51
|
Haynes WA, Kamath K, Bozekowski J, Baum-Jones E, Campbell M, Casanovas-Massana A, Daugherty PS, Dela Cruz CS, Dhal A, Farhadian SF, Fitzgibbons L, Fournier J, Jhatro M, Jordan G, Klein J, Lucas C, Kessler D, Luchsinger LL, Martinez B, Catherine Muenker M, Pischel L, Reifert J, Sawyer JR, Waitz R, Wunder EA, Zhang M, Iwasaki A, Ko A, Shon JC. High-resolution epitope mapping and characterization of SARS-CoV-2 antibodies in large cohorts of subjects with COVID-19. Commun Biol 2021; 4:1317. [PMID: 34811480 PMCID: PMC8608966 DOI: 10.1038/s42003-021-02835-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
As Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to spread, characterization of its antibody epitopes, emerging strains, related coronaviruses, and even the human proteome in naturally infected patients can guide the development of effective vaccines and therapies. Since traditional epitope identification tools are dependent upon pre-defined peptide sequences, they are not readily adaptable to diverse viral proteomes. The Serum Epitope Repertoire Analysis (SERA) platform leverages a high diversity random bacterial display library to identify proteome-independent epitope binding specificities which are then analyzed in the context of organisms of interest. When evaluating immune response in the context of SARS-CoV-2, we identify dominant epitope regions and motifs which demonstrate potential to classify mild from severe disease and relate to neutralization activity. We highlight SARS-CoV-2 epitopes that are cross-reactive with other coronaviruses and demonstrate decreased epitope signal for mutant SARS-CoV-2 strains. Collectively, the evolution of SARS-CoV-2 mutants towards reduced antibody response highlight the importance of data-driven development of the vaccines and therapies to treat COVID-19.
Collapse
Affiliation(s)
| | | | | | | | - Melissa Campbell
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Arnau Casanovas-Massana
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | | | - Charles S Dela Cruz
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Shelli F Farhadian
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | | | - John Fournier
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | | | | | - Jon Klein
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | - M Catherine Muenker
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Lauren Pischel
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | - Elsio A Wunder
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | | | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Albert Ko
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | | |
Collapse
|
52
|
Kumar A, Narayan RK, Prasoon P, Kumari C, Kaur G, Kumar S, Kulandhasamy M, Sesham K, Pareek V, Faiq MA, Pandey SN, Singh HN, Kant K, Shekhawat PS, Raza K, Kumar S. COVID-19 Mechanisms in the Human Body-What We Know So Far. Front Immunol 2021; 12:693938. [PMID: 34790191 PMCID: PMC8592035 DOI: 10.3389/fimmu.2021.693938] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023] Open
Abstract
More than one and a half years have elapsed since the commencement of the coronavirus disease 2019 (COVID-19) pandemic, and the world is struggling to contain it. Being caused by a previously unknown virus, in the initial period, there had been an extreme paucity of knowledge about the disease mechanisms, which hampered preventive and therapeutic measures against COVID-19. In an endeavor to understand the pathogenic mechanisms, extensive experimental studies have been conducted across the globe involving cell culture-based experiments, human tissue organoids, and animal models, targeted to various aspects of the disease, viz., viral properties, tissue tropism and organ-specific pathogenesis, involvement of physiological systems, and the human immune response against the infection. The vastly accumulated scientific knowledge on all aspects of COVID-19 has currently changed the scenario from great despair to hope. Even though spectacular progress has been made in all of these aspects, multiple knowledge gaps are remaining that need to be addressed in future studies. Moreover, multiple severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have emerged across the globe since the onset of the first COVID-19 wave, with seemingly greater transmissibility/virulence and immune escape capabilities than the wild-type strain. In this review, we narrate the progress made since the commencement of the pandemic regarding the knowledge on COVID-19 mechanisms in the human body, including virus-host interactions, pulmonary and other systemic manifestations, immunological dysregulations, complications, host-specific vulnerability, and long-term health consequences in the survivors. Additionally, we provide a brief review of the current evidence explaining molecular mechanisms imparting greater transmissibility and virulence and immune escape capabilities to the emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Ravi K. Narayan
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anatomy, Andaman and Nicobar Islands Institute of Medical Sciences, Port Blair, India
| | - Pranav Prasoon
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Pittsburgh Center for Pain Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Chiman Kumari
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anatomy, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Gurjot Kaur
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- School of Pharmaceutical Sciences, Shoolini University, Solan, India
| | - Santosh Kumar
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Maheswari Kulandhasamy
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Biochemistry, Maulana Azad Medical College (MAMC), New Delhi, India
| | - Kishore Sesham
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Vijayawada, India
| | - Vikas Pareek
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Center for Cognitive and Brain Sciences, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, India
| | - Muneeb A. Faiq
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- New York University (NYU) Langone Health Center, NYU Robert I. Grossman School of Medicine, New York, NY, United States
| | - Sada N. Pandey
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Zoology, Banaras Hindu University (BHU), Varanasi, India
| | - Himanshu N. Singh
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - Kamla Kant
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Bathinda, India
| | - Prakash S. Shekhawat
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Clinical Hematology, National Institute of Medical Sciences, Jaipur, India
| | - Khursheed Raza
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Deoghar, India
| | - Sujeet Kumar
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Center for Proteomics and Drug Discovery, Amity Institute of Biotechnology, Amity University, Maharashtra, India
| |
Collapse
|
53
|
Huijghebaert S, Hoste L, Vanham G. Essentials in saline pharmacology for nasal or respiratory hygiene in times of COVID-19. Eur J Clin Pharmacol 2021; 77:1275-1293. [PMID: 33772626 PMCID: PMC7998085 DOI: 10.1007/s00228-021-03102-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/28/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Nasal irrigation or nebulizing aerosol of isotonic or hypertonic saline is a traditional method for respiratory or nasal care. A recent small study in outpatients with COVID-19 without acute respiratory distress syndrome suggests substantial symptom resolution. We therefore analyzed pharmacological/pharmacodynamic effects of isotonic or hypertonic saline, relevant to SARS-CoV-2 infection and respiratory care. METHODS Mixed search method. RESULTS Due to its wetting properties, saline achieves an improved spreading of alveolar lining fluid and has been shown to reduce bio-aerosols and viral load. Saline provides moisture to respiratory epithelia and gels mucus, promotes ciliary beating, and improves mucociliary clearance. Coronaviruses and SARS-CoV-2 damage ciliated epithelium in the nose and airways. Saline inhibits SARS-CoV-2 replication in Vero cells; possible interactions involve the viral ACE2-entry mechanism (chloride-dependent ACE2 configuration), furin and 3CLpro (inhibition by NaCl), and the sodium channel ENaC. Saline shifts myeloperoxidase activity in epithelial or phagocytic cells to produce hypochlorous acid. Clinically, nasal or respiratory airway care with saline reduces symptoms of seasonal coronaviruses and other common cold viruses. Its use as aerosol reduces hospitalization rates for bronchiolitis in children. Preliminary data suggest symptom reduction in symptomatic COVID-19 patients if saline is initiated within 48 h of symptom onset. CONCLUSIONS Saline interacts at various levels relevant to nasal or respiratory hygiene (nasal irrigation, gargling or aerosol). If used from the onset of common cold symptoms, it may represent a useful add-on to first-line interventions for COVID-19. Formal evaluation in mild COVID-19 is desirable as to establish efficacy and optimal treatment regimens.
Collapse
Affiliation(s)
| | - Levi Hoste
- Pediatric Pulmonology, Infectious Diseases and Immunology, Ghent University Hospital, Ghent, Belgium
- Primary Immunodeficiency Research Lab, Center for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Center, Ghent University Hospital, Ghent, Belgium
| | - Guido Vanham
- Department of Biomedical Sciences, Institute of Tropical Medicine and University of Antwerp, Antwerp, Belgium
| |
Collapse
|
54
|
Anand H, Ende V, Singh G, Qureshi I, Duong TQ, Mehler MF. Nervous System-Systemic Crosstalk in SARS-CoV-2/COVID-19: A Unique Dyshomeostasis Syndrome. Front Neurosci 2021; 15:727060. [PMID: 34512253 PMCID: PMC8430330 DOI: 10.3389/fnins.2021.727060] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/30/2021] [Indexed: 01/05/2023] Open
Abstract
SARS-CoV-2 infection is associated with a spectrum of acute neurological syndromes. A subset of these syndromes promotes higher in-hospital mortality than is predicted by traditional parameters defining critical care illness. This suggests that deregulation of components of the central and peripheral nervous systems compromises the interplay with systemic cellular, tissue and organ interfaces to mediate numerous atypical manifestations of COVID-19 through impairments in organismal homeostasis. This unique dyshomeostasis syndrome involves components of the ACE-2/1 lifecycles, renin-angiotensin system regulatory axes, integrated nervous system functional interactions and brain regions differentially sculpted by accelerated evolutionary processes and more primordial homeostatic functions. These biological contingencies suggest a mechanistic blueprint to define long-term neurological sequelae and systemic manifestations such as premature aging phenotypes, including organ fibrosis, tissue degeneration and cancer. Therapeutic initiatives must therefore encompass innovative combinatorial agents, including repurposing FDA-approved drugs targeting components of the autonomic nervous system and recently identified products of SARS-CoV-2-host interactions.
Collapse
Affiliation(s)
- Harnadar Anand
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Victoria Ende
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Gurinder Singh
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Irfan Qureshi
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, United States
- Biohaven Pharmaceuticals, New Haven, CT, United States
| | - Tim Q. Duong
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Mark F. Mehler
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, United States
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY, United States
- Rose F. Kennedy Center for Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY, United States
- Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, United States
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
- Center for Epigenomics, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
55
|
Durdagi S, Dağ Ç, Dogan B, Yigin M, Avsar T, Buyukdag C, Erol I, Ertem FB, Calis S, Yildirim G, Orhan MD, Guven O, Aksoydan B, Destan E, Sahin K, Besler SO, Oktay L, Shafiei A, Tolu I, Ayan E, Yuksel B, Peksen AB, Gocenler O, Yucel AD, Can O, Ozabrahamyan S, Olkan A, Erdemoglu E, Aksit F, Tanisali G, Yefanov OM, Barty A, Tolstikova A, Ketawala GK, Botha S, Dao EH, Hayes B, Liang M, Seaberg MH, Hunter MS, Batyuk A, Mariani V, Su Z, Poitevin F, Yoon CH, Kupitz C, Sierra RG, Snell EH, DeMirci H. Near-physiological-temperature serial crystallography reveals conformations of SARS-CoV-2 main protease active site for improved drug repurposing. Structure 2021; 29:1382-1396.e6. [PMID: 34403647 PMCID: PMC8367086 DOI: 10.1016/j.str.2021.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/19/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023]
Abstract
The COVID-19 pandemic has resulted in 198 million reported infections and more than 4 million deaths as of July 2021 (covid19.who.int). Research to identify effective therapies for COVID-19 includes: (1) designing a vaccine as future protection; (2) de novo drug discovery; and (3) identifying existing drugs to repurpose them as effective and immediate treatments. To assist in drug repurposing and design, we determine two apo structures of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease at ambient temperature by serial femtosecond X-ray crystallography. We employ detailed molecular simulations of selected known main protease inhibitors with the structures and compare binding modes and energies. The combined structural and molecular modeling studies not only reveal the dynamics of small molecules targeting the main protease but also provide invaluable opportunities for drug repurposing and structure-based drug design strategies against SARS-CoV-2.
