1951
|
Rizk JG, Kalantar-Zadeh K, Mehra MR, Lavie CJ, Rizk Y, Forthal DN. Authors' Reply to Vrachatis et al. "Pharmaco-Immunomodulatory Therapy I COVID-19". Drugs 2020; 80:1501-1503. [PMID: 32880806 PMCID: PMC7471515 DOI: 10.1007/s40265-020-01396-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- John G Rizk
- Edson College, Arizona State University, Phoenix, AZ, USA.
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology, Hypertension and Kidney Transplantation, University of California, Irvine, School of Medicine, Irvine, CA, USA.,Department of Epidemiology, University of California, Los Angeles, UCLA Fielding School of Public Health, Los Angeles, CA, USA.,Tibor Rubin VA Long Beach Healthcare System, Long Beach, CA, USA
| | - Mandeep R Mehra
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-the University of Queensland School of Medicine, New Orleans, LA, USA
| | - Youssef Rizk
- Department of Family Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Donald N Forthal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, Irvine, CA, USA.,Department of Molecular Biology and Biochemistry, University of California, Irvine, School of Medicine, Irvine, CA, USA
| |
Collapse
|
1952
|
Liu W, Guan WJ, Zhong NS. Strategies and Advances in Combating COVID-19 in China. ENGINEERING (BEIJING, CHINA) 2020; 6:1076-1084. [PMID: 33078078 PMCID: PMC7558233 DOI: 10.1016/j.eng.2020.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 05/02/2023]
Abstract
Coronavirus disease 2019 (COVID-19)-the third in a series of coronavirus infections-has caused a global public health event in the 21st century, resulting in substantial global morbidity and mortality. Building on its legacy of managing severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS), China has played a key role in the scientific community by revealing the viral transmission routes and clinical characteristics of COVID-19 and developing novel therapeutic interventions and vaccines. Despite these rapid scientific and technological advances, uncertainties remain in tracing the original sources of infection, determining the routes of transmission and pathogenesis, and addressing the lack of targeted clinical management of COVID-19. Here, we summarize the major COVID-19 research advances in China in order to provide useful information for global pandemic control.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
1953
|
Wilson MG, Hull JH, Rogers J, Pollock N, Dodd M, Haines J, Harris S, Loosemore M, Malhotra A, Pieles G, Shah A, Taylor L, Vyas A, Haddad FS, Sharma S. Cardiorespiratory considerations for return-to-play in elite athletes after COVID-19 infection: a practical guide for sport and exercise medicine physicians. Br J Sports Med 2020; 54:1157-1161. [PMID: 32878870 PMCID: PMC7513247 DOI: 10.1136/bjsports-2020-102710] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2020] [Indexed: 12/12/2022]
Abstract
SARS-CoV-2 is the causative virus responsible for the COVID-19 pandemic. This pandemic has necessitated that all professional and elite sport is either suspended, postponed or cancelled altogether to minimise the risk of viral spread. As infection rates drop and quarantine restrictions are lifted, the question how athletes can safely resume competitive sport is being asked. Given the rapidly evolving knowledge base about the virus and changing governmental and public health recommendations, a precise answer to this question is fraught with complexity and nuance. Without robust data to inform policy, return-to-play (RTP) decisions are especially difficult for elite athletes on the suspicion that the COVID-19 virus could result in significant cardiorespiratory compromise in a minority of afflicted athletes. There are now consistent reports of athletes reporting persistent and residual symptoms many weeks to months after initial COVID-19 infection. These symptoms include cough, tachycardia and extreme fatigue. To support safe RTP, we provide sport and exercise medicine physicians with practical recommendations on how to exclude cardiorespiratory complications of COVID-19 in elite athletes who place high demand on their cardiorespiratory system. As new evidence emerges, guidance for a safe RTP should be updated.
Collapse
Affiliation(s)
- Mathew G Wilson
- Institute for Sport Exercise and Health (ISEH), University College Hospital London, London, UK .,The Princess Grace Hospital (HCA Healthcare UK), London, United Kingdom
| | - James H Hull
- Institute for Sport Exercise and Health (ISEH), University College Hospital London, London, UK.,Department of Respiratory Medicine, Royal Brompton Hospital, London, UK.,English Institute of Sport, London, United Kingdom
| | - John Rogers
- Manchester Institute of Health & Performance (MHIP), Manchester, United Kingdom.,Manchester University NHS Foundation Trust, Manchester, United Kingdom.,The Wilmslow Hospital (HCAHealthcareUK), Wilmslow, United Kingdom
| | - Noel Pollock
- Institute for Sport Exercise and Health (ISEH), University College Hospital London, London, UK.,British Athletics, London, United Kingdom
| | - Miranda Dodd
- The Princess Grace Hospital (HCA Healthcare UK), London, United Kingdom
| | - Jemma Haines
- Manchester Institute of Health & Performance (MHIP), Manchester, United Kingdom.,Manchester University NHS Foundation Trust, Manchester, United Kingdom.,NIHR Manchester BRC and University of Manchester, Manchester, United Kingdom
| | - Sally Harris
- Manchester Institute of Health & Performance (MHIP), Manchester, United Kingdom.,The Wilmslow Hospital (HCAHealthcareUK), Wilmslow, United Kingdom
| | - Mike Loosemore
- Institute for Sport Exercise and Health (ISEH), University College Hospital London, London, UK.,English Institute of Sport, London, United Kingdom
| | - Aneil Malhotra
- Manchester Institute of Health & Performance (MHIP), Manchester, United Kingdom.,Manchester University NHS Foundation Trust, Manchester, United Kingdom.,Department of Cardiovascular Science, University of Manchester, Manchester, United Kingdom
| | - Guido Pieles
- Institute for Sport Exercise and Health (ISEH), University College Hospital London, London, UK.,Cardiovascular Biomedical Research Centre, Bristol Heart Institute, Bristol, UK
| | - Anand Shah
- Department of Respiratory Medicine, Royal Brompton Hospital, London, UK.,Department of Infectious Diseases Epidemiology, Imperial College London, London, United Kingdom
| | - Lesley Taylor
- Manchester Institute of Health & Performance (MHIP), Manchester, United Kingdom.,The Wilmslow Hospital (HCAHealthcareUK), Wilmslow, United Kingdom
| | - Aashish Vyas
- Manchester Institute of Health & Performance (MHIP), Manchester, United Kingdom.,Manchester University NHS Foundation Trust, Manchester, United Kingdom.,Lancashire Teaching Hospitals Trust, Lancashire, United Kingdom
| | - Fares S Haddad
- Institute for Sport Exercise and Health (ISEH), University College Hospital London, London, UK.,The Princess Grace Hospital (HCA Healthcare UK), London, United Kingdom.,Department of Trauma and Orthopaedics, University College London Hospitals NHS Foundation Trust, London, London, United Kingdom
| | - Sanjay Sharma
- St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
1954
|
Bertoletti L, Huisman MV. How can we better predict pulmonary blood clots in patients hospitalised for COVID-19? Eur Respir J 2020; 56:2003092. [PMID: 32907888 PMCID: PMC7487270 DOI: 10.1183/13993003.03092-2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/21/2022]
Abstract
The emergence of coronavirus disease 2019 (COVID-19) has put pressure on health systems around the world [1, 2]. This coronavirus has also questioned much of our medical knowledge, with each day seeing the appearance of a new possible clinical expression of the virus [3]. Although its physiopathology is still poorly understood, the vascular tropism of the disease now seems to be a major pathway [4]. Recent studies highlight the development of a specific pulmonary vascular endothelialitis associated with thrombosis and angiogenesis [5]. D-dimer threshold at 2590 ng·mL−1 is able to predict pulmonary embolism in COVID-19 patients with clinical deterioration https://bit.ly/3kYObc3
Collapse
Affiliation(s)
- Laurent Bertoletti
- Service de Médecine Vasculaire et Thérapeutique, CHU de St-Etienne, Saint-Etienne, France
- INSERM, UMR1059, Equipe Dysfonction Vasculaire et Hémostase, Université Jean-Monnet, Saint-Etienne, France
- INSERM, CIC-1408, CHU Saint-Etienne, Saint-Etienne, France
- F-CRIN INNOVTE network, Saint-Etienne, France
| | - Menno V. Huisman
- Dept of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
1955
|
Deigendesch N, Sironi L, Kutza M, Wischnewski S, Fuchs V, Hench J, Frank A, Nienhold R, Mertz KD, Cathomas G, Matter MS, Siegemund M, Tolnay M, Schirmer L, Pröbstel AK, Tzankov A, Frank S. Correlates of critical illness-related encephalopathy predominate postmortem COVID-19 neuropathology. Acta Neuropathol 2020; 140:583-586. [PMID: 32851506 PMCID: PMC7449525 DOI: 10.1007/s00401-020-02213-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Nikolaus Deigendesch
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Lara Sironi
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Michael Kutza
- Department of Neurology and Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Sven Wischnewski
- Department of Neurology and Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Vidmante Fuchs
- Departments of Medicine and Biomedicine, Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jürgen Hench
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Angela Frank
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Ronny Nienhold
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Gieri Cathomas
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Matthias S Matter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Martin Siegemund
- Department of Intensive Care, University Hospital, University of Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Markus Tolnay
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Lucas Schirmer
- Department of Neurology and Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anne-Katrin Pröbstel
- Departments of Medicine and Biomedicine, Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland
| | - Stephan Frank
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Schönbeinstrasse 40, 4031, Basel, Switzerland.
| |
Collapse
|
1956
|
Heffernan KS, Ranadive SM, Jae SY. Exercise as medicine for COVID-19: On PPAR with emerging pharmacotherapy. Med Hypotheses 2020; 143:110197. [PMID: 33017906 PMCID: PMC7430295 DOI: 10.1016/j.mehy.2020.110197] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022]
Abstract
Coronavirus disease 2019 (COVID-19) may have a metabolic origin given strong links with risk factors such as lipids and glucose and co-morbidities such as obesity and type 2 diabetes mellitus. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein mediates viral cellular entry via the ACE2 receptor. The cytoplasmic tail of this spike protein is heavily palmitoylated. Emerging studies suggest that SARS-CoV-2 alters lipid metabolism in the lung epithelial cells by modulating peroxisome proliferator-activated receptor alpha (PPARα), possibly contributing to lipotoxicity, inflammation and untoward respiratory effects. Disruption of this process may affect palmitoylation of SARS-CoV spike protein and thus infectivity and viral assembly. COVID-19 is also increasingly being recognized as a vascular disease, with several studies noting prominent systemic endothelial dysfunction. The pathogenesis of endothelial dysfunction may also be linked to COVID-19-mediated metabolic and inflammatory effects. Herein, exercise will be compared to fenofibrate as a possible therapeutic strategy to bolster resilience against (and help manage recovery from) COVID-19. This paper will explore the hypothesis that exercise may be a useful adjuvant in a setting of COVID-19 management/rehabilitation due to its effects on PPARα and vascular endothelial function.
