Coronavirus Disease-19 (COVID-19): Different Models and Treatment Strategies -  - E-Book

Coronavirus Disease-19 (COVID-19): Different Models and Treatment Strategies E-Book

0,0
72,24 €

-100%
Sammeln Sie Punkte in unserem Gutscheinprogramm und kaufen Sie E-Books und Hörbücher mit bis zu 100% Rabatt.
Mehr erfahren.
Beschreibung

With the emergence of new coronavirus variants, epidemiology, host tropism presents an opportunity for a thorough analysis of their evolution and acquired adaptability to their host. These studies require different animal models and treatment approaches. No studies are complete without animal models closely related to human physiology to replicate the disease and observe the pathological conditions for comparison to human cases. Such animal models play a vital role in virus pathogenesis and prepare a therapeutic immune response in the organism studied.
This volume focuses on two aspects of COVID-19 infection: Models for SARS-CoV-2 infection and Treatment Strategies for SARS-CoV-2 infection, respectively. Chapters in the first part describe bio-engineered transgenic mouse models with specific genes (including models created with CRISPR-Cas9 gene editing tools used previously for SARS-CoV and MERS-CoV), cell culture lines for COVID-19 studies and the histopathology of COVID-19. The section also includes a chapter covering essential proteins that up or down-regulate SARS-CoV-2 multiplication. The last chapter of this part describes other diseases having similar signs and symptoms and their differential diagnosis. The next section covers different treatment strategies for COVID-19. There is no specific treatment available to date, just symptomatic therapy. However, scientists have been testing antiviral drugs in clinical trials, phytochemicals, photomedicine such as UV A & B light, homemade remedies, blood plasma transfusion, stem cell therapy, and computational approaches in both in vivo and in vitro trials. This book serves as an ideal resource on these aspects of COVID-19 for academicians, scientists, and health professionals.

Das E-Book können Sie in Legimi-Apps oder einer beliebigen App lesen, die das folgende Format unterstützen:

EPUB

Seitenzahl: 669

Veröffentlichungsjahr: 2021

Bewertungen
0,0
0
0
0
0
0
Mehr Informationen
Mehr Informationen
Legimi prüft nicht, ob Rezensionen von Nutzern stammen, die den betreffenden Titel tatsächlich gekauft oder gelesen/gehört haben. Wir entfernen aber gefälschte Rezensionen.



