1
|
Hboub H, Ben Mrid R, Bouchmaa N, Oukkache N, El Fatimy R. An in-depth exploration of snake venom-derived molecules for drug discovery in advancing antiviral therapeutics. Heliyon 2024; 10:e37321. [PMID: 39323826 PMCID: PMC11422003 DOI: 10.1016/j.heliyon.2024.e37321] [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: 02/26/2024] [Revised: 07/20/2024] [Accepted: 09/01/2024] [Indexed: 09/27/2024] Open
Abstract
Snake venom is a cocktail and rich source of various bioactive compounds that have been extensively studied for their potential as pharmaceutical agents due to their diverse chemical structures and wide range of biological activities. In light of the emergency and the re-emergence of viral infectious diseases that threaten human health and economic systems, exploring new fertile and rich fields such as snake venom is an attractive path for anti-viral drug discovery, especially in the lack of effective vaccines. Although 85 % of reported antiviral molecules belong to the phospholipase A2 (PLA2) family, other protein families including L-amino acid oxidases (LAAO), disintegrins, metalloproteases (SVMPs), and cathelicidins have also shown antiviral activity. Thus, in this review, we have highlighted the antiviral properties of compounds derived from snake venom and their mechanisms of action against virus classes like HIV, Coronaviridae, Flaviviridae, and Paramyxoviridae. Although the initial research emphasis has been on Retroviridae (HIV) and Flaviviridae viruses, it is crucial to extend the exploration of the potential of these compounds to other viruses. The utilization of snake venom-derived compounds as antivirals shows significant promise for the development of novel therapeutics to address viral infections. However, a more in-depth investigation is necessary to fully assess the potential of these compounds against other viruses and unveil the mechanisms underlying their action.
Collapse
Affiliation(s)
- Hicham Hboub
- Institute of Biological Sciences (ISSB), Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Reda Ben Mrid
- Institute of Biological Sciences (ISSB), Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Najat Bouchmaa
- Institute of Biological Sciences (ISSB), Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| | - Naoual Oukkache
- Laboratory of Venoms and Toxins, Pasteur Institute of Morocco, Casablanca, 20360, Morocco
| | - Rachid El Fatimy
- Institute of Biological Sciences (ISSB), Faculty of Medical Sciences (FMS), Mohammed VI Polytechnic University (UM6P), Ben Guerir, 43150, Morocco
| |
Collapse
|
2
|
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
|
3
|
Žáčková S, Pávová M, Trylčová J, Chalupová J, Priss A, Lukšan O, Weber J. Upregulation of mRNA Expression of ADGRD1/GPR133 and ADGRG7/GPR128 in SARS-CoV-2-Infected Lung Adenocarcinoma Calu-3 Cells. Cells 2024; 13:791. [PMID: 38786015 PMCID: PMC11119037 DOI: 10.3390/cells13100791] [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: 03/26/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Adhesion G protein-coupled receptors (aGPCRs) play an important role in neurodevelopment, immune defence and cancer; however, their role throughout viral infections is mostly unexplored. We have been searching for specific aGPCRs involved in SARS-CoV-2 infection of mammalian cells. In the present study, we infected human epithelial cell lines derived from lung adenocarcinoma (Calu-3) and colorectal carcinoma (Caco-2) with SARS-CoV-2 in order to analyse changes in the level of mRNA encoding individual aGPCRs at 6 and 12 h post infection. Based on significantly altered mRNA levels, we identified four aGPCR candidates-ADGRB3/BAI3, ADGRD1/GPR133, ADGRG7/GPR128 and ADGRV1/GPR98. Of these receptors, ADGRD1/GPR133 and ADGRG7/GPR128 showed the largest increase in mRNA levels in SARS-CoV-2-infected Calu-3 cells, whereas no increase was observed with heat-inactivated SARS-CoV-2 and virus-cleared conditioned media. Next, using specific siRNA, we downregulated the aGPCR candidates and analysed SARS-CoV-2 entry, replication and infectivity in both cell lines. We observed a significant decrease in the amount of SARS-CoV-2 newly released into the culture media by cells with downregulated ADGRD1/GPR133 and ADGRG7/GPR128. In addition, using a plaque assay, we observed a reduction in SARS-CoV-2 infectivity in Calu-3 cells. In summary, our data suggest that selected aGPCRs might play a role during SARS-CoV-2 infection of mammalian cells.
Collapse
Affiliation(s)
- Sandra Žáčková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic; (S.Ž.); (M.P.); (J.T.); (J.C.); (A.P.); (O.L.)
- Department of Genetics and Microbiology, Charles University, Faculty of Sciences, 128 44 Prague, Czech Republic
| | - Marcela Pávová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic; (S.Ž.); (M.P.); (J.T.); (J.C.); (A.P.); (O.L.)
| | - Jana Trylčová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic; (S.Ž.); (M.P.); (J.T.); (J.C.); (A.P.); (O.L.)
| | - Jitka Chalupová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic; (S.Ž.); (M.P.); (J.T.); (J.C.); (A.P.); (O.L.)
| | - Anastasiia Priss
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic; (S.Ž.); (M.P.); (J.T.); (J.C.); (A.P.); (O.L.)
| | - Ondřej Lukšan
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic; (S.Ž.); (M.P.); (J.T.); (J.C.); (A.P.); (O.L.)
| | - Jan Weber
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic; (S.Ž.); (M.P.); (J.T.); (J.C.); (A.P.); (O.L.)
| |
Collapse
|
4
|
Bessonov N, Volpert V. Airway obstruction in respiratory viral infections due to impaired mucociliary clearance. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2023; 39:e3707. [PMID: 37073098 DOI: 10.1002/cnm.3707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/14/2023] [Accepted: 04/02/2023] [Indexed: 05/03/2023]
Abstract
Respiratory viral infections, such as SARS-CoV-2 or influenza, can lead to impaired mucociliary clearance in the bronchial tree due to increased mucus viscosity and its hyper-secretion. We develop in this work a mathematical model to study the interplay between viral infection and mucus motion. The results of numerical simulations show that infection progression can be characterized by three main stages. At the first stage, infection spreads through the most part of mucus producing airways (about 90% of the length) without significant changes in mucus velocity and thickness layer. During the second stage, when it passes through the remaining generations, mucus viscosity increases, its velocity drops down, and it forms a plug. At the last stage, the thickness of the mucus layer gradually increases because mucus is still produced but not removed by the flow. After some time, the thickness of the mucus layer in the small airways becomes comparable with their diameter leading to their complete obstruction.
Collapse
Affiliation(s)
- N Bessonov
- Institute of Problems of Mechanical Engineering, Russian Academy of Sciences, Saint Petersburg, Russian Federation
| | - V Volpert
- Institut Camille Jordan, UMR 5208 CNRS, University Lyon 1, Villeurbanne, France
- S.M. Nikolskii Mathematical Institute, Peoples Friendship University of Russia (RUDN University), Moscow, Russian Federation
| |
Collapse
|
5
|
Drews SJ, O’Brien SF. Lessons Learned from the COVID-19 Pandemic and How Blood Operators Can Prepare for the Next Pandemic. Viruses 2022; 14:2126. [PMID: 36298680 PMCID: PMC9608827 DOI: 10.3390/v14102126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Humans interact with virus-infected animal hosts, travel globally, and maintain social networks that allow for novel viruses to emerge and develop pandemic potential. There are key lessons-learned from the coronavirus diseases 2019 (COVID-19) pandemic that blood operators can apply to the next pandemic. Warning signals to the COVID-19 pandemic included outbreaks of Severe acute respiratory syndrome-related coronavirus-1 (SARS-CoV-1) and Middle East respiratory syndrome-related coronavirus (MERS-CoV) in the prior two decades. It will be critical to quickly determine whether there is a risk of blood-borne transmission of a new pandemic virus. Prior to the next pandemic blood operators should be prepared for changes in activities, policies, and procedures at all levels of the organization. Blood operators can utilize "Plan-Do-Study-Act" cycles spanning from: vigilance for emerging viruses, surveillance activities and studies, operational continuity, donor engagement and trust, and laboratory testing if required. Occupational health and donor safety issues will be key areas of focus even if the next pandemic virus is not transfusion transmitted. Blood operators may also be requested to engage in new activities such as the development of therapeutics or supporting public health surveillance activities. Activities such as scenario development, tabletop exercises, and drills will allow blood operators to prepare for the unknowns of the next pandemic.
