1
|
Khalid K, Ahmad F, Anwar A, Ong SK. A Bibliometric Analysis on Multi-epitope Vaccine Development Against SARS-CoV-2: Current Status, Development, and Future Directions. Mol Biotechnol 2025:10.1007/s12033-024-01358-5. [PMID: 39789401 DOI: 10.1007/s12033-024-01358-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
The etiological agent for the coronavirus disease 2019 (COVID-19), the SARS-CoV-2, caused a global pandemic. Although mRNA, viral-vectored, DNA, and recombinant protein vaccine candidates were effective against the SARS-CoV-2 Wuhan strain, the emergence of SARS-CoV-2 variants of concern (VOCs) reduced the protective efficacies of these vaccines. This necessitates the need for effective and accelerated vaccine development against mutated VOCs. The development of multi-epitope vaccines against SARS-CoV-2 based on in silico identification of highly conserved and immunogenic epitopes is a promising strategy for future SARS-CoV-2 vaccine development. Considering the evolving landscape of the COVID-19 pandemic, we have conducted a bibliometric analysis to consolidate current findings and research trends in multi-epitope vaccine development to provide insights for future vaccine development strategies. Analysis of 102 publications on multi-epitope vaccine development against SARS-CoV-2 revealed significant growth and global collaboration, with India leading in the number of publications, along with an identification of the most prolific authors. Key journals included the Journal of Biomolecular Structure and Dynamics, while top collaborations involved Pakistan-China and India-USA. Keyword analysis showed a prominent focus on immunoinformatics, epitope prediction, and spike glycoprotein. Advances in immunoinformatics, including AI-driven epitope prediction, offer promising avenues for the development of safe and effective multi-epitope vaccines. Immunogenicity may be further improved through nanoparticle-based systems or the use of adjuvants along with real-time genomic surveillance to tailor vaccines against emerging variants.
Collapse
Affiliation(s)
- Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Petaling Jaya, Selangor, Malaysia.
| | - Fiaz Ahmad
- Department of Economics and Finance, Sunway Business School, Sunway University, Bandar Sunway, 47500, Petaling Jaya, Selangor, Malaysia
| | - Ayaz Anwar
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Petaling Jaya, Selangor, Malaysia
| | - Seng-Kai Ong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Petaling Jaya, Selangor, Malaysia
| |
Collapse
|
2
|
Ong YC, Tejo BA, Yap WB. An Immunoinformatic Approach for Identifying and Designing Conserved Multi-Epitope Vaccines for Coronaviruses. Biomedicines 2024; 12:2530. [PMID: 39595095 PMCID: PMC11592158 DOI: 10.3390/biomedicines12112530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The COVID-19 pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus has exposed the vulnerabilities and unpreparedness of the global healthcare system in dealing with emerging zoonoses. In the past two decades, coronaviruses (CoV) have been responsible for three major viral outbreaks, and the likelihood of future outbreaks caused by these viruses is high and nearly inevitable. Therefore, effective prophylactic universal vaccines targeting multiple circulating and emerging coronavirus strains are warranted. METHODS This study utilized an immunoinformatic approach to identify evolutionarily conserved CD4+ (HTL) and CD8+ (CTL) T cells, and B-cell epitopes in the coronaviral spike (S) glycoprotein. RESULTS A total of 132 epitopes were identified, with the majority of them found to be conserved across the bat CoVs, pangolin CoVs, endemic coronaviruses, SARS-CoV-2, and Middle East respiratory syndrome coronavirus (MERS-CoV). Their peptide sequences were then aligned and assembled to identify the overlapping regions. Eventually, two major peptide assemblies were derived based on their promising immune-stimulating properties. CONCLUSIONS In this light, they can serve as lead candidates for universal coronavirus vaccine development, particularly in the search for pan-coronavirus multi-epitope universal vaccines that can confer protection against current and novel coronaviruses.
