251
|
Caragliano E, Bonazza S, Frascaroli G, Tang J, Soh TK, Grünewald K, Bosse JB, Brune W. Human cytomegalovirus forms phase-separated compartments at viral genomes to facilitate viral replication. Cell Rep 2022; 38:110469. [PMID: 35263605 PMCID: PMC8924372 DOI: 10.1016/j.celrep.2022.110469] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/29/2021] [Accepted: 02/10/2022] [Indexed: 12/21/2022] Open
Abstract
Human cytomegalovirus (HCMV) replicates its DNA genome in specialized replication compartments (RCs) in the host cell nucleus. These membrane-less organelles originate as spherical structures and grow in size over time. However, the mechanism of RC biogenesis has remained understudied. Using live-cell imaging and photo-oligomerization, we show that a central component of RCs, the UL112-113 proteins, undergo liquid-liquid phase separation (LLPS) to form RCs in the nucleus. We show that the self-interacting domain and large intrinsically disordered regions of UL112-113 are required for LLPS. Importantly, viral DNA induces local clustering of these proteins and lowers the threshold for phase separation. The formation of phase-separated compartments around viral genomes is necessary to recruit the viral DNA polymerase for viral genome replication. Thus, HCMV uses its UL112-113 proteins to generate RCs around viral genomes by LLPS to ensure the formation of a pro-replicative environment.
Collapse
Affiliation(s)
- Enrico Caragliano
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; Centre for Structural Systems Biology, 22607 Hamburg, Germany; Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Stefano Bonazza
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany
| | - Giada Frascaroli
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany
| | - Jiajia Tang
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany
| | - Timothy K Soh
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; Centre for Structural Systems Biology, 22607 Hamburg, Germany; Institute of Virology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Kay Grünewald
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; Centre for Structural Systems Biology, 22607 Hamburg, Germany; Institute of Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Jens B Bosse
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; Centre for Structural Systems Biology, 22607 Hamburg, Germany; Institute of Virology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| | - Wolfram Brune
- Leibniz Institute for Experimental Virology (HPI), 20251 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.
| |
Collapse
|
252
|
Abstract
Biomolecular condensates are intracellular organelles that are not bounded by membranes and often show liquid-like, dynamic material properties. They typically contain various types of proteins and nucleic acids. How the interaction of proteins and nucleic acids finally results in dynamic condensates is not fully understood. Here we use optical tweezers and fluorescence microscopy to study how the prototypical prion-like protein Fused-in-Sarcoma (FUS) condenses with individual molecules of single- and double-stranded DNA. We find that FUS adsorbs on DNA in a monolayer and hence generates an effectively sticky FUS–DNA polymer that collapses and finally forms a dynamic, reversible FUS–DNA co-condensate. We speculate that protein monolayer-based protein–nucleic acid co-condensation is a general mechanism for forming intracellular membraneless organelles. Biomolecular condensates provide distinct compartments that can localize and organize biochemistry inside cells. Recent evidence suggests that condensate formation is prevalent in the cell nucleus. To understand how different components of the nucleus interact during condensate formation is an important challenge. In particular, the physics of co-condensation of proteins together with nucleic acids remains elusive. Here we use optical tweezers to study how the prototypical prion-like protein Fused-in-Sarcoma (FUS) forms liquid-like assemblies in vitro, by co-condensing together with individual DNA molecules. Through progressive force-induced peeling of dsDNA, buffer exchange, and force measurements, we show that FUS adsorbing in a single layer on DNA effectively generates a sticky FUS–DNA polymer that can collapse to form a liquid-like FUS–DNA co-condensate. Condensation occurs at constant DNA tension for double-stranded DNA, which is a signature of phase separation. We suggest that co-condensation mediated by protein monolayer adsorption on nucleic acids is an important mechanism for intracellular compartmentalization.
Collapse
|
253
|
Vo HTM, Maestri A, Auerswald H, Sorn S, Lay S, Seng H, Sann S, Ya N, Pean P, Dussart P, Schwartz O, Ly S, Bruel T, Ly S, Duong V, Karlsson EA, Cantaert T. Robust and Functional Immune Memory Up to 9 Months After SARS-CoV-2 Infection: A Southeast Asian Longitudinal Cohort. Front Immunol 2022; 13:817905. [PMID: 35185909 PMCID: PMC8853741 DOI: 10.3389/fimmu.2022.817905] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/10/2022] [Indexed: 01/14/2023] Open
Abstract
The duration of humoral and cellular immune memory following SARS-CoV-2 infection in populations in least developed countries remains understudied but is key to overcome the current SARS-CoV-2 pandemic. Sixty-four Cambodian individuals with laboratory-confirmed infection with asymptomatic or mild/moderate clinical presentation were evaluated for Spike (S)-binding and neutralizing antibodies and antibody effector functions during acute phase of infection and at 6-9 months follow-up. Antigen-specific B cells, CD4+ and CD8+ T cells were characterized, and T cells were interrogated for functionality at late convalescence. Anti-S antibody titers decreased over time, but effector functions mediated by S-specific antibodies remained stable. S- and nucleocapsid (N)-specific B cells could be detected in late convalescence in the activated memory B cell compartment and are mostly IgG+. CD4+ and CD8+ T cell immune memory was maintained to S and membrane (M) protein. Asymptomatic infection resulted in decreased antibody-dependent cellular cytotoxicity (ADCC) and frequency of SARS-CoV-2-specific CD4+ T cells at late convalescence. Whereas anti-S antibodies correlated with S-specific B cells, there was no correlation between T cell response and humoral immune memory. Hence, all aspects of a protective immune response are maintained up to nine months after SARS-CoV-2 infection and in the absence of re-infection.
Collapse
Affiliation(s)
- Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Alvino Maestri
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sopheak Sorn
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Heng Seng
- Department of Communicable Disease Control, Ministry of Health (CDC-MoH), Phnom Penh, Cambodia
| | - Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Nisa Ya
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Polidy Pean
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Sovann Ly
- Department of Communicable Disease Control, Ministry of Health (CDC-MoH), Phnom Penh, Cambodia
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France.,Vaccine Research Institute, Créteil, France
| | - Sowath Ly
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Erik A Karlsson
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| |
Collapse
|
254
|
Cascarina SM, Ross ED. Phase separation by the SARS-CoV-2 nucleocapsid protein: Consensus and open questions. J Biol Chem 2022; 298:101677. [PMID: 35131265 PMCID: PMC8813722 DOI: 10.1016/j.jbc.2022.101677] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 01/09/2023] Open
Abstract
In response to the recent SARS-CoV-2 pandemic, a number of labs across the world have reallocated their time and resources to better our understanding of the virus. For some viruses, including SARS-CoV-2, viral proteins can undergo phase separation: a biophysical process often related to the partitioning of protein and RNA into membraneless organelles in vivo. In this review, we discuss emerging observations of phase separation by the SARS-CoV-2 nucleocapsid (N) protein-an essential viral protein required for viral replication-and the possible in vivo functions that have been proposed for N-protein phase separation, including viral replication, viral genomic RNA packaging, and modulation of host-cell response to infection. Additionally, since a relatively large number of studies examining SARS-CoV-2 N-protein phase separation have been published in a short span of time, we take advantage of this situation to compare results from similar experiments across studies. Our evaluation highlights potential strengths and pitfalls of drawing conclusions from a single set of experiments, as well as the value of publishing overlapping scientific observations performed simultaneously by multiple labs.
Collapse
Affiliation(s)
- Sean M Cascarina
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Eric D Ross
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA.
| |
Collapse
|
255
|
Khan WH, Khan N, Mishra A, Gupta S, Bansode V, Mehta D, Bhambure R, Ansari MA, Das S, Rathore AS. Dimerization of SARS-CoV-2 nucleocapsid protein affects sensitivity of ELISA based diagnostics of COVID-19. Int J Biol Macromol 2022; 200:428-437. [PMID: 35051498 PMCID: PMC8762837 DOI: 10.1016/j.ijbiomac.2022.01.094] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 12/27/2022]
Abstract
Nucleocapsid protein (N protein) is the primary antigen of the virus for development of sensitive diagnostic assays of COVID-19. In this paper, we demonstrate the significant impact of dimerization of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) N-protein on sensitivity of enzyme-linked immunosorbent assay (ELISA) based diagnostics. The expressed purified protein from E. coli is composed of dimeric and monomeric forms, which have been further characterized using biophysical and immunological techniques. Indirect ELISA indicated elevated susceptibility of the dimeric form of the nucleocapsid protein for identification of protein-specific monoclonal antibody as compared to the monomeric form. This finding also confirmed with the modelled structure of monomeric and dimeric nucleocapsid protein via HHPred software and its solvent accessible surface area, which indicates higher stability and antigenicity of the dimeric type as compared to the monomeric form. The sensitivity and specificity of the ELISA at 95% CI are 99.0% (94.5-99.9) and 95.0% (83.0-99.4), respectively, for the highest purified dimeric form of the N protein. As a result, using the highest purified dimeric form will improve the sensitivity of the current nucleocapsid-dependent ELISA for COVID-19 diagnosis, and manufacturers should monitor and maintain the monomer-dimer composition for accurate and robust diagnostics.
Collapse
Affiliation(s)
- Wajihul Hasan Khan
- Department of Chemical Engineering, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Nida Khan
- Department of Chemical Engineering, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Avinash Mishra
- Department of Chemical Engineering, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Surbhi Gupta
- Department of Chemical Engineering, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Vikrant Bansode
- Department of Chemical Engineering, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Deepa Mehta
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Dr Homi Bhabha Rd, Pune, Maharashtra 411008, India
| | - Rahul Bhambure
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Dr Homi Bhabha Rd, Pune, Maharashtra 411008, India
| | - M. Ahmad Ansari
- Department of Microbiology, The University College of Medical Sciences (UCMS) and Guru Teg Bahadur Hospital (GTB), Dilshad Garden, Delhi 110095, India
| | - Shukla Das
- Department of Microbiology, The University College of Medical Sciences (UCMS) and Guru Teg Bahadur Hospital (GTB), Dilshad Garden, Delhi 110095, India
| | - Anurag S. Rathore
- Department of Chemical Engineering, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India,Corresponding author at: DBT Centre of Excellence for Biopharmaceutical Technology, Department of Chemical Engineering, Indian Institute of Technology, Delhi Hauz Khas, New Delhi 110016, India
| |
Collapse
|
256
|
Obermeyer F, Jankowiak M, Barkas N, Schaffner SF, Pyle JD, Yurkovetskiy L, Bosso M, Park DJ, Babadi M, MacInnis BL, Luban J, Sabeti PC, Lemieux JE. Analysis of 6.4 million SARS-CoV-2 genomes identifies mutations associated with fitness. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2021.09.07.21263228. [PMID: 35194619 PMCID: PMC8863165 DOI: 10.1101/2021.09.07.21263228] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Repeated emergence of SARS-CoV-2 variants with increased fitness necessitates rapid detection and characterization of new lineages. To address this need, we developed PyR 0 , a hierarchical Bayesian multinomial logistic regression model that infers relative prevalence of all viral lineages across geographic regions, detects lineages increasing in prevalence, and identifies mutations relevant to fitness. Applying PyR 0 to all publicly available SARS-CoV-2 genomes, we identify numerous substitutions that increase fitness, including previously identified spike mutations and many non-spike mutations within the nucleocapsid and nonstructural proteins. PyR 0 forecasts growth of new lineages from their mutational profile, identifies viral lineages of concern as they emerge, and prioritizes mutations of biological and public health concern for functional characterization. ONE SENTENCE SUMMARY A Bayesian hierarchical model of all SARS-CoV-2 viral genomes predicts lineage fitness and identifies associated mutations.
Collapse
Affiliation(s)
- Fritz Obermeyer
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
- Pyro Committee, Linux AI & Data Foundation; 548 Market St San Francisco, California 94104
| | - Martin Jankowiak
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
- Pyro Committee, Linux AI & Data Foundation; 548 Market St San Francisco, California 94104
| | - Nikolaos Barkas
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
| | - Stephen F. Schaffner
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
- Department of Organismic and Evolutionary Biology, Harvard University; Cambridge, MA 02138, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University; Boston, MA, USA
| | - Jesse D. Pyle
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
| | - Lonya Yurkovetskiy
- Program in Molecular Medicine, University of Massachusetts Medical School; Worcester, MA 01605, USA
| | - Matteo Bosso
- Program in Molecular Medicine, University of Massachusetts Medical School; Worcester, MA 01605, USA
| | - Daniel J. Park
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
| | - Mehrtash Babadi
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
| | - Bronwyn L. MacInnis
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University; Boston, MA, USA
- Massachusetts Consortium on Pathogen Readiness; Boston, MA 02115, USA
| | - Jeremy Luban
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
- Program in Molecular Medicine, University of Massachusetts Medical School; Worcester, MA 01605, USA
- Massachusetts Consortium on Pathogen Readiness; Boston, MA 02115, USA
- Ragon Institute of MGH, MIT, and Harvard; 400 Technology Square, Cambridge, MA 02139, USA
| | - Pardis C. Sabeti
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
- Department of Organismic and Evolutionary Biology, Harvard University; Cambridge, MA 02138, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University; Boston, MA, USA
- Massachusetts Consortium on Pathogen Readiness; Boston, MA 02115, USA
- Howard Hughes Medical Institute; 4000 Jones Bridge Rd, Chevy Chase, MD 20815, USA
| | - Jacob E. Lemieux
- Broad Institute of MIT and Harvard; 415 Main Street, Cambridge, MA 02142, USA
- Division of Infectious Diseases, Massachusetts General Hospital; Boston, MA, USA
| |
Collapse
|
257
|
Kumar N, Shetti NP, Jagannath S, Aminabhavi TM. Electrochemical sensors for the detection of SARS-CoV-2 virus. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2022; 430:132966. [PMID: 34690533 PMCID: PMC8525496 DOI: 10.1016/j.cej.2021.132966] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/29/2021] [Accepted: 10/10/2021] [Indexed: 05/09/2023]
Abstract
Coronavirus (COVID-19), a deadly pandemic has spread worldwide and created many global health issues. Though methods of its detection are being continuously developed for the early detection and monitoring of COVID-19, still there is need for more novel methods. The presently used methods include rapid antigen tests, serological surveys, reverse transcription-polymerase chain reaction (RT-PCR), artificial intelligence-based techniques, and assays based on sensors/biosensors. Of all these, RT-PCR test has high sensitivity and specificity though it requires more time for testing and need for skilled technicians. Recently, electrochemical sensors have been developed for rapid monitoring and detection of SARS-CoV-2 from the patient's biological fluid samples. This review covers the recently developed electrochemical sensors that are focused on the detection of viral nucleic acid, immunoglobulin, antigen, and the entire viral particles. In addition, we also compare and assess their detection limits, sensitivities and specificities for the identification and monitoring of COVID-19. Furthermore, this review will address the best practices for the development of electrochemical sensors such as electrode fouling, limit of detection/limit of quantification determination and verification.