Collapse
Affiliation(s)
- Steven J. Drews
- Canadian Blood Services, Microbiology, Donation and Policy Studies, Canadian Blood Services, Edmonton, AB T6G 2R8, Canada
- Division of Applied and Diagnostic Microbiology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sheila F. O’Brien
- Epidemiology and Surveillance, Donation Policy and Studies, Canadian Blood Services, Ottawa, ON K1G 4J5, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
6
|
Masanam MK, Cheney SM, Sutton W, Keyloun JW, Fitzgibbons S. COVID-19 infection and large intestinal perforation: A case series. Int J Surg Case Rep 2022; 98:107538. [PMID: 36027834 PMCID: PMC9395222 DOI: 10.1016/j.ijscr.2022.107538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/14/2022] [Accepted: 08/18/2022] [Indexed: 11/28/2022] Open
Abstract
Introduction COVID-19 is a viral illness characterized primarily by respiratory symptoms. However, patients with COVID-19 infection may also present with gastrointestinal symptoms. Subsequent complications can be associated with high morbidity and mortality. Methods This is a retrospective observational study of three COVID-19 positive patients who developed large intestinal perforations and an analysis of their clinical characteristics, diagnosis, surgical treatment and outcomes. Three patients aged 45, 51 and 82 years old presented to our institution between November 2021 and March 2022 and were diagnosed with COVID-19 pneumonia requiring admission to the intensive care unit (ICU). All three patients received steroids and underwent surgery during their admission. None of our patients had prior history of bowel perforation or risks factors justifying their presentation. Presentation of cases Our first patient was found to have an ascending colon perforation and underwent right colon resection and end ileostomy. Our second patient was found to have a cecal perforation and underwent ileocecectomy with end ileostomy and mucus fistula creation. Our third patient was found to have a large cecal perforation and underwent right hemicolectomy and was left in discontinuity during the index operation. Discussion GI perforation is a less common but serious extra-pulmonary complication of COVID-19. The cases in the present study involve ascending colon perforations in the setting of active COVID-19 infection that occurred within two to five weeks after initial COVID-19 diagnosis. Given viral replication in GI cells, the local inflammatory effect of viral infection in the GI may play a role in bowel perforation. Providers should additionally be aware of the risk of perforation with steroids and immunomodulators. Immunosuppressive effects of these therapies may mask the classical signs of abdominal sepsis and lead to possible missed diagnoses. Conclusion Gastrointestinal perforation is a rare but serious complication of COVID-19 infection. A high degree of clinical suspicion is necessary for timely diagnosis and management. Gastrointestinal perforation is a rare but serious complication of COVID-19 infection In this case series, ascending colon perforation occurs in each case within two to five weeks of initial COVID-19 diagnosis Steroids and immunomodulators pose additional risk of perforation and may mask the classical signs of abdominal sepsis
Collapse
Affiliation(s)
- Monika K Masanam
- Department of Surgery, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Sarah M Cheney
- Georgetown University School of Medicine, Washington, DC, USA
| | - Whitney Sutton
- Department of Surgery, MedStar Georgetown University Hospital, Washington, DC, USA
| | - John W Keyloun
- Department of Surgery, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Shimae Fitzgibbons
- Department of Surgery, MedStar Georgetown University Hospital, Washington, DC, USA.
| |
Collapse
|
7
|
Ait Mahiout L, Bessonov N, Kazmierczak B, Volpert V. Mathematical modeling of respiratory viral infection and applications to SARS-CoV-2 progression. MATHEMATICAL METHODS IN THE APPLIED SCIENCES 2022; 46:MMA8606. [PMID: 36247228 PMCID: PMC9538414 DOI: 10.1002/mma.8606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 06/16/2023]
Abstract
Viral infection in cell culture and tissue is modeled with delay reaction-diffusion equations. It is shown that progression of viral infection can be characterized by the viral replication number, time-dependent viral load, and the speed of infection spreading. These three characteristics are determined through the original model parameters including the rates of cell infection and of virus production in the infected cells. The clinical manifestations of viral infection, depending on tissue damage, correlate with the speed of infection spreading, while the infectivity of a respiratory infection depends on the viral load in the upper respiratory tract. Parameter determination from the experiments on Delta and Omicron variants allows the estimation of the infection spreading speed and viral load. Different variants of the SARS-CoV-2 infection are compared confirming that Omicron is more infectious and has less severe symptoms than Delta variant. Within the same variant, spreading speed (symptoms) correlates with viral load allowing prognosis of disease progression.
Collapse
Affiliation(s)
- Latifa Ait Mahiout
- Laboratoire d'équations aux dérivées partielles non linéaires et histoire des mathématiquesEcole Normale SupérieureAlgiersAlgeria
| | - Nikolai Bessonov
- Institute of Problems of Mechanical EngineeringRussian Academy of SciencesSaint PetersburgRussia
| | - Bogdan Kazmierczak
- Institute of Fundamental Technological ResearchPolish Academy of SciencesWarsawPoland
| | - Vitaly Volpert
- Institut Camille Jordan, UMR 5208 CNRSUniversity Lyon 1VilleurbanneFrance
- Peoples' Friendship University of Russia6 Miklukho‐Maklaya StMoscowRussia
| |
Collapse
|
8
|
Chapuy-Regaud S, Allioux C, Capelli N, Migueres M, Lhomme S, Izopet J. Vectorial Release of Human RNA Viruses from Epithelial Cells. Viruses 2022; 14:231. [PMID: 35215825 PMCID: PMC8875463 DOI: 10.3390/v14020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/07/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Epithelial cells are apico-basolateral polarized cells that line all tubular organs and are often targets for infectious agents. This review focuses on the release of human RNA virus particles from both sides of polarized human cells grown on transwells. Most viruses that infect the mucosa leave their host cells mainly via the apical side while basolateral release is linked to virus propagation within the host. Viruses do this by hijacking the cellular factors involved in polarization and trafficking. Thus, understanding epithelial polarization is essential for a clear understanding of virus pathophysiology.
Collapse
Affiliation(s)
- Sabine Chapuy-Regaud
- Department of Virology, CHU Purpan, F-31059 Toulouse, France; (N.C.); (M.M.); (S.L.); (J.I.)
- INFINITy (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM UMR1291, CNRS UMR5051, Université Toulouse III, CHU Purpan, F-31024 Toulouse, France;
| | - Claire Allioux
- INFINITy (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM UMR1291, CNRS UMR5051, Université Toulouse III, CHU Purpan, F-31024 Toulouse, France;
| | - Nicolas Capelli
- Department of Virology, CHU Purpan, F-31059 Toulouse, France; (N.C.); (M.M.); (S.L.); (J.I.)
- INFINITy (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM UMR1291, CNRS UMR5051, Université Toulouse III, CHU Purpan, F-31024 Toulouse, France;
| | - Marion Migueres
- Department of Virology, CHU Purpan, F-31059 Toulouse, France; (N.C.); (M.M.); (S.L.); (J.I.)
- INFINITy (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM UMR1291, CNRS UMR5051, Université Toulouse III, CHU Purpan, F-31024 Toulouse, France;
| | - Sébastien Lhomme
- Department of Virology, CHU Purpan, F-31059 Toulouse, France; (N.C.); (M.M.); (S.L.); (J.I.)
- INFINITy (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM UMR1291, CNRS UMR5051, Université Toulouse III, CHU Purpan, F-31024 Toulouse, France;
| | - Jacques Izopet
- Department of Virology, CHU Purpan, F-31059 Toulouse, France; (N.C.); (M.M.); (S.L.); (J.I.)
- INFINITy (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM UMR1291, CNRS UMR5051, Université Toulouse III, CHU Purpan, F-31024 Toulouse, France;
| |
Collapse
|
9
|
Pizuorno A, Brim H, Ashktorab H. Gastrointestinal manifestations and SARS-CoV-2 infection. Curr Opin Pharmacol 2021; 61:114-119. [PMID: 34688995 PMCID: PMC8463306 DOI: 10.1016/j.coph.2021.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/18/2021] [Accepted: 09/14/2021] [Indexed: 12/23/2022]
Abstract
Since COVID-19 occurrence in late 2019, intense research efforts on an unprecedented scale have focused on the study of named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry mechanisms and clinical presentations. As for other coronaviruses, SARS-CoV-2 presents with extrarespiratory clinical manifestations such as diarrhea, nausea, vomiting, and abdominal pain which highlight that the gastrointestinal (GI) system as another viral target along with the typical presentations of COVID-19 which is characterized primarily by respiratory symptoms. The digestive system is involved in many systemic functions through the gut-brain axis and systemic immunity modulation. Therefore, the GI system plays an important role in the presentation of the disease, pathogenesis, and possibly treatment outcomes. This minireview summarizes recent work to study SARS-CoV-2 infection as it relates to comorbidities, GI symptoms. This will help to strategize the priorities in understanding the impact of the virus on outcomes in various aspects.
