1
|
Grycová A, Vyhlídalová B, Dvořák Z. The role of aryl hydrocarbon receptor in antiviral immunity: a focus on RNA viruses. Crit Rev Microbiol 2025:1-15. [PMID: 40299755 DOI: 10.1080/1040841x.2025.2497789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/02/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
Aryl hydrocarbon receptor (AhR) is a ligand-dependent transcriptional factor that is activated by a plethora of exogenous and endogenous compounds, including environmental pollutants, drugs, and microbial metabolites. The AhR plays an important role in modulating immunity. Current findings suggest that AhR activation serves as a mechanism for evasion of host antiviral immune response and promotes viral replication. This review will focus on AhR's role in RNA virus infection because they show high mutation rates compared with DNA viruses, and therefo pose one of the greatest threats to humans in terms of potential pandemic risk. Indeed, they include human immunodeficiency virus (HIV), influenza A virus (IAV), coronaviruses (CoVs), Zika virus, and others. Understanding the mechanisms by which AhR influences the immune response to these viruses is critical for developing effective therapeutic strategies. By focusing on the interplay between AhR signaling and RNA virus infections, this review aims to contribute to the growing body of knowledge regarding host-pathogen interactions and the implications for antiviral immunity.
Collapse
Affiliation(s)
- Aneta Grycová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| | - Barbora Vyhlídalová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czech Republic
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
2
|
Li H, Song C, Li Y, Zhang T, Yang X, Wang H. Genome-wide CRISPR screen reveals host factors for gama- and delta-coronavirus infection in Huh7 cells. Int J Biol Macromol 2025; 304:140728. [PMID: 39920943 DOI: 10.1016/j.ijbiomac.2025.140728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Genome-wide CRISPR screening has emerged as a powerful tool for identifying novel host factors involved in viral infections. In recent years, host factors for several Alpha- and Beta-coronaviruses have been systematically screened and characterized. However, knowledge regarding Gamma- and Delta-coronavirus infections remains limited. In this study, we conducted genome-scale CRISPR knockout (KO) screening in Huh7 cells infected with infectious bronchitis virus (IBV), a Gamma-coronavirus, and porcine deltacoronavirus (PDCoV), a Delta-coronavirus. We identified known host factors for PDCoV, including APN and TMEM41B. We confirmed that human APN does not serve as a critical host factor for IBV. Notably, SPPL3 was identified as a key factor involved in viral particle entry and S protein-induced syncytium formation through the modulation of cellular N-glycosylation. Furthermore, we performed a meta-analysis integrating all Huh7 cell-based genome-wide CRISPR screens across the four genera of coronaviruses (Alpha-, Beta-, Gamma-, and Delta-coronaviruses). Our analysis highlighted conserved host pathways, particularly those related to proteoglycans, glycoproteins, and vesicle trafficking. TMEM41B, SCAP, and FAM98A emerged as the most frequently targeted host genes. These findings provide valuable insights into the life cycles of IBV and PDCoV infections and facilitate the development of host-directed therapeutic strategies.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Cailiang Song
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yuqing Li
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Tiejun Zhang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Xin Yang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Hongning Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| |
Collapse
|
3
|
Barreira-Silva P, Lian Y, Kaufmann SHE, Moura-Alves P. The role of the AHR in host-pathogen interactions. Nat Rev Immunol 2025; 25:178-194. [PMID: 39415055 DOI: 10.1038/s41577-024-01088-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/18/2024]
Abstract
Host-microorganism encounters take place in many different ways and with different types of outcomes. Three major types of microorganisms need to be distinguished: (1) pathogens that cause harm to the host and must be controlled; (2) environmental microorganisms that can be ignored but must be controlled at higher abundance; and (3) symbiotic microbiota that require support by the host. Recent evidence indicates that the aryl hydrocarbon receptor (AHR) senses and initiates signalling and gene expression in response to a plethora of microorganisms and infectious conditions. It was originally identified as a receptor that binds xenobiotics. However, it was subsequently found to have a critical role in numerous biological processes, including immunity and inflammation and was recently classified as a pattern recognition receptor. Here we review the role of the AHR in host-pathogen interactions, focusing on AHR sensing of different microbial classes, the ligands involved, responses elicited and disease outcomes. Moreover, we explore the therapeutic potential of targeting the AHR in the context of infection.
Collapse
Affiliation(s)
- Palmira Barreira-Silva
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Yilong Lian
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, USA
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pedro Moura-Alves
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
4
|
Andreu S, Ripa I, López-Guerrero JA, Bello-Morales R. Human Coronavirus 229E Uses Clathrin-Mediated Endocytosis as a Route of Entry in Huh-7 Cells. Biomolecules 2024; 14:1232. [PMID: 39456165 PMCID: PMC11505773 DOI: 10.3390/biom14101232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Human coronavirus 229E (HCoV-229E) is an endemic coronavirus responsible for approximately one-third of "common cold" cases. To infect target cells, HCoV-229E first binds to its receptor on the cell surface and then can follow different pathways, entering by direct fusion or by taking advantage of host cell mechanisms such as endocytosis. Based on the role of clathrin, the process can be classified into clathrin-dependent or -independent endocytosis. This study characterizes the role of clathrin-mediated endocytosis (CME) in HCoV-229E infection of the human hepatoma cell line Huh-7. Using specific CME inhibitory drugs, we demonstrated that blocking CME significantly reduces HCoV-229E infection. Additionally, CRISPR/Cas9-mediated knockout of the µ subunit of adaptor protein complex 2 (AP-2) further corroborated the role of CME, as KOs showed over a 50% reduction in viral infection. AP-2 plays an important role in clathrin recruitment and the maturation of clathrin-coated vesicles. Our study also confirmed that in Huh-7 cells, HCoV-229E requires endosomal acidification for successful entry, as viral entry decreased when treated with lysomotropic agents. Furthermore, the colocalization of HCoV-229E with early endosome antigen 1 (EEA-1), only present in early endosomes, suggested that the virus uses an endosomal route for entry. These findings highlight, for the first time, the role of CME in HCoV-229E infection and confirm previous data of the use of the endosomal route at a low pH in the experimental cell model Huh-7. Our results provide new insights into the mechanisms of entry of HCoV-229E and provide a new basis for the development of targeted antiviral therapies.
Collapse
Affiliation(s)
- Sabina Andreu
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), 28049 Madrid, Spain
| | - Inés Ripa
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), 28049 Madrid, Spain
| | - José Antonio López-Guerrero
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), 28049 Madrid, Spain
| | - Raquel Bello-Morales
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas), 28049 Madrid, Spain
| |
Collapse
|
5
|
Cerracchio C, Del Sorbo L, Serra F, Staropoli A, Amoroso MG, Vinale F, Fiorito F. Fungal metabolite 6-pentyl-α-pyrone reduces canine coronavirus infection. Heliyon 2024; 10:e28351. [PMID: 38545179 PMCID: PMC10966688 DOI: 10.1016/j.heliyon.2024.e28351] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 11/11/2024] Open
Abstract
Canine coronavirus (CCoV) can produce a self-limited enteric disease in dogs but, because of notable biological plasticity of coronaviruses (CoVs), numerous mutations as well as recombination events happen leading to the emergence of variants often more dangerous for both animals and humans. Indeed, the emergence of new canine-feline recombinant alphacoronaviruses, recently isolated from humans, highlight the cross-species transmission potential of CoVs. Consequently, new effective antiviral agents are required to treat CoV infections. Among the candidates for the development of drugs against CoVs infection, fungal secondary metabolites (SMs) represent an important source to investigate. Herein, antiviral ability of 6-pentyl-α-pyrone (6 PP), a SM obtained by Trichoderma atroviride, was assessed against CCoV. During in vitro infection, nontoxic concentration of 6 PP significantly increased cell viability, reduced morphological signs of cell death, and inhibited viral replication of CCoV. In addition, we found a noticeable lessening in the expression of aryl hydrocarbon receptor (AhR), a strategic modulator of CoVs infection. Overall, due to the variety of their chemical and biological properties, fungal SMs can decrease the replication of CoVs, thus identifying a suitable in vitro model to screen for potential drugs against CoVs, using a reference strain of CCoV (S/378), non-pathogenic for humans.
Collapse
Affiliation(s)
- Claudia Cerracchio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | - Luca Del Sorbo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | - Francesco Serra
- Department of Animal Health, Unit of Virology, Istituto Zooprofilattico del Mezzogiorno, Portici, 80055 Naples, Italy
| | - Alessia Staropoli
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Naples, Italy
- Institute for Sustainable Plant Protection, National Research Council, Portici, Naples, Italy
| | - Maria Grazia Amoroso
- Department of Animal Health, Unit of Virology, Istituto Zooprofilattico del Mezzogiorno, Portici, 80055 Naples, Italy
| | - Francesco Vinale
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, Portici, Naples, Italy
| | - Filomena Fiorito
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, Portici, Naples, Italy
| |
Collapse
|
6
|
Gallucci L, Bazire J, Davidson AD, Shytaj IL. Broad-spectrum antiviral activity of two structurally analogous CYP3A inhibitors against pathogenic human coronaviruses in vitro. Antiviral Res 2024; 221:105766. [PMID: 38042417 DOI: 10.1016/j.antiviral.2023.105766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/19/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Coronaviruses pose a permanent risk of outbreaks, with three highly pathogenic species and strains (SARS-CoV, MERS-CoV, SARS-CoV-2) having emerged in the last twenty years. Limited antiviral therapies are currently available and their efficacy in randomized clinical trials enrolling SARS-CoV-2 patients has not been consistent, highlighting the need for more potent treatments. We previously showed that cobicistat, a clinically approved inhibitor of Cytochrome P450-3A (CYP3A), has direct antiviral activity against early circulating SARS-CoV-2 strains in vitro and in Syrian hamsters. Cobicistat is a derivative of ritonavir, which is co-administered as pharmacoenhancer with the SARS-CoV-2 protease inhibitor nirmatrelvir, to inhibit its metabolization by CPY3A and preserve its antiviral efficacy. Here, we used automated image analysis for a screening and parallel comparison of the anti-coronavirus effects of cobicistat and ritonavir. Our data show that both drugs display antiviral activity at low micromolar concentrations against multiple SARS-CoV-2 variants in vitro, including epidemiologically relevant Omicron subvariants. Despite their close structural similarity, we found that cobicistat is more potent than ritonavir, as shown by significantly lower EC50 values in monotherapy and higher levels of viral suppression when used in combination with nirmatrelvir. Finally, we show that the antiviral activity of both cobicistat and ritonavir is maintained against other human coronaviruses, including HCoV-229E and the highly pathogenic MERS-CoV. Overall, our results demonstrate that cobicistat has more potent anti-coronavirus activity than ritonavir and suggest that dose adjustments could pave the way to the use of both drugs as broad-spectrum antivirals against highly pathogenic human coronaviruses.
Collapse
Affiliation(s)
- Lara Gallucci
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - James Bazire
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Andrew D Davidson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK.
| | - Iart Luca Shytaj
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK.
| |
Collapse
|
7
|
Bjørklund G, Lysiuk R, Semenova Y, Lenchyk L, Dub N, Doşa MD, Hangan T. Herbal Substances with Antiviral Effects: Features and Prospects for the Treatment of Viral Diseases with Emphasis on Pro-Inflammatory Cytokines. Curr Med Chem 2024; 31:393-409. [PMID: 36698239 DOI: 10.2174/0929867330666230125121758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 01/26/2023]
Abstract
Viral diseases have a significant impact on human health, and three novel coronaviruses (CoV) have emerged during the 21st century. In this review, we have emphasized the potential of herbal substances with antiviral effects. Our investigation focused on the features and prospects of viral disease treatment, with a particular emphasis on proinflammatory cytokines. We conducted comprehensive searches of various databases, including Science Direct, CABI Direct, Web of Science, PubMed, and Scopus. Cytokine storm mechanisms play a crucial role in inducing a pro-inflammatory response by triggering the expression of cytokines and chemokines. This response leads to the recruitment of leukocytes and promotes antiviral effects, forming the first line of defense against viruses. Numerous studies have investigated the use of herbal medicine candidates as immunomodulators or antivirals. However, cytokine-storm-targeted therapy is recommended for patients with acute respiratory distress syndrome caused by SARS-CoV to survive severe pulmonary failure. Our reviews have demonstrated that herbal formulations could serve as alternative medicines and significantly reduce complicated viral infections. Furthermore, they hold promising potential as specific antiviral agents in experimental animal models.