Table of Contents
BENTHAM SCIENCE PUBLISHERS LTD.
End User License Agreement (for non-institutional, personal use)
Usage Rules:
Disclaimer:
Limitation of Liability:
General:
FOREWORD
PREFACE
List of Contributors
Part I: Models for COVID-19
Genetically Engineered Mouse Models for COVID-19
Abstract
INTRODUCTION
Emerging Role of CRISPR/Cas9
Human ACE2 Expression
ACE2 Knockout Mouse for Acute respiratory Distress Syndrome (ARDS) Investigation
K18-hACE2 Transgenic Mouse
TMPRSS2 Knockout Mouse for Viral Entry and Pathogenesis Studies
STAT1 Knockout Mouse for Studying Pneumonia and Antiviral Strategies
Standard Mouse Strains (BALB/c mice, C57BL/6, and 129S6)
Inbred Mice for Pathogenesis and Therapeutic Response Studies
Humanized Immune System Mice for Vaccine Approaches
CONCLUSION AND FUTURE PERSPECTIVES
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Different Cell Lines for SARS-CoV-2
Abstract
INTRODUCTION
Role of Cell Lines in Scientific Research
Caco-2 cells
Huh-7 cells
A549 cells
Human Intestinal Organoids
HAE cells
HEK 293T cells
Calu-3 cells
Vero Cells
Therapeutic Agents Using in vitro Models Especially Vero and Vero Derived Cell Lines
Remdesivir
Lopinavir
Homorringtonine
Emetine hydrochloride
Ivermectin
Chloroquine/Hydroxychloroquine
Nelfinavir
Indomethacin
Merimepodib
Scutellaria baicalensis
Carmofur
Arbidol
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Histopathologic Evaluation and Scoring of SARS-CoV-2 Infection
Abstract
INTRODUCTION
Importance of Histopathology Technique
Principles
Techniques and Types
Technique for Immuno-histopathology, RNA scope, or immunofluorescent staining
Scoring Methods
Recent Case Studies of Coronavirus and its Types
Virological Investigations
Blood Cell Examination and Counts
Pathological Characteristics of COVID-19
Lung
Gastrointestinal (GI) tract
Liver
Kidney
Skin
Heart
Brain
Blood vessels and placenta
Animal Models and Real Situation
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Key Proteins and Their Roles in COVID-19
Abstract
INTRODUCTION
Classification Track of the SARS-CoV-2
Structural Biology of COVID-19 Proteins
The Structural Proteins
Spike Protein
Envelope Protein
Membrane Protein
Nucleocapsid Protein
Non-Structural Proteins (NSPs)
NSP1 (Leader Protein)
NSP2
NSP3 (Papain-like Protease)
NSP4 (Contains Transmembrane Domain 2) and NSP5 (3C-like Proteinase)
NSP6 (Putative Transmembrane Domain)
NSP7, NSP8, and NSP9
NSP10, NSP11, and NSP12 (RNA-dependent RNA polymerase)
NSP13 (Helicase)
NSP14 (3′ to 5′ Endonuclease, N7-Methyltransferase)
NSP15 (endoRNAse)
NSP16 (2′-O-Ribose-Methyltransferase)
Accessory Proteins
ORF3a Protein
ORF6 Protein
ORF7a and ORF7b Protein
ORF8 Protein
ORF10 Protein
Pathogenesis in Real-Time Replication of COVID-19
Receptor Recognition by Spike Protein
Mechanism of Membrane Fusion by Spike Protein
Activators for Membrane Fusion by Spike Protein
Evolution of Spike Proteins
Invasion and Replication of SARS-CoV-2
Antigen Appearance in COVID-19 Infection
Activation of Humoral and Cellular Immunity against Covid-19 Infection
Cytokine Storm in COVID-19
Coronavirus Immune Evasion
Factors Affecting SARS-CoV-2 Pathogenesis
Host Factors on the Genetic Basis
Viral Factors
RNA-dependent RNA-polymerase (RdRp)
Papain-like Protease (PLpro)
Coronavirus Protease (3CLpro)
Spike Protein (S Protein)
Comparison of Different Coronaviruses
Virology and Genome
Epidemiology
Key Protein Differences in Clinical Findings
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Differential Diagnosis of COVID-19
Abstract
INTRODUCTION
Differential Diagnosis of COVID-19
Differential Etiology
COVID-19 and other Coronaviruses
Adenovirus
Influenza Virus
Human Metapneumovirus (HMPV)
Parainfluenza Virus
Respiratory Syncytial Virus (RSV)
Rhinovirus (Common Cold)
Bacterial Pneumonia
Streptococcus Pneumoniae
Haemophilus Influenza
Moraxella Catarrhalis Pneumonia
Chlamydial Pneumonia
M. Pneumonia
Comparative Modes of Transmission and Epidemiology
COVID-19 and other Coronaviruses (SARS, MERS)
Adenovirus
Influenza Virus
Human Metapneumovirus (HMPV)
Parainfluenza Virus
Respiratory Syncytial Virus (RSV)
Rhinovirus (Common cold)
Bacterial Pneumonia
Mycoplasma pneumonia
Comparative Pathogenesis
COVID-19 and Other Coronaviruses (SARS, MERS)
Adenovirus
Influenza Virus
Human Metapneumovirus (HMPV)
Parainfluenza Virus
Respiratory Syncytial Virus (RSV)
Rhinovirus (Common Cold)
Bacterial Pneumonia
Mycoplasma pneumonia
Differential Signs and Symptoms
COVID-19 and other Coronaviruses (SARS, MERS)
Adenovirus
Influenza Virus
Human Metapneumovirus (HMPV)
Parainfluenza Virus
Respiratory Syncytial Virus (RSV)
Rhinovirus (Common cold)
Bacterial Pneumonia
Mycoplasma pneumonia
Differential Diagnostic Approaches
COVID-19 and other Coronaviruses (SARS, MERS)
Adenovirus
Influenza Virus
Human Metapneumovirus (HMPV)
Parainfluenza Virus
Respiratory Syncytial Virus (RSV)
Rhinovirus (Common cold)
Bacterial Pneumonia
Mycoplasma Pneumonia
Differential Response Therapy
COVID-19 and other Coronaviruses (SARS, MERS)
Adenovirus
Influenza Virus
Human Metapneumovirus (HMPV)
Parainfluenza Virus
Respiratory Syncytial Virus (RSV)
Rhinovirus (Common cold)
Bacterial pneumonia
Mycoplasma pneumonia
Mathematical Modelling in the Prediction of COVID-19
Salient and Latest Models Applied
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Part II: Treatment Strategies for COVID-19
Different Antiviral Drugs against COVID-19 and Future Perspective
Abstract
INTRODUCTION
Mechanisms of Different Antivirals Actions in COVID-19 Treatments
Different Antiviral Drugs used in COVID-19 Treatment
Broad-spectrum Antiviral Agents (BSAA)
Remdesivir
Umifenovir (Arbidol)
Favipiravir (T-705 or Avigan)
Oseltamivir (Tamiflu)
Ribavirin
Lopinavir-Ritonavir
Baloxavir Marboxil
Chloroquine and Hydroxychloroquine
Other Antiviral Agents
BSAA Combination Therapy
Clinical Trials on Treatments for COVID-19
FUTURE PERSPECTIVE
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Phytochemicals Effective against COVID-19 and Future Perspective
Abstract
INTRODUCTION
Various Phytochemicals Activities against COVID-19
Allicin (garlic compounds)
Glycyrrhizin
Quercetin
Flavonoids
Catechin
Curcumin
Miscellaneous Phytochemicals against COVID-19
Carvacrol, Oleanolic Acid and Ursolic Acid
Mechanism of Phytochemicals in Altering SARS-CoV-2 Infection
a. Inhibition of Spike Proteins to Bind with Angiotensin-converting Enzyme 2 (ACE2)
b. Inhibition of Viral Replication
Plant Extracts and their Possible Anti-Viral Activities in COVID-19
FUTURE PERSPECTIVE OF PHYTOCHEMICALS IN COVID-19
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Ultraviolet Radiation A (UVA): Modulates the Production of Nitric Oxide (NO) to Combat COVID-19
Abstract
INTRODUCTION
Solar Light and Ultraviolet Radiation System
Long term Environmental Exposure of Ambient Solar UVA Radiation and COVID-19
Coronavirus
What is SARS-CoV-2?
Mode of Transmission of SARS-CoV-2
Prevention Strategies Adopted
UVA Mediates Production of NO
Effect of NO on SARS-CoV-2
NO: A Key Player in the Airways
NO Blocks Virus Replication Inside Host Cells
NO Reduces the Inflammatory Influx and Oxidative Stress
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Herbal Remedies Effective against COVID-19
Abstract
INTRODUCTION
Symptomatic Comparison of SARS-CoV, MERS-CoV, and COVID-19
Medicinal Plants against Coronaviruses
Traditional Chinese Medicine (TCM) against Coronaviruses
TCM Prescription for COVID 19
TCM to Prevent COVID-19 Infection
TCM to Treat the Mild Infection
TCM to Treat Severe Infection
Wild Plants against COVID-19
Indian Subcontinents Plants against COVID-19
Ashwagandha (Withania somnifera)
Harsinghar (Nyctanthes Arbortristis)
Giloy (Tinospora Cordifolia)
Neem (Azadirachta indica)
Tulsi (Ocimum sanctum)
AYUSH Recommended Medicinal Plants or Extracts against COVID-19
Ayurveda Medicines
Unani Medicinal Plants for the Prevention and Management of COVID-19
Telemedicine and COVID-19
Types of Telemedicine
1-Communication Time
2- Communication mode
3-Persons engaged in teleconsultation
4-Consultation purpose
Telemedicine During the Previous Outbreak of Diseases
Telemedicine During the Pandemic of COVID-19
FUTURE PROSPECTIVE
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Role of Blood Plasma Transfusion Against COVID-19
Abstract
INTRODUCTION
Therapeutic Agents (Convalescent Plasma and Immune Globulin)
Safety and Efficacy of CP for COVID-19 patients
Pathways Use for Investigational COVID-19 CP
Collection of COVID-19 Plasma and Eligibility to Donate Blood Plasma (Precautionary Measures)
Eligibility of Donors
Pre-screening of Donors
Antibody Screening
Blood Collection and Testing
Dosing and Transfusion
Patient Eligibility
Labeling and Recordkeeping
Adverse Effects of CP Therapy
Optimal Dose and Clinical Benefits of CP Therapy
FUTURE PERSPECTIVE
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Stem Cell Therapy for COVID-19
Abstract
INTRODUCTION
Overview of Stem Cells
Different Stem Cells
Stem Cells and Immune System Responses During COVID-19
Mesenchymal Stem Cells in COVID-19
Synthetic Stem Cells for COVID-19
Stem Cells in Clinical and Preclinical Research Stage
Serum Cytokine/Chemokine/Growth factors Up- and Down-Regulation During Stem Cell Therapy
Role of ACE2-Mesenchymal Stem Cells for COVID-19
Clinical Outcomes of Stem Cells for COVID-19
FDA/WHO Approved Stem Cells Therapy for COVID-19
FUTURE PERSPECTIVE OF HSC FOR COVID-19
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Computational Methods for COVID-19 Treatment
Abstract
INTRODUCTION
Integrated Pipelines of COVID-19
Virtual Screening of COVID-19
Drug Libraries for COVID-19
Drug Repurposing Approach for COVID-19
Molecular Databases of COVID-19
Genomic Information of SARS-CoV-2 (COVID-19)
Homology Modeling and Molecular Docking of COVID-19
Molecular Docking
Protein Modeling
Fold and Function Assignment System
Protein Data Bank
Pharmacophore Modeling
Pharmacophore Fingerprinting
Pharmacophore Model or Query
Modeling Softwares
Monte Carlo Method
The Stochastic Global Optimization Procedure
Pseudo-Brownian Positional/Torsional
Protein-protein Docking
Drug-target Interaction
Molecular Dynamic Simulation
Binding Affinity
Binding Free Energy Calculation
Method Validation
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Vaccine Development, Advantages, and Disadvantages of Group Immunity and Future Perspective for COVID-19
Abstract
INTRODUCTION
Clinical-Phase Vaccine Candidates for COVID-19
Diversity of Technology
Nucleic Acid (DNA and RNA)
Virus-like Particle
Peptide
Live Attenuated Virus
Viral Vector (replicating and non-replicating
Recombinant Protein
Monoclonal Antibodies (mAbs)
Inactivated Virus
Clinical Trials in Terms of Vaccine Development
Preclinical Research
Non-Specific Vaccine
Group Immunity
Herd Immunity and COVID-19
Advantages and Disadvantages of Herd Immunity
FUTURE PERSPECTIVE OF VACCINE DEVELOPMENT
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENT
REFERENCES
Coronavirus Disease-19 (COVID-19): A Perspective of New Scenario
(Volume 2)
Coronavirus Disease-19 (COVID-19):
Different Models and Treatment Strategies
Edited by
Kamal Niaz
Department of Pharmacology and Toxicology
Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences
Bahawalpur-63100
Pakistan
&
Muhammad Farrukh Nisar
Department of Physiology and Biochemistry
Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences (CUVAS)
Bahawalpur-63100
Pakistan

BENTHAM SCIENCE PUBLISHERS LTD.

End User License Agreement (for non-institutional, personal use)

This is an agreement between you and Bentham Science Publishers Ltd. Please read this License Agreement carefully before using the ebook/echapter/ejournal (“Work”). Your use of the Work constitutes your agreement to the terms and conditions set forth in this License Agreement. If you do not agree to these terms and conditions then you should not use the Work.

Bentham Science Publishers agrees to grant you a non-exclusive, non-transferable limited license to use the Work subject to and in accordance with the following terms and conditions. This License Agreement is for non-library, personal use only. For a library / institutional / multi user license in respect of the Work, please contact: [email protected].

Usage Rules:

All rights reserved: The Work is 1. the subject of copyright and Bentham Science Publishers either owns the Work (and the copyright in it) or is licensed to distribute the Work. You shall not copy, reproduce, modify, remove, delete, augment, add to, publish, transmit, sell, resell, create derivative works from, or in any way exploit the Work or make the Work available for others to do any of the same, in any form or by any means, in whole or in part, in each case without the prior written permission of Bentham Science Publishers, unless stated otherwise in this License Agreement.You may download a copy of the Work on one occasion to one personal computer (including tablet, laptop, desktop, or other such devices). You may make one back-up copy of the Work to avoid losing it.The unauthorised use or distribution of copyrighted or other proprietary content is illegal and could subject you to liability for substantial money damages. You will be liable for any damage resulting from your misuse of the Work or any violation of this License Agreement, including any infringement by you of copyrights or proprietary rights.

Disclaimer:

Bentham Science Publishers does not guarantee that the information in the Work is error-free, or warrant that it will meet your requirements or that access to the Work will be uninterrupted or error-free. The Work is provided "as is" without warranty of any kind, either express or implied or statutory, including, without limitation, implied warranties of merchantability and fitness for a particular purpose. The entire risk as to the results and performance of the Work is assumed by you. No responsibility is assumed by Bentham Science Publishers, its staff, editors and/or authors for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products instruction, advertisements or ideas contained in the Work.