Collapse
Affiliation(s)
- Kevin S Heffernan
- Department of Exercise Science, Syracuse University, Syracuse, NY, USA.
| | | | - Sae Young Jae
- Department of Sports Sciences, University of Seoul, Seoul, South Korea
| |
Collapse
|
1957
|
Jones GW, Monopoli MP, Campagnolo L, Pietroiusti A, Tran L, Fadeel B. No small matter: a perspective on nanotechnology-enabled solutions to fight COVID-19. Nanomedicine (Lond) 2020; 15:2411-2427. [PMID: 32873192 PMCID: PMC7488724 DOI: 10.2217/nnm-2020-0286] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
There is an urgent need for safe and effective approaches to combat COVID-19. Here, we asked whether lessons learned from nanotoxicology and nanomedicine could shed light on the current pandemic. SARS-CoV-2, the causative agent, may trigger a mild, self-limiting disease with respiratory symptoms, but patients may also succumb to a life-threatening systemic disease. The host response to the virus is equally complex and studies are now beginning to unravel the immunological correlates of COVID-19. Nanotechnology can be applied for the delivery of antiviral drugs or other repurposed drugs. Moreover, recent work has shown that synthetic nanoparticles wrapped with host-derived cellular membranes may prevent virus infection. We posit that nanoparticles decorated with ACE2, the receptor for SARS-CoV-2, could be exploited as decoys to intercept the virus before it infects cells in the respiratory tract. However, close attention should be paid to biocompatibility before such nano-decoys are deployed in the clinic.
Collapse
Affiliation(s)
| | - Marco P Monopoli
- Department of Chemistry, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Luisa Campagnolo
- Department of Biomedicine & Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonio Pietroiusti
- Department of Biomedicine & Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Lang Tran
- Institute of Occupational Medicine, Edinburgh, EH14 4AP, UK
| | - Bengt Fadeel
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
1958
|
Dai YJ, Zhang WN, Wang WD, He SY, Liang CC, Wang DW. Comprehensive analysis of two potential novel SARS-CoV-2 entries, TMPRSS2 and IFITM3, in healthy individuals and cancer patients. Int J Biol Sci 2020; 16:3028-3036. [PMID: 33061814 PMCID: PMC7545701 DOI: 10.7150/ijbs.51234] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/20/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2, with acute respiratory failure as the most significant symptom, has led to a global pandemic. Angiotensin-converting enzyme 2 (ACE2) is considered as the most important receptor of SARS-CoV-2 and wildly expressed in human tissues. Whereas, the extremely low expression of ACE2 in lung could hardly interpret the severe symptom of pneumonia in COVID-19 patients. Here we profiled two SARS-CoV-2 infection related genes, the transmembrane serine protease 2 (TMPRSS2) and the interferon-inducible transmembrane protein 3 (IFITM3), in human tissues and organs. Consistent with the expression and distribution of ACE2, TMPRSS2 was also highly expressed in digestive, urinary and reproductive systems, but low expressed in lung. Notably, the anti-virus protein IFITM3 also expressed much lower in lung than other tissues, which might be related to the severe lung symptoms of COVID-19. In addition, the low expression of IFITM3 in immune cells suggested that SARS-CoV-2 might attack lymphocytes and induce the cytokine release syndrome (CRS). Furthermore, cancer patients were considered as more susceptible to SARS-CoV-2 infection. Our data supposed that fourteen types of tumors might have different susceptibility to the virus according to ACE2, TMPRSS2 and IFITM3 expression patterns. Interestingly the prognosis of six types of cancers including breast carcinoma (BRCA), lung adenocarcinoma (LUAD), uterine corpus endometrial carcinoma (UCEC), renal clear cell carcinoma (KIRC), prostate adenocarcinoma (PRAD), and hepatocellular carcinoma (LIHC) were closely related to these gene expressions. Our study explored the expression and distribution profiles of two potential novel molecules that might participate in SARS-CoV-2 infection and involved in immunity, which may provide a functional basis for preventing infection of SARS-CoV-2.
Collapse
Affiliation(s)
- Yu-Jun Dai
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wei-Na Zhang
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wei-Da Wang
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Si-Yuan He
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Cheng-Cai Liang
- Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China, Guangzhou, 510060, China
| | - Da-Wei Wang
- National Research Center for Translational Medicine, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 197, Ruijin Road II, Shanghai, 200025, China
| |
Collapse
|
1959
|
Caruso A, Caccuri F, Bugatti A, Zani A, Vanoni M, Bonfanti P, Cazzaniga ME, Perno CF, Messa C, Alberghina L. Methotrexate inhibits SARS-CoV-2 virus replication "in vitro". J Med Virol 2020; 93:1780-1785. [PMID: 32926453 PMCID: PMC7891346 DOI: 10.1002/jmv.26512] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/31/2022]
Abstract
In early 2020 the new respiratory syndrome COVID‐19 (caused by the zoonotic SARS‐CoV‐2 virus) spread like a pandemic, starting from Wuhan, China, causing severe economic depression. Despite some advances in drug treatments of medical complications in the later stages of the disease, the pandemic's death toll is tragic, as no vaccine or specific antiviral treatment is currently available. By using a systems approach, we identify the host‐encoded pathway, which provides ribonucleotides to viral RNA synthesis, as a possible target. We show that methotrexate, an FDA‐approved inhibitor of purine biosynthesis, potently inhibits viral RNA replication, viral protein synthesis, and virus release. The effective antiviral methotrexate concentrations are similar to those used for established human therapies using the same drug. Methotrexate should be most effective in patients at the earliest appearance of symptoms to effectively prevent viral replication, diffusion of the infection, and possibly fatal complications. The COVID‐19 pandemic caused by the SARS‐CoV‐2 virus is a global public health threat causing over 800000 deaths, millions of infected people, and severe economic depression. No vaccine or specific antiviral treatments are currently available. By inhibiting the host‐encoded pathway, which provides ribonucleotides to viral RNA synthesis, the FDA‐approved drug methotrexate efficiently blocks SARS‐CoV‐2 replication.
Collapse
Affiliation(s)
- Arnaldo Caruso
- Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Francesca Caccuri
- Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Antonella Bugatti
- Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Alberto Zani
- Department of Molecular and Translational Medicine, Section of Microbiology and Virology, University of Brescia Medical School, Brescia, Italy
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, ISBE.IT-SYSBIO/Centre of Systems Biology, University of Milano Bicocca, Milano, Italy
| | - Paolo Bonfanti
- School of Medicine and Surgery, University of Milano Bicocca, Milano, Italy.,Clinic of Infectious Diseases-ASST Monza, Monza, Italy
| | - Marina E Cazzaniga
- School of Medicine and Surgery, University of Milano Bicocca, Milano, Italy.,ASST Monza, Phase 1 Research Unit, Monza, Italy
| | | | - Cristina Messa
- School of Medicine and Surgery, University of Milano Bicocca, Milano, Italy.,Tecnomed Foundation, Milan, Italy
| | - Lilia Alberghina
- Department of Biotechnology and Biosciences, ISBE.IT-SYSBIO/Centre of Systems Biology, University of Milano Bicocca, Milano, Italy
| |
Collapse
|
1960
|
Development and Optimization of In-house ELISA for Detection of Human IgG Antibody to SARS-CoV-2 Full Length Spike Protein. Pathogens 2020; 9:pathogens9100803. [PMID: 32998438 PMCID: PMC7601663 DOI: 10.3390/pathogens9100803] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 12/18/2022] Open
Abstract
The ongoing coronavirus disease 19 (COVID-19) pandemic, caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), poses a threat to human health. Despite this, many affected countries are now in the process of gradual lifting of COVID-19 restrictions that were initially implemented in response to the pandemic. The success of the so-called “exit strategy” requires continued surveillance of virus circulation in the community and evaluation of the prevalence of protective immunity among population. Serology tests are valuable tools for these purposes. Herein, SARS-CoV-2 full-length spike (S) recombinant protein was utilized to develop and optimize an indirect enzyme-linked immunoassay (ELISA) that enables a reliable detection of virus-specific IgG antibody in human sera. Importantly, the performance of this assay was evaluated utilizing micro-neutralization (MN) assay as a reference test. Our developed ELISA offers 100% sensitivity, 98.4% specificity, 98.8% agreement, and high overall accuracy. Moreover, the optical density (OD) values of positive samples significantly correlated with their MN titers. The assay specifically detects human IgG antibodies directed against SARS-CoV-2, but not those to Middle East respiratory syndrome coronavirus (MERS-CoV) or human coronavirus HKU1 (HCoV-HKU1). The availability of this in-house ELISA protocol would be valuable for various diagnostic and epidemiological applications.