Collapse
Affiliation(s)
- Geir Bjørklund
- Department of Research, Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Roman Lysiuk
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Yuliya Semenova
- School of Medicine, Nazarbayev University , Astana, Kazakhstan
| | - Larysa Lenchyk
- Department of Research, National University of Pharmacy, Kharkiv, Ukraine
- CONEM Ukraine Pharmacognosy and Natural Product Chemistry Research Group, National University of Pharmacy, Kharkiv, Ukraine
| | - Natalia Dub
- Andrei Krupynskyi Lviv Medical Academy, Lviv, Ukraine
| | | | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| |
Collapse
|
8
|
Cerracchio C, Amoroso MG, Piccolo M, Ferraro MG, Nocera FP, De Martino L, Serra F, Irace C, Tenore GC, Novellino E, Santamaria R, Fiorito F. Antiviral activity of Taurisolo® during bovine alphaherpesvirus 1 infection. Virus Res 2023; 336:199217. [PMID: 37666327 PMCID: PMC10504091 DOI: 10.1016/j.virusres.2023.199217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Bovine alphaherpesvirus 1 (BoAHV-1), the pathogen causing Infectious Bovine Rhinotracheitis (IBR) and predisposing to polymicrobial infections in cattle, provokes farm economic losses and trading restrictions in the world. However, nontoxic antiviral agents for BoAHV-1 infection are still unavailable, but plant extracts, such as flavonoid derivatives possess activity against BoAHV-1. Taurisolo®, a nutraceutical produced by Aglianico grape pomace, has recently shown promising antiviral activity. Herein, the potential activity of Taurisolo® during BoAHV-1 infection in Madin Darby bovine kidney (MDBK) cells was tested. Taurisolo® enhanced cell viability and reduced morphological death signs in BoAHV-1-infected cells. Moreover, Taurisolo® influenced the expression of bICP0, the key regulatory protein of BoAHV-1, and it strongly diminished virus yield. These effects were associated with an up-regulation of aryl hydrocarbon receptor (AhR), a transcription factor involved in microbial metabolism and immune response. In conclusion, our findings indicate that Taurisolo® may represent a potential antiviral agent against BoAHV-1 infection. Noteworthy, AhR could be involved in the observed effects and become a new target in antiviral therapy.
Collapse
Affiliation(s)
- Claudia Cerracchio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | | | - Marialuisa Piccolo
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | | | - Francesca Paola Nocera
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | - Luisa De Martino
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | - Francesco Serra
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Naples, Italy
| | - Carlo Irace
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy.
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Ettore Novellino
- Department of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Rita Santamaria
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Filomena Fiorito
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy.
| |
Collapse
|
9
|
Mohseni Afshar Z, Tavakoli Pirzaman A, Hosseinzadeh R, Babazadeh A, Taghizadeh Moghadam MA, Miri SR, Sio TT, Sullman MJM, Barary M, Ebrahimpour S. Anticoagulant therapy in COVID-19: A narrative review. Clin Transl Sci 2023; 16:1510-1525. [PMID: 37326220 PMCID: PMC10499427 DOI: 10.1111/cts.13569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/17/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can manifest itself in several ways, including coagulopathy and thrombosis. These complications can be the first and sometimes only manifestations of SARS-CoV-2 infection and can occur early or late in the course of the disease. However, these symptoms are more prevalent in hospitalized patients with venous thromboembolism, particularly those admitted to intensive care units. Moreover, various forms of arterial and venous thrombosis, or micro- or macro-vasculature embolisms, have been reported during the current pandemic. They have led to harmful consequences, such as neurological and cardiac events, nearly all resulting from the hypercoagulable state caused by this viral infection. The severe hypercoagulability observed in patients with COVID-19 accounts for most cases of the disease that become critical. Therefore, anticoagulants seem to be one of the most vital therapeutics for treating this potentially life-threatening condition. In the current paper, we present a thorough review of the pathophysiology of COVID-19-induced hypercoagulable state and the use of anticoagulants to treat SARS-CoV-2 infections in different patient groups, as well as their pros and cons.
Collapse
Affiliation(s)
- Zeinab Mohseni Afshar
- Clinical Research Development Center, Imam Reza HospitalKermanshah University of Medical SciencesKermanshahIran
| | | | | | - Arefeh Babazadeh
- Infectious Diseases and Tropical Medicine Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| | | | - Seyed Rouhollah Miri
- Cancer Research CenterCancer Institute of Iran, Tehran University of Medical ScienceTehranIran
| | - Terence T. Sio
- Department of Radiation OncologyMayo ClinicPhoenixArizonaUSA
| | - Mark J. M. Sullman
- Department of Social SciencesUniversity of NicosiaNicosiaCyprus
- Department of Life and Health SciencesUniversity of NicosiaNicosiaCyprus
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and ManagementShahid Beheshti University of Medical SciencesTehranIran
| | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research CenterHealth Research Institute, Babol University of Medical SciencesBabolIran
| |
Collapse
|
10
|
Reddy KR, Sahni C, Sharma S. Antiviral Activity of Svarnvir-IV Tablet Assayed for Activity Against SARS-CoV-2 In Vitro. Cureus 2023; 15:e39421. [PMID: 37362483 PMCID: PMC10288389 DOI: 10.7759/cureus.39421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
The coronavirus disease (COVID-19), caused by the virus SARS-CoV-2, has become a global pandemic in a very short time span. While several vaccines have been developed in the last year, specific treatments for CoV infection are still being explored. Thus, the situation highlights the need to develop safe and efficacious antiviral therapeutics. Ayurvedic Rasayana therapy has been traditionally used in India for its holistic healing systems and proven history of empirical use. There is emerging evidence that Ayurvedic treatment methodologies and herbal medicines may be effective strategies in combating COVID-19. The present study is aimed at evaluating the antiviral and therapeutic activity of an Ayurvedic herbomineral formulation (Svarnvir-IV tablet, 450 mg) against the SARS-CoV-2 virus in vitro. A cell-based assay was conducted to evaluate the cytotoxicity of the Svarnvir-IV tablets (Aimil Pharmaceuticals, Delhi, India) for the determination of virucidal activity assessment (at 2 hours) and therapeutic activity assessment (at 1 hour, 2 hours, and 4 hours). When incubated with SARS-CoV-2 virus at 0.1 multiplicity of infection (MoI) for two hours, Svarnvir-IV tablet exhibited virucidal activity against SARS-CoV-2 with an EC50 value of 0.0058 mg/ml. It also exhibited therapeutic activity when treated with cells infected with the SARS-CoV-2 virus (0.1 MoI) for 1 hour, 2 hours and 4 hours post-infection, with an EC50 value of 0.094 mg/ml, 0.023 mg/ml, and 0.05 mg/ml, respectively. The original supporting data obtained from this study, along with existing Ayurvedic traditional information, has shown encouraging results.
Collapse
Affiliation(s)
- K Ramachandra Reddy
- Department of Rasa Shastra, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Chetan Sahni
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Sanchit Sharma
- Research and Development, Aimil Pharmaceuticals, Delhi, IND
| |
Collapse
|
11
|
Sameni M, Mirmotalebisohi SA, Dehghan Z, Abooshahab R, Khazaei-Poul Y, Mozafar M, Zali H. Deciphering molecular mechanisms of SARS-CoV-2 pathogenesis and drug repurposing through GRN motifs: a comprehensive systems biology study. 3 Biotech 2023; 13:117. [PMID: 37070032 PMCID: PMC10090260 DOI: 10.1007/s13205-023-03518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/13/2023] [Indexed: 03/28/2023] Open
Abstract
The world has recently been plagued by a new coronavirus infection called SARS-CoV-2. This virus may lead to severe acute respiratory syndrome followed by multiple organ failure. SARS-CoV-2 has approximately 80-90% genetic similarity to SARS-CoV. Given the limited omics data available for host response to the viruses (more limited data for SARS-CoV-2), we attempted to unveil the crucial molecular mechanisms underlying the SARS-CoV-2 pathogenesis by comparing its regulatory network motifs with SARS-CoV. We also attempted to identify the non-shared crucial molecules and their functions to predict the specific mechanisms for each infection and the processes responsible for their different manifestations. Deciphering the crucial shared and non-shared mechanisms at the molecular level and signaling pathways underlying both diseases may help shed light on their pathogenesis and pave the way for other new drug repurposing against COVID-19. We constructed the GRNs for host response to SARS-CoV and SARS-CoV-2 pathogens (in vitro) and identified the significant 3-node regulatory motifs by analyzing them topologically and functionally. We attempted to identify the shared and non-shared regulatory elements and signaling pathways between their host responses. Interestingly, our findings indicated that NFKB1, JUN, STAT1, FOS, KLF4, and EGR1 were the critical shared TFs between motif-related subnetworks in both SARS and COVID-1, which are considered genes with specific functions in the immune response. Enrichment analysis revealed that the NOD-like receptor signaling, TNF signaling, and influenza A pathway were among the first significant pathways shared between SARS and COVID-19 up-regulated DEGs networks, and the term "metabolic pathways" (hsa01100) among the down-regulated DEGs networks. WEE1, PMAIP1, and TSC22D2 were identified as the top three hubs specific to SARS. However, MYPN, SPRY4, and APOL6 were the tops specific to COVID-19 in vitro. The term "Complement and coagulation cascades" pathway was identified as the first top non-shared pathway for COVID-19 and the MAPK signaling pathway for SARS. We used the identified crucial DEGs to construct a drug-gene interaction network to propose some drug candidates. Zinc chloride, Fostamatinib, Copper, Tirofiban, Tretinoin, and Levocarnitine were the six drugs with higher scores in our drug-gene network analysis. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03518-x.
Collapse
Affiliation(s)
- Marzieh Sameni
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Mirmotalebisohi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Yalda Khazaei-Poul
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mozafar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Hakimeh Zali
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Zeng W, Ren J, Yang G, Jiang C, Dong L, Sun Q, Hu Y, Li W, He Q. Porcine Epidemic Diarrhea Virus and Its nsp14 Suppress ER Stress Induced GRP78. Int J Mol Sci 2023; 24:ijms24054936. [PMID: 36902365 PMCID: PMC10003387 DOI: 10.3390/ijms24054936] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), a member of the α-coronavirus genus, can cause vomiting, diarrhea, and dehydration in piglets. Neonatal piglets infected with PEDV have a mortality rate as high as 100%. PEDV has caused substantial economic losses to the pork industry. Endoplasmic reticulum (ER) stress, which can alleviate the accumulation of unfolded or misfolded proteins in ER, involves in coronavirus infection. Previous studies have indicated that ER stress could inhibit the replication of human coronaviruses, and some human coronaviruses in turn could suppress ER stress-related factors. In this study, we demonstrated that PEDV could interact with ER stress. We determined that ER stress could potently inhibit the replication of GⅠ, GⅡ-a, and GⅡ-b PEDV strains. Moreover, we found that these PEDV strains can dampen the expression of the 78 kDa glucose-regulated protein (GRP78), an ER stress marker, while GRP78 overexpression showed antiviral activity against PEDV. Among different PEDV proteins, PEDV non-structural protein 14 (nsp14) was revealed to play an essential role in the inhibition of GRP78 by PEDV, and its guanine-N7-methyltransferase domain is necessary for this role. Further studies show that both PEDV and its nsp14 negatively regulated host translation, which could account for their inhibitory effects against GRP78. In addition, we found that PEDV nsp14 could inhibit the activity of GRP78 promotor, helping suppress GRP78 transcription. Our results reveal that PEDV possesses the potential to antagonize ER stress, and suggest that ER stress and PEDV nsp14 could be the targets for developing anti-PEDV drugs.
Collapse
Affiliation(s)
- Wei Zeng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jingping Ren
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Gan Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Changsheng Jiang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Ling Dong
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qi Sun
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yaofang Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Wentao Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (W.Z.); (J.R.); (G.Y.); (C.J.); (L.D.); (Q.S.); (Y.H.); (W.L.)
- The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
13
|
Identification of novel antiviral drug candidates using an optimized SARS-CoV-2 phenotypic screening platform. iScience 2023; 26:105944. [PMID: 36644320 PMCID: PMC9822553 DOI: 10.1016/j.isci.2023.105944] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/07/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
Reliable, easy-to-handle phenotypic screening platforms are needed for the identification of anti-SARS-CoV-2 compounds. Here, we present caspase 3/7 activity as a readout for monitoring the replication of SARS-CoV-2 isolates from different variants, including a remdesivir-resistant strain, and of other coronaviruses in numerous cell culture models, independently of cytopathogenic effect formation. Compared to other models, the Caco-2 subline Caco-2-F03 displayed superior performance. It possesses a stable SARS-CoV-2 susceptibility phenotype and does not produce false-positive hits due to drug-induced phospholipidosis. A proof-of-concept screen of 1,796 kinase inhibitors identified known and novel antiviral drug candidates including inhibitors of phosphoglycerate dehydrogenase (PHGDH), CDC like kinase 1 (CLK-1), and colony stimulating factor 1 receptor (CSF1R). The activity of the PHGDH inhibitor NCT-503 was further increased in combination with the hexokinase II (HK2) inhibitor 2-deoxy-D-glucose, which is in clinical development for COVID-19. In conclusion, caspase 3/7 activity detection in SARS-CoV-2-infected Caco-2-F03 cells provides a simple phenotypic high-throughput screening platform for SARS-CoV-2 drug candidates that reduces false-positive hits.