Collapse
Affiliation(s)
- Antonio Pizuorno
- La Universidad del Zulia, Faculty of Medicine, School of Medicine, 4002, Maracaibo, Zulia state, Venezuela
| | - Hassan Brim
- Pathology and Cancer Center, Howard University College of Medicine, Washington, DC, USA
| | - Hassan Ashktorab
- Department of Medicine, Gastroenterology division, and Cancer Center, Howard University College of Medicine, Washington, DC, USA.
| |
Collapse
|
10
|
Jiao Y, Kong N, Wang H, Sun D, Dong S, Chen X, Zheng H, Tong W, Yu H, Yu L, Huang Y, Wang H, Sui B, Zhao L, Liao Y, Zhang W, Tong G, Shan T. PABPC4 Broadly Inhibits Coronavirus Replication by Degrading Nucleocapsid Protein through Selective Autophagy. Microbiol Spectr 2021; 9:e0090821. [PMID: 34612687 PMCID: PMC8510267 DOI: 10.1128/spectrum.00908-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022] Open
Abstract
Emerging coronaviruses (CoVs) can cause severe diseases in humans and animals, and, as of yet, none of the currently available broad-spectrum drugs or vaccines can effectively control these diseases. Host antiviral proteins play an important role in inhibiting viral proliferation. One of the isoforms of cytoplasmic poly(A)-binding protein (PABP), PABPC4, is an RNA-processing protein, which plays an important role in promoting gene expression by enhancing translation and mRNA stability. However, its function in viruses remains poorly understood. Here, we report that the host protein, PABPC4, could be regulated by transcription factor SP1 and broadly inhibits the replication of CoVs, covering four genera (Alphacoronavirus, Betacoronavirus, Gammacoronavirus, and Deltacoronavirus) of the Coronaviridae family by targeting the nucleocapsid (N) protein through the autophagosomes for degradation. PABPC4 recruited the E3 ubiquitin ligase MARCH8/MARCHF8 to the N protein for ubiquitination. Ubiquitinated N protein was recognized by the cargo receptor NDP52/CALCOCO2, which delivered it to the autolysosomes for degradation, resulting in impaired viral proliferation. In addition to regulating gene expression, these data demonstrate a novel antiviral function of PABPC4, which broadly suppresses CoVs by degrading the N protein via the selective autophagy pathway. This study will shed light on the development of broad anticoronaviral therapies. IMPORTANCE Emerging coronaviruses (CoVs) can cause severe diseases in humans and animals, but none of the currently available drugs or vaccines can effectively control these diseases. During viral infection, the host will activate the interferon (IFN) signaling pathways and host restriction factors in maintaining the innate antiviral responses and suppressing viral replication. This study demonstrated that the host protein, PABPC4, interacts with the nucleocapsid (N) proteins from eight CoVs covering four genera (Alphacoronavirus, Betacoronavirus, Gammacoronavirus, and Deltacoronavirus) of the Coronaviridae family. PABPC4 could be regulated by SP1 and broadly inhibits the replication of CoVs by targeting the nucleocapsid (N) protein through the autophagosomes for degradation. This study significantly increases our understanding of the novel host restriction factor PABPC4 against CoV replication and will help develop novel antiviral strategies.
Collapse
Affiliation(s)
- Yajuan Jiao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Ning Kong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Hua Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Dage Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Sujie Dong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Xiaoyong Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Hao Zheng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Wu Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Hai Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Lingxue Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yaowei Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, People’s Republic of China
| | - Huan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Baokun Sui
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Ling Zhao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Wen Zhang
- School of Medicine, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Guangzhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Tongling Shan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
11
|
Barros MT, Veletić M, Kanada M, Pierobon M, Vainio S, Balasingham I, Balasubramaniam S. Molecular Communications in Viral Infections Research: Modeling, Experimental Data, and Future Directions. IEEE TRANSACTIONS ON MOLECULAR, BIOLOGICAL, AND MULTI-SCALE COMMUNICATIONS 2021; 7:121-141. [PMID: 35782714 PMCID: PMC8544950 DOI: 10.1109/tmbmc.2021.3071780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 12/22/2022]
Abstract
Hundreds of millions of people worldwide are affected by viral infections each year, and yet, several of them neither have vaccines nor effective treatment during and post-infection. This challenge has been highlighted by the COVID-19 pandemic, showing how viruses can quickly spread and impact society as a whole. Novel interdisciplinary techniques must emerge to provide forward-looking strategies to combat viral infections, as well as possible future pandemics. In the past decade, an interdisciplinary area involving bioengineering, nanotechnology and information and communication technology (ICT) has been developed, known as Molecular Communications. This new emerging area uses elements of classical communication systems to molecular signalling and communication found inside and outside biological systems, characterizing the signalling processes between cells and viruses. In this paper, we provide an extensive and detailed discussion on how molecular communications can be integrated into the viral infectious diseases research, and how possible treatment and vaccines can be developed considering molecules as information carriers. We provide a literature review on molecular communications models for viral infection (intra-body and extra-body), a deep analysis on their effects on immune response, how experimental can be used by the molecular communications community, as well as open issues and future directions.
Collapse
Affiliation(s)
- Michael Taynnan Barros
- CBIG/BioMediTechTampere University33014TampereFinland
- School of Computer Science and Electronic EngineeringUniversity of EssexColchesterCO4 3SQU.K.
| | - Mladen Veletić
- Intervention CentreOslo University Hospital0424OsloNorway
- Department of Electronic SystemsNorwegian University of Science and Technology7491TrondheimNorway
| | - Masamitsu Kanada
- Department of Pharmacology and ToxicologyInstitute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingMI48824USA
| | - Massimiliano Pierobon
- Department of Computer Science and EngineeringUniversity of Nebraska–LincolnLincolnNE68588USA
| | - Seppo Vainio
- InfoTech OuluKvantum Institute, Faculty of Biochemistry and Molecular Medicine, Laboratory of Developmental Biology, Oulu University90570OuluFinland
| | - Ilangko Balasingham
- Intervention CentreOslo University Hospital0424OsloNorway
- Department of Electronic SystemsNorwegian University of Science and Technology7491TrondheimNorway
| | | |
Collapse
|
12
|
Kesheh MM, Hosseini P, Soltani S, Zandi M. An overview on the seven pathogenic human coronaviruses. Rev Med Virol 2021; 32:e2282. [PMID: 34339073 DOI: 10.1002/rmv.2282] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023]
Abstract
To date, seven human coronaviruses (HCoVs) have been detected: HCoV-NL63, HCoV-229E, HCoV-HKU1, HCoV-OC43, severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV) and SARS-CoV-2. Four of these viruses, including HCoV-NL63, -229E, -HKU1 and -OC43, usually cause mild-to-moderate respiratory diseases with a seasonal pattern. Since 2000, three new HCoVs have emerged with a significant mortality rate. Although SARS-CoV and MERS-CoV caused an epidemic in some countries, SARS-CoV-2 escalated into a pandemic. All HCoVs can cause severe complications in the elderly and immunocompromised individuals. The bat origin of HCoVs, the presence of intermediate hosts and the nature of their viral replication suggest that other new coronaviruses may emerge in the future. Despite the fact that all HCoVs share similarities in viral replication, they differ in their accessory proteins, incubation period and pathogenicity. This study aims to review these differences between the seven HCoVs.
Collapse
Affiliation(s)
- Mina Mobini Kesheh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parastoo Hosseini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Saber Soltani
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Mishra D, Maurya RR, Kumar K, Munjal NS, Bahadur V, Sharma S, Singh P, Bahadur I. Structurally modified compounds of hydroxychloroquine, remdesivir and tetrahydrocannabinol against main protease of SARS-CoV-2, a possible hope for COVID-19: Docking and molecular dynamics simulation studies. J Mol Liq 2021; 335:116185. [PMID: 33879934 PMCID: PMC8051003 DOI: 10.1016/j.molliq.2021.116185] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/03/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023]
Abstract
Now a days, more than 200 countries faces the health crisis due to epidemiological disease COVID-19 caused by SARS-CoV-2 virus. It will cause a very high impact on world's economy and global health sector. Earlier the structure of main protease (Mpro) protein was deposited in the RCSB protein repository. Hydroxychloroquine (HCQ) and remdesivir were found to effective in treatment of COVID-19 patients. Here we have performed docking and molecule dynamic (MD) simulation study of HCQ and remdesivir with Mpro protein which gave promising results to inhibit Mpro protein in SARS-CoV-2. On the basis of results obtained we designed structurally modified 18 novel derivatives of HCQ, remdesivir and tetrahydrocannabinol (THC) and carried out docking studies of all the derivatives. From the docking studies six molecules DK4, DK7, DK10, DK16, DK17 and DK19 gave promising results and can be use as inhibitor for Mpro of SARS-CoV-2 to control COVID-19 very effectively. Further, molecular dynamics simulation of one derivative of HCQ and one derivative of tetrahydrocannabinol showing excellent docking score was performed along with the respective parent molecules. The two derivatives gave excellent docking score and higher stability than the parent molecule as validated with molecular dynamics (MD) simulation for the binding affinities towards Mpro of SARS-CoV-2 thus represented as strong inhibitors at very low concentration.
