51
|
Majid S, Farooq R, Khan MS, Rashid S, Bhat SA, Wani HA, Qureshi W. Managing the COVID-19 Pandemic: Research Strategies Based on the Evolutionary and Molecular Characteristics of Coronaviruses. SN COMPREHENSIVE CLINICAL MEDICINE 2020; 2:1767-1776. [PMID: 32864575 PMCID: PMC7445016 DOI: 10.1007/s42399-020-00457-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/11/2020] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19), an ongoing global health emergency, is a highly transmittable and pathogenic viral infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Emerging in Wuhan, China, in December 2019, it spread widely across the world causing panic-worst ever economic depression is visibly predictable. Coronaviruses (CoVs) have emerged as a major public health concern having caused three zoonotic outbreaks; severe acute respiratory syndrome-CoV (SARS-CoV) in 2002-2003, Middle East respiratory syndrome-CoV (MERS-CoV) in 2012, and currently this devastating COVID-19. Research strategies focused on understanding the evolutionary origin, transmission, and molecular basis of SARS-CoV-2 and its pathogenesis need to be urgently formulated to manage the current and possible future coronaviral outbreaks. Current response to the COVID-19 outbreak has been largely limited to monitoring/containment. Although frantic global efforts for developing safe and effective prophylactic and therapeutic agents are on, no licensed antiviral treatment or vaccine exists till date. In this review, research strategies for coping with COVID-19 based on evolutionary and molecular aspects of coronaviruses have been proposed.
Collapse
Affiliation(s)
- Sabhiya Majid
- Department of Biochemistry, Government Medical College Srinagar and Associated Hospitals , Srinagar, J&K 190010 India
| | - Rabia Farooq
- Department of Biochemistry, Government Medical College Srinagar and Associated Hospitals , Srinagar, J&K 190010 India
- Department of Basic Medical Sciences, University of Bisha, Bisha, 67714 Saudi Arabia
| | - Mosin S. Khan
- Department of Biochemistry, Government Medical College Srinagar and Associated Hospitals , Srinagar, J&K 190010 India
| | - Samia Rashid
- Department of Medicine, Government Medical College Srinagar and Associated Hospitals , Srinagar, J&K 190010 India
| | - Showkat A. Bhat
- Department of Biochemistry, Government Medical College Doda, Doda, J&K 182202 India
| | - Hilal A. Wani
- Department of Biochemistry, Government Medical College Srinagar and Associated Hospitals , Srinagar, J&K 190010 India
| | - Waseem Qureshi
- Registrar Academics, Government Medical College Srinagar, Srinagar, J&K 190010 India
| |
Collapse
|
52
|
Otto DP, de Villiers MM. Layer-By-Layer Nanocoating of Antiviral Polysaccharides on Surfaces to Prevent Coronavirus Infections. Molecules 2020; 25:E3415. [PMID: 32731428 PMCID: PMC7435837 DOI: 10.3390/molecules25153415] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/24/2020] [Accepted: 07/26/2020] [Indexed: 12/28/2022] Open
Abstract
In 2020, the world is being ravaged by the coronavirus, SARS-CoV-2, which causes a severe respiratory disease, Covid-19. Hundreds of thousands of people have succumbed to the disease. Efforts at curing the disease are aimed at finding a vaccine and/or developing antiviral drugs. Despite these efforts, the WHO warned that the virus might never be eradicated. Countries around the world have instated non-pharmaceutical interventions such as social distancing and wearing of masks in public to curb the spreading of the disease. Antiviral polysaccharides provide the ideal opportunity to combat the pathogen via pharmacotherapeutic applications. However, a layer-by-layer nanocoating approach is also envisioned to coat surfaces to which humans are exposed that could harbor pathogenic coronaviruses. By coating masks, clothing, and work surfaces in wet markets among others, these antiviral polysaccharides can ensure passive prevention of the spreading of the virus. It poses a so-called "eradicate-in-place" measure against the virus. Antiviral polysaccharides also provide a green chemistry pathway to virus eradication since these molecules are primarily of biological origin and can be modified by minimal synthetic approaches. They are biocompatible as well as biodegradable. This surface passivation approach could provide a powerful measure against the spreading of coronaviruses.
Collapse
Affiliation(s)
- Daniel P. Otto
- Research Focus Area for Chemical Resource Beneficiation, Laboratory for Analytical Services, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom 2531, South Africa
| | - Melgardt M. de Villiers
- Division of Pharmaceutical Sciences–Drug Delivery, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Ave, Madison, WI 53705, USA;
| |
Collapse
|
53
|
Liya G, Yuguang W, Jian L, Huaiping Y, Xue H, Jianwei H, Jiaju M, Youran L, Chen M, Yiqing J. Studies on viral pneumonia related to novel coronavirus SARS-CoV-2, SARS-CoV, and MERS-CoV: a literature review. APMIS 2020; 128:423-432. [PMID: 32363707 DOI: 10.1111/apm.13047] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/23/2020] [Indexed: 01/08/2023]
Abstract
Coronaviruses are a class of RNA viruses that can cause respiratory and intestinal infections in animals and humans. SARS-CoV, MERS-CoV, and a novel coronavirus (SARS-CoV-2 [2019-nCoV]) belong to the family Coronaviridae and the genus Betacoronavirus. At present, the understanding of SARS-CoV-2 is getting deeper and deeper. In order to better prevent and treat SARS-CoV-2, this article compares the infectivity, pathogenicity, and related clinical characteristics of the three human pathogenic coronaviruses, SARS-CoV-2, SARS-CoV, and MERS-CoV to help us further understand the pathogenic characteristics of novel coronaviruses.
Collapse
Affiliation(s)
- Guo Liya
- Department of Respiratory, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wang Yuguang
- Department of Respiratory, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Liu Jian
- Department of Respiratory, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yuan Huaiping
- Department of Radiology, Fuyang Hospital of Anhui Medical University, China, Anhui
| | - Han Xue
- Department of Respiratory, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Huo Jianwei
- Department of Respiratory, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ma Jiaju
- Department of Respiratory, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Lu Youran
- Department of Respiratory, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ming Chen
- Department of Clinical Microbiology, Hospital of Southern Jutland, Aabenraa, Denmark
| | - Jiao Yiqing
- Department of Respiratory, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
54
|
Schaller T, Hirschbühl K, Burkhardt K, Braun G, Trepel M, Märkl B, Claus R. Postmortem Examination of Patients With COVID-19. JAMA 2020; 323:2518-2520. [PMID: 32437497 PMCID: PMC7243161 DOI: 10.1001/jama.2020.8907] [Citation(s) in RCA: 422] [Impact Index Per Article: 84.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This case series describes autopsy findings in 10 patients with proven severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection who died at a university medical center in Germany.
Collapse
Affiliation(s)
- Tina Schaller
- Institute of Pathology and Molecular Diagnostics, University Medical Center Augsburg, Augsburg, Germany
| | - Klaus Hirschbühl
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, Augsburg, Germany
| | - Katrin Burkhardt
- Institute of Laboratory Medicine and Microbiology, University Medical Center Augsburg, Augsburg, Germany
| | - Georg Braun
- Department of Gastroenterology, University Medical Center Augsburg, Augsburg, Germany
| | - Martin Trepel
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Institute of Pathology and Molecular Diagnostics, University Medical Center Augsburg, Augsburg, Germany
| | - Rainer Claus
- Department of Hematology and Clinical Oncology, University Medical Center Augsburg, Augsburg, Germany
| |
Collapse
|
55
|
Lechowicz K, Drożdżal S, Machaj F, Rosik J, Szostak B, Zegan-Barańska M, Biernawska J, Dabrowski W, Rotter I, Kotfis K. COVID-19: The Potential Treatment of Pulmonary Fibrosis Associated with SARS-CoV-2 Infection. J Clin Med 2020; 9:E1917. [PMID: 32575380 PMCID: PMC7356800 DOI: 10.3390/jcm9061917] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 01/08/2023] Open
Abstract
In December 2019, a novel coronavirus, SARS-CoV-2, appeared, causing a wide range of symptoms, mainly respiratory infection. In March 2020, the World Health Organization (WHO) declared Coronavirus Disease 2019 (COVID-19) a pandemic, therefore the efforts of scientists around the world are focused on finding the right treatment and vaccine for the novel disease. COVID-19 has spread rapidly over several months, affecting patients across all age groups and geographic areas. The disease has a diverse course; patients may range from asymptomatic to those with respiratory failure, complicated by acute respiratory distress syndrome (ARDS). One possible complication of pulmonary involvement in COVID-19 is pulmonary fibrosis, which leads to chronic breathing difficulties, long-term disability and affects patients' quality of life. There are no specific mechanisms that lead to this phenomenon in COVID-19, but some information arises from previous severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS) epidemics. The aim of this narrative review is to present the possible causes and pathophysiology of pulmonary fibrosis associated with COVID-19 based on the mechanisms of the immune response, to suggest possible ways of prevention and treatment.
Collapse
Affiliation(s)
- Kacper Lechowicz
- Department of Anaesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (M.Z.-B.)
| | - Sylwester Drożdżal
- Department of Pharmacokinetics and Monitored Therapy, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Filip Machaj
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (F.M.); (J.R.); (B.S.)
| | - Jakub Rosik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (F.M.); (J.R.); (B.S.)
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (F.M.); (J.R.); (B.S.)
| | - Małgorzata Zegan-Barańska
- Department of Anaesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (M.Z.-B.)
| | - Jowita Biernawska
- Department of Anaesthesiology and Intensive Therapy, Pomeranian Medical University, 71-242 Szczecin, Poland;
| | - Wojciech Dabrowski
- Department of Anaesthesiology and Intensive Care, Medical University, 20-090 Lublin, Poland;
| | - Iwona Rotter
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University, 71-210 Szczecin, Poland;
| | - Katarzyna Kotfis
- Department of Anaesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (M.Z.-B.)
| |
Collapse
|
56
|
Deng M, Jiang L, Ren Y, Liao J. Can We Reduce Mortality of COVID-19 if We do Better in Glucose Control? MEDICINE IN DRUG DISCOVERY 2020; 7:100048. [PMID: 32551437 PMCID: PMC7266598 DOI: 10.1016/j.medidd.2020.100048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 02/08/2023] Open
Abstract
The SARS-CoV-2 has infected more than 3 million people and caused more than 240,000 death globally. Among the COVID-19 patients, the prevalence of people with other chronic diseases, such as diabetes, high blood pressure, and coronary heart disease is much higher than others. More strikingly, the survival rate of diabetic patients is also much lower than in non-diabetic patients. In addition to the general damage of high glucose to cells and tissues, a recent discovery that high glucose activates interferon regulatory factor 15 promotes influenza virus -induced cytokine storm. This discovery may shed light on the high incidence of diabetes in COVID-19. Several diabetes prevention strategies together with recent significant data-driven diabetes prediction approaches, which may help COVID-19 treatments, have been proposed. The mortality rate of COVID-19 patients is about 6% worldwide. Out of the ease patients, one third have diabetes, which has significant impact on the disease progress and mortality. If we can prevent and even cure diabetes, the mortality rate will be significantly reduced. We summarize the potential reasons that high glucose increases the damage of human cells and immune system together with the infection of SARS-CoV-2 virus, and give suggestions for prevention and prediction of diabetes in the AI era.