Collapse
Affiliation(s)
- Neeraj Kumar
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012, India
| | - Nagaraj P Shetti
- School of Advanced Sciences, KLE Technological University, Hubballi 580 031, India
| | - Somanath Jagannath
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Tejraj M Aminabhavi
- School of Advanced Sciences, KLE Technological University, Hubballi 580 031, India
| |
Collapse
|
258
|
Abstract
The 14-3-3 family proteins are vital scaffold proteins that ubiquitously expressed in various tissues. They interact with numerous protein targets and mediate many cellular signaling pathways. The 14-3-3 binding motifs are often embedded in intrinsically disordered regions which are closely associated with liquid-liquid phase separation (LLPS). In the past ten years, LLPS has been observed for a variety of proteins and biological processes, indicating that LLPS plays a fundamental role in the formation of membraneless organelles and cellular condensates. While extensive investigations have been performed on 14-3-3 proteins, its involvement in LLPS is overlooked. To date, 14-3-3 proteins have not been reported to undergo LLPS alone or regulate LLPS of their binding partners. To reveal the potential involvement of 14-3-3 proteins in LLPS, in this review, we summarized the LLPS propensity of 14-3-3 binding partners and found that about one half of them may undergo LLPS spontaneously. We further analyzed the phase separation behavior of representative 14-3-3 binders and discussed how 14-3-3 proteins may be involved. By modulating the conformation and valence of interactions and recruiting other molecules, we speculate that 14-3-3 proteins can efficiently regulate the functions of their targets in the context of LLPS. Considering the critical roles of 14-3-3 proteins, there is an urgent need for investigating the involvement of 14-3-3 proteins in the phase separation process of their targets and the underling mechanisms.
Collapse
|
259
|
Zhao H, Nguyen A, Wu D, Li Y, Hassan SA, Chen J, Shroff H, Piszczek G, Schuck P. Plasticity in structure and assembly of SARS-CoV-2 nucleocapsid protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.02.08.479556. [PMID: 35169797 PMCID: PMC8845419 DOI: 10.1101/2022.02.08.479556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Worldwide SARS-CoV-2 sequencing efforts track emerging mutations in its spike protein, as well as characteristic mutations in other viral proteins. Besides their epidemiological importance, the observed SARS-CoV-2 sequences present an ensemble of viable protein variants, and thereby a source of information on viral protein structure and function. Charting the mutational landscape of the nucleocapsid (N) protein that facilitates viral assembly, we observe variability exceeding that of the spike protein, with more than 86% of residues that can be substituted, on average by 3-4 different amino acids. However, mutations exhibit an uneven distribution that tracks known structural features but also reveals highly protected stretches of unknown function. One of these conserved regions is in the central disordered linker proximal to the N-G215C mutation that has become dominant in the Delta variant, outcompeting G215 variants without further spike or N-protein substitutions. Structural models suggest that the G215C mutation stabilizes conserved transient helices in the disordered linker serving as protein-protein interaction interfaces. Comparing Delta variant N-protein to its ancestral version in biophysical experiments, we find a significantly more compact and less disordered structure. N-G215C exhibits substantially stronger self-association, shifting the unliganded protein from a dimeric to a tetrameric oligomeric state, which leads to enhanced co-assembly with nucleic acids. This suggests that the sequence variability of N-protein is mirrored by high plasticity of N-protein biophysical properties, which we hypothesize can be exploited by SARS-CoV-2 to achieve greater efficiency of viral assembly, and thereby enhanced infectivity.
Collapse
Affiliation(s)
- Huaying Zhao
- Laboratory of Dynamics of Macromolecular Assembly, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ai Nguyen
- Laboratory of Dynamics of Macromolecular Assembly, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Di Wu
- Biophysics Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Li
- Proteomics Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sergio A. Hassan
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jiji Chen
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hari Shroff
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Grzegorz Piszczek
- Biophysics Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter Schuck
- Laboratory of Dynamics of Macromolecular Assembly, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
260
|
Gray-Rodriguez S, Jensen MP, Otero-Jimenez M, Hanley B, Swann OC, Ward PA, Salguero FJ, Querido N, Farkas I, Velentza-Almpani E, Weir J, Barclay WS, Carroll MW, Jaunmuktane Z, Brandner S, Pohl U, Allinson K, Thom M, Troakes C, Al-Sarraj S, Sastre M, Gveric D, Gentleman S, Roufosse C, Osborn M, Alegre-Abarrategui J. Multisystem screening reveals SARS-CoV-2 in neurons of the myenteric plexus and in megakaryocytes. J Pathol 2022; 257:198-217. [PMID: 35107828 PMCID: PMC9325073 DOI: 10.1002/path.5878] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/12/2021] [Accepted: 01/29/2022] [Indexed: 11/30/2022]
Abstract
SARS‐CoV‐2, the causative agent of COVID‐19, typically manifests as a respiratory illness, although extrapulmonary involvement, such as in the gastrointestinal tract and nervous system, as well as frequent thrombotic events, are increasingly recognised. How this maps onto SARS‐CoV‐2 organ tropism at the histological level, however, remains unclear. Here, we perform a comprehensive validation of a monoclonal antibody against the SARS‐CoV‐2 nucleocapsid protein (NP) followed by systematic multisystem organ immunohistochemistry analysis of the viral cellular tropism in tissue from 36 patients, 16 postmortem cases and 16 biopsies with polymerase chain reaction (PCR)‐confirmed SARS‐CoV‐2 status from the peaks of the pandemic in 2020 and four pre‐COVID postmortem controls. SARS‐CoV‐2 anti‐NP staining in the postmortem cases revealed broad multiorgan involvement of the respiratory, digestive, haematopoietic, genitourinary and nervous systems, with a typical pattern of staining characterised by punctate paranuclear and apical cytoplasmic labelling. The average time from symptom onset to time of death was shorter in positively versus negatively stained postmortem cases (mean = 10.3 days versus mean = 20.3 days, p = 0.0416, with no cases showing definitive staining if the interval exceeded 15 days). One striking finding was the widespread presence of SARS‐CoV‐2 NP in neurons of the myenteric plexus, a site of high ACE2 expression, the entry receptor for SARS‐CoV‐2, and one of the earliest affected cells in Parkinson's disease. In the bone marrow, we observed viral SARS‐CoV‐2 NP within megakaryocytes, key cells in platelet production and thrombus formation. In 15 tracheal biopsies performed in patients requiring ventilation, there was a near complete concordance between immunohistochemistry and PCR swab results. Going forward, our findings have relevance to correlating clinical symptoms with the organ tropism of SARS‐CoV‐2 in contemporary cases as well as providing insights into potential long‐term complications of COVID‐19. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sandra Gray-Rodriguez
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Melanie P Jensen
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK
| | - Maria Otero-Jimenez
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Brian Hanley
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK.,Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK
| | - Olivia C Swann
- Department of Infectious Disease, Imperial College London, London, UK
| | - Patrick A Ward
- Chelsea and Westminster NHS Foundation Trust, London, UK
| | - Francisco J Salguero
- National Infection Service, United Kingdom Health Security Agency, Porton Down, Salisbury, UK
| | - Nadira Querido
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Ildiko Farkas
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | | | - Justin Weir
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK
| | - Wendy S Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Miles W Carroll
- National Infection Service, United Kingdom Health Security Agency, Porton Down, Salisbury, UK.,Pandemic Sciences Centre, Nuffield Department of Medicine, Oxford University, OX3 7BN, UK
| | - Zane Jaunmuktane
- Department of Neuropathology, UCL Queen Square Institute of Neurology, London, UK
| | - Sebastian Brandner
- Department of Neuropathology, UCL Queen Square Institute of Neurology, London, UK
| | - Ute Pohl
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham/University Hospitals Birmingham, Birmingham, UK
| | - Kieren Allinson
- Department of Neuropathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Maria Thom
- Department of Neuropathology, UCL Queen Square Institute of Neurology, London, UK
| | - Claire Troakes
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Safa Al-Sarraj
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Djordje Gveric
- Multiple Sclerosis and Parkinson's Tissue Bank, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Steve Gentleman
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.,Multiple Sclerosis and Parkinson's Tissue Bank, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Candice Roufosse
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK.,Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK
| | - Michael Osborn
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK
| | - Javier Alegre-Abarrategui
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.,Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK.,Multiple Sclerosis and Parkinson's Tissue Bank, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| |
Collapse
|
261
|
Active site prediction of phosphorylated SARS-CoV-2 N-Protein using molecular simulation. INFORMATICS IN MEDICINE UNLOCKED 2022; 29:100889. [PMID: 35224174 PMCID: PMC8860464 DOI: 10.1016/j.imu.2022.100889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 11/23/2022] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) nucleocapsid protein (N-protein) is responsible for viral replication by assisting in viral RNA synthesis and attaching the viral genome to the replicase-transcriptase complex (RTC). Numerous studies suggested the N-protein as a drug target. However, the specific N-protein active sites for SARS-CoV-2 drug treatments are yet to be discovered. The purpose of this study was to determine active sites of the SARS-CoV-2 N-protein by identifying torsion angle classifiers for N-protein structural changes that correlated with the respective angle differences between the active and inactive N-protein. In the study, classifiers with a minimum accuracy of 80% determined from molecular simulation data were analyzed by Principal Component Analysis and cross-validated by Logistic Regression, Support Vector Machine, and Random Forest Classification. The ability of torsion angles ψ252 and φ375 to differentiate between phosphorylated and unphosphorylated structures suggested that residues 252 and 375 in the RNA binding domain might be important in N-protein activation. Furthermore, the φ and ψ angles of residue S189 correlated to a 90.7% structural determination accuracy. The key residues involved in the structural changes identified here might suggest possible important functional sites on the N-protein that could be the focus of further study to understand their potential as drug targets.
Collapse
|
262
|
Kankariya RA, Chaudhari AB, Dandi ND. Inhibitory efficacy of 2, 4-diacetylphloroglucinol against SARS-COV-2 proteins: in silico study. Biologia (Bratisl) 2022; 77:815-828. [PMID: 35125499 PMCID: PMC8800849 DOI: 10.1007/s11756-021-00979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/24/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Raksha A. Kankariya
- School of Life Sciences, Kavayitri Bahinabai Chaudhari North Maharashtra University, Jalgaon, MS 425001 India
| | - Ambalal B. Chaudhari
- School of Life Sciences, Kavayitri Bahinabai Chaudhari North Maharashtra University, Jalgaon, MS 425001 India
| | - Navin D. Dandi
- School of Life Sciences, Kavayitri Bahinabai Chaudhari North Maharashtra University, Jalgaon, MS 425001 India
| |
Collapse
|
263
|
Chen M, Yan C, Zheng L, Zhang XE. The smallest near-infrared fluorescence complementation system for imaging protein-protein and RNA-protein interactions. Chem Sci 2022; 13:1119-1129. [PMID: 35211278 PMCID: PMC8790895 DOI: 10.1039/d1sc04839b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Bimolecular fluorescence complementation (BiFC) and its derivative molecular biosensor systems provide effective tools for visualizing biomolecular interactions. The introduction of red and near-infrared fluorescence emission proteins has expanded the spectrum of signal generating modules, enabling BiFC for in vivo imaging. However, the large size of the signal module of BiFC can hinder the interaction between proteins under investigation. In this study, we constructed the near-infrared BiFC and TriFC systems by splitting miRFP670nano, the smallest cyanobacteriochrome-evolved phytochrome available. The miRFP670nano-BiFC sensor system identified and enabled visualization of protein–protein interactions in living cells and live mice, and afforded a faster maturation rate and higher photostability and cellular stability when compared with those of reported near-infrared BiFC systems. We used the miRFP670nano-BiFC sensor system to identify interactions between the nucleocapsid (N) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and cellular stress granule proteins in living cells and found that the N protein downregulated the expression level of granule protein G3BP1. With the advantages of small size and long wavelength emission of the signal module, the proposed molecular biosensor system should be suitable for various applications in cell imaging studies. The smallest near-infrared fluorescence complementation system for imaging protein–protein and RNA–protein interactions in living cells and live mice.![]()
Collapse
Affiliation(s)
- Minghai Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences Shenzhen 518055 China
| | - Chuang Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences Shenzhen 518055 China
| | - Luping Zheng
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences Shenzhen 518055 China
| | - Xian-En Zhang
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen 518055 China .,National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences Beijing 100101 China
| |
Collapse
|
264
|
Hyun YS, Lee YH, Jo HA, Baek IC, Kim SM, Sohn HJ, Kim TG. Comprehensive Analysis of CD4 + T Cell Response Cross-Reactive to SARS-CoV-2 Antigens at the Single Allele Level of HLA Class II. Front Immunol 2022; 12:774491. [PMID: 35069546 PMCID: PMC8770530 DOI: 10.3389/fimmu.2021.774491] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
Common human coronaviruses have been circulating undiagnosed worldwide. These common human coronaviruses share partial sequence homology with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); therefore, T cells specific to human coronaviruses are also cross-reactive with SARS-CoV-2 antigens. Herein, we defined CD4+ T cell responses that were cross-reactive with SARS-CoV-2 antigens in blood collected in 2016–2018 from healthy donors at the single allele level using artificial antigen-presenting cells (aAPC) expressing a single HLA class II allotype. We assessed the allotype-restricted responses in the 42 individuals using the aAPCs matched 22 HLA-DR alleles, 19 HLA-DQ alleles, and 13 HLA-DP alleles. The response restricted by the HLA-DR locus showed the highest magnitude, and that by HLA-DP locus was higher than that by HLA-DQ locus. Since two alleles of HLA-DR, -DQ, and -DP loci are expressed co-dominantly in an individual, six different HLA class II allotypes can be used to the cross-reactive T cell response. Of the 16 individuals who showed a dominant T cell response, five, one, and ten showed a dominant response by a single allotype of HLA-DR, -DQ, and -DP, respectively. The single allotype-restricted T cells responded to only one antigen in the five individuals and all the spike, membrane, and nucleocapsid proteins in the six individuals. In individuals heterozygous for the HLA-DPA and HLA-DPB loci, four combinations of HLA-DP can be expressed, but only one combination showed a dominant response. These findings demonstrate that cross-reactive T cells to SARS-CoV-2 respond with single-allotype dominance.