Collapse
|
1961
|
Rauch A, Dupont A, Goutay J, Caplan M, Staessens S, Moussa M, Jeanpierre E, Corseaux D, Lefevre G, Lassalle F, Faure K, Lambert M, Duhamel A, Labreuche J, Garrigue D, De Meyer SF, Staels B, Van Belle E, Vincent F, Kipnis E, Lenting PJ, Poissy J, Susen S. Endotheliopathy Is Induced by Plasma From Critically Ill Patients and Associated With Organ Failure in Severe COVID-19. Circulation 2020; 142:1881-1884. [PMID: 32970476 PMCID: PMC7643783 DOI: 10.1161/circulationaha.120.050907] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Antoine Rauch
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen)
| | - Annabelle Dupont
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen)
| | - Julien Goutay
- Intensive Care Department, Pôle de Réanimation (J.G., M.C.), Centre Hospitalier Universitaire Lille, France
| | - Morgan Caplan
- Intensive Care Department, Pôle de Réanimation (J.G., M.C.), Centre Hospitalier Universitaire Lille, France
| | - Senna Staessens
- Centre national de la recherche scientifique, Surgical Critical Care, Department of Anesthesiology and Critical Care, U1019 - Unité mixte de recherche 9017 - Center for Infection and Immunity of Lille (E.K.), Institut Pasteur de Lille, UMR1011-EGID (S. Staessens, D.C., B.S.), Univ. Lille, CHU Lille, France.,Laboratory for Thrombosis Research, Katholieke Universiteit Leuven Campus Kulak Kortrijk, Belgium (S. Staessens, S.F.D.M.)
| | - Mouhamed Moussa
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen).,Surgical Critical Care, Department of Anesthesiology and Critical Care (M.M.), Univ. Lille, CHU Lille, France
| | - Emmanuelle Jeanpierre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen)
| | - Delphine Corseaux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen).,Centre national de la recherche scientifique, Surgical Critical Care, Department of Anesthesiology and Critical Care, U1019 - Unité mixte de recherche 9017 - Center for Infection and Immunity of Lille (E.K.), Institut Pasteur de Lille, UMR1011-EGID (S. Staessens, D.C., B.S.), Univ. Lille, CHU Lille, France
| | - Guillaume Lefevre
- Institut d'Immunologie, U1286 - INFINITE - Institute for Translational Research in Inflammation (G.L.), ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales (A. Duhamel, J.L.), Univ. Lille, CHU Lille, France
| | - Fanny Lassalle
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen)
| | - Karine Faure
- Department of Infectious Diseases (K.F.), Univ. Lille, CHU Lille, France
| | - Marc Lambert
- Department of Internal Medicine, Institut national de la santé et de la recherche médicale U 1167 (M.L.), Univ. Lille, CHU Lille, France
| | - Alain Duhamel
- Institut d'Immunologie, U1286 - INFINITE - Institute for Translational Research in Inflammation (G.L.), ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales (A. Duhamel, J.L.), Univ. Lille, CHU Lille, France
| | - Julien Labreuche
- Institut d'Immunologie, U1286 - INFINITE - Institute for Translational Research in Inflammation (G.L.), ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales (A. Duhamel, J.L.), Univ. Lille, CHU Lille, France
| | - Delphine Garrigue
- Surgical Critical Care, Department of Anesthesiology and Critical Care (D.G.), Centre Hospitalier Universitaire Lille, France
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, Katholieke Universiteit Leuven Campus Kulak Kortrijk, Belgium (S. Staessens, S.F.D.M.)
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen).,Centre national de la recherche scientifique, Surgical Critical Care, Department of Anesthesiology and Critical Care, U1019 - Unité mixte de recherche 9017 - Center for Infection and Immunity of Lille (E.K.), Institut Pasteur de Lille, UMR1011-EGID (S. Staessens, D.C., B.S.), Univ. Lille, CHU Lille, France
| | - Eric Van Belle
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen).,Department Cardiology (E.V.B., F.V.), Univ. Lille, CHU Lille, France
| | - Flavien Vincent
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen)
| | - Eric Kipnis
- Centre national de la recherche scientifique, Surgical Critical Care, Department of Anesthesiology and Critical Care, U1019 - Unité mixte de recherche 9017 - Center for Infection and Immunity of Lille (E.K.), Institut Pasteur de Lille, UMR1011-EGID (S. Staessens, D.C., B.S.), Univ. Lille, CHU Lille, France
| | | | - Julien Poissy
- Institut national de la santé et de la recherche médicale U1285, Pôle de réanimation, Centre national de la recherche scientifique, Unité mixte de recherche 8576 - Unité de Glycobiologie Structurale et Fonctionnelle (J.P.), Univ. Lille, CHU Lille, France
| | - Sophie Susen
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France (A.R., A. Dupont, M.M., E.J., D.C., F.L., B.S., E.V.B., F.V., S. Susen)
| | | | | |
Collapse
|
1962
|
Affiliation(s)
- Eric J. Topol
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA, USA
| |
Collapse
|
1963
|
Affiliation(s)
- Rajesh Bhatia
- Former Director, Communicable Diseases, World Health Organization South-East Asia Regional Office, New Delhi 110 002, India
| | - Priya Abraham
- Director, ICMR-National Institute of Virology, Pune 411 001, Maharashtra, India
| |
Collapse
|
1964
|
Sheha D, El-Shayeb M, Eid Y, Amin M, Saeed A, Abdou D, Aly T, Samy S, Elziaty R, Aboelyazed S, Osman A, Sheha A. Unfolding of sickle cell trait by coronavirus disease 2019 (COVID-19) infection. Br J Haematol 2020; 191:e38-e40. [PMID: 32966591 PMCID: PMC7537001 DOI: 10.1111/bjh.17089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/16/2020] [Indexed: 11/27/2022]
Affiliation(s)
- Dina Sheha
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed El-Shayeb
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Yara Eid
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mariam Amin
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Alia Saeed
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Dina Abdou
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tarek Aly
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherief Samy
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Rahma Elziaty
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherihan Aboelyazed
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed Osman
- Department of Radiodiagnosis, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Aliaa Sheha
- Department of Radiodiagnosis, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
1965
|
Singh M, Bansal V, Feschotte C. A Single-Cell RNA Expression Map of Human Coronavirus Entry Factors. Cell Rep 2020; 32:108175. [PMID: 32946807 PMCID: PMC7470764 DOI: 10.1016/j.celrep.2020.108175] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/15/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
To predict the tropism of human coronaviruses, we profile 28 SARS-CoV-2 and coronavirus-associated receptors and factors (SCARFs) using single-cell transcriptomics across various healthy human tissues. SCARFs include cellular factors both facilitating and restricting viral entry. Intestinal goblet cells, enterocytes, and kidney proximal tubule cells appear highly permissive to SARS-CoV-2, consistent with clinical data. Our analysis also predicts non-canonical entry paths for lung and brain infections. Spermatogonial cells and prostate endocrine cells also appear to be permissive to SARS-CoV-2 infection, suggesting male-specific vulnerabilities. Both pro- and anti-viral factors are highly expressed within the nasal epithelium, with potential age-dependent variation, predicting an important battleground for coronavirus infection. Our analysis also suggests that early embryonic and placental development are at moderate risk of infection. Lastly, SCARF expression appears broadly conserved across a subset of primate organs examined. Our study establishes a resource for investigations of coronavirus biology and pathology.
Collapse
Affiliation(s)
- Manvendra Singh
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| | - Vikas Bansal
- Biomedical Data Science and Machine Learning Group, German Center for Neurodegenerative Diseases, Tübingen 72076, Germany; Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen 37075, Germany.
| | - Cédric Feschotte
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
1966
|
Mlcochova P, Collier D, Ritchie A, Assennato SM, Hosmillo M, Goel N, Meng B, Chatterjee K, Mendoza V, Temperton N, Kiss L, James LC, Ciazynska KA, Xiong X, Briggs JA, Nathan JA, Mescia F, Bergamaschi L, Zhang H, Barmpounakis P, Demeris N, Skells R, Lyons PA, Bradley J, Baker S, Allain JP, Smith KG, Bousfield R, Wilson M, Sparkes D, Amoroso G, Gkrania-Klotsas E, Hardwick S, Boyle A, Goodfellow I, Gupta RK. Combined Point-of-Care Nucleic Acid and Antibody Testing for SARS-CoV-2 following Emergence of D614G Spike Variant. Cell Rep Med 2020; 1:100099. [PMID: 32905045 PMCID: PMC7462534 DOI: 10.1016/j.xcrm.2020.100099] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/05/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022]
Abstract
Rapid COVID-19 diagnosis in the hospital is essential, although this is complicated by 30%-50% of nose/throat swabs being negative by SARS-CoV-2 nucleic acid amplification testing (NAAT). Furthermore, the D614G spike mutant dominates the pandemic and it is unclear how serological tests designed to detect anti-spike antibodies perform against this variant. We assess the diagnostic accuracy of combined rapid antibody point of care (POC) and nucleic acid assays for suspected COVID-19 disease due to either wild-type or the D614G spike mutant SARS-CoV-2. The overall detection rate for COVID-19 is 79.2% (95% CI 57.8-92.9) by rapid NAAT alone. The combined point of care antibody test and rapid NAAT is not affected by D614G and results in very high sensitivity for COVID-19 diagnosis with very high specificity.