Collapse
Affiliation(s)
- Mingyan Deng
- West China-California Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ling Jiang
- Biochemistry and Molecular biology Department, School Basic Medical Science, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, 150040, P.R.China
| | - Yan Ren
- Division of Endocrinology and Metabolism, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Jiayu Liao
- Department of Bioengineering, College of Engineering, University of California at Riverside, 900 University Avenue, Riverside, CA 92521, United States of America.,West China-California Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
57
|
Liu H, Chen S, Liu M, Nie H, Lu H. Comorbid Chronic Diseases are Strongly Correlated with Disease Severity among COVID-19 Patients: A Systematic Review and Meta-Analysis. Aging Dis 2020; 11:668-678. [PMID: 32489711 PMCID: PMC7220287 DOI: 10.14336/ad.2020.0502] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/02/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has resulted in considerable morbidity and mortality worldwide since December 2019. In order to explore the effects of comorbid chronic diseases on clinical outcomes of COVID-19, a search was conducted in PubMed, Ovid MEDLINE, EMBASE, CDC, and NIH databases to April 25, 2020. A total of 24 peer-reviewed articles, including 10948 COVID-19 cases were selected. We found diabetes was present in 10.0%, coronary artery disease/cardiovascular disease (CAD/CVD) was in 8.0%, and hypertension was in 20.0%, which were much higher than that of chronic pulmonary disease (3.0%). Specifically, preexisting chronic conditions are strongly correlated with disease severity [Odds ratio (OR) 3.50, 95% CI 1.78 to 6.90], and being admitted to intensive care unit (ICU) (OR 3.36, 95% CI 1.67 to 6.76); in addition, compared to COVID-19 patients with no preexisting chronic diseases, COVID-19 patients who present with either diabetes, hypertension, CAD/CVD, or chronic pulmonary disease have a higher risk of developing severe disease, with an OR of 2.61 (95% CI 1.93 to 3.52), 2.84 (95% CI 2.22 to 3.63), 4.18 (95% CI 2.87 to 6.09) and 3.83 (95% CI 2.15 to 6.80), respectively. Surprisingly, we found no correlation between chronic conditions and increased risk of mortality (OR 2.09, 95% CI 0.26 to16.67). Taken together, cardio-metabolic diseases, such as diabetes, hypertension and CAD/CVD were more common than chronic pulmonary disease in COVID-19 patients, however, each comorbid disease was correlated with increased disease severity. After active treatment, increased risk of mortality in patients with preexisting chronic diseases may reduce.
Collapse
Affiliation(s)
- Hong Liu
- Department of Nutrition, the Third Xiangya Hospital of Central South University, Changsha, China.
| | - Shiyan Chen
- Department of Endocrinology & Metabolism, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.
| | - Min Liu
- Department of Nutrition, the Third Xiangya Hospital of Central South University, Changsha, China.
| | - Hao Nie
- Department of Geriatrics, the First Affiliated Hospital of Hunan Normal University, Changsha, China.
| | - Hongyun Lu
- Department of Endocrinology & Metabolism, Zhuhai Hospital Affiliated with Jinan University, Zhuhai People’s Hospital, Zhuhai, China.
| |
Collapse
|
58
|
Selection of viral variants during persistent infection of insectivorous bat cells with Middle East respiratory syndrome coronavirus. Sci Rep 2020; 10:7257. [PMID: 32350357 PMCID: PMC7190632 DOI: 10.1038/s41598-020-64264-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/09/2020] [Indexed: 02/06/2023] Open
Abstract
Coronaviruses that cause severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) are speculated to have originated in bats. The mechanisms by which these viruses are maintained in individuals or populations of reservoir bats remain an enigma. Mathematical models have predicted long-term persistent infection with low levels of periodic shedding as a likely route for virus maintenance and spillover from bats. In this study, we tested the hypothesis that bat cells and MERS coronavirus (CoV) can co-exist in vitro. To test our hypothesis, we established a long-term coronavirus infection model of bat cells that are persistently infected with MERS-CoV. We infected cells from Eptesicus fuscus with MERS-CoV and maintained them in culture for at least 126 days. We characterized the persistently infected cells by detecting virus particles, protein and transcripts. Basal levels of type I interferon in the long-term infected bat cells were higher, relative to uninfected cells, and disrupting the interferon response in persistently infected bat cells increased virus replication. By sequencing the whole genome of MERS-CoV from persistently infected bat cells, we identified that bat cells repeatedly selected for viral variants that contained mutations in the viral open reading frame 5 (ORF5) protein. Furthermore, bat cells that were persistently infected with ΔORF5 MERS-CoV were resistant to superinfection by wildtype virus, likely due to reduced levels of the virus receptor, dipeptidyl peptidase 4 (DPP4) and higher basal levels of interferon in these cells. In summary, our study provides evidence for a model of coronavirus persistence in bats, along with the establishment of a unique persistently infected cell culture model to study MERS-CoV-bat interactions.
Collapse
|
59
|
Pathological evidence for residual SARS-CoV-2 in pulmonary tissues of a ready-for-discharge patient. Cell Res 2020; 30:541-543. [PMID: 32346074 PMCID: PMC7186763 DOI: 10.1038/s41422-020-0318-5] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/08/2020] [Indexed: 12/19/2022] Open
|
60
|
Wang H, He S, Deng W, Zhang Y, Li G, Sun J, Zhao W, Guo Y, Yin Z, Li D, Shang L. Comprehensive Insights into the Catalytic Mechanism of Middle East Respiratory Syndrome 3C-Like Protease and Severe Acute Respiratory Syndrome 3C-Like Protease. ACS Catal 2020; 10:5871-5890. [PMID: 32391184 PMCID: PMC7202269 DOI: 10.1021/acscatal.0c00110] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/28/2020] [Indexed: 12/11/2022]
Abstract
Coronavirus 3C-like protease (3CLPro) is a highly conserved cysteine protease employing a catalytic dyad for its functions. 3CLPro is essential to the viral life cycle and, therefore, is an attractive target for developing antiviral agents. However, the detailed catalytic mechanism of coronavirus 3CLPro remains largely unknown. We took an integrated approach of employing X-ray crystallography, mutational studies, enzyme kinetics study, and inhibitors to gain insights into the mechanism. Such experimental work is supplemented by computational studies, including the prereaction state analysis, the ab initio calculation of the critical catalytic step, and the molecular dynamic simulation of the wild-type and mutant enzymes. Taken together, such studies allowed us to identify a residue pair (Glu-His) and a conserved His as critical for binding; a conserved GSCGS motif as important for the start of catalysis, a partial negative charge cluster (PNCC) formed by Arg-Tyr-Asp as essential for catalysis, and a conserved water molecule mediating the remote interaction between PNCC and catalytic dyad. The data collected and our insights into the detailed mechanism have allowed us to achieve a good understanding of the difference in catalytic efficiency between 3CLPro from SARS and MERS, conduct mutational studies to improve the catalytic activity by 8-fold, optimize existing inhibitors to improve the potency by 4-fold, and identify a potential allosteric site for inhibitor design. All such results reinforce each other to support the overall catalytic mechanism proposed herein.
Collapse
Affiliation(s)
- Hao Wang
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Drug Discovery Center for Infectious
Disease, Nankai University, 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| | - Shuai He
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Drug Discovery Center for Infectious
Disease, Nankai University, 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| | - Weilong Deng
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Drug Discovery Center for Infectious
Disease, Nankai University, 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| | - Ying Zhang
- Laboratory of Structural Biological
& Ministry of Education and Laboratory of Protein Science, School
of Medicine and Life Sciences, Tsinghua
University, Beijing 100084,
People’s Republic of China
| | - Guobang Li
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Drug Discovery Center for Infectious
Disease, Nankai University, 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| | - Jixue Sun
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| | - Wei Zhao
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Drug Discovery Center for Infectious
Disease, Nankai University, 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| | - Yu Guo
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Drug Discovery Center for Infectious
Disease, Nankai University, 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| | - Zheng Yin
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Drug Discovery Center for Infectious
Disease, Nankai University, 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Center of Basic Molecular Science,
Department of Chemistry, Tsinghua
University, Beijing 100084,
People’s Republic of China
| | - Dongmei Li
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| | - Luqing Shang
- State Key Laboratory of Medicinal
Chemical Biology, College of Pharmacy and KLMDASR of Tianjin,
Nankai University, No. 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
- Drug Discovery Center for Infectious
Disease, Nankai University, 38 Tongyan
Road, Haihe Education Park, Tianjin 300350, People’s
Republic of China
| |
Collapse
|
61
|
Elfiky AA, Azzam EB. Novel guanosine derivatives against MERS CoV polymerase: An in silico perspective. J Biomol Struct Dyn 2020; 39:2923-2931. [PMID: 32306854 PMCID: PMC7189410 DOI: 10.1080/07391102.2020.1758789] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The Middle East Respiratory Syndrome Coronavirus (MERS CoV), also termed camel flu, is a new viral infection that first reported in the year 2012 in the Middle East region and further spread during the last seven years. MERS CoV is characterized by its high mortality rate among different human coronaviruses. MERS CoV polymerase shares more than 20% sequence identity with the Hepatitis C Virus (HCV) Non-structural 5b (NS5b) RNA dependent RNA polymerase (RdRp). Despite the low sequence identity, the active site is conserved between the two proteins, with two consecutive aspartates that are crucial in the nucleotide transfer reaction. In this study, seven nucleotide inhibitors have been tested against MERS CoV RdRp using molecular modeling and docking simulations, from which four are novel compounds. Molecular Dynamics Simulation for 260 nanoseconds is performed on the MERS CoV RdRp model to test the effect of protein dynamics on the binding affinities to the tested nucleotide inhibitors. Results support the hypothesis of using the anti-polymerases (Anti-HCV drugs) against MERS CoV RdRp as a potent candidates. Besides four novel compounds are suggested as a seed for high performance inhibitors against MERS CoV RdRp.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdo A Elfiky
- Biophysics Department, Faculty of Sciences, Cairo University, Giza, Egypt
| | - Eman B Azzam
- Biophysics Section of Physics Department, Faculty of Sciences, Helwan University, Cairo, Egypt
| |
Collapse
|
62
|
Horowitz RI, Freeman PR, Bruzzese J. Efficacy of glutathione therapy in relieving dyspnea associated with COVID-19 pneumonia: A report of 2 cases. Respir Med Case Rep 2020; 30:101063. [PMID: 32322478 PMCID: PMC7172740 DOI: 10.1016/j.rmcr.2020.101063] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/19/2020] [Accepted: 04/19/2020] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Infection with COVID-19 potentially can result in severe outcomes and death from "cytokine storm syndrome", resulting in novel coronavirus pneumonia (NCP) with severe dyspnea, acute respiratory distress syndrome (ARDS), fulminant myocarditis and multiorgan dysfunction with or without disseminated intravascular coagulation. No published treatment to date has been shown to adequately control the inflammation and respiratory symptoms associated with COVID-19, apart from oxygen therapy and assisted ventilation. We evaluated the effects of using high dose oral and/or IV glutathione in the treatment of 2 patients with dyspnea secondary to COVID-19 pneumonia. METHODS Two patients living in New York City (NYC) with a history of Lyme and tick-borne co-infections experienced a cough and dyspnea and demonstrated radiological findings consistent with novel coronavirus pneumonia (NCP). A trial of 2 g of PO or IV glutathione was used in both patients and improved their dyspnea within 1 h of use. Repeated use of both 2000 mg of PO and IV glutathione was effective in further relieving respiratory symptoms. CONCLUSION Oral and IV glutathione, glutathione precursors (N-acetyl-cysteine) and alpha lipoic acid may represent a novel treatment approach for blocking NF-κB and addressing "cytokine storm syndrome" and respiratory distress in patients with COVID-19 pneumonia.