Collapse
Affiliation(s)
- Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey.
| | - Çağdaş Dağ
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Berna Dogan
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey
| | - Merve Yigin
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Timucin Avsar
- Department of Medical Biology, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey
| | - Cengizhan Buyukdag
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Ismail Erol
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey; Department of Chemistry, Gebze Technical University, Kocaeli 41400, Turkey
| | - Fatma Betul Ertem
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Seyma Calis
- Department of Medical Biology, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey; Department of Molecular Biology - Genetics and Biotechnology, Istanbul Technical University, Istanbul 34469, Turkey
| | - Gunseli Yildirim
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Muge D Orhan
- Department of Medical Biology, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey
| | - Omur Guven
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Busecan Aksoydan
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey; Neuroscience Program, Graduate School of Health Sciences, Bahcesehir University, Istanbul 34734, Turkey
| | - Ebru Destan
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Kader Sahin
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey
| | - Sabri O Besler
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Lalehan Oktay
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey
| | - Alaleh Shafiei
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Ilayda Tolu
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey
| | - Esra Ayan
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Busra Yuksel
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Ayse B Peksen
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Oktay Gocenler
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Ali D Yucel
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Ozgur Can
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Serena Ozabrahamyan
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Alpsu Olkan
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey; School of Medicine, Bahcesehir University, Istanbul 34734, Turkey
| | - Ece Erdemoglu
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34734, Turkey; Faculty of Medicine, Mersin University, Mersin 33070, Turkey
| | - Fulya Aksit
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | - Gokhan Tanisali
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey
| | | | - Anton Barty
- Deutsches Elektronen-Synchrotron, Notkestrasse 85, Hamburg 22607, Germany
| | | | - Gihan K Ketawala
- Department of Physics, Arizona State University, Tempe, AZ 85287-1504, USA; Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85287-5001, USA
| | - Sabine Botha
- Department of Physics, Arizona State University, Tempe, AZ 85287-1504, USA; Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85287-5001, USA
| | - E Han Dao
- Stanford PULSE Institute, SLAC National Laboratory, Menlo Park, CA 94025, USA
| | - Brandon Hayes
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Mengning Liang
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Matthew H Seaberg
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Mark S Hunter
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Alex Batyuk
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Valerio Mariani
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Zhen Su
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA; Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Frederic Poitevin
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Chun Hong Yoon
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Christopher Kupitz
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Raymond G Sierra
- Linac Coherent Light Source, SLAC National Accelerator Laboratory, 2575 Sand Hill Road, Menlo Park, CA 94025, USA
| | - Edward H Snell
- Hauptman-Woodward Medical Research Institute, University at Buffalo, 700 Ellicott St, Buffalo, NY, USA; Materials Design and Innovation, SUNY at Buffalo, 700 Ellicott St., Buffalo, NY, USA
| | - Hasan DeMirci
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkey; Stanford PULSE Institute, SLAC National Laboratory, Menlo Park, CA 94025, USA; Koc University Isbank Center for Infectious Diseases (KUISCID), 34450, Istanbul, Turkey.
| |
Collapse
|
56
|
Noori M, Nejadghaderi SA, Sullman MJM, Carson-Chahhoud K, Ardalan M, Kolahi AA, Safiri S. How SARS-CoV-2 might affect potassium balance via impairing epithelial sodium channels? Mol Biol Rep 2021; 48:6655-6661. [PMID: 34392451 PMCID: PMC8364628 DOI: 10.1007/s11033-021-06642-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/10/2021] [Indexed: 01/15/2023]
Abstract
Severe acute respiratory syndrome coronaviruses 2 (SARS-CoV-2) is the causative agent of current coronavirus disease 2019 (COVID-19) pandemic. Electrolyte disorders particularly potassium abnormalities have been repeatedly reported as common clinical manifestations of COVID-19. Here, we discuss how SARS-CoV-2 may affect potassium balance by impairing the activity of epithelial sodium channels (ENaC). The first hypothesis could justify the incidence of hypokalemia. SARS-CoV-2 cell entry through angiotensin-converting enzyme 2 (ACE2) may enhance the activity of renin–angiotensin–aldosterone system (RAAS) classical axis and further leading to over production of aldosterone. Aldosterone is capable of enhancing the activity of ENaC and resulting in potassium loss from epithelial cells. However, type II transmembrane serine protease (TMPRSS2) is able to inhibit the ENaC, but it is utilized in the case of SARS-CoV-2 cell entry, therefore the ENaC remains activated. The second hypothesis describe the incidence of hyperkalemia based on the key role of furin. Furin is necessary for cleaving both SARS-CoV-2 spike protein and ENaC subunits. While the furin is hijacked by the virus, the decreased activity of ENaC would be expected, which causes retention of potassium ions and hyperkalemia. Given that the occurrence of hypokalemia is higher than hyperkalemia in COVID-19 patients, the first hypothesis may have greater impact on potassium levels. Further investigations are warranted to determine the exact role of ENaC in SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Maryam Noori
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Aria Nejadghaderi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mark J M Sullman
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus.,Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - Kristin Carson-Chahhoud
- Australian Centre for Precision Health, University of South Australia, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| | | | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeid Safiri
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
57
|
John G, Sahajpal NS, Mondal AK, Ananth S, Williams C, Chaubey A, Rojiani AM, Kolhe R. Next-Generation Sequencing (NGS) in COVID-19: A Tool for SARS-CoV-2 Diagnosis, Monitoring New Strains and Phylodynamic Modeling in Molecular Epidemiology. Curr Issues Mol Biol 2021; 43:845-867. [PMID: 34449545 PMCID: PMC8929009 DOI: 10.3390/cimb43020061] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022] Open
Abstract
This review discusses the current testing methodologies for COVID-19 diagnosis and explores next-generation sequencing (NGS) technology for the detection of SARS-CoV-2 and monitoring phylogenetic evolution in the current COVID-19 pandemic. The review addresses the development, fundamentals, assay quality control and bioinformatics processing of the NGS data. This article provides a comprehensive review of the obstacles and opportunities facing the application of NGS technologies for the diagnosis, surveillance, and study of SARS-CoV-2 and other infectious diseases. Further, we have contemplated the opportunities and challenges inherent in the adoption of NGS technology as a diagnostic test with real-world examples of its utility in the fight against COVID-19.
Collapse
Affiliation(s)
- Goldin John
- Department of Pathology, Medical College of Georgia, Augusta University, BAE 2576, 1120 15th Street, Augusta, GA 30912, USA; (G.J.); (N.S.S.); (A.K.M.); (S.A.); (C.W.); (A.C.)
| | - Nikhil Shri Sahajpal
- Department of Pathology, Medical College of Georgia, Augusta University, BAE 2576, 1120 15th Street, Augusta, GA 30912, USA; (G.J.); (N.S.S.); (A.K.M.); (S.A.); (C.W.); (A.C.)
| | - Ashis K. Mondal
- Department of Pathology, Medical College of Georgia, Augusta University, BAE 2576, 1120 15th Street, Augusta, GA 30912, USA; (G.J.); (N.S.S.); (A.K.M.); (S.A.); (C.W.); (A.C.)
| | - Sudha Ananth
- Department of Pathology, Medical College of Georgia, Augusta University, BAE 2576, 1120 15th Street, Augusta, GA 30912, USA; (G.J.); (N.S.S.); (A.K.M.); (S.A.); (C.W.); (A.C.)
| | - Colin Williams
- Department of Pathology, Medical College of Georgia, Augusta University, BAE 2576, 1120 15th Street, Augusta, GA 30912, USA; (G.J.); (N.S.S.); (A.K.M.); (S.A.); (C.W.); (A.C.)
| | - Alka Chaubey
- Department of Pathology, Medical College of Georgia, Augusta University, BAE 2576, 1120 15th Street, Augusta, GA 30912, USA; (G.J.); (N.S.S.); (A.K.M.); (S.A.); (C.W.); (A.C.)
- Bionano Genomics Inc., San Diego, CA 92121, USA
| | - Amyn M. Rojiani
- Department of Pathology, Penn State University College of Medicine, Hershey, PA 16802, USA;
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, BAE 2576, 1120 15th Street, Augusta, GA 30912, USA; (G.J.); (N.S.S.); (A.K.M.); (S.A.); (C.W.); (A.C.)
| |
Collapse
|
58
|
Grant SN, Lester HA. Regulation of epithelial sodium channel activity by SARS-CoV-1 and SARS-CoV-2 proteins. Biophys J 2021; 120:2805-2813. [PMID: 34197807 PMCID: PMC8238646 DOI: 10.1016/j.bpj.2021.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/21/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) coronavirus (CoV) 2 (SARS-CoV-2), which causes the coronavirus disease 2019, encodes several proteins whose roles are poorly understood. We tested their ability either to directly form plasma membrane ion channels or to change functions of two mammalian plasma membrane ion channels, the epithelial sodium channel (ENaC) and the α3β4 nicotinic acetylcholine receptor. In mRNA-injected Xenopus oocytes, none of nine SARS-CoV-2 proteins or two SARS-CoV-1 proteins produced conductances, nor did co-injection of several combinations. Immunoblots for ORF8, spike (S), and envelope (E) proteins revealed that the proteins are expressed at appropriate molecular weights. In experiments on coexpression with ENaC, three tested SARS proteins (SARS-CoV-1 E, SARS-CoV-2 E, and SARS-CoV-2 S) markedly decrease ENaC currents. SARS-CoV-1 S protein decreases ENaC currents modestly. Coexpressing the E proteins but not the S proteins with α3β4 nicotinic acetylcholine receptors significantly reduces acetylcholine-induced currents. ENaC inhibition does not occur if the SARS-CoV protein mRNAs are injected 24 h after the ENaC mRNAs, suggesting that SARS-CoV proteins affect early step(s) in functional expression of channel proteins. Consistent with the hypothesis that the SARS-CoV-2 S protein-induced ENaC inhibition involves competition for available protease, mutating the furin cleavage site in SARS-CoV-2 S protein partially relieves inhibition of ENaC currents. Extending previous suggestions that SARS proteins affect ENaC currents via protein kinase C (PKC) activation, PKC activation via phorbol 12-myristate 13-acetate decreases ENaC and α3β4 activity. Phorbol 12-myristate 13-acetate application reduced membrane capacitance ∼5%, presumably via increased endocytosis, but this decrease is much smaller than the SARS proteins' effects on conductances. Also, incubating oocytes in Gö-6976, a PKCα and PKCβ inhibitor, did not alter E or S protein-induced channel inhibition. We conclude that SARS-CoV-1 and SARS-CoV-2 proteins alter the function of human plasma membrane channels, via incompletely understood mechanisms. These interactions may play a role in the coronavirus 2019 pathophysiology.
Collapse
Affiliation(s)
- Stephen N Grant
- Division of Chemistry and Chemical Engineering, Pasadena, California
| | - Henry A Lester
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California.
| |
Collapse
|
59
|
Dvornikova KA, Bystrova EY, Churilov LP, Lerner A. Pathogenesis of the inflammatory bowel disease in context of SARS-COV-2 infection. Mol Biol Rep 2021; 48:5745-5758. [PMID: 34296352 PMCID: PMC8297608 DOI: 10.1007/s11033-021-06565-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
To date, the latest research results suggest that the novel severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) can enter host cells directly via the gastrointestinal tract by binding to the enterocyte-expressed ACE2 receptor, or indirectly as a result of infection of type II alveolar epithelial cells. At the same time, entry of SARS-CoV-2 through the gastrointestinal tract initiates the activation of innate and adaptive immune responses, the formation of an excessive inflammatory reaction and critical increase in the expression of proinflammatory cytokines, which, subsequently, can presumably increase inflammation and induce intestinal damage in patients suffering from inflammatory bowel disease (IBD). The aims of the present review were to reveal and analyze possible molecular pathways and consequences of the induction of an innate and adaptive immune response during infection with SARS-CoV-2 in patients with IBD. A thorough literature search was carried out by using the keywords: IBD, SARS-CoV-2, COVID-19. Based on the screening, a number of intracellular and extracellular pathways were considered and discussed, which can impact the immune response during SARS-CoV-2 infection in IBD patients. Additionally, the possible consequences of the infection for such patients were estimated. We further hypothesize that any virus, including the new SARS-CoV-2, infecting intestinal tissues and/or entering the host's body through receptors located on intestinal enterocytes may be a trigger for the onset of IBD in individuals with a genetic predisposition and/or the risk of developing IBD associated with other factors.