Collapse
|
14
|
Cerracchio C, Serra F, Amoroso MG, Fiorito F. Canine Coronavirus Activates Aryl Hydrocarbon Receptor during In Vitro Infection. Viruses 2022; 14:v14112437. [PMID: 36366535 PMCID: PMC9692492 DOI: 10.3390/v14112437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that interacts with substrates, including microbial metabolites. Recent advances reveal that AhR is involved in the host response to coronaviruses (CoVs) infection. Particularly, AhR antagonists decrease the expression of angiotensin-converting enzyme 2 (ACE2) via AhR up-regulation, resulting in suppression of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in mammalian cells. Herein, we report that AhR is expressed in canine fibrosarcoma (A72) cells, where it is considerably activated by infection with genotype II of canine coronavirus (CCoV-II). The pharmacological inhibition of AhR, by CH223191, suppressed cell death signs and increased cell viability. Furthermore, the AhR antagonist induced a meaningful decline in virus yield, accompanied by the inhibition of the expression of viral nuclear protein (NP). Fascinatingly, during CCoV infection, a novel co-expression of NP and AhR expression was found. Taken together, our preliminary findings show that infection with CCoV activates AhR, and pharmacologic AhR inhibition reduces CCoV replication, identifying AhR as a possible candidate target for CCoV antiviral therapy.
Collapse
Affiliation(s)
- Claudia Cerracchio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | - Francesco Serra
- Department of Animal Health, Unit of Virology, Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Naples, Italy
| | - Maria Grazia Amoroso
- Department of Animal Health, Unit of Virology, Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Naples, Italy
| | - Filomena Fiorito
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
- Correspondence: (F.F.); Tel.: +39-0812536180
| |
Collapse
|
15
|
Plant Molecular Pharming and Plant-Derived Compounds towards Generation of Vaccines and Therapeutics against Coronaviruses. Vaccines (Basel) 2022; 10:vaccines10111805. [DOI: 10.3390/vaccines10111805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
The current century has witnessed infections of pandemic proportions caused by Coronaviruses (CoV) including severe acute respiratory syndrome-related CoV (SARS-CoV), Middle East respiratory syndrome-related CoV (MERS-CoV) and the recently identified SARS-CoV2. Significantly, the SARS-CoV2 outbreak, declared a pandemic in early 2020, has wreaked devastation and imposed intense pressure on medical establishments world-wide in a short time period by spreading at a rapid pace, resulting in high morbidity and mortality. Therefore, there is a compelling need to combat and contain the CoV infections. The current review addresses the unique features of the molecular virology of major Coronaviruses that may be tractable towards antiviral targeting and design of novel preventative and therapeutic intervention strategies. Plant-derived vaccines, in particular oral vaccines, afford safer, effectual and low-cost avenues to develop antivirals and fast response vaccines, requiring minimal infrastructure and trained personnel for vaccine administration in developing countries. This review article discusses recent developments in the generation of plant-based vaccines, therapeutic/drug molecules, monoclonal antibodies and phytochemicals to preclude and combat infections caused by SARS-CoV, MERS-CoV and SARS-CoV-2 viruses. Efficacious plant-derived antivirals could contribute significantly to combating emerging and re-emerging pathogenic CoV infections and help stem the tide of any future pandemics.
Collapse
|
16
|
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) pandemic. Of particular interest for this topic are the signaling cascades that regulate cell survival and death, two opposite cell programs whose control is hijacked by viral infections. The AKT and the Unfolded Protein Response (UPR) pathways, which maintain cell homeostasis by regulating these two programs, have been shown to be deregulated during SARS-CoVs infection as well as in the development of cancer, one of the most important comorbidities in relation to COVID-19. Recent evidence revealed two way crosstalk mechanisms between the AKT and the UPR pathways, suggesting that they might constitute a unified homeostatic control system. Here, we review the role of the AKT and UPR pathways and their interaction in relation to SARS-CoV-2 infection as well as in tumor onset and progression. Feedback regulation between AKT and UPR pathways emerges as a master control mechanism of cell decision making in terms of survival or death and therefore represents a key potential target for developing treatments for both viral infection and cancer. In particular, drug repositioning, the investigation of existing drugs for new therapeutic purposes, could significantly reduce time and costs compared to de novo drug discovery.
Collapse
|
17
|
Wifi MN, Morad MA, El Sheemy R, Abdeen N, Afify S, Abdalgaber M, Abdellatef A, Zaghloul M, Alboraie M, El-Kassas M. Hemostatic system and COVID-19 crosstalk: A review of the available evidence. World J Methodol 2022; 12:331-349. [PMID: 36186748 PMCID: PMC9516549 DOI: 10.5662/wjm.v12.i5.331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/17/2022] [Accepted: 07/19/2022] [Indexed: 02/08/2023] Open
Abstract
Since the discovery of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its resultant coronavirus disease 2019 (COVID-19) pandemic, respiratory manifestations have been the mainstay of clinical diagnosis, laboratory evaluations, and radiological investigations. As time passed, other pathological aspects of SARS-CoV-2 have been revealed. Various hemostatic abnormalities have been reported since the rise of the pandemic, which was sometimes superficial, transient, or fatal. Mild thrombocytopenia, thrombocytosis, venous, arterial thromboembolism, and disseminated intravascular coagulation are among the many hemostatic events associated with COVID-19. Venous thromboembolism necessitating therapeutic doses of anticoagulants is more frequently seen in severe cases of COVID-19, especially in patients admitted to intensive care units. Hemorrhagic complications rarely arise in COVID-19 patients either due to a hemostatic imbalance resulting from severe disease or as a complication of over anticoagulation. Although the pathogenesis of coagulation disturbance in SARS-CoV-2 infection is not yet understood, professional societies recommend prophylactic antithrombotic therapy in severe cases, especially in the presence of abnormal coagulation indices. The review article discusses the various available evidence on coagulation disorders, management strategies, outcomes, and prognosis associated with COVID-19 coagulopathy, which raises awareness about the importance of anticoagulation therapy for COVID-19 patients to guard against possible thromboembolic events.
Collapse
Affiliation(s)
- Mohamed-Naguib Wifi
- Department of Internal Medicine, Hepatogastro- enterology Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo 11451, Egypt
| | - Mohamed Abdelkader Morad
- Clinical Hematology Unit, Department of Internal Medicine, Kasr Al-Ainy, Faculty of Medicine, Cairo University, Cairo 11451, Egypt
| | - Reem El Sheemy
- Department of Tropical Medicine, Minia Faculty of Medicine, Minia University, Minia 61511, Egypt
| | - Nermeen Abdeen
- Department of Tropical Medicine, Faculty of Medicine, Alexandria University, Alexandria 21523, Egypt
| | - Shimaa Afify
- Department of Gastroenterology, National Hepatology and Tropical Medicine, National Hepatology and Tropical Medicine Research Institute, Cairo 11451, Egypt
| | - Mohammad Abdalgaber
- Department of Gastroenterology and Hepatology, Police Authority Hospital, Agoza, Giza 12511, Egypt
| | - Abeer Abdellatef
- Department of Internal Medicine, Hepatogastro- enterology Unit, Kasr Al-Ainy School of Medicine, Cairo University, Cairo 11451, Egypt
| | - Mariam Zaghloul
- Department of Hepatology, Gastroenterology and Infectious Diseases, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh 33511, Egypt
| | - Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo 11884, Egypt
| | - Mohamed El-Kassas
- Department of Endemic Medicine, Faculty of Medicine, Helwan University, Helwan 11731, Egypt
| |
Collapse
|
18
|
Thiopurines inhibit coronavirus Spike protein processing and incorporation into progeny virions. PLoS Pathog 2022; 18:e1010832. [PMID: 36121863 PMCID: PMC9522307 DOI: 10.1371/journal.ppat.1010832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/29/2022] [Accepted: 08/24/2022] [Indexed: 11/19/2022] Open
Abstract
There is an outstanding need for broadly acting antiviral drugs to combat emerging viral diseases. Here, we report that thiopurines inhibit the replication of the betacoronaviruses HCoV-OC43 and SARS-CoV-2. 6-Thioguanine (6-TG) disrupted early stages of infection, limiting accumulation of full-length viral genomes, subgenomic RNAs and structural proteins. In ectopic expression models, we observed that 6-TG increased the electrophoretic mobility of Spike from diverse betacoronaviruses, matching the effects of enzymatic removal of N-linked oligosaccharides from Spike in vitro. SARS-CoV-2 virus-like particles (VLPs) harvested from 6-TG-treated cells were deficient in Spike. 6-TG treatment had a similar effect on production of lentiviruses pseudotyped with SARS-CoV-2 Spike, yielding pseudoviruses deficient in Spike and unable to infect ACE2-expressing cells. Together, these findings from complementary ectopic expression and infection models strongly indicate that defective Spike trafficking and processing is an outcome of 6-TG treatment. Using biochemical and genetic approaches we demonstrated that 6-TG is a pro-drug that must be converted to the nucleotide form by hypoxanthine phosphoribosyltransferase 1 (HPRT1) to achieve antiviral activity. This nucleotide form has been shown to inhibit small GTPases Rac1, RhoA, and CDC42; however, we observed that selective chemical inhibitors of these GTPases had no effect on Spike processing or accumulation. By contrast, the broad GTPase agonist ML099 countered the effects of 6-TG, suggesting that the antiviral activity of 6-TG requires the targeting of an unknown GTPase. Overall, these findings suggest that small GTPases are promising targets for host-targeted antivirals. The COVID-19 pandemic has ignited efforts to repurpose existing drugs as safe and effective antivirals. Rather than directly inhibiting viral enzymes, host-targeted antivirals inhibit host cell processes to indirectly impede viral replication and/or stimulate antiviral responses. Here, we describe a new antiviral mechanism of action for an FDA-approved thiopurine known as 6-thioguanine (6-TG). We demonstrate that 6-TG is a pro-drug that must be metabolized by host enzymes to gain antiviral activity. We show that it can inhibit the replication of human coronaviruses, including SARS-CoV-2, at least in part by interfering with the processing and accumulation of Spike glycoproteins, thereby impeding assembly of infectious progeny viruses. We provide evidence implicating host cell GTPase enzymes in the antiviral mechanism of action.
Collapse
|
19
|
Wolf A, Khimani F, Yoon B, Gerhart C, Endsley D, Ray AK, Yango AF, Flynn SD, Lip GYH, Gonzalez SA, Sathyamoorthy M. The mechanistic basis linking cytokine storm to thrombosis in COVID-19. THROMBOSIS UPDATE 2022; 8:100110. [PMID: 38620974 PMCID: PMC9116969 DOI: 10.1016/j.tru.2022.100110] [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: 02/01/2022] [Revised: 04/26/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022] Open
Abstract
It is now well established that infection with SARS-CoV-2 resulting in COVID-19 disease includes a severely symptomatic subset of patients in whom an aggressive and/or dysregulated host immune response leads to cytokine storm syndrome (CSS) that may be further complicated by thrombotic events, contributing to the severe morbidity and mortality observed in COVID-19. This review provides a brief overview of cytokine storm in COVID-19, and then presents a mechanistic discussion of how cytokine storm affects integrated pathways in thrombosis involving the endothelium, platelets, the coagulation cascade, eicosanoids, auto-antibody mediated thrombosis, and the fibrinolytic system.