Collapse
Affiliation(s)
- Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dami Collier
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Allyson Ritchie
- Diagnostics for the Real World EU, Chesterford Research Park, UK
| | | | - Myra Hosmillo
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Neha Goel
- Diagnostics for the Real World EU, Chesterford Research Park, UK
| | - Bo Meng
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Vivien Mendoza
- NIHR Cambridge Clinical Research Facility, Cambridge, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Kent, UK
| | - Leo Kiss
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Leo C. James
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | | - Xiaoli Xiong
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - John A.G. Briggs
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - James A. Nathan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Federica Mescia
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hongyi Zhang
- Clinical Microbiology & Public Health Laboratory, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
| | - Petros Barmpounakis
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
| | - Nikos Demeris
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
- Cambridge Clinical Trials Unit-Cancer Theme, University of Cambridge, Cambridge, UK
| | - Richard Skells
- NIHR Cambridge Clinical Research Facility, Cambridge, UK
- Cambridge Clinical Trials Unit-Cancer Theme, University of Cambridge, Cambridge, UK
| | - Paul A. Lyons
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - John Bradley
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
- National Institutes for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Steven Baker
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Kenneth G.C. Smith
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rachel Bousfield
- Department of Infectious Diseases, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
| | - Michael Wilson
- Department of Infectious Diseases, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
| | - Dominic Sparkes
- Department of Infectious Diseases, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
| | - Glenn Amoroso
- Department of Emergency Medicine, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
| | - Effrosyni Gkrania-Klotsas
- Department of Infectious Diseases, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
| | - Susie Hardwick
- Department of Emergency Medicine, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
| | - Adrian Boyle
- Department of Emergency Medicine, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
| | - Ian Goodfellow
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ravindra K. Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Infectious Diseases, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
- Africa Health Research Institute, Durban, South Africa
| |
Collapse
|
1967
|
Affiliation(s)
- Elia J Duh
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
1968
|
Timmis K, Huang WE, Timmis J. Strategies to minimize preventable morbidity and mortality resulting from pandemics like
COVID
‐19. Environ Microbiol 2020; 22:4085-4092. [DOI: 10.1111/1462-2920.15217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Kenneth Timmis
- Institute of Microbiology, Technical University Braunschweig Braunschweig Germany
| | - Wei E. Huang
- Department of Engineering Science University of Oxford Oxford UK
| | - James Timmis
- Athena Institute, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| |
Collapse
|
1969
|
Abstract
The pandemic of Coronavirus disease 2019 (COVID-19) is rapidly progressing, causing significant morbidity and mortality. Various antiviral drugs, anti-inflammatory drugs and immunomodulators have been tried without substantial clinical benefits. The severe and critical cases of COVID-19 disease are characterised by gut microbiome dysbiosis, immune dysregulation, hyper-inflammation and hypercytokinaemia (cytokine storm). Therefore, the strategies which target these pathophysiological processes may be beneficial. Probiotics are one such strategy that exerts beneficial effects by manipulation of the gut microbiota, suppression of opportunistic pathogens in the gut, decreasing translocation of opportunistic organisms, activation of mucosal immunity and modulation of the innate and adaptive immune response. Probiotics are the potential candidates to be tested in moderate and severe cases of COVID-19 due to several beneficial effects, including easy availability, easy to administer, safe and economical to use.
Collapse
|
1970
|
Inde Z, Yapp C, Joshi GN, Spetz J, Fraser C, Deskin B, Ghelfi E, Sodhi C, Hackam DJ, Kobzik L, Croker BA, Brownfield D, Jia H, Sarosiek KA. Age-dependent regulation of SARS-CoV-2 cell entry genes and cell death programs correlates with COVID-19 disease severity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.09.13.276923. [PMID: 32935109 PMCID: PMC7491524 DOI: 10.1101/2020.09.13.276923] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) maintains cardiovascular and renal homeostasis but also serves as the entry receptor for the novel severe acute respiratory syndrome coronavirus (SARS-CoV-2), the causal agent of novel coronavirus disease 2019 (COVID-19). COVID-19 disease severity is typically lower in pediatric patients than adults (particularly the elderly), but higher rates of hospitalizations requiring intensive care are observed in infants than in older children - the reasons for these differences are unknown. ACE2 is expressed in several adult tissues and cells, including alveolar type 2 cells of the distal lung epithelium, but expression at other ages is largely unexplored. Here we show that ACE2 transcripts are expressed in the lung and trachea shortly after birth, downregulated during childhood, and again expressed at high levels in late adulthood. Notably, the repertoire of cells expressing ACE2 protein in the mouse lung and airways shifts during key phases of lung maturation. In particular, podoplanin-positive cells, which are likely alveolar type I cells responsible for gas exchange, express ACE2 only in advanced age. Similar patterns of expression were evident in analysis of human lung tissue from over 100 donors, along with extreme inter- and intra-individual heterogeneity in ACE2 protein expression in epithelial cells. Furthermore, we find that apoptosis, which is a natural host defense system against viral infection, is dynamically regulated during lung maturation, resulting in periods of heightened apoptotic priming and dependence on pro-survival BCL-2 family proteins including MCL-1. Infection of human lung cells with SARS-CoV-2 triggers an unfolded protein stress response and upregulation of the endogenous MCL-1 inhibitor Noxa; in young individuals, MCL-1 inhibition is sufficient to trigger apoptosis in lung epithelial cells and may thus limit virion production and inflammatory signaling. Overall, we identify strong and distinct correlates of COVID-19 disease severity across lifespan and advance our understanding of the regulation of ACE2 and cell death programs in the mammalian lung. Furthermore, our work provides the framework for translation of apoptosis modulating drugs as novel treatments for COVID-19.
Collapse
Affiliation(s)
- Zintis Inde
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
- John B. Little Center for Radiation Sciences, Harvard School of Public Health, Boston, MA
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA
| | - Clarence Yapp
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA
- Image and Data Analysis Core, Harvard Medical School, Boston, MA
| | - Gaurav N. Joshi
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
- John B. Little Center for Radiation Sciences, Harvard School of Public Health, Boston, MA
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA
- Integrated Cellular Imaging Core, Emory University, Atlanta, GA
| | - Johan Spetz
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
- John B. Little Center for Radiation Sciences, Harvard School of Public Health, Boston, MA
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA
| | - Cameron Fraser
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
- John B. Little Center for Radiation Sciences, Harvard School of Public Health, Boston, MA
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA
| | - Brian Deskin
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
| | - Elisa Ghelfi
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
| | - Chhinder Sodhi
- Department of Surgery, Johns Hopkins University, Baltimore, MD
| | - David J. Hackam
- Department of Surgery, Johns Hopkins University, Baltimore, MD
| | - Lester Kobzik
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
| | - Ben A. Croker
- Division of Allergy, Immunology and Rheumatology, University of California, San Diego, CA
| | - Douglas Brownfield
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
| | - Hongpeng Jia
- Department of Surgery, Johns Hopkins University, Baltimore, MD
| | - Kristopher A. Sarosiek
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA
- John B. Little Center for Radiation Sciences, Harvard School of Public Health, Boston, MA
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA
| |
Collapse
|
1971
|
Algassim AA, Elghazaly AA, Alnahdi AS, Mohammed-Rahim OM, Alanazi AG, Aldhuwayhi NA, Alanazi MM, Almutairi MF, Aldeailej IM, Kamli NA, Aljurf MD. Prognostic significance of hemoglobin level and autoimmune hemolytic anemia in SARS-CoV-2 infection. Ann Hematol 2020; 100:37-43. [PMID: 32918594 PMCID: PMC7486165 DOI: 10.1007/s00277-020-04256-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
Higher levels of D-dimer, LDH, and ferritin, all have been associated with the poor prognosis of COVID-19. In a disease where there are acute inflammation and compromised oxygenation, we investigated the impact of initial hemoglobin (Hgb) levels at Emergency Department (ED) triage on the severity and the clinical course of COVID-19. We conducted a cross-sectional study on 601 COVID-19 patients in a COVID-19 national referral center between 13 and 27 June 2020. All adult patients presented at our hospital that required admission or hotel isolation were included in this study. Patients admitted to the intensive care unit (ICU) had a lower initial Hgb than those admitted outside the ICU (12.84 g/dL vs. 13.31 g/dL, p = 0.026) and over the course of admission; the prevalence of anemia (Hgb < 12.5 g/dL) was 65% in patients admitted to ICU, whereas it was only 43% in non-ICU patients (odds ratio of 2.464, 95% CI 1.71-3.52). Anemic ICU patients had a higher mortality compared with non-anemic ICU patients (hazard ratio = 1.88, log-rank p = 0.0104). A direct agglutination test (DAT) for all anemic patients showed that 14.7% of ICU patients and 9% of non-ICU patients had autoimmune hemolytic anemia (AIHA). AIHA patients had significantly longer length of hospital stay compared with anemic patients without AIHA (17.1 days vs. 14.08 days, p = 0.034). Lower Hgb level at hospital presentation could be a potential surrogate for COVID-19 severity.