Collapse
Affiliation(s)
- Deepak Mishra
- Department of Chemistry, SRM University, Delhi-NCR Sonepa t, Haryana 131029, India
| | - Radha Raman Maurya
- Department of Chemistry, Ramjas College, University of Delhi, University Enclave, Delhi 110007, India
| | - Kamlesh Kumar
- Department of Chemistry, Kumaun University, Nainital 263001, UK, India,Corresponding authors
| | - Nupur S. Munjal
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | - Vijay Bahadur
- Department of Chemistry, SRM University, Delhi-NCR Sonepa t, Haryana 131029, India
| | - Sandeep Sharma
- Department of Chemistry, SRM University, Delhi-NCR Sonepa t, Haryana 131029, India
| | - Prashant Singh
- Department of Chemistry, Atma Ram Sanatan Dharma (ARSD) College, Delhi University, New Delhi 110021, India
| | - Indra Bahadur
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, North-West University, South Africa,Corresponding authors
| |
Collapse
|
14
|
Mondeja B, Valdes O, Resik S, Vizcaino A, Acosta E, Montalván A, Paez A, Mune M, Rodríguez R, Valdés J, Gonzalez G, Sanchez D, Falcón V, González Y, Kourí V, Díaz A, Guzmán M. SARS-CoV-2: preliminary study of infected human nasopharyngeal tissue by high resolution microscopy. Virol J 2021; 18:149. [PMID: 34275492 PMCID: PMC8286443 DOI: 10.1186/s12985-021-01620-1] [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: 06/17/2020] [Accepted: 07/08/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The novel coronavirus SARS-CoV-2 is the etiological agent of COVID-19. This virus has become one of the most dangerous in recent times with a very high rate of transmission. At present, several publications show the typical crown-shape of the novel coronavirus grown in cell cultures. However, an integral ultramicroscopy study done directly from clinical specimens has not been published. METHODS Nasopharyngeal swabs were collected from 12 Cuban individuals, six asymptomatic and RT-PCR negative (negative control) and six others from a COVID-19 symptomatic and RT-PCR positive for SARS CoV-2. Samples were treated with an aldehyde solution and processed by scanning electron microscopy (SEM), confocal microscopy (CM) and, atomic force microscopy. Improvement and segmentation of coronavirus images were performed by a novel mathematical image enhancement algorithm. RESULTS The images of the negative control sample showed the characteristic healthy microvilli morphology at the apical region of the nasal epithelial cells. As expected, they do not display virus-like structures. The images of the positive sample showed characteristic coronavirus-like particles and evident destruction of microvilli. In some regions, virions budding through the cell membrane were observed. Microvilli destruction could explain the anosmia reported by some patients. Virus-particles emerging from the cell-surface with a variable size ranging from 80 to 400 nm were observed by SEM. Viral antigen was identified in the apical cells zone by CM. CONCLUSIONS The integral microscopy study showed that SARS-CoV-2 has a similar image to SARS-CoV. The application of several high-resolution microscopy techniques to nasopharyngeal samples awaits future use.
Collapse
Affiliation(s)
- Brian Mondeja
- Center for Advanced Studies of Cuba (CEA), La Habana, Cuba.
| | - Odalys Valdes
- Institute of Tropical Medicine "Pedro Kourí" (IPK), La Habana, Cuba
| | - Sonia Resik
- Institute of Tropical Medicine "Pedro Kourí" (IPK), La Habana, Cuba
| | | | - Emilio Acosta
- Center for Advanced Studies of Cuba (CEA), La Habana, Cuba
| | | | - Amira Paez
- Center for Advanced Studies of Cuba (CEA), La Habana, Cuba
| | - Mayra Mune
- Institute of Tropical Medicine "Pedro Kourí" (IPK), La Habana, Cuba
| | - Roberto Rodríguez
- Institute of Cybernetics, Mathematics, and Physics of Cuba (ICIMAF), La Habana, Cuba
| | - Juan Valdés
- Center for Advanced Studies of Cuba (CEA), La Habana, Cuba
| | - Guelsys Gonzalez
- Institute of Tropical Medicine "Pedro Kourí" (IPK), La Habana, Cuba
| | - Daisy Sanchez
- Center for Advanced Studies of Cuba (CEA), La Habana, Cuba
| | - Viviana Falcón
- Center of Genetic Engineer and Biotechnology of Cuba (CIGB), La Habana, Cuba
| | | | - Vivian Kourí
- Institute of Tropical Medicine "Pedro Kourí" (IPK), La Habana, Cuba
| | | | - Angelina Díaz
- Center for Advanced Studies of Cuba (CEA), La Habana, Cuba
| | - María Guzmán
- Institute of Tropical Medicine "Pedro Kourí" (IPK), La Habana, Cuba
| |
Collapse
|
15
|
Dobson L, Zeke A, Tusnády GE. PolarProtPred: Predicting apical and basolateral localization of transmembrane proteins using putative short linear motifs and deep learning. Bioinformatics 2021; 37:4328-4335. [PMID: 34185052 PMCID: PMC8384406 DOI: 10.1093/bioinformatics/btab480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/03/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Motivation Cell polarity refers to the asymmetric organization of cellular components in various cells. Epithelial cells are the best-known examples of polarized cells, featuring apical and basolateral membrane domains. Mounting evidence suggests that short linear motifs play a major role in protein trafficking to these domains, although the exact rules governing them are still elusive. Results In this study we prepared neural networks that capture recurrent patterns to classify transmembrane proteins localizing into apical and basolateral membranes. Asymmetric expression of drug transporters results in vectorial drug transport, governing the pharmacokinetics of numerous substances, yet the data on how proteins are sorted in epithelial cells is very scattered. The provided method may offer help to experimentalists to identify or better characterize molecular networks regulating the distribution of transporters or surface receptors (including viral entry receptors like that of COVID-19). Availability The prediction server PolarProtPred is available at http://polarprotpred.ttk.hu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Laszlo Dobson
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary.,Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, Heidelberg, 69117, Germany
| | - András Zeke
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
| | - Gábor E Tusnády
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, Budapest, 1117, Hungary
| |
Collapse
|
16
|
Rehman HA, Ramzan F, Basharat Z, Shakeel M, Khan MUG, Khan IA. Comprehensive comparative genomic and microsatellite analysis of SARS, MERS, BAT-SARS, and COVID-19 coronaviruses. J Med Virol 2021; 93:4382-4391. [PMID: 33782990 PMCID: PMC8250465 DOI: 10.1002/jmv.26974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/02/2021] [Accepted: 03/24/2021] [Indexed: 01/11/2023]
Abstract
The coronavirus disease 2019 (COVID‐19) pandemic has spread around the globe very rapidly. Previously, the evolution pattern and similarity among the COVID‐19 causative organism severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) and causative organisms of other similar infections have been determined using a single type of genetic marker in different studies. Herein, the SARS‐CoV‐2 and related β coronaviruses Middle East respiratory syndrome coronavirus (MERS‐CoV), SARS‐CoV, bat coronavirus (BAT‐CoV) were comprehensively analyzed using a custom‐built pipeline that employed phylogenetic approaches based on multiple types of genetic markers including the whole genome sequences, mutations in nucleotide sequences, mutations in protein sequences, and microsatellites. The whole‐genome sequence‐based phylogeny revealed that the strains of SARS‐CoV‐2 are more similar to the BAT‐CoV strains. The mutational analysis showed that on average MERS‐CoV and BAT‐CoV genomes differed at 134.21 and 136.72 sites, respectively, whereas the SARS‐CoV genome differed at 26.64 sites from the reference genome of SARS‐CoV‐2. Furthermore, the microsatellite analysis highlighted a relatively higher number of average microsatellites for MERS‐CoV and SARS‐CoV‐2 (106.8 and 107, respectively), and a lower number for SARS‐CoV and BAT‐CoV (95.8 and 98.5, respectively). Collectively, the analysis of multiple genetic markers of selected β viral genomes revealed that the newly born SARS‐COV‐2 is closely related to BAT‐CoV, whereas, MERS‐CoV is more distinct from the SARS‐CoV‐2 than BAT‐CoV and SARS‐CoV.
Collapse
Affiliation(s)
- Hafiz Abdul Rehman
- Intelligent Criminology Research Lab (ICRL), National Center of Artificial Intelligence (NCAI), Al-Khawarizmi Institute of Computer Science (KICS), University of Engineering and Technology (UET), Lahore, Pakistan
| | - Farheen Ramzan
- Department of Computer Science, University of Engineering and Technology (UET), Lahore, Pakistan
| | - Zarrin Basharat
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi, Pakistan
| | - Muhammad Shakeel
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi, Pakistan
| | - Muhammad Usman Ghani Khan
- Intelligent Criminology Research Lab (ICRL), National Center of Artificial Intelligence (NCAI), Al-Khawarizmi Institute of Computer Science (KICS), University of Engineering and Technology (UET), Lahore, Pakistan.,Department of Computer Science, University of Engineering and Technology (UET), Lahore, Pakistan
| | - Ishtiaq Ahmad Khan
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi, Pakistan
| |
Collapse
|
17
|
Hernandez-Gonzalez M, Larocque G, Way M. Viral use and subversion of membrane organization and trafficking. J Cell Sci 2021; 134:jcs252676. [PMID: 33664154 PMCID: PMC7610647 DOI: 10.1242/jcs.252676] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Membrane trafficking is an essential cellular process conserved across all eukaryotes, which regulates the uptake or release of macromolecules from cells, the composition of cellular membranes and organelle biogenesis. It influences numerous aspects of cellular organisation, dynamics and homeostasis, including nutrition, signalling and cell architecture. Not surprisingly, malfunction of membrane trafficking is linked to many serious genetic, metabolic and neurological disorders. It is also often hijacked during viral infection, enabling viruses to accomplish many of the main stages of their replication cycle, including entry into and egress from cells. The appropriation of membrane trafficking by viruses has been studied since the birth of cell biology and has helped elucidate how this integral cellular process functions. In this Review, we discuss some of the different strategies viruses use to manipulate and take over the membrane compartments of their hosts to promote their replication, assembly and egress.
