapeutic strategies based on the S1P-mediated signal transduction, and the adjuvant therapeutic effects of S1P analogues to limit or prevent the interaction between the host and SARS-CoV-2, block the spread of the SARS-CoV-2, and consequently treat related complications in the CNS.
Collapse
Affiliation(s)
- Yuehai Pan
- Department of Hand and Foot SurgeryThe Affiliated Hospital of Qingdao UniversityShangdongChina
| | - Fei Gao
- Department of Hand and Foot SurgeryThe Affiliated Hospital of Qingdao UniversityShangdongChina
| | - Shuai Zhao
- Department of AnesthesiologyBonn UniversityBonnGermany
| | - Jinming Han
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Fan Chen
- Department of NeurosurgeryThe Affiliated Hospital of Qingdao UniversityShangdongChina
| |
Collapse
|
261
|
Djokovic N, Ruzic D, Djikic T, Cvijic S, Ignjatovic J, Ibric S, Baralic K, Buha Djordjevic A, Curcic M, Djukic‐Cosic D, Nikolic K. An Integrative in silico Drug Repurposing Approach for Identification of Potential Inhibitors of SARS-CoV-2 Main Protease. Mol Inform 2021; 40:e2000187. [PMID: 33787066 PMCID: PMC8250230 DOI: 10.1002/minf.202000187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/15/2021] [Indexed: 12/25/2022]
Abstract
Considering the urgent need for novel therapeutics in ongoing COVID-19 pandemic, drug repurposing approach might offer rapid solutions comparing to de novo drug design. In this study, we designed an integrative in silico drug repurposing approach for rapid selection of potential candidates against SARS-CoV-2 Main Protease (Mpro ). To screen FDA-approved drugs, we implemented structure-based molecular modelling techniques, physiologically-based pharmacokinetic (PBPK) modelling of drugs disposition and data mining analysis of drug-gene-COVID-19 association. Through presented approach, we selected the most promising FDA approved drugs for further COVID-19 drug development campaigns and analysed them in context of available experimental data. To the best of our knowledge, this is unique in silico study which integrates structure-based molecular modeling of Mpro inhibitors with predictions of their tissue disposition, drug-gene-COVID-19 associations and prediction of pleiotropic effects of selected candidates.
Collapse
Affiliation(s)
- Nemanja Djokovic
- Department of Pharmaceutical ChemistryFaculty of PharmacyUniversity of BelgradeVojvode Stepe 45011221BelgradeSerbia
| | - Dusan Ruzic
- Department of Pharmaceutical ChemistryFaculty of PharmacyUniversity of BelgradeVojvode Stepe 45011221BelgradeSerbia
| | - Teodora Djikic
- Department of Pharmaceutical ChemistryFaculty of PharmacyUniversity of BelgradeVojvode Stepe 45011221BelgradeSerbia
| | - Sandra Cvijic
- Department of Pharmaceutical Technology and CosmetologyUniversity of BelgradeFaculty of PharmacyVojvode Stepe 45011221BelgradeSerbia
| | - Jelisaveta Ignjatovic
- Department of Pharmaceutical Technology and CosmetologyUniversity of BelgradeFaculty of PharmacyVojvode Stepe 45011221BelgradeSerbia
| | - Svetlana Ibric
- Department of Pharmaceutical Technology and CosmetologyUniversity of BelgradeFaculty of PharmacyVojvode Stepe 45011221BelgradeSerbia
| | - Katarina Baralic
- Department of Toxicology “Akademik Danilo Soldatovic”Faculty of PharmacyUniversity of BelgradeVojvode Stepe 45011221BelgradeSerbia
| | - Aleksandra Buha Djordjevic
- Department of Toxicology “Akademik Danilo Soldatovic”Faculty of PharmacyUniversity of BelgradeVojvode Stepe 45011221BelgradeSerbia
| | - Marijana Curcic
- Department of Toxicology “Akademik Danilo Soldatovic”Faculty of PharmacyUniversity of BelgradeVojvode Stepe 45011221BelgradeSerbia
| | - Danijela Djukic‐Cosic
- Department of Toxicology “Akademik Danilo Soldatovic”Faculty of PharmacyUniversity of BelgradeVojvode Stepe 45011221BelgradeSerbia
| | - Katarina Nikolic
- Department of Pharmaceutical ChemistryFaculty of PharmacyUniversity of BelgradeVojvode Stepe 45011221BelgradeSerbia
| |
Collapse
|
262
|
Santana FPR, Thevenard F, Gomes KS, Taguchi L, Câmara NOS, Stilhano RS, Ureshino RP, Prado CM, Lago JHG. New perspectives on natural flavonoids on COVID-19-induced lung injuries. Phytother Res 2021; 35:4988-5006. [PMID: 33928690 PMCID: PMC8242604 DOI: 10.1002/ptr.7131] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/11/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022]
Abstract
The SARS-CoV-2 virus, responsible for COVID-19, spread rapidly worldwide and became a pandemic in 2020. In some patients, the virus remains in the respiratory tract, causing pneumonia, respiratory failure, acute respiratory distress syndrome (ARDS), and sepsis, leading to death. Natural flavonoids (aglycone and glycosides) possess broad biological activities encompassing antiinflammatory, antiviral, antitumoral, antiallergic, antiplatelet, and antioxidant effects. While many studies have focused on the effects of natural flavonoids in experimental models, reports based on clinical trials are still insufficient. In this review, we highlight the effects of flavonoids in controlling pulmonary diseases, particularly the acute respiratory distress syndrome, a consequence of COVID-19, and their potential use in coronavirus-related diseases. Furthermore, we also focus on establishing a relationship between biological potential and chemical aspects of related flavonoids and discuss several possible mechanisms of action, pointing out some possible effects on COVID-19.
Collapse
Affiliation(s)
- Fernanda Paula R Santana
- Department of Bioscience, Federal University of São Paulo, Santos, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Fernanda Thevenard
- Center of Natural Sciences and Humanities, Federal University of ABC, São Paulo, São Paulo, Brazil
| | - Kaio S Gomes
- Center of Natural Sciences and Humanities, Federal University of ABC, São Paulo, São Paulo, Brazil
| | - Laura Taguchi
- Department of Bioscience, Federal University of São Paulo, Santos, São Paulo, Brazil
| | - Niels Olsen S Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Roberta S Stilhano
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, São Paulo, Brazil
| | - Rodrigo P Ureshino
- Department of Biological Science, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Carla Maximo Prado
- Department of Bioscience, Federal University of São Paulo, Santos, São Paulo, Brazil
| | | |
Collapse
|
263
|
Premkumar M, Kedarisetty CK. Cytokine Storm of COVID-19 and Its Impact on Patients with and without Chronic Liver Disease. J Clin Transl Hepatol 2021; 9:256-264. [PMID: 34007808 PMCID: PMC8111101 DOI: 10.14218/jcth.2021.00055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/11/2021] [Accepted: 04/01/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus pandemic has resulted in increased rates of hepatic decompensation, morbidity and mortality in patients suffering from existing liver disease, and deranged liver biochemistries in those without liver disease. In patients with cirrhosis with coronavirus disease 2019 (COVID-19), new onset organ failures manifesting as acute-on-chronic liver failure have also been reported. The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) also directly binds to enterocytes and cholangiocytes via the angiotensin converting enzyme receptor 2, although the lung remains the portal of entry. Superadded with the COVID-19 related bystander hepatitis, a systemic inflammatory response is noted due to unregulated macrophage activation syndrome and cytokine storm. However, the exact definition and diagnostic criteria of the 'cytokine storm' in COVID-19 are yet unclear. In addition, inflammatory markers like C-reactive protein, ferritin, D-dimer and procalcitonin are frequently elevated. This in turn leads to disease progression, activation of the coagulation cascade, vascular microthrombi and immune-mediated injury in different organ systems. Deranged liver chemistries are also noted due to the cytokine storm, and synergistic hypoxic or ischemic liver injury, drug-induced liver injury, and use of hepatotoxic antiviral agents all contribute to deranged liver chemistry. Control of an unregulated cytokine storm at an early stage may avert disease morbidity and mortality. Several immunomodulator drugs and repurposed immunosuppressive agents have been used in COVID-19 with varying degrees of success.
Collapse
Affiliation(s)
- Madhumita Premkumar
- Departments of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | |
Collapse
|
264
|
Li P, Wu W, Zhang T, Wang Z, Li J, Zhu M, Liang Y, You W, Li K, Ding R, Huang B, Wu L, Duan W, Han Y, Li X, Tang X, Wang X, Shen H, Wang Q, Yan H, Xia X, Ji Y, Chen H. Implications of cardiac markers in risk-stratification and management for COVID-19 patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2021; 25:158. [PMID: 33902676 PMCID: PMC8074282 DOI: 10.1186/s13054-021-03555-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/30/2021] [Indexed: 01/08/2023]
Abstract
Background COVID-19 has resulted in high mortality worldwide. Information regarding cardiac markers for precise risk-stratification is limited. We aim to discover sensitive and reliable early-warning biomarkers for optimizing management and improving the prognosis of COVID-19 patients. Methods A total of 2954 consecutive COVID-19 patients who were receiving treatment from the Wuhan Huoshenshan Hospital in China from February 4 to April 10 were included in this retrospective cohort. Serum levels of cardiac markers were collected after admission. Coronary artery disease diagnosis and survival status were recorded. Single-cell RNA-sequencing and bulk RNA-sequencing from different cohorts of non-COVID-19 were performed to analyze SARS-CoV-2 receptor expression. Results Among 2954 COVID-19 patients in the analysis, the median age was 60 years (50–68 years), 1461 (49.5%) were female, and 1515 (51.3%) were severe/critical. Compared to mild/moderate (1439, 48.7%) patients, severe/critical patients showed significantly higher levels of cardiac markers within the first week after admission. In severe/critical COVID-19 patients, those with abnormal serum levels of BNP (42 [24.6%] vs 7 [1.1%]), hs-TNI (38 [48.1%] vs 6 [1.0%]), α- HBDH (55 [10.4%] vs 2 [0.2%]), CK-MB (45 [36.3%] vs 12 [0.9%]), and LDH (56 [12.5%] vs 1 [0.1%]) had a significantly higher mortality rate compared to patients with normal levels. The same trend was observed in the ICU admission rate. Severe/critical COVID-19 patients with pre-existing coronary artery disease (165/1,155 [10.9%]) had more cases of BNP (52 [46.5%] vs 119 [16.5%]), hs-TNI (24 [26.7%] vs 9.6 [%], α- HBDH (86 [55.5%] vs 443 [34.4%]), CK-MB (27 [17.4%] vs 97 [7.5%]), and LDH (65 [41.9%] vs 382 [29.7%]), when compared with those without coronary artery disease. There was enhanced SARS-CoV-2 receptor expression in coronary artery disease compared with healthy controls. From regression analysis, patients with five elevated cardiac markers were at a higher risk of death (hazards ratio 3.4 [95% CI 2.4–4.8]). Conclusions COVID-19 patients with pre-existing coronary artery disease represented a higher abnormal percentage of cardiac markers, accompanied by high mortality and ICU admission rate. BNP together with hs-TNI, α- HBDH, CK-MB and LDH act as a prognostic biomarker in COVID-19 patients with or without pre-existing coronary artery disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13054-021-03555-z.