Collapse
Affiliation(s)
- K. A. Dvornikova
- Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russian Federation
| | - E. Yu. Bystrova
- Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russian Federation
| | - L. P. Churilov
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - A. Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| |
Collapse
|
60
|
Lenehan PJ, Ramudu E, Venkatakrishnan AJ, Berner G, McMurry R, O'Horo JC, Badley AD, Morice W, Halamka J, Soundararajan V. Anemia during SARS-CoV-2 infection is associated with rehospitalization after viral clearance. iScience 2021; 24:102780. [PMID: 34189429 PMCID: PMC8225287 DOI: 10.1016/j.isci.2021.102780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/01/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022] Open
Abstract
Patients with COVID-19 can experience symptoms and complications after viral clearance. It is important to identify clinical features of patients who are likely to experience these prolonged effects. We conducted a retrospective study to compare longitudinal laboratory test measurements (hemoglobin, hematocrit, estimated glomerular filtration rate, serum creatinine, and blood urea nitrogen) in patients rehospitalized after PCR-confirmed SARS-CoV-2 clearance (n = 104) versus patients not rehospitalized after viral clearance (n = 278). Rehospitalized patients had lower median hemoglobin levels in the year prior to COVID-19 diagnosis (Cohen's D = -0.50; p = 1.2 × 10-3) and during their active SARS-CoV-2 infection (Cohen's D = -0.71; p = 4.6 × 10-8). Rehospitalized patients were also more likely to be diagnosed with moderate or severe anemia during their active infection (Odds Ratio = 4.07; p = 4.99 × 10-9). These findings suggest that anemia-related laboratory tests should be considered in risk stratification algorithms for patients with COVID-19.
Collapse
Affiliation(s)
| | | | | | | | - Reid McMurry
- nference, One Main Street, Cambridge, MA 02142, USA
| | | | | | - William Morice
- Mayo Clinic, Rochester, MN 55902, USA.,Mayo Clinic Laboratories, Rochester, MN 55902, USA
| | | | | |
Collapse
|
61
|
Chen Y, Lear TB, Evankovich JW, Larsen MB, Lin B, Alfaras I, Kennerdell JR, Salminen L, Camarco DP, Lockwood KC, Tuncer F, Liu J, Myerburg MM, McDyer JF, Liu Y, Finkel T, Chen BB. A high-throughput screen for TMPRSS2 expression identifies FDA-approved compounds that can limit SARS-CoV-2 entry. Nat Commun 2021; 12:3907. [PMID: 34162861 PMCID: PMC8222394 DOI: 10.1038/s41467-021-24156-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 (2019-nCoV) is the pathogenic coronavirus responsible for the global pandemic of COVID-19 disease. The Spike (S) protein of SARS-CoV-2 attaches to host lung epithelial cells through the cell surface receptor ACE2, a process dependent on host proteases including TMPRSS2. Here, we identify small molecules that reduce surface expression of TMPRSS2 using a library of 2,560 FDA-approved or current clinical trial compounds. We identify homoharringtonine and halofuginone as the most attractive agents, reducing endogenous TMPRSS2 expression at sub-micromolar concentrations. These effects appear to be mediated by a drug-induced alteration in TMPRSS2 protein stability. We further demonstrate that halofuginone modulates TMPRSS2 levels through proteasomal-mediated degradation that involves the E3 ubiquitin ligase component DDB1- and CUL4-associated factor 1 (DCAF1). Finally, cells exposed to homoharringtonine and halofuginone, at concentrations of drug known to be achievable in human plasma, demonstrate marked resistance to SARS-CoV-2 infection in both live and pseudoviral in vitro models. Given the safety and pharmacokinetic data already available for the compounds identified in our screen, these results should help expedite the rational design of human clinical trials designed to combat active COVID-19 infection.
Collapse
Affiliation(s)
- Yanwen Chen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Travis B Lear
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - John W Evankovich
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mads B Larsen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
| | - Bo Lin
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
| | - Irene Alfaras
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
| | | | - Laura Salminen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
| | - Daniel P Camarco
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
| | | | - Ferhan Tuncer
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
| | - Jie Liu
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA
| | - Michael M Myerburg
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
| | - John F McDyer
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuan Liu
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA.
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Toren Finkel
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA.
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Medicine, Division of Cardiology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Bill B Chen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA, USA.
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA.
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
62
|
Raghav PK, Kalyanaraman K, Kumar D. Human cell receptors: potential drug targets to combat COVID-19. Amino Acids 2021; 53:813-842. [PMID: 33950300 PMCID: PMC8097256 DOI: 10.1007/s00726-021-02991-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 04/21/2021] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the coronavirus disease 2019 (COVID-19). The World Health Organization (WHO) has announced that COVID-19 is a pandemic having a higher spread rate rather than the mortality. Identification of a potential approach or therapy against COVID-19 is still under consideration. Therefore, it is essential to have an insight into SARS-CoV-2, its interacting partner, and domains for an effective treatment. The present study is divided into three main categories, including SARS-CoV-2 prominent receptor and its expression levels, other interacting partners, and their binding domains. The first section focuses primarily on coronaviruses' general aspects (SARS-CoV-2, SARS-CoV, and the Middle East Respiratory Syndrome Coronaviruses (MERS-CoV)) their structures, similarities, and mode of infections. The second section discusses the host receptors which includes the human targets of coronaviruses like dipeptidyl peptidase 4 (DPP4), CD147, CD209L, Angiotensin-Converting Enzyme 2 (ACE2), and other miscellaneous targets (type-II transmembrane serine proteases (TTSPs), furin, trypsin, cathepsins, thermolysin, elastase, phosphatidylinositol 3-phosphate 5-kinase, two-pore segment channel, and epithelium sodium channel C-α subunit). The human cell receptor, ACE2 plays an essential role in the Renin-Angiotensin system (RAS) pathway and COVID-19. Thus, this section also discusses the ACE2 expression and risk of COVID-19 infectivity in various organs and tissues such as the liver, lungs, intestine, heart, and reproductive system in the human body. Absence of ACE2 protein expression in immune cells could be used for limiting the SARS-CoV-2 infection. The third section covers the current available approaches for COVID-19 treatment. Overall, this review focuses on the critical role of human cell receptors involved in coronavirus pathogenesis, which would likely be used in designing target-specific drugs to combat COVID-19.
Collapse
Affiliation(s)
| | - Keerthana Kalyanaraman
- Amity Institute of Biotechnology, Amity University, Sector-125, Noida, Uttar Pradesh, India
| | - Dinesh Kumar
- ICMR-National Institute of Cancer Prevention & Research, Noida, 201301, India.
| |
Collapse
|
63
|
Abstract
The Epithelial Na+ Channel, ENaC, comprised of 3 subunits (αβγ, or sometimes δβγENaC), plays a critical role in regulating salt and fluid homeostasis in the body. It regulates fluid reabsorption into the blood stream from the kidney to control blood volume and pressure, fluid absorption in the lung to control alveolar fluid clearance at birth and maintenance of normal airway surface liquid throughout life, and fluid absorption in the distal colon and other epithelial tissues. Moreover, recent studies have also revealed a role for sodium movement via ENaC in nonepithelial cells/tissues, such as endothelial cells in blood vessels and neurons. Over the past 25 years, major advances have been made in our understanding of ENaC structure, function, regulation, and role in human disease. These include the recently solved three-dimensional structure of ENaC, ENaC function in various tissues, and mutations in ENaC that cause a hereditary form of hypertension (Liddle syndrome), salt-wasting hypotension (PHA1), or polymorphism in ENaC that contributes to other diseases (such as cystic fibrosis). Moreover, great strides have been made in deciphering the regulation of ENaC by hormones (e.g., the mineralocorticoid aldosterone, glucocorticoids, vasopressin), ions (e.g., Na+ ), proteins (e.g., the ubiquitin-protein ligase NEDD4-2, the kinases SGK1, AKT, AMPK, WNKs & mTORC2, and proteases), and posttranslational modifications [e.g., (de)ubiquitylation, glycosylation, phosphorylation, acetylation, palmitoylation]. Characterization of ENaC structure, function, regulation, and role in human disease, including using animal models, are described in this article, with a special emphasis on recent advances in the field. © 2021 American Physiological Society. Compr Physiol 11:1-29, 2021.
Collapse
Affiliation(s)
- Daniela Rotin
- The Hospital for Sick Children, and The University of Toronto, Toronto, Canada
| | - Olivier Staub
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
64
|
Dessie G, Malik T. Role of Serine Proteases and Host Cell Receptors Involved in Proteolytic Activation, Entry of SARS-CoV-2 and Its Current Therapeutic Options. Infect Drug Resist 2021; 14:1883-1892. [PMID: 34079299 PMCID: PMC8163626 DOI: 10.2147/idr.s308176] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/22/2021] [Indexed: 12/16/2022] Open
Abstract
The current global pandemic of a novel severe acute respiratory syndrome coronavirus-2 continues with its public health disaster beginning from late December 2019 in Wuhan, Hubei province, China. The scientific community has tried to fight against this novel coronavirus through vaccine development and designing different candidate drugs. However, there is no well-defined therapy to prevent 2019-nCov infection, thus complete prevention of the virus remains difficult. Therefore, it is a critical factor for death of millions worldwide. Many clinical trials and insights are ongoing in the struggle with this pandemic of SARS-CoV-2. SARS-CoV-2 entry into the host cell requires host cell angiotensin-converting enzyme-2 (ACE2) and glucose regulated protein 78 (GRP78). On the other hand, proteolytic activation of the viral spike protein (S protein) needs the host cell serine proteases, including transmembrane serine protease 2 (TMPRSS2), cathepsins, trypsin and furin. This review focuses on the protein involved in the mechanism of entry, and proteolytic activation. In addition, it looks at current therapeutic options for SARS-CoV-2.
Collapse
Affiliation(s)
- Gashaw Dessie
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tabarak Malik
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
65
|
Gruca A, Ziemska-Legiecka J, Jarnot P, Sarnowska E, Sarnowski TJ, Grynberg M. Common low complexity regions for SARS-CoV-2 and human proteomes as potential multidirectional risk factor in vaccine development. BMC Bioinformatics 2021; 22:182. [PMID: 33832440 PMCID: PMC8027979 DOI: 10.1186/s12859-021-04017-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The rapid spread of the COVID-19 demands immediate response from the scientific communities. Appropriate countermeasures mean thoughtful and educated choice of viral targets (epitopes). There are several articles that discuss such choices in the SARS-CoV-2 proteome, other focus on phylogenetic traits and history of the Coronaviridae genome/proteome. However none consider viral protein low complexity regions (LCRs). Recently we created the first methods that are able to compare such fragments. RESULTS We show that five low complexity regions (LCRs) in three proteins (nsp3, S and N) encoded by the SARS-CoV-2 genome are highly similar to regions from human proteome. As many as 21 predicted T-cell epitopes and 27 predicted B-cell epitopes overlap with the five SARS-CoV-2 LCRs similar to human proteins. Interestingly, replication proteins encoded in the central part of viral RNA are devoid of LCRs. CONCLUSIONS Similarity of SARS-CoV-2 LCRs to human proteins may have implications on the ability of the virus to counteract immune defenses. The vaccine targeted LCRs may potentially be ineffective or alternatively lead to autoimmune diseases development. These findings are crucial to the process of selection of new epitopes for drugs or vaccines which should omit such regions.