Collapse
Affiliation(s)
- Miguel Hernandez-Gonzalez
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Gabrielle Larocque
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Infectious Disease, Imperial College, London W2 1PG, UK
| |
Collapse
|
18
|
Assembly and Cellular Exit of Coronaviruses: Hijacking an Unconventional Secretory Pathway from the Pre-Golgi Intermediate Compartment via the Golgi Ribbon to the Extracellular Space. Cells 2021; 10:cells10030503. [PMID: 33652973 PMCID: PMC7996754 DOI: 10.3390/cells10030503] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
Coronaviruses (CoVs) assemble by budding into the lumen of the intermediate compartment (IC) at the endoplasmic reticulum (ER)-Golgi interface. However, why CoVs have chosen the IC as their intracellular site of assembly and how progeny viruses are delivered from this compartment to the extracellular space has remained unclear. Here we address these enigmatic late events of the CoV life cycle in light of recently described properties of the IC. Of particular interest are the emerging spatial and functional connections between IC elements and recycling endosomes (REs), defined by the GTPases Rab1 and Rab11, respectively. The establishment of IC-RE links at the cell periphery, around the centrosome and evidently also at the noncompact zones of the Golgi ribbon indicates that—besides traditional ER-Golgi communication—the IC also promotes a secretory process that bypasses the Golgi stacks, but involves its direct connection with the endocytic recycling system. The initial confinement of CoVs to the lumen of IC-derived large transport carriers and their preferential absence from Golgi stacks is consistent with the idea that they exit cells following such an unconventional route. In fact, CoVs may share this pathway with other intracellularly budding viruses, lipoproteins, procollagen, and/or protein aggregates experimentally introduced into the IC lumen.
Collapse
|
19
|
Zhang Y, Greer RA, Song Y, Praveen H, Song Y. In silico identification of available drugs targeting cell surface BiP to disrupt SARS-CoV-2 binding and replication: Drug repurposing approach. Eur J Pharm Sci 2021; 160:105771. [PMID: 33617948 PMCID: PMC7894100 DOI: 10.1016/j.ejps.2021.105771] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/03/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022]
Abstract
Aims Cell surface binding immunoglobin protein (csBiP) is predicted to be susceptible to SARS-CoV-2 binding. With a substrate-binding domain (SBD) that binds to polypeptides and a nucleotide-binding domain (NBD) that can initiate extrinsic caspase-dependent apoptosis, csBiP may be a promising therapeutic target for COVID-19. This study aims to identify FDA-approved drugs that can neutralize viral binding and prevent viral replication by targeting the functional domains of csBiP. Methods In silico screening of 1999 FDA-approved drugs against the functional domains of BiP were performed using three molecular docking programs to avoid bias from individual docking programs. Top ligands were selected by averaging the ligand rankings from three programs. Interactions between top ligands and functional domains of BiP were analyzed. Key findings The top 10 SBD-binding candidates are velpatasvir, irinotecan, netupitant, lapatinib, doramectin, conivaptan, fenoverine, duvelisib, irbesartan, and pazopanib. The top 10 NBD-binding candidates are nilotinib, eltrombopag, grapiprant, topotecan, acetohexamide, vemurafenib, paritaprevir, pixantrone, azosemide, and piperaquine-phosphate. Among them, Velpatasvir and paritaprevir are antiviral agents that target the protease of hepatitis C virus. Netupitant is an anti-inflammatory drug that inhibits neurokinin-1 receptor, which contributes to acute inflammation. Grapiprant is an anti-inflammatory drug that inhibits the prostaglandin E2 receptor protein subtype 4, which is expressed on immune cells and triggers inflammation. These predicted SBD-binding drugs could disrupt SARS-CoV-2 binding to csBiP, and NBD-binding drugs may falter viral attachment and replication by locking the SBD in closed conformation and triggering apoptosis in infected cells. Significance csBiP appears to be a novel therapeutic target against COVID-19 by preventing viral attachment and replication. These identified drugs could be repurposed to treat COVID-19 patients.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, United States
| | - Rory A Greer
- Department of Biomedical Engineering, The University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, United States
| | - Yuwei Song
- Department of Dermatology, The University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, United States
| | - Hrithik Praveen
- Department of Biomedical Engineering, The University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, United States
| | - Yuhua Song
- Department of Biomedical Engineering, The University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, United States.
| |
Collapse
|
20
|
Sorice M, Misasi R, Riitano G, Manganelli V, Martellucci S, Longo A, Garofalo T, Mattei V. Targeting Lipid Rafts as a Strategy Against Coronavirus. Front Cell Dev Biol 2021; 8:618296. [PMID: 33614627 PMCID: PMC7890255 DOI: 10.3389/fcell.2020.618296] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Lipid rafts are functional membrane microdomains containing sphingolipids, including gangliosides, and cholesterol. These regions are characterized by highly ordered and tightly packed lipid molecules. Several studies revealed that lipid rafts are involved in life cycle of different viruses, including coronaviruses. Among these recently emerged the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The main receptor for SARS-CoV-2 is represented by the angiotensin-converting enzyme-2 (ACE-2), although it also binds to sialic acids linked to host cell surface gangliosides. A new type of ganglioside-binding domain within the N-terminal portion of the SARS-CoV-2 spike protein was identified. Lipid rafts provide a suitable platform able to concentrate ACE-2 receptor on host cell membranes where they may interact with the spike protein on viral envelope. This review is focused on selective targeting lipid rafts components as a strategy against coronavirus. Indeed, cholesterol-binding agents, including statins or methyl-β-cyclodextrin (MβCD), can affect cholesterol, causing disruption of lipid rafts, consequently impairing coronavirus adhesion and binding. Moreover, these compounds can block downstream key molecules in virus infectivity, reducing the levels of proinflammatory molecules [tumor necrosis factor alpha (TNF-α), interleukin (IL)-6], and/or affecting the autophagic process involved in both viral replication and clearance. Furthermore, cyclodextrins can assemble into complexes with various drugs to form host-guest inclusions and may be used as pharmaceutical excipients of antiviral compounds, such as lopinavir and remdesivir, by improving bioavailability and solubility. In conclusion, the role of lipid rafts-affecting drugs in the process of coronavirus entry into the host cells prompts to introduce a new potential task in the pharmacological approach against coronavirus.
Collapse
Affiliation(s)
- Maurizio Sorice
- Department of Experimental Medicine, “Sapienza” University, Rome, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, “Sapienza” University, Rome, Italy
| | - Gloria Riitano
- Department of Experimental Medicine, “Sapienza” University, Rome, Italy
| | | | - Stefano Martellucci
- Biomedicine and Advanced Technologies Rieti Center, “Sabina Universitas”, Rieti, Italy
| | - Agostina Longo
- Department of Experimental Medicine, “Sapienza” University, Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, “Sapienza” University, Rome, Italy
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, “Sabina Universitas”, Rieti, Italy
| |
Collapse
|
21
|
Bhandari R, Khanna G, Kuhad A. Pharmacological insight into potential therapeutic agents for the deadly Covid-19 pandemic. Eur J Pharmacol 2021; 890:173643. [PMID: 33065092 PMCID: PMC7550915 DOI: 10.1016/j.ejphar.2020.173643] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/03/2020] [Accepted: 10/08/2020] [Indexed: 12/15/2022]
Abstract
Coronaviruses are pleomorphic, enveloped, or spherical viruses, which have a size ranging from 80 to 120 nm. These viruses act on receptors that cause the triggering of fusion. Coronaviruses were first described after cultivation from patients with common colds by Tyrell and Bynoe in 1966. There are various subtypes of coronavirus, 7 out of these can cause infection in human beings. The Alpha subtype is responsible for mild infection showing symptoms or infection without any prevailing symptoms. On the other hand, the beta subtype is responsible for very serious diseases leading to fatality. The lineage of this novel SARS-CoV-2 falls under the beta lineage of the beta coronavirus which has been observed to have a relation to the MERS and SARS coronavirus. In the Huanan market selling seafood, the transition of this novel virus in humans from animals has occurred. It has the potential to be the cause of widespread fatality amongst the people of the globe. On August 16, 2020, the World Health Organisation had reported 2,1294,845 cases which are confirmed to date out of which 413,372 deaths have occurred. Currently, no targeted antiviral vaccines or drugs to fight against COVID-19 infection have been approved for use in humans. This pandemic is fast emerging and drug repurposing is the only ray of hope which can ensure quick availability. Vaccine development is progressing each day with various platforms such as DNA, Live Attenuated Virus, Non-Replicating Viral Vector, Protein Subunit, and RNA, being utilized for the development. COVID-19 attacks the immune system of the host & this can result in a cytokine storm. As a result, various herbal agents both acting as antivirals and immunomodulatory can also be used. Convalescent Plasma Therapy and Mesenchymal Stem Cell therapy are also being explored as a plausible therapeutic. There remains a considerable unmet need for therapeutics to be addressed. The development and availability of accessible and efficient therapy are essential for the treatment of patients. This review discusses the epidemiology, pathogenesis, the tale of origin, and transmission of COVID-19 or Sars-Cov2 virus and gives evidence of potential therapeutic agents that can be explored to cast away this pandemic.