Collapse
Affiliation(s)
- Richard I. Horowitz
- HHS Babesia and Tickborne Pathogen Subcommittee, Washington, D.C., 20201, USA
- Hudson Valley Healing Arts Center, 4232 Albany Post Road, Hyde Park, NY, 12538, USA
| | - Phyllis R. Freeman
- Hudson Valley Healing Arts Center, 4232 Albany Post Road, Hyde Park, NY, 12538, USA
| | - James Bruzzese
- Sophie Davis School of Biomedical Education/CUNY School of Medicine, New York, NY, 10031, USA
| |
Collapse
|
63
|
He F, Deng Y, Li W. Coronavirus disease 2019: What we know? J Med Virol 2020; 92:719-725. [PMID: 32170865 PMCID: PMC7228340 DOI: 10.1002/jmv.25766] [Citation(s) in RCA: 432] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
Abstract
In late December 2019, a cluster of unexplained pneumonia cases has been reported in Wuhan, China. A few days later, the causative agent of this mysterious pneumonia was identified as a novel coronavirus. This causative virus has been temporarily named as severe acute respiratory syndrome coronavirus 2 and the relevant infected disease has been named as coronavirus disease 2019 (COVID-19) by the World Health Organization, respectively. The COVID-19 epidemic is spreading in China and all over the world now. The purpose of this review is primarily to review the pathogen, clinical features, diagnosis, and treatment of COVID-19, but also to comment briefly on the epidemiology and pathology based on the current evidence.
Collapse
Affiliation(s)
- Feng He
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Deng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weina Li
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
64
|
Willman M, Kobasa D, Kindrachuk J. A Comparative Analysis of Factors Influencing Two Outbreaks of Middle Eastern Respiratory Syndrome (MERS) in Saudi Arabia and South Korea. Viruses 2019; 11:v11121119. [PMID: 31817037 PMCID: PMC6950189 DOI: 10.3390/v11121119] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 01/06/2023] Open
Abstract
In 2012, an emerging viral infection was identified in Saudi Arabia that subsequently spread to 27 additional countries globally, though cases may have occurred elsewhere. The virus was ultimately named Middle Eastern Respiratory Syndrome Coronavirus (MERS-CoV), and has been endemic in Saudi Arabia since 2012. As of September 2019, 2468 laboratory-confirmed cases with 851 associated deaths have occurred with a case fatality rate of 34.4%, according to the World Health Organization. An imported case of MERS occurred in South Korea in 2015, stimulating a multi-month outbreak. Several distinguishing factors emerge upon epidemiological and sociological analysis of the two outbreaks including public awareness of the MERS outbreak, and transmission and synchronization of governing healthcare bodies. South Korea implemented a stringent healthcare model that protected patients and healthcare workers alike through prevention and high levels of public information. In addition, many details about MERS-CoV virology, transmission, pathological progression, and even the reservoir, remain unknown. This paper aims to delineate the key differences between the two regional outbreaks from both a healthcare and personal perspective including differing hospital practices, information and public knowledge, cultural practices, and reservoirs, among others. Further details about differing emergency outbreak responses, public information, and guidelines put in place to protect hospitals and citizens could improve the outcome of future MERS outbreaks.
Collapse
Affiliation(s)
- Marnie Willman
- High Containment Respiratory Viruses, Special Pathogens, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.W.); (D.K.)
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Darwyn Kobasa
- High Containment Respiratory Viruses, Special Pathogens, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.W.); (D.K.)
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Jason Kindrachuk
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Correspondence: ; Tel.: +1-204-789-3807
| |
Collapse
|
65
|
Alosaimi B, Hamed ME, Naeem A, Alsharef AA, AlQahtani SY, AlDosari KM, Alamri AA, Al-Eisa K, Khojah T, Assiri AM, Enani MA. MERS-CoV infection is associated with downregulation of genes encoding Th1 and Th2 cytokines/chemokines and elevated inflammatory innate immune response in the lower respiratory tract. Cytokine 2019; 126:154895. [PMID: 31706200 PMCID: PMC7128721 DOI: 10.1016/j.cyto.2019.154895] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/31/2022]
Abstract
MERS-CoV infection downregulates Th1 and Th2 cytokines and chemokines. MERS-CoV infection provokes high levels of IL-1α, IL-1β and IL-8 (CXCL8). Inflammatory cytokines/chemokines correlate with MERS-CoV case fatality rate. Th1/Th2 downregulation may contribute to severe infection and evolution of ARDS.
MERS-CoV, a highly pathogenic virus in humans, is associated with high morbidity and case fatality. Inflammatory responses have a significant impact on MERS-CoV pathogenesis and disease outcome. However, CD4+ T-cell induced immune responses during acute MERS-CoV infection are barely detectable, with potent inhibition of effector T cells and downregulation of antigen presentation. The local pulmonary immune response, particularly the Th1 and Th2-related immune response during acute severe MERS-CoV infection is not fully understood. In this study, we offer the first insights into the pulmonary gene expression profile of Th1 and Th2-related cytokines/chemokines (Th1 & Th2 responses) during acute MERS-CoV infection using RT2 Profiler PCR Arrays. We also quantified the expression level of primary inflammatory cytokines/chemokines. Our results showed a downregulation of Th2, inadequate (partial) Th1 immune response and high expression levels of inflammatory cytokines IL-1α and IL-1β and the neutrophil chemoattractant chemokine IL-8 (CXCL8) in the lower respiratory tract of MERS-CoV infected patients. Moreover, we identified a high viral load in all included patients. We also observed a correlation between inflammatory cytokines, Th1, and Th2 downregulation and the case fatality rate. Th1 and Th2 response downregulation, high expression of inflammatory cytokines, and high viral load may contribute to lung inflammation, severe infection, the evolution of pneumonia and ARDS, and a higher case fatality rate. Further study of the molecular mechanisms underlying the Th1 and Th2 regulatory pathways will be vital for active vaccine development and the identification of novel therapeutic strategies.
Collapse
Affiliation(s)
- Bandar Alosaimi
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia; College of Medicine, King Fahad Medical City, Riyadh, Saudi Arabia.
| | - Maaweya E Hamed
- College of Science, King Saud University, Department of Botany and Microbiology, Riyadh, Saudi Arabia
| | - Asif Naeem
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ali A Alsharef
- General Directorate of Laboratories and Blood Banks, Ministry of Health, Saudi Arabia
| | - Saeed Y AlQahtani
- General Directorate of Laboratories and Blood Banks, Ministry of Health, Saudi Arabia
| | - Kamel M AlDosari
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Aref A Alamri
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Kholoud Al-Eisa
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Taghreed Khojah
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Abdullah M Assiri
- Preventive Medicine Assistant Deputyship, Ministry of Health, Riyadh, Saudi Arabia
| | - Mushira A Enani
- Medical Specialties Department, Section of Infectious Diseases, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
66
|
Kuiken T, Breitbart M, Beer M, Grund C, Höper D, van den Hoogen B, Kerkhoffs JLH, Kroes ACM, Rosario K, van Run P, Schwarz M, Svraka S, Teifke J, Koopmans M. Zoonotic Infection With Pigeon Paramyxovirus Type 1 Linked to Fatal Pneumonia. J Infect Dis 2019; 218:1037-1044. [PMID: 29373675 PMCID: PMC7107406 DOI: 10.1093/infdis/jiy036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/17/2018] [Indexed: 02/06/2023] Open
Abstract
The characteristics and risk factors of pigeon paramyxovirus type 1 (PPMV-1) infection in humans are poorly known. We performed virological, pathological, and epidemiological analyses of a Dutch case, and compared the results with those of a US case. Both infections occurred in transplant patients under immunosuppressive therapy and caused fatal respiratory failure. Both virus isolates clustered with PPMV-1, which has pigeons and doves as reservoir. Experimentally inoculated pigeons became infected and transmitted the virus to naive pigeons. Both patients were likely infected by contact with infected pigeons or doves. Given the large populations of feral pigeons with PPMV-1 infection in cities, increasing urbanization, and a higher proportion of immunocompromised individuals, the risk of severe human PPMV-1 infections may increase. We recommend testing for avian paramyxovirus type 1, including PPMV-1, in respiratory disease cases where common respiratory pathogens cannot be identified.
Collapse
Affiliation(s)
- Thijs Kuiken
- Department of Viroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Mya Breitbart
- College of Marine Science, University of South Florida, Saint Petersburg
| | - Martin Beer
- Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Christian Grund
- Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Dirk Höper
- Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | | | - Jean-Louis H Kerkhoffs
- Department of Medical Microbiology, Leiden University Medical Centre, Bilthoven, The Netherlands
| | - Aloys C M Kroes
- Department of Medical Microbiology, Leiden University Medical Centre, Bilthoven, The Netherlands
| | - Karyna Rosario
- College of Marine Science, University of South Florida, Saint Petersburg
| | - Peter van Run
- Department of Viroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Sanela Svraka
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Jens Teifke
- Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Marion Koopmans
- Department of Viroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands.,Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
67
|
Lee JY, Bae S, Myoung J. Middle East respiratory syndrome coronavirus-encoded ORF8b strongly antagonizes IFN-β promoter activation: its implication for vaccine design. J Microbiol 2019; 57:803-811. [PMID: 31452044 PMCID: PMC7091237 DOI: 10.1007/s12275-019-9272-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is a causative agent of severe-to-fatal pneumonia especially in patients with pre-existing conditions, such as smoking and chronic obstructive pulmonary disease (COPD). MERS-CoV transmission continues to be reported in the Saudi Arabian Peninsula since its discovery in 2012. However, it has rarely been epidemic outside the area except one large outbreak in South Korea in May 2015. The genome of the epidemic MERS-CoV isolated from a Korean patient revealed its homology to previously reported strains. MERS-CoV encodes 5 accessory proteins and generally, they do not participate in the genome transcription and replication but rather are involved in viral evasion of the host innate immune responses. Here we report that ORF8b, an accessory protein of MERS-CoV, strongly inhibits both MDA5- and RIG-I-mediated activation of interferon beta promoter activity while downstream signaling molecules were left largely unaffected. Of note, MDA5 protein levels were significantly down-regulated by ORF8b and co-expression of ORF4a and ORF4b. These novel findings will facilitate elucidation of mechanisms of virus-encoded evasion strategies, thus helping design rationale antiviral countermeasures against deadly MERS-CoV infection.
Collapse
Affiliation(s)
- Jeong Yoon Lee
- Korea Zoonosis Research Institute, Genetic Engineering Research Institute & Department of Bioactive Material Science, College of Natural Science, Chonbuk National University, Jeonju, 54531, Republic of Korea
| | - Sojung Bae
- Korea Zoonosis Research Institute, Genetic Engineering Research Institute & Department of Bioactive Material Science, College of Natural Science, Chonbuk National University, Jeonju, 54531, Republic of Korea
| | - Jinjong Myoung
- Korea Zoonosis Research Institute, Genetic Engineering Research Institute & Department of Bioactive Material Science, College of Natural Science, Chonbuk National University, Jeonju, 54531, Republic of Korea.
| |
Collapse
|
68
|
Han Y, Du J, Su H, Zhang J, Zhu G, Zhang S, Wu Z, Jin Q. Identification of Diverse Bat Alphacoronaviruses and Betacoronaviruses in China Provides New Insights Into the Evolution and Origin of Coronavirus-Related Diseases. Front Microbiol 2019; 10:1900. [PMID: 31474969 PMCID: PMC6702311 DOI: 10.3389/fmicb.2019.01900] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 07/31/2019] [Indexed: 12/29/2022] Open
Abstract
Outbreaks of severe acute respiratory syndrome (SARS) in 2002, Middle East respiratory syndrome in 2012 and fatal swine acute diarrhea syndrome in 2017 caused serious infectious diseases in humans and in livestock, resulting in serious public health threats and huge economic losses. All such coronaviruses (CoVs) were confirmed to originate from bats. To continuously monitor the epidemic-related CoVs in bats, virome analysis was used to classify CoVs from 831 bats of 15 species in Yunnan, Guangxi, and Sichuan Provinces between August 2016 and May 2017. We identified 11 CoV strains from 22 individual samples of four bat species. Identification of four alpha-CoVs from Scotophilus kuhlii in Guangxi, which was closely related to a previously reported bat CoV and porcine epidemic diarrhea virus (PEDV), revealed a bat-swine lineage under the genus Alphacoronavirus. A recombinant CoV showed that the PEDV probably originated from the CoV of S. kuhlii. Another alpha-CoV, α-YN2018, from Rhinolophus sinicus in Yunnan, suggested that this alpha-CoV lineage had multiple host origins, and α-YN2018 had recombined with CoVs of other bat species over time. We identified five SARS-related CoVs (SARSr-CoVs) in Rhinolophus bats from Sichuan and Yunnan and confirmed that angiotensin-converting enzyme 2 usable SARSr-CoVs were continuously circulating in Rhinolophus spp. in Yunnan. The other beta-CoV, strain β-GX2018, found in Cynopterus sphinx of Guangxi, represented an independently evolved lineage different from known CoVs of Rousettus and Eonycteris bats. The identification of diverse CoVs here provides new genetic data for understanding the distribution and source of pathogenic CoVs in China.