Collapse
Affiliation(s)
- You-Seok Hyun
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yong-Hun Lee
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyeong-A Jo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - In-Cheol Baek
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sun-Mi Kim
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyun-Jung Sohn
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Tai-Gyu Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
265
|
Bessa LM, Guseva S, Camacho-Zarco AR, Salvi N, Maurin D, Perez LM, Botova M, Malki A, Nanao M, Jensen MR, Ruigrok RWH, Blackledge M. The intrinsically disordered SARS-CoV-2 nucleoprotein in dynamic complex with its viral partner nsp3a. SCIENCE ADVANCES 2022; 8:eabm4034. [PMID: 35044811 PMCID: PMC8769549 DOI: 10.1126/sciadv.abm4034] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/24/2021] [Indexed: 05/10/2023]
Abstract
The processes of genome replication and transcription of SARS-CoV-2 represent important targets for viral inhibition. Betacoronaviral nucleoprotein (N) is a highly dynamic cofactor of the replication-transcription complex (RTC), whose function depends on an essential interaction with the amino-terminal ubiquitin-like domain of nsp3 (Ubl1). Here, we describe this complex (dissociation constant - 30 to 200 nM) at atomic resolution. The interaction implicates two linear motifs in the intrinsically disordered linker domain (N3), a hydrophobic helix (219LALLLLDRLNQL230) and a disordered polar strand (243GQTVTKKSAAEAS255), that mutually engage to form a bipartite interaction, folding N3 around Ubl1. This results in substantial collapse in the dimensions of dimeric N, forming a highly compact molecular chaperone, that regulates binding to RNA, suggesting a key role of nsp3 in the association of N to the RTC. The identification of distinct linear motifs that mediate an important interaction between essential viral factors provides future targets for development of innovative strategies against COVID-19.
Collapse
Affiliation(s)
| | - Serafima Guseva
- Université Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | | | - Nicola Salvi
- Université Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | - Damien Maurin
- Université Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | | | - Maiia Botova
- Université Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | - Anas Malki
- Université Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | - Max Nanao
- Structural Biology Group, European Synchrotron Radiation Facility, F-38000 Grenoble, France
| | | | | | | |
Collapse
|
266
|
The Broad-Spectrum Antiviral Potential of the Amphibian Peptide AR-23. Int J Mol Sci 2022; 23:ijms23020883. [PMID: 35055066 PMCID: PMC8779559 DOI: 10.3390/ijms23020883] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/23/2022] Open
Abstract
Viral infections represent a serious threat to the world population and are becoming more frequent. The search and identification of broad-spectrum antiviral molecules is necessary to ensure new therapeutic options, since there is a limited availability of effective antiviral drugs able to eradicate viral infections, and consequently due to the increase of strains that are resistant to the most used drugs. Recently, several studies on antimicrobial peptides identified them as promising antiviral agents. In detail, amphibian skin secretions serve as a rich source of natural antimicrobial peptides. Their antibacterial and antifungal activities have been widely reported, but their exploitation as potential antiviral agents have yet to be fully investigated. In the present study, the antiviral activity of the peptide derived from the secretion of Rana tagoi, named AR-23, was evaluated against both DNA and RNA viruses, with or without envelope. Different assays were performed to identify in which step of the infectious cycle the peptide could act. AR-23 exhibited a greater inhibitory activity in the early stages of infection against both DNA (HSV-1) and RNA (MeV, HPIV-2, HCoV-229E, and SARS-CoV-2) enveloped viruses and, on the contrary, it was inactive against naked viruses (PV-1). Altogether, the results indicated AR-23 as a peptide with potential therapeutic effects against a wide variety of human viruses.
Collapse
|
267
|
Pizzato M, Baraldi C, Boscato Sopetto G, Finozzi D, Gentile C, Gentile MD, Marconi R, Paladino D, Raoss A, Riedmiller I, Ur Rehman H, Santini A, Succetti V, Volpini L. SARS-CoV-2 and the Host Cell: A Tale of Interactions. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2021.815388] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ability of a virus to spread between individuals, its replication capacity and the clinical course of the infection are macroscopic consequences of a multifaceted molecular interaction of viral components with the host cell. The heavy impact of COVID-19 on the world population, economics and sanitary systems calls for therapeutic and prophylactic solutions that require a deep characterization of the interactions occurring between virus and host cells. Unveiling how SARS-CoV-2 engages with host factors throughout its life cycle is therefore fundamental to understand the pathogenic mechanisms underlying the viral infection and to design antiviral therapies and prophylactic strategies. Two years into the SARS-CoV-2 pandemic, this review provides an overview of the interplay between SARS-CoV-2 and the host cell, with focus on the machinery and compartments pivotal for virus replication and the antiviral cellular response. Starting with the interaction with the cell surface, following the virus replicative cycle through the characterization of the entry pathways, the survival and replication in the cytoplasm, to the mechanisms of egress from the infected cell, this review unravels the complex network of interactions between SARS-CoV-2 and the host cell, highlighting the knowledge that has the potential to set the basis for the development of innovative antiviral strategies.
Collapse
|
268
|
Scherer KM, Mascheroni L, Carnell GW, Wunderlich LCS, Makarchuk S, Brockhoff M, Mela I, Fernandez-Villegas A, Barysevich M, Stewart H, Suau Sans M, George CL, Lamb JR, Kaminski-Schierle GS, Heeney JL, Kaminski CF. SARS-CoV-2 nucleocapsid protein adheres to replication organelles before viral assembly at the Golgi/ERGIC and lysosome-mediated egress. SCIENCE ADVANCES 2022; 8:eabl4895. [PMID: 34995113 PMCID: PMC10954198 DOI: 10.1126/sciadv.abl4895] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 11/15/2021] [Indexed: 06/14/2023]
Abstract
Despite being the target of extensive research efforts due to the COVID-19 (coronavirus disease 2019) pandemic, relatively little is known about the dynamics of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication within cells. We investigate and characterize the tightly orchestrated virus assembly by visualizing the spatiotemporal dynamics of the four structural SARS-CoV-2 proteins at high resolution. The nucleoprotein is expressed first and accumulates around folded endoplasmic reticulum (ER) membranes in convoluted layers that contain viral RNA replication foci. We find that, of the three transmembrane proteins, the membrane protein appears at the Golgi apparatus/ER-to-Golgi intermediate compartment before the spike and envelope proteins. Relocation of a lysosome marker toward the assembly compartment and its detection in transport vesicles of viral proteins confirm an important role of lysosomes in SARS-CoV-2 egress. These data provide insights into the spatiotemporal regulation of SARS-CoV-2 assembly and refine the current understanding of SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Katharina M. Scherer
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Luca Mascheroni
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - George W. Carnell
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Lucia C. S. Wunderlich
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | | | - Marius Brockhoff
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Ioanna Mela
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Ana Fernandez-Villegas
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Max Barysevich
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Hazel Stewart
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Maria Suau Sans
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | | - Jacob R. Lamb
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | | | - Jonathan L. Heeney
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Clemens F. Kaminski
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| |
Collapse
|
269
|
Semerdzhiev SA, Fakhree MAA, Segers-Nolten I, Blum C, Claessens MMAE. Interactions between SARS-CoV-2 N-Protein and α-Synuclein Accelerate Amyloid Formation. ACS Chem Neurosci 2022; 13:143-150. [PMID: 34860005 PMCID: PMC8739828 DOI: 10.1021/acschemneuro.1c00666] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
![]()
First cases that
point at a correlation between SARS-CoV-2 infections
and the development of Parkinson’s disease (PD) have been reported.
Currently, it is unclear if there is also a direct causal link between
these diseases. To obtain first insights into a possible molecular
relation between viral infections and the aggregation of α-synuclein
protein into amyloid fibrils characteristic for PD, we investigated
the effect of the presence of SARS-CoV-2 proteins on α-synuclein
aggregation. We show, in test tube experiments, that SARS-CoV-2 spike
protein (S-protein) has no effect on α-synuclein aggregation,
while SARS-CoV-2 nucleocapsid protein (N-protein) considerably speeds
up the aggregation process. We observe the formation of multiprotein
complexes and eventually amyloid fibrils. Microinjection of N-protein
in SH-SY5Y cells disturbed the α-synuclein proteostasis and
increased cell death. Our results point toward direct interactions
between the N-protein of SARS-CoV-2 and α-synuclein as molecular
basis for the observed correlation between SARS-CoV-2 infections and
Parkinsonism.
Collapse
Affiliation(s)
- Slav A. Semerdzhiev
- Nanobiophysics, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Mohammad A. A. Fakhree
- Nanobiophysics, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Ine Segers-Nolten
- Nanobiophysics, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Christian Blum
- Nanobiophysics, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Mireille M. A. E. Claessens
- Nanobiophysics, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
270
|
Fertig TE, Chitoiu L, Terinte‐Balcan G, Peteu V, Marta D, Gherghiceanu M. The atomic portrait of SARS-CoV-2 as captured by cryo-electron microscopy. J Cell Mol Med 2022; 26:25-34. [PMID: 34904376 PMCID: PMC8742239 DOI: 10.1111/jcmm.17103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 01/18/2023] Open
Abstract
Transmission electron microscopy has historically been indispensable for virology research, as it offers unique insight into virus function. In the past decade, as cryo-electron microscopy (cryo-EM) has matured and become more accessible, we have been able to peer into the structure of viruses at the atomic level and understand how they interact with the host cell, with drugs or with antibodies. Perhaps, there was no time in recent history where cryo-EM was more needed, as SARS-CoV-2 has spread around the globe, causing millions of deaths and almost unquantifiable economic devastation. In this concise review, we aim to mark the most important contributions of cryo-EM to understanding the structure and function of SARS-CoV-2 proteins, from surface spikes to the virus core and from virus-receptor interactions to antibody binding.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/chemistry
- Angiotensin-Converting Enzyme 2/immunology
- Angiotensin-Converting Enzyme 2/metabolism
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/chemistry
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/biosynthesis
- COVID-19 Vaccines/chemistry
- Cryoelectron Microscopy
- Epitopes/chemistry
- Epitopes/immunology
- Epitopes/metabolism
- Humans
- Models, Molecular
- Protein Binding
- Protein Interaction Domains and Motifs
- Protein Structure, Secondary
- Receptors, Virus/chemistry
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- SARS-CoV-2/drug effects
- SARS-CoV-2/pathogenicity
- SARS-CoV-2/ultrastructure
- Serine Endopeptidases/chemistry
- Serine Endopeptidases/immunology
- Serine Endopeptidases/metabolism
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Virion/drug effects
- Virion/pathogenicity
- Virion/ultrastructure
Collapse
Affiliation(s)
- Tudor Emanuel Fertig
- Ultrastructural Pathology and Bioimaging LabVictor Babeș National Institute of PathologyBucharestRomania
- Carol Davila University of Medicine and PharmacyBucharestRomania
| | - Leona Chitoiu
- Ultrastructural Pathology and Bioimaging LabVictor Babeș National Institute of PathologyBucharestRomania
| | - George Terinte‐Balcan
- Ultrastructural Pathology and Bioimaging LabVictor Babeș National Institute of PathologyBucharestRomania
| | - Victor‐Eduard Peteu
- Ultrastructural Pathology and Bioimaging LabVictor Babeș National Institute of PathologyBucharestRomania
| | - Daciana Marta
- Ultrastructural Pathology and Bioimaging LabVictor Babeș National Institute of PathologyBucharestRomania
| | - Mihaela Gherghiceanu
- Ultrastructural Pathology and Bioimaging LabVictor Babeș National Institute of PathologyBucharestRomania
- Carol Davila University of Medicine and PharmacyBucharestRomania
| |
Collapse
|
271
|
Ino Y, Nishi M, Yamaoka Y, Miyakawa K, Jeremiah SS, Osada M, Kimura Y, Ryo A. Phosphopeptide enrichment using Phos-tag technology reveals functional phosphorylation of the nucleocapsid protein of SARS-CoV-2. J Proteomics 2022; 255:104501. [PMID: 35093569 PMCID: PMC8800104 DOI: 10.1016/j.jprot.2022.104501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/19/2022] [Accepted: 01/23/2022] [Indexed: 11/28/2022]
Abstract
Phosphorylation of viral proteins serves as a regulatory mechanism during the intracellular life cycle of infected viruses. There is therefore a pressing need to develop a method to efficiently purify and enrich phosphopeptides derived from viral particles in biological samples. In this study, we utilized Phos-tag technology to analyze the functional phosphorylation of the nucleocapsid protein (N protein; NP) of severe respiratory syndrome coronavirus 2 (SARS-CoV-2). Viral particles were collected from culture supernatants of SARS-CoV-2-infected VeroE6/TMPRSS2 cells by ultracentrifugation, and phosphopeptides were purified by Phos-tag magnetic beads for LC-MS/MS analysis. Analysis revealed that NP was reproducibly phosphorylated at serine 79 (Ser79). Multiple sequence alignment and phylogenetic analysis showed that the Ser79 was a distinct phospho-acceptor site in SARS-CoV-2 but not in other beta-coronaviruses. We also found that the prolyl-isomerase Pin1 bound to the phosphorylated Ser79 in NP and positively regulated the production of viral particles. These results suggest that SARS-CoV-2 may have acquired the potent virus-host interaction during its evolution mediated by viral protein phosphorylation. Moreover, Phos-tag technology can provide a useful means for analyzing the functional phosphorylation of viral proteins. Significance In this study, we aimed to investigate the functional phosphorylation of SARS-CoV-2 NP. For this purpose, we used Phos-tag technology to purify and enrich virus-derived phosphopeptides with high selectivity and reproducibility. This method can be particularly useful in analyzing viral phosphopeptides from cell culture supernatants that often contain high concentrations of fetal bovine serum and supplements. We newly identified an NP phosphorylation site at Ser79, which is important for Pin1 binding. Furthermore, we showed that the interaction between Pin1 and phosphorylated NP could enhance viral replication in a cell culture model.