Collapse
Affiliation(s)
- Pengping Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Wei Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Tingting Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Ziyu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Jie Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Yuan Liang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Wenhua You
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Rong Ding
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Weiwei Duan
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Yi Han
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuesong Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xin Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xin Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Hong Yan
- Laboratory Medicine Center, the Second Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| | - Xinyi Xia
- COVID-19 Research Center, Institute of Laboratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China. .,Department of Laboratory Medicine & Blood Transfusion, Wuhan Huoshenshan Hospital, Wuhan, China. .,Joint Expert Group for COVID-19, Wuhan Huoshenshan Hospital, Wuhan, China.
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China. .,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China. .,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.
| | - Hongshan Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China. .,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China. .,Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
265
|
Orlando V, Coscioni E, Guarino I, Mucherino S, Perrella A, Trama U, Limongelli G, Menditto E. Drug-utilisation profiles and COVID-19. Sci Rep 2021; 11:8913. [PMID: 33903671 PMCID: PMC8076316 DOI: 10.1038/s41598-021-88398-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has substantially challenged healthcare systems worldwide. By investigating population characteristics and prescribing profiles, it is possible to generate hypotheses about the associations between specific drug-utilisation profiles and susceptibility to COVID-19 infection. A retrospective drug-utilisation study was carried out using routinely collected information from a healthcare database in Campania (Southern Italy). We aimed to discover the prevalence of drug utilisation (monotherapy and polytherapy) in COVID-19 versus non-COVID-19 patients in Campania (~ 6 million inhabitants). The study cohort comprised 1532 individuals who tested positive for COVID-19. Drugs were grouped according to the Anatomical Therapeutic Chemical (ATC) classification system. We noted higher prevalence rates of the use of drugs in the ATC categories C01, B01 and M04, which was probably linked to related comorbidities (i.e., cardiovascular and metabolic). Nevertheless, the prevalence of the use of drugs acting on the renin-angiotensin system, such as antihypertensive drugs, was not higher in COVID-19 patients than in non-COVID-19 patients after adjustments for age and sex. These results highlight the need for further case-control studies to define the effects of medications and comorbidities on susceptibility to and associated mortality from COVID-19.
Collapse
Affiliation(s)
- Valentina Orlando
- CIRFF, Center of Drug Utilisation and Pharmacoeconomics, University of Naples Federico II, 80131, Naples, Italy.
- Department of Pharmacy, Center of Drug Utilisation and Pharmacoeconomics, University of Naples Federico II, 80131, Naples, Italy.
| | - Enrico Coscioni
- Division of Cardiac Surgery, AOU San Giovanni di Dio E Ruggi d'Aragona, 84131, Salerno, Italy
| | - Ilaria Guarino
- CIRFF, Center of Drug Utilisation and Pharmacoeconomics, University of Naples Federico II, 80131, Naples, Italy
| | - Sara Mucherino
- CIRFF, Center of Drug Utilisation and Pharmacoeconomics, University of Naples Federico II, 80131, Naples, Italy
- Department of Pharmacy, Center of Drug Utilisation and Pharmacoeconomics, University of Naples Federico II, 80131, Naples, Italy
| | - Alessandro Perrella
- Infectious Disease of Healthcare Direction, AORN Antonio Cardarelli, 80131, Naples, Italy
| | - Ugo Trama
- Regional Pharmaceutical Unit, Campania Region, 80143, Naples, Italy
| | - Giuseppe Limongelli
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Monaldi Hospital, 80131, Naples, Italy
| | - Enrica Menditto
- CIRFF, Center of Drug Utilisation and Pharmacoeconomics, University of Naples Federico II, 80131, Naples, Italy.
- Department of Pharmacy, Center of Drug Utilisation and Pharmacoeconomics, University of Naples Federico II, 80131, Naples, Italy.
| |
Collapse
|
266
|
Medvedev VE. Anxiety and depression in COVID-19: treatment options. NEUROLOGY, NEUROPSYCHIATRY, PSYCHOSOMATICS 2021. [DOI: 10.14412/2074-2711-2021-2-111-116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- V. E. Medvedev
- Department of Psychiatry, Psychotherapy, and Psychosomatic Pathology, Faculty of Advanced Training of Medical Workers, Medical Institute, Peoples' Friendship University of Russia (RUDN University)
| |
Collapse
|
267
|
Derington CG, Cohen JB, Mohanty AF, Greene TH, Cook J, Ying J, Wei G, Herrick JS, Stevens VW, Jones BE, Wang L, Zheutlin AR, South AM, Hanff TC, Smith SM, Cooper-DeHoff RM, King JB, Alexander GC, Berlowitz DR, Ahmad FS, Penrod MJ, Hess R, Conroy MB, Fang JC, Rubin MA, Beddhu S, Cheung AK, Xian W, Weintraub WS, Bress AP. Angiotensin II receptor blocker or angiotensin-converting enzyme inhibitor use and COVID-19-related outcomes among US Veterans. PLoS One 2021; 16:e0248080. [PMID: 33891615 PMCID: PMC8064574 DOI: 10.1371/journal.pone.0248080] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/19/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Angiotensin II receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs) may positively or negatively impact outcomes in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We investigated the association of ARB or ACEI use with coronavirus disease 2019 (COVID-19)-related outcomes in US Veterans with treated hypertension using an active comparator design, appropriate covariate adjustment, and negative control analyses. METHODS AND FINDINGS In this retrospective cohort study of Veterans with treated hypertension in the Veterans Health Administration (01/19/2020-08/28/2020), we compared users of (A) ARB/ACEI vs. non-ARB/ACEI (excluding Veterans with compelling indications to reduce confounding by indication) and (B) ARB vs. ACEI among (1) SARS-CoV-2+ outpatients and (2) COVID-19 hospitalized inpatients. The primary outcome was all-cause hospitalization or mortality (outpatients) and all-cause mortality (inpatients). We estimated hazard ratios (HR) using propensity score-weighted Cox regression. Baseline characteristics were well-balanced between exposure groups after weighting. Among outpatients, there were 5.0 and 6.0 primary outcomes per 100 person-months for ARB/ACEI (n = 2,482) vs. non-ARB/ACEI (n = 2,487) users (HR 0.85, 95% confidence interval [CI] 0.73-0.99, median follow-up 87 days). Among outpatients who were ARB (n = 4,877) vs. ACEI (n = 8,704) users, there were 13.2 and 14.8 primary outcomes per 100 person-months (HR 0.91, 95%CI 0.86-0.97, median follow-up 85 days). Among inpatients who were ARB/ACEI (n = 210) vs. non-ARB/ACEI (n = 275) users, there were 3.4 and 2.0 all-cause deaths per 100 person months (HR 1.25, 95%CI 0.30-5.13, median follow-up 30 days). Among inpatients, ARB (n = 1,164) and ACEI (n = 2,014) users had 21.0 vs. 17.7 all-cause deaths, per 100 person-months (HR 1.13, 95%CI 0.93-1.38, median follow-up 30 days). CONCLUSIONS This observational analysis supports continued ARB or ACEI use for patients already using these medications before SARS-CoV-2 infection. The novel beneficial association observed among outpatients between users of ARBs vs. ACEIs on hospitalization or mortality should be confirmed with randomized trials.
Collapse
Affiliation(s)
- Catherine G. Derington
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Jordana B. Cohen
- Department of Medicine, Renal-Electrolyte and Hypertension Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - April F. Mohanty
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Tom H. Greene
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - James Cook
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Jian Ying
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
| | - Guo Wei
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
| | - Jennifer S. Herrick
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
| | - Vanessa W. Stevens
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
| | - Barbara E. Jones
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Libo Wang
- Department of Medicine, Division of Cardiology, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Alexander R. Zheutlin
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Andrew M. South
- Department of Pediatrics, Section of Nephrology, Brenner Children’s Hospital, Wake Forest School of Medicine, Winston Salem, NC, United States of America
- Division of Public Health Sciences, Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston Salem, NC, United States of America
| | - Thomas C. Hanff
- Department of Medicine, Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Steven M. Smith
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States of America
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, United States of America
- Department of Medicine, University of Florida, College of Medicine, Gainesville, FL, United States of America
| | - Jordan B. King
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO, United States of America
| | - G. Caleb Alexander
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Dan R. Berlowitz
- Department of Public Health; University of Massachusetts Lowell, Lowell, MA, United States of America
- Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States of America
| | - Faraz S. Ahmad
- Department of Medicine, Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - M. Jason Penrod
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Rachel Hess
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Molly B. Conroy
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - James C. Fang
- Department of Medicine, Division of Cardiology, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Michael A. Rubin
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Srinivasan Beddhu
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Alfred K. Cheung
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Weiming Xian
- Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States of America
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States of America
| | | | - Adam P. Bress
- Department of Population Health Sciences, Division of Health System Innovation and Research, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, United States of America
| |
Collapse
|
268
|
Mascolo A, Scavone C, Rafaniello C, De Angelis A, Urbanek K, di Mauro G, Cappetta D, Berrino L, Rossi F, Capuano A. The Role of Renin-Angiotensin-Aldosterone System in the Heart and Lung: Focus on COVID-19. Front Pharmacol 2021; 12:667254. [PMID: 33959029 PMCID: PMC8093861 DOI: 10.3389/fphar.2021.667254] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
The renin-angiotensin-aldosterone system (RAAS) firstly considered as a cardiovascular circulating hormonal system, it is now accepted as a local tissue system that works synergistically or independently with the circulating one. Evidence states that tissue RAAS locally generates mediators with regulatory homeostatic functions, thus contributing, at some extent, to organ dysfunction or disease. Specifically, RAAS can be divided into the traditional RAAS pathway (or classic RAAS) mediated by angiotensin II (AII), and the non-classic RAAS pathway mediated by angiotensin 1–7. Both pathways operate in the heart and lung. In the heart, the classic RAAS plays a role in both hemodynamics and tissue remodeling associated with cardiomyocyte and endothelial dysfunction, leading to progressive functional impairment. Moreover, the local classic RAAS may predispose the onset of atrial fibrillation through different biological mechanisms involving inflammation, accumulation of epicardial adipose tissue, and electrical cardiac remodeling. In the lung, the classic RAAS regulates cell proliferation, immune-inflammatory response, hypoxia, and angiogenesis, contributing to lung injury and different pulmonary diseases (including COVID-19). Instead, the local non-classic RAAS counteracts the classic RAAS effects exerting a protective action on both heart and lung. Moreover, the non-classic RAAS, through the angiotensin-converting enzyme 2 (ACE2), mediates the entry of the etiological agent of COVID-19 (SARS-CoV-2) into cells. This may cause a reduction in ACE2 and an imbalance between angiotensins in favor of AII that may be responsible for the lung and heart damage. Drugs blocking the classic RAAS (angiotensin-converting enzyme inhibitors and angiotensin receptor blockers) are well known to exert a cardiovascular benefit. They are recently under evaluation for COVID-19 for their ability to block AII-induced lung injury altogether with drugs stimulating the non-classic RAAS. Herein, we discuss the available evidence on the role of RAAS in the heart and lung, summarizing all clinical data related to the use of drugs acting either by blocking the classic RAAS or stimulating the non-classic RAAS.