Limitation of Liability:

In no event will Bentham Science Publishers, its staff, editors and/or authors, be liable for any damages, including, without limitation, special, incidental and/or consequential damages and/or damages for lost data and/or profits arising out of (whether directly or indirectly) the use or inability to use the Work. The entire liability of Bentham Science Publishers shall be limited to the amount actually paid by you for the Work.

General:

Any dispute or claim arising out of or in connection with this License Agreement or the Work (including non-contractual disputes or claims) will be governed by and construed in accordance with the laws of the U.A.E. as applied in the Emirate of Dubai. Each party agrees that the courts of the Emirate of Dubai shall have exclusive jurisdiction to settle any dispute or claim arising out of or in connection with this License Agreement or the Work (including non-contractual disputes or claims).Your rights under this License Agreement will automatically terminate without notice and without the need for a court order if at any point you breach any terms of this License Agreement. In no event will any delay or failure by Bentham Science Publishers in enforcing your compliance with this License Agreement constitute a waiver of any of its rights.You acknowledge that you have read this License Agreement, and agree to be bound by its terms and conditions. To the extent that any other terms and conditions presented on any website of Bentham Science Publishers conflict with, or are inconsistent with, the terms and conditions set out in this License Agreement, you acknowledge that the terms and conditions set out in this License Agreement shall prevail.

Bentham Science Publishers Ltd. Executive Suite Y - 2 PO Box 7917, Saif Zone Sharjah, U.A.E. Email: [email protected]

FOREWORD

According to World Health Organization, 122 million cases and over 2.69 million deaths of coronavirus disease-2019 (COVID-19) pandemic have impacted nearly every region of the globe. The pandemic has a huge global economic influence, and successfully fighting against COVID-19 needs efforts. Newly emerging respiratory diseases such as Severe Acute Respiratory Syndrome-coronavirus (SARS-CoV), Middle East Respiratory Syndrome (MERS), and severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) posing a serious threat to the human population and reported in the years 2003, 2012, and 2019, respectively. Furthermore, SARS-CoV-2 new strains shocked the world due to genetic mutation over time.

This book is very relevant and consists of two key modules. The first module provides clear information regarding different animals and cell line models and differentiation from other respiratory diseases. The second part focuses on therapeutic agents (antivirals, natural compounds, ultraviolet radiation, herbal remedies, blood plasma, stem cell therapy, and vaccines. It will give step-by-step awareness to the scholars about SARS-CoV-2. Basic and advanced knowledge about the disease is arranged into clear and easy-to-read chapters for general readers, scholars, and molecular biology teachers, making this book a valuable and thorough guide for all.

Prof. Dr. Zahir Shah University of Chitral Chitral Khyber Pakhtunkhwa Pakistan

PREFACE

Coronavirus disease-19 (COVID-19) is a complex disease that causes illnesses ranging from mild to severe respiratory problems. It is caused by a novel coronavirus severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), an enveloped, positive-sense, and single-stranded RNA (+ssRNA) virus that belongs to the coronavirus family. It has a fast-spreading potential worldwide, which leads to high death cases regardless of lows death rates. Yet, there are no animal models or specific drugs for disease prevention and/or treatment. Therefore, it is highly demanded to identify the known drugs and test them as a possible therapeutic approach. In this critical situation, one or more of these drugs may represent the only option to treat or reduce the severity of the disease until a specific drug or vaccine is developed and approved. A wide variety of therapeutics have been explored to treat COVID-19, initially suggested for other diseases and already established safety profiles, and approved by the Food and Drug Administration (FDA). Such treatments are referred to by the World Health Organization (WHO) as repurpose medications. Still, there are many ongoing clinical trials regarding the safety and effectiveness of repurposing immune-therapeutics to mitigate the symptoms of COVID-19.

In this book volume-2 proposal, we consolidate the various animal models and treatment strategies widely used for the global emergency of COVID-19. Since SARS-CoV-2 is the closest to SARS-CoV and MERS-CoV, the approaches brought here will be similar and/or varying with a slight degree. It is cleared that in the last 17-18 years, this is the third outbreak of the same coronavirus with a small mutation that shock the whole world. The chapters in this book should be prioritized as up-to-date literature of techniques used in the study SARS-CoV-2 and will act as a suitable reference if any such wary appear soon.

The 2nd volume of the proposed book proposal has been classified into Part IV: Models for SARS-CoV-2 and Part V: Treatment Strategies for SARS-CoV-2. With the emergence of new coronavirus variants, epidemiology, different host tropism permits a thorough analysis of their evolution and acquired adaptability to their host. The 1st volume already discussed the entry, epidemiology, genetic alteration, and diagnostic approaches. In the 2nd volume, part IV, we have planned to describe chapter-wise models used in COVID-19. No studies are complete without animal models closely related to human physiology to replicate the disease and observe the pathology conditions as in human cases. Such animal models play a vital role in virus pathogenesis and prepare a therapeutic immune response. Here describe bio-engineered transgenic mouse model inserting with specific genes, or CRISPR-Case9 gene-editing tool has been used previously for SARS-CoV and MERS-CoV. The chapter will deal with culture techniques or cell lines for COVID-19-also histopathology of COVID-19, essential proteins that up or down-regulate SARS-CoV-2. The last chapter of this part will describe other diseases having similar signs and symptoms and their differentiation. In the last part of the book proposal, part V, chapters will deal with therapeutic approaches to attenuate SARS-CoV-2 as there is no specific treatment available to date, just symptomatic therapy. However, scientists will elucidate effective antiviral drugs in clinical trials, phytochemicals, photomedicine such as ultraviolet A & B, homemade remedies, blood plasma transfusion, stem cell therapy, and computational approaches in vivo and in vitro trials.

This book will appear as a baseline for academicians, scientists, and health professionals as still, research is going to overcome this outbreak of COVID-19, the novelty of best animal models, and find an effective treatment. However, just a single book proposal like this wouldn't have flourished without enthusiasm and determined publishers' and investigators' strength to take time from their busy schedule and subsidize on time.

We thank the whole investigators who contributed, directly and indirectly, to bring it to reality.

Kamal Niaz Department of Pharmacology and Toxicology Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences Bahawalpur-63100 Pakistan &Muhammad Farrukh Nisar Department of Physiology and Biochemistry Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences (CUVAS) Bahawalpur-63100 Pakistan

List of Contributors

AbdullahDepartment of Pharmacy, University of Malakand, Chakdara Dir Lower, KPK, PakistanAhmad FarooqDepartment of Biological Sciences, International Islamic University, Islamabad, PakistanAadil JavedDepartment of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Bornova, Izmir, TurkeyAbhinav AnandSchool of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, PIN- 144 411, India CT Institute of Pharmaceutical Sciences, CT Group of Institutions, Shahpur, Jalandhar, Punjab, PIN- 144 020, IndiaAmjad Islam AqibDepartment of Medicine, Faculty of Veterinary Science, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, PakistanAmna AhmadFaculty of Veterinary Science, University of Agriculture, Faisalabad, PakistanAhmed Abdelmajed AlkhodaryMinistry of Health, Gaza Governorates, Palestine International Centre for Casemix and Clinical Coding, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, MalaysiaAfshan ShafiDepartment of Food Science and Technology, MNS University of Agriculture, Multan, PakistanAhmad UddinDrug Discovery Research Centre, Southwest Medical University, Luzhou, ChinaDevesh TewariSchool of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, PIN- 144 411, IndiaFaisal SiddiqueDepartment of Microbiology, Faculty of Veterinary Science, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, PakistanFirasat HussainDepartment of Microbiology, Faculty of Veterinary Science, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, PakistanFazlullah KhanDepartment of Allied Health Sciences, Bashir Institute of Health Sciences, Bharakahu, Islamabad, PakistanIhtisham UlhaqDepartment of Biosciences, COMSATS University Islamabad (CUI), 45550, PakistanIqra MuzammilDepartment of Clinical Medicine and Surgery, University of Agriculture, Faisalabad, PakistanIsmail ShahDepartment of Pharmacy, Garden Campus, Abdul Wali Khan University, Mardan, PakistanIyad Ibrahim ShaquraDepartment of Health Management and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran Ministry of Health, Gaza Governorates, PalestineKamal NiazDepartment of Pharmacology & Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur-63100, PakistanKashif AkramDepartment of Food Sciences, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, PakistanKashif RahimDepartment of Microbiology, Faculty of Veterinary Science, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, PakistanLaiba HassanDepartment of Microbiology, The University of Haripur, Haripur, PakistanMaqsood ur RehmanDepartment of Pharmacy University of Malakand, Chakdara Dir Lower, Cholistan University of Veterinary & Animal Sciences, PakistanMuhammad Mazhar MunirDepartment of Pharmacology & Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary & Animal Sciences, Bahawalpur, PakistanMuhammad Mazhar AyazDepartment of Parasitology, Faculty of Veterinary Science, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, PakistanMuhammad IbrarDepartment of Allied Health Sciences, Iqra University Peshawar, Swat Campus, KPK, PakistanMuhammad Farrukh NisarDepartment of Physiology and Biochemistry, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, PakistanMuhammad Yasir WaqasDepartment of Physiology and Biochemistry, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, PakistanMuhammad Ali SyedDepartment of Microbiology, The University of Haripur, Haripur, PakistanMohammad EjazDepartment of Microbiology, The University of Haripur, Haripur, PakistanMuhammad Ali SyedDepartment of Microbiology, The University of Haripur, Haripur, PakistanNavneet KhuranaSchool of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, PIN- 144 411, IndiaNeha SharmaSchool of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, PIN- 144 411, IndiaOvais SideeqSchool of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, IranQaisar TanveerInstitute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, PakistanQudratullulahDepartment of Surgery, Faculty of Veterinary Science, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, PakistanSana MuzammalDepartment of Mathematics, National College of Business Administration and Economics, Sub-Campus Bahawalpur, PakistanShaukat Hussain MunawarDepartment of Pharmacology and Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, PakistanSonali BajajSchool of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, PIN- 144 411, IndiaTapan BehlChitkara College of Pharmacy, Chitkara University, Punjab, IndiaTean ZaheerDepartment of Parasitology, University of Agriculture, Faisalabad, PakistanZahid ManzoorDepartment of Pharmacology & Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary & Animal Sciences, Bahawalpur, Pakistan