Collapse
Affiliation(s)
- Yoko Ino
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Graduate School of Health Sciences, Gunma Paz University, Tonyamachi 1-7-1, Takasaki-shi, Gunma 370-0006, Japan
| | - Mayuko Nishi
- Department of Microbiology, School of Medicine, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Yutaro Yamaoka
- Department of Microbiology, School of Medicine, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Life Science Laboratory, Technology and Development Division, Kanto Chemical Co., Inc., Suzukawa 21, Isehara-shi, Kanagawa 259-1146, Japan
| | - Kei Miyakawa
- Department of Microbiology, School of Medicine, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Sundararaj Stanleyraj Jeremiah
- Department of Microbiology, School of Medicine, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Makoto Osada
- Graduate School of Health Sciences, Gunma Paz University, Tonyamachi 1-7-1, Takasaki-shi, Gunma 370-0006, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Akihide Ryo
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Department of Microbiology, School of Medicine, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan.
| |
Collapse
|
272
|
Wade H, Duan Q, Su Q. Interaction between Sars-CoV-2 structural proteins and host cellular receptors: From basic mechanisms to clinical perspectives. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:243-277. [PMID: 36088078 PMCID: PMC9182089 DOI: 10.1016/bs.apcsb.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (Sars-CoV-2) has caused a global pandemic that has affected the lives of billions of individuals. Sars-CoV-2 primarily infects human cells by binding of the viral spike protein to angiotensin-converting enzyme 2 (ACE2). In addition, novel means of viral entry are currently being investigated, including Neuropillin 1, toll-like receptors (TLRs), cluster of differentiation 147 (CD147), and integrin α5β1. Enriched expression of these proteins across metabolic regulatory organs/tissues, including the circulatory system, liver, pancreas, and intestine contributes to major clinical complications among COVID-19 patients, particularly the development of hypertension, myocardial injury, arrhythmia, acute coronary syndrome and increased coagulation in the circulatory system during and post-infection. Pre-existing metabolic disease, such as cardiovascular disease, obesity, diabetes, and non-alcoholic fatty liver disease, is associated with increased risk of hospitalization, persistent post-infection complications and worse outcomes in patients with COVID-19. This review overviews the biological features of Sars-CoV-2, highlights recent findings that delineate the pathological mechanisms of COVID-19 and the consequent clinical diseases.
Collapse
|
273
|
Qu N, Ying Y, Qin J, Chen AK. Rational design of self-assembled RNA nanostructures for HIV-1 virus assembly blockade. Nucleic Acids Res 2021; 50:e44. [PMID: 34967412 PMCID: PMC9071489 DOI: 10.1093/nar/gkab1282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/16/2021] [Accepted: 12/15/2021] [Indexed: 11/14/2022] Open
Abstract
Many pathological processes are driven by RNA-protein interactions, making such interactions promising targets for molecular interventions. HIV-1 assembly is one such process, in which the viral genomic RNA interacts with the viral Gag protein and serves as a scaffold to drive Gag multimerization that ultimately leads to formation of a virus particle. Here, we develop self-assembled RNA nanostructures that can inhibit HIV-1 virus assembly, achieved through hybridization of multiple artificial small RNAs with a stem-loop structure (STL) that we identify as a prominent ligand of Gag that can inhibit virus particle production via STL-Gag interactions. The resulting STL-decorated nanostructures (double and triple stem-loop structures denoted as Dumbbell and Tribell, respectively) can elicit more pronounced viral blockade than their building blocks, with the inhibition arising as a result of nanostructures interfering with Gag multimerization. These findings could open up new avenues for RNA-based therapy.
Collapse
Affiliation(s)
- Na Qu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Yachen Ying
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.,Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China.,National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Jinshan Qin
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.,Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China.,National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Antony K Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.,National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
274
|
Syed AM, Taha TY, Tabata T, Chen IP, Ciling A, Khalid MM, Sreekumar B, Chen PY, Hayashi JM, Soczek KM, Ott M, Doudna JA. Rapid assessment of SARS-CoV-2-evolved variants using virus-like particles. Science 2021; 374:1626-1632. [PMID: 34735219 PMCID: PMC9005165 DOI: 10.1126/science.abl6184] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/29/2021] [Indexed: 01/16/2023]
Abstract
Efforts to determine why new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants demonstrate improved fitness have been limited to analyzing mutations in the spike (S) protein with the use of S-pseudotyped particles. In this study, we show that SARS-CoV-2 virus-like particles (SC2-VLPs) can package and deliver exogenous transcripts, enabling analysis of mutations within all structural proteins and at multiple steps in the viral life cycle. In SC2-VLPs, four nucleocapsid (N) mutations found universally in more-transmissible variants independently increased messenger RNA delivery and expression ~10-fold, and in a reverse genetics model, the serine-202→arginine (S202R) and arginine-203→methionine (R203M) mutations each produced >50 times as much virus. SC2-VLPs provide a platform for rapid testing of viral variants outside of a biosafety level 3 setting and demonstrate N mutations and particle assembly to be mechanisms that could explain the increased spread of variants, including B.1.617.2 (Delta, which contains the R203M mutation).
Collapse
Affiliation(s)
- Abdullah M. Syed
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Taha Y. Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
| | - Takako Tabata
- Gladstone Institute of Virology, San Francisco, CA, USA
| | - Irene P. Chen
- Gladstone Institute of Virology, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Alison Ciling
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Mir M. Khalid
- Gladstone Institute of Virology, San Francisco, CA, USA
| | | | - Pei-Yi Chen
- Gladstone Institute of Virology, San Francisco, CA, USA
| | | | - Katarzyna M. Soczek
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Melanie Ott
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- Gladstone Institute of Virology, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, CA, USA
| | - Jennifer A. Doudna
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| |
Collapse
|
275
|
Lesbon JCC, Poleti MD, de Mattos Oliveira EC, Patané JSL, Clemente LG, Viala VL, Ribeiro G, Giovanetti M, de Alcantara LCJ, Teixeira O, Nonato MC, de Lima LPO, Martins AJ, dos Santos Barros CR, Marqueze EC, de Souza Todão Bernardino J, Moretti DB, Brassaloti RA, de Lello Rocha Campos Cassano R, Mariani PDSC, Slavov SN, dos Santos RB, Rodrigues ES, Santos EV, Borges JS, de La Roque DGL, Kitajima JP, Santos B, Assato PA, da Silva da Costa FA, Banho CA, Sacchetto L, Moraes MM, Palmieri M, da Silva FEV, Grotto RMT, Souza-Neto JA, Nogueira ML, Coutinho LL, Calado RT, Neto RM, Covas DT, Kashima S, Elias MC, Sampaio SC, Fukumasu H. Nucleocapsid (N) Gene Mutations of SARS-CoV-2 Can Affect Real-Time RT-PCR Diagnostic and Impact False-Negative Results. Viruses 2021; 13:v13122474. [PMID: 34960743 PMCID: PMC8707239 DOI: 10.3390/v13122474] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 01/17/2023] Open
Abstract
The current COVID-19 pandemic demands massive testing by Real-time RT-PCR (Reverse Transcription Polymerase Chain Reaction), which is considered the gold standard diagnostic test for the detection of the SARS-CoV-2 virus. However, the virus continues to evolve with mutations that lead to phenotypic alterations as higher transmissibility, pathogenicity or vaccine evasion. Another big issue are mutations in the annealing sites of primers and probes of RT-PCR diagnostic kits leading to false-negative results. Therefore, here we identify mutations in the N (Nucleocapsid) gene that affects the use of the GeneFinder COVID-19 Plus RealAmp Kit. We sequenced SARS-CoV-2 genomes from 17 positive samples with no N gene detection but with RDRP (RNA-dependent RNA polymerase) and E (Envelope) genes detection, and observed a set of three different mutations affecting the N detection: a deletion of 18 nucleotides (Del28877-28894), a substitution of GGG to AAC (28881-28883) and a frameshift mutation caused by deletion (Del28877-28878). The last one cause a deletion of six AAs (amino acids) located in the central intrinsic disorder region at protein level. We also found this mutation in 99 of the 14,346 sequenced samples by the Sao Paulo state Network for Pandemic Alert of Emerging SARS-CoV-2 variants, demonstrating the circulation of the mutation in Sao Paulo, Brazil. Continuous monitoring and characterization of mutations affecting the annealing sites of primers and probes by genomic surveillance programs are necessary to maintain the effectiveness of the diagnosis of COVID-19.
Collapse
Affiliation(s)
- Jéssika Cristina Chagas Lesbon
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, São Paulo, Brazil; (J.C.C.L.); (M.D.P.); (E.C.d.M.O.)
| | - Mirele Daiana Poleti
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, São Paulo, Brazil; (J.C.C.L.); (M.D.P.); (E.C.d.M.O.)
| | - Elisângela Chicaroni de Mattos Oliveira
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, São Paulo, Brazil; (J.C.C.L.); (M.D.P.); (E.C.d.M.O.)
| | - José Salvatore Leister Patané
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Luan Gaspar Clemente
- Functional Genomic Center, Department of Animal Science, Luiz de Queiroz School of Agriculture, University of Sao Paulo, Piracicaba 13418-900, São Paulo, Brazil; (L.G.C.); (R.A.B.); (R.d.L.R.C.C.); (L.L.C.)
| | - Vincent Louis Viala
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Gabriela Ribeiro
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Marta Giovanetti
- Fundação Oswaldo Cruz, FIOCRUZ, Manguinhos 21040-900, Rio de Janeiro, Brazil; (M.G.); (L.C.J.d.A.)
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | | | - Olivia Teixeira
- Ribeirao Preto Protein Crystallography Laboratory, School of Pharmaceutical Sciences, University of São Paulo, Ribeirao Preto 14040-903, São Paulo, Brazil; (O.T.); (M.C.N.)
| | - Maria Cristina Nonato
- Ribeirao Preto Protein Crystallography Laboratory, School of Pharmaceutical Sciences, University of São Paulo, Ribeirao Preto 14040-903, São Paulo, Brazil; (O.T.); (M.C.N.)
| | - Loyze Paola Oliveira de Lima
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Antonio Jorge Martins
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Claudia Renata dos Santos Barros
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Elaine Cristina Marqueze
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Jardelina de Souza Todão Bernardino
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Debora Botequio Moretti
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Ricardo Augusto Brassaloti
- Functional Genomic Center, Department of Animal Science, Luiz de Queiroz School of Agriculture, University of Sao Paulo, Piracicaba 13418-900, São Paulo, Brazil; (L.G.C.); (R.A.B.); (R.d.L.R.C.C.); (L.L.C.)
| | - Raquel de Lello Rocha Campos Cassano
- Functional Genomic Center, Department of Animal Science, Luiz de Queiroz School of Agriculture, University of Sao Paulo, Piracicaba 13418-900, São Paulo, Brazil; (L.G.C.); (R.A.B.); (R.d.L.R.C.C.); (L.L.C.)
| | | | - Svetoslav Nanev Slavov
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Rafael Bezerra dos Santos
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Evandra Strazza Rodrigues
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Elaine Vieira Santos
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Josiane Serrano Borges
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Debora Glenda Lima de La Roque
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Joao Paulo Kitajima
- Mendelics Genomic Analysis, São Paulo 02511-000, São Paulo, Brazil; (J.P.K.); (B.S.)
| | - Bibiana Santos
- Mendelics Genomic Analysis, São Paulo 02511-000, São Paulo, Brazil; (J.P.K.); (B.S.)
| | - Patricia Akemi Assato
- School of Agricultural Sciences, São Paulo State University, Botucatu 18618-970, São Paulo, Brazil; (P.A.A.); (F.A.d.S.d.C.); (R.M.T.G.); (J.A.S.-N.)