Collapse
Affiliation(s)
- Annamaria Mascolo
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy.,Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Cristina Scavone
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy.,Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Concetta Rafaniello
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy.,Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy.,Department of Experimental and Clinical Medicine, Molecular and Cellular Cardiology, Magna Graecia University, Catanzaro, Italy
| | - Gabriella di Mauro
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy.,Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Rossi
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy.,Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annalisa Capuano
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy.,Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
269
|
Zhan Y, Ta W, Tang W, Hua R, Wang J, Wang C, Lu W. Potential antiviral activity of isorhamnetin against SARS-CoV-2 spike pseudotyped virus in vitro. Drug Dev Res 2021; 82:1124-1130. [PMID: 33847382 PMCID: PMC8251057 DOI: 10.1002/ddr.21815] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus Disease 2019 (COVID-19) cases and deaths are still rising worldwide, there is currently no effective treatment for severe inflammation and acute lung injury caused by new coronavirus (SARS-COV-2) infection. Therapies to prevent or treat COVID-19, including antiviral drug and several vaccines, are still being development. Human angiotensin-converting enzyme 2 (ACE2), expressing in lung, has been confirmed to be a receptor for SARS-COV-2 infection, interventions for attachment of spike protein of SARS-CoV-2 to ACE2 may be a potential approach to prevent viral infections and it is considered as a potential target for drug development. In this study, we observed that seabuckthorn and its flavonoid compounds quercetin and isorhamnetin were shown strong retention to ACE2 overexpression HEK293 (ACE2h ) cells by CMC analysis. Based on drug receptor interaction analysis and viral entry studies in vitro, we evaluated the interaction of two flavonoid compounds and ACE2 as well as the inhibitory effect of the two compounds on viral entry. Surface plasmon resonance assay proved the effect that isorhamnetin bound to the ACE2, and its affinity (KD value) was at the micromolar level, that was, 2.51 ± 0.68 μM. Viral entry studies in vitro indicated that isorhamnetin inhibited SARS-CoV-2 spike pseudotyped virus entering ACE2h cells. Based on promising in vitro results, we proposed isorhamnetin to be a potential therapeutic candidate compound against COVID-19.
Collapse
Affiliation(s)
- Yingzhuan Zhan
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Wenjing Ta
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Wenjuan Tang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Ruochen Hua
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Jue Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Cheng Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Wen Lu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
270
|
Wang Y, Zheng J, Islam MS, Yang Y, Hu Y, Chen X. The role of CD4 +FoxP3 + regulatory T cells in the immunopathogenesis of COVID-19: implications for treatment. Int J Biol Sci 2021; 17:1507-1520. [PMID: 33907514 PMCID: PMC8071774 DOI: 10.7150/ijbs.59534] [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: 02/18/2021] [Accepted: 03/15/2021] [Indexed: 01/08/2023] Open
Abstract
The severe cases of Coronavirus Disease 2019 (COVID-19) frequently exhibit excessive inflammatory responses, acute respiratory distress syndrome (ARDS), coagulopathy, and organ damage. The most striking immunopathology of advanced COVID-19 is cytokine release syndrome or "cytokine storm" that is attributable to the deficiencies in immune regulatory mechanisms. CD4+FoxP3+ regulatory T cells (Tregs) are central regulators of immune responses and play an indispensable role in the maintenance of immune homeostasis. Tregs are likely involved in the attenuation of antiviral defense at the early stage of infection and ameliorating inflammation-induced organ injury at the late stage of COVID-19. In this article, we review and summarize the current understanding of the change of Tregs in patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and discuss the potential role of Tregs in the immunopathology of COVID-19. The emerging concept of Treg-targeted therapies, including both adoptive Treg transfer and low dose of IL-2 treatment, is introduced. Furthermore, the potential Treg-boosting effect of therapeutic agents used in the treatment of COVID-19, including dexamethasone, vitamin D, tocilizumab and sarilumab, chloroquine, hydroxychloroquine, azithromycin, adalimumab and tetrandrine, is discussed. The problems in the current study of Treg cells in COVID-19 and future perspectives are also addressed.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| |
Collapse
|
271
|
Saavedra LPJ, Prates KV, Gonçalves GD, Piovan S, Matafome P, Mathias PCDF. COVID-19 During Development: A Matter of Concern. Front Cell Dev Biol 2021; 9:659032. [PMID: 33898461 PMCID: PMC8058409 DOI: 10.3389/fcell.2021.659032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
A new infectious disease, COVID-19, has spread around the world. The most common symptoms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are cough and fever, but severe cases can develop acute respiratory distress syndrome. The main receptor for SARS-CoV-2 in human tissue is angiotensin-converting enzyme 2, and the lungs, heart, and kidneys are the most affected organs. Besides the inflammatory process and tissue damage, the presence of a cytokine "storm" has been related to a higher mortality rate. Other infectious viral diseases, such as Zika, chikungunya, and influenza, were associated with complications in pregnant women, such as growth restriction, malformation, preterm birth, low birth weight, miscarriage, and death, although they can also cause developmental disorders in infants and adolescents. Evidence points out that stressors during pregnancy and infancy may lead to the development of obesity, diabetes, and cardiovascular disease. Therefore, we hypothesize that COVID-19 infection during the critical phases of development can program the individual to chronic diseases in adulthood. It is important that COVID-19 patients receive proper monitoring as a way to avoid expensive costs to public health in the future.
Collapse
Affiliation(s)
- Lucas Paulo Jacinto Saavedra
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Kelly Valério Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Gessica Dutra Gonçalves
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Silvano Piovan
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| | - Paulo Matafome
- Institute of Physiology and Institute of Clinical and Biomedical Research, Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Coimbra Health School, ESTeSC, Instituto Politécnico de Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Brazil
| |
Collapse
|
272
|
Cao Q, Lei H, Yang M, Wei L, Dong Y, Xu J, Nasser M, Liu M, Zhu P, Xu L, Zhao M. Impact of Cardiovascular Diseases on COVID-19: A Systematic Review. Med Sci Monit 2021; 27:e930032. [PMID: 33820904 PMCID: PMC8035813 DOI: 10.12659/msm.930032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In December 2019, pneumonia of unknown cause broke out, and currently more than 150 countries around the world have been affected. Globally, as of 5: 46 pm CET, 6 November 2020, the World Health Organization (WHO) had reported 48 534 508 confirmed cases of COVID-19, including 1 231 017 deaths. The novel coronavirus disease (COVID-19) outbreak, caused by the SARS-CoV-2 virus, is the most important medical challenge in decades. Previous research mainly focused on the exploration of lung changes. However, with development of the disease and deepening research, more and more patients showed cardiovascular diseases, even in those without respiratory symptoms, and some researchers have found that underlying cardiovascular diseases increase the risk of infection. Although the related mechanism is not thoroughly studied, based on existing research, we speculate that the interaction between the virus and its receptor, inflammatory factors, various forms of the stress response, hypoxic environment, and drug administration could all induce the development of cardiac adverse events. Interventions to control these pathogenic factors may effectively reduce the occurrence of cardiovascular complications. This review summarizes the latest research on the relationship between COVID-19 and its associated cardiovascular complications, and we also explore possible mechanisms and treatments.
Collapse
Affiliation(s)
- Qingtai Cao
- Hunan Normal University School of Medicine, Changsha, Hunan, China (mainland)
| | - HanYu Lei
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - MengLing Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Le Wei
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - YinMiao Dong
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - JiaHao Xu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Mi Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China (mainland)
| | - MengQi Liu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China (mainland)
| | - LinYong Xu
- School of Life Science, Central South University, Changsha, Hunan, China (mainland)
| | - MingYi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
273
|
The contrasting role of nasopharyngeal angiotensin converting enzyme 2 (ACE2) transcription in SARS-CoV-2 infection: A cross-sectional study of people tested for COVID-19 in British Columbia, Canada. EBioMedicine 2021; 66:103316. [PMID: 33819740 PMCID: PMC8016616 DOI: 10.1016/j.ebiom.2021.103316] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background Angiotensin converting enzyme 2 (ACE2) protein serves as the host receptor for SARS-CoV-2, with a critical role in viral infection. We aim to understand population level variation of nasopharyngeal ACE2 transcription in people tested for COVID-19 and the relationship between ACE2 transcription and SARS-CoV-2 viral load, while adjusting for expression of: (i) the complementary protease, Transmembrane serine protease 2 (TMPRSS2), (ii) soluble ACE2, (iii) age, and (iv) biological sex. The ACE2 gene was targeted to measure expression of transmembrane and soluble transcripts. Methods A cross-sectional study of n = 424 “participants” aged 1–104 years referred for COVID-19 testing was performed in British Columbia, Canada. Patients who tested positive for COVID-19 were matched by age and biological sex to patients who tested negative. Viral load and host gene expression were assessed by quantitative reverse-transcriptase polymerase chain reaction. Bivariate analysis and multiple linear regression were performed to understand the role of nasopharyngeal ACE2 expression in SARS-CoV-2 infection. Findings Analysis showed no association between age and nasopharyngeal ACE2 transcription in those who tested negative for COVID-19 (P = 0•092). Mean relative transcription of transmembrane (P = 0•00012) and soluble (P<0•0001) ACE2 isoforms, as well as TMPRSS2 (P<0•0001) was higher in COVID-19-negative participants than COVID--19 positive ones, yielding a negative correlation between targeted host gene expression and positive COVID-19 diagnosis. In bivariate analysis of COVID-19-positive participants, transcription of transmembrane ACE2 positively correlated with SARS-CoV-2 viral RNA load (B = 0•49, R2=0•14, P<0•0001), transcription of soluble ACE2 negatively correlated (B= -0•85, R2= 0•26, P<0•0001), and no correlation was found with TMPRSS2 transcription (B= -0•042, R2=<0•10, P = 0•69). Multivariable analysis showed that the greatest viral RNA loads were observed in participants with high transmembrane ACE2 transcription (Β= 0•89, 95%CI: [0•59 to 1•18]), while transcription of the soluble isoform appears to protect against high viral RNA load in the upper respiratory tract (Β= -0•099, 95%CI: [-0•18 to -0•022]). Interpretation Nasopharyngeal ACE2 transcription plays a dual, contrasting role in SARS-CoV-2 infection of the upper respiratory tract. Transcription of the transmembrane ACE2 isoform positively correlates, while transcription of the soluble isoform negatively correlates with viral RNA load after adjusting for age, biological sex, and transcription of TMPRSS2. Funding This project (COV-55) was funded by Genome British Columbia as part of their COVID-19 rapid response initiative.