Part I: Models for COVID-19

Genetically Engineered Mouse Models for COVID-19

Aadil Javed1,*,Kamal Niaz2,Ovais Sideeq3
1 Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Bornova, Izmir, Turkey
2 Department of Pharmacology & Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur-63100, Pakistan
3 School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran

Abstract

Previous severe acute respiratory syndrome-coronavirus (SARS-CoV) outbreaks resulted in a cohort of preclinical studies that utilized various mice models for determining the pathogenesis of the infection, including the viral replication, spread, and mortality of the disease. Such studies have provided a framework upon which new investigations have been launched for understanding the outbreak of new coronavirus disease-19 (COVID-19) causing viral agents and their interaction with the host and its body. Recent investigations showed that the previous SARS-CoV and the recently discovered severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) both require the spike S protein to enter the host cell upon infection the binding with the receptors on the surface of the cells. The viral entry also requires proteases from the host cells. Since there are key similarities between the structure of the viruses and the construct of the viral transmission along with the spread inside the host's body in animal models. They were developed for the previous viral agent. The disease can be emulated or manipulated to bring forth novel investigations leading to key data that can broaden the sphere of COVID-19 studies being conducted. There are several different options to choose the right animal model for the question being raised in the experimental design with the pathogenesis of COVID-19. This chapter focused on the already established animal models for other coronavirus outbreaks and some of the strategies that can be exploited to develop new animal models. For COVID-19, research aimed at targeting the therapy or basic investigations for understanding cellular or organ level mechanisms involved in the disease.

Keywords: COVID-19, Knockout, Mouse models, SARS-CoV-2, Transgenic.
*Corresponding author Aadil Javed: Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Bornova, Izmir, Turkey. E-mail: [email protected]

INTRODUCTION

In December 2019, a novel coronavirus outbreak was reported in Wuhan, China which caused a pandemic and is an ongoing public health concern worldwide [1]. The disease known as coronavirus disease-19 (COVID-19) is caused by the novel severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), which leads to deadly pneumonia and severe health issues related to lung damage [2]. The genomic properties of the viral agent responsible for the local epidemic in China were timely shared across the scientific community. Since then, a valiant effort to combat the disease and understanding the agent responsible for the pandemic has been ongoing. Across the world, millions of people have been affected by the pandemic leading to a change in lifestyle globally [3-5]. The diverse scientific community took upon understanding the pandemic and the viral agent responsible for the COVID-19 from the get-go during the earlier days of the pandemic in 2020 [6]. Millions of people have been infected, and hundreds of thousands of people have lost their lives due to the COVID-19 pandemic outbreak [7]. Various attempts have been reported to classify the epidemiological features of the pandemic, including the case fatality rate, reproduction number, and recovery rate. To date, it is being assumed that various factors that affect the rigorous testing and other socio-economic factors hamper any progress in determining the true features of the pandemic [8-13].

The SARS-CoV-2 virus belonging to the Coronaviridae family of the virus has been studied extensively due to the urgency of pandemics. The vaccine development approaches have been challenging so far, and various mouse models have been utilized for understanding different aspects of the physiology of this novel coronavirus. Previously, the Middle East Respiratory Syndrome (MERS) and SARS candidates for vaccines were evaluated in the different mouse models [14, 15]. The clinical features, including the pathogenesis of the coronavirus, can be studied in great detail upon the development of animal models that can efficiently mimic the properties of the disease. For this purpose, numerous models have been developed in animals such as hamsters, guinea pigs, mice, cats, and rabbits, etc., [16-20]. The major receptor of SAR-CoV is angiotensin-converting enzyme 2 (ACE2) and is utilized to develop transgenic mouse models containing the human ACE2 gene [21]. Rhesus macaques were the first animal model for vaccine development against MERS-CoV, showing the symptoms of the infections as seen in clinical patients [22]. Golden Syrian hamsters were also used as animal models for establishing the vaccine candidate's safety and viral pathogenesis for different strains of SARS-CoV [19]. NSP16 CoV attenuated vaccine development approaches utilized mice as well [23]. Developing animal models, MERS-CoV had several drawbacks due to the inefficiency of the virus being replicated in the respiratory system. Therefore, new approaches like gene targeting paved the way for demonstrating modified models containing genes of interest or lack thereof for more naturalized infectivity of the virus, e.g., human DPP4 transgenic mice [24]. Clustered regularly interspaced short palindromic repeats and associated protein 9 (CRISPR-Cas9) the system has been efficiently employed for developing mouse models that are engineered to get infected by the virus and show high replication [25, 26]. Small animals like rabbits and mice are used to develop mouse models specific for a particular infection such as SAR-CoV-2 for various reasons, including efficacy, cost issues, and manipulation approaches. There is still much work that is needed to better understand the behavior of COVID-19 and transgenic animal models can efficiently smoothen the process of understanding due to ease in receptor identification, protection, pathogenesis models and immune response studies [15, 27]. Various applications and interference points for different types of mouse models in understanding or combating SARS have been depicted in Fig. (1). The aim of this chapter is to summarize the well-known animal models that can be used for coronavirus along with some of the strategies that can be exploited to develop new animal models for COVID-19 as a model to target the therapy or basic investigations the underline molecular mechanisms involved in the disease.

Emerging Role of CRISPR/Cas9

In molecular diagnostics, CRISPR, and the Cas protein-containing CRISPR-Cas systems, have advanced the process of functional research. Since its discovery almost 30 years ago, the field of genome editing using CRISPR systems has been revolutionized [28]. In clinical sciences and modern molecular biology approaches, CRISPR-Cas9 systems are routinely applied for targeting the cells of mammalian origin involving gene editing methodology [29]. Such approaches have been utilized to detect Zika virus and methicillin-resistant Staphylococcus aureus along with nucleic acid detection-based diagnosis procedures that use RNA-guided or RNA-targeting CRISPR-Cas systems [30-32]. In the wake of the COVID-19 pandemic of 2020, CRISPR-Cas13 approached for diagnostic applications are being considered [33]. The potential for this technology is deemed as tremendous, as described by Xiang et al. [34]. A fast and accurate method for detecting SARS-CoV-2 is the need for the hour, and SHERLOCK protocols can provide the scientific community an approach that has huge potential in this area. A recently reported DETECTR assay for the detection of SARS-CoV-2 via employing CRISPR leads to a 95% positive prediction along with a 100% negative prediction agreement in a real-time RT-PCR [35]. Genome-wide CRISPR screens were employed to determine the therapeutic targets for COVID-19. Apart from already established protease cathepsin and receptor ACE2, SWI/SNF chromatin complex proteins that comprise the transforming growth factor-beta (TGF-β) signaling network were also identified using the technology [36]. The protocol for designing a transgenic mouse model for SARS-CoV-2 that conforms to the CRISPR-Cas9 technology has also been reported recently [37]. Such models can successfully provide representativeness of the infection with tissue specificity of the human genes as well [37].

Fig. (1)) Experimental animals of SARS-CoV, MERS-CoV, and SARS-CoV-2. The coronaviruses with high infectivity and pathogenicity break the species barrier and infect humans in the past two decades. Besides NHP, mice, hamsters, ferrets, and rabbits, the other possible natural hosts might be able to support the studies of coronavirus infection, pathogenesis, and drug discovery.