Collapse
Affiliation(s)
- Abdulrahman A Algassim
- Department of Medicine, King Saud Medical City, Riyadh, Saudi Arabia. .,Department of Medicine, Prince Sultan Military Medical City, Riyadh, Saudi Arabia.
| | - Assem A Elghazaly
- Department of Hematology and Oncology, King Saud Medical City, Riyadh, Saudi Arabia
| | | | | | - Abdulaziz G Alanazi
- Department of Medicine, King Saud Medical City, Riyadh, Saudi Arabia.,Department of Medicine, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Nawaf A Aldhuwayhi
- Department of Medicine, King Saud Medical City, Riyadh, Saudi Arabia.,Department of Medicine, Prince Mohammed Medical City, Sakaka, Al-Jouf, Saudi Arabia
| | - Mashael M Alanazi
- Regional Laboratory and Blood Bank, Ministry of Health, Riyadh, Saudi Arabia
| | | | - Ibrahim M Aldeailej
- Regional Laboratory and Blood Bank, Ministry of Health, Riyadh, Saudi Arabia
| | - Najeeb A Kamli
- Regional Laboratory and Blood Bank, Ministry of Health, Riyadh, Saudi Arabia
| | - Mahmoud D Aljurf
- Oncology Center, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
1972
|
Aragão MT, dos Santos EL, Ataide TD, Alves JS, Aragão NVBT. COVID-19 presenting as an exanthematic disease: a case report. Rev Soc Bras Med Trop 2020; 53:e20200533. [PMID: 32935788 PMCID: PMC7491557 DOI: 10.1590/0037-8682-0533-2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 01/10/2023] Open
Abstract
Coronavirus disease (COVID-19) is caused by SARS-CoV-2 and has spread rapidly and caused a global pandemic. Knowledge about clinical and laboratory manifestations in the pediatric population is necessary to guide and monitor such patients. A 3-year-old female patient diagnosed with COVID-19 presented with high fever. After defervescence, she experienced a maculopapular rash that worsened by the sixth day of the disease with self-limited evolution without relevant laboratory changes. The identification of rashes in children with COVID-19 is an unusual and important condition that must be recognized in view of the high transmissibility shown.
Collapse
Affiliation(s)
| | | | | | - José Seabra Alves
- Universidade Tiradentes, Departamento de Medicina, Aracaju, SE, Brasil
| | | |
Collapse
|
1973
|
Wendt R, Kalbitz S, Lübbert C, Kellner N, Macholz M, Schroth S, Ermisch J, Latosisnka A, Arnold B, Mischak H, Beige J, Metzger J. Urinary Proteomics Associates with COVID-19 Severity: Pilot Proof-of-Principle Data and Design of a Multicentric Diagnostic Study. Proteomics 2020; 20:e2000202. [PMID: 32960510 DOI: 10.1002/pmic.202000202] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/26/2020] [Indexed: 12/11/2022]
Abstract
SARS-CoV-2 infection results in a mild-to-moderate disease course in most patients, allowing outpatient self-care and quarantine. However, in approx. 10% of cases a two- or three-phasic critical disease course with starting from day 7 to 10 is observed. To facilitate and plan outpatient care, biomarkers prognosing such worsening at an early stage appear of outmost importance. In this accelerated article, we report on the identification of urinary peptides significantly associated with SARS-CoV-2 infection, and the development of a multi-marker urinary peptide based test, COVID20, that may enable prognosis of critical and fatal outcomes in COVID-19 patients. COVID20 is composed of 20 endogenous peptides mainly derived from various collagen chains that enable differentiating moderate or severe disease from critical state or death with 83% sensitivity at 100% specificity. Based on the performance in this pilot study, testing in a prospective study on 1000 patients has been initiated. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Leipzig, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Leipzig, Germany
- Department of Infectious Diseases and Tropical Medicine, Leipzig University Hospital, Leipzig, Germany
| | - Nils Kellner
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Leipzig, Germany
| | - Martin Macholz
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Leipzig, Germany
| | - Stefanie Schroth
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Leipzig, Germany
| | - Jörg Ermisch
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Leipzig, Germany
| | | | - Benjamin Arnold
- Department of Infectious Diseases/Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Leipzig, Germany
| | - Harald Mischak
- Mosaiques-Diagnostics GmbH, Hannover, Germany
- Institute of Cardiovascular and Medical Sciences, Glasgow, United Kingdom
| | - Joachim Beige
- Martin-Luther-University Halle/Wittenberg, Halle, Germany
| | | |
Collapse
|
1974
|
Hennighausen L, Lee HK. Activation of the SARS-CoV-2 Receptor Ace2 through JAK/STAT-Dependent Enhancers during Pregnancy. Cell Rep 2020; 32:108199. [PMID: 32966801 PMCID: PMC7474886 DOI: 10.1016/j.celrep.2020.108199] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/20/2020] [Accepted: 09/03/2020] [Indexed: 01/04/2023] Open
Abstract
ACE2 binds the coronavirus SARS-CoV-2 and facilitates its cellular entry. Interferons activate ACE2 expression in pneumocytes, suggesting a critical role of cytokines in SARS-CoV-2 target cells. Viral RNA was detected in breast milk in at least seven studies, raising the possibility that ACE2 is expressed in mammary tissue during lactation. Here, we show that Ace2 expression in mouse mammary tissue is induced during pregnancy and lactation, which coincides with the activation of intronic enhancers. These enhancers are occupied by the prolactin-activated transcription factor STAT5 and additional regulatory factors, including RNA polymerase II. Deletion of Stat5a results in decommissioning of the enhancers and an 83% reduction of Ace2 mRNA. We also demonstrate that Ace2 expression increases during lactation in lung, but not in kidney and intestine. JAK/STAT components are present in a range of SARS-CoV-2 target cells, opening the possibility that cytokines contribute to the viral load and extrapulmonary pathophysiology. Ace2 expression is induced in the mammary glands of pregnant and lactating mice Gene enhancers are activated by the prolactin-activated transcription factors STAT5A/B Deletion of the Stat5a gene mitigates enhancer formation and Ace2 expression Ace2 levels also increase in lung tissue during lactation
Collapse
Affiliation(s)
- Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, U.S. National Institutes of Health, Bethesda, MD 20892, USA.
| | - Hye Kyung Lee
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, U.S. National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
1975
|
National Psoriasis Foundation COVID-19 Task Force Guidance for Management of Psoriatic Disease During the Pandemic: Version 1. J Am Acad Dermatol 2020; 83:1704-1716. [PMID: 32891785 PMCID: PMC7471802 DOI: 10.1016/j.jaad.2020.09.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 02/08/2023]
Abstract
Objective To provide guidance about management of psoriatic disease during the coronavirus disease 2019 (COVID-19) pandemic. Study design A task force (TF) of 18 physician voting members with expertise in dermatology, rheumatology, epidemiology, infectious diseases, and critical care was convened. The TF was supplemented by nonvoting members, which included fellows and National Psoriasis Foundation (NPF) staff. Clinical questions relevant to the psoriatic disease community were informed by questions received by the NPF. A Delphi process was conducted. Results The TF approved 22 guidance statements. The average of the votes was within the category of agreement for all statements. All guidance statements proposed were recommended, 9 with high consensus and 13 with moderate consensus. Limitations The evidence behind many guidance statements is limited in quality. Conclusion These statements provide guidance for the management of patients with psoriatic disease on topics ranging from how the disease and its treatments impact COVID-19 risk and outcome, how medical care can be optimized during the pandemic, what patients should do to lower their risk of getting infected with severe acute respiratory syndrome coronavirus 2 and what they should do if they develop COVID-19. The guidance is intended to be a living document that will be updated by the TF as data emerge.
Collapse
|
1976
|
Reyes M, Filbin MR, Bhattacharyya RP, Sonny A, Mehta A, Billman K, Kays KR, Pinilla-Vera M, Benson ME, Cosimi LA, Hung DT, Levy BD, Villani AC, Sade-Feldman M, Baron RM, Goldberg MB, Blainey PC, Hacohen N. Induction of a regulatory myeloid program in bacterial sepsis and severe COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32908980 DOI: 10.1101/2020.09.02.280180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
A recent estimate suggests that one in five deaths globally are associated with sepsis 1 . To date, no targeted treatment is available for this syndrome, likely due to substantial patient heterogeneity 2,3 and our lack of insight into sepsis immunopathology 4 . These issues are highlighted by the current COVID-19 pandemic, wherein many clinical manifestations of severe SARS-CoV-2 infection parallel bacterial sepsis 5-8 . We previously reported an expanded CD14+ monocyte state, MS1, in patients with bacterial sepsis or non-infectious critical illness, and validated its expansion in sepsis across thousands of patients using public transcriptomic data 9 . Despite its marked expansion in the circulation of bacterial sepsis patients, its relevance to viral sepsis and association with disease outcomes have not been examined. In addition, the ontogeny and function of this monocyte state remain poorly characterized. Using public transcriptomic data, we show that the expression of the MS1 program is associated with sepsis mortality and is up-regulated in monocytes from patients with severe COVID-19. We found that blood plasma from bacterial sepsis or COVID-19 patients with severe disease induces emergency myelopoiesis and expression of the MS1 program, which are dependent on the cytokines IL-6 and IL-10. Finally, we demonstrate that MS1 cells are broadly immunosuppressive, similar to monocytic myeloid-derived suppressor cells (MDSCs), and have decreased responsiveness to stimulation. Our findings highlight the utility of regulatory myeloid cells in sepsis prognosis, and the role of systemic cytokines in inducing emergency myelopoiesis during severe bacterial and SARS-CoV-2 infections.