| | - Felipe Allan da Silva da Costa
- School of Agricultural Sciences, São Paulo State University, Botucatu 18618-970, São Paulo, Brazil; (P.A.A.); (F.A.d.S.d.C.); (R.M.T.G.); (J.A.S.-N.)
| | - Cecilia Artico Banho
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto 15090-000, São Paulo, Brazil; (C.A.B.); (L.S.); (M.M.M.); (M.L.N.)
| | - Livia Sacchetto
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto 15090-000, São Paulo, Brazil; (C.A.B.); (L.S.); (M.M.M.); (M.L.N.)
| | - Marilia Mazzi Moraes
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto 15090-000, São Paulo, Brazil; (C.A.B.); (L.S.); (M.M.M.); (M.L.N.)
| | - Melissa Palmieri
- Coordenação de Vigilância em Saúde—Secretaria Municipal da Saúde, São Paulo 01223-906, São Paulo, Brazil;
| | | | - Rejane Maria Tommasini Grotto
- School of Agricultural Sciences, São Paulo State University, Botucatu 18618-970, São Paulo, Brazil; (P.A.A.); (F.A.d.S.d.C.); (R.M.T.G.); (J.A.S.-N.)
| | - Jayme A. Souza-Neto
- School of Agricultural Sciences, São Paulo State University, Botucatu 18618-970, São Paulo, Brazil; (P.A.A.); (F.A.d.S.d.C.); (R.M.T.G.); (J.A.S.-N.)
| | - Mauricio Lacerda Nogueira
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto 15090-000, São Paulo, Brazil; (C.A.B.); (L.S.); (M.M.M.); (M.L.N.)
| | - Luiz Lehman Coutinho
- Functional Genomic Center, Department of Animal Science, Luiz de Queiroz School of Agriculture, University of Sao Paulo, Piracicaba 13418-900, São Paulo, Brazil; (L.G.C.); (R.A.B.); (R.d.L.R.C.C.); (L.L.C.)
| | - Rodrigo Tocantins Calado
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Raul Machado Neto
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Dimas Tadeu Covas
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Simone Kashima
- Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, São Paulo, Brazil; (S.N.S.); (R.B.d.S.); (E.S.R.); (E.V.S.); (J.S.B.); (D.G.L.d.L.R.); (R.T.C.); (S.K.)
| | - Maria Carolina Elias
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Sandra Coccuzzo Sampaio
- Butantan Institute, São Paulo 05503-000, São Paulo, Brazil; (J.S.L.P.); (V.L.V.); (G.R.); (L.P.O.d.L.); (A.J.M.); (C.R.d.S.B.); (E.C.M.); (J.d.S.T.B.); (D.B.M.); (R.M.N.); (D.T.C.); (M.C.E.); (S.C.S.)
| | - Heidge Fukumasu
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, São Paulo, Brazil; (J.C.C.L.); (M.D.P.); (E.C.d.M.O.)
- Correspondence: ; Tel.: +55-19-35656864
| |
Collapse
|
276
|
Hayashi M, Schultz EP, Lanchy JM, Lodmell JS. Time-Resolved Analysis of N-RNA Interactions during RVFV Infection Shows Qualitative and Quantitative Shifts in RNA Encapsidation and Packaging. Viruses 2021; 13:2417. [PMID: 34960686 PMCID: PMC8704896 DOI: 10.3390/v13122417] [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: 10/15/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a negative-sense, tripartite RNA virus that is endemic to Africa and the Arabian Peninsula. It can cause severe disease and mortality in humans and domestic livestock and is a concern for its potential to spread more globally. RVFV's nucleocapsid protein (N) is an RNA-binding protein that is necessary for viral transcription, replication, and the production of nascent viral particles. We have conducted crosslinking, immunoprecipitation, and sequencing (CLIP-seq) to characterize N interactions with host and viral RNAs during infection. In parallel, to precisely measure intracellular N levels, we employed multiple reaction monitoring mass spectrometry (MRM-MS). Our results show that N binds mostly to host RNAs at early stages of infection, yielding nascent virus particles of reduced infectivity. The expression of N plateaus 10 h post-infection, whereas the intracellular viral RNA concentration continues to increase. Moreover, the virions produced later in infection have higher infectivity. Taken together, the detailed examination of these N-RNA interactions provides insight into how the regulated expression of N and viral RNA produces both infectious and incomplete, noninfectious particles.
Collapse
Affiliation(s)
- Miyuki Hayashi
- Department of Chemistry and Biochemistry, University of Montana, Missoula, MT 59812, USA;
- Center for Biomolecular Structure and Dynamics, Missoula, MT 59812, USA;
| | - Eric P. Schultz
- Center for Biomolecular Structure and Dynamics, Missoula, MT 59812, USA;
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA;
| | - Jean-Marc Lanchy
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA;
| | - J. Stephen Lodmell
- Center for Biomolecular Structure and Dynamics, Missoula, MT 59812, USA;
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA;
| |
Collapse
|
277
|
Forchette L, Sebastian W, Liu T. A Comprehensive Review of COVID-19 Virology, Vaccines, Variants, and Therapeutics. Curr Med Sci 2021; 41:1037-1051. [PMID: 34241776 PMCID: PMC8267225 DOI: 10.1007/s11596-021-2395-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative pathogen of the coronavirus disease 2019 (COVID-19), has caused more than 179 million infections and 3.8 million deaths worldwide. Throughout the past year, multiple vaccines have already been developed and used, while some others are in the process of being developed. However, the emergence of new mutant strains of SARS-CoV-2 that have demonstrated immune-evading characteristics and an increase in infective capabilities leads to potential ineffectiveness of the vaccines against these variants. The purpose of this review article is to highlight the current understanding of the immunological mechanisms of the virus and vaccines, as well as to investigate some key variants and mutations of the virus driving the current pandemic and their impacts on current management guidelines. We also discussed new technologies being developed for the prevention, treatment, and detection of SARS-CoV-2. In this paper, we thoroughly reviewed and provided crucial information on SARS-CoV-2 virology, vaccines and drugs being used and developed for its prevention and treatment, as well as important variant strains. Our review paper will be beneficial to health care professionals and researchers so they can have a better understanding of the basic sciences, prevention, and clinical treatment of COVID-19 during the pandemic. This paper consists of the most updated information that has been available as of June 21, 2021.
Collapse
Affiliation(s)
- Lauren Forchette
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, 400 Lee Street North, Lewisburg, West Virginia 24901 USA
| | - William Sebastian
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, 400 Lee Street North, Lewisburg, West Virginia 24901 USA
| | - Tuoen Liu
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, 400 Lee Street North, Lewisburg, West Virginia 24901 USA
| |
Collapse
|
278
|
Hirai Y, Tomonaga K, Horie M. Borna disease virus phosphoprotein triggers the organization of viral inclusion bodies by liquid-liquid phase separation. Int J Biol Macromol 2021; 192:55-63. [PMID: 34606793 DOI: 10.1016/j.ijbiomac.2021.09.153] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/02/2021] [Accepted: 09/20/2021] [Indexed: 12/19/2022]
Abstract
Inclusion bodies (IBs) are characteristic biomolecular condensates organized by the non-segmented negative-strand RNA viruses belonging to the order Mononegavirales. Although recent studies have revealed the characteristics of IBs formed by cytoplasmic mononegaviruses, that of Borna disease virus 1 (BoDV-1), a unique mononegavirus that forms IBs in the cell nucleus and establishes persistent infection remains elusive. Here, we characterize the IBs of BoDV-1 in terms of liquid-liquid phase separation (LLPS). The BoDV-1 phosphoprotein (P) alone induces LLPS and the nucleoprotein (N) is incorporated into the P droplets in vitro. In contrast, co-expression of N and P is required for the formation of IB-like structure in cells. Furthermore, while BoDV-1 P binds to RNA, an excess amount of RNA dissolves the liquid droplets formed by N and P in vitro. Notably, the intrinsically disordered N-terminal region of BoDV-1 P is essential to drive LLPS and to bind to RNA, suggesting that both abilities could compete with one another. These features are unique among mononegaviruses, and thus this study will contribute to a deeper understanding of LLPS-driven organization and RNA-mediated regulation of biomolecular condensates.
Collapse
Affiliation(s)
- Yuya Hirai
- Department of Biology, Osaka Dental University, 8-1, Kuzuha Hanazono-cho, Hirakata, Osaka 573-1121, Japan.
| | - Keizo Tomonaga
- Laboratory of RNA viruses, Department of Virus Research, Institute for Frontier Life and Medical Sciences (InFRONT), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Laboratory of RNA Viruses, Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, 606-8507 Kyoto, Japan; Department of Molecular Virology, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, 606-8507 Kyoto, Japan.
| | - Masayuki Horie
- Laboratory of RNA viruses, Department of Virus Research, Institute for Frontier Life and Medical Sciences (InFRONT), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Hakubi Center for Advanced Research, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Laboratory of Veterinary Microbiology, Division of Veterinary Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-oraikita, Izumisano, Osaka 598-8531, Japan.
| |
Collapse
|
279
|
Srinivasan K, Pandey AK, Livingston A, Venkatesh S. Roles of host mitochondria in the development of COVID-19 pathology: Could mitochondria be a potential therapeutic target? MOLECULAR BIOMEDICINE 2021; 2:38. [PMID: 34841263 PMCID: PMC8608434 DOI: 10.1186/s43556-021-00060-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
The recent emergence of severe acute respiratory syndrome-Corona Virus 2 (SARS-CoV-2) in late 2019 and its spread worldwide caused an acute pandemic of Coronavirus disease 19 (COVID-19). Since then, COVID-19 has been under intense scrutiny as its outbreak led to significant changes in healthcare, social activities, and economic settings worldwide. Although angiotensin-converting enzyme-2 (ACE-2) receptor is shown to be the primary port of SARS-CoV-2 entry in cells, the mechanisms behind the establishment and pathologies of COVID-19 are poorly understood. As recent studies have shown that host mitochondria play an essential role in virus-mediated innate immune response, pathologies, and infection, in this review, we will discuss in detail the entry and progression of SARS-CoV-2 and how mitochondria could play roles in COVID-19 disease. We will also review the potential interactions between SARS-CoV-2 and mitochondria and discuss possible treatments, including whether mitochondria as a potential therapeutic target in COVID-19. Understanding SARS-CoV-2 and mitochondrial interactions mediated virus establishment, inflammation, and other consequences may provide a unique mechanism and conceptual advancement in finding a novel treatment for COVID-19.
Collapse
Affiliation(s)
- Kavya Srinivasan
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers -New Jersey Medical School, The State University of New Jersey, Newark, NJ USA
- New York Institute of Technology, Old Westbury, NY USA
| | - Ashutosh Kumar Pandey
- Department of Pharmacology, Physiology and Neuroscience, Rutgers -New Jersey Medical School, The State University of New Jersey, Newark, NJ USA
| | | | - Sundararajan Venkatesh
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers -New Jersey Medical School, The State University of New Jersey, Newark, NJ USA
| |
Collapse
|
280
|
Singla RK, He X, Chopra H, Tsagkaris C, Shen L, Kamal MA, Shen B. Natural Products for the Prevention and Control of the COVID-19 Pandemic: Sustainable Bioresources. Front Pharmacol 2021; 12:758159. [PMID: 34925017 PMCID: PMC8671886 DOI: 10.3389/fphar.2021.758159] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/27/2021] [Indexed: 02/05/2023] Open
Abstract
Background: The world has been unprecedentedly hit by a global pandemic which broke the record of deadly pandemics that faced humanity ever since its existence. Even kids are well-versed in the terminologies and basics of the SARS-CoV-2 virus and COVID-19 now. The vaccination program has been successfully launched in various countries, given that the huge global population of concern is still far behind to be vaccinated. Furthermore, the scarcity of any potential drug against the COVID-19-causing virus forces scientists and clinicians to search for alternative and complementary medicines on a war-footing basis. Aims and Objectives: The present review aims to cover and analyze the etiology and epidemiology of COVID-19, the role of intestinal microbiota and pro-inflammatory markers, and most importantly, the natural products to combat this deadly SARS-CoV-2 virus. Methods: A primary literature search was conducted through PubMed and Google Scholar using relevant keywords. Natural products were searched from January 2020 to November 2020. No timeline limit has been imposed on the search for the biological sources of those phytochemicals. Interactive mapping has been done to analyze the multi-modal and multi-target sources. Results and Discussion: The intestinal microbiota and the pro-inflammatory markers that can serve the prognosis, diagnosis, and treatment of COVID-19 were discussed. The literature search resulted in yielding 70 phytochemicals and ten polyherbal formulations which were scientifically analyzed against the SARS-CoV-2 virus and its targets and found significant. Retrospective analyses led to provide information about 165 biological sources that can also be screened if not done earlier. Conclusion: The interactive analysis mapping of biological sources with phytochemicals and targets as well as that of phytochemical class with phytochemicals and COVID-19 targets yielded insights into the multitarget and multimodal evidence-based complementary medicines.