Human ACE2 Expression

A carboxypeptidase named ACE2, which is encoded by the gene ACE2 resides on chromosome Xp22. It gives rise to a type I type transmembrane protein containing an extracellular N-terminal domain with carboxypeptidase which is heavy, and a short C-terminal domain having a cytoplasmic tail intracellularly [38, 39]. The SARS-CoV binding site in ACE2 is present on the N-terminal peptidase domain [40]. Cellular ACE2 manifests in either soluble or membrane-bound form and is usually expressed in pneumocytes and enterocytes located in the small intestine [41]. Vascular endothelial cells located in the heart, brain, and kidneys also exhibit ACE2 expression [42, 43]. The major site of infection for the coronavirus, especially SARS-CoV, is the respiratory tract. In human airway epithelia and lung parenchyma, ACE2 is expressed and is considered the receptor for these coronaviruses. SARS-CoV also seemed to infect the differentiated epithelial cells more than undifferentiated cells as the differentiated epithelia are reported to have higher expression of ACE2 receptor via SARS S protein [44].

ACE2 gene in humans produces the protein receptor that is identified by SARS coronaviruses, including SARS-CoV and NL63, along with the novel SARS-CoV-2 virus that caused the pandemic of 2020 [45]. In vitro studies suggest that SARS-CoV infection correlates with the expression of the ACE2 receptor. The variations or mutations in the ACE2 sequence lead to a reduced association or linkage between the spike protein on the SARS-CoV or NL63 with the ACE2 receptor [40, 46]. It has been concluded that the severity, susceptibility, or even the symptoms of SARS-CoV-2 infection is dependent on the expression of the receptor ACE2. Recently, ACE2 expression levels from single-cell RNA-sequencing experiments concluded that Asian males show higher expression of the receptor [47]. However, more studies need to be conducted to profile a linkage with ethnicities based on ACE2 expression and COVID-19 epidemiological features. The overall function of ACE2 in different populations and the genetic background correlations, are yet to be understood in detail [48]. A systematic analysis of variants of ACE2 (coding region variants) that can influence the expression pattern has been performed. The genetic analysis from microarray data showed that the East Asian populations have a higher allelic frequency of different eQTL variants associated with higher expression of ACE2. More studies on the pilot data need to be performed in other populations to report concrete evidence related to receptor and virus spike protein [49].

ACE2 Knockout Mouse for Acute respiratory Distress Syndrome (ARDS) Investigation

Major causes of death resulting from SARS include acute lung injury along with ARDS. ACE2 knockout mice are reported to be protected from infections by SARS [50]. Moreover, studies showing data from knockout ACE2 receptors have depicted that the spike protein of the virus can cause the downregulation of the ACE2 expression and ACE2 being involved in the protection of lungs from acute injury of the lungs [50, 51]. The introduction of recombinant ACE2 protein can reduce ARDS in mice due to the angiotensin and renin pathway [51]. Moreover, knockout ACE2 mice have been reported to be more prone to impaired lung functionality and exacerbated fibrosis of the lung along with reduced exercise capacity when compared with wild-type mice [52]. In another investigation, knockout ACE2 mice depicted an increased level of lung edema, increased vascular permeability and demonstrated severe lung disease [51].

On the one hand, knockout ACE2 shows severe lung disease, the introduction of recombinant ACE leads to elevated symptoms related to ARDS [53]. It can be deduced from the studies on knockout ACE2 models that ACE2 is a key factor involved in the pathogenesis of SARS viruses and could be the most promising target for therapy that does not include the immunization procedures that can target ARDS. Based on the data obtained through knockout studies, recombinant human ACE2 studies showed promising results in improving the pulmonary circulatory hemodynamics and arterial hypoxemia associated with ARDS [54]. ACE2 knockout mice are known to be resistant to SARS infection as the viral titers determined from the knockout model were a hundred thousand folds lower than the wild-type mice lungs [50].

K18-hACE2 Transgenic Mouse

Coronaviruses, especially those that cause SARS, infect many animal species, including ferrets, mice, cynomolgus, hamsters, and rhesus macaques, among others [55]. Apart from different types of coronaviruses available for studies that demonstrate the infectivity comparable to human coronaviruses. The clinical disease that arises from infection by the SARS-CoV is difficult to replicate in animals in terms of clinical signs and severity as observed in the patients. To determine the vaccine efficacy along with the overall pathogenesis of SARS-CoV, a reproducible and equivalent animal model is the need of the hour that can mimic the immune response in the host as well. Transgenic models are preferred to overcome this drawback, e.g., hACE2 transgenic mice, which show overexpression of the receptor ACE against the SARS-CoV [56]. Since mACE2 does not bind as efficiently with the virus as hACE2. Therefore hACE2 is the specific target for generating transgenic mice. Moreover, the expression of ACE2 being higher in transgenic hACE2 mice also result in a severe disease upon infection with the SARS-CoV in the epithelia of mice [57]. pK18-hACE2 transgenic mice were generated by cloning into pCR2.1-TOPO vector the amplified hACE2 coding sequence from IMAGE consortium clone ID 5243048. The final construct for generating transgenic mice contained a promoter and intron for human cytokeratin 18 (K18 gene) and the enhancer sequence obtained from the alfalfa mosaic virus upstream to the hACE2 coding sequence. Besides, 2 exons and 1 intron of K18 along with polyA signal tail were included for bringing higher specificity towards the epithelia in terms of selective translation in the downstream sequence [57, 58]. For the final generation of the mice, the transgene was injected into pronuclei of the fertilized mice eggs (C57BL/6J) for embryo generation and validated by PCR specific for transgene primers. In one of the recent reports, the SARS-CoV-2 viral agent responsible for the COVID-19 pandemic (one of the earlier isolates) was infected in transgenic hACE2 mice for determining the pathogenicity of the novel SARS-CoV-2 in hACE2 mice [59]. As a result of this experimental infection, the mice lost weight and demonstrated interstitial pneumonia, known as the clinical signs of COVID-19 [59, 60]. Specific immunoglobulins (IgG) targeting SARS-CoV-2 were also observed in the lungs of transgenic hACE2 mice along with virus replication. More studies are needed to determine the immune response against the infection in these animals.

TMPRSS2 Knockout Mouse for Viral Entry and Pathogenesis Studies

Coronavirus fusion glycoproteins which are also known as spike proteins, are cleaved by transmembrane protease serine-type 2 (TMPSS2) belongs to a family of transmembrane serine protease type II. Thus activates the spike protein resulting in the fusion of the cell-virus membrane to facilitate the next step of infection further; the entry of the virus in the host cell [61-63]. There are reports which suggest that TMPRSS2 knockout mice are resistant to severe outcomes from infection by the influenza virus. At the same time, higher expression of this gene leads to severe outcomes after infection with the influenza virus [64, 65]. Mice deficient in TMPRSS2 are reported to have shown a reduced loss of weight and replication of viral agents in the lungs. It spreads the virus in the airways of the animals when infected with SARS-CoV and MERS-CoV [61]. The viral entry of coronaviruses and influenza viruses depends critically on TMPRSS2 as the viral protein interacts and binds with ACE2 localized in the epithelia. Viral protein is cleaved by special proteases such as TMPRSS2 to activate the process of internalization of the virus, leading to the next phases of the infection [66]. SARS-CoV-2 and other coronaviruses and influenza viruses require TMPRSS2 to activate the virus and its entry into the host cell for carrying the infection [62, 67, 68]. Therefore, TMPRSS2 can be placed at the center of the pathogenesis of COVID-19 and other pandemics of the past [69].

Since it has been established that TMPRSS2 is vital for entry of SARS-CoV-2 and involved in the dysregulation of ACE2. Therefore, it has been hypothesized by various accounts that it can play a vital role in preventing the critical circumstances of COVID-19 disease if used in a well-defined elaborated therapeutic strategy [70]. TMPRSS2 mediated entry of the SARS-CoV-2 can be inhibited by the serine protease inhibitor known as camostat mesylate [71, 72]. The envelope and plasma membrane fusion between SARS-CoV-2 mediated by TMPRSS2 and ACE2 can also be inhibited by nafamostat mesylate showing 10-times more efficacy than the camostat mesylate [73]. TMPRSS2 is now considered one of the most promising candidates as a therapeutic target for COVID-19. TMPRSS2 knockout mice model study indicated that the influenza virus entry into the cell is inhibited compared to wild-type mice, further implying the role of TMPRSS2 in regulating the pathogenesis of viruses as the crucial step of viral entry inhibition can lead to reduced mortality [74]. TMPRSS2 knockout mouse model can be generated in C57BL/6 Embryonic Stem Cells (ESCs) usually obtained from KOMP (knockout Mouse Project) repository. It is followed by injection of ESCs into blastocyst for generating chimeric mice, followed by male and female mice for creating homologous genotype [61, 75]. The viral kinetics observed in the lungs of TMPRSS2 knockout mice upon infection by SARS-CoV were reduced along with a reduced weight loss upon infection. Moreover, the inflammatory cytokine and chemokine response usually associated with such infections were also observed to be weakened in TMPRSS2 knockout mice [61]. For SARS-CoV-2 experimental infections in mice models, more studies need to be performed to understand the underlying mechanisms of the inflammation-mediated responses and immune responses generated via infections that lead to severe clinical circumstances of COVID-19 disease.