Collapse
Affiliation(s)
- Yelin Han
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Du
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haoxiang Su
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junpeng Zhang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | | | - Shuyi Zhang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Zhiqiang Wu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Jin
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
69
|
Skariyachan S, Challapilli SB, Packirisamy S, Kumargowda ST, Sridhar VS. Recent Aspects on the Pathogenesis Mechanism, Animal Models and Novel Therapeutic Interventions for Middle East Respiratory Syndrome Coronavirus Infections. Front Microbiol 2019; 10:569. [PMID: 30984127 PMCID: PMC6448012 DOI: 10.3389/fmicb.2019.00569] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 03/05/2019] [Indexed: 12/17/2022] Open
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is an emerging zoonotic virus considered as one of the major public threat with a total number of 2 298 laboratory-confirmed cases and 811 associated deaths reported by World Health Organization as of January 2019. The transmission of the virus was expected to be from the camels found in Middle Eastern countries via the animal and human interaction. The genome structure provided information about the pathogenicity and associated virulent factors present in the virus. Recent studies suggested that there were limited insight available on the development of novel therapeutic strategies to induce immunity against the virus. The severities of MERS-CoV infection highlight the necessity of effective approaches for the development of various therapeutic remedies. Thus, the present review comprehensively and critically illustrates the recent aspects on the epidemiology of the virus, the structural and functional features of the viral genome, viral entry and transmission, major mechanisms of pathogenesis and associated virulent factors, current animal models, detection methods and novel strategies for the development of vaccines against MERS-CoV. The review further illustrates the molecular and computational virtual screening platforms which provide insights for the identification of putative drug targets and novel lead molecules toward the development of therapeutic remedies.
Collapse
Affiliation(s)
- Sinosh Skariyachan
- R&D Centre, Department of Biotechnology, Dayananda Sagar College of Engineering, Bengaluru, India
| | | | | | | | | |
Collapse
|
70
|
Acute Respiratory Infection in Human Dipeptidyl Peptidase 4-Transgenic Mice Infected with Middle East Respiratory Syndrome Coronavirus. J Virol 2019; 93:JVI.01818-18. [PMID: 30626685 PMCID: PMC6401458 DOI: 10.1128/jvi.01818-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) infections are endemic in the Middle East and a threat to public health worldwide. Rodents are not susceptible to the virus because they do not express functional receptors; therefore, we generated a new animal model of MERS-CoV infection based on transgenic mice expressing human DPP4 (hDPP4). The pattern of hDPP4 expression in this model was similar to that in human tissues (except lymphoid tissue). In addition, MERS-CoV was limited to the respiratory tract. Here, we focused on host factors involved in immunopathology in MERS-CoV infection and clarified differences in antiviral immune responses between young and adult transgenic mice. This new small-animal model could contribute to more in-depth study of the pathology of MERS-CoV infection and aid development of suitable treatments. Middle East respiratory syndrome coronavirus (MERS-CoV) infection can manifest as a mild illness, acute respiratory distress, organ failure, or death. Several animal models have been established to study disease pathogenesis and to develop vaccines and therapeutic agents. Here, we developed transgenic (Tg) mice on a C57BL/6 background; these mice expressed human CD26/dipeptidyl peptidase 4 (hDPP4), a functional receptor for MERS-CoV, under the control of an endogenous hDPP4 promoter. We then characterized this mouse model of MERS-CoV. The expression profile of hDPP4 in these mice was almost equivalent to that in human tissues, including kidney and lung; however, hDPP4 was overexpressed in murine CD3-positive cells within peripheral blood and lymphoid tissues. Intranasal inoculation of young and adult Tg mice with MERS-CoV led to infection of the lower respiratory tract and pathological evidence of acute multifocal interstitial pneumonia within 7 days, with only transient loss of body weight. However, the immunopathology in young and adult Tg mice was different. On day 5 or 7 postinoculation, lungs of adult Tg mice contained higher levels of proinflammatory cytokines and chemokines associated with migration of macrophages. These results suggest that the immunopathology of MERS-CoV infection in the Tg mouse is age dependent. The mouse model described here will increase our understanding of disease pathogenesis and host mediators that protect against MERS-CoV infection. IMPORTANCE Middle East respiratory syndrome coronavirus (MERS-CoV) infections are endemic in the Middle East and a threat to public health worldwide. Rodents are not susceptible to the virus because they do not express functional receptors; therefore, we generated a new animal model of MERS-CoV infection based on transgenic mice expressing human DPP4 (hDPP4). The pattern of hDPP4 expression in this model was similar to that in human tissues (except lymphoid tissue). In addition, MERS-CoV was limited to the respiratory tract. Here, we focused on host factors involved in immunopathology in MERS-CoV infection and clarified differences in antiviral immune responses between young and adult transgenic mice. This new small-animal model could contribute to more in-depth study of the pathology of MERS-CoV infection and aid development of suitable treatments.
Collapse
|
71
|
TMPRSS2 Contributes to Virus Spread and Immunopathology in the Airways of Murine Models after Coronavirus Infection. J Virol 2019; 93:JVI.01815-18. [PMID: 30626688 PMCID: PMC6401451 DOI: 10.1128/jvi.01815-18] [Citation(s) in RCA: 472] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/28/2018] [Indexed: 11/25/2022] Open
Abstract
Broad-spectrum antiviral drugs against highly pathogenic coronaviruses and other emerging viruses are desirable to enable a rapid response to pandemic threats. Transmembrane protease serine type 2 (TMPRSS2), a protease belonging to the type II transmembrane serine protease family, cleaves the coronavirus spike protein, making it a potential therapeutic target for coronavirus infections. Here, we examined the role of TMPRSS2 using animal models of SARS-CoV and MERS-CoV infection. The results suggest that lack of TMPRSS2 in the airways reduces the severity of lung pathology after infection by SARS-CoV and MERS-CoV. Taken together, the results will facilitate development of novel targets for coronavirus therapy. Transmembrane serine protease TMPRSS2 activates the spike protein of highly pathogenic human coronaviruses such as severe acute respiratory syndrome-related coronavirus (SARS-CoV) and Middle East respiratory syndrome-related coronavirus (MERS-CoV). In vitro, activation induces virus-cell membrane fusion at the cell surface. However, the roles of TMPRSS2 during coronavirus infection in vivo are unclear. Here, we used animal models of SARS-CoV and MERS-CoV infection to investigate the role of TMPRSS2. Th1-prone C57BL/6 mice and TMPRSS2-knockout (KO) mice were used for SARS-CoV infection, and transgenic mice expressing the human MERS-CoV receptor DPP4 (hDPP4-Tg mice) and TMPRSS2-KO hDPP4-Tg mice were used for MERS-CoV infection. After experimental infection, TMPRSS2-deficient mouse strains showed reduced body weight loss and viral kinetics in the lungs. Lack of TMPRSS2 affected the primary sites of infection and virus spread within the airway, accompanied by less severe immunopathology. However, TMPRSS2-KO mice showed weakened inflammatory chemokine and/or cytokine responses to intranasal stimulation with poly(I·C), a Toll-like receptor 3 agonist. In conclusion, TMPRSS2 plays a crucial role in viral spread within the airway of murine models infected by SARS-CoV and MERS-CoV and in the resulting immunopathology. IMPORTANCE Broad-spectrum antiviral drugs against highly pathogenic coronaviruses and other emerging viruses are desirable to enable a rapid response to pandemic threats. Transmembrane protease serine type 2 (TMPRSS2), a protease belonging to the type II transmembrane serine protease family, cleaves the coronavirus spike protein, making it a potential therapeutic target for coronavirus infections. Here, we examined the role of TMPRSS2 using animal models of SARS-CoV and MERS-CoV infection. The results suggest that lack of TMPRSS2 in the airways reduces the severity of lung pathology after infection by SARS-CoV and MERS-CoV. Taken together, the results will facilitate development of novel targets for coronavirus therapy.
Collapse
|
72
|
Kim JI, Park S, Bae JY, Park MS. Evolutionary relationship analysis of Middle East respiratory syndrome coronavirus 4a and 4b protein coding sequences. J Vet Sci 2019; 20:e1. [PMID: 30944524 PMCID: PMC6441811 DOI: 10.4142/jvs.2019.20.e1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/26/2018] [Accepted: 12/16/2018] [Indexed: 02/02/2023] Open
Abstract
The 4a and 4b proteins of the Middle East respiratory syndrome coronavirus (MERS-CoV) have been described for their antagonism on host innate immunity. However, unlike clustering patterns of the complete gene sequences of human and camel MERS-CoVs, the 4a and 4b protein coding regions did not constitute species-specific phylogenetic groups. Moreover, given the estimated evolutionary rates of the complete, 4a, and 4b gene sequences, the 4a and 4b proteins might be less affected by species-specific innate immune pressures. These results suggest that the 4a and 4b proteins of MERS-CoV may function against host innate immunity in a manner independent of host species and/or evolutionary clustering patterns.
Collapse
Affiliation(s)
- Jin Il Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Sehee Park
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Joon-Yong Bae
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
73
|
Tambyah P, Isa MS, Tan CXT. New and Emerging Infections of the Lung. KENDIG'S DISORDERS OF THE RESPIRATORY TRACT IN CHILDREN 2019. [PMCID: PMC7151841 DOI: 10.1016/b978-0-323-44887-1.00028-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this era of rapid globalization and frequent travel, emerging viral infections have gained an immense potential to spread at an unprecedented speed and scale compared with the past. This poses a significant challenge to coordinated international efforts in global surveillance and infection control. Significantly, respiratory viral infections, spread mostly via droplet transmission, are extremely contagious and have caused significant morbidity and mortality during outbreaks in the last decade. Molecular diagnostics via reverse transcriptase polymerase chain reaction (RT-PCR) have been key in the rapid diagnosis of most of these viral infections. However, a high index of suspicion and early institution of appropriate isolation measures remain as the mainstay in the control and containment of the spread of these viral infections. Although treatment for most of the viral infections remains supportive, efficacious antiviral agents against influenza infections exist. The infections discussed in this chapter include those first described in the 2000s: Middle East respiratory syndrome coronavirus (MERS-CoV) and metapneumovirus and rhinovirus C as well as those that have been described in the past but have reemerged in the last decade in outbreaks resulting in significant morbidity and mortality, including adenovirus, influenza virus, and enterovirus D68 (EV-D68).