Collapse
Affiliation(s)
- Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Xuefei He
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, India
| | | | - Li Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Mohammad Amjad Kamal
- West China School of Nursing/Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Enzymoics; Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
281
|
Monoclonal Antibodies against Nucleocapsid Protein of SARS-CoV-2 Variants for Detection of COVID-19. Int J Mol Sci 2021; 22:ijms222212412. [PMID: 34830291 PMCID: PMC8623253 DOI: 10.3390/ijms222212412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/13/2021] [Accepted: 11/14/2021] [Indexed: 02/04/2023] Open
Abstract
Mitigation strategies of the coronavirus disease 2019 (COVID-19) pandemic have been greatly hindered by the continuous emergence of SARS-CoV-2 variants. New sensitive, rapid diagnostic tests for the wide-spectrum detection of viral variants are needed. We generated a panel of 41 monoclonal antibodies against the SARS-CoV-2 nucleocapsid protein (NP) by using mice hybridoma techniques. Of these mAbs, nine exhibited high binding activities and were applied in latex-based lateral flow immunoassays (LFIAs). The LFIAs utilizing NP-mAb-7 and -40 had the best sensitivity and lowest limit of detection: 8 pg for purified NP and 625 TCID50/mL for the authentic virus (hCoV-19/Taiwan/4/2020). The specificity tests showed that the NP-mAb-40/7 LFIA strips did not cross-react with five human coronavirus strains or 20 other common respiratory pathogens. Importantly, we found that 10 NP mutants, including alpha (B.1.1.7), beta (B.1.351), gamma (P.1), and delta (B.1.617.2) variants, could be detected by NP-mAb-40/7 LFIA strips. A clinical study (n = 60) of the NP-mAb-40/7 LFIA strips demonstrated a specificity of 100% and sensitivity of 90% in infected individuals with cycle threshold (Ct) values < 29.5. These anti-NP mAbs have strong potential for use in the clinical detection of SARS-CoV-2 infection, whether the virus is wild-type or a variant of concern.
Collapse
|
282
|
Somasekharan SP, Gleave M. SARS-CoV-2 nucleocapsid protein interacts with immunoregulators and stress granules and phase separates to form liquid droplets. FEBS Lett 2021; 595:2872-2896. [PMID: 34780058 PMCID: PMC8652540 DOI: 10.1002/1873-3468.14229] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/16/2022]
Abstract
The current work investigated SARS‐CoV‐2 Nucleocapsid (NCAP or N protein) interactors in A549 human lung cancer cells using a SILAC‐based mass spectrometry approach. NCAP interactors included proteins of the stress granule (SG) machinery and immunoregulators. NCAP showed specific interaction with the SG proteins G3BP1, G3BP2, YTHDF3, USP10 and PKR, and translocated to SGs following oxidative stress and heat shock. Treatment of recombinant NCAP with RNA isolated from A549 cells exposed to oxidative stress‐stimulated NCAP to undergo liquid–liquid phase separation (LLPS). RNA degradation using RNase A treatment completely blocked the LLPS property of NCAP as well as its SG association. The RNA intercalator mitoxantrone also disrupted NCAP assembly in vitro and in cells. This study provides insight into the biological processes and biophysical properties of the SARS‐CoV‐2 NCAP.
Collapse
Affiliation(s)
- Syam Prakash Somasekharan
- Department of Urologic Sciences, Faculty of Medicine, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- Department of Urologic Sciences, Faculty of Medicine, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
283
|
Al-Karmalawy AA, Soltane R, Abo Elmaaty A, Tantawy MA, Antar SA, Yahya G, Chrouda A, Pashameah RA, Mustafa M, Abu Mraheil M, Mostafa A. Coronavirus Disease (COVID-19) Control between Drug Repurposing and Vaccination: A Comprehensive Overview. Vaccines (Basel) 2021; 9:1317. [PMID: 34835248 PMCID: PMC8622998 DOI: 10.3390/vaccines9111317] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023] Open
Abstract
Respiratory viruses represent a major public health concern, as they are highly mutated, resulting in new strains emerging with high pathogenicity. Currently, the world is suffering from the newly evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This virus is the cause of coronavirus disease 2019 (COVID-19), a mild-to-severe respiratory tract infection with frequent ability to give rise to fatal pneumonia in humans. The overwhelming outbreak of SARS-CoV-2 continues to unfold all over the world, urging scientists to put an end to this global pandemic through biological and pharmaceutical interventions. Currently, there is no specific treatment option that is capable of COVID-19 pandemic eradication, so several repurposed drugs and newly conditionally approved vaccines are in use and heavily applied to control the COVID-19 pandemic. The emergence of new variants of the virus that partially or totally escape from the immune response elicited by the approved vaccines requires continuous monitoring of the emerging variants to update the content of the developed vaccines or modify them totally to match the new variants. Herein, we discuss the potential therapeutic and prophylactic interventions including repurposed drugs and the newly developed/approved vaccines, highlighting the impact of virus evolution on the immune evasion of the virus from currently licensed vaccines for COVID-19.
Collapse
Affiliation(s)
- Ahmed A Al-Karmalawy
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Raya Soltane
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
- Department of Biology, Faculty of Sciences, Tunis El Manar University, Tunis 1068, Tunisia
| | - Ayman Abo Elmaaty
- Department of Medicinal Chemistry, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt
| | - Mohamed A Tantawy
- Hormones Department, Medical Research and Clinical Studies Research Institute, National Research Centre, Dokki 12622, Egypt
- Stem Cells Laboratory, Center of Excellence for Advanced Sciences, National Research Centre, Dokki 12622, Egypt
| | - Samar A Antar
- Department of Pharmacology, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Galal Yahya
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Amani Chrouda
- Department of Chemistry, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11932, Saudi Arabia
- Laboratory of Interfaces and Advanced Materials, Faculty of Sciences, Monastir University, Monastir 5000, Tunisia
- Institute of Analytical Sciences, UMR CNRS-UCBL-ENS 5280, 5 Rue la Doua, CEDEX, 69100 Villeurbanne, France
| | - Rami Adel Pashameah
- Department of Basic Sciences, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Muhamad Mustafa
- Department of Medicinal Chemistry, Deraya University, Minia 61111, Egypt
| | - Mobarak Abu Mraheil
- German Center for Infection Research (DZIF), Institute of Medical Microbiology, Justus-Liebig University, 35392 Giessen, Germany
| | - Ahmed Mostafa
- German Center for Infection Research (DZIF), Institute of Medical Microbiology, Justus-Liebig University, 35392 Giessen, Germany
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Dokki 12622, Egypt
| |
Collapse
|
284
|
Insights into the SARS-CoV-2-Mediated Alteration in the Stress Granule Protein Regulatory Networks in Humans. Pathogens 2021; 10:pathogens10111459. [PMID: 34832615 PMCID: PMC8624858 DOI: 10.3390/pathogens10111459] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 12/15/2022] Open
Abstract
The rapidly and constantly evolving coronavirus, SARS-CoV-2, imposes a great threat to human health causing severe lung disease and significant mortality. Cytoplasmic stress granules (SGs) exert anti-viral activities due to their involvement in translation inhibition and innate immune signaling. SARS-CoV-2 sequesters important SG nucleator proteins and impairs SG formation, thus evading the host response for efficient viral replication. However, the significance of SGs in COVID-19 infection remains elusive. In this study, we utilize a protein-protein interaction network approach to systematically dissect the crosstalk of human post-translational regulatory networks governed by SG proteins due to SARS-CoV-2 infection. We uncovered that 116 human SG proteins directly interact with SARS-CoV-2 proteins and are involved in 430 different brain disorders including COVID-19. Further, we performed gene set enrichment analysis to identify the drugs against three important key SG proteins (DYNC1H1, DCTN1, and LMNA) and also looked for potential microRNAs (miRNAs) targeting these proteins. We identified bexarotene as a potential drug molecule and miRNAs, hsa-miR-615-3p, hsa-miR-221-3p, and hsa-miR-124-3p as potential candidates for the treatment of COVID-19 and associated manifestations.
Collapse
|
285
|
Sankaranarayanan M, Emenecker RJ, Wilby EL, Jahnel M, Trussina IREA, Wayland M, Alberti S, Holehouse AS, Weil TT. Adaptable P body physical states differentially regulate bicoid mRNA storage during early Drosophila development. Dev Cell 2021; 56:2886-2901.e6. [PMID: 34655524 PMCID: PMC8555633 DOI: 10.1016/j.devcel.2021.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/22/2021] [Accepted: 09/23/2021] [Indexed: 01/01/2023]
Abstract
Ribonucleoprotein condensates can exhibit diverse physical states in vitro and in vivo. Despite considerable progress, the relevance of condensate physical states for in vivo biological function remains limited. Here, we investigated the physical properties of processing bodies (P bodies) and their impact on mRNA storage in mature Drosophila oocytes. We show that the conserved DEAD-box RNA helicase Me31B forms viscous P body condensates, which adopt an arrested physical state. We demonstrate that structurally distinct proteins and protein-protein interactions, together with RNA, regulate the physical properties of P bodies. Using live imaging and in situ hybridization, we show that the arrested state and integrity of P bodies support the storage of bicoid (bcd) mRNA and that egg activation modulates P body properties, leading to the release of bcd for translation in the early embryo. Together, this work provides an example of how physical states of condensates regulate cellular function in development.
Collapse
Affiliation(s)
- M Sankaranarayanan
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| | - Ryan J Emenecker
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Center for Science and Engineering of Living Systems, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA
| | - Elise L Wilby
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Marcus Jahnel
- Center for Molecular and Cellular Bioengineering, Biotechnology Center, Technische Universität Dresden, Tatzberg 47/49, 01307 Dresden, Germany
| | - Irmela R E A Trussina
- Center for Molecular and Cellular Bioengineering, Biotechnology Center, Technische Universität Dresden, Tatzberg 47/49, 01307 Dresden, Germany
| | - Matt Wayland
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Simon Alberti
- Center for Molecular and Cellular Bioengineering, Biotechnology Center, Technische Universität Dresden, Tatzberg 47/49, 01307 Dresden, Germany
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Center for Science and Engineering of Living Systems, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA
| | - Timothy T Weil
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.
| |
Collapse
|
286
|
Jung HE, Lee HK. Current Understanding of the Innate Control of Toll-like Receptors in Response to SARS-CoV-2 Infection. Viruses 2021; 13:2132. [PMID: 34834939 PMCID: PMC8622567 DOI: 10.3390/v13112132] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection, threatens the entire world. It has affected every aspect of life and increased the burden on both healthcare and socioeconomic systems. Current studies have revealed that excessive inflammatory immune responses are responsible for the severity of COVID-19, which suggests that anti-inflammatory drugs may be promising therapeutic treatments. However, there are currently a limited number of approved therapeutics for COVID-19. Toll-like receptors (TLRs), which recognize microbial components derived from invading pathogens, are involved in both the initiation of innate responses against SARS-CoV-2 infection and the hyperinflammatory phenotype of COVID-19. In this review, we provide current knowledge on the pivotal role of TLRs in immune responses against SARS-CoV-2 infection and demonstrate the potential effectiveness of TLR-targeting drugs on the control of hyperinflammation in patients with COVID-19.
Collapse
Affiliation(s)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea;
| |
Collapse
|
287
|
Montalvo Villalba MC, Sosa Glaria E, Rodriguez Lay LDLA, Valdés Ramirez O, Vallina García D, Arencibia Garcia A, Martinez Alfonso J, Menes Llerena DM, Torres Pérez L, Resik Aguirre SR, Guzman Tirado MG. Performance evaluation of Elecsys SARS-CoV-2 Antigen immunoassay for diagnostic of COVID-19. J Med Virol 2021; 94:1001-1008. [PMID: 34676585 PMCID: PMC8662245 DOI: 10.1002/jmv.27412] [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: 06/04/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022]
Abstract
One of the challenges for control and prevention of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is the early diagnostic at the point of care. Several tests based on qualitative antigen detection have been developed; one of these is Elecsys SARS-CoV-2 Antigen immunoassay (Roche Diagnostics). In total, 523 nasopharyngeal swabs were randomly selected with the aims to evaluate sensitivity, specificity, cross-reactivity, positive and negative predictive value (PPV, NPV), and agreement of Elecsys SARS-CoV-2 Antigen immunoassay using reverse transcription-polymerase chain reaction (RT-PCR) STAT-NAT® coronavirus disease-2019 as reference test. Cross-reactivity was estimated using samples positive by RT-PCR to other respiratory viruses (influenza virus, parainfluenza virus, rhinovirus, coronavirus OC43, and HKU1). The overall sensitivity of Elecsys SARS-CoV-2 Antigen was 89.72% (288/321); specificity was 90.59% (183/202); and cross-reactivity to other respiratory viruses were not detected. Elecsys SARS-CoV-2 Antigen immunoassay showed a high sensitivity in samples with cycle threshold value <30, which ranged from 92.81% to 95.40%, independently of symptoms. PPV and NPV were 93.81% and 84.72%, respectively. The κ coefficient was 0.79 (95% confidence interval: 0.73-0.84), showing substantial agreement between both tests. The results suggest Elecsys SARS-CoV-2 Antigen immunoassay could be used as an alternative to RT-PCR testing, or in complement with it, to identify infectious individuals and reduce SARS-CoV-2 transmission.
Collapse
Affiliation(s)
| | - Elena Sosa Glaria
- Department of Virology, Institute of Tropical Medicine Pedro Kouri, Havana, Cuba
| | | | | | | | | | | | | | - Loida Torres Pérez
- Department of Clincal Laboratory, Health International Centre ¨Las Praderas¨, Siboney, La Habana, Cuba
| | | | | |
Collapse
|
288
|
Pajkos M, Dosztányi Z. Functions of intrinsically disordered proteins through evolutionary lenses. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 183:45-74. [PMID: 34656334 DOI: 10.1016/bs.pmbts.2021.06.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein sequences are the result of an evolutionary process that involves the balancing act of experimenting with novel mutations and selecting out those that have an undesirable functional outcome. In the case of globular proteins, the function relies on a well-defined conformation, therefore, there is a strong evolutionary pressure to preserve the structure. However, different evolutionary rules might apply for the group of intrinsically disordered regions and proteins (IDR/IDPs) that exist as an ensemble of fluctuating conformations. The function of IDRs can directly originate from their disordered state or arise through different types of molecular recognition processes. There is an amazing variety of ways IDRs can carry out their functions, and this is also reflected in their evolutionary properties. In this chapter we give an overview of the different types of evolutionary behavior of disordered proteins and associated functions in normal and disease settings.