Collapse
Affiliation(s)
- Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Garima Khanna
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, 160 014, India.
| |
Collapse
|
22
|
Stolfi P, Manni L, Soligo M, Vergni D, Tieri P. Designing a Network Proximity-Based Drug Repurposing Strategy for COVID-19. Front Cell Dev Biol 2020; 8:545089. [PMID: 33123533 PMCID: PMC7573309 DOI: 10.3389/fcell.2020.545089] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
The ongoing COVID-19 pandemic still requires fast and effective efforts from all fronts, including epidemiology, clinical practice, molecular medicine, and pharmacology. A comprehensive molecular framework of the disease is needed to better understand its pathological mechanisms, and to design successful treatments able to slow down and stop the impressive pace of the outbreak and harsh clinical symptomatology, possibly via the use of readily available, off-the-shelf drugs. This work engages in providing a wider picture of the human molecular landscape of the SARS-CoV-2 infection via a network medicine approach as the ground for a drug repurposing strategy. Grounding on prior knowledge such as experimentally validated host proteins known to be viral interactors, tissue-specific gene expression data, and using network analysis techniques such as network propagation and connectivity significance, the host molecular reaction network to the viral invasion is explored and exploited to infer and prioritize candidate target genes, and finally to propose drugs to be repurposed for the treatment of COVID-19. Ranks of potential target genes have been obtained for coherent groups of tissues/organs, potential and distinct sites of interaction between the virus and the organism. The normalization and the aggregation of the different scores allowed to define a preliminary, restricted list of genes candidates as pharmacological targets for drug repurposing, with the aim of contrasting different phases of the virus infection and viral replication cycle.
Collapse
Affiliation(s)
- Paola Stolfi
- National Research Council (CNR), Institute for Applied Computing (IAC), Rome, Italy
| | - Luigi Manni
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Marzia Soligo
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Davide Vergni
- National Research Council (CNR), Institute for Applied Computing (IAC), Rome, Italy
| | - Paolo Tieri
- National Research Council (CNR), Institute for Applied Computing (IAC), Rome, Italy
| |
Collapse
|
23
|
Song ZG, Chen YM, Wu F, Xu L, Wang BF, Shi L, Chen X, Dai FH, She JL, Chen JM, Holmes EC, Zhu TY, Zhang YZ. Identifying the Risk of SARS-CoV-2 Infection and Environmental Monitoring in Airborne Infectious Isolation Rooms (AIIRs). Virol Sin 2020; 35:785-792. [PMID: 32986229 PMCID: PMC7521197 DOI: 10.1007/s12250-020-00301-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/31/2020] [Indexed: 01/22/2023] Open
Abstract
Healthcare workers (HCWs) are at high risk of occupational exposure to the new pandemic human coronavirus, SARS-CoV-2, and are a source of nosocomial transmission in airborne infectious isolation rooms (AIIRs). Here, we performed comprehensive environmental contamination surveillance to evaluate the risk of viral transmission in AIIRs with 115 rooms in three buildings at the Shanghai Public Health Clinical Center, Shanghai, during the treatment of 334 patients infected with SARS-CoV-2. The results showed that the risk of airborne transmission of SARS-CoV-2 in AIIRs was low (1.62%, 25/1544) due to the directional airflow and strong environmental hygiene procedures. However, we detected viral RNA on the surface of foot-operated openers and bathroom sinks in AIIRs (viral load: 55.00–3154.50 copies/mL). This might be a source of contamination to connecting corridors and object surfaces through the footwear and gloves used by HCWs. The risk of infection was eliminated by the use of disposable footwear covers and the application of more effective environmental and personal hygiene measures. With the help of effective infection control procedures, none of 290 HCWs was infected when working in the AIIRs at this hospital. This study has provided information pertinent for infection control in AIIRs during the treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Zhi-Gang Song
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
| | - Yan-Mei Chen
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
| | - Fan Wu
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
| | - Lin Xu
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
| | - Bang-Fang Wang
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
| | - Lei Shi
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
| | - Xiao Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Fa-Hui Dai
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
| | - Jia-Lei She
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
| | - Jian-Min Chen
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200438, China
| | - Edward C Holmes
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and School of Medical Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Tong-Yu Zhu
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China.
| | - Yong-Zhen Zhang
- Shanghai Public Health Clinical Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
24
|
De Maio F, Lo Cascio E, Babini G, Sali M, Della Longa S, Tilocca B, Roncada P, Arcovito A, Sanguinetti M, Scambia G, Urbani A. Improved binding of SARS-CoV-2 Envelope protein to tight junction-associated PALS1 could play a key role in COVID-19 pathogenesis. Microbes Infect 2020; 22:592-597. [PMID: 32891874 PMCID: PMC7473260 DOI: 10.1016/j.micinf.2020.08.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
The Envelope (E) protein of SARS-CoV-2 is the most enigmatic protein among the four structural ones. Most of its current knowledge is based on the direct comparison to the SARS E protein, initially mistakenly undervalued and subsequently proved to be a key factor in the ER-Golgi localization and in tight junction disruption. We compared the genomic sequences of E protein of SARS-CoV-2, SARS-CoV and the closely related genomes of bats and pangolins obtained from the GISAID and GenBank databases. When compared to the known SARS E protein, we observed a significant difference in amino acid sequence in the C-terminal end of SARS-CoV-2 E protein. Subsequently, in silico modelling analyses of E proteins conformation and docking provide evidences of a strengthened binding of SARS-CoV-2 E protein with the tight junction-associated PALS1 protein. Based on our computational evidences and on data related to SARS-CoV, we believe that SARS-CoV-2 E protein interferes more stably with PALS1 leading to an enhanced epithelial barrier disruption, amplifying the inflammatory processes, and promoting tissue remodelling. These findings raise a warning on the underestimated role of the E protein in the pathogenic mechanism and open the route to detailed experimental investigations.
Collapse
Affiliation(s)
- Flavio De Maio
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Ettore Lo Cascio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Gabriele Babini
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy.
| | - Michela Sali
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Stefano Della Longa
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Bruno Tilocca
- Department of Health Science, University "Magna Græcia" of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Paola Roncada
- Department of Health Science, University "Magna Græcia" of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy; Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy
| | - Maurizio Sanguinetti
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Giovanni Scambia
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze della Vita e Sanità Pubblica - Sezione di Ginecologia ed Ostetricia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Andrea Urbani
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| |
Collapse
|
25
|
Bhowmik D, Nandi R, Jagadeesan R, Kumar N, Prakash A, Kumar D. Identification of potential inhibitors against SARS-CoV-2 by targeting proteins responsible for envelope formation and virion assembly using docking based virtual screening, and pharmacokinetics approaches. INFECTION GENETICS AND EVOLUTION 2020; 84:104451. [PMID: 32640381 PMCID: PMC7335633 DOI: 10.1016/j.meegid.2020.104451] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
WHO has declared the outbreak of COVID-19 as a public health emergency of international concern. The ever-growing new cases have called for an urgent emergency for specific anti-COVID-19 drugs. Three structural proteins (Membrane, Envelope and Nucleocapsid protein) play an essential role in the assembly and formation of the infectious virion particles. Thus, the present study was designed to identify potential drug candidates from the unique collection of 548 anti-viral compounds (natural and synthetic anti-viral), which target SARS-CoV-2 structural proteins. High-end molecular docking analysis was performed to characterize the binding affinity of the selected drugs-the ligand, with the SARS-CoV-2 structural proteins, while high-level Simulation studies analyzed the stability of drug-protein interactions. The present study identified rutin, a bioflavonoid and the antibiotic, doxycycline, as the most potent inhibitor of SARS-CoV-2 envelope protein. Caffeic acid and ferulic acid were found to inhibit SARS-CoV-2 membrane protein while the anti-viral agent's simeprevir and grazoprevir showed a high binding affinity for nucleocapsid protein. All these compounds not only showed excellent pharmacokinetic properties, absorption, metabolism, minimal toxicity and bioavailability but were also remain stabilized at the active site of proteins during the MD simulation. Thus, the identified lead compounds may act as potential molecules for the development of effective drugs against SARS-CoV-2 by inhibiting the envelope formation, virion assembly and viral pathogenesis.
Collapse
Affiliation(s)
- Deep Bhowmik
- Department of Microbiology, Assam University, Silchar 788011, Assam, India
| | - Rajat Nandi
- Department of Microbiology, Assam University, Silchar 788011, Assam, India
| | - Rahul Jagadeesan
- CAS in Crystallography and Biophysics, Guindy Campus, University of Madras, Chennai 600025, India
| | - Niranjan Kumar
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon 122413, India
| | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar 788011, Assam, India.
| |
Collapse
|
26
|
Chen L, Hu C, Hood M, Zhang X, Zhang L, Kan J, Du J. A Novel Combination of Vitamin C, Curcumin and Glycyrrhizic Acid Potentially Regulates Immune and Inflammatory Response Associated with Coronavirus Infections: A Perspective from System Biology Analysis. Nutrients 2020; 12:E1193. [PMID: 32344708 PMCID: PMC7230237 DOI: 10.3390/nu12041193] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/21/2022] Open
Abstract
Novel coronaviruses (CoV) have emerged periodically around the world in recent years. The recurrent spreading of CoVs imposes an ongoing threat to global health and the economy. Since no specific therapy for these CoVs is available, any beneficial approach (including nutritional and dietary approach) is worth investigation. Based on recent advances in nutrients and phytonutrients research, a novel combination of vitamin C, curcumin and glycyrrhizic acid (VCG Plus) was developed that has potential against CoV infection. System biology tools were applied to explore the potential of VCG Plus in modulating targets and pathways relevant to immune and inflammation responses. Gene target acquisition, gene ontology and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment were conducted consecutively along with network analysis. The results show that VCG Plus can act on 88 hub targets which are closely connected and associated with immune and inflammatory responses. Specifically, VCG Plus has the potential to regulate innate immune response by acting on NOD-like and Toll-like signaling pathways to promote interferons production, activate and balance T-cells, and regulate the inflammatory response by inhibiting PI3K/AKT, NF-κB and MAPK signaling pathways. All these biological processes and pathways have been well documented in CoV infections studies. Therefore, our findings suggest that VCG Plus may be helpful in regulating immune response to combat CoV infections and inhibit excessive inflammatory responses to prevent the onset of cytokine storm. However, further in vitro and in vivo experiments are warranted to validate the current findings with system biology tools. Our current approach provides a new strategy in predicting formulation rationale when developing new dietary supplements.