Collapse
|
74
|
Toosy AH, O'sullivan S. An Overview of Middle East Respiratory Syndrome in the Middle East. FOWLER'S ZOO AND WILD ANIMAL MEDICINE CURRENT THERAPY, VOLUME 9 2019. [PMCID: PMC7152387 DOI: 10.1016/b978-0-323-55228-8.00042-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
75
|
Jia W, Channappanavar R, Zhang C, Li M, Zhou H, Zhang S, Zhou P, Xu J, Shan S, Shi X, Wang X, Zhao J, Zhou D, Perlman S, Zhang L. Single intranasal immunization with chimpanzee adenovirus-based vaccine induces sustained and protective immunity against MERS-CoV infection. Emerg Microbes Infect 2019; 8:760-772. [PMID: 31130102 PMCID: PMC6542157 DOI: 10.1080/22221751.2019.1620083] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/16/2019] [Accepted: 05/09/2019] [Indexed: 01/19/2023]
Abstract
The recently identified Middle East Respiratory Syndrome Coronavirus (MERS-CoV) causes severe and fatal acute respiratory illness in humans. However, no approved prophylactic and therapeutic interventions are currently available. The MERS-CoV envelope spike protein serves as a crucial target for neutralizing antibodies and vaccine development, as it plays a critical role in mediating viral entry through interactions with the cellular receptor, dipeptidyl peptidase 4 (DPP4). Here, we constructed a recombinant rare serotype of the chimpanzee adenovirus 68 (AdC68) that expresses full-length MERS-CoV S protein (AdC68-S). Single intranasal immunization with AdC68-S induced robust and sustained neutralizing antibody and T cell responses in BALB/c mice. In a human DPP4 knock-in (hDPP4-KI) mouse model, it completely protected against lethal challenge with a mouse-adapted MERS-CoV (MERS-CoV-MA). Passive transfer of immune sera to naïve hDPP4-KI mice also provided survival advantages from lethal MERS-CoV-MA challenge. Analysis of sera absorption and isolated monoclonal antibodies from immunized mice demonstrated that the potent and broad neutralizing activity was largely attributed to antibodies targeting the receptor binding domain (RBD) of the S protein. These results show that AdC68-S can induce protective immune responses in mice and represent a promising candidate for further development against MERS-CoV infection in both dromedaries and humans.
Collapse
MESH Headings
- Adenoviridae/genetics
- Administration, Intranasal
- Animals
- Animals, Genetically Modified
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Coronavirus Infections/prevention & control
- Drug Carriers/administration & dosage
- Humans
- Immunization, Passive
- Mice, Inbred BALB C
- Middle East Respiratory Syndrome Coronavirus/genetics
- Middle East Respiratory Syndrome Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Survival Analysis
- T-Lymphocytes/immunology
- Treatment Outcome
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Wenxu Jia
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Rudragouda Channappanavar
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, USA
- Department of Acute and Tertiary Care, and the Institute for the Study of Host–Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Chao Zhang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Key Laboratory of Molecular Virology & Immunology, Vaccine Research Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Mingxi Li
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Haixia Zhou
- The Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Biotherapy, School of Life Sciences, Tsinghua University, Beijing, People’s Republic of China
| | - Shuyuan Zhang
- The Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Biotherapy, School of Life Sciences, Tsinghua University, Beijing, People’s Republic of China
| | - Panpan Zhou
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Jiuyang Xu
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Sisi Shan
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Xuanling Shi
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Xinquan Wang
- The Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Biotherapy, School of Life Sciences, Tsinghua University, Beijing, People’s Republic of China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Dongming Zhou
- Key Laboratory of Molecular Virology & Immunology, Vaccine Research Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Stanley Perlman
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, USA
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Linqi Zhang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People’s Republic of China
| |
Collapse
|
76
|
Chen J, Wang H, Jin L, Wang L, Huang X, Chen W, Yan M, Liu G. Profile analysis of circRNAs induced by porcine endemic diarrhea virus infection in porcine intestinal epithelial cells. Virology 2018; 527:169-179. [PMID: 30530223 PMCID: PMC7112103 DOI: 10.1016/j.virol.2018.11.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 01/16/2023]
Abstract
The circRNA is a newly defined noncoding RNA and characterized by its unique splicing reactions to form circles. However, the function of circRNAs during viral infection remains largely unknown. In this study, the circRNA expression profile during porcine endemic diarrhea virus (PEDV) infection in IPEC-J2 cell line was investigated using the next-generation sequencing technology. A total of 26670 circRNA candidates were identified. The functional annotation analysis revealed that the parent genes of differentially expressed circRNAs might be associated with host response to PEDV infection. Further analysis verified the existence of eight selected circRNAs and confirmed that PEDV infection alerted the expression patterns of circRNAs and their linear parent genes in IPEC-J2 cell line. The circRNA-miRNA interaction network was also constructed to elucidate their potential targets. Our results provided not only the first large-scale profile analysis of circRNAs associated with PEDV infection but also a novel direction to elucidate host-PEDV interactions.
Collapse
Affiliation(s)
- Jianing Chen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Haiwen Wang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Li Jin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Liyuan Wang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Xin Huang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Wenwen Chen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Miaomiao Yan
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Guangliang Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China.
| |
Collapse
|
77
|
Lee K, Ko HL, Lee EY, Park HJ, Kim YS, Kim YS, Cho NH, Park MS, Lee SM, Kim J, Kim H, Seong BL, Nam JH. Development of a diagnostic system for detection of specific antibodies and antigens against Middle East respiratory syndrome coronavirus. Microbiol Immunol 2018; 62:574-584. [PMID: 30117617 PMCID: PMC7168444 DOI: 10.1111/1348-0421.12643] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/22/2018] [Accepted: 08/08/2018] [Indexed: 12/25/2022]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is a single-stranded RNA virus that causes severe respiratory disease in humans with a high fatality rate. Binding of the receptor binding domain (RBD) of the spike (S) glycoprotein to dipeptidyl peptidase 4 is the critical step in MERS-CoV infection of a host cell. No vaccines or clinically applicable treatments are currently available for MERS-CoV. Therefore, rapid diagnosis is important for improving patient outcomes through prompt treatment and protection against viral outbreaks. In this study, the aim was to establish two ELISA systems for detecting antigens and antibodies against MERS-CoV. Using a recombinant full-length S protein, an indirect ELISA was developed and found to detect MERS-CoV-specific antibodies in animal sera and sera of patient with MERS. Moreover, MAbs were induced with the recombinant S protein and RBD and used for sandwich ELISA to detect the MERS-CoV S protein. Neither ELISA system exhibited significant intra-assay or inter-assay variation, indicating good reproducibility. Moreover, the inter-day precision and sensitivity were adequate for use as a diagnostic kit. Thus, these ELISAs can be used clinically to diagnose MERS-CoV.
Collapse
Affiliation(s)
- Kunse Lee
- Department of Biotechnology, Catholic University of Korea, Bucheon 14662, Korea.,Bio Research and Development, SK Chemicals, Seongnam, Gyeonggi-do 13493, Korea
| | - Hae Li Ko
- Department of Biotechnology, Catholic University of Korea, Bucheon 14662, Korea
| | - Eun-Young Lee
- Department of Biotechnology, Catholic University of Korea, Bucheon 14662, Korea
| | - Hyo-Jung Park
- Department of Biotechnology, Catholic University of Korea, Bucheon 14662, Korea
| | - Young Seok Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Yeon-Sook Kim
- Division of Infectious Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Man-Seong Park
- Department of Microbiology, College of Medicine, Institute for Viral Diseases, Korea University, Seoul 02841, Korea
| | - Sang-Myeong Lee
- Department of, Biotechnology, Chonbuk National University, Iksan 570-752, Korea
| | - Jihye Kim
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Korea
| | - Hun Kim
- Bio Research and Development, SK Chemicals, Seongnam, Gyeonggi-do 13493, Korea
| | - Baik Lin Seong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jae-Hwan Nam
- Department of Biotechnology, Catholic University of Korea, Bucheon 14662, Korea
| |
Collapse
|
78
|
Overexpression of the nucleocapsid protein of Middle East respiratory syndrome coronavirus up-regulates CXCL10. Biosci Rep 2018; 38:BSR20181059. [PMID: 30242057 PMCID: PMC6200698 DOI: 10.1042/bsr20181059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 11/17/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) causes respiratory diseases in humans and has a high mortality rate. During infection, MERS-CoV regulates several host cellular processes including antiviral response genes. In order to determine if the nucleocapsid protein of MERS-CoV (MERS-N) plays a role in viral-host interactions, a murine monoclonal antibody was generated so as to allow detection of the protein in infected cells as well as in overexpression system. Then, MERS-N was stably overexpressed in A549 cells, and a PCR array containing 84 genes was used to screen for genes transcriptionally regulated by it. Several up-regulated antiviral genes, namely TNF, IL6, IL8, and CXCL10, were selected for independent validation in transiently transfected 293FT cells. Out of these, the overexpression of MERS-N was found to up-regulate CXCL10 at both transcriptional and translational levels. Interestingly, CXCL10 has been reported to be up-regulated in MERS-CoV infected airway epithelial cells and lung fibroblast cells, as well as monocyte-derived macrophages and dendritic cells. High secretions and persistent increase of CXCL10 in MERS-CoV patients have been also associated with severity of disease. To our knowledge, this is the first report showing that the MERS-N protein is one of the contributing factors for CXCL10 up-regulation during infection. In addition, our results showed that a fragment consisting of residues 196-413 in MERS-N is sufficient to up-regulate CXCL10, while the N-terminal domain and serine-arginine (SR)-rich motif of MERS-N do not play a role in this up-regulation.
Collapse
|
79
|
Castino G, Guillemet M, Joly A, Vignon A. Le système CRISPR/Cas : un outil d’édition des génomes pour le développement de modèles animaux d’infections virales. Med Sci (Paris) 2018; 34:403-405. [DOI: 10.1051/medsci/20183405011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
80
|
Galasiti Kankanamalage AC, Kim Y, Damalanka VC, Rathnayake AD, Fehr AR, Mehzabeen N, Battaile KP, Lovell S, Lushington GH, Perlman S, Chang KO, Groutas WC. Structure-guided design of potent and permeable inhibitors of MERS coronavirus 3CL protease that utilize a piperidine moiety as a novel design element. Eur J Med Chem 2018; 150:334-346. [PMID: 29544147 PMCID: PMC5891363 DOI: 10.1016/j.ejmech.2018.03.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 01/17/2023]
Abstract
There are currently no approved vaccines or small molecule therapeutics available for the prophylaxis or treatment of Middle East Respiratory Syndrome coronavirus (MERS-CoV) infections. MERS-CoV 3CL protease is essential for viral replication; consequently, it is an attractive target that provides a potentially effective means of developing small molecule therapeutics for combatting MERS-CoV. We describe herein the structure-guided design and evaluation of a novel class of inhibitors of MERS-CoV 3CL protease that embody a piperidine moiety as a design element that is well-suited to exploiting favorable subsite binding interactions to attain optimal pharmacological activity and PK properties. The mechanism of action of the compounds and the structural determinants associated with binding were illuminated using X-ray crystallography.