Collapse
|
274
|
Affiliation(s)
- Mark C Chappell
- Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Correspondence: Mark C. Chappell, PhD, Cardiovascular Sciences Center, Wake Forest University School of Medicine, 575 Patterson Ave, Biotech Pl, Winston Salem, NC, 27101 USA.
| |
Collapse
|
275
|
Okpechi SC, Fong JT, Gill SS, Harman JC, Nguyen TH, Chukwurah QC, Onor IO, Alahari SK. Global Sex Disparity of COVID-19: A Descriptive Review of Sex Hormones and Consideration for the Potential Therapeutic Use of Hormone Replacement Therapy in Older Adults. Aging Dis 2021; 12:671-683. [PMID: 33815890 PMCID: PMC7990361 DOI: 10.14336/ad.2020.1211] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
The 2019-2020 SARS-related coronavirus-2 (SARS-CoV-2) pandemic has brought unprecedented challenges to healthcare sectors around the world. As of November 2020, there have been over 64 million confirmed cases and approaching 2 million deaths globally. Despite the large number of positive cases, there are very limited established standards of care and therapeutic options available. To date, there is still no Food and Drug Administration (FDA) approved vaccine for COVID-19, although there are several options in various clinical trial stages. Herein, we have performed a global review evaluating the roles of age and sex on COVID-19 hospitalizations, ICU admissions, deaths in hospitals, and deaths in nursing homes. We have identified a trend in which elderly and male patients are significantly affected by adverse outcomes. There is evidence suggesting that sex hormone levels can influence immune system function against SARS-CoV-2 infection, thus reducing the adverse effects of COVID-19. Since older adults have lower levels of these sex hormones, we therefore speculate, within rational scientific context, that sex steroids, such as estrogen and progesterone, needs further consideration for use as alternative therapeutic option for treating COVID-19 elderly patients. To our knowledge, this is the first comprehensive article evaluating the significance of sex hormones in COVID-19 outcomes in older adults.
Collapse
Affiliation(s)
- Samuel C Okpechi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Jordyn T Fong
- Department of Biological Sciences, College of Science, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Shawn S Gill
- Ochsner Clinical School, University of Queensland, Brisbane, Queensland, Australia.
| | - Jarrod C Harman
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Tina H Nguyen
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | | | - IfeanyiChukwu O Onor
- College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
- Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
276
|
Cohen JB, D'Agostino McGowan L, Jensen ET, Rigdon J, South AM. Evaluating sources of bias in observational studies of angiotensin-converting enzyme inhibitor/angiotensin II receptor blocker use during COVID-19: beyond confounding. J Hypertens 2021; 39:795-805. [PMID: 33186321 PMCID: PMC8164085 DOI: 10.1097/hjh.0000000000002706] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Concerns over ACE inhibitor or ARB use to treat hypertension during COVID-19 remain unresolved. Although studies using more robust methodologies provided some clarity, sources of bias persist and it remains critical to quickly address this question. In this review, we discuss pernicious sources of bias using a causal model framework, including time-varying confounder, collider, information, and time-dependent bias, in the context of recently published studies. We discuss causal inference methodologies that can address these issues, including causal diagrams, time-to-event analyses, sensitivity analyses, and marginal structural modeling. We discuss effect modification and we propose a role for causal mediation analysis to estimate indirect effects via mediating factors, especially components of the renin--angiotensin system. Thorough knowledge of these sources of bias and the appropriate methodologies to address them is crucial when evaluating observational studies to inform patient management decisions regarding whether ACE inhibitors or ARBs are associated with greater risk from COVID-19.
Collapse
Affiliation(s)
- Jordana B Cohen
- Renal-Electrolyte and Hypertension Division, Department of Medicine
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Elizabeth T Jensen
- Department of Epidemiology and Prevention, Division of Public Health Sciences
| | - Joseph Rigdon
- Department of Biostatistics and Data Science, Division of Public Health Sciences
| | - Andrew M South
- Department of Epidemiology and Prevention, Division of Public Health Sciences
- Section of Nephrology, Department of Pediatrics, Brenner Children's Hospital
- Department of Surgery-Hypertension & Vascular Research
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
277
|
Lionetti V, Bollini S, Coppini R, Gerbino A, Ghigo A, Iaccarino G, Madonna R, Mangiacapra F, Miragoli M, Moccia F, Munaron L, Pagliaro P, Parenti A, Pasqua T, Penna C, Quaini F, Rocca C, Samaja M, Sartiani L, Soda T, Tocchetti CG, Angelone T. Understanding the heart-brain axis response in COVID-19 patients: A suggestive perspective for therapeutic development. Pharmacol Res 2021; 168:105581. [PMID: 33781873 PMCID: PMC7997688 DOI: 10.1016/j.phrs.2021.105581] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/19/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
In-depth characterization of heart-brain communication in critically ill patients with severe acute respiratory failure is attracting significant interest in the COronaVIrus Disease 19 (COVID-19) pandemic era during intensive care unit (ICU) stay and after ICU or hospital discharge. Emerging research has provided new insights into pathogenic role of the deregulation of the heart-brain axis (HBA), a bidirectional flow of information, in leading to severe multiorgan disease syndrome (MODS) in patients with confirmed infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Noteworthy, HBA dysfunction may worsen the outcome of the COVID-19 patients. In this review, we discuss the critical role HBA plays in both promoting and limiting MODS in COVID-19. We also highlight the role of HBA as new target for novel therapeutic strategies in COVID-19 in order to open new translational frontiers of care. This is a translational perspective from the Italian Society of Cardiovascular Researches.
Collapse
Affiliation(s)
- Vincenzo Lionetti
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; UOSVD Anesthesia and Intensive Care, Fondazione Toscana G. Monasterio, Pisa, Italy.
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Raffaele Coppini
- Department of NEUROFARBA, Center of Molecular Medicine, University of Firenze, 50139 Firenze, Italy
| | - Andrea Gerbino
- Department of Bioscience, Biotechnology and Biopharmaceuticals, University of Bari, Bari, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, Federico II University, Italy
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy; Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fabio Mangiacapra
- Unit of Cardiovascular Science, Campus Bio-Medico University, Rome, Italy
| | - Michele Miragoli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology, Laboratory of General Physiology, University of Pavia, Pavia, Italy.
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Pasquale Pagliaro
- Clinical and Biological Sciences Department, University of Turin, Orbassano, Turin, Italy
| | - Astrid Parenti
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Teresa Pasqua
- Department of Health Science, University of Magna Graecia, Catanzaro, Italy
| | - Claudia Penna
- Clinical and Biological Sciences Department, University of Turin, Orbassano, Turin, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, E. and E.S., University of Calabria, Arcavacata di Rende, CS, Italy
| | - Michele Samaja
- Department of Health Science, University of Milano, Milan, Italy
| | - Laura Sartiani
- Department of NEUROFARBA, Center of Molecular Medicine, University of Firenze, 50139 Firenze, Italy
| | - Teresa Soda
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Carlo Gabriele Tocchetti
- Interdepartmental Center of Clinical and Translational Research, Federico II University, Naples, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, E. and E.S., University of Calabria, Arcavacata di Rende, CS, Italy
| |
Collapse
|
278
|
ACE2 as therapeutic agent. Clin Sci (Lond) 2021; 134:2581-2595. [PMID: 33063820 DOI: 10.1042/cs20200570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
The angiotensin-converting enzyme 2 (ACE2) has emerged as a critical regulator of the renin-angiotensin system (RAS), which plays important roles in cardiovascular homeostasis by regulating vascular tone, fluid and electrolyte balance. ACE2 functions as a carboxymonopeptidase hydrolyzing the cleavage of a single C-terminal residue from Angiotensin-II (Ang-II), the key peptide hormone of RAS, to form Angiotensin-(1-7) (Ang-(1-7)), which binds to the G-protein-coupled Mas receptor and activates signaling pathways that counteract the pathways activated by Ang-II. ACE2 is expressed in a variety of tissues and overwhelming evidence substantiates the beneficial effects of enhancing ACE2/Ang-(1-7)/Mas axis under many pathological conditions in these tissues in experimental models. This review will provide a succinct overview on current strategies to enhance ACE2 as therapeutic agent, and discuss limitations and future challenges. ACE2 also has other functions, such as acting as a co-factor for amino acid transport and being exploited by the severe acute respiratory syndrome coronaviruses (SARS-CoVs) as cellular entry receptor, the implications of these functions in development of ACE2-based therapeutics will also be discussed.
Collapse
|
279
|
Kaseb AO, Mohamed YI, Malek AE, Raad II, Altameemi L, Li D, Kaseb OA, Kaseb SA, Selim A, Ma Q. The Impact of Angiotensin-Converting Enzyme 2 (ACE2) Expression on the Incidence and Severity of COVID-19 Infection. Pathogens 2021; 10:379. [PMID: 33809851 PMCID: PMC8004186 DOI: 10.3390/pathogens10030379] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 01/08/2023] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic has led to an unprecedented threat to the international community and raised major concerns in terms of public health safety. Although our current understanding of the complexity of COVID-19 pathogenesis remains limited, the infection is largely mediated by the interaction of viral spike protein and angiotensin-converting enzyme 2 (ACE2). The functional importance of ACE2 in different demographic and comorbid conditions may explain the significant variation in incidence and mortality of COVID-19 in vulnerable groups, and highlights its candidacy as a potential therapeutic target. We provide evidence supporting the idea that differences in incidence and severity of COVID-19 infection may be related to ACE2. Emerging data based on the prevalence and severity of COVID-19 among those with established high levels of ACE2 expression strongly support our hypothesis. Considering the burden of COVID-19 infection in these vulnerable groups and the impact of the potential therapeutic and preventive measures that would result from adopting ACE2-driven anti-viral strategies, our hypothesis may expedite global efforts to control the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Ahmed O. Kaseb
- Departments of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.I.M.); (L.A.)
| | - Yehia I. Mohamed
- Departments of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.I.M.); (L.A.)
| | - Alexandre E. Malek
- Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.E.M.); (I.I.R.)
| | - Issam I. Raad
- Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.E.M.); (I.I.R.)
| | - Lina Altameemi
- Departments of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.I.M.); (L.A.)
| | - Dan Li
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.L.); (Q.M.)
| | - Omar A. Kaseb
- Iman Academy, Webster, TX 77598, USA; (O.A.K.); (S.A.K.)
| | - Safa A. Kaseb
- Iman Academy, Webster, TX 77598, USA; (O.A.K.); (S.A.K.)
| | - Abdelhafez Selim
- Philadelphia College of Osteopathic Medicine (PCOM), Philadelphia, PA 19131, USA;
| | - Qing Ma
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.L.); (Q.M.)