Collapse
|
1977
|
Zisook RE, Monnot A, Parker J, Gaffney S, Dotson S, Unice K. Assessing and managing the risks of COVID-19 in the workplace: Applying industrial hygiene (IH)/occupational and environmental health and safety (OEHS) frameworks. Toxicol Ind Health 2020; 36:607-618. [PMID: 33085585 PMCID: PMC7578844 DOI: 10.1177/0748233720967522] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/02/2022]
Abstract
As businesses attempt to reopen to varying degrees amid the current coronavirus disease (COVID-19) pandemic, industrial hygiene (IH) and occupational and environmental health and safety (OEHS) professionals have been challenged with assessing and managing the risks of COVID-19 in the workplace. In general, the available IH/OEHS tools were designed to control hazards originating in the workplace; however, attempts to tailor them specifically to the control of infectious disease outbreaks have been limited. This analysis evaluated the IH decision-making framework (Anticipate, Recognize, Evaluate, Control, and Confirm ("ARECC")) as it relates to biological hazards, in general, and to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), specifically. Available IH/OEHS risk assessment and risk management tools (e.g. control banding and the hierarchy of controls) are important components of the ARECC framework. These conceptual models, however, were primarily developed for controlling chemical hazards and must be adapted to the unique characteristics of highly infectious and virulent pathogens, such as SARS-CoV-2. This assessment provides an overview of the key considerations for developing occupational infection control plans, selecting the best available controls, and applying other emerging tools (e.g. quantitative microbial risk assessment), with the ultimate goal of facilitating risk management decisions during the current global pandemic.
Collapse
|
1978
|
Smith R, Geary SM, Salem AK. Implications of current and future approaches to coronavirus disease 2019 testing. Future Virol 2020; 15:551-556. [PMID: 33193806 PMCID: PMC7560716 DOI: 10.2217/fvl-2020-0318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/30/2020] [Indexed: 11/21/2022]
Affiliation(s)
- Rasheid Smith
- Division of Pharmaceutics & Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Sean M Geary
- Division of Pharmaceutics & Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Aliasger K Salem
- Division of Pharmaceutics & Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
1979
|
Abstract
This article explains how the mortality rate of an illness such as Coronavirus Disease 2019 (COVID-19) is calculated as well as how the definition of what is a "case" has changed from the earliest days of the pandemic to now. Many factors were not known about The Sudden Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) which causes COVID-19 at the beginning of the pandemic because it is a novel human pathogen. One key factor that was not known in the earliest days of the pandemic was that many patients are either asymptomatic or have symptoms so mild that they may not seek medical attention and hence these patients would not be identified as a "case" if that term is defined as being sufficiently symptomatic to be seeking medical attention. Cases in the earliest days of the pandemic were defined as based on having symptoms (eg, fever, cough, respiratory distress) after ruling out other possible causes. Cases now are defined by tests confirming that the person is shedding the SARS-CoV-2 (ie, a laboratory vs. a symptomatic diagnosis). The mortality rate of this virus dropped as a function of this change. On the basis of the results of an unintended, naturalistic experiment on an expeditionary cruise in March of 2020, there was more than a 5-fold drop in the calculated mortality rate due to this definitional change in what constituted a case. This column explains this issue and discusses its implications for effectively dealing with the SARS-CoV-2 (or COVID-19) pandemic.
Collapse
|
1980
|
Rajdev K, Lahan S, Klein K, Piquette CA, Thi M. Acute Ischemic and Hemorrhagic Stroke in COVID-19: Mounting Evidence. Cureus 2020; 12:e10157. [PMID: 33014653 PMCID: PMC7526955 DOI: 10.7759/cureus.10157] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The novel coronavirus disease of 2019 (COVID-19) is caused by the binding of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) to angiotensin-converting enzyme 2 (ACE2) receptors present on various locations such as the pulmonary alveolar epithelium and vascular endothelium. In COVID-19 patients, the interaction of SARS-CoV-2 with these receptors in the cerebral blood vessels has been attributed to stroke. Although the incidence of acute ischemic stroke is relatively low, ranging from 1% to 6%, the mortality associated with it is substantially high, reaching as high as 38%. This case series describes three distinct yet similar scenarios of COVID-19 positive patients with several underlying comorbidities, wherein two of the patients presented to our hospital with sudden onset right-sided weakness, later diagnosed with ischemic stroke, and one patient who developed an acute intracerebral hemorrhage during his hospital stay. The patients were diagnosed with acute stroke as a complication of COVID-19 infection. We also provide an insight into the possible mechanisms responsible for the life-threatening complication. Physicians should have a low threshold for suspecting stroke in COVID-19 patients, and close observation should be kept on such patients particularly those with clinical evidence of traditional risk factors.
Collapse
Affiliation(s)
- Kartikeya Rajdev
- Pulmonary and Critical Care Medicine, University of Nebraska Medical Center, Omaha, USA
| | - Shubham Lahan
- Internal Medicine, University College of Medical Sciences, Delhi, IND
| | - Kate Klein
- Internal Medicine, University of Nebraska Medical Center, Omaha, USA
| | - Craig A Piquette
- Pulmonary and Critical Care Medicine, University of Nebraska Medical Center, Omaha, USA
| | - Meilinh Thi
- Pulmonary and Critical Care Medicine, University of Nebraska Medical Center, Omaha, USA
| |
Collapse
|
1981
|
Aslam M, Ladilov Y. Targeting the sAC-Dependent cAMP Pool to Prevent SARS-Cov-2 Infection. Cells 2020; 9:cells9091962. [PMID: 32854430 PMCID: PMC7563949 DOI: 10.3390/cells9091962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
An outbreak of the novel coronavirus (CoV) SARS-CoV-2, the causative agent of COVID-19 respiratory disease, infected millions of people since the end of 2019, led to high-level morbidity and mortality and caused worldwide social and economic disruption. There are currently no antiviral drugs available with proven efficacy or vaccines for its prevention. An understanding of the underlying cellular mechanisms involved in virus replication is essential for repurposing the existing drugs and/or the discovery of new ones. Endocytosis is the important mechanism of entry of CoVs into host cells. Endosomal maturation followed by the fusion with lysosomes are crucial events in endocytosis. Late endosomes and lysosomes are characterized by their acidic pH, which is generated by a proton transporter V-ATPase and required for virus entry via endocytic pathway. The cytoplasmic cAMP pool produced by soluble adenylyl cyclase (sAC) promotes V-ATPase recruitment to endosomes/lysosomes and thus their acidification. In this review, we discuss targeting the sAC-specific cAMP pool as a potential strategy to impair the endocytic entry of the SARS-CoV-2 into the host cell. Furthermore, we consider the potential impact of sAC inhibition on CoV-induced disease via modulation of autophagy and apoptosis.
Collapse
Affiliation(s)
- Muhammad Aslam
- Experimental Cardiology, Department of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany;
- DZHK (German Centre for Cardiovascular Research), Department of Cardiology, Kerckhoff Clinic GmbH partner site Rhein-Main, 61231 Bad Nauheim, Germany
| | - Yury Ladilov
- Independent Researcher, 42929 Wermelskirchen, Germany
- Correspondence:
| |
Collapse
|
1982
|
Hussain N, Chung E, Heyl JJ, Hussain B, Oh MC, Pinon C, Boral S, Chun D, Babu B. A Meta-Analysis on the Effects of Hydroxychloroquine on COVID-19. Cureus 2020; 12:e10005. [PMID: 32983702 PMCID: PMC7515154 DOI: 10.7759/cureus.10005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/24/2020] [Indexed: 11/27/2022] Open
Abstract
Introduction Since December 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread throughout the world with a large medical and economic impact. On March 12, 2020, the World Health Organization (WHO) classified SARS-CoV-2 as a pandemic. As a result of this worldwide public health crisis, politicians, elected officials, and healthcare professionals emergently began trialing hydroxychloroquine (HCQ) in efforts to treat and prevent the transmission of the virus. This meta-analysis was performed to assess the effects of HCQ on patients with COVID-19. Methods This meta-analysis adheres to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRIMA) guidelines. Selected articles published between December 2019 and July 2020 were found utilizing the following search engines: PubMed, Google Scholar, Cochrane Library, DisasterLit, Clinicaltrials.gov, Medrxiv, and Embase. Two independent physician reviewers screened eligible articles that met the inclusion and exclusion criteria of the analysis. The outcome measures analyzed were mortality rate, rate of disease progression/improvement, rate of disease severity, and adverse effects of treatment. Six out of 14 studies that met the study's eligibility criteria were selected and further analyzed, with a total of 381 participants (n= 381). Conclusion From the studies analyzed, it was found that groups treated with HCQ had an overall mortality rate that was 2.5 times greater than that of the control group. HCQ treated patients had higher rates of adverse clinical outcomes and side effects compared with the control populations. Lastly, there was a 1.2 times higher rate of improvement in the group of HCQ treated patients with mild to moderate symptoms as compared to the control group.