Collapse
Affiliation(s)
- Adam Wolf
- Sathyamoorthy Laboratory, TCU School of Medicine, Fort Worth, TX, United States
- TCU School of Medicine, Fort Worth, TX, United States
| | - Faria Khimani
- Sathyamoorthy Laboratory, TCU School of Medicine, Fort Worth, TX, United States
- TCU School of Medicine, Fort Worth, TX, United States
| | - Braian Yoon
- Sathyamoorthy Laboratory, TCU School of Medicine, Fort Worth, TX, United States
- TCU School of Medicine, Fort Worth, TX, United States
| | - Coltin Gerhart
- Sathyamoorthy Laboratory, TCU School of Medicine, Fort Worth, TX, United States
- TCU School of Medicine, Fort Worth, TX, United States
| | - Dakota Endsley
- Sathyamoorthy Laboratory, TCU School of Medicine, Fort Worth, TX, United States
- TCU School of Medicine, Fort Worth, TX, United States
| | - Anish K Ray
- Cook Children's Medical Center, Fort Worth, TX, United States
- Department of Pediatrics, TCU School of Medicine, Fort Worth, TX, United States
| | - Angelito F Yango
- Department of Medicine, TCU School of Medicine, Fort Worth, TX, United States
- Annette C. and Harold C. Simmons Transplant Institute, Baylor All Saints Medical Center, Fort Worth, TX, USA
| | | | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
- Department of Clinical Medicine, Aalborg, Denmark
| | - Stevan A Gonzalez
- Department of Medicine, TCU School of Medicine, Fort Worth, TX, United States
- Annette C. and Harold C. Simmons Transplant Institute, Baylor All Saints Medical Center, Fort Worth, TX, USA
| | - Mohanakrishnan Sathyamoorthy
- Sathyamoorthy Laboratory, TCU School of Medicine, Fort Worth, TX, United States
- Department of Medicine, TCU School of Medicine, Fort Worth, TX, United States
- Consultants in Cardiovascular Medicine and Science - Fort Worth, PLLC, Fort Worth, TX, United States
| |
Collapse
|
20
|
Weil T, Lawrenz J, Seidel A, Münch J, Müller JA. Immunodetection assays for the quantification of seasonal common cold coronaviruses OC43, NL63, or 229E infection confirm nirmatrelvir as broad coronavirus inhibitor. Antiviral Res 2022; 203:105343. [PMID: 35598779 PMCID: PMC9119192 DOI: 10.1016/j.antiviral.2022.105343] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/05/2022] [Accepted: 05/16/2022] [Indexed: 01/21/2023]
Abstract
Besides pandemic SARS-CoV-2, also endemic seasonal human common cold coronaviruses (hCoVs) have a significant impact on human health and economy. Studies on hCoVs and the identification of antivirals are therefore crucial to improve human well-being. However, hCoVs have long been neglected and the methodology to study virus infection, replication and inhibition warrants being updated. We here evaluated the established plaque-based assays to determine viral titers and cell-to-cell spread and developed protocols for the immunodetection of the viral nucleocapsid protein by flow cytometry and in-cell ELISA to study infection rates at early time points. The developed protocols allow detection of hCoV-229E infection after 2, and hCoV-NL63 and -OC43 infection after 3 days at a single cell level or in a 96 well microtiter format, in large sample numbers without being laborious or expensive. Both assays can be applied to assess the susceptibility of cells to hCoV infection and replication, and to determine the efficacy of antiviral compounds as well as neutralizing antibodies in a sensitive and quantitative manner. Application revealed that clinically applied SARS-CoV-2 targeting monoclonal antibodies are inactive against hCoVs, but that the viral polymerase targeting antivirals remdesivir and molnupiravir are broadly active also against all three hCoVs. Further, the in-cell ELISA provided evidence that nirmatrelvir, previously shown to broadly inhibit coronavirus proteases, also prevents replication of authentic hCoVs. Importantly, the protocols described here can be easily adapted to other coronavirus strains and species as well as viruses of other families within a short time. This will facilitate future research on known and emerging (corona)viruses, support the identification of antivirals and increase the preparedness for future virus outbreaks.
Collapse
Affiliation(s)
- Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Jan Lawrenz
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany; Core Facility Functional Peptidomics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany; Institute of Virology, Philipps University of Marburg, 35043, Marburg, Germany.
| |
Collapse
|
21
|
Zhao X, Chen D, Li X, Griffith L, Chang J, An P, Guo JT. Interferon Control of Human Coronavirus Infection and Viral Evasion: Mechanistic Insights and Implications for Antiviral Drug and Vaccine Development. J Mol Biol 2022; 434:167438. [PMID: 34990653 PMCID: PMC8721920 DOI: 10.1016/j.jmb.2021.167438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022]
Abstract
Recognition of viral infections by various pattern recognition receptors (PRRs) activates an inflammatory cytokine response that inhibits viral replication and orchestrates the activation of adaptive immune responses to control the viral infection. The broadly active innate immune response puts a strong selective pressure on viruses and drives the selection of variants with increased capabilities to subvert the induction and function of antiviral cytokines. This revolutionary process dynamically shapes the host ranges, cell tropism and pathogenesis of viruses. Recent studies on the innate immune responses to the infection of human coronaviruses (HCoV), particularly SARS-CoV-2, revealed that HCoV infections can be sensed by endosomal toll-like receptors and/or cytoplasmic RIG-I-like receptors in various cell types. However, the profiles of inflammatory cytokines and transcriptome response induced by a specific HCoV are usually cell type specific and determined by the virus-specific mechanisms of subverting the induction and function of interferons and inflammatory cytokines as well as the genetic trait of the host genes of innate immune pathways. We review herein the recent literatures on the innate immune responses and their roles in the pathogenesis of HCoV infections with emphasis on the pathobiological roles and therapeutic effects of type I interferons in HCoV infections and their antiviral mechanisms. The knowledge on the mechanism of innate immune control of HCoV infections and viral evasions should facilitate the development of therapeutics for induction of immune resolution of HCoV infections and vaccines for efficient control of COVID-19 pandemics and other HCoV infections.
Collapse
Affiliation(s)
- Xuesen Zhao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Institute of Infectious Diseases, Beijing 100015, China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| | - Danying Chen
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Institute of Infectious Diseases, Beijing 100015, China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xinglin Li
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Institute of Infectious Diseases, Beijing 100015, China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Lauren Griffith
- Baruch S. Blumberg Institute, Hepatitis B Foundation, 3805 Old Easton Road, Doylestown, PA 18902, USA
| | - Jinhong Chang
- Baruch S. Blumberg Institute, Hepatitis B Foundation, 3805 Old Easton Road, Doylestown, PA 18902, USA
| | - Ping An
- Basic Research Laboratory, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Ju-Tao Guo
- Baruch S. Blumberg Institute, Hepatitis B Foundation, 3805 Old Easton Road, Doylestown, PA 18902, USA.
| |
Collapse
|
22
|
Cimolai N. Comment on: "COVID-19 and acute limb ischemia: a systematic review". Reconsidering human-associated coronavirus infections and idiopathic thrombosis. THE JOURNAL OF CARDIOVASCULAR SURGERY 2022; 63:114. [PMID: 35179339 DOI: 10.23736/s0021-9509.21.12115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Affiliation(s)
- Nevio Cimolai
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada -
- Children's and Women's Health Centre of British Columbia, Vancouver, BC, Canada -
| |
Collapse
|
23
|
Ahmad F, Kannan M, Ansari AW. Role of SARS-CoV-2 -induced cytokines and growth factors in coagulopathy and thromboembolism. Cytokine Growth Factor Rev 2022; 63:58-68. [PMID: 34750061 PMCID: PMC8541834 DOI: 10.1016/j.cytogfr.2021.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/08/2023]
Abstract
Severe COVID-19 patients frequently present thrombotic complications which commonly lead to multiorgan failure and increase the risk of death. Severe SARS-CoV-2 infection induces the cytokine storm and is often associated with coagulation dysfunction. D-dimer, a hallmark of venous thromboembolism (VTE), is observed at a higher level in the majority of hospitalized COVID-19 patients. The precise molecular mechanism of the disproportionate effect of SARS-CoV-2 infection on the coagulation system is largely undefined. SARS-CoV-2 -induced endotheliopathy and, induction of cytokines and growth factors (GFs) most likely play important roles in platelet activation, coagulopathy, and VTE. Generally, viral infections lead to systemic inflammation and induction of numerous cytokines and GFs and many of them are reported to be associated with increased VTE. Most importantly, platelets play key thromboinflammatory roles linking coagulation to immune mediators in a variety of infections including response to viral infection. Since the pathomechanism of coagulopathy and VTE in COVID-19 is largely undefined, herein we highlight the association of dysregulated inflammatory cytokines and GFs with thrombotic complications and coagulopathy in COVID-19.
Collapse
Affiliation(s)
- Firdos Ahmad
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE.
| | - Meganathan Kannan
- Blood and Vascular Biology Research Lab, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610005, India
| | - Abdul W Ansari
- Dermatology Institute, Translational Research Institute, Academic Health Systems, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| |
Collapse
|
24
|
Anaeigoudari A, Mollaei HR, Arababadi MK, Nosratabadi R. Severe Acute Respiratory Syndrome Coronavirus 2: The Role of the Main Components of the Innate Immune System. Inflammation 2021; 44:2151-2169. [PMID: 34524614 PMCID: PMC8442517 DOI: 10.1007/s10753-021-01519-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/08/2021] [Accepted: 07/08/2021] [Indexed: 02/08/2023]
Abstract
At the end of December 2019, the COVID-19 pandemic began in Wuhan of China. COVID-19 affects different people with a wide spectrum of clinical manifestations, ranging from asymptomatic with recovery without hospitalization up to a severe acute respiratory syndrome (SARS). The innate and adaptive immunity appears responsible for the defense against the virus and recovery from the disease. The innate immune system, as the first line of defense, is essential for the detection of virus and subsequent activation of acquired immunity. The innate immune response is carried out by sentinel cells such as monocytes/macrophages and dendritic cells and by receptors known as pattern recognition receptors (PRR). These receptors can recognize various components of the virus, which lead to intracellular signaling and subsequently the synthesis of various cytokines. These cytokines then recruit other immune cells, activate adaptive immune responses, and inhibit viral spreading. The most common receptors include Toll-like receptors, C-type lectin receptors, and RIG-I like receptors. This review describes the current knowledge about the interplay between innate immune responses and SARS-CoV-2 with a focus on the innate immune cells and the role of their receptors in viral RNA recognition, as well as their mechanisms for recognizing SARS-CoV-2.
Collapse
Affiliation(s)
- Akbar Anaeigoudari
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Hamid Reza Mollaei
- Department of Medical Microbiology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Kazemi Arababadi
- Department of Laboratory Sciences, Faculty of Paramedicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Reza Nosratabadi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
25
|
Kundu S, Saadi F, Sengupta S, Antony GR, Raveendran VA, Kumar R, Kamble MA, Sarkar L, Burrows A, Pal D, Sen GC, Sarma JD. DJ-1-Nrf2 axis is activated upon murine β-coronavirus infection in the CNS. BRAIN DISORDERS 2021; 4:100021. [PMID: 34514445 PMCID: PMC8418700 DOI: 10.1016/j.dscb.2021.100021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/03/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Coronaviruses have emerged as alarming pathogens owing to their inherent ability of genetic variation and cross-species transmission. Coronavirus infection burdens the endoplasmic reticulum (ER.), causes reactive oxygen species production and induces host stress responses, including unfolded protein response (UPR) and antioxidant system. In this study, we have employed a neurotropic murine β-coronavirus (M-CoV) infection in the Central Nervous System (CNS) of experimental mice model to study the role of host stress responses mediated by interplay of DJ-1 and XBP1. DJ-1 is an antioxidant molecule with established functions in neurodegeneration. However, its regulation in virus-induced cellular stress response is less explored. Our study showed that M-CoV infection activated the glial cells and induced antioxidant and UPR genes during the acute stage when the viral titer peaks. As the virus particles decreased and acute neuroinflammation diminished at day ten p.i., a significant up-regulation in UPR responsive XBP1, antioxidant DJ-1, and downstream signaling molecules, including Nrf2, was recorded in the brain tissues. Additionally, preliminary in silico analysis of the binding between the DJ-1 promoter and a positively charged groove of XBP1 is also investigated, thus hinting at a mechanism behind the upregulation of DJ-1 during MHV-infection. The current study thus attempts to elucidate a novel interplay between the antioxidant system and UPR in the outcome of coronavirus infection.