STAT1 Knockout Mouse for Studying Pneumonia and Antiviral Strategies

Signal transducer and activator of transcription 1 (STAT1) is considered one of the STAT family members of proteins involved in the signaling responses of the innate immune system. The signaling works by cytokines or growth factors that elicit response inside the cells, leading to phosphorylation of these proteins by the kinases associated with certain receptors, further causing the translocation of these proteins to the nucleus. One of the most important cytokine-mediated signalings in immune responses caused by viruses is interferon receptor signaling, which requires STAT1 responses [76]. The severe cases of COVID-19 show a cytokine storm phenotype, which is considered to be caused by SARS-CoV-2 nsp3 due to the de-mono-ADP-ribosylation of STAT1. It further highlights the importance of STAT1 as a therapeutic target in the precarious times of the COVID-19 pandemic [77]. A group of cytokines known as type 1 interferons (IFN-1) having further subtypes is known to be secreted by different cells like dendritic cells when pattern recognition receptors (PRRs) recognize the viral antigens or components [78]. IFN-1 is considered the first type of cytokine produced and released upon infection by a viral agent.

Interferon gets fixated upon interferon-α/β receptor (IFNAR), transcription factors including STAT1 are phosphorylated, leading to their translocation to the nucleus, further causing activation interferon-stimulated genes (ISGs). ISGs are known for the modulation of immune responses and signaling related to inflammatory pathways. After activation, ISGs lead to the further secretion of cytokines promoting adaptive immunity and interference with viral replication or spread and slowing down the cell's metabolism [79, 80]. Previously available IFN-1 treatment data on SARS-CoV and MERS-CoV can be valuable in determining the SARS-CoV-2 related mechanisms of pathogenesis and immune responses, as both of these viruses led to the disruption of the interferon signaling pathway. SARS-CoV Orf6 protein has been reported to inhibit the STAT1 transport to the nucleus upon interferon response by disrupting karyopherin transport [81, 82]. Therefore, STAT1 knockout models are important in understanding the role of viral proteins and their subsequent action in determining the immune response resulting from the infection [83]. The innate immune response that leads to clearance of the SARS-CoV also requires STAT1 activity, which modulates the IFN signaling [84]. IFN deficient mice showed that these components of innate immune responses are required STAT1 deficiency to prolong the viral shedding or susceptibility, allowing for lethal infection outcomes. STAT1 deficient mice data revealed the mechanistic role of these proteins in regulating SARS-CoV pathogenesis in the lungs.

Moreover, the pathogenesis of SARS-CoV seemed to be dependent on STAT1 and not on IFN subtypes, highlighting the role of STAT1 in controlling SARS especially viral replication, wound repairing, or cell proliferation in the lethal type disease [85]. Profibrotic phenotype resulting from the lethal outcome of SARS-CoV in the lungs of STAT1 knockout mice was also reported [86]. Therefore, the role of STAT1 in clearing the SARS-CoV-2 infection cannot be avoided and can be used in determining the therapeutic strategies for lethal conditions of COVID-19 [87]. Moreover, the role of STAT1 knockout, as highlighted in studies, demonstrated the role of an innate immune response about interferon signaling, can be utilized for potential therapeutic approaches for COVID-19.

Standard Mouse Strains (BALB/c mice, C57BL/6, and 129S6)

The development of COVID-19 therapy approaches along with vaccine studies, and preclinical investigations can be performed in these mice as they are known to support the replication of viral agents (SARS-CoV and MERS). The standard mice used in laboratory research, like BALB/c mice, C57BL/6, and 129S6, demonstrate the SARS-CoV replication along with other mice models in conjunction with the experimental designs [55]. Mouse-adapted SARS-CoV-2 infection in BALB/c mice showed severe symptoms compared to young mice, showing the clinical similarities observed in human patients of COVID-19 [88, 89]. Moreover, acute lung injury models have also been developed using C57BL/6 mice, which may further understand the therapeutic agent selection against ARDS associated with the severity of COVID-19 [90]. Similarly, 126S6 mice have been used in viral studies investigating the immune responses elicited upon viral infection [91].

Inbred Mice for Pathogenesis and Therapeutic Response Studies

Mouse strains of inbred nature that are available widely can also be adapted in the experimental designs to understand various parameters associated with the pathogenesis and pathophysiology of COVID-19. For example, age-related mortality can be studied in more detail in mouse models that are infected with SARS-CoV-2 [19]. The pneumonitis and clinical symptoms of SARS have been mimicked in several inbred mouse species, including BALB/c, C57BL/6, and 129S [84, 92-94]. The main reason for inbred mice being animals of choice for COVID-19 disease is their cost-effectiveness, smaller size, larger numbers to include a valid statistical output, and ease of manipulation at the genetic level [95]. 129S6 strain of mice is observed to report SARS susceptibility higher than BALB/c or B6 mice because the infection led to mild interstitial pneumonitis 2 days post-infection [84].

Similarly, TNF-alpha, a known pro-inflammatory cytokine, was reported at higher levels in BALB/c mice when recorded 2-9 days post-infection [94]. Age-related severity of SARS has already been demonstrated in aged BALB/c mice, which is observed in previous SARS outbreaks as well [93]. As seen in COVID-19 patients, viral replication is observed in the respiratory tract, and BALB/c also shows the same pattern of viral replication upon infection with viral agents in experimental conditions, leading to respiratory distress. Other clinical signs of weight loss, edema, dehydration, and pneumonitis were also observed in such mice strains when infected with SARS-CoV [96].

Humanized Immune System Mice for Vaccine Approaches

COVID-19 has overwhelmed many healthcare systems and has impacted economies negatively since its spread globally. If a COVID-19 vaccine becomes available for public immunization, life globally can go back to normalcy. Many vaccine candidates for COVID-19 are under investigation, and some are in clinical trials [74, 97]. Preclinical studies to develop an approach against SARS-CoV-2 require animal models corresponding to the immune responses as elicited in humans against the viral agent [98]. The antigens presented to the Major Histocompatibility Complex (MHC), however, vary between different species. Therefore, the mice models that can carry human leukocyte antigen genes can mimic the human response to the epitope-based vaccines and assist in developing or discovering the vaccines for COVID-19 in animal models [99].

Moreover, the vaccine discovery process can become rapid if the humanized immune system containing mice model is used for this purpose [100]. They can enhance the discovery process by speedily eliciting the response for efficacy studies and the immune response or memory generated upon vaccinated and un-immunized mice [37, 101-103]. Human immune cell engrafted mice have also been instrumental in determining the immune response for a vaccine candidate and include a promising approach in combating COVID-19 [104, 105]. Immuno-deficient mice, when engrafted with peripheral blood mononuclear cells, exhibit another vaccine modeling approach [106].

CONCLUSION AND FUTURE PERSPECTIVES

Although there are many mice strains available that have assisted in understanding the pathogenesis of SARS and SARS-CoV previously, COVID-19 caused by SARS-CoV-2 presents a new challenge to the scientific community. Although mice and humans have shared genes, mice as in vivo replicas of COVID-19 or disease models require overcoming the challenges that are unique to the virus. The challenges include the model not possessing the receptor for the virus as human cells have mouse protein expression not being similar to human proteins. The binding affinity is different among viral and animal proteins. It also has a slightly distinct immune system among different species. Due to the aforementioned challenges, certain approaches have been utilized to overcome certain challenges, including transgenic mice representing the human version of certain aspects of the pathogenesis, knockout mice revealing the functions of intracellular programs required for the disease patterns, and inbred mice as age-related mortality models for disease, etc. In the end, mice models can also provide a great tool in discovering the therapeutic targets for the disease, which are not obvious at first but can be unearthed upon functional studies. Moreover, the information obtained through studies conducted in various types of mouse models consequently leads to the development phases of the vaccine or a drug target. Therefore, recently used immunized or immuno-deficient mice for investigating vaccine candidates or drug targets will ultimately lead to our understanding of rapidly concluding the different phases of discovery, therefore providing a gateway to quick response in times like COVID-19.

CONSENT FOR PUBLICATION

Not applicable.

CONFLICT OF INTEREST

The author confirms that this chapter contents have no conflict of interest.