Collapse
Affiliation(s)
- Liang Chen
- Nutrilite Health Institute, 720 Cailun Road, Shanghai 201203, China; (L.C.); (X.Z.); (L.Z.); (J.K.)
| | - Chun Hu
- Nutrilite Health Institute, 5600 Beach Boulevard, Buena Park, CA 90621, USA;
| | - Molly Hood
- Nutrilite Health Institute, 7575 East Fulton Avenue, Ada, MI 49355, USA;
| | - Xue Zhang
- Nutrilite Health Institute, 720 Cailun Road, Shanghai 201203, China; (L.C.); (X.Z.); (L.Z.); (J.K.)
| | - Lu Zhang
- Nutrilite Health Institute, 720 Cailun Road, Shanghai 201203, China; (L.C.); (X.Z.); (L.Z.); (J.K.)
| | - Juntao Kan
- Nutrilite Health Institute, 720 Cailun Road, Shanghai 201203, China; (L.C.); (X.Z.); (L.Z.); (J.K.)
| | - Jun Du
- Nutrilite Health Institute, 720 Cailun Road, Shanghai 201203, China; (L.C.); (X.Z.); (L.Z.); (J.K.)
| |
Collapse
|
27
|
Kim JM, Chung YS, Jo HJ, Lee NJ, Kim MS, Woo SH, Park S, Kim JW, Kim HM, Han MG. Identification of Coronavirus Isolated from a Patient in Korea with COVID-19. Osong Public Health Res Perspect 2020; 11:3-7. [PMID: 32149036 PMCID: PMC7045880 DOI: 10.24171/j.phrp.2020.11.1.02] [Citation(s) in RCA: 295] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objectives Following reports of patients with unexplained pneumonia at the end of December 2019 in Wuhan, China, the causative agent was identified as coronavirus (SARS-CoV-2), and the 2019 novel coronavirus disease was named COVID-19 by the World Health Organization. Putative patients with COVID-19 have been identified in South Korea, and attempts have been made to isolate the pathogen from these patients. Methods Upper and lower respiratory tract secretion samples from putative patients with COVID-19 were inoculated onto cells to isolate the virus. Full genome sequencing and electron microscopy were used to identify the virus. Results The virus replicated in Vero cells and cytopathic effects were observed. Full genome sequencing showed that the virus genome exhibited sequence homology of more than 99.9% with SARS-CoV-2 which was isolated from patients from other countries, for instance China. Sequence homology of SARS-CoV-2 with SARS-CoV, and MERS-CoV was 77.5% and 50%, respectively. Coronavirus-specific morphology was observed by electron microscopy in virus-infected Vero cells. Conclusion SARS-CoV-2 was isolated from putative patients with unexplained pneumonia and intermittent coughing and fever. The isolated virus was named BetaCoV/Korea/KCDC03/2020.
Collapse
Affiliation(s)
- Jeong-Min Kim
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Yoon-Seok Chung
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Hye Jun Jo
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Nam-Joo Lee
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Mi Seon Kim
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Sang Hee Woo
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Sehee Park
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Jee Woong Kim
- Division of Biosafety Evaluation and Control, National Institute of Health, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Heui Man Kim
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| | - Myung-Guk Han
- Division of Viral Diseases, Center for Laboratory Control of Infectious Diseases, Korea Centers for Disease Control and Prevention, Cheongju, Korea
| |
Collapse
|
28
|
Koonpaew S, Teeravechyan S, Frantz PN, Chailangkarn T, Jongkaewwattana A. PEDV and PDCoV Pathogenesis: The Interplay Between Host Innate Immune Responses and Porcine Enteric Coronaviruses. Front Vet Sci 2019; 6:34. [PMID: 30854373 PMCID: PMC6395401 DOI: 10.3389/fvets.2019.00034] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 01/28/2019] [Indexed: 12/24/2022] Open
Abstract
Enteropathogenic porcine epidemic diarrhea virus (PEDV) and porcine deltacoronavirus (PDCoV), members of the coronavirus family, account for the majority of lethal watery diarrhea in neonatal pigs in the past decade. These two viruses pose significant economic and public health burdens, even as both continue to emerge and reemerge worldwide. The ability to evade, circumvent or subvert the host’s first line of defense, namely the innate immune system, is the key determinant for pathogen virulence, survival, and the establishment of successful infection. Unfortunately, we have only started to unravel the underlying viral mechanisms used to manipulate host innate immune responses. In this review, we gather current knowledge concerning the interplay between these viruses and components of host innate immunity, focusing on type I interferon induction and signaling in particular, and the mechanisms by which virus-encoded gene products antagonize and subvert host innate immune responses. Finally, we provide some perspectives on the advantages gained from a better understanding of host-pathogen interactions. This includes their implications for the future development of PEDV and PDCoV vaccines and how we can further our knowledge of the molecular mechanisms underlying virus pathogenesis, virulence, and host coevolution.
Collapse
Affiliation(s)
- Surapong Koonpaew
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Samaporn Teeravechyan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Phanramphoei Namprachan Frantz
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Thanathom Chailangkarn
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| |
Collapse
|
29
|
Influenza A Virus M2 Protein Apical Targeting Is Required for Efficient Virus Replication. J Virol 2018; 92:JVI.01425-18. [PMID: 30158290 DOI: 10.1128/jvi.01425-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 11/20/2022] Open
Abstract
The influenza A virus (IAV) M2 protein is a multifunctional protein with critical roles in virion entry, assembly, and budding. M2 is targeted to the apical plasma membrane of polarized epithelial cells, and the interaction of the viral proteins M2, M1, HA, and NA near glycolipid rafts in the apical plasma membrane is hypothesized to coordinate the assembly of infectious virus particles. To determine the role of M2 protein apical targeting in IAV replication, a panel of M2 proteins with basolateral plasma membrane (M2-Baso) or endoplasmic reticulum (M2-ER) targeting sequences was generated. MDCK II cells stably expressing M2-Baso, but not M2-ER, complemented the replication of M2-stop viruses. However, in primary human nasal epithelial cell (hNEC) cultures, viruses encoding M2-Baso and M2-ER replicated to negligible titers compared to those of wild-type virus. M2-Baso replication was negatively correlated with cell polarization. These results demonstrate that M2 apical targeting is essential for IAV replication: targeting M2 to the ER results in a strong, cell type-independent inhibition of virus replication, and targeting M2 to the basolateral membrane has greater effects in hNECs than in MDCK cells.IMPORTANCE Influenza A virus assembly and particle release occur at the apical membrane of polarized epithelial cells. The integral membrane proteins encoded by the virus, HA, NA, and M2, are all targeted to the apical membrane and believed to recruit the other structural proteins to sites of virus assembly. By targeting M2 to the basolateral or endoplasmic reticulum membranes, influenza A virus replication was significantly reduced. Basolateral targeting of M2 reduced the infectious virus titers with minimal effects on virus particle release, while targeting to the endoplasmic reticulum resulted in reduced infectious and total virus particle release. Therefore, altering the expression and the intracellular targeting of M2 has major effects on virus replication.
Collapse
|
30
|
Thomas M, Banks L. Upsetting the Balance: When Viruses Manipulate Cell Polarity Control. J Mol Biol 2018; 430:3481-3503. [PMID: 29680664 PMCID: PMC7094317 DOI: 10.1016/j.jmb.2018.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/12/2018] [Accepted: 04/14/2018] [Indexed: 12/20/2022]
Abstract
The central importance of cell polarity control is emphasized by the frequency with which it is targeted by many diverse viruses. It is clear that in targeting key polarity control proteins, viruses affect not only host cell polarity, but also influence many cellular processes, including transcription, replication, and innate and acquired immunity. Examination of the interactions of different virus proteins with the cell and its polarity controls during the virus life cycles, and in virally-induced cell transformation shows ever more clearly how intimately all cellular processes are linked to the control of cell polarity.
Collapse
|
31
|
Coronavirus nucleocapsid proteins assemble constitutively in high molecular oligomers. Sci Rep 2017; 7:5740. [PMID: 28720894 PMCID: PMC5515880 DOI: 10.1038/s41598-017-06062-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/07/2017] [Indexed: 01/19/2023] Open
Abstract
Coronaviruses (CoV) are enveloped viruses and rely on their nucleocapsid N protein to incorporate the positive-stranded genomic RNA into the virions. CoV N proteins form oligomers but the mechanism and relevance underlying their multimerization remain to be fully understood. Using in vitro pull-down experiments and density glycerol gradients, we found that at least 3 regions distributed over its entire length mediate the self-interaction of mouse hepatitis virus (MHV) and severe acute respiratory syndrome coronavirus (SARS-CoV) N protein. The fact that these regions can bind reciprocally between themselves provides a possible molecular basis for N protein oligomerization. Interestingly, cytoplasmic N molecules of MHV-infected cells constitutively assemble into oligomers through a process that does not require binding to genomic RNA. Based on our data, we propose a model where constitutive N protein oligomerization allows the optimal loading of the genomic viral RNA into a ribonucleoprotein complex via the presentation of multiple viral RNA binding motifs.