Collapse
Affiliation(s)
- Soumya Kundu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Sourodip Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Gisha Rose Antony
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Vineeth A Raveendran
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Rahul Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Mithila Ashok Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Lucky Sarkar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Amy Burrows
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
| | - Ganes C Sen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| |
Collapse
|
26
|
Perri F, Crispo A, Ionna F, Muto P, Caponigro F, Longo F, Montagnese C, Franco P, Pavone E, Aversa C, Guida A, Bimonte S, Ottaiano A, Di Marzo M, Porciello G, Amore A, Celentano E, Della Vittoria Scarpati G, Cascella M. Patients affected by squamous cell carcinoma of the head and neck: A population particularly prone to developing severe forms of COVID-19. Exp Ther Med 2021; 22:1298. [PMID: 34630653 PMCID: PMC8461515 DOI: 10.3892/etm.2021.10733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/17/2021] [Indexed: 01/09/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the recent Coronavirus Disease 2019 (COVID-19) pandemic, which has spread all over the world over the past year. Comorbidities appear to affect the prognosis of patients with such diseases, but the impact of cancer on the course of SARS-CoV2 has remained largely elusive. The aim of the present study is to analyze the outcome of patients affected by squamous cell carcinoma of the head and neck (SCCHN) and a number of their comorbidities, if infected with SARS-CoV2. The clinical data of 100 patients affected by SCCHN, who were undergoing treatment or who had finished their oncologic treatment in the past 6 months, were retrospectively collected and analysed. For each patient, the Charlson Comorbidity Index (CCI) was calculated to provide a score assessing the real weight of comorbidities on the patient's outcome at the time of diagnosis. It was discovered that these patients, besides the SCCHN, frequently presented at diagnosis with several other comorbidities, including hypertension, type 2 diabetes, cardiac arrhytmia, chronic obstructive pulmonary disease and various forms of vasculopathy (and thus a poor CCI). This feature suggest that, given the high frequency of various comorbidities in patients with SCCHN, additional SARS-CoV2 infection could have particularly devastating consequences.
Collapse
Affiliation(s)
- Francesco Perri
- Head and Neck Medical and Experimental Oncology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy,Correspondence to: Dr Francesco Perri, Head and Neck Medical and Experimental Oncology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, 8031 Via M. Semmola, I-80131 Naples, Italy
| | - Anna Crispo
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Franco Ionna
- Otolaryngology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Paolo Muto
- Radiation Oncology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Francesco Caponigro
- Head and Neck Medical and Experimental Oncology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Francesco Longo
- Otolaryngology Unit, Casa Sollievo della Sofferenza di San Giovanni Rotondo, I-71013 Foggia, Italy
| | - Concetta Montagnese
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Pierluigi Franco
- Otolaryngology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Ettore Pavone
- Otolaryngology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Corrado Aversa
- Otolaryngology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Agostino Guida
- Otolaryngology Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Sabrina Bimonte
- Department of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Alessandro Ottaiano
- Innovative Therapies for Abdominal Metastases, Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Massimiliano Di Marzo
- Colorectal and Abdominal Surgery Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Giuseppe Porciello
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Alfonso Amore
- Department Melanoma, Soft Tissue, Muscle-Skeletal and Head-Neck, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | - Egidio Celentano
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| | | | - Marco Cascella
- Department of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS ‘Fondazione G. Pascale’, I-80131 Naples, Italy
| |
Collapse
|
27
|
Gavriilaki E, Eftychidis I, Papassotiriou I. Update on endothelial dysfunction in COVID-19: severe disease, long COVID-19 and pediatric characteristics. J LAB MED 2021. [DOI: 10.1515/labmed-2021-0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract
Objectives
To review current literature on the role of endothelial dysfunction in coronavirus disease-2019 (COVID-19) infection in terms of pathophysiology, laboratory features and markers, clinical phenotype in adults and children, as well as long COVID-19.
Content
We conducted a thorough assessment of the literature and critically analyzed current data, mostly utilizing the PubMed and Medline search engines to find original studies published in the previous decade.
Summary and Outlook
Accumulating evidence suggests that endothelial dysfunction may be a common denominator of severe COVID-19 in adults and children, as well as long COVID-19, implicating mutual pathophysiological pathways. This narrative review summarizes the up-to-date knowledge of endothelial dysfunction caused by COVID-19, including novel aspects of long COVID-19 and pediatric disease. This knowledge is important in order not only to understand the multisystemic attack of COVID-19, but also to improve patient management and prognosis.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department – BMT Unit , G. Papanikolaou Hospital , Thessaloniki , Greece
| | - Ioannis Eftychidis
- Hematology Department – BMT Unit , G. Papanikolaou Hospital , Thessaloniki , Greece
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry , “Aghia Sophia” Children’s Hospital , Athens , Greece
- IFCC Emerging Technologies Division , Emerging Technologies in Pediatric Laboratory Medicine (C-ETPLM) , Milan , Italy
| |
Collapse
|
28
|
Nahalka J. Theoretical Analysis of S, M and N Structural Proteins by the Protein-RNA Recognition Code Leads to Genes/proteins that Are Relevant to the SARS-CoV-2 Life Cycle and Pathogenesis. Front Genet 2021; 12:763995. [PMID: 34659373 PMCID: PMC8511677 DOI: 10.3389/fgene.2021.763995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/15/2021] [Indexed: 12/14/2022] Open
Abstract
In this conceptual review, based on the protein-RNA recognition code, some theoretical sequences were detected in the spike (S), membrane (M) and capsid (N) proteins that may post-transcriptionally regulate the host genes/proteins in immune homeostasis, pulmonary epithelial tissue homeostasis, and lipid homeostasis. According to the review of literature, the spectrum of identified genes/proteins shows that the virus promotes IL1α/β-IL1R1 signaling (type 1 immunity) and immunity defense against helminths and venoms (type 2 immunity). In the alteration of homeostasis in the pulmonary epithelial tissue, the virus blocks the function of cilia and the molecular programs that are involved in wound healing (EMT and MET). Additionally, the protein-RNA recognition method described here identifies compatible sequences in the S1A-domain for the post-transcriptional promotion of PIKFYVE, which is one of the critical factors for SARS-CoV-2 entry to the host cell, and for the post-transcriptional repression of xylulokinase XYLB. A decrease in XYLB product (Xu5P) in plasma was proposed as one of the potential metabolomics biomarkers of COVID-19. In summary, the protein-RNA recognition code leads to protein genes relevant to the SARS-CoV-2 life cycle and pathogenesis.
Collapse
Affiliation(s)
- Jozef Nahalka
- Institute of Chemistry, Centre for Glycomics, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Chemistry, Centre of Excellence for White-green Biotechnology, Slovak Academy of Sciences, Nitra, Slovakia
| |
Collapse
|
29
|
Giovannoni F, Li Z, Remes-Lenicov F, Dávola ME, Elizalde M, Paletta A, Ashkar AA, Mossman KL, Dugour AV, Figueroa JM, Barquero AA, Ceballos A, Garcia CC, Quintana FJ. AHR signaling is induced by infection with coronaviruses. Nat Commun 2021; 12:5148. [PMID: 34446714 PMCID: PMC8390748 DOI: 10.1038/s41467-021-25412-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Coronavirus infection in humans is usually associated to respiratory tract illnesses, ranging in severity from mild to life-threatening respiratory failure. The aryl hydrocarbon receptor (AHR) was recently identified as a host factor for Zika and dengue viruses; AHR antagonists boost antiviral immunity, decrease viral titers and ameliorate Zika-induced pathology in vivo. Here we report that AHR is activated by infection with different coronaviruses, potentially impacting antiviral immunity and lung epithelial cells. Indeed, the analysis of single-cell RNA-seq from lung tissue detected increased expression of AHR and AHR transcriptional targets, suggesting AHR signaling activation in SARS-CoV-2-infected epithelial cells from COVID-19 patients. Moreover, we detected an association between AHR expression and viral load in SARS-CoV-2 infected patients. Finally, we found that the pharmacological inhibition of AHR suppressed the replication in vitro of one of the causative agents of the common cold, HCoV-229E, and the causative agent of the COVID-19 pandemic, SARS-CoV-2. Taken together, these findings suggest that AHR activation is a common strategy used by coronaviruses to evade antiviral immunity and promote viral replication, which may also contribute to lung pathology. Future studies should further evaluate the potential of AHR as a target for host-directed antiviral therapy.
Collapse
Affiliation(s)
- Federico Giovannoni
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhaorong Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Federico Remes-Lenicov
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María E Dávola
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Mercedes Elizalde
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Paletta
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ali A Ashkar
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Karen L Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Andrea V Dugour
- Instituto de Ciencia y Tecnología Dr. Cesar Milstein (Consejo Nacional de Investigaciones Científicas y Técnicas-Fundacion Cassara), Buenos Aires, Argentina
| | - Juan M Figueroa
- Instituto de Ciencia y Tecnología Dr. Cesar Milstein (Consejo Nacional de Investigaciones Científicas y Técnicas-Fundacion Cassara), Buenos Aires, Argentina
| | - Andrea A Barquero
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. CONICET- Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Ana Ceballos
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cybele C Garcia
- Laboratorio de Estrategias Antivirales, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. CONICET- Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina.
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
30
|
Kaczmarczyk LS, Marsay KS, Shevchenko S, Pilossof M, Levi N, Einat M, Oren M, Gerlitz G. Corona and polio viruses are sensitive to short pulses of W-band gyrotron radiation. ENVIRONMENTAL CHEMISTRY LETTERS 2021; 19:3967-3972. [PMID: 34456659 PMCID: PMC8385265 DOI: 10.1007/s10311-021-01300-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/28/2021] [Indexed: 06/03/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has raised the need of versatile means for virus decontamination. Millimeter waves are used in biochemical research in dynamic nuclear polarization enhanced nuclear magnetic resonance (DNP/NMR) spectroscopy. However, their efficiency in object decontamination for viruses has not been tested yet. Here we report the high efficiency of 95 GHz waves in killing both coronavirus 229E and poliovirus. An exposure of 2 s to 95 GHz waves reduced the titer of these viruses by 99.98% and 99.375%, respectively, and formed holes in the envelope of 229E virions as detected by scanning electron microscopy (SEM) analysis. The ability of 95 GHz waves to reduce the coronavirus titer to a range of limited infective dose of SARS-CoV-2 for humans and animal models along with precise focusing capabilities for these waves suggest 95 GHz waves as an effective way to decontaminate objects.
Collapse
Affiliation(s)
- Lukasz S. Kaczmarczyk
- Department of Molecular Biology, Faculty of Life Sciences, Faculty of Natural Sciences, Ariel University, Kiryat Hamada, 40700 Ariel, Israel
- Ariel Center for Applied Cancer Research, Ariel University, Ariel, Israel
| | - Katherine S. Marsay
- Department of Molecular Biology, Faculty of Life Sciences, Faculty of Natural Sciences, Ariel University, Kiryat Hamada, 40700 Ariel, Israel
| | - Sergey Shevchenko
- Department of Electrical Engineering and Electronics, Faculty of Engineering, Ariel University, Ariel, Israel
| | - Moritz Pilossof
- Department of Electrical Engineering and Electronics, Faculty of Engineering, Ariel University, Ariel, Israel
| | - Nehora Levi
- Department of Molecular Biology, Faculty of Life Sciences, Faculty of Natural Sciences, Ariel University, Kiryat Hamada, 40700 Ariel, Israel
- Ariel Center for Applied Cancer Research, Ariel University, Ariel, Israel
| | - Moshe Einat
- Department of Electrical Engineering and Electronics, Faculty of Engineering, Ariel University, Ariel, Israel
| | - Matan Oren
- Department of Molecular Biology, Faculty of Life Sciences, Faculty of Natural Sciences, Ariel University, Kiryat Hamada, 40700 Ariel, Israel
| | - Gabi Gerlitz
- Department of Molecular Biology, Faculty of Life Sciences, Faculty of Natural Sciences, Ariel University, Kiryat Hamada, 40700 Ariel, Israel
- Ariel Center for Applied Cancer Research, Ariel University, Ariel, Israel
| |
Collapse
|
31
|
Ultraviolet-A light reduces cellular cytokine release from human endotracheal cells infected with Coronavirus. Photodiagnosis Photodyn Ther 2021; 35:102457. [PMID: 34314863 PMCID: PMC8310417 DOI: 10.1016/j.pdpdt.2021.102457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/12/2021] [Accepted: 07/22/2021] [Indexed: 01/08/2023]
Abstract
Background An important clinical feature of coronavirus disease 2019 (COVID-19) is hypercytokinemia (cytokine storm). We previously showed that narrow band ultraviolet-A (NB-UVA) treatment salvages coronavirus (CoV)-229E-infected human tracheal cells, and that daily endotracheal NB-UVA therapy reduced severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) levels in human subjects, with improved clinical outcomes. Here, we examined NB-UVA effects on cytokine release during CoV-229E infection. Methods Primary human tracheal epithelial cells were transfected with CoV-229E, then exposed to 2 mW/cm2 NB-UVA for 20 minutes every 24h, either 3 or 4 times. Secreted cytokine/chemokine levels were analyzed in supernatants collected from CoV-229E-infected/UVA-exposed cells 24h after the last UVA treatment, and from matched non-infected/UVA-exposed controls, CoV-229E-infected/non-exposed controls, and non-infected/non-exposed (naïve) controls. Metabolic pathway/downstream prediction analyses were also performed. Results Pro-inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF), and chemokines IL-8, monocyte chemoattractant protein-1 (MCP1), and interferon gamma-induced protein 10 (IP-10), were significantly increased in CoV-229E-infected cells, and significantly decreased following NB-UVA treatment. Interferon (IFN)-α2, IFN-γ, and IL-10 were not upregulated in response to CoV-229E. Metabolic pathway predictions indicated hypercytokinemia as the top inflammatory response in CoV-229E-infected cells, whereas the top predicted pathway in CoV-229E-infected/UVA-exposed cells was the recovery stage of severe acute respiratory syndrome. Conclusions Human tracheal epithelial cells infected with CoV-229E showed reduced cytokine secretions including IL-6, TNF, IL-8, and MCP-1, following NB-UVA exposure. This reduction of cytokine levels in vitro, coupled with previously identified reduced cell death in CoV-229E-infected/UVA-exposed cells, suggests that determining UVA effects on cytokine storm in human SARS-Co-V2 patients is warranted.