Collapse
Affiliation(s)
- Aleksandra Gruca
- Department of Computer Networks and Systems, Silesian University of Technology, Gliwice, Poland
| | | | - Patryk Jarnot
- Department of Computer Networks and Systems, Silesian University of Technology, Gliwice, Poland
| | - Elzbieta Sarnowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Tomasz J Sarnowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Marcin Grynberg
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
66
|
Mishra S, Bassi G, Nyomba BLG. Inter-proteomic posttranslational modifications of the SARS-CoV-2 and the host proteins ‒ A new frontier. Exp Biol Med (Maywood) 2021; 246:749-757. [PMID: 33467896 PMCID: PMC8719024 DOI: 10.1177/1535370220986785] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
Posttranslational modification of proteins, which include both the enzymatic alterations of protein side chains and main-chain peptide bond connectivity, is a fundamental regulatory process that is crucial for almost every aspects of cell biology, including the virus-host cell interaction and the SARS-CoV-2 infection. The posttranslational modification of proteins has primarily been studied in cells and tissues in an intra-proteomic context (where both substrates and enzymes are part of the same species). However, the inter-proteomic posttranslational modifications of most of the SARS-CoV-2 proteins by the host enzymes and vice versa are largely unexplored in virus pathogenesis and in the host immune response. It is now known that the structural spike (S) protein of the SARS-CoV-2 undergoes proteolytic priming by the host serine proteases for entry into the host cells, and N- and O-glycosylation by the host cell enzymes during virion packaging, which enable the virus to spread. New evidence suggests that both SARS-CoV-2 and the host proteins undergo inter-proteomic posttranslational modifications, which play roles in virus pathogenesis and infection-induced immune response by hijacking the host cell signaling. The purpose of this minireview is to bring attention of the scientific community to recent cutting-edge discoveries in this understudied area. It is likely that a better insight into the molecular mechanisms involved may open new research directions, and thereby contribute to novel therapeutic modality development against the SARS-CoV-2. Here we briefly discuss the rationale and touch upon some unanswered questions in this context, especially those that require attention from the scientific community.
Collapse
Affiliation(s)
- Suresh Mishra
- Department of Internal Medicine, College of Medicine, Faculty of Health Sciences, University of Manitoba, Manitoba R3E 3P4, Canada
- Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Manitoba R3E 3P4, Canada
| | - Geetika Bassi
- Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Manitoba R3E 3P4, Canada
| | - BL Grégoire Nyomba
- Department of Internal Medicine, College of Medicine, Faculty of Health Sciences, University of Manitoba, Manitoba R3E 3P4, Canada
| |
Collapse
|
67
|
Yapici-Eser H, Koroglu YE, Oztop-Cakmak O, Keskin O, Gursoy A, Gursoy-Ozdemir Y. Neuropsychiatric Symptoms of COVID-19 Explained by SARS-CoV-2 Proteins' Mimicry of Human Protein Interactions. Front Hum Neurosci 2021; 15:656313. [PMID: 33833673 PMCID: PMC8021734 DOI: 10.3389/fnhum.2021.656313] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/23/2021] [Indexed: 12/16/2022] Open
Abstract
The first clinical symptoms focused on the presentation of coronavirus disease 2019 (COVID-19) have been respiratory failure, however, accumulating evidence also points to its presentation with neuropsychiatric symptoms, the exact mechanisms of which are not well known. By using a computational methodology, we aimed to explain the molecular paths of COVID-19 associated neuropsychiatric symptoms, based on the mimicry of the human protein interactions with SARS-CoV-2 proteins. Methods: Available 11 of the 29 SARS-CoV-2 proteins' structures have been extracted from Protein Data Bank. HMI-PRED (Host-Microbe Interaction PREDiction), a recently developed web server for structural PREDiction of protein-protein interactions (PPIs) between host and any microbial species, was used to find the "interface mimicry" through which the microbial proteins hijack host binding surfaces. Classification of the found interactions was conducted using the PANTHER Classification System. Results: Predicted Human-SARS-CoV-2 protein interactions have been extensively compared with the literature. Based on the analysis of the molecular functions, cellular localizations and pathways related to human proteins, SARS-CoV-2 proteins are found to possibly interact with human proteins linked to synaptic vesicle trafficking, endocytosis, axonal transport, neurotransmission, growth factors, mitochondrial and blood-brain barrier elements, in addition to its peripheral interactions with proteins linked to thrombosis, inflammation and metabolic control. Conclusion: SARS-CoV-2-human protein interactions may lead to the development of delirium, psychosis, seizures, encephalitis, stroke, sensory impairments, peripheral nerve diseases, and autoimmune disorders. Our findings are also supported by the previous in vivo and in vitro studies from other viruses. Further in vivo and in vitro studies using the proteins that are pointed here, could pave new targets both for avoiding and reversing neuropsychiatric presentations.
Collapse
Affiliation(s)
- Hale Yapici-Eser
- Department of Psychiatry, School of Medicine, Koç University, Istanbul, Turkey
- Research Center for Translational Medicine, Koç University, Istanbul, Turkey
| | - Yunus Emre Koroglu
- Research Center for Translational Medicine, Koç University, Istanbul, Turkey
- Graduate School of Sciences and Engineering, College of Engineering, Koç University, Istanbul, Turkey
| | - Ozgur Oztop-Cakmak
- Research Center for Translational Medicine, Koç University, Istanbul, Turkey
- Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
| | - Ozlem Keskin
- Research Center for Translational Medicine, Koç University, Istanbul, Turkey
- College of Engineering, Chemical and Biological Engineering, Koç University, Istanbul, Turkey
| | - Attila Gursoy
- Research Center for Translational Medicine, Koç University, Istanbul, Turkey
- Department of Computer Science and Engineering, College of Engineering, Koç University, Istanbul, Turkey
| | - Yasemin Gursoy-Ozdemir
- Research Center for Translational Medicine, Koç University, Istanbul, Turkey
- Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
68
|
Kumar A, Prasoon P, Kumari C, Pareek V, Faiq MA, Narayan RK, Kulandhasamy M, Kant K. SARS-CoV-2-specific virulence factors in COVID-19. J Med Virol 2021; 93:1343-1350. [PMID: 33085084 DOI: 10.1002/jmv.26615] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/25/2022]
Abstract
The paucity of knowledge about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific virulence factors has greatly hampered the therapeutic management of patients with coronavirus disease 2019 (COVID-19). Recently, a cluster of studies appeared, which presented empirical evidence for SARS-CoV-2-specific virulence factors that can explain key elements of COVID-19 pathology. These studies unravel multiple structural and nonstructural specifics of SARS-CoV-2, such as a unique FURIN cleavage site, papain-like protease (SCoV2-PLpro), ORF3b and nonstructural proteins, and dynamic conformational changes in the structure of spike protein during host cell fusion, which give it an edge in infectivity and virulence over previous coronaviruses causing pandemics. Investigators provided robust evidence that SARS-CoV-2-specific virulence factors may have an impact on viral infectivity and transmissibility and disease severity as well as the development of immunity against the infection, including response to the vaccines. In this article, we are presenting a summarized account of the newly reported studies.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Pranav Prasoon
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Pittsburgh Center for Pain Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chiman Kumari
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anatomy, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Vikas Pareek
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- National Brain Research Center, Manesar, Haryana, India
| | - Muneeb A Faiq
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- New York University (NYU) Langone Health Center, NYU Robert I Grossman School of Medicine, New York, New York, USA
| | - Ravi K Narayan
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Maheswari Kulandhasamy
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Biochemistry, Maulana Azad Medical College (MAMC), New Delhi, India
| | - Kamla Kant
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Bathinda, India
| |
Collapse
|
69
|
Abstract
PURPOSE OF REVIEW The aim of this study was to evaluate the relationship between infection with SARS-CoV-2 and autoimmunity. RECENT FINDINGS Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome (SARS) associated coronavirus 2 (SARS-CoV-2). Although most of the infected individuals are asymptomatic, a proportion of patients with COVID-19 develop severe disease with multiple organ injuries. Evidence suggests that some medications used to treat autoimmune rheumatologic diseases might have therapeutic effect in patients with severe COVID-19 infections, drawing attention to the relationship between COVID-19 and autoimmune diseases. COVID-19 shares similarities with autoimmune diseases in clinical manifestations, immune responses and pathogenic mechanisms. Robust immune reactions participate in the pathogenesis of both disease conditions. Autoantibodies as a hallmark of autoimmune diseases can also be detected in COVID-19 patients. Moreover, some patients have been reported to develop autoimmune diseases, such as Guillain--Barré syndrome or systemic lupus erythematosus, after COVID-19 infection. It is speculated that SARS-CoV-2 can disturb self-tolerance and trigger autoimmune responses through cross-reactivity with host cells. The infection risk and prognosis of COVID-19 in patients with autoimmune diseases remains controversial, but patient adherence to medication regimens to prevent autoimmune disease flares is strongly recommended. SUMMARY We present a review of the association between COVID-19 and autoimmune diseases, focusing on similarities in immune responses, cross-reactivity of SARS-CoV-2, the development of autoimmune diseases in COVID-19 patients and the risk of COVID-19 infection in patients with preexisting autoimmune conditions.
Collapse
Affiliation(s)
- Yu Liu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenetics, Changsha, Hunan, PR China
| | - Amr H. Sawalha
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenetics, Changsha, Hunan, PR China
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
70
|
Casey W, Massey SE, Mishra B. How signalling games explain mimicry at many levels: from viral epidemiology to human sociology. J R Soc Interface 2021; 18:20200689. [PMID: 33622145 DOI: 10.1098/rsif.2020.0689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mimicry is exhibited in multiple scales, ranging from molecular, to organismal, and then to human society. 'Batesian'-type mimicry entails a conflict of interest between sender and receiver, reflected in a deceptive mimic signal. 'Müllerian'-type mimicry occurs when there is perfect common interest between sender and receiver in a particular type of encounter, manifested by an honest co-mimic signal. Using a signalling games approach, simulations show that invasion by Batesian mimics will make Müllerian mimicry unstable, in a coevolutionary chase. We use these results to better understand the deceptive strategies of SARS-CoV-2 and their key role in the COVID-19 pandemic. At the biomolecular level, we explain how cellularization promotes Müllerian molecular mimicry, and discourages Batesian molecular mimicry. A wide range of processes analogous to cellularization are presented; these might represent a manner of reducing oscillatory instabilities. Lastly, we identify examples of mimicry in human society that might be addressed using a signalling game approach.
Collapse
Affiliation(s)
- William Casey
- United States Naval Academy, Annapolis, MD 21402, USA
| | - Steven E Massey
- Biology Department, University of Puerto Rico - Rio Piedras, San Juan, PR 00931, USA
| | - Bud Mishra
- New York University Courant Institute of Mathematical Sciences, New York, NY 10012-1110, USA
| |
Collapse
|
71
|
Corona-Cov-2 (COVID-19) and ginseng: Comparison of possible use in COVID-19 and influenza. J Ginseng Res 2021; 45:535-537. [PMID: 33623472 PMCID: PMC7891076 DOI: 10.1016/j.jgr.2020.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 01/27/2023] Open
Abstract
In the 1918 influenza pandemic, more than 95% of mortalities were ascribed to bacterial pneumonia. After the primary influenza infection, the innate immune system is attenuated, and the susceptibility to bacteria is increased. Subsequent bacterial pneumonia exacerbates morbidity and increases the mortality rate. Similarly, COVID-19 infection attenuates innate immunity and results in pneumonia. In addition, the current pneumococcal conjugate vaccine may have limited defense against secondary pneumococcal infection after influenza infection. Therefore, until a fully protective vaccine is available, a method of increasing immunity may be helpful. Ginseng has been shown to increase the defense against influenza in clinical trials and animal experiments, as well as the defense against pneumococcal pneumonia in animal experiments. Based on these findings, ginseng is suspected to be helpful for providing immunity against COVID-19.
Collapse
|
72
|
Stavropoulou E, Kantartzi K, Tsigalou C, Konstantinidis T, Voidarou C, Konstantinidis T, Bezirtzoglou E. Unraveling the Interconnection Patterns Across Lung Microbiome, Respiratory Diseases, and COVID-19. Front Cell Infect Microbiol 2021; 10:619075. [PMID: 33585285 PMCID: PMC7876344 DOI: 10.3389/fcimb.2020.619075] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/11/2020] [Indexed: 01/07/2023] Open
Abstract
Albeit the lungs were thought to be sterile, recent scientific data reported a microbial microbiota in the lungs of healthy individuals. Apparently, new developments in technological approachesincluding genome sequencing methodologies contributed in the identification of the microbiota and shed light on the role of the gut and lung microbiomes in the development of respiratory diseases. Moreover, knowledge of the human microbiome in health may act as a tool for evaluating characteristic shifts in the case of disease. This review paper discusses the development of respiratory disease linked to the intestinal dysbiosis which influences the lung immunity and microbiome. The gastrointestinal-lung dialogue provides interesting aspects in the pathogenesis of the respiratory diseases. Lastly, we were further interested on the role of this interconnection in the progression and physiopathology of newly emergedCOVID-19.