| |
Collapse
|
280
|
Kohler H, Bäuerle A, Schweda A, Weismüller B, Fink M, Musche V, Robitzsch A, Pfeiffer C, Benecke AV, Dörrie N, Führer D, Taube C, Rassaf T, Teufel M, Skoda EM. Increased COVID-19-related fear and subjective risk perception regarding COVID-19 affects behavior in individuals with internal high-risk diseases. J Prim Care Community Health 2021; 12:2150132721996898. [PMID: 33719697 PMCID: PMC8851367 DOI: 10.1177/2150132721996898] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Since December 2019, the coronavirus disease-2019 (COVID-19) has been keeping the world in suspense. Proven risk factors for a severe course of COVID-19 are common diseases like diabetes, hypertension, cardiovascular or respiratory disorders. Until today, little is known about the psychological burden of individuals suffering from these high-risk diseases regard to COVID-19. The aim of the study was to define the impact of the coronavirus pandemic on behavior and mental health in individuals at high risk for developing a severe COVID-19 course. Items assessed generalized anxiety (GAD-7), COVID-19-related fear, adherent/dysfunctional safety behavior, and the subjective risk perception of regarding symptoms, having a severe course and dying because of COVID-19. Data were compared between participants with the high risk diseases and individuals without any of those diseases. 16,983 respondents completed the study. Generalized anxiety, COVID-19-related fear, adherent/dysfunctional safety behavior and subjective risk perception were elevated in participants with high-risk diseases. The increased COVID-19-related fear as a functional concern is a conclusion on the increased risk of a severe course. The functionality of the fear is reflected in people’s increased need for security and includes an increase in both adherent and dysfunctional safety behavior that underlines the need for psychological support strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Nora Dörrie
- University of Duisburg-Essen, Essen, Germany
| | | | | | | | | | | |
Collapse
|
281
|
Dey A, Sen S, Maulik U. Unveiling COVID-19-associated organ-specific cell types and cell-specific pathway cascade. Brief Bioinform 2021; 22:914-923. [PMID: 32968798 PMCID: PMC7543283 DOI: 10.1093/bib/bbaa214] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/13/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
The novel coronavirus or COVID-19 has first been found in Wuhan, China, and became pandemic. Angiotensin-converting enzyme 2 (ACE2) plays a key role in the host cells as a receptor of Spike-I Glycoprotein of COVID-19 which causes final infection. ACE2 is highly expressed in the bladder, ileum, kidney and liver, comparing with ACE2 expression in the lung-specific pulmonary alveolar type II cells. In this study, the single-cell RNAseq data of the five tissues from different humans are curated and cell types with high expressions of ACE2 are identified. Subsequently, the protein-protein interaction networks have been established. From the network, potential biomarkers which can form functional hubs, are selected based on k-means network clustering. It is observed that angiotensin PPAR family proteins show important roles in the functional hubs. To understand the functions of the potential markers, corresponding pathways have been researched thoroughly through the pathway semantic networks. Subsequently, the pathways have been ranked according to their influence and dependency in the network using PageRank algorithm. The outcomes show some important facts in terms of infection. Firstly, renin-angiotensin system and PPAR signaling pathway can play a vital role for enhancing the infection after its intrusion through ACE2. Next, pathway networks consist of few basic metabolic and influential pathways, e.g. insulin resistance. This information corroborate the fact that diabetic patients are more vulnerable to COVID-19 infection. Interestingly, the key regulators of the aforementioned pathways are angiontensin and PPAR family proteins. Hence, angiotensin and PPAR family proteins can be considered as possible therapeutic targets. Contact: sagnik.sen2008@gmail.com, umaulik@cse.jdvu.ac.in Supplementary information: Supplementary data are available online.
Collapse
Affiliation(s)
- Ashmita Dey
- Department of Computer Science and Engineering, Jadavpur University, India
| | - Sagnik Sen
- Department of Computer Science and Engineering, Jadavpur University, India
| | - Ujjwal Maulik
- Department of Computer Science and Engineering, Jadavpur University, India
| |
Collapse
|
282
|
Exercise Training and Cardiac Rehabilitation in COVID-19 Patients with Cardiovascular Complications: State of Art. Life (Basel) 2021; 11:life11030259. [PMID: 33801080 PMCID: PMC8004041 DOI: 10.3390/life11030259] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Recent scientific literature has investigated the cardiovascular implications of COVID-19. The mechanisms of cardiovascular damage seem to involve the protein angiotensin-converting enzyme 2 (ACE2), to which severe acute respiratory syndrome (SARS) coronavirus-2 (CoV-2) binds to penetrate cells and other mechanisms, most of which are still under study. Cardiovascular sequelae of COVID-19 include heart failure, cardiomyopathy, acute coronary syndrome, arrhythmias, and venous thromboembolism. This article aims to collect scientific evidence by exploiting PubMed, Scopus, and Pedro databases to highlight the cardiovascular complications of COVID-19 and to define the physiotherapy treatment recommended for these patients. Exercise training (ET), an important part of cardiac rehabilitation, is a powerful tool in physiotherapy, capable of inducing significant changes in the cardiovascular system and functional in the recovery of endothelial dysfunction and for the containment of thromboembolic complications. In conclusion, due to the wide variety of possible exercise programs that can be obtained by combining intensity, duration, and speed in various ways, and by adjusting the program based on continuous patient monitoring, exercise training is well suited to the treatment of post-COVID patients with an impaired cardiovascular system of various degrees.
Collapse
|
283
|
Zhao S, Huang Z, Zeng H, Chen Z, Luo F, Zhang C, Song B. Combining initial chest CT with clinical variables in differentiating coronavirus disease 2019 (COVID-19) pneumonia from influenza pneumonia. Sci Rep 2021; 11:6422. [PMID: 33742041 PMCID: PMC7979799 DOI: 10.1038/s41598-021-85779-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 03/05/2021] [Indexed: 02/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has spread in more than 100 countries and regions around the world, raising grave global concerns. COVID-19 has a similar pattern of infection, clinical symptoms, and chest imaging findings to influenza pneumonia. In this retrospective study, we analysed clinical and chest CT data of 24 patients with COVID-19 and 79 patients with influenza pneumonia. Univariate analysis demonstrated that the temperature, systolic pressure, cough and sputum production could distinguish COVID-19 from influenza pneumonia. The diagnostic sensitivity and specificity for the clinical features are 0.783 and 0.747, and the AUC value is 0.819. Univariate analysis demonstrates that nine CT features, central-peripheral distribution, superior-inferior distribution, anterior-posterior distribution, patches of GGO, GGO nodule, vascular enlargement in GGO, air bronchogram, bronchiectasis within focus, interlobular septal thickening, could distinguish COVID-19 from influenza pneumonia. The diagnostic sensitivity and specificity for the CT features are 0.750 and 0.962, and the AUC value is 0.927. Finally, a multivariate logistic regression model combined the variables from the clinical variables and CT features models was made. The combined model contained six features: systolic blood pressure, sputum production, vascular enlargement in the GGO, GGO nodule, central-peripheral distribution and bronchiectasis within focus. The diagnostic sensitivity and specificity for the combined features are 0.87 and 0.96, and the AUC value is 0.961. In conclusion, some CT features or clinical variables can differentiate COVID-19 from influenza pneumonia. Moreover, CT features combined with clinical variables had higher diagnostic performance.
Collapse
Affiliation(s)
- Shuang Zhao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zixing Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Hanjiang Zeng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhixia Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fengming Luo
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chongwei Zhang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
284
|
Association Between Chronic Statin Use and 30-Day Mortality in Hospitalized Patients With COVID-19. Mayo Clin Proc Innov Qual Outcomes 2021; 5:442-446. [PMID: 33748678 PMCID: PMC7955930 DOI: 10.1016/j.mayocpiqo.2021.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective To determine the association between chronic statin use and mortality in patients hospitalized with coronavirus disease 2019 (COVID-19). Patients and Methods We identified a retrospective cohort of patients requiring admission at the Mayo Clinic using our enterprise-wide COVID-19 registry from March 1, 2020, through September 30, 2020. Available information included age, sex, use of statins, medical comorbidities, and 30-day mortality. We estimated the association of statins with 30-day mortality using odds ratios and 95% CIs from logistic regression modeling. Results Patients (N=1295) between the ages of 30 and 80 years tested positive for COVID-19 and required admission during the study period, of whom 500 (38.6%) were taking statins at admission. Patients taking statins were older and more likely to have diabetes mellitus or congestive heart failure. Within 30 days of diagnosis, 59 (4.6%) died. In multivariable analysis, statin users did not have statistically different odds of death within 30 days with an odds ratio of 1.14 (95% CI, 0.64 to 2.03; P=.67) compared to nonusers. Conclusion Patients with COVID-19 taking statins had similar 30-day mortality to those not taking statins after adjusting for relevant covariates. Although this is partly influenced by a higher prevalence of risk factors for more severe COVID-19 presentation not entirely adjusted for by the Charlson comorbidity index, these data would not support statins as a likely therapeutic intervention for COVID-19 in the hospital setting.
Collapse
|
285
|
Analyzing knowledge entities about COVID-19 using entitymetrics. Scientometrics 2021; 126:4491-4509. [PMID: 33746309 PMCID: PMC7953944 DOI: 10.1007/s11192-021-03933-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/26/2021] [Indexed: 12/24/2022]
Abstract
COVID-19 cases have surpassed the 109 + million markers, with deaths tallying up to 2.4 million. Tens of thousands of papers regarding COVID-19 have been published along with countless bibliometric analyses done on COVID-19 literature. Despite this, none of the analyses have focused on domain entities occurring in scientific publications. However, analysis of these bio-entities and the relations among them, a strategy called entity metrics, could offer more insights into knowledge usage and diffusion in specific cases. Thus, this paper presents an entitymetric analysis on COVID-19 literature. We construct an entity–entity co-occurrence network and employ network indicators to analyze the extracted entities. We find that ACE-2 and C-reactive protein are two very important genes and that lopinavir and ritonavir are two very important chemicals, regardless of the results from either ranking.
Collapse
|
286
|
Yue-liang X, Jiang-lin W, Hui-qin Y, Ge Z, Hongyu D, Wei-jin F, Xiao-cong Z. The risk factors for severe patients with COVID-19 in China: A systematic review and meta-analysis. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211000890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is spreading exponentially. In order to optimize medical resources allocation and reduce mortality, biomarkers are needed to differentiate between COVID-19 patients with or without severe diseases early as possible. We searched Ovid MEDLINE(R), Ovid EMBASE, CNKI, Wanfang, VIP databases, the Cochrane Library, and medRxiv for primary articles in English or Chinese up to March 30, 2020 to systematically evaluate the risk factors for severe patients in China. Mean difference or standardize mean difference and odds ratio with 95% confidence intervals were performed by random-effect or fixed models in cases of significant heterogeneity between studies. We used I2 to evaluate the magnitude of heterogeneity. A total of 54 articles involving about 7000 patients were eligible for this meta-analysis. In total, 52 of 67 parameters between severe and non-severe cases were significantly different. Elderly male patients with comorbidities including hypertension, diabetes, chronic obstructive pulmonary disease (COPD) cardiovascular disease, cerebrovascular disease, chronic kidney disease, or cancer were more common in severe COVID-19 patients. Regarding the clinical manifestations on admission, fever, cough, expectoration, dyspnea, chest distress, fatigue, headache, chills, anorexia, or abdominal pain were more prevalent in severe COVID-19 patients. The results of the clinical examination showed that high C-reactive protein (CRP), high lactate dehydrogenase (LDH), high D-dimer, and decreased T lymphocytes cells subsets, decreased lymphocyte may help clinicians predict the progression of severe illness in patients with COVID-19. Our findings will be conducive for clinician to stratify the COVID-19 patients to reduce mortality under the relative shortage of medical resources.