Collapse
|
32
|
Overcoming Culture Restriction for SARS-CoV-2 in Human Cells Facilitates the Screening of Compounds Inhibiting Viral Replication. Antimicrob Agents Chemother 2021; 65:e0009721. [PMID: 33903110 PMCID: PMC8406809 DOI: 10.1128/aac.00097-21] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Efforts to mitigate the coronavirus disease 2019 (COVID-19) pandemic include the screening of existing antiviral molecules that could be repurposed to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Although SARS-CoV-2 replicates and propagates efficiently in African green monkey kidney (Vero) cells, antivirals such as nucleos(t)ide analogs (NUCs) often show decreased activity in these cells due to inefficient metabolization. SARS-CoV-2 exhibits low viability in human cells in culture. Here, serial passages of a SARS-CoV-2 isolate (original-SARS2) in the human hepatoma cell clone Huh7.5 led to the selection of a variant (adapted-SARS2) with significantly improved infectivity in human liver (Huh7 and Huh7.5) and lung cancer (unmodified Calu-1 and A549) cells. The adapted virus exhibited mutations in the spike protein, including a 9-amino-acid deletion and 3 amino acid changes (E484D, P812R, and Q954H). E484D also emerged in Vero E6-cultured viruses that became viable in A549 cells. Original and adapted viruses were susceptible to scavenger receptor class B type 1 (SR-B1) receptor blocking, and adapted-SARS2 exhibited significantly less dependence on ACE2. Both variants were similarly neutralized by COVID-19 convalescent-phase plasma, but adapted-SARS2 exhibited increased susceptibility to exogenous type I interferon. Remdesivir inhibited original- and adapted-SARS2 similarly, demonstrating the utility of the system for the screening of NUCs. Among the tested NUCs, only remdesivir, molnupiravir, and, to a limited extent, galidesivir showed antiviral effects across human cell lines, whereas sofosbuvir, ribavirin, and favipiravir had no apparent activity. Analogously to the emergence of spike mutations in vivo, the spike protein is under intense adaptive selection pressure in cell culture. Our results indicate that the emergence of spike mutations will most likely not affect the activity of remdesivir.
Collapse
|
33
|
Salman AA, Waheed MH, Ali-Abdulsahib AA, Atwan ZW. Low type I interferon response in COVID-19 patients: Interferon response may be a potential treatment for COVID-19. Biomed Rep 2021; 14:43. [PMID: 33786172 PMCID: PMC7995242 DOI: 10.3892/br.2021.1419] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Interferons (IFN) are antiviral cytokines that mitigate the effects of invading viruses early on during the infection process. SARS-CoV and MERS induce weak IFN responses; hence, the clinical trials which included recombinant IFN accompanied with other antiviral drugs exhibited improved results in terms of shortening the duration of illness. The aim of the present study was to evaluate the type I IFN response in COVID-19 patients to determine whether it is sufficient to eliminate or reduce the severity of the infection, and whether it can be recommended as a potential therapy. Total RNA samples were converted to cDNA and used as templates to evaluate the gene expression levels of IFN regulatory factor (IRF)3 and IFN-β in COVID-19 patients or control. The results showed that IRF3 gene expression was upregulated ~250-fold compared with the negative samples. In contrast, IFN-β expression increased slightly in COVID-19 patients. Consistent with other coronaviruses, such as SARS-CoV and MERS, COVID-19 infection does not induce an efficient IFN response to reduce the severity of the virus. This may be attributed to an incomplete response of IRF3 in activating the IFN-β promoter in the infected patients. The results suggest IFN-β or α may be used as potential treatments.
Collapse
Affiliation(s)
| | | | | | - Zeenah Weheed Atwan
- Genetic Engineering Laboratory, Biology Department, College of Science, Basrah University, Basrah, Iraq
| |
Collapse
|
34
|
Trimarco JD, Heaton BE, Chaparian RR, Burke KN, Binder RA, Gray GC, Smith CM, Menachery VD, Heaton NS. TMEM41B is a host factor required for the replication of diverse coronaviruses including SARS-CoV-2. PLoS Pathog 2021; 17:e1009599. [PMID: 34043740 PMCID: PMC8189496 DOI: 10.1371/journal.ppat.1009599] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/09/2021] [Accepted: 04/29/2021] [Indexed: 12/23/2022] Open
Abstract
Antiviral therapeutics are a front-line defense against virally induced diseases. Because viruses frequently mutate to escape direct inhibition of viral proteins, there is interest in targeting the host proteins that the virus must co-opt to complete its replication cycle. However, a detailed understanding of the interactions between the virus and the host cell is necessary in order to facilitate development of host-directed therapeutics. As a first step, we performed a genome-wide loss of function screen using the alphacoronavirus HCoV-229E to better define the interactions between coronaviruses and host factors. We report the identification and validation of an ER-resident host protein, TMEM41B, as an essential host factor for not only HCoV-229E but also genetically distinct coronaviruses including the pandemic betacoronavirus SARS-CoV-2. We show that the protein is required at an early, but post-receptor engagement, stage of the viral lifecycle. Further, mechanistic studies revealed that although the protein was not enriched at replication complexes, it likely contributes to viral replication complex formation via mobilization of cholesterol and other lipids to facilitate host membrane expansion and curvature. Continued study of TMEM41B and the development of approaches to prevent its function may lead to broad spectrum anti-coronavirus therapeutics.
Collapse
Affiliation(s)
- Joseph D. Trimarco
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Brook E. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Ryan R. Chaparian
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Kaitlyn N. Burke
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Raquel A. Binder
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
| | - Gregory C. Gray
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
| | - Clare M. Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Vineet D. Menachery
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
35
|
Abstract
Severe Acute Respiratory Syndrome-Coronavirus-2 is responsible for the current pandemic that has led to more than 10 million confirmed cases of Coronavirus Disease-19 (COVID-19) and over 500,000 deaths worldwide (4 July 2020). Virus-mediated injury to multiple organs, mainly the respiratory tract, activation of immune response with the release of pro-inflammatory cytokines, and overactivation of the coagulation cascade and platelet aggregation leading to micro- and macrovascular thrombosis are the main pathological features of COVID-19. Empirical multidrug therapeutic approaches to treat COVID-19 are currently used with extremely uncertain outcomes, and many others are being tested in clinical trials. Acetylsalicylic acid (ASA) has both anti-inflammatory and antithrombotic effects. In addition, a significant ASA-mediated antiviral activity against DNA and RNA viruses, including different human coronaviruses, has been documented. The use of ASA in patients with different types of infections has been associated with reduced thrombo-inflammation and lower rates of clinical complications and in-hospital mortality. However, safety issues related both to the risk of bleeding and to that of developing rare but serious liver and brain damage mostly among children (i.e., Reye's syndrome) should be considered. Hence, whether ASA might be a safe and reasonable therapeutic candidate to be tested in clinical trials involving adults with COVID-19 deserves further attention. In this review we provide a critical appraisal of current evidence on the anti-inflammatory, antithrombotic, and antiviral effects of ASA, from both a pre-clinical and a clinical perspective. In addition, the potential benefits and risks of use of ASA have been put in the context of the adult-restricted COVID-19 population.
Collapse
|
36
|
Gao X, Zhang L, Zhou P, Zhang Y, Wei Y, Wang Y, Liu X. Tandem Mass Tag-Based Quantitative Proteome Analysis of Porcine Deltacoronavirus (PDCoV)-Infected LLC Porcine Kidney Cells. ACS OMEGA 2020; 5:21979-21987. [PMID: 32923756 PMCID: PMC7482077 DOI: 10.1021/acsomega.0c00886] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 08/12/2020] [Indexed: 05/12/2023]
Abstract
Porcine deltacoronavirus (PDCoV) is a newly emerging porcine pathogenic enteric coronavirus that can cause diarrhea, vomiting, dehydration, and a high mortality rate in piglets. At present, the understanding of PDCoV pathogenesis is very limited, which seriously hinders effective prevention and control. In this study, liquid chromatography tandem-mass spectrometry (LC-MS/MS) combined with tandem mass tag (TMT) labeling was performed to compare the differential expression of proteins in PDCoV-infected and mock-infected LLC-PK cells at 18 h post-infection (hpi). In addition, the parallel reaction monitoring (PRM) technique was used to verify the quantitative proteome data. A total of 4624 differentially expressed proteins (DEPs) were quantitated, of which 128 were significantly upregulated, and 147 were significantly downregulated. Bioinformatics analysis revealed that these DEPs were involved mainly in the defense response, apoptosis, and the immune system, and several DEPs may be related to interferon-stimulated genes and the immune system. Based on DEP bioinformatics analysis, we propose that PDCoV infection may utilize the apoptosis pathway of host cells to achieve maximum viral replication. Meanwhile, the host may be able to stimulate the transcription of interferon-stimulated genes (ISGs) through the JAK/STAT signaling pathway to resist the virus. Overall, in this study, we presented the first application of proteomics analysis to determine the protein profile of PDCoV-infected cells, which provides valuable information with respect to better understanding the host response to PDCoV infection and the specific pathogenesis of PDCoV infection.
Collapse
Affiliation(s)
- Xiang Gao
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- College
of Veterinary Medicine, Gansu Agricultural
University, Lanzhou 730070, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Liping Zhang
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Peng Zhou
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yongguang Zhang
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yanming Wei
- College
of Veterinary Medicine, Gansu Agricultural
University, Lanzhou 730070, China
| | - Yonglu Wang
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Xinsheng Liu
- State
Key Laboratory of Veterinary Etiological Biology, Key Laboratory of
Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research
Institute, Chinese Academy of Agricultural
Sciences, Lanzhou 730046, China
- Jiangsu
Co-innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou 225009, China
| |
Collapse
|
37
|
Abstract
Purpose of Review To review current literature on endothelial dysfunction with previous coronaviruses, and present available data on the role of endothelial dysfunction in coronavirus disease-2019 (COVID-19) infection in terms of pathophysiology and clinical phenotype Recent Findings Recent evidence suggests that signs and symptoms of severe COVID-19 infection resemble the clinical phenotype of endothelial dysfunction, implicating mutual pathophysiological pathways. Dysfunction of endothelial cells is believed to mediate a variety of viral infections, including those caused by previous coronaviruses. Experience from previous coronaviruses has triggered hypotheses on the role of endothelial dysfunction in the pathophysiology of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), which are currently being tested in preclinical and clinical studies. Summary Endothelial dysfunction is the common denominator of multiple clinical aspects of severe COVID-19 infection that have been problematic for treating physicians. Given the global impact of this pandemic, better understanding of the pathophysiology could significantly affect management of patients.
Collapse
|
38
|
Coagulation dysfunction in COVID-19: The interplay between inflammation, viral infection and the coagulation system. Blood Rev 2020; 46:100745. [PMID: 32868115 PMCID: PMC7444609 DOI: 10.1016/j.blre.2020.100745] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 is a new pandemic, caused by Severe Acute Respiratory Syndrome-CoronaVirus-2 (SARS-Cov2) infection and characterized by a broad spectrum of clinical manifestations. Inflammation and the innate immune system have been recently recognized as pivotal players in the most severe forms, characterized by significantly elevated levels of pro-inflammatory cytokines. In this setting, several studies have also reported the presence of abnormalities in coagulation parameters and platelets count, possibly identifying a subgroup of patients with poor prognosis. Some reports of full-blown thromboembolic events are emerging. Among the possible mechanisms underlying coagulation dysfunction, the so-called "cytokine storm" seems to play a pivotal role. Other candidate factors include virus-specific mechanisms, related to the virus interaction with renin angiotensin system (RAS) and the fibrinolytic pathway, but also comorbidities affecting these patients. Coagulation dysfunction is therefore a candidate risk factor for adverse outcomes in COVID-19 and should be carefully addressed in clinical practice.