Collapse
Affiliation(s)
| | - Yunjeong Kim
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Vishnu C Damalanka
- Department of Chemistry, Wichita State University, Wichita, KS 67260, USA
| | - Athri D Rathnayake
- Department of Chemistry, Wichita State University, Wichita, KS 67260, USA
| | - Anthony R Fehr
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Nurjahan Mehzabeen
- Protein Structure Laboratory, The University of Kansas, Lawrence, KS 66047, USA
| | - Kevin P Battaile
- IMCA-CAT, Hauptman-Woodward Medical Research Institute, APS Argonne National Laboratory, Argonne, IL 60439, USA
| | - Scott Lovell
- Protein Structure Laboratory, The University of Kansas, Lawrence, KS 66047, USA
| | | | - Stanley Perlman
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - William C Groutas
- Department of Chemistry, Wichita State University, Wichita, KS 67260, USA.
| |
Collapse
|
81
|
Jiang Y, Zhao G, Song N, Li P, Chen Y, Guo Y, Li J, Du L, Jiang S, Guo R, Sun S, Zhou Y. Blockade of the C5a-C5aR axis alleviates lung damage in hDPP4-transgenic mice infected with MERS-CoV. Emerg Microbes Infect 2018; 7:77. [PMID: 29691378 PMCID: PMC5915580 DOI: 10.1038/s41426-018-0063-8] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/13/2018] [Accepted: 02/25/2018] [Indexed: 02/07/2023]
Abstract
The pathogenesis of highly pathogenic Middle East respiratory syndrome coronavirus (MERS-CoV) remains poorly understood. In a previous study, we established an hDPP4-transgenic (hDPP4-Tg) mouse model in which MERS-CoV infection causes severe acute respiratory failure and high mortality accompanied by an elevated secretion of cytokines and chemokines. Since excessive complement activation is an important factor that contributes to acute lung injury after viral infection, in this study, we investigated the role of complement in MERS-CoV-induced lung damage. Our study showed that complement was excessively activated in MERS-CoV-infected hDPP4-Tg mice through observations of increased concentrations of the C5a and C5b-9 complement activation products in sera and lung tissues, respectively. Interestingly, blocking C5a production by targeting its receptor, C5aR, alleviated lung and spleen tissue damage and reduced inflammatory responses. More importantly, anti-C5aR antibody treatment led to decreased viral replication in lung tissues. Furthermore, compared with the sham treatment control, apoptosis of splenic cells was less pronounced in the splenic white pulp of treated mice, and greater number of proliferating splenic cells, particularly in the red pulp, was observed. These data indicate that (1) dysregulated host immune responses contribute to the severe outcome of MERS; (2) excessive complement activation, triggered by MERS-CoV infection, promote such dysregulation; and (3) blockade of the C5a-C5aR axis lead to the decreased tissue damage induced by MERS-CoV infection, as manifested by reduced apoptosis and T cell regeneration in the spleen. Therefore, the results of this study suggest a new strategy for clinical intervention and adjunctive treatment in MERS-CoV cases.
Collapse
Affiliation(s)
- Yuting Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Nianping Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Pei Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yuehong Chen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Junfeng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | | | - Shihui Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| |
Collapse
|
82
|
Cockrell AS, Beall A, Yount B, Baric R. Efficient Reverse Genetic Systems for Rapid Genetic Manipulation of Emergent and Preemergent Infectious Coronaviruses. Methods Mol Biol 2018; 1602:59-81. [PMID: 28508214 PMCID: PMC7120940 DOI: 10.1007/978-1-4939-6964-7_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Emergent and preemergent coronaviruses (CoVs) pose a global threat that requires immediate intervention. Rapid intervention necessitates the capacity to generate, grow, and genetically manipulate infectious CoVs in order to rapidly evaluate pathogenic mechanisms, host and tissue permissibility, and candidate antiviral therapeutic efficacy. CoVs encode the largest viral RNA genomes at about 28-32,000 nucleotides in length, and thereby complicate efficient engineering of the genome. Deconstructing the genome into manageable fragments affords the plasticity necessary to rapidly introduce targeted genetic changes in parallel and assort mutated fragments while maximizing genome stability over time. In this protocol we describe a well-developed reverse genetic platform strategy for CoVs that is comprised of partitioning the viral genome into 5-7 independent DNA fragments (depending on the CoV genome), each subcloned into a plasmid for increased stability and ease of genetic manipulation and amplification. Coronavirus genomes are conveniently partitioned by introducing type IIS or IIG restriction enzyme recognition sites that confer directional cloning. Since each restriction site leaves a unique overhang between adjoining fragments, reconstruction of the full-length genome can be achieved through a standard DNA ligation comprised of equal molar ratios of each fragment. Using this method, recombinant CoVs can be rapidly generated and used to investigate host range, gene function, pathogenesis, and candidate therapeutics for emerging and preemergent CoVs both in vitro and in vivo.
Collapse
Affiliation(s)
- Adam S Cockrell
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Anne Beall
- Departments of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Boyd Yount
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Ralph Baric
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA. .,Departments of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
83
|
Abstract
Since the identification of the first patients with Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012, over 1,600 cases have been reported as of February 2016. Most cases have occurred in Saudi Arabia or in other countries on or near the Arabian Peninsula, but travel-associated cases have also been seen in countries outside the Arabian Peninsula. MERS-CoV causes a severe respiratory illness in many patients, with a case fatality rate as high as 40%, although when contacts are investigated, a significant proportion of patients are asymptomatic or only have mild symptoms. At this time, no vaccines or treatments are available. Epidemiological and other data suggest that the source of most primary cases is exposure to camels. Person-to-person transmission occurs in household and health care settings, although sustained and efficient person-to-person transmission has not been observed. Strict adherence to infection control recommendations has been associated with control of previous outbreaks. Vigilance is needed because genomic changes in MERS-CoV could result in increased transmissibility, similar to what was seen in severe acute respiratory syndrome coronavirus (SARS-CoV).
Collapse
|
84
|
Rapid development of vaccines against emerging pathogens: The replication-deficient simian adenovirus platform technology. Vaccine 2017; 35:4461-4464. [PMID: 28576573 PMCID: PMC5571606 DOI: 10.1016/j.vaccine.2017.04.085] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 04/13/2017] [Accepted: 04/26/2017] [Indexed: 12/11/2022]
Abstract
Despite the fact that there had been multiple small outbreaks of Ebola Virus Disease, when a large outbreak occurred in 2014 there were no vaccines or drugs available for use. Clinical development of multiple candidate vaccines was then initiated in parallel with attempts to contain the outbreak but only one vaccine was eventually tested in a phase III trial. In order to be better prepared for future outbreaks of known human pathogens, platform technologies to accelerate vaccine development should be employed, allowing vaccine developers to take advantage of detailed knowledge of the vaccine platform and facilitating rapid progress to clinical trials and eventually to vaccine stockpiles. This review gives an example of one such vaccine platform, replication-deficient simian adenoviruses, and describes progress in human and livestock vaccine development for three outbreak pathogens, Ebola virus, Rift Valley Fever Virus and Middle East Respiratory Syndrome Coronavirus.
Collapse
|
85
|
Camp JV, Jonsson CB. A Role for Neutrophils in Viral Respiratory Disease. Front Immunol 2017; 8:550. [PMID: 28553293 PMCID: PMC5427094 DOI: 10.3389/fimmu.2017.00550] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 04/24/2017] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are immune cells that are well known to be present during many types of lung diseases associated with acute respiratory distress syndrome (ARDS) and may contribute to acute lung injury. Neutrophils are poorly studied with respect to viral infection, and specifically to respiratory viral disease. Influenza A virus (IAV) infection is the cause of a respiratory disease that poses a significant global public health concern. Influenza disease presents as a relatively mild and self-limiting although highly pathogenic forms exist. Neutrophils increase in the respiratory tract during infection with mild seasonal IAV, moderate and severe epidemic IAV infection, and emerging highly pathogenic avian influenza (HPAI). During severe influenza pneumonia and HPAI infection, the number of neutrophils in the lower respiratory tract is correlated with disease severity. Thus, comparative analyses of the relationship between IAV infection and neutrophils provide insights into the relative contribution of host and viral factors that contribute to disease severity. Herein, we review the contribution of neutrophils to IAV disease pathogenesis and to other respiratory virus infections.
Collapse
Affiliation(s)
- Jeremy V Camp
- Institute of Virology, University of Veterinary Medicine at Vienna, Vienna, Austria
| | - Colleen B Jonsson
- Department of Microbiology, University of Tennessee-Knoxville, Knoxville, TN, USA
| |
Collapse
|
86
|
Mouse-adapted MERS coronavirus causes lethal lung disease in human DPP4 knockin mice. Proc Natl Acad Sci U S A 2017; 114:E3119-E3128. [PMID: 28348219 DOI: 10.1073/pnas.1619109114] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The Middle East respiratory syndrome (MERS) emerged in Saudi Arabia in 2012, caused by a zoonotically transmitted coronavirus (CoV). Over 1,900 cases have been reported to date, with ∼36% fatality rate. Lack of autopsies from MERS cases has hindered understanding of MERS-CoV pathogenesis. A small animal model that develops progressive pulmonary manifestations when infected with MERS-CoV would advance the field. As mice are restricted to infection at the level of DPP4, the MERS-CoV receptor, we generated mice with humanized exons 10-12 of the mouse Dpp4 locus. Upon inoculation with MERS-CoV, human DPP4 knockin (KI) mice supported virus replication in the lungs, but developed no illness. After 30 serial passages through the lungs of KI mice, a mouse-adapted virus emerged (MERSMA) that grew in lungs to over 100 times higher titers than the starting virus. A plaque-purified MERSMA clone caused weight loss and fatal infection. Virus antigen was observed in airway epithelia, pneumocytes, and macrophages. Pathologic findings included diffuse alveolar damage with pulmonary edema and hyaline membrane formation associated with accumulation of activated inflammatory monocyte-macrophages and neutrophils in the lungs. Relative to the parental MERS-CoV, MERSMA viruses contained 13-22 mutations, including several within the spike (S) glycoprotein gene. S-protein mutations sensitized viruses to entry-activating serine proteases and conferred more rapid entry kinetics. Recombinant MERSMA bearing mutant S proteins were more virulent than the parental virus in hDPP4 KI mice. The hDPP4 KI mouse and the MERSMA provide tools to investigate disease causes and develop new therapies.
Collapse
|
87
|
Elfiky AA, Mahdy SM, Elshemey WM. Quantitative structure-activity relationship and molecular docking revealed a potency of anti-hepatitis C virus drugs against human corona viruses. J Med Virol 2017; 89:1040-1047. [PMID: 27864902 PMCID: PMC7167072 DOI: 10.1002/jmv.24736] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 12/30/2022]
Abstract
A number of human coronaviruses (HCoVs) were reported in the last and present centuries. Some outbreaks of which (eg, SARS and MERS CoVs) caused the mortality of hundreds of people worldwide. The problem of finding a potent drug against HCoV strains lies in the inability of finding a drug that stops the viral replication through inhibiting its important proteins. In spite of its limited efficacy and potential side effects, Ribavirin is extensively used as a first choice against HCoVs. Therefore, scientists reverted towards the investigation of different drugs that can more specifically target proteins. In this study, four anti‐HCV drugs (one approved by FDA and others under clinical trials) are tested against HCoV polymerases. Quantitative Structure‐Activity Relationship (QSAR) and molecular docking are both used to compare the performance of the selected nucleotide inhibitors to their parent nucleotides and Ribavirin. Both QSAR and molecular docking showed that IDX‐184 is superior compared to Ribavirin against MERS CoV, a result that was also reported for HCV. MK‐0608 showed a performance that is comparable to Ribavirin. We strongly suggest an in vitro study on the potency of these two drugs against MERS CoV.