Collapse
Affiliation(s)
- Mátyás Pajkos
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsanna Dosztányi
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
289
|
Abstract
Viruses have evolved precise mechanisms for using the cellular physiological pathways for their perpetuation. These virus-driven biochemical events must be separated in space and time from those of the host cell. In recent years, granular structures, known for over a century for rabies virus, were shown to host viral gene function and were named using terms such as viroplasms, replication sites, inclusion bodies, or viral factories (VFs). More recently, these VFs were shown to be liquid-like, sharing properties with membrane-less organelles driven by liquid–liquid phase separation (LLPS) in a process widely referred to as biomolecular condensation. Some of the best described examples of these structures come from negative stranded RNA viruses, where micrometer size VFs are formed toward the end of the infectious cycle. We here discuss some basic principles of LLPS in connection with several examples of VFs and propose a view, which integrates viral replication mechanisms with the biochemistry underlying liquid-like organelles. In this view, viral protein and RNA components gradually accumulate up to a critical point during infection where phase separation is triggered. This yields an increase in transcription that leads in turn to increased translation and a consequent growth of initially formed condensates. According to chemical principles behind phase separation, an increase in the concentration of components increases the size of the condensate. A positive feedback cycle would thus generate in which crucial components, in particular nucleoproteins and viral polymerases, reach their highest levels required for genome replication. Progress in understanding viral biomolecular condensation leads to exploration of novel therapeutics. Furthermore, it provides insights into the fundamentals of phase separation in the regulation of cellular gene function given that virus replication and transcription, in particular those requiring host polymerases, are governed by the same biochemical principles.
Collapse
|
290
|
Queirós-Reis L, Gomes da Silva P, Gonçalves J, Brancale A, Bassetto M, Mesquita JR. SARS-CoV-2 Virus-Host Interaction: Currently Available Structures and Implications of Variant Emergence on Infectivity and Immune Response. Int J Mol Sci 2021; 22:10836. [PMID: 34639178 PMCID: PMC8509653 DOI: 10.3390/ijms221910836] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 01/11/2023] Open
Abstract
Coronavirus disease 19, or COVID-19, is an infection associated with an unprecedented worldwide pandemic caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), which has led to more than 215 million infected people and more than 4.5 million deaths worldwide. SARS-CoV-2 cell infection is initiated by a densely glycosylated spike (S) protein, a fusion protein, binding human angiotensin converting enzyme 2 (hACE2), that acts as the functional receptor through the receptor binding domain (RBD). In this article, the interaction of hACE2 with the RBD and how fusion is initiated after recognition are explored, as well as how mutations influence infectivity and immune response. Thus, we focused on all structures available in the Protein Data Bank for the interaction between SARS-CoV-2 S protein and hACE2. Specifically, the Delta variant carries particular mutations associated with increased viral fitness through decreased antibody binding, increased RBD affinity and altered protein dynamics. Combining both existing mutations and mutagenesis studies, new potential SARS-CoV-2 variants, harboring advantageous S protein mutations, may be predicted. These include mutations S13I and W152C, decreasing antibody binding, N460K, increasing RDB affinity, or Q498R, positively affecting both properties.
Collapse
Affiliation(s)
- Luís Queirós-Reis
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal; (L.Q.-R.); (P.G.d.S.)
| | - Priscilla Gomes da Silva
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal; (L.Q.-R.); (P.G.d.S.)
- Epidemiology Research Unit (EPIunit), Institute of Public Health, University of Porto, 4050-091 Porto, Portugal
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - José Gonçalves
- Institute of Sustainable Processes, University of Valladolid, 47011 Valladolid, Spain;
| | - Andrea Brancale
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, UK;
| | - Marcella Bassetto
- Department of Chemistry, Faculty of Science and Engineering, Swansea University,
Swansea SA2 8PP, UK;
| | - João R. Mesquita
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal; (L.Q.-R.); (P.G.d.S.)
- Epidemiology Research Unit (EPIunit), Institute of Public Health, University of Porto, 4050-091 Porto, Portugal
| |
Collapse
|
291
|
Ryu JK, Hwang DE, Choi JM. Current Understanding of Molecular Phase Separation in Chromosomes. Int J Mol Sci 2021; 22:10736. [PMID: 34639077 PMCID: PMC8509192 DOI: 10.3390/ijms221910736] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
Biomolecular phase separation denotes the demixing of a specific set of intracellular components without membrane encapsulation. Recent studies have found that biomolecular phase separation is involved in a wide range of cellular processes. In particular, phase separation is involved in the formation and regulation of chromosome structures at various levels. Here, we review the current understanding of biomolecular phase separation related to chromosomes. First, we discuss the fundamental principles of phase separation and introduce several examples of nuclear/chromosomal biomolecular assemblies formed by phase separation. We also briefly explain the experimental and computational methods used to study phase separation in chromosomes. Finally, we discuss a recent phase separation model, termed bridging-induced phase separation (BIPS), which can explain the formation of local chromosome structures.
Collapse
Affiliation(s)
- Je-Kyung Ryu
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Da-Eun Hwang
- Department of Chemistry, Pusan National University, Busan 46241, Korea;
| | - Jeong-Mo Choi
- Department of Chemistry, Pusan National University, Busan 46241, Korea;
| |
Collapse
|
292
|
Tarczewska A, Kolonko-Adamska M, Zarębski M, Dobrucki J, Ożyhar A, Greb-Markiewicz B. The method utilized to purify the SARS-CoV-2 N protein can affect its molecular properties. Int J Biol Macromol 2021; 188:391-403. [PMID: 34371045 PMCID: PMC8343380 DOI: 10.1016/j.ijbiomac.2021.08.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/30/2022]
Abstract
One of the main structural proteins of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the nucleocapsid protein (N). The basic function of this protein is to bind genomic RNA and to form a protective nucleocapsid in the mature virion. The intrinsic ability of the N protein to interact with nucleic acids makes its purification very challenging. Therefore, typically employed purification methods appear to be insufficient for removing nucleic acid contamination. In this study, we present a novel purification protocol that enables the N protein to be prepared without any bound nucleic acids. We also performed comparative structural analysis of the N protein contaminated with nucleic acids and free of contamination and showed significant differences in the structural and phase separation properties of the protein. These results indicate that nucleic-acid contamination may severely affect molecular properties of the purified N protein. In addition, the notable ability of the N protein to form condensates whose morphology and behaviour suggest more ordered forms resembling gel-like or solid structures is described.
Collapse
Affiliation(s)
- Aneta Tarczewska
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| | - Marta Kolonko-Adamska
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| | - Mirosław Zarębski
- Department of Cell Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Cracow, Poland
| | - Jurek Dobrucki
- Department of Cell Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Cracow, Poland
| | - Andrzej Ożyhar
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland
| | - Beata Greb-Markiewicz
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland.
| |
Collapse
|
293
|
Spegg V, Altmeyer M. Biomolecular condensates at sites of DNA damage: More than just a phase. DNA Repair (Amst) 2021; 106:103179. [PMID: 34311273 PMCID: PMC7612016 DOI: 10.1016/j.dnarep.2021.103179] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022]
Abstract
Protein recruitment to DNA break sites is an integral part of the DNA damage response (DDR). Elucidation of the hierarchy and temporal order with which DNA damage sensors as well as repair and signaling factors assemble around chromosome breaks has painted a complex picture of tightly regulated macromolecular interactions that build specialized compartments to facilitate repair and maintenance of genome integrity. While many of the underlying interactions, e.g. between repair factors and damage-induced histone marks, can be explained by lock-and-key or induced fit binding models assuming fixed stoichiometries, structurally less well defined interactions, such as the highly dynamic multivalent interactions implicated in phase separation, also participate in the formation of multi-protein assemblies in response to genotoxic stress. Although much remains to be learned about these types of cooperative and highly dynamic interactions and their functional roles, the rapidly growing interest in material properties of biomolecular condensates and in concepts from polymer chemistry and soft matter physics to understand biological processes at different scales holds great promises. Here, we discuss nuclear condensates in the context of genome integrity maintenance, highlighting the cooperative potential between clustered stoichiometric binding and phase separation. Rather than viewing them as opposing scenarios, their combined effects can balance structural specificity with favorable physicochemical properties relevant for the regulation and function of multilayered nuclear condensates.
Collapse
Affiliation(s)
- Vincent Spegg
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
294
|
Jack A, Ferro LS, Trnka MJ, Wehri E, Nadgir A, Nguyenla X, Fox D, Costa K, Stanley S, Schaletzky J, Yildiz A. SARS-CoV-2 nucleocapsid protein forms condensates with viral genomic RNA. PLoS Biol 2021; 19:e3001425. [PMID: 34634033 PMCID: PMC8553124 DOI: 10.1371/journal.pbio.3001425] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 10/28/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection causes Coronavirus Disease 2019 (COVID-19), a pandemic that seriously threatens global health. SARS-CoV-2 propagates by packaging its RNA genome into membrane enclosures in host cells. The packaging of the viral genome into the nascent virion is mediated by the nucleocapsid (N) protein, but the underlying mechanism remains unclear. Here, we show that the N protein forms biomolecular condensates with viral genomic RNA both in vitro and in mammalian cells. While the N protein forms spherical assemblies with homopolymeric RNA substrates that do not form base pairing interactions, it forms asymmetric condensates with viral RNA strands. Cross-linking mass spectrometry (CLMS) identified a region that drives interactions between N proteins in condensates, and deletion of this region disrupts phase separation. We also identified small molecules that alter the size and shape of N protein condensates and inhibit the proliferation of SARS-CoV-2 in infected cells. These results suggest that the N protein may utilize biomolecular condensation to package the SARS-CoV-2 RNA genome into a viral particle.
Collapse
Affiliation(s)
- Amanda Jack
- Biophysics Graduate Group, University of California, Berkeley, California, United States of America
| | - Luke S. Ferro
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Michael J. Trnka
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, United States of America
| | - Eddie Wehri
- Center for Emerging and Neglected Diseases, University of California, Berkeley, California, United States of America
| | - Amrut Nadgir
- Physics Department, University of California, Berkeley, California, United States of America
| | - Xammy Nguyenla
- School of Public Health, Division of Infectious Diseases and Vaccinology, University of California, Berkeley, California, United States of America
| | - Douglas Fox
- School of Public Health, Division of Infectious Diseases and Vaccinology, University of California, Berkeley, California, United States of America
| | - Katelyn Costa
- Press West Illustrations, Boston, Massachusetts, United States of America
| | - Sarah Stanley
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- School of Public Health, Division of Infectious Diseases and Vaccinology, University of California, Berkeley, California, United States of America
| | - Julia Schaletzky
- Center for Emerging and Neglected Diseases, University of California, Berkeley, California, United States of America
| | - Ahmet Yildiz
- Biophysics Graduate Group, University of California, Berkeley, California, United States of America
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- Physics Department, University of California, Berkeley, California, United States of America
| |
Collapse
|
295
|
Taneja I, Holehouse AS. Folded domain charge properties influence the conformational behavior of disordered tails. Curr Res Struct Biol 2021; 3:216-228. [PMID: 34557680 PMCID: PMC8446786 DOI: 10.1016/j.crstbi.2021.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Intrinsically disordered proteins and protein regions (IDRs) make up around 30% of the human proteome where they play essential roles in dictating and regulating many core biological processes. While IDRs are often studied as isolated domains, in naturally occurring proteins most IDRs are found adjacent to folded domains, where they exist as either N- or C-terminal tails or as linkers connecting two folded domains. Prior work has shown that charge properties of IDRs can influence their conformational behavior, both in isolation and in the context of folded domains. In contrast, the converse scenario is less well-explored: how do the charge properties of folded domains influence IDR conformational behavior? To answer this question, we combined a large-scale structural bioinformatics analysis with all-atom implicit solvent simulations of both rationally designed and naturally occurring proteins. Our results reveal three key takeaways. Firstly, the relative position and accessibility of charged residues across the surface of a folded domain can dictate IDR conformational behavior, overriding expectations based on net surface charge properties. Secondly, naturally occurring proteins possess multiple charge patches that are physically accessible to local IDRs. Finally, even modest changes in the local electrostatic environment of a folded domain can substantially modulate IDR-folded domain interactions. Taken together, our results suggest that folded domain surfaces can act as local determinants of IDR conformational behavior. Intrinsically disordered regions (IDRs) are mostly found adjacent to folded domains. Here we propose that the folded domain surface properties influence IDR behavior. We combine all-atom simulations and sequence design of IDRs and folded domains. IDR conformational behavior is determined by a complex combination of factors. Folded domains can substantially alter IDR conformational biases.