Collapse
|
32
|
Cong Y, Verlhac P, Reggiori F. The Interaction between Nidovirales and Autophagy Components. Viruses 2017; 9:E182. [PMID: 28696396 PMCID: PMC5537674 DOI: 10.3390/v9070182] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 06/28/2017] [Accepted: 07/03/2017] [Indexed: 12/15/2022] Open
Abstract
Autophagy is a conserved intracellular catabolic pathway that allows cells to maintain homeostasis through the degradation of deleterious components via specialized double-membrane vesicles called autophagosomes. During the past decades, it has been revealed that numerous pathogens, including viruses, usurp autophagy in order to promote their propagation. Nidovirales are an order of enveloped viruses with large single-stranded positive RNA genomes. Four virus families (Arterividae, Coronaviridae, Mesoniviridae, and Roniviridae) are part of this order, which comprises several human and animal pathogens of medical and veterinary importance. In host cells, Nidovirales induce membrane rearrangements including autophagosome formation. The relevance and putative mechanism of autophagy usurpation, however, remain largely elusive. Here, we review the current knowledge about the possible interplay between Nidovirales and autophagy.
Collapse
Affiliation(s)
- Yingying Cong
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Pauline Verlhac
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Fulvio Reggiori
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
33
|
Kanner-Acerbo E, Lowe J. Review of immunological responses to porcine coronaviruses and implications on population based control strategies in epidemic and endemic infections. World J Immunol 2016; 6:60-66. [DOI: 10.5411/wji.v6.i1.60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/06/2015] [Accepted: 03/16/2016] [Indexed: 02/05/2023] Open
Abstract
Five major porcine coronaviruses (COVs) have been identified which cause severe gastrointestinal (GI) and respiratory disease in pigs. They include transmissible gastroenteritis (TGEV), porcine epidemic diarrhea virus (PEDV), porcine deltacoronavirus, porcine respiratory coronavirus, and porcine hemagglutinating encephalomyelitis. These diseases, especially TGEV and PEDV, have caused epidemics in Europe, Asia, and the Americas over the past 50 years, causing significant economic losses to swine producers. As pigs are a major protein source worldwide there is great interest in understanding, controlling, and preventing these diseases. These diseases have no cure, and current vaccines are not fully protective. On-farm prevention and biosecurity are difficult to enforce and have not stopped the spread of these diseases between herds. Recent advances in the immunology of porcine COVs has revealed that the immune response to porcine COVs shares many similarities with the response to human COVs, leading to increased interest in pigs as models for human disease. Highlights of these advances include the key role of local antigen presenting cells in the gastrointestinal tract in stimulating a protective immune response. This understanding has lead to new proposed vaccines. Advances in the understanding of the ways the viruses evade and degrade the host immune system have also lead to novel proposed therapies. Many of these therapies are in the early development stages, as researchers attempt to create efficacious, cost-effective, and practical therapies for these diseases.
Collapse
|
34
|
Warnes SL, Little ZR, Keevil CW. Human Coronavirus 229E Remains Infectious on Common Touch Surface Materials. mBio 2015; 6:e01697-15. [PMID: 26556276 PMCID: PMC4659470 DOI: 10.1128/mbio.01697-15] [Citation(s) in RCA: 285] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 10/13/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED The evolution of new and reemerging historic virulent strains of respiratory viruses from animal reservoirs is a significant threat to human health. Inefficient human-to-human transmission of zoonotic strains may initially limit the spread of transmission, but an infection may be contracted by touching contaminated surfaces. Enveloped viruses are often susceptible to environmental stresses, but the human coronaviruses responsible for severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) have recently caused increasing concern of contact transmission during outbreaks. We report here that pathogenic human coronavirus 229E remained infectious in a human lung cell culture model following at least 5 days of persistence on a range of common nonbiocidal surface materials, including polytetrafluoroethylene (Teflon; PTFE), polyvinyl chloride (PVC), ceramic tiles, glass, silicone rubber, and stainless steel. We have shown previously that noroviruses are destroyed on copper alloy surfaces. In this new study, human coronavirus 229E was rapidly inactivated on a range of copper alloys (within a few minutes for simulated fingertip contamination) and Cu/Zn brasses were very effective at lower copper concentration. Exposure to copper destroyed the viral genomes and irreversibly affected virus morphology, including disintegration of envelope and dispersal of surface spikes. Cu(I) and Cu(II) moieties were responsible for the inactivation, which was enhanced by reactive oxygen species generation on alloy surfaces, resulting in even faster inactivation than was seen with nonenveloped viruses on copper. Consequently, copper alloy surfaces could be employed in communal areas and at any mass gatherings to help reduce transmission of respiratory viruses from contaminated surfaces and protect the public health. IMPORTANCE Respiratory viruses are responsible for more deaths globally than any other infectious agent. Animal coronaviruses that "host jump" to humans result in severe infections with high mortality, such as severe acute respiratory syndrome (SARS) and, more recently, Middle East respiratory syndrome (MERS). We show here that a closely related human coronavirus, 229E, which causes upper respiratory tract infection in healthy individuals and serious disease in patients with comorbidities, remained infectious on surface materials common to public and domestic areas for several days. The low infectious dose means that this is a significant infection risk to anyone touching a contaminated surface. However, rapid inactivation, irreversible destruction of viral RNA, and massive structural damage were observed in coronavirus exposed to copper and copper alloy surfaces. Incorporation of copper alloy surfaces in conjunction with effective cleaning regimens and good clinical practice could help to control transmission of respiratory coronaviruses, including MERS and SARS.
Collapse
Affiliation(s)
- Sarah L Warnes
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Zoë R Little
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - C William Keevil
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
35
|
Cong Y, Li X, Bai Y, Lv X, Herrler G, Enjuanes L, Zhou X, Qu B, Meng F, Cong C, Ren X, Li G. Porcine aminopeptidase N mediated polarized infection by porcine epidemic diarrhea virus in target cells. Virology 2015; 478:1-8. [PMID: 25681796 PMCID: PMC7112081 DOI: 10.1016/j.virol.2015.01.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/15/2015] [Accepted: 01/23/2015] [Indexed: 12/16/2022]
Abstract
Infection of polarized intestinal epithelial cells by porcine epidemic diarrhea virus (PEDV) was characterized. Indirect immunofluorescence assay, real-time PCR, and transmission electron microscopy confirmed PEDV can be successfully propagated in immortalized swine small intestine epithelial cells (IECs). Infection involved porcine aminpeptidase N (pAPN), a reported cellular receptor for PEDV, transient expression of pAPN and siRNA targeted pAPN increased and decreased the infectivity of PEDV in IECs, respectively. Subsequently, polarized entry into and release from both Vero E6 and IECs was analyzed. PEDV entry into polarized cells and pAPN grown on membrane inserts occurs via apical membrane. The progeny virus released into the medium was also quantified which demonstrated that PEDV is preferentially released from the apical membrane. Collectively, our data demonstrate that pAPN, the cellular receptor for PEDV, mediates polarized PEDV infection. These results imply the possibility that PEDV infection may proceed by lateral spread of virus in intestinal epithelial cells.
Collapse
Affiliation(s)
- Yingying Cong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoxue Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yunyun Bai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiaonan Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience & Technology of China, Beijing 100090, China
| | - Georg Herrler
- Institute for Virology, University of Veterinary Medicine, Hannover D-30559, Germany
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Xingdong Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Bo Qu
- Faculty of Life Sciences, Northeast Agricultural University, Harbin 150030, China
| | - Fandan Meng
- Institute for Virology, University of Veterinary Medicine, Hannover D-30559, Germany
| | - Chengcheng Cong
- College Animal Husbandry and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161, China
| | - Xiaofeng Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Guangxing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
36
|
Cong Y, Ren X. Coronavirus entry and release in polarized epithelial cells: a review. Rev Med Virol 2014; 24:308-15. [PMID: 24737708 PMCID: PMC7169134 DOI: 10.1002/rmv.1792] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 03/20/2014] [Accepted: 03/20/2014] [Indexed: 12/03/2022]
Abstract
Most coronaviruses cause respiratory or intestinal infections in their animal or human host. Hence, their interaction with polarized epithelial cells plays a critical role in the onset and outcome of infection. In this paper, we review the knowledge regarding the entry and release of coronaviruses, with particular emphasis on the severe acute respiratory syndrome and Middle East respiratory syndrome coronaviruses. As these viruses approach the epithelial surfaces from the apical side, it is not surprising that coronavirus cell receptors are exposed primarily on the apical domain of polarized epithelial cells. With respect to release, all possibilities appear to occur. Thus, most coronaviruses exit through the apical surface, several through the basolateral one, although the Middle East respiratory syndrome coronavirus appears to use both sides. These observations help us understand the local or systematic spread of the infection within its host as well as the spread of the virus within the host population. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yingying Cong
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | | |
Collapse
|