Collapse
Affiliation(s)
- Elisavet Stavropoulou
- CHUV (Centre HospitalierUniversitaire Vaudois), Lausanne, Switzerland
- Department of Infectious Diseases, Central Institute, Valais Hospital, Sion, Switzerland
| | - Konstantia Kantartzi
- Nephrology Clinic, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Christina Tsigalou
- Laboratory of Microbiology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Theocharis Konstantinidis
- Laboratory of Microbiology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Theodoros Konstantinidis
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Eugenia Bezirtzoglou
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
73
|
Vojdani A, Vojdani E, Kharrazian D. Reaction of Human Monoclonal Antibodies to SARS-CoV-2 Proteins With Tissue Antigens: Implications for Autoimmune Diseases. Front Immunol 2021; 11:617089. [PMID: 33584709 PMCID: PMC7873987 DOI: 10.3389/fimmu.2020.617089] [Citation(s) in RCA: 210] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
We sought to determine whether immune reactivity occurs between anti-SARS-CoV-2 protein antibodies and human tissue antigens, and whether molecular mimicry between COVID-19 viral proteins and human tissues could be the cause. We applied both human monoclonal anti-SARS-Cov-2 antibodies (spike protein, nucleoprotein) and rabbit polyclonal anti-SARS-Cov-2 antibodies (envelope protein, membrane protein) to 55 different tissue antigens. We found that SARS-CoV-2 antibodies had reactions with 28 out of 55 tissue antigens, representing a diversity of tissue groups that included barrier proteins, gastrointestinal, thyroid and neural tissues, and more. We also did selective epitope mapping using BLAST and showed similarities and homology between spike, nucleoprotein, and many other SARS-CoV-2 proteins with the human tissue antigens mitochondria M2, F-actin and TPO. This extensive immune cross-reactivity between SARS-CoV-2 antibodies and different antigen groups may play a role in the multi-system disease process of COVID-19, influence the severity of the disease, precipitate the onset of autoimmunity in susceptible subgroups, and potentially exacerbate autoimmunity in subjects that have pre-existing autoimmune diseases. Very recently, human monoclonal antibodies were approved for use on patients with COVID-19. The human monoclonal antibodies used in this study are almost identical with these approved antibodies. Thus, our results can establish the potential risk for autoimmunity and multi-system disorders with COVID-19 that may come from cross-reactivity between our own human tissues and this dreaded virus, and thus ensure that the badly-needed vaccines and treatments being developed for it are truly safe to use against this disease.
Collapse
Affiliation(s)
- Aristo Vojdani
- Department of Immunology, Immunosciences Laboratory, Inc., Los Angeles, CA, United States.,Department of Preventive Medicine, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | | | - Datis Kharrazian
- Department of Preventive Medicine, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Neurology, Harvard Medical School, Boston, MA, United States.,Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| |
Collapse
|
74
|
Finkelstein MT, Mermelstein AG, Parker Miller E, Seth PC, Stancofski ESD, Fera D. Structural Analysis of Neutralizing Epitopes of the SARS-CoV-2 Spike to Guide Therapy and Vaccine Design Strategies. Viruses 2021; 13:134. [PMID: 33477902 PMCID: PMC7833398 DOI: 10.3390/v13010134] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/01/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus research has gained tremendous attention because of the COVID-19 pandemic, caused by the novel severe acute respiratory syndrome coronavirus (nCoV or SARS-CoV-2). In this review, we highlight recent studies that provide atomic-resolution structural details important for the development of monoclonal antibodies (mAbs) that can be used therapeutically and prophylactically and for vaccines against SARS-CoV-2. Structural studies with SARS-CoV-2 neutralizing mAbs have revealed a diverse set of binding modes on the spike's receptor-binding domain and N-terminal domain and highlight alternative targets on the spike. We consider this structural work together with mAb effects in vivo to suggest correlations between structure and clinical applications. We also place mAbs against severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) coronaviruses in the context of the SARS-CoV-2 spike to suggest features that may be desirable to design mAbs or vaccines capable of conferring broad protection.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniela Fera
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, PA 19081, USA; (M.T.F.); (A.G.M.); (E.P.M.); (P.C.S.); (E.-S.D.S.)
| |
Collapse
|
75
|
Hou Y, Ding Y, Nie H, Ji HL. Fibrinolysis influences SARS-CoV-2 infection in ciliated cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.07.425801. [PMID: 33442688 PMCID: PMC7805445 DOI: 10.1101/2021.01.07.425801] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Rapid spread of COVID-19 has caused an unprecedented pandemic worldwide, and an inserted furin site in SARS-CoV-2 spike protein (S) may account for increased transmissibility. Plasmin, and other host proteases, may cleave the furin site of SARS-CoV-2 S protein and γ subunits of epithelial sodium channels (γ ENaC), resulting in an increment in virus infectivity and channel activity. As for the importance of ENaC in the regulation of airway surface and alveolar fluid homeostasis, whether SARS-CoV-2 will share and strengthen the cleavage network with ENaC proteins at the single-cell level is urgently worthy of consideration. To address this issue, we analyzed single-cell RNA sequence (scRNA-seq) datasets, and found the PLAU (encoding urokinase plasminogen activator), SCNN1G (γENaC), and ACE2 (SARS-CoV-2 receptor) were co-expressed in alveolar epithelial, basal, club, and ciliated epithelial cells. The relative expression level of PLAU, TMPRSS2, and ACE2 were significantly upregulated in severe COVID-19 patients and SARS-CoV-2 infected cell lines using Seurat and DESeq2 R packages. Moreover, the increments in PLAU, FURIN, TMPRSS2, and ACE2 were predominately observed in different epithelial cells and leukocytes. Accordingly, SARS-CoV-2 may share and strengthen the ENaC fibrinolytic proteases network in ACE2 positive airway and alveolar epithelial cells, which may expedite virus infusion into the susceptible cells and bring about ENaC associated edematous respiratory condition.
Collapse
|
76
|
Kumar N, Golhar R, Sharma KS, Holloway JL, Sarangi S, Neuhaus I, Walsh AM, Pitluk ZW. Rapid single cell evaluation of human disease and disorder targets using REVEAL: SingleCell™. BMC Genomics 2021; 22:5. [PMID: 33407110 PMCID: PMC7785925 DOI: 10.1186/s12864-020-07300-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/02/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Single-cell (sc) sequencing performs unbiased profiling of individual cells and enables evaluation of less prevalent cellular populations, often missed using bulk sequencing. However, the scale and the complexity of the sc datasets poses a great challenge in its utility and this problem is further exacerbated when working with larger datasets typically generated by consortium efforts. As the scale of single cell datasets continues to increase exponentially, there is an unmet technological need to develop database platforms that can evaluate key biological hypotheses by querying extensive single-cell datasets. Large single-cell datasets like Human Cell Atlas and COVID-19 cell atlas (collection of annotated sc datasets from various human organs) are excellent resources for profiling target genes involved in human diseases and disorders ranging from oncology, auto-immunity, as well as infectious diseases like COVID-19 caused by SARS-CoV-2 virus. SARS-CoV-2 infections have led to a worldwide pandemic with massive loss of lives, infections exceeding 7 million cases. The virus uses ACE2 and TMPRSS2 as key viral entry associated proteins expressed in human cells for infections. Evaluating the expression profile of key genes in large single-cell datasets can facilitate testing for diagnostics, therapeutics, and vaccine targets, as the world struggles to cope with the on-going spread of COVID-19 infections. MAIN BODY In this manuscript we describe REVEAL: SingleCell, which enables storage, retrieval, and rapid query of single-cell datasets inclusive of millions of cells. The array native database described here enables selecting and analyzing cells across multiple studies. Cells can be selected using individual metadata tags, more complex hierarchical ontology filtering, and gene expression threshold ranges, including co-expression of multiple genes. The tags on selected cells can be further evaluated for testing biological hypotheses. One such example includes identifying the most prevalent cell type annotation tag on returned cells. We used REVEAL: SingleCell to evaluate the expression of key SARS-CoV-2 entry associated genes, and queried the current database (2.2 Million cells, 32 projects) to obtain the results in < 60 s. We highlighted cells expressing COVID-19 associated genes are expressed on multiple tissue types, thus in part explains the multi-organ involvement in infected patients observed worldwide during the on-going COVID-19 pandemic. CONCLUSION In this paper, we introduce the REVEAL: SingleCell database that addresses immediate needs for SARS-CoV-2 research and has the potential to be used more broadly for many precision medicine applications. We used the REVEAL: SingleCell database as a reference to ask questions relevant to drug development and precision medicine regarding cell type and co-expression for genes that encode proteins necessary for SARS-CoV-2 to enter and reproduce in cells.
Collapse
Affiliation(s)
- Namit Kumar
- Informatics & Predictive Sciences, Bristol Myers Squibb, Princeton, NJ, 08648, USA
| | - Ryan Golhar
- Informatics & Predictive Sciences, Bristol Myers Squibb, Princeton, NJ, 08648, USA
| | - Kriti Sen Sharma
- Paradigm4, Inc., Suite 360, 281 Winter Street, Waltham, MA, 02451, USA
| | - James L Holloway
- Informatics & Predictive Sciences, Bristol Myers Squibb, Redwood City, CA, 94063, USA
| | - Srikant Sarangi
- Paradigm4, Inc., Suite 360, 281 Winter Street, Waltham, MA, 02451, USA
| | - Isaac Neuhaus
- Informatics & Predictive Sciences, Bristol Myers Squibb, Princeton, NJ, 08648, USA
| | - Alice M Walsh
- Informatics & Predictive Sciences, Bristol Myers Squibb, Princeton, NJ, 08648, USA
| | - Zachary W Pitluk
- Paradigm4, Inc., Suite 360, 281 Winter Street, Waltham, MA, 02451, USA.
| |
Collapse
|
77
|
Bitossi C, Frasca F, Viscido A, Oliveto G, Scordio M, Belloni L, Cimino G, Pietropaolo V, Gentile M, d’Ettorre G, Midulla F, Trancassini M, Antonelli G, Pierangeli A, Scagnolari C. SARS-CoV-2 Entry Genes Expression in Relation with Interferon Response in Cystic Fibrosis Patients. Microorganisms 2021; 9:microorganisms9010093. [PMID: 33401565 PMCID: PMC7824643 DOI: 10.3390/microorganisms9010093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 01/14/2023] Open
Abstract
The expression rate of SARS-CoV-2 entry genes, angiotensin-converting enzyme 2 (ACE2), the main viral receptor and the proteases, furin and transmembrane serine protease 2 (TMPRSS2) in cystic fibrosis (CF) individuals is poorly known. Hence, we examined their levels in upper respiratory samples of CF patients (n = 46) and healthy controls (n = 45). Moreover, we sought to understand the interplay of type I interferon (IFN-I) with ACE2, furin and TMPRSS2 by evaluating their gene expression with respect to ISG15, a well-known marker of IFN activation, in upper respiratory samples and after ex vivo IFNβ exposure. Lower ACE2 levels and trends toward the reduction of furin and TMPRSS2 were found in CF patients compared with the healthy controls; decreased ACE2 amounts were also detected in CF individuals with pancreatic insufficiency and in those receiving inhaled antibiotics. Moreover, there was a strong positive correlation between ISG15 and ACE2 levels. However, after ex vivo IFNβ stimulation of nasopharyngeal cells, the truncated isoform (dACE2), recently demonstrated as the IFN stimulated one with respect to the full-length isoform (flACE2), slightly augmented in cells from CF patients whereas in those from healthy donors, dACE2 levels showed variable levels of upregulation. An altered expression of SARS-COV-2 entry genes and a poor responsiveness of dACE2 to IFN-I stimulation might be crucial in the diffusion of SARS-CoV-2 infection in CF.