Collapse
Affiliation(s)
- Yu Chuan Ong
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Bimo Ario Tejo
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Wei Boon Yap
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
- One Health UKM, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|
3
|
Nafian F, Soleymani G, Pourmanouchehri Z, Kiyanjam M, Nafian S, Mohammadi SM, Jeyroudi H, Berenji Jalaei S, Sabzpoushan F. In Silico Design of a Trans-Amplifying RNA-Based Vaccine against SARS-CoV-2 Structural Proteins. Adv Virol 2024; 2024:3418062. [PMID: 39380944 PMCID: PMC11459942 DOI: 10.1155/2024/3418062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 10/10/2024] Open
Abstract
Nucleic acid-based vaccines allow scalable, rapid, and cell-free vaccine production in response to an emerging disease such as the current COVID-19 pandemic. Here, we objected to the design of a multiepitope mRNA vaccine against the structural proteins of SARS-CoV-2. Through an immunoinformatic approach, promising epitopes were predicted for the spike (S), envelope (E), membrane (M), and nucleocapsid (N) proteins. Fragments rich in overlapping epitopes were selected based on binding affinities with HLA classes I and II for the specific presentation to B and T lymphocytes. Two constructs were designed by fusing the fragments in different arrangements via GG linkers. Construct 1 showed better structural properties and interactions with toll-like receptor 2 (TLR-2), TLR-3, and TLR-4 during molecular docking and dynamic simulation. A 50S ribosomal L7/L12 adjuvant was added to its N-terminus to improve stability and immunogenicity. The final RNA sequence was used to design a trans-amplifying RNA (taRNA) vaccine in a split-vector system. It consists of two molecules: a nonreplicating RNA encoding a trans-acting replicase to amplify the second one, a trans-replicon (TR) RNA encoding the vaccine protein. Overall, the immune response simulation detected that activated B and T lymphocytes and increased memory cell formation. Macrophages and dendritic cells proliferated continuously, and IFN-γ and cytokines like IL-2 were released highly.
Collapse
Affiliation(s)
- Fatemeh Nafian
- Department of Medical Laboratory SciencesFaculty of ParamedicsTehran Medical SciencesIslamic Azad University, Tehran, Iran
| | - Ghazal Soleymani
- Department of Biological SciencesVirginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Zahra Pourmanouchehri
- Department of BiologyTechnical University of Kaiserslautern, Kaiserslautern Technical University of Kaiserslautern, Kaiserslautern, Germany
| | - Mahnaz Kiyanjam
- Department of Cellular and Molecular BiologyFaculty of Advanced Sciences and TechnologyTehran Medical SciencesIslamic Azad University, Tehran, Iran
| | - Simin Nafian
- Department of Stem Cell and Regenerative MedicineNational Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Sayed Mohammad Mohammadi
- Department of BiotechnologyFaculty of Converging Sciences and TechnologiesScience and Research BranchIslamic Azad University, Tehran, Iran
| | - Hanie Jeyroudi
- Department of Cellular and Molecular BiologyFaculty of Advanced Sciences and TechnologyTehran Medical SciencesIslamic Azad University, Tehran, Iran
| | - Sharareh Berenji Jalaei
- Department of BiochemistryFaculty of Converging Sciences and TechnologiesScience and Research BranchIslamic Azad University, Tehran, Iran
| | - Fatemeh Sabzpoushan
- Department of Cellular and Molecular BiologyFaculty of Advanced Sciences and TechnologyTehran Medical SciencesIslamic Azad University, Tehran, Iran
| |
Collapse
|
4
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
- Ziyi Bai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
5
|
Samrat SK, Kumar P, Liu Y, Chen K, Lee H, Li Z, Chen Y, Li H. An ISG15-Based High-Throughput Screening Assay for Identification and Characterization of SARS-CoV-2 Inhibitors Targeting Papain-like Protease. Viruses 2024; 16:1239. [PMID: 39205213 PMCID: PMC11359932 DOI: 10.3390/v16081239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Emergence of newer variants of SARS-CoV-2 underscores the need for effective antivirals to complement the vaccination program in managing COVID-19. The multi-functional papain-like protease (PLpro) of SARS-CoV-2 is an essential viral protein that not only regulates the viral replication but also modulates the host immune system, making it a promising therapeutic target. To this end, we developed an in vitro interferon stimulating gene 15 (ISG15)-based Förster resonance energy transfer (FRET) assay and screened the National Cancer Institute (NCI) Diversity Set VI compound library, which comprises 1584 small molecules. Subsequently, we assessed the PLpro enzymatic activity in the presence of screened molecules. We identified three potential PLpro inhibitors, namely, NSC338106, 651084, and 679525, with IC50 values in the range from 3.3 to 6.0 µM. These molecules demonstrated in vitro inhibition of the enzyme activity and exhibited antiviral activity against SARS-CoV-2, with EC50 values ranging from 0.4 to 4.6 µM. The molecular docking of all three small molecules to PLpro suggested their specificity towards the enzyme's active site. Overall, our study contributes promising prospects for further developing potential antivirals to combat SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Subodh Kumar Samrat
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Prashant Kumar
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Yuchen Liu
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Ke Chen
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Hyun Lee
- Department of Pharmaceutical Sciences, College of Pharmacy and Biophysics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Zhong Li
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Yin Chen
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
| | - Hongmin Li
- Department of Pharmacology and Toxicology, R Ken Coit College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (P.K.); (Y.L.); (K.C.); (Z.L.); (Y.C.)