Collapse
Affiliation(s)
- Xie Yue-liang
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Pharmacy Administration Commission, Hospital Association of Hunan Province, Changsha, Hunan, China
| | - Wang Jiang-lin
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Hui-qin
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhou Ge
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Deng Hongyu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Fang Wei-jin
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zuo Xiao-cong
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Pharmacy and Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Pharmacy Administration Commission, Hospital Association of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
287
|
Migliaccio MG, Di Mauro M, Ricciolino R, Spiniello G, Carfora V, Verde N, Mottola FF, Coppola N. Renal Involvement in COVID-19: A Review of the Literature. Infect Drug Resist 2021; 14:895-903. [PMID: 33707958 PMCID: PMC7943324 DOI: 10.2147/idr.s288869] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/17/2020] [Indexed: 01/08/2023] Open
Abstract
Kidney injury may be a severe complication of acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and contributes to worsen the prognosis. Various pathophysiological mechanisms can contribute to organ damage and impair renal function, proving the complexity of the virus activity and the resulting immunity response. We summarized the evidence of the literature on the prevalence of kidney involvement, on the pathogenic pathways and on its management.
Collapse
Affiliation(s)
- Marco Giuseppe Migliaccio
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Marco Di Mauro
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Riccardo Ricciolino
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giorgio Spiniello
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Vincenzo Carfora
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Nicoletta Verde
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Filiberto Fausto Mottola
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Nicola Coppola
- Department of Mental Health and Public Medicine, Infectious Diseases Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - On behalf of the Vanvitelli COVID-19 Group
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Department of Mental Health and Public Medicine, Infectious Diseases Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
288
|
Silva RN, Goulart CDL, Oliveira MR, Tacao GY, Back GD, Severin R, Faghy MA, Arena R, Borghi-Silva A. Cardiorespiratory and skeletal muscle damage due to COVID-19: making the urgent case for rehabilitation. Expert Rev Respir Med 2021; 15:1107-1120. [PMID: 33606567 DOI: 10.1080/17476348.2021.1893169] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION It has become increasingly evident that COVID-19 contributes to multiorgan pathophysiology. The systemic inflammatory response increases both pro-inflammatory cytokine and chemokine levels, leading to immune dysregulation and increasing the likelihood of incurring cardiac and pulmonary injuries. AREAS COVERED Longer periods of hospitalization (~20 days) increase susceptibility to ICU-acquired muscle weakness and deconditioning, which decreases muscle function and functional capacity. These conditions affect the quality of life in the post-COVID-19 period and require multi-disciplinary approaches to rehabilitate the cardiopulmonary and musculoskeletal systems of these patients. In this context, this narrative review, which included articles published in the Embase, PEDro and PubMed databases up to December 2020, is focused on discussing the essential role of exercise and rehabilitation health professionals in the COVID-19 recovery process, from hospitalization to hospital discharge, addressing strategies for professionals to mitigate the cardiac and pulmonary impairments associated with hospitalization to home or ambulatory rehabilitation, purposing ways to conduct rehabilitation programs to restore their functional status and quality of life after the infection. EXPERT OPINION In the current environment, these findings further point to the vital role of rehabilitation health professionals in the coming years and the urgent need to develop strategies to assist COVID-19 survivors.
Collapse
Affiliation(s)
- Rebeca Nunes Silva
- Cardiopulmonary Physiotherapy Laboratory, Department of Physiotherapy, Federal University of São Carlos (Ufscar), Monjolinho, Zip-code: São Carlos, SP, Brazil
| | - Cássia Da Luz Goulart
- Cardiopulmonary Physiotherapy Laboratory, Department of Physiotherapy, Federal University of São Carlos (Ufscar), Monjolinho, Zip-code: São Carlos, SP, Brazil
| | - Murilo Rezende Oliveira
- Cardiopulmonary Physiotherapy Laboratory, Department of Physiotherapy, Federal University of São Carlos (Ufscar), Monjolinho, Zip-code: São Carlos, SP, Brazil
| | - Guilherme Yassuyuki Tacao
- Department of Physiotherapy, Postgraduate Program in Physiotherapy, Faculty of Science and Technology, São Paulo State University (UNESP), Zip-code: Presidente Prudente, SP, Brazil
| | - Guilherme Dionir Back
- Cardiopulmonary Physiotherapy Laboratory, Department of Physiotherapy, Federal University of São Carlos (Ufscar), Monjolinho, Zip-code: São Carlos, SP, Brazil
| | - Richard Severin
- Department of Physical Therapy, Healthy Living for Pandemic Event Protection (HL - PIVOT) Network, Chicago, IL, USA.,Department of Physical Therapy Department, College of Applied Health Science, the University of Illinois at Chicago, Chicago, IL, USA.,Department of Physical Therapy, Robbins College of Applied Health Sciences, Baylor University, Waco, TX, USA
| | - Mark A Faghy
- Department of Physical Therapy, Healthy Living for Pandemic Event Protection (HL - PIVOT) Network, Chicago, IL, USA.,Department School of Human Sciences, Human Science Research Centre, University of Derby, Derby, UK
| | - Ross Arena
- Department of Physical Therapy, Healthy Living for Pandemic Event Protection (HL - PIVOT) Network, Chicago, IL, USA.,Department of Physical Therapy Department, College of Applied Health Science, the University of Illinois at Chicago, Chicago, IL, USA
| | - Audrey Borghi-Silva
- Cardiopulmonary Physiotherapy Laboratory, Department of Physiotherapy, Federal University of São Carlos (Ufscar), Monjolinho, Zip-code: São Carlos, SP, Brazil.,Department of Physical Therapy, Healthy Living for Pandemic Event Protection (HL - PIVOT) Network, Chicago, IL, USA
| |
Collapse
|
289
|
Sheth A, Modi M, Dawson D, Dominic P. Prognostic value of cardiac biomarkers in COVID-19 infection. Sci Rep 2021; 11:4930. [PMID: 33654230 PMCID: PMC7925599 DOI: 10.1038/s41598-021-84643-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 02/10/2021] [Indexed: 01/08/2023] Open
Abstract
Multiple Biomarkers have recently been shown to be elevated in COVID-19, a respiratory infection with multi-organ dysfunction; however, information regarding the prognostic value of cardiac biomarkers as it relates to disease severity and cardiac injury are inconsistent. The goal of this meta-analysis was to summarize the evidence regarding the prognostic relevance of cardiac biomarkers from data available in published reports. PubMed, Embase and Web of Science were searched from inception through April 2020 for studies comparing median values of cardiac biomarkers in critically ill versus non-critically ill COVID-19 patients, or patients who died versus those who survived. The weighted mean differences (WMD) and 95% confidence interval (CI) between the groups were calculated for each study and combined using a random effects meta-analysis model. The odds ratio (OR) for mortality based on cardiac injury was combined from studies reporting it. Troponin levels were significantly higher in COVID-19 patients who died or were critically ill versus those who were alive or not critically ill (WMD 0.57, 95% CI 0.43-0.70, p < 0.001). Additionally, BNP levels were also significantly higher in patients who died or were critically ill (WMD 0.45, 95% CI - 0.21-0.69, p < 0.001). Cardiac injury was independently associated with significantly increased odds of mortality (OR 6.641, 95% CI 1.26-35.1, p = 0.03). A significant difference in levels of D-dimer was seen in those who died or were critically ill. CK levels were only significantly higher in those who died versus those who were alive (WMD 0.79, 95% CI 0.25-1.33, p = 0.004). Cardiac biomarkers add prognostic value to the determination of the severity of COVID-19 and can predict mortality.
Collapse
Affiliation(s)
- Aakash Sheth
- Division of Cardiology, The Department of Medicine and Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center-Shreveport (LSUHSC-S), 1501 Kings Hwy, Shreveport, LA, 71103, USA
| | - Malak Modi
- Division of Cardiology, The Department of Medicine and Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center-Shreveport (LSUHSC-S), 1501 Kings Hwy, Shreveport, LA, 71103, USA
| | - Desiree' Dawson
- Division of Cardiology, The Department of Medicine and Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center-Shreveport (LSUHSC-S), 1501 Kings Hwy, Shreveport, LA, 71103, USA
| | - Paari Dominic
- Division of Cardiology, The Department of Medicine and Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center-Shreveport (LSUHSC-S), 1501 Kings Hwy, Shreveport, LA, 71103, USA.
| |
Collapse
|
290
|
Rocha JLM, de Oliveira WCF, Noronha NC, Dos Santos NCD, Covas DT, Picanço-Castro V, Swiech K, Malmegrim KCR. Mesenchymal Stromal Cells in Viral Infections: Implications for COVID-19. Stem Cell Rev Rep 2021; 17:71-93. [PMID: 32895900 PMCID: PMC7476649 DOI: 10.1007/s12015-020-10032-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) constitute a heterogeneous population of stromal cells with immunomodulatory and regenerative properties that support their therapeutic use. MSCs isolated from many tissue sources replicate vigorously in vitro and maintain their main biological properties allowing their widespread clinical application. To date, most MSC-based preclinical and clinical trials targeted immune-mediated and inflammatory diseases. Nevertheless, MSCs have antiviral properties and have been used in the treatment of various viral infections in the last years. Here, we revised in detail the biological properties of MSCs and their preclinical and clinical applications in viral diseases, including the disease caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection (COVID-19). Notably, rapidly increasing numbers of MSC-based therapies for COVID-19 have recently been reported. MSCs are theoretically capable of reducing inflammation and promote lung regeneration in severe COVID-19 patients. We critically discuss the rationale, advantages and disadvantages of MSC-based therapies for viral infections and also specifically for COVID-19 and point out some directions in this field. Finally, we argue that MSC-based therapy may be a promising therapeutic strategy for severe COVID-19 and other emergent respiratory tract viral infections, beyond the viral infection diseases in which MSCs have already been clinically applied. Graphical Abstract ![]()
Collapse
Affiliation(s)
- José Lucas Martins Rocha
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Basic and Applied Immunology Program, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Waldir César Ferreira de Oliveira
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Bioscience and Biotecnology Program, Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Nádia Cássia Noronha
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Bioscience and Biotecnology Program, Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Natalia Cristine Dias Dos Santos
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Bioscience and Biotecnology Program, Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Virgínia Picanço-Castro
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kamilla Swiech
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n, Ribeirão Preto, 14040-903, São Paulo, Brazil
| | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil. .,School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n, Ribeirão Preto, 14040-903, São Paulo, Brazil.
| |
Collapse
|
291
|
Singh A, Khanna P, Sarkar S. High-Flow Nasal Cannula, a Boon or a Bane for COVID-19 Patients? An Evidence-Based Review. CURRENT ANESTHESIOLOGY REPORTS 2021; 11:101-106. [PMID: 33679254 PMCID: PMC7921283 DOI: 10.1007/s40140-021-00439-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW This review instantiates the efficacy and safety of HFNC in the context of COVID-19 pandemic. RECENT FINDINGS Globally, the healthcare system is facing an unprecedented crisis of resources due to the 2019 novel coronavirus disease (COVID-19) pandemic. Fever, cough, dyspnea, myalgia, fatigue, and pneumonia are the most common symptoms associated with it. The incidence of invasive mechanical ventilation in ICU patients ranges from 29.1 to 89.9%. Supplemental oxygen therapy is the main stay treatment for managing hypoxemic respiratory failure. The high-flow nasal cannula (HFNC) is a novel non-invasive strategy for better oxygenation and ventilation in critically ill patients. In this grim scenario, a reduction in mechanical ventilation by means of HFNC is of prime interest. SUMMARY HFNC is considered an aerosol-generating intervention with the risk of viral aerosolization with a concern of potential nosocomial transmission of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). However, there is no consensus regarding the use of HFNC in novel coronavirus-infected pneumonia (NCIP). HFNC seems to be an effective and safe treatment modality in acute respiratory failure with optimal settings and selection of ideal patients.