Collapse
Affiliation(s)
- Camilla Bitossi
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
| | - Federica Frasca
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
| | - Agnese Viscido
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
| | - Giuseppe Oliveto
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
| | - Mirko Scordio
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
| | - Laura Belloni
- Department of Internal Medicine, Sapienza University, 00161 Rome, Italy;
| | - Giuseppe Cimino
- Lazio Reference Center for Cystic Fibrosis, Policlinico Umberto I University Hospital, Sapienza University, 00161 Rome, Italy;
| | - Valeria Pietropaolo
- Department of Public Health and Infectious Diseases, Sapienza University, 00161 Rome, Italy; (V.P.); (G.d.); (M.T.)
- Microbiology and Virology Unit, Policlinico Umberto I University Hospital, Sapienza University, 00161 Rome, Italy
| | - Massimo Gentile
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
- Microbiology and Virology Unit, Policlinico Umberto I University Hospital, Sapienza University, 00161 Rome, Italy
| | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University, 00161 Rome, Italy; (V.P.); (G.d.); (M.T.)
| | - Fabio Midulla
- Department of Maternal Science, Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria Trancassini
- Department of Public Health and Infectious Diseases, Sapienza University, 00161 Rome, Italy; (V.P.); (G.d.); (M.T.)
- Microbiology and Virology Unit, Policlinico Umberto I University Hospital, Sapienza University, 00161 Rome, Italy
| | - Guido Antonelli
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
- Microbiology and Virology Unit, Policlinico Umberto I University Hospital, Sapienza University, 00161 Rome, Italy
| | - Alessandra Pierangeli
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, Istituto Pasteur Italia, Sapienza University, 00161 Rome, Italy; (C.B.); (F.F.); (A.V.); (G.O.); (M.S.); (M.G.); (G.A.); (A.P.)
- Correspondence:
| |
Collapse
|
78
|
Bain R, Cosgriff R, Zampoli M, Elbert A, Burgel PR, Carr SB, Castaños C, Colombo C, Corvol H, Faro A, Goss CH, Gutierrez H, Jung A, Kashirskaya N, Marshall BC, Melo J, Mondejar-Lopez P, de Monestrol I, Naehrlich L, Padoan R, Pastor-Vivero MD, Rizvi S, Salvatore M, Filho LVRFDS, Brownlee KG, Haq IJ, Brodlie M. Clinical characteristics of SARS-CoV-2 infection in children with cystic fibrosis: An international observational study. J Cyst Fibros 2021; 20:25-30. [PMID: 33309057 PMCID: PMC7713571 DOI: 10.1016/j.jcf.2020.11.021] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND The presence of co-morbidities, including underlying respiratory problems, has been identified as a risk factor for severe COVID-19 disease. Information on the clinical course of SARS-CoV-2 infection in children with cystic fibrosis (CF) is limited, yet vital to provide accurate advice for children with CF, their families, caregivers and clinical teams. METHODS Cases of SARS-CoV-2 infection in children with CF aged less than 18 years were collated by the CF Registry Global Harmonization Group across 13 countries between 1 February and 7 August 2020. RESULTS Data on 105 children were collated and analysed. Median age of cases was ten years (interquartile range 6-15), 54% were male and median percentage predicted forced expiratory volume in one second was 94% (interquartile range 79-104). The majority (71%) of children were managed in the community during their COVID-19 illness. Out of 24 children admitted to hospital, six required supplementary oxygen and two non-invasive ventilation. Around half were prescribed antibiotics, five children received antiviral treatments, four azithromycin and one additional corticosteroids. Children that were hospitalised had lower lung function and reduced body mass index Z-scores. One child died six weeks after testing positive for SARS-CoV-2 following a deterioration that was not attributed to COVID-19 disease. CONCLUSIONS SARS-CoV-2 infection in children with CF is usually associated with a mild illness in those who do not have pre-existing severe lung disease.
Collapse
Affiliation(s)
- Robert Bain
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Marco Zampoli
- Division of Paediatric Pulmonology, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, South Africa
| | | | - Pierre-Régis Burgel
- Respiratory Medicine and National Reference CF Center, AP-HP Hôpital Cochin, Paris, France; Université de Paris, Institut Cochin, Inserm U-1016, Paris, France
| | - Siobhán B Carr
- Royal Brompton Hospital and Imperial College London, United Kingdom
| | - Claudio Castaños
- Department of Pulmonology, Hospital de Pediatria JP Garrahan, Buenos Aires, Argentina
| | - Carla Colombo
- CF Regional Reference Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Italy
| | - Harriet Corvol
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Assistance Publique Hôpitaux de Paris (APHP), Hôpital Trousseau, Service de Pneumologie Pédiatrique, Paris, France
| | - Albert Faro
- Cystic Fibrosis Foundation, Bethesda, MD, United States
| | - Christopher H Goss
- Department of Medicine and Pediatrics, University of Washington, Seattle, WA, United States
| | - Hector Gutierrez
- Pediatric Pulmonary and Sleep Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andreas Jung
- Department of Pulmonology and Children's Research Centre, University Children's Hospital Zurich, Zurich, Switzerland
| | - Nataliya Kashirskaya
- Laboratory of Genetic Epidemiology, Research Centre for Medical Genetics, Moscow, Russian Federation
| | | | - Joel Melo
- Instituo Nacional del Tórax, Santiago, Chile
| | - Pedro Mondejar-Lopez
- Pediatric Pulmonology and Cystic Fibrosis Unit, Hospital Clinico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Isabelle de Monestrol
- Stockholm Cystic Fibrosis Centre Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Lutz Naehrlich
- Universities of Giessen and Marburg Lung Center, German Center of Lung Research, Justus-Liebig-University Giessen, Giessen, Germany
| | - Rita Padoan
- Cystic Fibrosis Support Center, Department of Paediatric, University of Brescia, Italy
| | - Maria Dolores Pastor-Vivero
- Pediatric Pulmonology and Cystic Fibrosis Unit, Osakidetza, Hospital Universitario Cruces, Barakado, Bizkaia, Spain
| | - Samar Rizvi
- Cystic Fibrosis Foundation, Bethesda, MD, United States
| | - Marco Salvatore
- National Center Rare Diseases, Undiagnosed Rare Diseases Interdepartmental Unit Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Iram J Haq
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Paediatric Respiratory Medicine, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Malcolm Brodlie
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Paediatric Respiratory Medicine, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
79
|
Aguirre García MM, Mancilla-Galindo J, Paredes-Paredes M, Tiburcio ÁZ, Ávila-Vanzzini N. Mechanisms of infection by SARS-CoV-2, inflammation and potential links with the microbiome. Future Virol 2021. [PMCID: PMC7876557 DOI: 10.2217/fvl-2020-0310] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The pandemic SARS coronavirus 2 utilizes efficient mechanisms to establish infection and evade the immune system. Established infection leads to severe inflammation in susceptible patients, the main hallmark of progression to severe coronavirus disease (COVID-19). Knowledge of the mechanisms of disease has expanded rapidly. As inflammation emerges as the central pathophysiological feature in COVID-19, elucidating how the immune system, lungs and gut communicate and interact with microbial components of the ecological niches that conform the human microbiome will shed light on how inflammation and disease progression are promoted. Studying the microbiome in COVID-19 could allow scientists to identify novel approaches to prevent severe inflammation by targeting components of the human microbiome. Innovation in the aforementioned is needed to combat this pandemic.
Collapse
Affiliation(s)
- María Magdalena Aguirre García
- División de Investigación, Facultad de Medicina, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan C.P. 14080, Ciudad de México, Mexico
| | - Javier Mancilla-Galindo
- División de Investigación, Facultad de Medicina, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan C.P. 14080, Ciudad de México, Mexico
| | - Mercedes Paredes-Paredes
- División de Investigación, Facultad de Medicina, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan C.P. 14080, Ciudad de México, Mexico
| | - Álvaro Zamudio Tiburcio
- Departamento de Gastroenterología, Unidad de Trasplante de Microbiota Intestinal, Especialidades Médicas Nápoles, Oficina 12, Pennsylvania No. 209 Esq. Kansas, Col. Nápoles, Benito Juárez C.P 03810, Ciudad de México, Mexico
| | - Nydia Ávila-Vanzzini
- Departamento de Consulta Externa, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Tlalpan C.P. 14080, Ciudad de México, Mexico
| |
Collapse
|
80
|
Abstract
Coronavirus infection (COVID-19) is an acute viral disease, which affects all vital organs and is caused by an RNA-genomic virus of the genus Betacoronavirus of the family Coronaviridae. This virus (SARS-CoV-2) enters the body through the respiratory tract and interacts primarily with Toll-like receptors of epithelial cells of the bronchi, alveoli, intestines and vascular endotheliocytes, as well as with angiotensin-converting enzyme 2 receptors. Toll-like receptors activate nuclear factor Kappa B in these cells, which initiates the formation of many cytokines (“cytokine storm”). SARS-CoV-2 affects type II pneumocytes by causing a termination of surfactant formation and, accordingly, alveolar shrinking and the formation of acute respiratory distress syndrome and also fibrosis on the interalveolar-capillary membrane and the formation of acute respiratory failure. SARS-CoV-2 and cytokines disrupt the function of vascular endothelial cells, which leads to endothelial dysfunction. In microvessels forms a mass formation of microthrombi, which causes the failure of organs and systems. “Cytokine storm” turns into cytokine sepsis with the formation of multiple organ dysfunction syndrome.
Collapse
Affiliation(s)
- S. B. Bolevich
- Sechenov First Moscow State Medical University (Sechenov University)
| | - S. S. Bolevich
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
81
|
Munir K, Ashraf S, Munir I, Khalid H, Muneer MA, Mukhtar N, Amin S, Ashraf S, Imran MA, Chaudhry U, Zaheer MU, Arshad M, Munir R, Ahmad A, Zhao X. Zoonotic and reverse zoonotic events of SARS-CoV-2 and their impact on global health. Emerg Microbes Infect 2020; 9:2222-2235. [PMID: 32967592 PMCID: PMC7594747 DOI: 10.1080/22221751.2020.1827984] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/25/2020] [Accepted: 09/20/2020] [Indexed: 12/12/2022]
Abstract
Coronaviruses (CoVs) are enveloped, positive sense, single-stranded RNA viruses. The viruses have adapted to infect a large number of animal species, ranging from bats to camels. At present, seven CoVs infect humans, of which Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is responsible for causing the Coronavirus Disease 2019 (COVID-19) in humans. Since its emergence in late 2019, SARS-CoV-2 has spread rapidly across the globe. Healthcare systems around the globe have been stretched beyond their limits posing new challenges to emergency healthcare services and critical care. The outbreak continues to jeopardize human health, social life and economy. All known human CoVs have zoonotic origins. Recent detection of SARS-CoV-2 in pet, zoo and certain farm animals has highlighted its potential for reverse zoonosis. This scenario is particularly alarming, since these animals could be potential reservoirs for secondary zoonotic infections. In this article, we highlight interspecies SARS-CoV-2 infections and focus on the reverse zoonotic potential of this virus. We also emphasize the importance of potential secondary zoonotic events and the One-Health and One-World approach to tackle such future pandemics.