Collapse
Affiliation(s)
| | - Emily Chung
- Medicine, St. John's Episcopal Hospital, New York, USA
| | | | - Bisma Hussain
- Medicine, St. John's Episcopal Hospital, New York, USA
| | - Michael C Oh
- Medicine, St. John's Episcopal Hospital, New York, USA
| | | | | | - David Chun
- Medicine, Northwell Health, New York, USA
| | - Benson Babu
- Hospital Medicine, Northwell Health, New York, USA
| |
Collapse
|
1983
|
Dandel M. Cardiac manifestations of COVID-19 infection: the role of echocardiography in patient management. Infection 2020; 49:187-189. [PMID: 32833108 PMCID: PMC7443532 DOI: 10.1007/s15010-020-01507-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Michael Dandel
- German Centre for Heart and Circulatory Research (DZHK) Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
1984
|
Faqihi F, Alharthy A, Memish ZA, Karakitsos D. Comment on Hu et al: The cytokine storm and COVID-19. J Med Virol 2020; 93:631-633. [PMID: 32761905 PMCID: PMC7436490 DOI: 10.1002/jmv.26396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 01/08/2023]
Affiliation(s)
| | | | - Ziad A Memish
- Department of Critical Care, King Saud Medical City, Riyadh, Saudi Arabia.,Research and Innovation Center, King Saud Medical City, Riyadh, Saudi Arabia
| | | |
Collapse
|
1985
|
Mohammad S, Bouchama A, Mohammad Alharbi B, Rashid M, Saleem Khatlani T, Gaber NS, Malik SS. SARS-CoV-2 ORF8 and SARS-CoV ORF8ab: Genomic Divergence and Functional Convergence. Pathogens 2020; 9:E677. [PMID: 32825438 PMCID: PMC7558349 DOI: 10.3390/pathogens9090677] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 01/18/2023] Open
Abstract
The COVID-19 pandemic, in the first seven months, has led to more than 15 million confirmed infected cases and 600,000 deaths. SARS-CoV-2, the causative agent for COVID-19, has proved to be a great challenge for its ability to spread in asymptomatic stages and the diverse disease spectrum it has generated. This has created a challenge of unimaginable magnitude, not only affecting human health and life but also potentially generating a long-lasting socioeconomic impact. Both medical sciences and biomedical research have also been challenged, consequently leading to a large number of clinical trials and vaccine initiatives. While known proteins of pathobiological importance are targets for these therapeutic approaches, it is imperative to explore other factors of viral significance. Accessory proteins are one such trait that have diverse roles in coronavirus pathobiology. Here, we analyze certain genomic characteristics of SARS-CoV-2 accessory protein ORF8 and predict its protein features. We have further reviewed current available literature regarding its function and comparatively evaluated these and other features of ORF8 and ORF8ab, its homolog from SARS-CoV. Because coronaviruses have been infecting humans repeatedly and might continue to do so, we therefore expect this study to aid in the development of holistic understanding of these proteins. Despite low nucleotide and protein identity and differentiating genome level characteristics, there appears to be significant structural integrity and functional proximity between these proteins pointing towards their high significance. There is further need for comprehensive genomics and structural-functional studies to lead towards definitive conclusions regarding their criticality and that can eventually define their relevance to therapeutics development.
Collapse
Affiliation(s)
- Sameer Mohammad
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, MNGHA, Riyadh 11426, Saudi Arabia; (S.M.); (A.B.); (B.M.A.); (N.S.G.)
| | - Abderrezak Bouchama
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, MNGHA, Riyadh 11426, Saudi Arabia; (S.M.); (A.B.); (B.M.A.); (N.S.G.)
| | - Bothina Mohammad Alharbi
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, MNGHA, Riyadh 11426, Saudi Arabia; (S.M.); (A.B.); (B.M.A.); (N.S.G.)
| | - Mamoon Rashid
- Bioinformatics and Biostatistics Department, King Abdullah International Medical Research Center, King~Saud bin Abdulaziz University for Health Sciences, MNGHA, Riyadh 11426, Saudi Arabia;
| | - Tanveer Saleem Khatlani
- Stem Cells Unit, Department of Cellular Therapy, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, MNGHA, Riyadh 11426, Saudi Arabia;
| | - Nusaibah S. Gaber
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, MNGHA, Riyadh 11426, Saudi Arabia; (S.M.); (A.B.); (B.M.A.); (N.S.G.)
| | - Shuja Shafi Malik
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, MNGHA, Riyadh 11426, Saudi Arabia; (S.M.); (A.B.); (B.M.A.); (N.S.G.)
| |
Collapse
|
1986
|
|
1987
|
Kopp K, Lichtenauer M, Motloch LJ, Hoppe UC, Egle A, Salzer HJF, Lamprecht B, Tomasits J, Müller HM, Dieplinger A. Interdisciplinary Model for Scheduling Post-discharge Cardiopulmonary Care of Patients Following Severe and Critical SARS-CoV-2 (Coronavirus) Infection. Front Cardiovasc Med 2020; 7:157. [PMID: 32923461 PMCID: PMC7457001 DOI: 10.3389/fcvm.2020.00157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/27/2020] [Indexed: 11/16/2022] Open
Affiliation(s)
- Kristen Kopp
- Department of Internal Medicine II, Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Lukas Jaroslaw Motloch
- Department of Internal Medicine II, Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Uta C. Hoppe
- Department of Internal Medicine II, Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Alexander Egle
- Department of Internal Medicine III, Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Paracelsus Medical University, Salzburg, Austria
| | - Helmut J. F. Salzer
- Department of Pulmonology, Kepler University Hospital, Linz, Austria
- Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Bernd Lamprecht
- Department of Pulmonology, Kepler University Hospital, Linz, Austria
- Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Josef Tomasits
- Institute for Medical and Chemical Laboratory Diagnostics, Kepler University Hospital, Linz, Austria
| | - Hannes M. Müller
- Department of Cardiac, Vascular, and Thoracic Surgery, Kepler University Hospital, Linz, Austria
| | - Anna Dieplinger
- Institute for Nursing and Practice, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
1988
|
Bellavite P, Donzelli A. Hesperidin and SARS-CoV-2: New Light on the Healthy Function of Citrus Fruits. Antioxidants (Basel) 2020; 9:E742. [PMID: 32823497 PMCID: PMC7465267 DOI: 10.3390/antiox9080742] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Among the many approaches to Coronavirus disease 2019 (COVID-19) prevention, the possible role of nutrition has so far been rather underestimated. Foods are very rich in substances, with a potential beneficial effect on health, and some of these could have an antiviral action or be important in modulating the immune system and in defending cells from the oxidative stress associated with infection. This short review draws the attention on some components of citrus fruits, and especially of the orange (Citrus sinensis), well known for its vitamin and flavonoid content. Among the flavonoids, hesperidin has recently attracted the attention of researchers, because it binds to the key proteins of the Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Several computational methods, independently applied by different researchers, showed that hesperidin has a low binding energy, both with the coronavirus "spike" protein, and with the main protease that transforms the early proteins of the virus (pp1a and ppa1b) into the complex responsible for viral replication. The binding energy of hesperidin to these important components is lower than that of lopinavir, ritonavir, and indinavir, suggesting that it could perform an effective antiviral action. Furthermore, both hesperidin and ascorbic acid counteract the cell damaging effects of the oxygen free radicals triggered by virus infection and inflammation. There is discussion about the preventive efficacy of vitamin C, at the dose achievable by the diet, but recent reviews suggest that this substance can be useful in the case of strong immune system burden caused by viral disease. Computational methods and laboratory studies support the need to undertake apposite preclinical, epidemiological, and experimental studies on the potential benefits of citrus fruit components for the prevention of infectious diseases, including COVID-19.
Collapse
Affiliation(s)
- Paolo Bellavite
- Department of Medicine, Section of General Pathology, University of Verona Medical School, Strada Le Grazie 8, 37134 Verona, Italy
| | - Alberto Donzelli
- Medical Doctor, Scientific Committee of Fondazione Allineare Sanità e Salute, 20122 Milano, Italy;
| |
Collapse
|
1989
|
Gupta A, Madhavan MV, Poterucha TJ, DeFilippis EM, Hennessey JA, Redfors B, Eckhardt C, Bikdeli B, Platt J, Nalbandian A, Elias P, Cummings MJ, Nouri SN, Lawlor M, Ranard LS, Li J, Boyle C, Givens R, Brodie D, Krumholz HM, Stone GW, Sethi SS, Burkhoff D, Uriel N, Schwartz A, Leon MB, Kirtane AJ, Wan EY, Parikh SA. Association Between Antecedent Statin Use and Decreased Mortality in Hospitalized Patients with COVID-19. RESEARCH SQUARE 2020. [PMID: 32818209 PMCID: PMC7430584 DOI: 10.21203/rs.3.rs-56210/v1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can result in a hyperinflammatory state, leading to acute respiratory distress syndrome (ARDS), myocardial injury, and thrombotic complications, among other sequelae. Statins, which are known to have anti-inflammatory and antithrombotic properties, have been studied in the setting of other viral infections and ARDS, but their benefit has not been assessed in COVID-19. Thus, we sought to determine whether antecedent statin use is associated with lower in-hospital mortality in patients hospitalized for COVID-19. This is a retrospective analysis of patients admitted with COVID-19 from February 1st through May 12th, 2020 with study period ending on June 11th, 2020. Antecedent statin use was assessed using medication information available in the electronic medical record. We constructed a multivariable logistic regression model to predict the propensity of receiving statins, adjusting for baseline socio-demographic and clinical characteristics, and outpatient medications. The primary endpoint included in-hospital mortality within 30 days. A total of 2626 patients were admitted during the study period, of whom 951 (36.2%) were antecedent statin users. Among 1296 patients (648 statin users, 648 non-statin users) identified with 1:1 propensity-score matching, demographic, baseline, and outpatient medication information were well balanced. Statin use was significantly associated with lower odds of the primary endpoint in the propensity-matched cohort (OR 0.48, 95% CI 0.36 – 0.64, p<0.001). We conclude that antecedent statin use in patients hospitalized with COVID-19 was associated with lower inpatient mortality. Randomized clinical trials evaluating the utility of statin therapy in patients with COVID-19 are needed.