Collapse
Affiliation(s)
- Abhishek Singh
- Department of Anaesthesia, Pain Medicine and Critical Care, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, 110029 India
| | - Puneet Khanna
- Department of Anaesthesia, Pain Medicine and Critical Care, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, 110029 India
| | - Soumya Sarkar
- Department of Anaesthesia, Pain Medicine and Critical Care, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, 110029 India
| |
Collapse
|
292
|
Ediz C, Tavukcu HH, Akan S, Kizilkan YE, Alcin A, Oz K, Yilmaz O. Is there any association of COVID-19 with testicular pain and epididymo-orchitis? Int J Clin Pract 2021; 75:e13753. [PMID: 33063899 PMCID: PMC7646040 DOI: 10.1111/ijcp.13753] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS This study aims to analyse the novel Coronavirus disease- (COVID-19) related testicular pain in hospitalised patients because of COVID-19 and to review as an aetiological factor for epididymitis, orchitis or both. METHODS A total of 91 patients were included in the study. A questionnaire was formed for the questioning of testicular pain or epididymo-orchitis in patients with COVID-19. Demographics and past medical history was also recorded. Patients' neutrophil and lymphocyte counts, neutrophil-lymphocyte ratios (NLR), C-reactive protein (CRP) levels and D-dimer values were recorded. Patients with COVID-19 were divided into two groups according to absence or presence of testicular pain or epididymo-orchitis as group 1 and group 2. All results were compared for both groups. RESULTS The median age of patients was similar in both groups. Testicular pain was occurred in 10.98% of the patients. Clinical presentation of epididymo-orchitis was diagnosed in only one patient. No statistically significant difference was reported in terms of patients' age, levels of CRP and D-Dimer or NLR and results of questionnaire form queries between the two groups (P > .05). CONCLUSION Testicular pain was observed more frequently in hospitalised COVID-19 cases. While no inflammation marker which is related to predict of testicular pain or epididymo-orchitis was found in patients with COVID-19.
Collapse
Affiliation(s)
- Caner Ediz
- Department of UrologySultan Abdulhamid Han Education and Research HospitalIstanbulTurkey
| | - Hasan Huseyin Tavukcu
- Department of UrologySultan Abdulhamid Han Education and Research HospitalIstanbulTurkey
| | - Serkan Akan
- Department of UrologySultan Abdulhamid Han Education and Research HospitalIstanbulTurkey
| | - Yunus Emre Kizilkan
- Department of UrologySultan Abdulhamid Han Education and Research HospitalIstanbulTurkey
| | - Adem Alcin
- Department of UrologySultan Abdulhamid Han Education and Research HospitalIstanbulTurkey
| | - Kerem Oz
- Department of UrologySultan Abdulhamid Han Education and Research HospitalIstanbulTurkey
| | - Omer Yilmaz
- Department of UrologySultan Abdulhamid Han Education and Research HospitalIstanbulTurkey
| |
Collapse
|
293
|
Akan S, Ediz C, Kızılkan YE, Alcin A, Tavukcu HH, Yilmaz O. COVID-19 infection threat in patients with high-risk non-muscle invasive bladder cancer receiving intravesical BCG therapy. Int J Clin Pract 2021; 75:e13752. [PMID: 33064918 PMCID: PMC7646056 DOI: 10.1111/ijcp.13752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/03/2020] [Indexed: 01/08/2023] Open
Abstract
AIM We evaluated the COVID-19 infection threat in patients receiving intravesical BCG therapy which has immunotherapeutic effects and is of vital importance in most of the individuals with high-risk non-muscle-invasive bladder cancer (NMIBC) and investigated the need for postponement of this therapy. METHODS A total of 71 patients, who were diagnosed with high-risk NMIBC and on intravesical BCG treatment regularly (induction or maintenance), were enrolled in the study. The patients were classified into two groups depending on whether they were diagnosed with COVID-19 during the pandemic period or not. RESULTS Of 71 patients, 26 underwent a COVID-19 polymerase chain reaction test with clinical suspicion during the pandemic period. Of these 26 patients, 4 were diagnosed with COVID-19. Age of the patients, working status (working/retired), compliance with containment measures against the pandemic, number of BCG courses, adverse effects after BCG therapy and systemic immune-inflammation index, which is an inflammation-related parameter, were not different between groups (P > .05). Neutrophil/lymphocyte ratio was significantly higher in the COVID-19 positive group (P < .05). COVID-19 positivity was higher in age groups 50-64 (6.6%) and 65-80 (5.8%) years than that in similar age groups of the normal population. CONCLUSION Every effort should be made to administer intravesical BCG treatment in high-risk NMIBC patients even during the pandemic period. However, increased risk of COVID-19 transmission should be kept in mind and protective measures against COVID-19 for healthcare providers and patients before the procedure should be taken optimally. The procedure should be postponed in patients with lymphopenia in recent complete blood count.
Collapse
Affiliation(s)
- Serkan Akan
- Department of UrologyUniversity of Health SciencesSultan Abdulhamid Han Training and Research HospitalIstanbulTurkey
| | - Caner Ediz
- Department of UrologyUniversity of Health SciencesSultan Abdulhamid Han Training and Research HospitalIstanbulTurkey
| | - Yunus Emre Kızılkan
- Department of UrologyUniversity of Health SciencesSultan Abdulhamid Han Training and Research HospitalIstanbulTurkey
| | - Adem Alcin
- Department of UrologyUniversity of Health SciencesSultan Abdulhamid Han Training and Research HospitalIstanbulTurkey
| | - Hasan Huseyin Tavukcu
- Department of UrologyUniversity of Health SciencesSultan Abdulhamid Han Training and Research HospitalIstanbulTurkey
| | - Omer Yilmaz
- Department of UrologyUniversity of Health SciencesSultan Abdulhamid Han Training and Research HospitalIstanbulTurkey
| |
Collapse
|
294
|
Chen Y, Huang WC, Yang CS, Cheng FJ, Chiu YF, Chen HF, Huynh TK, Huang CF, Chen CH, Wang HC, Hung MC. Screening strategy of TMPRSS2 inhibitors by FRET-based enzymatic activity for TMPRSS2-based cancer and COVID-19 treatment. Am J Cancer Res 2021; 11:827-836. [PMID: 33791156 PMCID: PMC7994159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/07/2020] [Indexed: 06/12/2023] Open
Abstract
Transmembrane serine protease (TMPRSS2) plays an oncogenic role in prostate cancer as the fusion gene with ERG, and has also been demonstrated to be essential for the cellular entry of severe acute respiratory syndrome coronaviruses (SARS-CoV). Thus, targeting TMPRSS2 is a promising strategy for therapies against both prostate cancer and coronavirus infection. Although Nafamostat and Camostat have been identified as TMPRSS2 inhibitors, severe side effects such as cerebral hemorrhage, anaphylactoid reaction, and cardiac arrest shock greatly hamper their clinical use. Therefore, more potent and safer drugs against this serine protease should be further developed. In this study, we developed a fluorescence resonance energy transfer (FRET)-based platform for effectively screening of inhibitors against TMPRSS2 protease activity. The disruption of FRET between green and red fluorescent proteins conjugated with the substrate peptide, which corresponds to the cleavage site of SARS-CoV-2 Spike protein, was measured to determine the enzymatic activity of TMPRSS2. Through an initiate pilot screening with around 100 compounds, Flupirtine, a selective neuronal potassium channel opener, was identified as a potential TMPRSS2 inhibitor from an FDA-approved drug library by using this screening platform, and showed inhibitory effect on the TMPRSS-dependent infection of SARS-CoV-2 Spike-pseudotyped lentiviral particles. This study describes a platform proven effective for rapidly screening of TMPRSS2 inhibitors, and suggests that Flupirtine may be worthy of further consideration of repurposing to treat COVID-19 patients.
Collapse
Affiliation(s)
- Yeh Chen
- Institute of New Drug Development, China Medical UniversityTaichung, Taiwan
- Research Center for Cancer Biology and Center for Molecular Medicine, China Medical UniversityTaichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichung 40402, Taiwan
| | - Wei-Chien Huang
- Institute of New Drug Development, China Medical UniversityTaichung, Taiwan
- Research Center for Cancer Biology and Center for Molecular Medicine, China Medical UniversityTaichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichung 40402, Taiwan
- Department of Biotechnology, Asia UniversityTaichung 413, Taiwan
| | - Chia-Shin Yang
- Institute of New Drug Development, China Medical UniversityTaichung, Taiwan
| | - Fang-Ju Cheng
- Graduate Institute of Basic Medical Sciences, China Medical UniversityTaichung, 40402, Taiwan
| | - Yi-Fang Chiu
- Institute of New Drug Development, China Medical UniversityTaichung, Taiwan
| | - Hsiao-Fan Chen
- Research Center for Cancer Biology and Center for Molecular Medicine, China Medical UniversityTaichung 40402, Taiwan
| | - Thanh Kieu Huynh
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichung 40402, Taiwan
| | - Chih-Feng Huang
- Institute of New Drug Development, China Medical UniversityTaichung, Taiwan
| | - Chia-Hung Chen
- School of Medicine, China Medical UniversityTaichung 404, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University HospitalTaichung 404, Taiwan
| | - Hsueh-Chun Wang
- Research Center for Cancer Biology and Center for Molecular Medicine, China Medical UniversityTaichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichung 40402, Taiwan
| | - Mien-Chie Hung
- Institute of New Drug Development, China Medical UniversityTaichung, Taiwan
- Research Center for Cancer Biology and Center for Molecular Medicine, China Medical UniversityTaichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical UniversityTaichung 40402, Taiwan
- Department of Biotechnology, Asia UniversityTaichung 413, Taiwan
| |
Collapse
|
295
|
Martin JH, Head R. A pharmacological framework for integrating treating the host, drug repurposing and the damage response framework in COVID-19. Br J Clin Pharmacol 2021; 87:875-885. [PMID: 32959913 PMCID: PMC7646655 DOI: 10.1111/bcp.14551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 12/19/2022] Open
Abstract
With any new disease a framework for the development of preventative or treatment therapeutics is key; the absence of such in COVID-19 has enabled ineffective and potentially unsafe treatments to be taken up by governments and clinicians desperate to have options for patients. As we still have few therapies and nil vaccines yet available, the void of a clear framework for research and practice is increasingly clear. We describe a framework that has been used to prioritise therapeutic research in previous pandemics which could be used to progress clinical pharmacology and therapeutics research in COVID-19. This is particularly relevant as discussion has already moved on from antiviral therapeutics to delineating the treatment of the host from treatment and elimination of the infective agent. Focussing on the host brings together three concepts: host treatment, the damage response framework and therapeutic repurposing. The integration of these three areas plays to the traditional strength of pharmaceuticals in providing a period of stabilization to permit time for the development of novel antiviral drugs and vaccines. In integrating approaches to repurposing, host treatment and damage response we identified three key properties that a potentially effective repurposed drug must possess by way of a framework. There must be homology, i.e., the same or similar relation with the pathogenesis of the disease, ideally targeted to the conserved pathophysiological outcomes of the viral attack; there must be a defined locus within the spectrum to prevention to severe disease and the framework must draw upon the historical dose and safety experience of the repurposed drug. To illustrate, we have mapped therapeutics that impact upon a key dysregulated pathway in COVID-19 - the renin angiotensin system - using this approach. Collectively this type of analysis reveals the importance of existing data (repurposed information and administrative observational data) and the importance of the details of the known pathophysiological response to viruses in approaches to treating the host.