Collapse
Affiliation(s)
- Khalid Munir
- PetLife Veterinary Professional Corporation, NJ, USA
- Department of Pathobiology, Riphah College of Veterinary Sciences, Riphah University, Lahore, Pakistan
| | - Shoaib Ashraf
- Department of Pathobiology, Riphah College of Veterinary Sciences, Riphah University, Lahore, Pakistan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isra Munir
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamna Khalid
- Department of Chemical and Biological Engineering, Villanova University, Villanova, PA, USA
| | - Mohammad Akram Muneer
- Department of Pathobiology, Riphah College of Veterinary Sciences, Riphah University, Lahore, Pakistan
| | | | - Shahid Amin
- Animal Hospital of Loves Park, Loves Park, IL, USA
| | - Sohaib Ashraf
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Shaikh Zayed Hospital Lahore, Lahore, Pakistan
| | | | - Umer Chaudhry
- ; Royal (Dick) School of Veterinary Studies and Roslin Institute, Edinburgh, UK
| | - Muhammad Usman Zaheer
- Food and Agriculture Organization of the United Nations, Country Office, Islamabad, Pakistan
| | | | - Rukhsana Munir
- Consultant Emergency Medicine, Russells Hall Hospital, Dudley Group of Hospitals NHS Trust, Dudley, UK
| | - Ali Ahmad
- CHU Sainte-Justine Research Center, Department of Microbiology, Infectious Diseases and Immunology, University of Montreal, Montreal, Canada
| | - Xin Zhao
- Department of Animal Sciences, McGill University, Sainte-Anne-de-Bellevue, Canada
| |
Collapse
|
82
|
Szabó GT, Kiss A, Csanádi Z, Czuriga D. Hypothetical dysfunction of the epithelial sodium channel may justify neurohumoral blockade in coronavirus disease 2019. ESC Heart Fail 2020; 8:171-174. [PMID: 33205539 PMCID: PMC7753344 DOI: 10.1002/ehf2.13078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 10/06/2020] [Indexed: 12/29/2022] Open
Affiliation(s)
- Gábor Tamás Szabó
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zs. krt., Debrecen, H-4032, Hungary
| | - Attila Kiss
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Zoltán Csanádi
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zs. krt., Debrecen, H-4032, Hungary
| | - Dániel Czuriga
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zs. krt., Debrecen, H-4032, Hungary
| |
Collapse
|
83
|
Goh GKM, Dunker AK, Foster JA, Uversky VN. Shell Disorder Analysis Suggests That Pangolins Offered a Window for a Silent Spread of an Attenuated SARS-CoV-2 Precursor among Humans. J Proteome Res 2020; 19:4543-4552. [PMID: 32790362 PMCID: PMC7640969 DOI: 10.1021/acs.jproteome.0c00460] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Indexed: 02/06/2023]
Abstract
A model to predict the relative levels of respiratory and fecal-oral transmission potentials of coronaviruses (CoVs) by measuring the percentage of protein intrinsic disorder (PID) of the M (Membrane) and N (Nucleoprotein) proteins in their outer and inner shells, respectively, was built before the MERS-CoV outbreak. With MPID = 8.6% and NPID = 50.2%, the 2003 SARS-CoV falls into group B, which consists of CoVs with intermediate levels of both fecal-oral and respiratory transmission potentials. Further validation of the model came with MERS-CoV (MPID = 9%, NPID = 44%) and SARS-CoV-2 (MPID = 5.5%, NPID = 48%) falling into the groups C and B, respectively. Group C contains CoVs with higher fecal-oral but lower respiratory transmission potentials. Unlike SARS-CoV, SARS-CoV-2 with MPID = 5.5% has one of the hardest outer shells among CoVs. Because the hard shell is able to resist the antimicrobial enzymes in body fluids, the infected person is able to shed large quantities of viral particles via saliva and mucus, which could account for the higher contagiousness of SARS-COV-2. Further searches have found that high rigidity of the outer shell is characteristic for the CoVs of burrowing animals, such as rabbits (MPID = 5.6%) and pangolins (MPID = 5-6%), which are in contact with the buried feces. A closer inspection of pangolin-CoVs from 2017 to 2019 reveals that pangolins provided a unique window of opportunity for the entry of an attenuated SARS-CoV-2 precursor into the human population in 2017 or earlier, with the subsequent slow and silent spread as a mild cold that followed by its mutations into the current more virulent form. Evidence of this lies in both the genetic proximity of the pangolin-CoVs to SARS-CoV-2 (∼90%) and differences in N disorder. A 2017 pangolin-CoV strain shows evidence of higher levels of attenuation and higher fecal-oral transmission associated with lower human infectivity via having lower NPID (44.8%). Our shell disorder model predicts this to be a SARS-CoV-2 vaccine strain, as lower inner shell disorder is associated with the lesser virulence in a variety of viruses.
Collapse
Affiliation(s)
| | - A. Keith Dunker
- Center
for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - James A. Foster
- Department
of Biological Sciences, University of Idaho, Moscow, Idaho 83844, United States
- Institute
for Bioinformatics and Evolutionary Studies, University of Idaho, Moscow, Idaho 83844, United States
| | - Vladimir N. Uversky
- Department
of Molecular Medicine, USF Health Byrd Alzheimer’s Research
Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida 33620, United States
- Laboratory
of New Methods in Biology, Institute for Biological Instrumentation
of the Russian Academy of Sciences, Federal
Research Center “Pushchino Scientific Center for Biological
Research of the Russian Academy of Sciences”, Pushchino, Moscow 119991, Russia
| |
Collapse
|
84
|
Muhanna D, Arnipalli SR, Kumar SB, Ziouzenkova O. Osmotic Adaptation by Na +-Dependent Transporters and ACE2: Correlation with Hemostatic Crisis in COVID-19. Biomedicines 2020; 8:E460. [PMID: 33142989 PMCID: PMC7693583 DOI: 10.3390/biomedicines8110460] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19 symptoms, including hypokalemia, hypoalbuminemia, ageusia, neurological dysfunctions, D-dimer production, and multi-organ microthrombosis reach beyond effects attributed to impaired angiotensin-converting enzyme 2 (ACE2) signaling and elevated concentrations of angiotensin II (Ang II). Although both SARS-CoV (Severe Acute Respiratory Syndrome Coronavirus) and SARS-CoV-2 utilize ACE2 for host entry, distinct COVID-19 pathogenesis coincides with the acquisition of a new sequence, which is homologous to the furin cleavage site of the human epithelial Na+ channel (ENaC). This review provides a comprehensive summary of the role of ACE2 in the assembly of Na+-dependent transporters of glucose, imino and neutral amino acids, as well as the functions of ENaC. Data support an osmotic adaptation mechanism in which osmotic and hemostatic instability induced by Ang II-activated ENaC is counterbalanced by an influx of organic osmolytes and Na+ through the ACE2 complex. We propose a paradigm for the two-site attack of SARS-CoV-2 leading to ENaC hyperactivation and inactivation of the ACE2 complex, which collapses cell osmolality and leads to rupture and/or necrotic death of swollen pulmonary, endothelial, and cardiac cells, thrombosis in infected and non-infected tissues, and aberrant sensory and neurological perception in COVID-19 patients. This dual mechanism employed by SARS-CoV-2 calls for combinatorial treatment strategies to address and prevent severe complications of COVID-19.
Collapse
Affiliation(s)
| | | | | | - Ouliana Ziouzenkova
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; (D.M.); (S.R.A.); (S.B.K.)
| |
Collapse
|
85
|
Kumar A, Prasoon P, Sekhawat PS, Pareek V, Faiq MA, Kumari C, Narayan RK, Kulandhasamy M, Kant K. Pathogenesis guided therapeutic management of COVID-19: an immunological perspective. Int Rev Immunol 2020; 40:54-71. [PMID: 33111578 DOI: 10.1080/08830185.2020.1840566] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lack of standardized therapeutic approaches is arguably the significant contributor to the high burden of mortality observed in the ongoing pandemic of the Coronavirus disease, 2019 (COVID-19). Evidence is accumulating on SARS-CoV-2 specific immune cell dysregulation and consequent tissue injury in COVID-19. Currently, no definite drugs or vaccines are available against the disease; however initial results of the ongoing clinical trials have raised some hope. In this article, taking insights from the emerging empirical evidence about host-virus interactions, we deliberate upon plausible pathogenic mechanisms and suitable therapeutic approaches for COVID-19.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Pranav Prasoon
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,Pittsburgh Center for Pain Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Prakash S Sekhawat
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,Department of Hematology, Nil RatanSircar Medical College and Hospital (NRSMCH), Kolkata, India
| | - Vikas Pareek
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,National Brain Research Center, Manesar, Haryana, India
| | - Muneeb A Faiq
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,NYU Robert I Grossman School of Medicine, New York University (NYU) Langone Health Center, New York, New York, USA
| | - Chiman Kumari
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,Department of Anatomy, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ravi K Narayan
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Maheswari Kulandhasamy
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,Department of Biochemistry, Maulana Azad Medical College (MAMC), New Delhi, India
| | - Kamla Kant
- Etiologically Elusive Disorders Research Network (EEDRN), New Delhi, India.,Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Bathinda, India
| |
Collapse
|
86
|
Ehsani S. COVID-19 and iron dysregulation: distant sequence similarity between hepcidin and the novel coronavirus spike glycoprotein. Biol Direct 2020; 15:19. [PMID: 33066821 PMCID: PMC7563913 DOI: 10.1186/s13062-020-00275-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
The spike glycoprotein of the SARS-CoV-2 virus, which causes COVID-19, has attracted attention for its vaccine potential and binding capacity to host cell surface receptors. Much of this research focus has centered on the ectodomain of the spike protein. The ectodomain is anchored to a transmembrane region, followed by a cytoplasmic tail. Here we report a distant sequence similarity between the cysteine-rich cytoplasmic tail of the coronavirus spike protein and the hepcidin protein that is found in humans and other vertebrates. Hepcidin is thought to be the key regulator of iron metabolism in humans through its inhibition of the iron-exporting protein ferroportin. An implication of this preliminary observation is to suggest a potential route of investigation in the coronavirus research field making use of an already-established literature on the interplay of local and systemic iron regulation, cytokine-mediated inflammatory processes, respiratory infections and the hepcidin protein. The question of possible homology and an evolutionary connection between the viral spike protein and hepcidin is not assessed in this report, but some scenarios for its study are discussed.
Collapse
Affiliation(s)
- Sepehr Ehsani
- Theoretical and Philosophical Biology, Department of Philosophy, University College London, Bloomsbury, London, WC1E 6BT, UK.
- Ronin Institute for Independent Scholarship, Montclair, NJ, 07043, USA.
| |
Collapse
|
87
|
Gentzsch M, Rossier BC. A Pathophysiological Model for COVID-19: Critical Importance of Transepithelial Sodium Transport upon Airway Infection. FUNCTION 2020; 1:zqaa024. [PMID: 33201937 PMCID: PMC7662147 DOI: 10.1093/function/zqaa024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 01/06/2023] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic remains a serious public health problem and will continue to be until effective drugs and/or vaccines are available. The rational development of drugs critically depends on our understanding of disease mechanisms, that is, the physiology and pathophysiology underlying the function of the organ targeted by the virus. Since the beginning of the pandemic, tireless efforts around the globe have led to numerous publications on the virus, its receptor, its entry into the cell, its cytopathic effects, and how it triggers innate and native immunity but the role of apical sodium transport mediated by the epithelial sodium channel (ENaC) during the early phases of the infection in the airways has received little attention. We propose a pathophysiological model that defines the possible role of ENaC in this process.
Collapse
Affiliation(s)
- Martina Gentzsch
- Department of Cell Biology and Physiology, Marsico Lung Institute, University of North Carolina, Chapel Hill, USA
- Department of Pediatric Pulmonology, Marsico Lung Institute, University of North Carolina, Chapel Hill, USA
| | - Bernard C Rossier
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
88
|
Endothelial Cell Contributions to COVID-19. Pathogens 2020; 9:pathogens9100785. [PMID: 32992810 PMCID: PMC7600722 DOI: 10.3390/pathogens9100785] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 01/08/2023] Open
Abstract
Understanding of the clinical, histological and molecular features of the novel coronavirus 2019 (Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)) has remained elusive. Coronavirus disease 2019 (COVID-19) caused by this virus has unusual clinical presentation with regard to other related coronaviruses. Recent reports suggest that SARS-CoV-2, unlike other related viruses, infects and replicates within endothelial cells, which may explain a significant portion of the observed clinical pathology. Likewise, mounting evidence associates vascular and endothelial cell dysfunction with increased mortality. This review focuses on understanding how endothelial cell pathology is caused by SARS-CoV-2 at the molecular and cellular levels and how these events relate to COVID-19. A detailed examination of current knowledge regarding canonical inflammatory reaction pathways as well as alteration of endothelial cell-derived exosomes and transdifferentiation by SARS-CoV-2 is included in this assessment. Additionally, given an understanding of endothelial contributions to COVID-19, potential therapeutic aims are discussed, particularly as would affect endothelial function and pathology.