Collapse
Affiliation(s)
- Abdo A Elfiky
- Faculty of Science, Department of Biophysics, Cairo University, Giza, Egypt.,The Abdus Salam International Center for Theoretical Physics ICTP, Trieste, Italy
| | - Samah M Mahdy
- Faculty of Science, Department of Biophysics, Cairo University, Giza, Egypt.,National Museum of Egyptian Civilization (NMEC), Ain Elsira-Elfustat, Cairo, Egypt
| | - Wael M Elshemey
- Faculty of Science, Department of Biophysics, Cairo University, Giza, Egypt
| |
Collapse
|
88
|
Choi J, Kim MG, Oh YK, Kim YB. Progress of Middle East respiratory syndrome coronavirus vaccines: a patent review. Expert Opin Ther Pat 2017; 27:721-731. [PMID: 28121202 DOI: 10.1080/13543776.2017.1281248] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Middle East respiratory syndrome coronavirus (MERS-CoV) has emerged as a new pathogen, causing severe complications and a high case fatality rate. No direct treatments are available as yet, highlighting the importance of prevention through suitable vaccination regimes. The viral spike (S) protein has been characterized as a key target antigen for vaccines. In particular, S protein domains have been utilized to produce high titers of neutralizing antibodies. Areas covered: Since the first report of MERS-CoV infection, a limited number of MERS-CoV-specific patents have been filed. Patents related to MERS-CoV are categorized into three areas: treatments, antibodies, and vaccines (receptor-related). This review mainly focuses on the types and efficacies of vaccines, briefly covering treatments and antibodies against the virus. MERS-CoV vaccine forms and delivery systems, together with comparable development strategies against SARS-CoV are additionally addressed. Expert opinion: Vaccines must be combined with delivery systems, administration routes, and adjuvants to maximize T-cell responses as well as neutralizing antibody production. High immune responses require further study in animal models, such as human receptor-expressing mice, non-human primates, and camels. Such a consideration of integrated actions should contribute to the rapid development of vaccines against MERS-CoV and related coronaviruses.
Collapse
Affiliation(s)
- Jiwon Choi
- a College of Animal Bioscience & Technology , Konkuk University , Seoul , Republic of Korea
| | - Mi-Gyeong Kim
- b College of Pharmacy , Seoul National University , Seoul , Republic of Korea
| | - Yu-Kyoung Oh
- b College of Pharmacy , Seoul National University , Seoul , Republic of Korea
| | - Young Bong Kim
- a College of Animal Bioscience & Technology , Konkuk University , Seoul , Republic of Korea
| |
Collapse
|
89
|
Abstract
The first major pandemic of the new millennium that arose from southern China in 2002 was of zoonotic origin from wild game animals, called severe acute respiratory syndrome [SARS]. The culprit was determined to be a coronavirus of animal origin [SARS-CoV]. The discovery of the SARS-CoV, which caused an outbreak of >8000 people in >30 countries with fatality of about 10%, resulted in intense search for novel coronaviruses with potentially high pathogenicity. Ten years later after the SARS pandemic, another novel coronavirus crossed the species barrier from bats to humans through an intermediate camel host, to produce a severe lower respiratory infection labeled the Middle East respiratory syndrome [MERS]. A novel coronavirus [MERS-CoV] was first identified in September 2012, from patients who resided or traveled to Saudi Arabia. The MERS-CoV spread through contacts with camel and subsequently from human to human via droplet transmission. MERS cases occurred in several other countries including in Europe and the United States, mainly from residence or travel to the Arabian Peninsula, but was not of pandemic potential. However, in the spring of 2015, a MERS outbreak started in South Korea which was initiated by a returning traveler from Saudi Arabia, and subsequently secondary infection of over 186 local residents occurred. Recent estimate in May 2015 indicates that the MERS-CoV have afflicted 1167 patients with MERS worldwide with 479 deaths [41% fatality]. Thus MERS is more deadly than SARS but appears to be less contagious. However, unlike SARS which has not reappeared since 2002–2003, MERS-CoV have the potential to cause sporadic or local outbreaks for many years as the virus may now be entrenched endemically in dromedary camels of the Middle East.
Collapse
|
90
|
Cockrell AS, Baric RS. An effective DNA vaccine platform for Middle East respiratory syndrome coronavirus. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:499. [PMID: 28149861 DOI: 10.21037/atm.2016.11.40] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Adam S Cockrell
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA; Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
91
|
Min J, Cella E, Ciccozzi M, Pelosi A, Salemi M, Prosperi M. The global spread of Middle East respiratory syndrome: an analysis fusing traditional epidemiological tracing and molecular phylodynamics. Glob Health Res Policy 2016; 1:14. [PMID: 29202063 PMCID: PMC5693564 DOI: 10.1186/s41256-016-0014-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/14/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Since its discovery in 2012, over 1700 confirmed cases of Middle East Respiratory Syndrome (MERS) have been documented worldwide and more than a third of those cases have died. While the greatest number of cases has occurred in Saudi Arabia, the recent export of MERS-coronavirus (MERS-CoV) to Republic of Korea showed that a pandemic is a possibility that cannot be ignored. Due to the deficit of knowledge in transmission methodology, targeted treatment and possible vaccines, understanding this virus should be a priority. Our aim was to combine epidemiological data from literature with genetic information from viruses sequenced around the world to present a phylodynamic picture of MERS spread molecular level to global scale. METHODS We performed a qualitative meta-analysis of all laboratory confirmed cases worldwide to date based on literature, with emphasis on international transmission and healthcare associated infections. In parallel, we used publicly available MERS-CoV genomes from GenBank to create a phylogeographic tree, detailing geospatial timeline of viral evolution. RESULTS Several healthcare associated outbreaks starting with the retrospectively identified hospital outbreak in Jordan to the most recent outbreak in Riyadh, Saudi Arabia have occurred. MERS has also crossed many oceans, entering multiple nations in eight waves between 2012 and 2015. In this paper, the spatiotemporal history of MERS cases, as documented epidemiologically, was examined by Bayesian phylogenetic analysis. Distribution of sequences into geographic clusters and interleaving of MERS-CoV sequences from camels among those isolated from humans indicated that multiple zoonotic introductions occurred in endemic nations. We also report a summary of basic reproduction numbers for MERS-CoV in humans and camels. CONCLUSION Together, these analyses can help us identify factors associated with viral evolution and spread as well as establish efficacy of infection control measures. The results are especially pertinent to countries without current MERS-CoV endemic, since their unfamiliarity makes them particularly susceptible to uncontrollable spread of a virus that may be imported by travelers.
Collapse
Affiliation(s)
- Jae Min
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, 2004 Mowry Rd, Gainesville, FL 32610-0231 USA
| | - Eleonora Cella
- Department of Infectious, Parasitic and Immune-mediated Diseases, National Institute of Health, Viale Regina Elena, 299, 00161 Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
- Department of Pathology, Immunology and Laboratory Medicine, Emerging Pathogens Institute, University of Florida, 2055 Mowry Rd, Gainesville, FL 32611 USA
| | - Massimo Ciccozzi
- Department of Infectious, Parasitic and Immune-mediated Diseases, National Institute of Health, Viale Regina Elena, 299, 00161 Rome, Italy
- Department of Clinical Pathology and Microbiology Laboratory, University of Biomedical Campus, Via Alvaro del Portillo, 21, Rome, Italy
| | - Antonello Pelosi
- Department of Infectious, Parasitic and Immune-mediated Diseases, National Institute of Health, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Marco Salemi
- Department of Pathology, Immunology and Laboratory Medicine, Emerging Pathogens Institute, University of Florida, 2055 Mowry Rd, Gainesville, FL 32611 USA
| | - Mattia Prosperi
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, 2004 Mowry Rd, Gainesville, FL 32610-0231 USA
| |
Collapse
|
92
|
Use of the Human Coronavirus 2012 ( MERS) GeneSig kit for MERS-CoV detection. GENE REPORTS 2016; 4:67-69. [PMID: 32289095 PMCID: PMC7104146 DOI: 10.1016/j.genrep.2016.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 04/05/2016] [Accepted: 04/05/2016] [Indexed: 12/04/2022]
Abstract
Introduction Mortality due to MERS-CoV infection is common especially among immunocompromised patients. The pathogenesis and the transmission mode of this virus are still not well understood. The name of the virus is derived from the area of its appearance and the genomic sequence that was used in the development of qRT-PCR assays for MERS-CoV detection was retrieved from the first detected case isolate. The employed assays target various regions including the area upstream of the envelope gene (upE) that is used for screening and the open reading frames (ORF) 1a and 1b used for confirmation. Aim This study assesses the use of a MERS-CoV specific assay for screening of respiratory samples in anticipation of the possible spread of the virus in the region. Methods 46 respiratory specimens were tested using the qualitative one-step qRT-PCR GeneSig Human Coronavirus 2012 (MERS) kit (PrimerDesign™). Results Out of the 46 tested samples, 45 were negative for MERS-CoV and one sample was found MERS-CoV positive. Conclusion The GeneSig Human Coronavirus 2012 (MERS) kit is very useful for the screening of suspected respiratory cases in the Middle East area as well as other regions. Mortality due to MERS-CoV infection is common especially among immunocompromised patients Several essays have been utilized to detect the virus in an attempt to diagnose it as early as possible The Human Coronavirus 2012 GeneSig kit is a new kit that can be used for MERS-CoV detection in respiratory samples
Collapse
|
93
|
Abstract
Coronaviruses (CoVs) are enveloped RNA viruses that infect birds, mammals, and humans. Infections caused by human coronaviruses (hCoVs) are mostly associated with the respiratory, enteric, and nervous systems. The hCoVs only occasionally induce lower respiratory tract disease, including bronchitis, bronchiolitis, and pneumonia. In 2002 to 2003, a global outbreak of severe acute respiratory syndrome (SARS) was the seminal detection of a novel CoV (SARS-CoV). A decade later (June 2012), another novel CoV was implicated as the cause of Middle East respiratory syndrome (MERS) in Saudi Arabia. Although bats might serve as a reservoir of MERS-CoV, it is unlikely that they are the direct source for most human cases. Severe lines of evidence suggest that dromedary camels have been the major cause of transmission to humans. The emergence of MERS-CoV has triggered serious concerns about the potential for a widespread outbreak. All MERS cases were linked directly or indirectly to the Middle East region including Saudi Arabia, Jordan, Qatar, Oman, Kuwait, and UAE. MERS cases have also been reported in the later phases in the United Kingdom, France, Germany, Italy, Spain, and Tunisia. Most of these MERS cases were linked with the Middle East. The high mortality rates in family-based and hospital-based outbreaks were reported among patients with comorbidities such as diabetes and renal failure. MERS-CoV causes an acute, highly lethal pneumonia and renal dysfunction. The major complications reported in fatal cases are hyperkalemia with associated ventricular tachycardia, disseminated intravascular coagulation, pericarditis, and multiorgan failure. The case-fatality rate seems to be higher for MERS-CoV (around 30%) than for SARS-CoV (9.6%). The combination regimen of type 1 interferon + lopinavir/ritonavir is considered as the first-line therapy for MERS. Antiviral treatment is generally recommended for 10 to 14 days in patients with MERS-CoV infection. Convalescent plasma therapy has shown some efficacy among patients refractory to antiviral drugs if administered within 2 weeks of the onset of the disease.
Collapse
Affiliation(s)
- Sunit K Singh
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh, India
| |
Collapse
|
94
|
Abstract
Human rhinovirus (HRV) and coronavirus (HCoV) infections are associated with both upper respiratory tract illness (“the common cold”) and lower respiratory tract illness (pneumonia). New species of HRVs and HCoVs have been diagnosed in the past decade. More sensitive diagnostic tests such as reverse transcription-polymerase chain reaction have expanded our understanding of the role these viruses play in both immunocompetent and immunosuppressed hosts. Recent identification of severe acute respiratory syndrome and Middle East respiratory syndrome viruses causing serious respiratory illnesses has led to renewed efforts for vaccine development. The role these viruses play in patients with chronic lung disease such as asthma makes the search for antiviral agents of increased importance.