Collapse
|
39
|
Appelberg S, Gupta S, Svensson Akusjärvi S, Ambikan AT, Mikaeloff F, Saccon E, Végvári Á, Benfeitas R, Sperk M, Ståhlberg M, Krishnan S, Singh K, Penninger JM, Mirazimi A, Neogi U. Dysregulation in Akt/mTOR/HIF-1 signaling identified by proteo-transcriptomics of SARS-CoV-2 infected cells. Emerg Microbes Infect 2020; 9:1748-1760. [PMID: 32691695 PMCID: PMC7473213 DOI: 10.1080/22221751.2020.1799723] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
How severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections engage cellular host pathways and innate immunity in infected cells remains largely elusive. We performed an integrative proteo-transcriptomics analysis in SARS-CoV-2 infected Huh7 cells to map the cellular response to the invading virus over time. We identified four pathways, ErbB, HIF-1, mTOR and TNF signaling, among others that were markedly modulated during the course of the SARS-CoV-2 infection in vitro. Western blot validation of the downstream effector molecules of these pathways revealed a dose-dependent activation of Akt, mTOR, S6K1 and 4E-BP1 at 24 hours post infection (hpi). However, we found a significant inhibition of HIF-1α through 24hpi and 48hpi of the infection, suggesting a crosstalk between the SARS-CoV-2 and the Akt/mTOR/HIF-1 signaling pathways. Inhibition of the mTOR signaling pathway using Akt inhibitor MK-2206 showed a significant reduction in virus production. Further investigations are required to better understand the molecular sequelae in order to guide potential therapy in the management of severe coronavirus disease 2019 (COVID-19) patients.
Collapse
Affiliation(s)
| | - Soham Gupta
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Sara Svensson Akusjärvi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anoop T Ambikan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Flora Mikaeloff
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Elisa Saccon
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rui Benfeitas
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University Stockholm, Sweden
| | - Maike Sperk
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Marie Ståhlberg
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Shuba Krishnan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Kamal Singh
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Veterinary Pathobiology and the Bond Life Science Center, University of Missouri, Columbia, MO, USA
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, Canada
| | - Ali Mirazimi
- Public Health Agency of Sweden, Solna, Sweden.,Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,National Veterinary Institute, Uppsala, Sweden
| | - Ujjwal Neogi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Veterinary Pathobiology and the Bond Life Science Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
40
|
Ultraviolet A light effectively reduces bacteria and viruses including coronavirus. PLoS One 2020; 15:e0236199. [PMID: 32673355 PMCID: PMC7365468 DOI: 10.1371/journal.pone.0236199] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
Antimicrobial-resistant and novel pathogens continue to emerge, outpacing efforts to contain and treat them. Therefore, there is a crucial need for safe and effective therapies. Ultraviolet-A (UVA) phototherapy is FDA-approved for several dermatological diseases but not for internal applications. We investigated UVA effects on human cells in vitro, mouse colonic tissue in vivo, and UVA efficacy against bacteria, yeast, coxsackievirus group B and coronavirus-229E. Several pathogens and virally transfected human cells were exposed to a series of specific UVA exposure regimens. HeLa, alveolar and primary human tracheal epithelial cell viability was assessed after UVA exposure, and 8-Oxo-2'-deoxyguanosine was measured as an oxidative DNA damage marker. Furthermore, wild-type mice were exposed to intracolonic UVA as an in vivo model to assess safety of internal UVA exposure. Controlled UVA exposure yielded significant reductions in Pseudomonas aeruginosa, Klebsiella pneumoniae, Escherichia coli, Enterococcus faecalis, Clostridioides difficile, Streptococcus pyogenes, Staphylococcus epidermidis, Proteus mirabilis and Candida albicans. UVA-treated coxsackievirus-transfected HeLa cells exhibited significantly increased cell survival compared to controls. UVA-treated coronavirus-229E-transfected tracheal cells exhibited significant coronavirus spike protein reduction, increased mitochondrial antiviral-signaling protein and decreased coronavirus-229E-induced cell death. Specific controlled UVA exposure had no significant effect on growth or 8-Oxo-2'-deoxyguanosine levels in three types of human cells. Single or repeated in vivo intraluminal UVA exposure produced no discernible endoscopic, histologic or dysplastic changes in mice. These findings suggest that, under specific conditions, UVA reduces various pathogens including coronavirus-229E, and may provide a safe and effective treatment for infectious diseases of internal viscera. Clinical studies are warranted to further elucidate the safety and efficacy of UVA in humans.
Collapse
|
41
|
Giannis D, Ziogas IA, Gianni P. Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol 2020; 127:104362. [PMID: 32305883 PMCID: PMC7195278 DOI: 10.1016/j.jcv.2020.104362] [Citation(s) in RCA: 649] [Impact Index Per Article: 129.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 04/05/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus strain disease, has recently emerged in China and rapidly spread worldwide. This novel strain is highly transmittable and severe disease has been reported in up to 16% of hospitalized cases. More than 600,000 cases have been confirmed and the number of deaths is constantly increasing. COVID-19 hospitalized patients, especially those suffering from severe respiratory or systemic manifestations, fall under the spectrum of the acutely ill medical population, which is at increased venous thromboembolism risk. Thrombotic complications seem to emerge as an important issue in patients infected with COVID-19. Preliminary reports on COVID-19 patients' clinical and laboratory findings include thrombocytopenia, elevated D-dimer, prolonged prothrombin time, and disseminated intravascular coagulation. As the pandemic is spreading and the whole picture is yet unknown, we highlight the importance of coagulation disorders in COVID-19 infected patients and review relevant data of previous coronavirus epidemics caused by the severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) and the Middle East Respiratory Syndrome coronavirus (MERS-CoV).
Collapse
Affiliation(s)
- Dimitrios Giannis
- Institute of Health Innovations and Outcomes Research, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA; Surgery Working Group, Society of Junior Doctors, Athens, Greece.
| | - Ioannis A Ziogas
- Surgery Working Group, Society of Junior Doctors, Athens, Greece; Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Panagiota Gianni
- Department of Internal Medicine III, Hematology, Oncology, Palliative Medicine, Rheumatology and Infectious Diseases, University Hospital Ulm, Ulm 89070, Germany.
| |
Collapse
|
42
|
The FDA-approved gold drug auranofin inhibits novel coronavirus (SARS-COV-2) replication and attenuates inflammation in human cells. Virology 2020; 547:7-11. [PMID: 32442105 PMCID: PMC7236683 DOI: 10.1016/j.virol.2020.05.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 11/25/2022]
Abstract
SARS-COV-2 has recently emerged as a new public health threat. Herein, we report that the FDA-approved drug, auranofin, inhibits SARS-COV-2 replication in human cells at low micro molar concentration. Treatment of cells with auranofin resulted in a 95% reduction in the viral RNA at 48 h after infection. Auranofin treatment dramatically reduced the expression of SARS-COV-2-induced cytokines in human cells. These data indicate that auranofin could be a useful drug to limit SARS-CoV-2 infection and associated lung injury due to its antiviral, anti-inflammatory and anti-reactive oxygen species (ROS) properties. Further animal studies are warranted to evaluate the safety and efficacy of auranofin for the management of SARS-COV-2 associated disease. Auranofin inhibits replication of SARS-COV-2 in human cells at low micro molar concentration. Auranofin treatment resulted in significant reduction in SARS-COV-2-induced cytokines in human cells. Auranofin could mitigate SARS-COV-2 infection and lung damage due to its anti-viral and anti-inflammatory properties. Auranofin is a gold-containing FDA-approved drug.
Collapse
|
43
|
Fung SY, Yuen KS, Ye ZW, Chan CP, Jin DY. A tug-of-war between severe acute respiratory syndrome coronavirus 2 and host antiviral defence: lessons from other pathogenic viruses. Emerg Microbes Infect 2020; 9:558-570. [PMID: 32172672 PMCID: PMC7103735 DOI: 10.1080/22221751.2020.1736644] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
World Health Organization has declared the ongoing outbreak of coronavirus disease 2019 (COVID-19) a Public Health Emergency of International Concern. The virus was named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by the International Committee on Taxonomy of Viruses. Human infection with SARS-CoV-2 leads to a wide range of clinical manifestations ranging from asymptomatic, mild, moderate to severe. The severe cases present with pneumonia, which can progress to acute respiratory distress syndrome. The outbreak provides an opportunity for real-time tracking of an animal coronavirus that has just crossed species barrier to infect humans. The outcome of SARS-CoV-2 infection is largely determined by virus-host interaction. Here, we review the discovery, zoonotic origin, animal hosts, transmissibility and pathogenicity of SARS-CoV-2 in relation to its interplay with host antiviral defense. A comparison with SARS-CoV, Middle East respiratory syndrome coronavirus, community-acquired human coronaviruses and other pathogenic viruses including human immunodeficiency viruses is made. We summarize current understanding of the induction of a proinflammatory cytokine storm by other highly pathogenic human coronaviruses, their adaptation to humans and their usurpation of the cell death programmes. Important questions concerning the interaction between SARS-CoV-2 and host antiviral defence, including asymptomatic and presymptomatic virus shedding, are also discussed.
Collapse
Affiliation(s)
- Sin-Yee Fung
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kit-San Yuen
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Zi-Wei Ye
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
44
|
Siu KL, Yuen KS, Castaño-Rodriguez C, Ye ZW, Yeung ML, Fung SY, Yuan S, Chan CP, Yuen KY, Enjuanes L, Jin DY. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. FASEB J 2019; 33:8865-8877. [PMID: 31034780 DOI: 10.1096/fj.201802418r] [Citation(s) in RCA: 395] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) is capable of inducing a storm of proinflammatory cytokines. In this study, we show that the SARS-CoV open reading frame 3a (ORF3a) accessory protein activates the NLRP3 inflammasome by promoting TNF receptor-associated factor 3 (TRAF3)-mediated ubiquitination of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). SARS-CoV and its ORF3a protein were found to be potent activators of pro-IL-1β gene transcription and protein maturation, the 2 signals required for activation of the NLRP3 inflammasome. ORF3a induced pro-IL-1β transcription through activation of NF-κB, which was mediated by TRAF3-dependent ubiquitination and processing of p105. ORF3a-induced elevation of IL-1β secretion was independent of its ion channel activity or absent in melanoma 2 but required NLRP3, ASC, and TRAF3. ORF3a interacted with TRAF3 and ASC, colocalized with them in discrete punctate structures in the cytoplasm, and facilitated ASC speck formation. TRAF3-dependent K63-linked ubiquitination of ASC was more pronounced in SARS-CoV-infected cells or when ORF3a was expressed. Taken together, our findings reveal a new mechanism by which SARS-CoV ORF3a protein activates NF-κB and the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of p105 and ASC.-Siu, K.-L., Yuen, K.-S., Castaño-Rodriguez, C., Ye, Z.-W., Yeung, M.-L., Fung, S.-Y., Yuan, S., Chan, C.-P., Yuen, K.-Y., Enjuanes, L., Jin, D.-Y. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC.