Collapse
|
89
|
Bohn MK, Hall A, Sepiashvili L, Jung B, Steele S, Adeli K. Pathophysiology of COVID-19: Mechanisms Underlying Disease Severity and Progression. Physiology (Bethesda) 2020; 35:288-301. [PMID: 32783610 PMCID: PMC7426542 DOI: 10.1152/physiol.00019.2020] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The global epidemiology of coronavirus disease 2019 (COVID-19) suggests a wide spectrum of clinical severity, ranging from asymptomatic to fatal. Although the clinical and laboratory characteristics of COVID-19 patients have been well characterized, the pathophysiological mechanisms underlying disease severity and progression remain unclear. This review highlights key mechanisms that have been proposed to contribute to COVID-19 progression from viral entry to multisystem organ failure, as well as the central role of the immune response in successful viral clearance or progression to death.
Collapse
Affiliation(s)
- Mary Kathryn Bohn
- Molecular Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Alexandra Hall
- Molecular Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Lusia Sepiashvili
- Molecular Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Jung
- Molecular Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Shannon Steele
- Molecular Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Khosrow Adeli
- Molecular Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
90
|
Chen Y, Lear TB, Evankovich JW, Larsen MB, Lin B, Alfaras I, Kennerdell JR, Salminen L, Camarco DP, Lockwood KC, Liu J, Myerburg MM, McDyer JF, Liu Y, Finkel T, Chen BB. A high throughput screen for TMPRSS2 expression identifies FDA-approved and clinically advanced compounds that can limit SARS-CoV-2 entry. RESEARCH SQUARE 2020:rs.3.rs-48659. [PMID: 32818215 PMCID: PMC7430593 DOI: 10.21203/rs.3.rs-48659/v1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
SARS-CoV-2 (2019-nCoV) is the pathogenic coronavirus responsible for the global pandemic of COVID-19 disease. The Spike (S) protein of SARS-CoV-2 attaches to host lung epithelial cells through the cell surface receptor ACE2, a process dependent on host proteases including TMPRSS2. Here, we identified small molecules that can reduce surface expression of TMPRSS2 using a 2,700 FDA-approved or current clinical trial compounds. Among these, homoharringtonine and halofuginone appear the most potent agents, reducing endogenous TMPRSS2 expression at sub-micromolar concentrations. These effects appear to be mediated by a drug-induced alteration in TMPRSS2 protein stability. We further demonstrate that halofuginone modulates TMPRSS2 levels through proteasomal-mediated degradation that involves the E3 ubiquitin ligase component DDB1- and CUL4-associated factor 1 (DCAF1). Finally, cells exposed to homoharringtonine and halofuginone, at concentrations of drug known to be achievable in human plasma, demonstrated marked resistance to SARS-CoV-2 pseudoviral infection. Given the safety and pharmacokinetic data already available for the compounds identified in our screen, these results should help expedite the rational design of human clinical trials designed to combat COVID-19 infection.
Collapse
Affiliation(s)
- Yanwen Chen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Travis B. Lear
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - John W. Evankovich
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mads B. Larsen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Bo Lin
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Irene Alfaras
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | | | - Laura Salminen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Daniel P. Camarco
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Karina C. Lockwood
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Jie Liu
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
| | - Michael M. Myerburg
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - John F. McDyer
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yuan Liu
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Toren Finkel
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Medicine, Division of Cardiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Bill B. Chen
- Aging Institute, University of Pittsburgh/UPMC, Pittsburgh, PA 15219, USA
- Department of Medicine, Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
91
|
Casey W, Massey SE, Mishra B. How Signaling Games Explain Mimicry at Many Levels: From Viral Epidemiology to Human Sociology. RESEARCH SQUARE 2020:rs.3.rs-51959. [PMID: 32793895 PMCID: PMC7418725 DOI: 10.21203/rs.3.rs-51959/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mimicry is exhibited in multiple scales, ranging from molecular, to organismal, and then to human society. 'Batesian' type mimicry entails a conflict of interest between sender and receiver, reflected in a deceptive mimic signal. 'Mullerian' type mimicry occurs when there is perfect common interest between sender and receiver, manifested by an honest co-mimic signal. Using a signaling games approach, simulations show that invasion by Batesian mimics will make Mullerian mimicry unstable, in a coevolutionary chase. We use these results to better understand the deceptive strategies of SARS-CoV-2 and their key role in the COVID-19 pandemic. At the biomolecular level, we explain how cellularization promotes Mullerian molecular mimicry, and discourages Batesian molecular mimicry. A wide range of processes analogous to cellularization are presented; these might represent a manner of reducing oscillatory instabilities. Lastly, we identify examples of mimicry in human society, that might be addressed using a signaling game approach.
Collapse
|
92
|
Xing Y, Li X, Gao X, Dong Q. Natural Polymorphisms Are Present in the Furin Cleavage Site of the SARS-CoV-2 Spike Glycoprotein. Front Genet 2020; 11:783. [PMID: 32765596 PMCID: PMC7379507 DOI: 10.3389/fgene.2020.00783] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/01/2020] [Indexed: 11/26/2022] Open
Abstract
The furin cleavage site in the spike glycoprotein of the SARS-CoV-2 coronavirus is considered important for the virus to enter the host cells. By analyzing 45828 SARS-CoV-2 genome sequences, we identified 103 strains of SARS-CoV-2 with various DNA mutations including 18 unique non-synonymous point mutations, one deletion, and six gains of premature stop codon that may affect the furin cleavage site. Our results revealed that the furin cleavage site might not be required for SARS-CoV-2 to enter human cells in vivo. The identified mutants may represent a new subgroup of SARS-CoV-2 coronavirus with reduced tropism and transmissibility as potential live-attenuated vaccine candidates.
Collapse
Affiliation(s)
- Yue Xing
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Xiao Li
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States
| | - Xiang Gao
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Qunfeng Dong
- Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Center for Biomedical Informatics, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
93
|
Wagner T, Shweta FNU, Murugadoss K, Awasthi S, Venkatakrishnan AJ, Bade S, Puranik A, Kang M, Pickering BW, O'Horo JC, Bauer PR, Razonable RR, Vergidis P, Temesgen Z, Rizza S, Mahmood M, Wilson WR, Challener D, Anand P, Liebers M, Doctor Z, Silvert E, Solomon H, Anand A, Barve R, Gores G, Williams AW, Morice WG, Halamka J, Badley A, Soundararajan V. Augmented curation of clinical notes from a massive EHR system reveals symptoms of impending COVID-19 diagnosis. eLife 2020; 9:e58227. [PMID: 32633720 PMCID: PMC7410498 DOI: 10.7554/elife.58227] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/06/2020] [Indexed: 01/09/2023] Open
Abstract
Understanding temporal dynamics of COVID-19 symptoms could provide fine-grained resolution to guide clinical decision-making. Here, we use deep neural networks over an institution-wide platform for the augmented curation of clinical notes from 77,167 patients subjected to COVID-19 PCR testing. By contrasting Electronic Health Record (EHR)-derived symptoms of COVID-19-positive (COVIDpos; n = 2,317) versus COVID-19-negative (COVIDneg; n = 74,850) patients for the week preceding the PCR testing date, we identify anosmia/dysgeusia (27.1-fold), fever/chills (2.6-fold), respiratory difficulty (2.2-fold), cough (2.2-fold), myalgia/arthralgia (2-fold), and diarrhea (1.4-fold) as significantly amplified in COVIDpos over COVIDneg patients. The combination of cough and fever/chills has 4.2-fold amplification in COVIDpos patients during the week prior to PCR testing, in addition to anosmia/dysgeusia, constitutes the earliest EHR-derived signature of COVID-19. This study introduces an Augmented Intelligence platform for the real-time synthesis of institutional biomedical knowledge. The platform holds tremendous potential for scaling up curation throughput, thus enabling EHR-powered early disease diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - William G Morice
- Mayo ClinicRochesterUnited States
- Mayo Clinic LaboratoriesRochesterUnited States
| | | | | | | |
Collapse
|
94
|
Wagner T, Shweta F, Murugadoss K, Awasthi S, Venkatakrishnan AJ, Bade S, Puranik A, Kang M, Pickering BW, O'Horo JC, Bauer PR, Razonable RR, Vergidis P, Temesgen Z, Rizza S, Mahmood M, Wilson WR, Challener D, Anand P, Liebers M, Doctor Z, Silvert E, Solomon H, Anand A, Barve R, Gores G, Williams AW, Morice WG, Halamka J, Badley A, Soundararajan V. Augmented curation of clinical notes from a massive EHR system reveals symptoms of impending COVID-19 diagnosis. eLife 2020; 9:58227. [PMID: 32633720 DOI: 10.1101/2020.04.19.20067660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/06/2020] [Indexed: 05/27/2023] Open
Abstract
Understanding temporal dynamics of COVID-19 symptoms could provide fine-grained resolution to guide clinical decision-making. Here, we use deep neural networks over an institution-wide platform for the augmented curation of clinical notes from 77,167 patients subjected to COVID-19 PCR testing. By contrasting Electronic Health Record (EHR)-derived symptoms of COVID-19-positive (COVIDpos; n = 2,317) versus COVID-19-negative (COVIDneg; n = 74,850) patients for the week preceding the PCR testing date, we identify anosmia/dysgeusia (27.1-fold), fever/chills (2.6-fold), respiratory difficulty (2.2-fold), cough (2.2-fold), myalgia/arthralgia (2-fold), and diarrhea (1.4-fold) as significantly amplified in COVIDpos over COVIDneg patients. The combination of cough and fever/chills has 4.2-fold amplification in COVIDpos patients during the week prior to PCR testing, in addition to anosmia/dysgeusia, constitutes the earliest EHR-derived signature of COVID-19. This study introduces an Augmented Intelligence platform for the real-time synthesis of institutional biomedical knowledge. The platform holds tremendous potential for scaling up curation throughput, thus enabling EHR-powered early disease diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - William G Morice
- Mayo Clinic, Rochester, United States
- Mayo Clinic Laboratories, Rochester, United States
| | | | | | | |
Collapse
|
95
|
|
96
|
Vojdani A, Vojdani E, Kharrazian D. Reaction of Human Monoclonal Antibodies to SARS-CoV-2 Proteins With Tissue Antigens: Implications for Autoimmune Diseases. Front Immunol 2020; 11:617089. [PMID: 33584709 DOI: 10.3389/fimmu.2020.617089/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/14/2020] [Indexed: 05/20/2023] Open
Abstract
We sought to determine whether immune reactivity occurs between anti-SARS-CoV-2 protein antibodies and human tissue antigens, and whether molecular mimicry between COVID-19 viral proteins and human tissues could be the cause. We applied both human monoclonal anti-SARS-Cov-2 antibodies (spike protein, nucleoprotein) and rabbit polyclonal anti-SARS-Cov-2 antibodies (envelope protein, membrane protein) to 55 different tissue antigens. We found that SARS-CoV-2 antibodies had reactions with 28 out of 55 tissue antigens, representing a diversity of tissue groups that included barrier proteins, gastrointestinal, thyroid and neural tissues, and more. We also did selective epitope mapping using BLAST and showed similarities and homology between spike, nucleoprotein, and many other SARS-CoV-2 proteins with the human tissue antigens mitochondria M2, F-actin and TPO. This extensive immune cross-reactivity between SARS-CoV-2 antibodies and different antigen groups may play a role in the multi-system disease process of COVID-19, influence the severity of the disease, precipitate the onset of autoimmunity in susceptible subgroups, and potentially exacerbate autoimmunity in subjects that have pre-existing autoimmune diseases. Very recently, human monoclonal antibodies were approved for use on patients with COVID-19. The human monoclonal antibodies used in this study are almost identical with these approved antibodies. Thus, our results can establish the potential risk for autoimmunity and multi-system disorders with COVID-19 that may come from cross-reactivity between our own human tissues and this dreaded virus, and thus ensure that the badly-needed vaccines and treatments being developed for it are truly safe to use against this disease.
Collapse
Affiliation(s)
- Aristo Vojdani
- Department of Immunology, Immunosciences Laboratory, Inc., Los Angeles, CA, United States
- Department of Preventive Medicine, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | | | - Datis Kharrazian
- Department of Preventive Medicine, Loma Linda University School of Medicine, Loma Linda, CA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| |
Collapse
|