Collapse
Affiliation(s)
- Stephen B Greenberg
- Department of Medicine, Ben Taub Hospital, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
95
|
Meyerholz DK. Modeling Emergent Diseases: Lessons From Middle East Respiratory Syndrome. Vet Pathol 2016; 53:517-8. [PMID: 27000399 DOI: 10.1177/0300985816634811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- D K Meyerholz
- University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
96
|
Middle East Respiratory Syndrome Coronavirus: What Does a Radiologist Need to Know? AJR Am J Roentgenol 2016; 206:1193-201. [PMID: 26998804 DOI: 10.2214/ajr.15.15363] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE The overarching goal of this article is to provide radiologists with the most up-to-date information regarding the underlying epidemiology, pathophysiology, clinical features, and imaging findings related to Middle East respiratory syndrome coronavirus (MERS-CoV), a potentially deadly new infection. CONCLUSION An increased awareness of MERS-CoV and an understanding of the radiologic features of MERS-CoV can improve the early assessment and monitoring of this new infection. Radiologists can provide information based on chest radiographic and CT scores that can be helpful for patient management and predicting prognosis.
Collapse
|
97
|
Virlogeux V, Yang J, Fang VJ, Feng L, Tsang TK, Jiang H, Wu P, Zheng J, Lau EHY, Qin Y, Peng Z, Peiris JSM, Yu H, Cowling BJ. Association between the Severity of Influenza A(H7N9) Virus Infections and Length of the Incubation Period. PLoS One 2016; 11:e0148506. [PMID: 26885816 PMCID: PMC4757028 DOI: 10.1371/journal.pone.0148506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/19/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In early 2013, a novel avian-origin influenza A(H7N9) virus emerged in China, and has caused sporadic human infections. The incubation period is the delay from infection until onset of symptoms, and varies from person to person. Few previous studies have examined whether the duration of the incubation period correlates with subsequent disease severity. METHODS AND FINDINGS We analyzed data of period of exposure on 395 human cases of laboratory-confirmed influenza A(H7N9) virus infection in China in a Bayesian framework using a Weibull distribution. We found a longer incubation period for the 173 fatal cases with a mean of 3.7 days (95% credibility interval, CrI: 3.4-4.1), compared to a mean of 3.3 days (95% CrI: 2.9-3.6) for the 222 non-fatal cases, and the difference in means was marginally significant at 0.47 days (95% CrI: -0.04, 0.99). There was a statistically significant correlation between a longer incubation period and an increased risk of death after adjustment for age, sex, geographical location and underlying medical conditions (adjusted odds ratio 1.70 per day increase in incubation period; 95% credibility interval 1.47-1.97). CONCLUSIONS We found a significant association between a longer incubation period and a greater risk of death among human H7N9 cases. The underlying biological mechanisms leading to this association deserve further exploration.
Collapse
Affiliation(s)
- Victor Virlogeux
- Department of Biology, Ecole Normale Supérieure de Lyon, 15 parvis René Descartes, 69007 Lyon, France
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong Special Administrative Region, China
| | - Juan Yang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-Warning on Infectious Disease, Chinese Center for Disease Control and Prevention, 155# Changbai Road, Beijing, 102206, China
| | - Vicky J. Fang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong Special Administrative Region, China
| | - Luzhao Feng
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-Warning on Infectious Disease, Chinese Center for Disease Control and Prevention, 155# Changbai Road, Beijing, 102206, China
| | - Tim K. Tsang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong Special Administrative Region, China
| | - Hui Jiang
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-Warning on Infectious Disease, Chinese Center for Disease Control and Prevention, 155# Changbai Road, Beijing, 102206, China
| | - Peng Wu
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong Special Administrative Region, China
| | - Jiandong Zheng
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-Warning on Infectious Disease, Chinese Center for Disease Control and Prevention, 155# Changbai Road, Beijing, 102206, China
| | - Eric H. Y. Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong Special Administrative Region, China
| | - Ying Qin
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-Warning on Infectious Disease, Chinese Center for Disease Control and Prevention, 155# Changbai Road, Beijing, 102206, China
| | - Zhibin Peng
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-Warning on Infectious Disease, Chinese Center for Disease Control and Prevention, 155# Changbai Road, Beijing, 102206, China
| | - J. S. Malik Peiris
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong Special Administrative Region, China
| | - Hongjie Yu
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-Warning on Infectious Disease, Chinese Center for Disease Control and Prevention, 155# Changbai Road, Beijing, 102206, China
- * E-mail:
| | - Benjamin J. Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong Special Administrative Region, China
| |
Collapse
|
98
|
Zhao G, Jiang Y, Qiu H, Gao T, Zeng Y, Guo Y, Yu H, Li J, Kou Z, Du L, Tan W, Jiang S, Sun S, Zhou Y. Multi-Organ Damage in Human Dipeptidyl Peptidase 4 Transgenic Mice Infected with Middle East Respiratory Syndrome-Coronavirus. PLoS One 2015; 10:e0145561. [PMID: 26701103 PMCID: PMC4689477 DOI: 10.1371/journal.pone.0145561] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/04/2015] [Indexed: 01/08/2023] Open
Abstract
The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) causes severe acute respiratory failure and considerable extrapumonary organ dysfuction with substantial high mortality. For the limited number of autopsy reports, small animal models are urgently needed to study the mechanisms of MERS-CoV infection and pathogenesis of the disease and to evaluate the efficacy of therapeutics against MERS-CoV infection. In this study, we developed a transgenic mouse model globally expressing codon-optimized human dipeptidyl peptidase 4 (hDPP4), the receptor for MERS-CoV. After intranasal inoculation with MERS-CoV, the mice rapidly developed severe pneumonia and multi-organ damage, with viral replication being detected in the lungs on day 5 and in the lungs, kidneys and brains on day 9 post-infection. In addition, the mice exhibited systemic inflammation with mild to severe pneumonia accompanied by the injury of liver, kidney and spleen with neutrophil and macrophage infiltration. Importantly, the mice exhibited symptoms of paralysis with high viral burden and viral positive neurons on day 9. Taken together, this study characterizes the tropism of MERS-CoV upon infection. Importantly, this hDPP4-expressing transgenic mouse model will be applicable for studying the pathogenesis of MERS-CoV infection and investigating the efficacy of vaccines and antiviral agents designed to combat MERS-CoV infection.
Collapse
Affiliation(s)
- Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yuting Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Hongjie Qiu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Tongtong Gao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yang Zeng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Hong Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Junfeng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Zhihua Kou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, 10065, United States of America
| | - Wenjie Tan
- Key Laboratory of Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, China CDC, Beijing, 102206, China
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, 10065, United States of America
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Shihui Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
- * E-mail: (Y. Zhou); (SS)
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
- * E-mail: (Y. Zhou); (SS)
| |
Collapse
|
99
|
Zhou J, Chu H, Chan JFW, Yuen KY. Middle East respiratory syndrome coronavirus infection: virus-host cell interactions and implications on pathogenesis. Virol J 2015; 12:218. [PMID: 26690369 PMCID: PMC4687146 DOI: 10.1186/s12985-015-0446-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/01/2015] [Indexed: 12/27/2022] Open
Abstract
Middle-East Respiratory Syndrome coronavirus (MERS-CoV) was identified to cause severe respiratory infection in humans since 2012. The continuing MERS epidemic with a case-fatality of more than 30 % poses a major threat to public health worldwide. Currently, the pathogenesis of human MERS-CoV infection remains poorly understood. We reviewed experimental findings from human primary cells and ex vivo human lung tissues, as well as those from animal studies, so as to understand the pathogenesis and high case-fatality of MERS. Human respiratory epithelial cells are highly susceptible to MERS-CoV and can support productive viral replication. However, the induction of antiviral cytokines and proinflammatory cytokines/chemokines are substantially dampened in the infected epithelial cells, due to the antagonistic mechanisms evolved by the virus. MERS-CoV can readily infect and robustly replicate in human macrophages and dendritic cells, triggering the aberrant production of proinflammatory cytokines/chemokines. MERS-CoV can also effectively infect human primary T cells and induce massive apoptosis in these cells. Although data from clinical, in vitro and ex vivo studies suggested the potential for virus dissemination, extrapulmonary involvement in MERS patients has not been ascertained due to the lack of autopsy study. In MERS-CoV permissive animal models, although viral RNA can be detected from multiple organs of the affected animals, the brain of human DPP4-transgenic mouse was the only extrapulmonary organ from which the infectious virus can be recovered. More research findings on the pathogenesis of MERS and the tissue tropisms of MERS-CoV may help to improve the treatment and infection control of MERS.
Collapse
Affiliation(s)
- Jie Zhou
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, China. .,Department of Microbiology, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong Special Administrative Region, China. .,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, China. .,Department of Microbiology, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong Special Administrative Region, China. .,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, China. .,Department of Microbiology, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong Special Administrative Region, China. .,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong Special Administrative Region, China. .,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong Special Administrative Region, China. .,Department of Microbiology, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong Special Administrative Region, China. .,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong Special Administrative Region, China. .,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
100
|
Warnes SL, Little ZR, Keevil CW. Human Coronavirus 229E Remains Infectious on Common Touch Surface Materials. mBio 2015; 6:e01697-15. [PMID: 26556276 PMCID: PMC4659470 DOI: 10.1128/mbio.01697-15] [Citation(s) in RCA: 302] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 10/13/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED The evolution of new and reemerging historic virulent strains of respiratory viruses from animal reservoirs is a significant threat to human health. Inefficient human-to-human transmission of zoonotic strains may initially limit the spread of transmission, but an infection may be contracted by touching contaminated surfaces. Enveloped viruses are often susceptible to environmental stresses, but the human coronaviruses responsible for severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) have recently caused increasing concern of contact transmission during outbreaks. We report here that pathogenic human coronavirus 229E remained infectious in a human lung cell culture model following at least 5 days of persistence on a range of common nonbiocidal surface materials, including polytetrafluoroethylene (Teflon; PTFE), polyvinyl chloride (PVC), ceramic tiles, glass, silicone rubber, and stainless steel. We have shown previously that noroviruses are destroyed on copper alloy surfaces. In this new study, human coronavirus 229E was rapidly inactivated on a range of copper alloys (within a few minutes for simulated fingertip contamination) and Cu/Zn brasses were very effective at lower copper concentration. Exposure to copper destroyed the viral genomes and irreversibly affected virus morphology, including disintegration of envelope and dispersal of surface spikes. Cu(I) and Cu(II) moieties were responsible for the inactivation, which was enhanced by reactive oxygen species generation on alloy surfaces, resulting in even faster inactivation than was seen with nonenveloped viruses on copper. Consequently, copper alloy surfaces could be employed in communal areas and at any mass gatherings to help reduce transmission of respiratory viruses from contaminated surfaces and protect the public health. IMPORTANCE Respiratory viruses are responsible for more deaths globally than any other infectious agent. Animal coronaviruses that "host jump" to humans result in severe infections with high mortality, such as severe acute respiratory syndrome (SARS) and, more recently, Middle East respiratory syndrome (MERS). We show here that a closely related human coronavirus, 229E, which causes upper respiratory tract infection in healthy individuals and serious disease in patients with comorbidities, remained infectious on surface materials common to public and domestic areas for several days. The low infectious dose means that this is a significant infection risk to anyone touching a contaminated surface. However, rapid inactivation, irreversible destruction of viral RNA, and massive structural damage were observed in coronavirus exposed to copper and copper alloy surfaces. Incorporation of copper alloy surfaces in conjunction with effective cleaning regimens and good clinical practice could help to control transmission of respiratory coronaviruses, including MERS and SARS.
Collapse
Affiliation(s)
- Sarah L Warnes
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Zoë R Little
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - C William Keevil
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|