Collapse
Affiliation(s)
- Jennifer H. Martin
- Centre for Human Drug Repurposing and Medicines ResearchUniversity of NewcastleNSW2305Australia
| | - Richard Head
- University of South AustraliaSouth AustraliaAustralia
| |
Collapse
|
296
|
Fedson DS. COVID-19, host response treatment, and the need for political leadership. J Public Health Policy 2021; 42:6-14. [PMID: 33268844 PMCID: PMC7708736 DOI: 10.1057/s41271-020-00266-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 12/16/2022]
Abstract
Health officials and scientists have warned that we face the threat of a potentially devastating influenza pandemic. Instead, we are now in the midst of a global coronavirus (COVID-19) pandemic. National and international pandemic preparedness plans have focused on developing vaccines and antiviral treatments. Another way to confront the COVID-19 pandemic (and future pandemics) might be to treat patients with inexpensive and widely available generic drugs that target the host response to infection, not the virus itself. The feasibility of this idea was tested during the Ebola outbreak in West Africa in 2014. This experience should inform our approach to treating COVID-19 patients. It could also save lives during outbreaks of other emerging infectious diseases and episodes of everyday acute critical illness. If this "bottom up" syndromic approach to treating acute critical illness were shown to be effective, it could have a dramatic impact on health, equity and security throughout the world. HIGHLIGHTS: Uncertainty about the outcome of COVID-19 is driving the social, economic and political distress associated with the pandemic. Treating the host response to COVID-19 with inexpensive and widely available generic drugs might save lives and mitigate this distress. Undertaking research on this idea will require political leadership.
Collapse
|
297
|
Darwesh AM, Bassiouni W, Sosnowski DK, Seubert JM. Can N-3 polyunsaturated fatty acids be considered a potential adjuvant therapy for COVID-19-associated cardiovascular complications? Pharmacol Ther 2021; 219:107703. [PMID: 33031856 PMCID: PMC7534795 DOI: 10.1016/j.pharmthera.2020.107703] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has currently led to a global pandemic with millions of confirmed and increasing cases around the world. The novel SARS-CoV-2 not only affects the lungs causing severe acute respiratory dysfunction but also leads to significant dysfunction in multiple organs and physiological systems including the cardiovascular system. A plethora of studies have shown the viral infection triggers an exaggerated immune response, hypercoagulation and oxidative stress, which contribute significantly to poor cardiovascular outcomes observed in COVID-19 patients. To date, there are no approved vaccines or therapies for COVID-19. Accordingly, cardiovascular protective and supportive therapies are urgent and necessary to the overall prognosis of COVID-19 patients. Accumulating literature has demonstrated the beneficial effects of n-3 polyunsaturated fatty acids (n-3 PUFA) toward the cardiovascular system, which include ameliorating uncontrolled inflammatory reactions, reduced oxidative stress and mitigating coagulopathy. Moreover, it has been demonstrated the n-3 PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are precursors to a group of potent bioactive lipid mediators, generated endogenously, which mediate many of the beneficial effects attributed to their parent compounds. Considering the favorable safety profile for n-3 PUFAs and their metabolites, it is reasonable to consider n-3 PUFAs as potential adjuvant therapies for the clinical management of COVID-19 patients. In this article, we provide an overview of the pathogenesis of cardiovascular complications secondary to COVID-19 and focus on the mechanisms that may contribute to the likely benefits of n-3 PUFAs and their metabolites.
Collapse
Affiliation(s)
- Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Wesam Bassiouni
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
298
|
Gu SX, Tyagi T, Jain K, Gu VW, Lee SH, Hwa JM, Kwan JM, Krause DS, Lee AI, Halene S, Martin KA, Chun HJ, Hwa J. Thrombocytopathy and endotheliopathy: crucial contributors to COVID-19 thromboinflammation. Nat Rev Cardiol 2021; 18:194-209. [PMID: 33214651 PMCID: PMC7675396 DOI: 10.1038/s41569-020-00469-1] [Citation(s) in RCA: 263] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
The core pathology of coronavirus disease 2019 (COVID-19) is infection of airway cells by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that results in excessive inflammation and respiratory disease, with cytokine storm and acute respiratory distress syndrome implicated in the most severe cases. Thrombotic complications are a major cause of morbidity and mortality in patients with COVID-19. Patients with pre-existing cardiovascular disease and/or traditional cardiovascular risk factors, including obesity, diabetes mellitus, hypertension and advanced age, are at the highest risk of death from COVID-19. In this Review, we summarize new lines of evidence that point to both platelet and endothelial dysfunction as essential components of COVID-19 pathology and describe the mechanisms that might account for the contribution of cardiovascular risk factors to the most severe outcomes in COVID-19. We highlight the distinct contributions of coagulopathy, thrombocytopathy and endotheliopathy to the pathogenesis of COVID-19 and discuss potential therapeutic strategies in the management of patients with COVD-19. Harnessing the expertise of the biomedical and clinical communities is imperative to expand the available therapeutics beyond anticoagulants and to target both thrombocytopathy and endotheliopathy. Only with such collaborative efforts can we better prepare for further waves and for future coronavirus-related pandemics.
Collapse
Affiliation(s)
- Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Vivian W Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Seung Hee Lee
- Division of Cardiovascular Diseases, Center for Biomedical Sciences, National Institute of Health, Cheongju, Chungbuk, Korea
| | - Jonathan M Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jennifer M Kwan
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Diane S Krause
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Alfred I Lee
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hyung J Chun
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
299
|
Polak Y, Speth RC. Metabolism of angiotensin peptides by angiotensin converting enzyme 2 (ACE2) and analysis of the effect of excess zinc on ACE2 enzymatic activity. Peptides 2021; 137:170477. [PMID: 33400951 PMCID: PMC7887068 DOI: 10.1016/j.peptides.2020.170477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/21/2020] [Accepted: 12/15/2020] [Indexed: 01/02/2023]
Abstract
After decades of notoriety for its adverse cardiovascular, proinflammatory and profibrotic actions, the renin-angiotensin system (RAS) began to be cast in a more favorable light with the discovery of angiotensin-converting enzyme-2 (ACE2) in 2000. This monocarboxypeptidase, best known for its ability to metabolize angiotensin (Ang) II to Ang 1-7, counteracts the adverse effects of Ang II mediated by the AT1 Ang II receptor. Ang peptides are classically considered to be metabolized by aminopeptidases, by which the nomenclature Ang III (des-Asp1Ang II, 2-8 heptapeptide) and Ang IV (des-Asp1des-Arg2Ang II, 3-8 hexapeptide) are derived. This report compares the ability of recombinant human ACE2 (rhACE2) to metabolize Ang III, Ang IV and Ang V, (4-8 pentapeptide) relative to Ang II to form corresponding des-omega-Phe metabolites. rhACE2 has highest affinity (lowest Km) for Ang III, followed by Ang II ∼ Ang V, followed by Ang IV. However, rhACE2 has the highest Kcat for metabolising Ang IV followed by Ang V, Ang III and Ang II. The enzymatic efficiency (Kcat/Km) is highest for Ang V and Ang III followed by Ang IV and is lowest for Ang II. As a gluzincin metallopeptidase, ACE2 requires a zinc molecule at its active site for catalysis. This report also documents inhibition of ACE2 activity by concentrations of zinc exceeding 10 μM. These observations extend the functional significance of ACE2 to include the metabolic inactivation of Ang III, Ang IV and Ang V, reemphasizing the importance of monitoring zinc intake to maintain metabolic homeostasis.
Collapse
Affiliation(s)
- Yasmin Polak
- College of Pharmacy, University of Utrecht, Universiteitsweg 99, 3584 CG, Utrecht, the Netherlands; College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, United States.
| | - Robert C Speth
- College of Pharmacy, University of Utrecht, Universiteitsweg 99, 3584 CG, Utrecht, the Netherlands; College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, United States.
| |
Collapse
|
300
|
Olaleye OA, Kaur M, Onyenaka C, Adebusuyi T. Discovery of Clioquinol and analogues as novel inhibitors of Severe Acute Respiratory Syndrome Coronavirus 2 infection, ACE2 and ACE2 - Spike protein interaction in vitro. Heliyon 2021; 7:e06426. [PMID: 33732940 PMCID: PMC7951571 DOI: 10.1016/j.heliyon.2021.e06426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/05/2021] [Accepted: 05/02/2021] [Indexed: 01/08/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the etiological agent for coronavirus disease 2019 (COVID-19), has resulted in an ongoing pandemic. Presently, there are no clinically approved drugs for COVID-19. Hence, there is an urgent need to accelerate the development of effective antivirals. Herein, we discovered Clioquinol (5-chloro-7-iodo-8-quinolinol (CLQ)), a Food and Drug Administration (FDA) approved drug, and two of its analogues (7-bromo-5-chloro-8-hydroxyquinoline (CLBQ14); and 5, 7-Dichloro-8-hydroxyquinoline (CLCQ)) as potent inhibitors of SARS-CoV-2 infection-induced cytopathic effect in vitro. In addition, all three compounds showed potent anti-exopeptidase activity against recombinant human angiotensin-converting enzyme 2 (rhACE2) and inhibited the binding of rhACE2 with SARS-CoV-2 Spike (RBD) protein. CLQ displayed the highest potency in the low micromolar range, with its antiviral activity showing a strong correlation with inhibition of rhACE2 and rhACE2-RBD interaction. Altogether, our findings provide a new mode of action and molecular target for CLQ and validates this pharmacophore as a promising lead series for the clinical development of potential therapeutics for COVID-19.
Collapse
Affiliation(s)
- Omonike A. Olaleye
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, TX 77004, USA
| | - Manvir Kaur
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, TX 77004, USA
| | - Collins Onyenaka
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, TX 77004, USA
| | - Tolulope Adebusuyi
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, TX 77004, USA
| |
Collapse
|