Collapse
Affiliation(s)
- Ishan Taneja
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO, 63110, USA.,Center for Science and Engineering of Living Systems (CSELS), Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO, 63110, USA.,Center for Science and Engineering of Living Systems (CSELS), Washington University in St. Louis, St. Louis, MO, 63130, USA
| |
Collapse
|
296
|
Kumar R, Christensen ND, Kaddis Maldonado RJ, Bewley MC, Ostman A, Sudol M, Chen EC, Buchkovich NW, Gontu A, Surendran Nair M, Nissly RH, Minns AM, Kapur V, Rossi R, Kuchipudi SV, Lindner SE, Parent LJ, Flanagan JM, Buchkovich NJ. Monoclonal Antibodies to S and N SARS-CoV-2 Proteins as Probes to Assess Structural and Antigenic Properties of Coronaviruses. Viruses 2021; 13:v13101899. [PMID: 34696329 PMCID: PMC8537396 DOI: 10.3390/v13101899] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/08/2021] [Accepted: 09/17/2021] [Indexed: 01/18/2023] Open
Abstract
Antibodies targeting the spike (S) and nucleocapsid (N) proteins of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are essential tools. In addition to important roles in the treatment and diagnosis of infection, the availability of high-quality specific antibodies for the S and N proteins is essential to facilitate basic research of virus replication and in the characterization of mutations responsible for variants of concern. We have developed panels of mouse and rabbit monoclonal antibodies (mAbs) to the SARS-CoV-2 spike receptor-binding domain (S-RBD) and N protein for functional and antigenic analyses. The mAbs to the S-RBD were tested for neutralization of native SARS-CoV-2, with several exhibiting neutralizing activity. The panels of mAbs to the N protein were assessed for cross-reactivity with the SARS-CoV and Middle East respiratory syndrome (MERS)-CoV N proteins and could be subdivided into sets that showed unique specificity for SARS-CoV-2 N protein, cross-reactivity between SARS-CoV-2 and SARS-CoV N proteins only, or cross-reactivity to all three coronavirus N proteins tested. Partial mapping of N-reactive mAbs were conducted using truncated fragments of the SARS-CoV-2 N protein and revealed near complete coverage of the N protein. Collectively, these sets of mouse and rabbit monoclonal antibodies can be used to examine structure/function studies for N proteins and to define the surface location of virus neutralizing epitopes on the RBD of the S protein.
Collapse
Affiliation(s)
- Rinki Kumar
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (R.K.); (A.O.); (N.W.B.); (L.J.P.)
| | - Neil D. Christensen
- Department of Pathology, Penn State College of Medicine, Hershey, PA 17033, USA
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
- Correspondence: (N.D.C.); (N.J.B.)
| | - Rebecca J. Kaddis Maldonado
- Department of Medicine, Penn State College of Medicine, Hershey, PA 17033, USA; (R.J.K.M.); (M.S.); (E.C.C.)
| | - Maria C. Bewley
- Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Alexandria Ostman
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (R.K.); (A.O.); (N.W.B.); (L.J.P.)
| | - Malgorzata Sudol
- Department of Medicine, Penn State College of Medicine, Hershey, PA 17033, USA; (R.J.K.M.); (M.S.); (E.C.C.)
| | - Eunice C. Chen
- Department of Medicine, Penn State College of Medicine, Hershey, PA 17033, USA; (R.J.K.M.); (M.S.); (E.C.C.)
| | - Natalie W. Buchkovich
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (R.K.); (A.O.); (N.W.B.); (L.J.P.)
| | - Abhinay Gontu
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA; (A.G.); (M.S.N.)
| | - Meera Surendran Nair
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA; (A.G.); (M.S.N.)
| | - Ruth H. Nissly
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA; (A.G.); (M.S.N.)
| | - Allen M. Minns
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
- Department of Animal Science, Penn State University, University Park, PA 16802, USA
| | - Vivek Kapur
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
- Department of Animal Science, Penn State University, University Park, PA 16802, USA
| | - Randall Rossi
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
| | - Suresh V. Kuchipudi
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
- Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA 16802, USA; (A.G.); (M.S.N.)
| | - Scott E. Lindner
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, PA 16802, USA
| | - Leslie J. Parent
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (R.K.); (A.O.); (N.W.B.); (L.J.P.)
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
- Department of Medicine, Penn State College of Medicine, Hershey, PA 17033, USA; (R.J.K.M.); (M.S.); (E.C.C.)
| | - John M. Flanagan
- Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; (R.H.N.); (A.M.M.); (V.K.); (R.R.); (S.V.K.); (S.E.L.); (J.M.F.)
- Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Nicholas J. Buchkovich
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (R.K.); (A.O.); (N.W.B.); (L.J.P.)
- Correspondence: (N.D.C.); (N.J.B.)
| |
Collapse
|
297
|
Jahangirian E, Jamal GA, Nouroozi M, Mohammadpour A. A reverse vaccinology and immunoinformatics approach for designing a multiepitope vaccine against SARS-CoV-2. Immunogenetics 2021; 73:459-477. [PMID: 34542663 PMCID: PMC8450176 DOI: 10.1007/s00251-021-01228-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/05/2021] [Indexed: 12/16/2022]
Abstract
Since 2019, the world was involved with SARS-CoV-2 and consequently, with the announcement by the World Health Organization that COVID-19 was a pandemic, scientific were an effort to obtain the best approach to combat this global dilemma. The best way to prevent the pandemic from spreading further is to use a vaccine against COVID-19. Here, we report the design of a recombinant multi-epitope vaccine against the four proteins spike or crown (S), membrane (M), nucleocapsid (N), and envelope (E) of SARS-CoV-2 using immunoinformatics tools. We evaluated the most antigenic epitopes that bind to HLA class 1 subtypes, along with HLA class 2, as well as B cell epitopes. Beta-defensin 3 and PADRE sequence were used as adjuvants in the structure of the vaccine. KK, GPGPG, and AAY linkers were used to fuse the selected epitopes. The nucleotide sequence was cloned into pET26b(+) vector using restriction enzymes XhoI and NdeI, and HisTag sequence was considered in the C-terminal of the construct. The results showed that the proposed candidate vaccine is a 70.87 kDa protein with high antigenicity and immunogenicity as well as non-allergenic and non-toxic. A total of 95% of the selected epitopes have conservancy with similar sequences. Molecular docking showed a strong binding between the vaccine structure and tool-like receptor (TLR) 7/8. The docking, molecular dynamics, and MM/PBSA analysis showed that the vaccine established a stable interaction with both structures of TLR7 and TLR8. Simulation of immune stimulation by this vaccine showed that it evokes immune responses related to humoral and cellular immunity.
Collapse
Affiliation(s)
- Ehsan Jahangirian
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Ghadir A Jamal
- Faculty of Allied Health Sciences, Kuwait University, Kuwait City, Kuwait.
| | - MohammadReza Nouroozi
- Department of Animal Science and Food Technology, Agriculture Science and Natural Resources University Khouzestan, Ahwaz, Iran
| | - Alemeh Mohammadpour
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
298
|
Quayum ST, Hasan S. Analysing the impact of the two most common SARS-CoV-2 nucleocapsid protein variants on interactions with membrane protein in silico. J Genet Eng Biotechnol 2021; 19:138. [PMID: 34542740 PMCID: PMC8451389 DOI: 10.1186/s43141-021-00233-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/21/2021] [Indexed: 12/23/2022]
Abstract
As the body of scientific research focusing on the severe acute respiratory syndrome coronavirus 2 or SARS-CoV-2 continues to grow, several mutations have been reported as very common across the globe. In this study, we analysed the SARS-CoV-2 nucleocapsid protein (N protein) with respect to the widely observed 28881-28883 GGG to AAC variant. One of the major functions of the SARS-CoV-2 nucleocapsid protein is virion packaging through its interactions with the membrane protein (M protein). Our goal was to investigate, using in silico studies, the interaction between the mutant nucleocapsid protein and the M protein and how it differed from that of wild type N-M protein interaction. The results showed significant differences in interactions between the two. The mutant protein was predicted to form 3 salt bridges with the M protein, while the wild type only formed 2. The mutant protein was also predicted to display less temperature sensitivity than its wild type counterpart.
Collapse
|
299
|
Vázquez-Valadez VH, Hernández-Serda A, Jiménez-Cabiedes MF, Aguirre-Vidal P, González-Tapia I, Carreño-Vargas L, Alarcón-López YA, Espejel-Fuentes A, Martínez-Soriano P, Lugo Álvarez M, Velázquez-Sánchez AM, Markarian NM, Angeles E, Abrahamyan L. Evaluation of Inhibitory Activity In Silico of In-House Thiomorpholine Compounds between the ACE2 Receptor and S1 Subunit of SARS-CoV-2 Spike. Pathogens 2021; 10:1208. [PMID: 34578240 PMCID: PMC8468748 DOI: 10.3390/pathogens10091208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/22/2022] Open
Abstract
At the end of 2019, the world was struck by the COVID-19 pandemic, which resulted in dire repercussions of unimaginable proportions. From the beginning, the international scientific community employed several strategies to tackle the spread of this disease. Most notably, these consisted of the development of a COVID-19 vaccine and the discovery of antiviral agents through the repositioning of already known drugs with methods such as de novo design. Previously, methylthiomorphic compounds, designed by our group as antihypertensive agents, have been shown to display an affinity with the ACE2 (angiotensin converting enzyme) receptor, a key mechanism required for SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) entry into target cells. Therefore, the objective of this work consists of evaluating, in silico, the inhibitory activity of these compounds between the ACE2 receptor and the S1 subunit of the SARS-CoV-2 spike protein. Supported by the advances of different research groups on the structure of the coronavirus spike and the interaction of the latter with its receptor, ACE2, we carried out a computational study that examined the effect of in-house designed compounds on the inhibition of said interaction. Our results indicate that the polyphenol LQM322 is one of the candidates that should be considered as a possible anti-COVID-19 agent.
Collapse
Affiliation(s)
- Victor H. Vázquez-Valadez
- Departamento de Ciencias Biológicas FES Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico;
| | - Alejandro Hernández-Serda
- Departamento de Ciencias Químicas FES Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (A.H.-S.); (P.M.-S.); (A.M.V.-S.); (E.A.)
| | - Ma. Fernanda Jiménez-Cabiedes
- Laboratorio de Química Medicinal y Teórica FESC, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (M.F.J.-C.); (P.A.-V.); (I.G.-T.); (L.C.-V.); (Y.A.A.-L.); (A.E.-F.); (M.L.Á.)
| | - Pablo Aguirre-Vidal
- Laboratorio de Química Medicinal y Teórica FESC, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (M.F.J.-C.); (P.A.-V.); (I.G.-T.); (L.C.-V.); (Y.A.A.-L.); (A.E.-F.); (M.L.Á.)
| | - Ingrid González-Tapia
- Laboratorio de Química Medicinal y Teórica FESC, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (M.F.J.-C.); (P.A.-V.); (I.G.-T.); (L.C.-V.); (Y.A.A.-L.); (A.E.-F.); (M.L.Á.)
| | - Laura Carreño-Vargas
- Laboratorio de Química Medicinal y Teórica FESC, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (M.F.J.-C.); (P.A.-V.); (I.G.-T.); (L.C.-V.); (Y.A.A.-L.); (A.E.-F.); (M.L.Á.)
| | - Yoshio A. Alarcón-López
- Laboratorio de Química Medicinal y Teórica FESC, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (M.F.J.-C.); (P.A.-V.); (I.G.-T.); (L.C.-V.); (Y.A.A.-L.); (A.E.-F.); (M.L.Á.)
| | - Andrea Espejel-Fuentes
- Laboratorio de Química Medicinal y Teórica FESC, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (M.F.J.-C.); (P.A.-V.); (I.G.-T.); (L.C.-V.); (Y.A.A.-L.); (A.E.-F.); (M.L.Á.)
| | - Pablo Martínez-Soriano
- Departamento de Ciencias Químicas FES Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (A.H.-S.); (P.M.-S.); (A.M.V.-S.); (E.A.)
| | - Miguel Lugo Álvarez
- Laboratorio de Química Medicinal y Teórica FESC, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (M.F.J.-C.); (P.A.-V.); (I.G.-T.); (L.C.-V.); (Y.A.A.-L.); (A.E.-F.); (M.L.Á.)
| | - Ana María Velázquez-Sánchez
- Departamento de Ciencias Químicas FES Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (A.H.-S.); (P.M.-S.); (A.M.V.-S.); (E.A.)
| | - Nathan Marko Markarian
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| | - Enrique Angeles
- Departamento de Ciencias Químicas FES Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Av. 1 de Mayo SN Cuautitlán Izcalli, Estado de México, México CP 54750, Mexico; (A.H.-S.); (P.M.-S.); (A.M.V.-S.); (E.A.)
| | - Levon Abrahamyan
- Swine and Poultry Infectious Diseases Research Center (CRIPA) and Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| |
Collapse
|
300
|
Zheng X, Sun Z, Yu L, Shi D, Zhu M, Yao H, Li L. Interactome Analysis of the Nucleocapsid Protein of SARS-CoV-2 Virus. Pathogens 2021; 10:1155. [PMID: 34578187 PMCID: PMC8465953 DOI: 10.3390/pathogens10091155] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 infection has caused a global pandemic that has severely damaged both public health and the economy. The nucleocapsid protein of SARS-CoV-2 is multifunctional and plays an important role in ribonucleocapsid formation and viral genome replication. In order to elucidate its functions, interaction partners of the SARS-CoV-2 N protein in human cells were identified via affinity purification and mass spectrometry. We identified 160 cellular proteins as interaction partners of the SARS-CoV-2 N protein in HEK293T and/or Calu-3 cells. Functional analysis revealed strong enrichment for ribosome biogenesis and RNA-associated processes, including ribonucleoprotein complex biogenesis, ribosomal large and small subunits biogenesis, RNA binding, catalysis, translation and transcription. Proteins related to virus defence responses, including MOV10, EIF2AK2, TRIM25, G3BP1, ZC3HAV1 and ZCCHC3 were also identified in the N protein interactome. This study comprehensively profiled the viral-host interactome of the SARS-CoV-2 N protein in human cells, and the findings provide the basis for further studies on the pathogenesis and antiviral strategies for this emerging infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Hangping Yao
- Correspondence: (H.Y.); (L.L.); Tel.: +86-571-87236580 (H.Y.); +86-571-87236458 (L.L.)
| | - Lanjuan Li
- Correspondence: (H.Y.); (L.L.); Tel.: +86-571-87236580 (H.Y.); +86-571-87236458 (L.L.)
| |
Collapse
|