- Department of Chemistry and Biochemistry, College of Science & College of Medicine, The University of Arizona, Tucson, AZ 85721, USA
- The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
6
|
Liu J, Han H, Yang B, Zhang N, Li J, Chen X, Wu J, Zhao Y, Yang Y. Immunogenicity and protective efficacy of the HC009 mRNA vaccine against SARS-CoV-2. Front Immunol 2024; 15:1416375. [PMID: 39131158 PMCID: PMC11310568 DOI: 10.3389/fimmu.2024.1416375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/27/2024] [Indexed: 08/13/2024] Open
Abstract
With the rapid global spread of COVID-19 and the continuous emergence of variants, there is an urgent need to develop safe and effective vaccines. Here, we developed a novel mRNA vaccine, HC009, based on new formulation by the QTsome delivery platform. Immunogenicity results showed that the prime-boost immunization strategy with HC009 was able to induce robust and durable humoral immunity, as well as Th1-biased cellular responses in rodents or non-human primates (NHPs). After further challenge with live SARS-CoV-2 virus, HC009 provided adequate protection against virus infection in hACE2 transgenic mice. Therefore, HC009 could provide significant immune protection against SARS-CoV-2.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Mice
- mRNA Vaccines/immunology
- Mice, Transgenic
- Immunogenicity, Vaccine
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Humans
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Immunity, Humoral
- Female
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice, Inbred BALB C
- Vaccine Efficacy
Collapse
Affiliation(s)
- Juan Liu
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | | - Yongsheng Yang
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Pscheidt VM, de Souza PO, Fazolo T, Modena JLP, Simeoni C, Teixeira D, Silva NB, Dos Santos KB, Júnior LR, Bonorino C. A flow cytometry-based assay to measure neutralizing antibodies against SARS-CoV-2 virus. Cytometry A 2024; 105:446-457. [PMID: 38624015 DOI: 10.1002/cyto.a.24838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/11/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024]
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus has highlighted the need for serological assays that can accurately evaluate the neutralizing efficiency of antibodies produced during infection or induced by vaccines. However, conventional assays often require the manipulation of live viruses on a level-three biosafety (BSL3) facility, which presents practical and safety challenges. Here, we present a novel, alternative assay that measures neutralizing antibodies (NAbs) against SARS-CoV-2 in plasma using flow cytometry. This assay is based on antibody binding to the S protein and has demonstrated precision in both intra- and inter-assay measurements at a dilution of 1:50. The cut-off was determined using Receiver Operating Characteristic (ROC) analysis and the value of 36.01% has shown high sensitivity and specificity in distinguishing between pre-pandemic sera, COVID-19 patients, and vaccinated individuals. The efficiency significantly correlates with the gold standard test, PRNT. Our new assay offers a safe and efficient alternative to conventional assays for evaluating NAbs against SARS-CoV-2.