ACKNOWLEDGEMENT

Declared none.

REFERENCES

[1]Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020; 395(10223): 497-506.[http://dx.doi.org/10.1016/S0140-6736(20)30183-5] [PMID: 31986264][2]Li Q, Guan X, Wu P, et al. Early transmission dynamics in Wuhan, China, of novel coronavirus-infected pneumonia. N Engl J Med 2020; 382(13): 1199-207.[http://dx.doi.org/10.1056/NEJMoa2001316] [PMID: 31995857][3]Saadat S, Rawtani D, Hussain CM. Environmental perspective of COVID-19. Sci Total Environ 2020; 728: 138870.[http://dx.doi.org/10.1016/j.scitotenv.2020.138870] [PMID: 32335408][4]Ross J. The exacerbation of burnout during COVID19: A major concern for nurse safety 2020. 35(4): 439–440.[http://dx.doi.org/10.1016/j.jopan.2020.04.001][5]Paraskevis D, Kostaki EG, Magiorkinis G, Panayiotakopoulos G, Sourvinos G, Tsiodras S. Full-genome evolutionary analysis of the novel corona virus (2019-nCoV) rejects the hypothesis of emergence as a result of a recent recombination event. Infect Genet Evol 2020; 79: 104212.[http://dx.doi.org/10.1016/j.meegid.2020.104212] [PMID: 32004758][6]Wu F, Zhao S, Yu B, et al. A new coronavirus associated with human respiratory disease in China. Nature 2020; 579(7798): 265-9.[http://dx.doi.org/10.1038/s41586-020-2008-3] [PMID: 32015508][7]World Health OrganizationWHO Coronavirus Disease (COVID-19) 2020.[8]Verity R, Okell LC, Dorigatti I, et al. Estimates of the severity of coronavirus disease 2019: a model-based analysis. Lancet Infect Dis 2020; 20(6): 669-77.[http://dx.doi.org/10.1016/S1473-3099(20)30243-7] [PMID: 32240634][9]Onder G, Rezza G, Brusaferro S. Case-fatality rate and characteristics of patients dying in relation to covid-19 in Italy. JAMA 2020; 323(18): 1775-6.[http://dx.doi.org/10.1001/jama.2020.4683] [PMID: 32203977][10]Rajgor DD, Lee MH, Archuleta S, Bagdasarian N, Quek SC. The many estimates of the COVID-19 case fatality rate. Lancet Infect Dis 2020; 20(7): 776-7.[http://dx.doi.org/10.1016/S1473-3099(20)30244-9] [PMID: 32224313][11]He W, Yi GY, Zhu Y. Estimation of the basic reproduction number, average incubation time, asymptomatic infection rate, and case fatality rate for COVID-19: Meta-analysis and sensitivity analysis. J Med Virol 2020; 92(11): 2543-50.[http://dx.doi.org/10.1002/jmv.26041] [PMID: 32470164][12]He F, Deng Y, Li W. Coronavirus disease 2019: what we know? J Med Virol 2020; 92(7): 719-25.[http://dx.doi.org/10.1002/jmv.25766] [PMID: 32170865][13]Wang Y, Wang Y, Chen Y, Qin Q. Unique epidemiological and clinical features of the emerging 2019 novel coronavirus pneumonia (COVID-19) implicate special control measures. J Med Virol 2020; 92(6): 568-76.[http://dx.doi.org/10.1002/jmv.25748] [PMID: 32134116][14]Gretebeck LM, Subbarao K. Animal models for SARS and MERS coronaviruses. Curr Opin Virol 2015; 13: 123-9.[http://dx.doi.org/10.1016/j.coviro.2015.06.009] [PMID: 26184451][15]Yong CY, Ong HK, Yeap SK, Ho KL, Tan WS. Recent advances in the vaccine development against middle east respiratory syndrome-coronavirus. Front Microbiol 2019; 10: 1781.[http://dx.doi.org/10.3389/fmicb.2019.01781] [PMID: 31428074][16]Bartlett JG. Virology: SARS virus infection of cats and ferrets. Infect Dis Clin Pract 2004.[http://dx.doi.org/10.1097/01.idc.0000129853.80250.2c][17]Roberts A, Wood J, Subbarao K, Ferguson M, Wood D, Cherian T. Animal models and antibody assays for evaluating candidate SARS vaccines: summary of a technical meeting 25-26 August 2005, London, UK. Vaccine 2006; 24(49-50): 7056-65.[http://dx.doi.org/10.1016/j.vaccine.2006.07.009] [PMID: 16930781][18]Lamirande EW, DeDiego ML, Roberts A, et al. A live attenuated severe acute respiratory syndrome coronavirus is immunogenic and efficacious in golden Syrian hamsters. J Virol 2008; 82(15): 7721-4.[http://dx.doi.org/10.1128/JVI.00304-08] [PMID: 18463152][19]Roberts A, Lamirande EW, Vogel L, et al. Animal models and vaccines for SARS-CoV infection. Virus Res 2008; 133(1): 20-32.[http://dx.doi.org/10.1016/j.virusres.2007.03.025] [PMID: 17499378][20]Zhao J, Li K, Wohlford-Lenane C, et al. Rapid generation of a mouse model for Middle East respiratory syndrome. Proc Natl Acad Sci USA 2014; 111(13): 4970-5.[http://dx.doi.org/10.1073/pnas.1323279111] [PMID: 24599590][21]Yang XH, Deng W, Tong Z, et al. Mice transgenic for human angiotensin-converting enzyme 2 provide a model for SARS coronavirus infection. Comp Med 2007; 57(5): 450-9.[PMID: 17974127][22]Munster VJ, de Wit E, Feldmann H. Pneumonia from human coronavirus in a macaque model. N Engl J Med 2013; 368(16): 1560-2.[http://dx.doi.org/10.1056/NEJMc1215691] [PMID: 23550601][23]Menachery VD, Gralinski LE, Mitchell HD, et al. Combination attenuation offers strategy for live attenuated coronavirus vaccines. J Virol 2018; 92(17): e00710-18.[http://dx.doi.org/10.1128/JVI.00710-18] [PMID: 29976657][24]Zhou Y, Jiang S, Du L. Prospects for a MERS-CoV spike vaccine. Expert Rev Vaccines 2018; 17(8): 677-86.[http://dx.doi.org/10.1080/14760584.2018.1506702] [PMID: 30058403][25]Cockrell AS, Yount BL, Scobey T, et al. A mouse model for MERS coronavirus-induced acute respiratory distress syndrome. Nat Microbiol 2016; 2: 16226.[http://dx.doi.org/10.1038/nmicrobiol.2016.226] [PMID: 27892925][26]Leist SR, Cockrell AS. Genetically engineering a susceptible mouse model for MERS-CoV-induced acute respiratory distress syndrome. Methods Mol Biol 2020; 2099: 137-59.[http://dx.doi.org/10.1007/978-1-0716-0211-9_12] [PMID: 31883094][27]Dhama K, Sharun K, Tiwari R. COVID-19, an emerging coronavirus infection: advances and prospects in designing and developing vaccines, immunotherapeutics, and therapeutics 2020; 16(6): 1232-38.[http://dx.doi.org/10.1080/21645515.2020.1735227][28]Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science 2014. 346(6213).[http://dx.doi.org/10.1126/science.1258096][29]Cong L, Ran FA, Cox D. Multiplex genome engineering using CRISPR/Cas systems. Science 2013. 339(6121): 819-23.[http://dx.doi.org/10.1126/science.1231143][30]Pardee K, Green AA, Takahashi MK, et al. Rapid, low-cost detection of zika virus using programmable biomolecular components. Cell 2016; 165(5): 1255-66.[http://dx.doi.org/10.1016/j.cell.2016.04.059] [PMID: 27160350][31]Guk K, Keem JO, Hwang SG, et al. A facile, rapid and sensitive detection of MRSA using a CRISPR-mediated DNA FISH method, antibody-like dCas9/sgRNA complex. Biosens Bioelectron 2017; 95: 67-71.[http://dx.doi.org/10.1016/j.bios.2017.04.016] [PMID: 28412663][32]Myhrvold C, Freije CA, Gootenberg JS. Field-deployable viral diagnostics using CRISPR-Cas13. Science 2018. 360(6387): 444-448.[http://dx.doi.org/10.1126/science.aas8836][33]Kellner MJ, Koob JG, Gootenberg JS, Abudayyeh OO, Zhang F. SHERLOCK: nucleic acid detection with CRISPR nucleases. Nat Protoc 2019; 14(10): 2986-3012.[http://dx.doi.org/10.1038/s41596-019-0210-2] [PMID: 31548639][34]Xiang X, Qian K, Zhang Z, et al. CRISPR-cas systems based molecular diagnostic tool for infectious diseases and emerging 2019 novel coronavirus (COVID-19) pneumonia. J Drug Target 2020; 28(7-8): 727-31.