Collapse
Affiliation(s)
- Kam-Leung Siu
- School of Biomedical Sciences, The University of Hong Kong, PokFuLam, Hong Kong
| | - Kit-San Yuen
- School of Biomedical Sciences, The University of Hong Kong, PokFuLam, Hong Kong
| | - Carlos Castaño-Rodriguez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Zi-Wei Ye
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Man-Lung Yeung
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Sin-Yee Fung
- School of Biomedical Sciences, The University of Hong Kong, PokFuLam, Hong Kong
| | - Shuofeng Yuan
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, PokFuLam, Hong Kong
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, PokFuLam, Hong Kong
| |
Collapse
|
45
|
Yan B, Chu H, Yang D, Sze KH, Lai PM, Yuan S, Shuai H, Wang Y, Kao RYT, Chan JFW, Yuen KY. Characterization of the Lipidomic Profile of Human Coronavirus-Infected Cells: Implications for Lipid Metabolism Remodeling upon Coronavirus Replication. Viruses 2019; 11:v11010073. [PMID: 30654597 PMCID: PMC6357182 DOI: 10.3390/v11010073] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/19/2022] Open
Abstract
Lipids play numerous indispensable cellular functions and are involved in multiple steps in the replication cycle of viruses. Infections by human-pathogenic coronaviruses result in diverse clinical outcomes, ranging from self-limiting flu-like symptoms to severe pneumonia with extrapulmonary manifestations. Understanding how cellular lipids may modulate the pathogenicity of human-pathogenic coronaviruses remains poor. To this end, we utilized the human coronavirus 229E (HCoV-229E) as a model coronavirus to comprehensively characterize the host cell lipid response upon coronavirus infection with an ultra-high performance liquid chromatography-mass spectrometry (UPLC–MS)-based lipidomics approach. Our results revealed that glycerophospholipids and fatty acids (FAs) were significantly elevated in the HCoV-229E-infected cells and the linoleic acid (LA) to arachidonic acid (AA) metabolism axis was markedly perturbed upon HCoV-229E infection. Interestingly, exogenous supplement of LA or AA in HCoV-229E-infected cells significantly suppressed HCoV-229E virus replication. Importantly, the inhibitory effect of LA and AA on virus replication was also conserved for the highly pathogenic Middle East respiratory syndrome coronavirus (MERS-CoV). Taken together, our study demonstrated that host lipid metabolic remodeling was significantly associated with human-pathogenic coronavirus propagation. Our data further suggested that lipid metabolism regulation would be a common and druggable target for coronavirus infections.
Collapse
Affiliation(s)
- Bingpeng Yan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Dong Yang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Kong-Hung Sze
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Pok-Man Lai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Huiping Shuai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Yixin Wang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Richard Yi-Tsun Kao
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Hainan-Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 96708, China.
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Hainan-Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 96708, China.
- The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
| |
Collapse
|
46
|
Cervera H, Ambrós S, Bernet GP, Rodrigo G, Elena SF. Viral Fitness Correlates with the Magnitude and Direction of the Perturbation Induced in the Host's Transcriptome: The Tobacco Etch Potyvirus-Tobacco Case Study. Mol Biol Evol 2018; 35:1599-1615. [PMID: 29562354 PMCID: PMC5995217 DOI: 10.1093/molbev/msy038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Determining the fitness of viral genotypes has become a standard practice in virology as it is essential to evaluate their evolutionary potential. Darwinian fitness, defined as the advantage of a given genotype with respect to a reference one, is a complex property that captures, in a single figure, differences in performance at every stage of viral infection. To what extent does viral fitness result from specific molecular interactions with host factors and regulatory networks during infection? Can we identify host genes in functional classes whose expression depends on viral fitness? Here, we compared the transcriptomes of tobacco plants infected with seven genotypes of tobacco etch potyvirus that differ in fitness. We found that the larger the fitness differences among genotypes, the more dissimilar the transcriptomic profiles are. Consistently, two different mutations, one in the viral RNA polymerase and another in the viral suppressor of RNA silencing, resulted in significantly similar gene expression profiles. Moreover, we identified host genes whose expression showed a significant correlation, positive or negative, with the virus' fitness. Differentially expressed genes which were positively correlated with viral fitness activate hormone- and RNA silencing-mediated pathways of plant defense. In contrast, those that were negatively correlated with fitness affect metabolism, reducing growth, and development. Overall, these results reveal the high information content of viral fitness and suggest its potential use to predict differences in genomic profiles of infected hosts.
Collapse
Affiliation(s)
- Héctor Cervera
- Instituto de Biología Molecular y Celular de Plantas (IBMCP), CSIC-Universitat Politècnia de València, Campus UPV CPI 8E, València, Spain
| | - Silvia Ambrós
- Instituto de Biología Molecular y Celular de Plantas (IBMCP), CSIC-Universitat Politècnia de València, Campus UPV CPI 8E, València, Spain
| | - Guillermo P Bernet
- Instituto de Biología Molecular y Celular de Plantas (IBMCP), CSIC-Universitat Politècnia de València, Campus UPV CPI 8E, València, Spain
| | - Guillermo Rodrigo
- Instituto de Biología Molecular y Celular de Plantas (IBMCP), CSIC-Universitat Politècnia de València, Campus UPV CPI 8E, València, Spain
- Instituto de Biología Integrativa de Sistemas (ISysBio), CSIC-Universitat de València, Parc Científic UV, Catedrático Agustín Escardino 9, Paterna, València, Spain
| | - Santiago F Elena
- Instituto de Biología Molecular y Celular de Plantas (IBMCP), CSIC-Universitat Politècnia de València, Campus UPV CPI 8E, València, Spain
- Instituto de Biología Integrativa de Sistemas (ISysBio), CSIC-Universitat de València, Parc Científic UV, Catedrático Agustín Escardino 9, Paterna, València, Spain
- The Santa Fe Institute, Santa Fe, NM
| |
Collapse
|
47
|
The NF-κB-dependent and -independent transcriptome and chromatin landscapes of human coronavirus 229E-infected cells. PLoS Pathog 2017; 13:e1006286. [PMID: 28355270 PMCID: PMC5386326 DOI: 10.1371/journal.ppat.1006286] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/10/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
Coronavirus replication takes place in the host cell cytoplasm and triggers inflammatory gene expression by poorly characterized mechanisms. To obtain more insight into the signals and molecular events that coordinate global host responses in the nucleus of coronavirus-infected cells, first, transcriptome dynamics was studied in human coronavirus 229E (HCoV-229E)-infected A549 and HuH7 cells, respectively, revealing a core signature of upregulated genes in these cells. Compared to treatment with the prototypical inflammatory cytokine interleukin(IL)-1, HCoV-229E replication was found to attenuate the inducible activity of the transcription factor (TF) NF-κB and to restrict the nuclear concentration of NF-κB subunits by (i) an unusual mechanism involving partial degradation of IKKβ, NEMO and IκBα and (ii) upregulation of TNFAIP3 (A20), although constitutive IKK activity and basal TNFAIP3 expression levels were shown to be required for efficient virus replication. Second, we characterized actively transcribed genomic regions and enhancers in HCoV-229E-infected cells and systematically correlated the genome-wide gene expression changes with the recruitment of Ser5-phosphorylated RNA polymerase II and prototypical histone modifications (H3K9ac, H3K36ac, H4K5ac, H3K27ac, H3K4me1). The data revealed that, in HCoV-infected (but not IL-1-treated) cells, an extensive set of genes was activated without inducible p65 NF-κB being recruited. Furthermore, both HCoV-229E replication and IL-1 were shown to upregulate a small set of genes encoding immunomodulatory factors that bind p65 at promoters and require IKKβ activity and p65 for expression. Also, HCoV-229E and IL-1 activated a common set of 440 p65-bound enhancers that differed from another 992 HCoV-229E-specific enhancer regions by distinct TF-binding motif combinations. Taken together, the study shows that cytoplasmic RNA viruses fine-tune NF-κB signaling at multiple levels and profoundly reprogram the host cellular chromatin landscape, thereby orchestrating the timely coordinated expression of genes involved in multiple signaling, immunoregulatory and metabolic processes. Coronaviruses are major human and animal pathogens. They belong to a family of plus-strand RNA viruses that have extremely large genomes and encode a variety of proteins involved in virus-host interactions. The four common coronaviruses (HCoV-229E, NL63, OC43, HKU1) cause mainly upper respiratory tract infections, while zoonotic coronaviruses (SARS-CoV and MERS-CoV) cause severe lung disease, including acute respiratory distress syndrome (ARDS). The molecular basis for this fundamentally different pathology is incompletely understood. Our study provides a genome-wide investigation of epigenetic changes occurring in response to HCoV-229E. We identify at high resolution a large number of regulatory regions in the genome of infected cells that coordinate de novo gene transcription. Many of these genes have immunomodulatory functions and, most likely, contribute to limiting viral replication, while other factors may promote viral replication. The study provides an intriguing example of a virus that completes its entire life cycle in the cytoplasm while sending multiple signals to the nuclear chromatin compartment to adjust the host cell repertoire of transcribed genes. The approach taken in this study is expected to provide a suitable framework for future studies aimed at dissecting and comparing host responses to representative coronaviruses with different pathogenic potential in humans.
Collapse
|
48
|
Li W, Luo R, He Q, van Kuppeveld FJM, Rottier PJM, Bosch BJ. Aminopeptidase N is not required for porcine epidemic diarrhea virus cell entry. Virus Res 2017; 235:6-13. [PMID: 28363778 PMCID: PMC7114539 DOI: 10.1016/j.virusres.2017.03.018] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 12/14/2022]
Abstract
Overexpression of porcine APN in cells does not confer susceptibility to PEDV. Knockout APN expression in PEDV-susceptible cells has no effect on PEDV infection. Results demonstrate that APN is not essential for PEDV cell entry.
Porcine epidemic diarrhea virus (PEDV) is an emerging pathogenic coronavirus that causes a significant economic burden to the swine industry. The virus infects the intestinal epithelium and causes villous atrophy, resulting in diarrhea and dehydration. Interaction of the viral spike (S) surface glycoprotein − through its S1 subunit − with the host cell receptor is the first step in infection and the main determinant for virus tropism. As for several other alphacoronaviruses including the porcine transmissible gastroenteritis virus (TGEV) and the human coronavirus 229E (HCoV-229E), the aminopeptidase N (APN) protein was reported to be a functional receptor for PEDV. In this study we examined the role of APN as a receptor. We show that overexpression of porcine APN renders MDCK cells susceptible to TGEV, but not to PEDV. Consistently, unlike TGEV-S1, PEDV-S1 exhibited no binding to cell-surface expressed APN or to a soluble version of APN. Moreover, preincubation of these viruses with soluble APN or pretreatment of APN expressing ST cells with soluble TGEV-S1 blocked TGEV infection, but had no effect on infection by PEDV. The combined observations indicated that APN is not required for PEDV infection. To definitively prove this conclusion, we applied CRISPR/Cas9 genome engineering to knock out APN expression in PEDV-susceptible porcine (ST) and human cell lines (Huh7 and HeLa). As a consequence these cells no longer bound TGEV-S1 and HCoV-229E-S1 at their surface and were resistant to infection by the corresponding viruses. However, genetic ablation of APN expression had no effect on their infectability by PEDV, demonstrating that APN is not essential for PEDV cell entry.
Collapse
Affiliation(s)
- Wentao Li
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Rui Luo
- State Key laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Qigai He
- State Key laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, China
| | - Frank J M van Kuppeveld
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Peter J M Rottier
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Berend-Jan Bosch
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
49
|
Lim YX, Ng YL, Tam JP, Liu DX. Human Coronaviruses: A Review of Virus-Host Interactions. Diseases 2016; 4:E26. [PMID: 28933406 PMCID: PMC5456285 DOI: 10.3390/diseases4030026] [Citation(s) in RCA: 390] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/18/2016] [Accepted: 07/18/2016] [Indexed: 12/19/2022] Open
Abstract
Human coronaviruses (HCoVs) are known respiratory pathogens associated with a range of respiratory outcomes. In the past 14 years, the onset of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) have thrust HCoVs into spotlight of the research community due to their high pathogenicity in humans. The study of HCoV-host interactions has contributed extensively to our understanding of HCoV pathogenesis. In this review, we discuss some of the recent findings of host cell factors that might be exploited by HCoVs to facilitate their own replication cycle. We also discuss various cellular processes, such as apoptosis, innate immunity, ER stress response, mitogen-activated protein kinase (MAPK) pathway and nuclear factor kappa B (NF-κB) pathway that may be modulated by HCoVs.
Collapse
Affiliation(s)
- Yvonne Xinyi Lim
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Yan Ling Ng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - James P Tam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Ding Xiang Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
50
|
Fung TS, Liao Y, Liu DX. Regulation of Stress Responses and Translational Control by Coronavirus. Viruses 2016; 8:v8070184. [PMID: 27384577 PMCID: PMC4974519 DOI: 10.3390/v8070184] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 01/02/2023] Open
Abstract
Similar to other viruses, coronavirus infection triggers cellular stress responses in infected host cells. The close association of coronavirus replication with the endoplasmic reticulum (ER) results in the ER stress responses, which impose a challenge to the viruses. Viruses, in turn, have come up with various mechanisms to block or subvert these responses. One of the ER stress responses is inhibition of the global protein synthesis to reduce the amount of unfolded proteins inside the ER lumen. Viruses have evolved the capacity to overcome the protein translation shutoff to ensure viral protein production. Here, we review the strategies exploited by coronavirus to modulate cellular stress response pathways. The involvement of coronavirus-induced stress responses and translational control in viral pathogenesis will also be briefly discussed.
Collapse
Affiliation(s)
- To Sing Fung
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Ying Liao
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Ziyue Road 518, Shanghai 200241, China.
| | - Ding Xiang Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|