Collapse
Affiliation(s)
- Aakriti Gupta
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center; Cardiovascular Research Foundation; Yale Center for Outcomes Research and Evaluation
| | - Mahesh V Madhavan
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center; Cardiovascular Research Foundation
| | - Timothy J Poterucha
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | | | - Jessica A Hennessey
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Bjorn Redfors
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center; Cardiovascular Research Foundation; Sahlgrenska University Hospital
| | - Christina Eckhardt
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Behnood Bikdeli
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center; Cardiovascular Research Foundation; Yale Center for Outcomes Research and Evaluation
| | - Jonathan Platt
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Ani Nalbandian
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Pierre Elias
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Matthew J Cummings
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Shayan N Nouri
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Matthew Lawlor
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Lauren S Ranard
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Jianhua Li
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Claudia Boyle
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Raymond Givens
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Daniel Brodie
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | | | | | - Sanjum S Sethi
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Daniel Burkhoff
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center; Cardiovascular Research Foundation
| | - Nir Uriel
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Allan Schwartz
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Martin B Leon
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center; Cardiovascular Research Foundation
| | - Ajay J Kirtane
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center; Cardiovascular Research Foundation
| | - Elaine Y Wan
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center
| | - Sahil A Parikh
- NewYork-Presbyterian Hospital and the Columbia University Irving Medical Center; Cardiovascular Research Foundation
| |
Collapse
|
1990
|
Luban J, Sattler R, Mühlberger E, Graci JD, Cao L, Weetall M, Trotta C, Colacino JM, Bavari S, Strambio-De-Castillia C, Suder EL, Wang Y, Soloveva V, Cintron-Lue K, Naryshkin NA, Pykett M, Welch EM, O'Keefe K, Kong R, Goodwin E, Jacobson A, Paessler S, Peltz S. The DHODH Inhibitor PTC299 Arrests SARS-CoV-2 Replication and Suppresses Induction of Inflammatory Cytokines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32793904 DOI: 10.1101/2020.08.05.238394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has created an urgent need for therapeutics that inhibit the SARS-CoV-2 virus and suppress the fulminant inflammation characteristic of advanced illness. Here, we describe the anti-COVID-19 potential of PTC299, an orally available compound that is a potent inhibitor of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme of the de novo pyrimidine biosynthesis pathway. In tissue culture, PTC299 manifests robust, dose-dependent, and DHODH-dependent inhibition of SARS CoV-2 replication (EC 50 range, 2.0 to 31.6 nM) with a selectivity index >3,800. PTC299 also blocked replication of other RNA viruses, including Ebola virus. Consistent with known DHODH requirements for immunomodulatory cytokine production, PTC299 inhibited the production of interleukin (IL)-6, IL-17A (also called IL-17), IL-17F, and vascular endothelial growth factor (VEGF) in tissue culture models. The combination of anti-SARS-CoV-2 activity, cytokine inhibitory activity, and previously established favorable pharmacokinetic and human safety profiles render PTC299 a promising therapeutic for COVID-19.
Collapse
|
1991
|
Kaur S, Tripathi DM, Yadav A. The Enigma of Endothelium in COVID-19. Front Physiol 2020; 11:989. [PMID: 32848893 PMCID: PMC7417426 DOI: 10.3389/fphys.2020.00989] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/20/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has affected millions of people globally. Clinically, it presents with mild flu-like symptoms in most cases but can cause respiratory failure in high risk population. With the aim of unearthing newer treatments, scientists all over the globe are striving hard to comprehend the underlying mechanisms of COVID-19. Several studies till date have indicated a dysregulated host immune response as the major cause of COVID-19 induced mortality. In this Perspective, we propose a key role of endothelium, particularly pulmonary endothelium in the pathogenesis of COVID-19. We draw parallels and divergences between COVID-19-induced respiratory distress and bacterial sepsis-induced lung injury and recommend the road ahead with respect to identification of endothelium-based biomarkers and plausible treatments for COVID-19.
Collapse
Affiliation(s)
- Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Dinesh M. Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Angeera Yadav
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Guwahati, India
| |
Collapse
|
1992
|
Cheng AP, Cheng MP, Gu W, Lenz JS, Hsu E, Schurr E, Bourque G, Bourgey M, Ritz J, Marty F, Chiu CY, Vinh DC, Vlaminck ID. Cell-Free DNA in Blood Reveals Significant Cell, Tissue and Organ Specific injury and Predicts COVID-19 Severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.07.27.20163188. [PMID: 32766608 PMCID: PMC7402071 DOI: 10.1101/2020.07.27.20163188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
COVID-19 primarily affects the lungs, but evidence of systemic disease with multi-organ involvement is emerging. Here, we developed a blood test to broadly quantify cell, tissue, and organ specific injury due to COVID-19, using genome-wide methylation profiling of circulating cell-free DNA in plasma. We assessed the utility of this test to identify subjects with severe disease in two independent, longitudinal cohorts of hospitalized patients. Cell-free DNA profiling was performed on 104 plasma samples from 33 COVID-19 patients and compared to samples from patients with other viral infections and healthy controls. We found evidence of injury to the lung and liver and involvement of red blood cell progenitors associated with severe COVID-19. The concentration of cfDNA correlated with the WHO ordinal scale for disease progression and was significantly increased in patients requiring intubation. This study points to the utility of cell-free DNA as an analyte to monitor and study COVID-19.
Collapse
Affiliation(s)
| | | | - Wei Gu
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbot Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Joan Sesing Lenz
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Elaine Hsu
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Erwin Schurr
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Mathieu Bourgey
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical school, Boston, MA, USA
| | - Francisco Marty
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Infectious Disease, Brigham and Women’s Hospital, Boston, MA, USA
| | - Charles Y. Chiu
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbot Viral Diagnostics and Discovery Center, San Francisco, CA, USA
- Department of Medicine, Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | | | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
1993
|
Kim J, Mukherjee A, Nelson D, Jozic A, Sahay G. Rapid generation of circulating and mucosal decoy ACE2 using mRNA nanotherapeutics for the potential treatment of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32743574 DOI: 10.1101/2020.07.24.205583] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enters through the airways and infects the lungs, causing lethal pulmonary damage in vulnerable patients. This virus contains spike proteins on its envelope that binds to human angiotensin-converting enzyme 2 (hACE2) expressed on the surface of airway cells, enabling entry of the virus for causing infection 1,2 . In severe cases, the virus enters the circulatory system, contributing to multiorgan failure. Soluble form of hACE2 binds to SARS-CoV-2 spike protein and prevents viral entry into target cells 3 . Moreover, soluble recombinant ACE2 ameliorates lung injury 4 but its short half-life limits its therapeutic utility 5 . Here, we engineered synthetic mRNA to encode a soluble form of hACE2 (hsACE2) to prevent viral infection. Novel lipid nanoparticles (LNPs) were used to package mRNA and transfect mammalian cells for enhanced production of secreted proteins. Intravenously administered LNP led to hepatic delivery of the mRNA. This elicited secretion of hsACE2 into the blood circulation within 2 h, and levels of circulating hsACE2 peaked at 6 h and gradually decreased over several days. Since the primary site of entry and pathogenesis for SARS-CoV-2 is the lungs, we instilled LNPs into the lungs and were able to detect hsACE2 in the bronchoalveolar lavage fluid within 24 h and lasted for 48 h. Through co-immunoprecipitation, we found that mRNA-generated hsACE2 was able to bind with the receptor binding domain of the SARS-CoV-2 spike protein. Furthermore, hsACE2 was able to strongly inhibit (over 90%) SARS-CoV-2 pseudovirus infection. Our proof of principle study shows that mRNA-based nanotherapeutics can be potentially deployed for pulmonary and extrapulmonary neutralization of SARS-CoV-2 and open new treatment opportunities for COVID-19.
Collapse
|
1994
|
Micallef J, Soeiro T, Jonville-Béra AP. COVID-19 and NSAIDs: Primum non nocere. Therapie 2020; 75:514-515. [PMID: 32839015 PMCID: PMC7375282 DOI: 10.1016/j.therap.2020.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Joëlle Micallef
- Aix-Marseille université, INSERM, UMR 1106, Assistance publique-Hôpitaux de Marseille, service de pharmacologie clinique, centre régional de pharmacovigilance, 264, rue Saint-Pierre, 13005 Marseille, France.
| | - Thomas Soeiro
- Aix-Marseille université, INSERM, UMR 1106, Assistance publique-Hôpitaux de Marseille, service de pharmacologie clinique, centre régional de pharmacovigilance, 264, rue Saint-Pierre, 13005 Marseille, France
| | - Annie-Pierre Jonville-Béra
- Centre hospitalier régional universitaire de Tours, service de pharmacosurveillance, centre régional de pharmacovigilance, 37000 Tours, France
| |
Collapse
|
1995
|
[Pathology and Immunology of COVID-19]. Uirusu 2020; 70:167-174. [PMID: 34544931 DOI: 10.2222/jsv.70.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Since the first case of COVID-19 was reported from Wuhan, China in December 2019, SARS-CoV-2 has been spreading globally and has become major public health concern. At present, development of specific treatment for COVID-19 is in progress and several countermeasures have been subjected to clinical trials. However, efficacy of these countermeasures is limited. For development of effective medicines or vaccines against infectious diseases, it is mandatory to elucidate its etiology and pathogenesis by means of pathological analysis. Pathological studies revealed that the COVID-19 mainly affects respiratory tracts although other organs are also involved. In addition, immunological studies demonstrated that host immune response may exacerbates COVID-19 through systemic inflammation. In this review, we would like to overview pathology and immunology of COVID-19.
Collapse
|