[http://dx.doi.org/10.1080/1061186X.2020.1769637] [PMID: 32401064][35]Broughton JP, Deng X, Yu G, et al. CRISPR-Cas12-based detection of SARS-CoV-2. Nat Biotechnol 2020; 38(7): 870-4.[http://dx.doi.org/10.1038/s41587-020-0513-4] [PMID: 32300245][36]Wei J, Alfajaro M, Hanna R. Genome-wide CRISPR screen reveals host genes that regulate SARS-CoV-2 infection BioRxiv 2020.[http://dx.doi.org/10.1101/2020.06.16.155101][37]Soldatov VO, Kubekina MV, Silaeva YY, Bruter AV, Deykin AV. On the way from SARS-CoV-sensitive mice to murine COVID-19 model 2020. 6(2): 1-7[http://dx.doi.org/10.3897/rrpharmacology.6.53633][38]Tipnis SR, Hooper NM, Hyde R, Karran E, Christie G, Turner AJ. A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J Biol Chem 2000; 275(43): 33238-43.[http://dx.doi.org/10.1074/jbc.M002615200] [PMID: 10924499][39]Lambert DW, Yarski M, Warner FJ, et al. Tumor necrosis factor-α convertase (ADAM17) mediates regulated ectodomain shedding of the severe-acute respiratory syndrome-coronavirus (SARS-CoV) receptor, angiotensin-converting enzyme-2 (ACE2). J Biol Chem 2005; 280(34): 30113-9.[http://dx.doi.org/10.1074/jbc.M505111200] [PMID: 15983030][40]Li W, Zhang C, Sui J, et al. Receptor and viral determinants of SARS-coronavirus adaptation to human ACE2. EMBO J 2005; 24(8): 1634-43.[http://dx.doi.org/10.1038/sj.emboj.7600640] [PMID: 15791205][41]To KF, Lo AWI. Exploring the pathogenesis of severe acute respiratory syndrome (SARS): the tissue distribution of the coronavirus (SARS-CoV) and its putative receptor, angiotensin-converting enzyme 2 (ACE2). J Pathol 2004; 203(3): 740-3.[http://dx.doi.org/10.1002/path.1597] [PMID: 15221932][42]Crackower MA, Sarao R, Oudit GY, et al. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature 2002; 417(6891): 822-8.[http://dx.doi.org/10.1038/nature00786] [PMID: 12075344][43]Hamming I, Timens W, Bulthuis MLC, Lely AT, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 2004; 203(2): 631-7.[http://dx.doi.org/10.1002/path.1570] [PMID: 15141377][44]Jia HP, Look DC, Shi L, et al. ACE2 receptor expression and severe acute respiratory syndrome coronavirus infection depend on differentiation of human airway epithelia. J Virol 2005; 79(23): 14614-21.[http://dx.doi.org/10.1128/JVI.79.23.14614-14621.2005] [PMID: 16282461][45]Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 2020; 395(10224): 565-74.[http://dx.doi.org/10.1016/S0140-6736(20)30251-8] [PMID: 32007145][46]Hofmann H, Geier M, Marzi A, et al. Susceptibility to SARS coronavirus S protein-driven infection correlates with expression of angiotensin converting enzyme 2 and infection can be blocked by soluble receptor. Biochem Biophys Res Commun 2004; 319(4): 1216-21.[http://dx.doi.org/10.1016/j.bbrc.2004.05.114] [PMID: 15194496][47]Zhao Y, Zhao Z, Wang Y, Zhou Y, Ma Y, Zuo W. Single-Cell RNA expression profiling of ACE2, the receptor of SARS-CoV-2. Am J Respir Crit Care Med 2020; 202(5): 756-9.[http://dx.doi.org/10.1164/rccm.202001-0179LE] [PMID: 32663409][48]Rothe C, Schunk M, Sothmann P, et al. Transmission of 2019-NCOV infection from an asymptomatic contact in Germany. N Engl J Med 2020; 382(10): 970-1.[http://dx.doi.org/10.1056/NEJMc2001468] [PMID: 32003551][49]Cao Y, Li L, Feng Z, et al. Comparative genetic analysis of the novel coronavirus (2019-nCoV/SARS-CoV-2) receptor ACE2 in different populations. Cell Discov 2020; 6: 11.[http://dx.doi.org/10.1038/s41421-020-0147-1] [PMID: 32133153][50]Kuba K, Imai Y, Rao S, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med 2005; 11(8): 875-9.[http://dx.doi.org/10.1038/nm1267] [PMID: 16007097][51]Imai Y, Kuba K, Rao S, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 2005; 436(7047): 112-6.[http://dx.doi.org/10.1038/nature03712] [PMID: 16001071][52]Rey-Parra GJ, Vadivel A, Coltan L, et al. Angiotensin converting enzyme 2 abrogates bleomycin-induced lung injury. J Mol Med (Berl) 2012; 90(6): 637-47.[http://dx.doi.org/10.1007/s00109-012-0859-2] [PMID: 22246130][53]Treml B, Neu N, Kleinsasser A, et al. Recombinant angiotensin-converting enzyme 2 improves pulmonary blood flow and oxygenation in lipopolysaccharide-induced lung injury in piglets. Crit Care Med 2010; 38(2): 596-601.[http://dx.doi.org/10.1097/CCM.0b013e3181c03009] [PMID: 19851091][54]Khan A, Benthin C, Zeno B, et al. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit Care 2017; 21(1): 234.[http://dx.doi.org/10.1186/s13054-017-1823-x] [PMID: 28877748][55]Subbarao K, Roberts A. Is there an ideal animal model for SARS? Trends Microbiol 2006; 14(7): 299-303.[http://dx.doi.org/10.1016/j.tim.2006.05.007] [PMID: 16759866][56]Li W, Greenough TC, Moore MJ, et al. Efficient replication of severe acute respiratory syndrome coronavirus in mouse cells is limited by murine angiotensin-converting enzyme 2. J Virol 2004; 78(20): 11429-33.[http://dx.doi.org/10.1128/JVI.78.20.11429-11433.2004] [PMID: 15452268][57]McCray PB, Jr, Pewe L, Wohlford-Lenane C, et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J Virol 2007; 81(2): 813-21.[http://dx.doi.org/10.1128/JVI.02012-06] [PMID: 17079315][58]Koehler DR, Chow YH, Plumb J, et al. A human epithelium-specific vector optimized in rat pneumocytes for lung gene therapy. Pediatr Res 2000; 48(2): 184-90.[http://dx.doi.org/10.1203/00006450-200008000-00011] [PMID: 10926293][59]Bao L, Deng W, Huang B, et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature 2020; 583(7818): 830-3.[http://dx.doi.org/10.1038/s41586-020-2312-y] [PMID: 32380511][60]Peeri NC, Shrestha N, Rahman MS, et al. The SARS, MERS and novel coronavirus (COVID-19) epidemics, the newest and biggest global health threats: what lessons have we learned? Int J Epidemiol 2020; 49(3): 717-26.[http://dx.doi.org/10.1093/ije/dyaa033] [PMID: 32086938][61]Iwata-Yoshikawa N, Okamura T, Shimizu Y, Hasegawa H, Takeda M, Nagata N. TMPRSS2 contributes to virus spread and immunopathology in the airways of murine models after coronavirus infection. J Virol 2019; 93(6): e01815-18.[http://dx.doi.org/10.1128/JVI.01815-18] [PMID: 30626688][62]Matsuyama S, Nagata N, Shirato K, Kawase M, Takeda M, Taguchi F. Efficient activation of the severe acute respiratory syndrome coronavirus spike protein by the transmembrane protease TMPRSS2. J Virol 2010; 84(24): 12658-64.[http://dx.doi.org/10.1128/JVI.01542-10] [PMID: 20926566][63]Glowacka I, Bertram S, Müller MA, et al. Evidence that TMPRSS2 activates the severe acute respiratory syndrome coronavirus spike protein for membrane fusion and reduces viral control by the humoral immune response. J Virol 2011; 85(9): 4122-34.[http://dx.doi.org/10.1128/JVI.02232-10] [PMID: 21325420][64]Tarnow C, Engels G, Arendt A, et al. TMPRSS2 is a host factor that is essential for pneumotropism and pathogenicity of H7N9 influenza A virus in mice. J Virol 2014; 88(9): 4744-51.[http://dx.doi.org/10.1128/JVI.03799-13] [PMID: 24522916][65]Cheng Z, Zhou J, To KKW, et al. Identification of TMPRSS2 as a susceptibility gene for severe 2009 pandemic A(H1N1) influenza and A(H7N9) influenza. J Infect Dis 2015; 212(8): 1214-21.[http://dx.doi.org/10.1093/infdis/jiv246] [PMID: 25904605][66]