Collapse
Affiliation(s)
- Veridiane M Pscheidt
- Immunotherapy Laboratory - (LAIT) - Department of Basic Health Sciences of Federal University of Health Sciences of Porto Alegre, UFCSPA, Porto Alegre, Brazil
| | - Priscila Oliveira de Souza
- Immunotherapy Laboratory - (LAIT) - Department of Basic Health Sciences of Federal University of Health Sciences of Porto Alegre, UFCSPA, Porto Alegre, Brazil
| | - Tiago Fazolo
- Immunotherapy Laboratory - (LAIT) - Department of Basic Health Sciences of Federal University of Health Sciences of Porto Alegre, UFCSPA, Porto Alegre, Brazil
| | - José Luiz Proença Modena
- Laboratory of Emerging Viruses (LEVE) - Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas - UNICAMP - Campinas, São Paulo, Brazil
| | - Camila Simeoni
- Laboratory of Emerging Viruses (LEVE) - Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas - UNICAMP - Campinas, São Paulo, Brazil
| | - Daniel Teixeira
- Laboratory of Emerging Viruses (LEVE) - Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas - UNICAMP - Campinas, São Paulo, Brazil
| | - Natália Brunetti Silva
- Laboratory of Emerging Viruses (LEVE) - Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas - UNICAMP - Campinas, São Paulo, Brazil
| | - Karina Bispo Dos Santos
- Laboratory of Emerging Viruses (LEVE) - Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas - UNICAMP - Campinas, São Paulo, Brazil
| | - Luiz Rodrigues Júnior
- Immunotherapy Laboratory - (LAIT) - Department of Basic Health Sciences of Federal University of Health Sciences of Porto Alegre, UFCSPA, Porto Alegre, Brazil
| | - Cristina Bonorino
- Immunotherapy Laboratory - (LAIT) - Department of Basic Health Sciences of Federal University of Health Sciences of Porto Alegre, UFCSPA, Porto Alegre, Brazil
- Department of Surgery, University of California at San Diego - UCSD, La Jolla, California, USA
| |
Collapse
|
8
|
Sarnelli G, Del Re A, Palenca I, Franzin SB, Lu J, Seguella L, Zilli A, Pesce M, Rurgo S, Esposito G, Sanseverino W, Esposito G. Intranasal administration of Escherichia coli Nissle expressing the spike protein of SARS-CoV-2 induces long-term immunization and prevents spike protein-mediated lung injury in mice. Biomed Pharmacother 2024; 174:116441. [PMID: 38518597 DOI: 10.1016/j.biopha.2024.116441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
While current anti-Spike protein (SP) vaccines have been pivotal in managing the pandemic, their limitations in delivery, storage, and the inability to provide mucosal immunization (preventing infections) highlight the ongoing necessity for research and innovation. To tackle these constraints, our research group developed a bacterial-based vaccine using a non-pathogenic E. coli Nissle 1917 (EcN) strain genetically modified to express the SARS-CoV-2 spike protein on its surface (EcN-pAIDA1-SP). We intranasally delivered the EcN-pAIDA1-SP in two doses and checked specific IgG/IgA production as well as the key immune mediators involved in the process. Moreover, following the initial and booster vaccine doses, we exposed both immunized and non-immunized mice to intranasal delivery of SARS-CoV-2 SP to assess the effectiveness of EcN-pAIDA1-SP in protecting lung tissue from the inflammation damage. We observed detectable levels of anti-SARS-CoV-2 spike IgG in serum samples and IgA in bronchoalveolar lavage fluid two weeks after the initial treatment, with peak concentrations in the respective samples on the 35th day. Moreover, immunoglobulins displayed a progressively enhanced avidity index, suggesting a selective binding to the spike protein. Finally, the pre-immunized group displayed a decrease in proinflammatory markers (TLR4, NLRP3, ILs) following SP challenge, compared to the non-immunized groups, along with better preservation of tissue morphology. Our probiotic-based technology provides an effective immunobiotic tool to protect individuals against disease and control infection spread.
Collapse
Affiliation(s)
- Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy; Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy.
| | - Alessandro Del Re
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Irene Palenca
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Silvia Basili Franzin
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Jie Lu
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Anatomy and Cell Biology, China Medical University, Shenyang 110122, China.
| | - Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy
| | - Sara Rurgo
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy.
| | - Giovanni Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Molecular Medicine and Medical Biotechnologies, Centro Ingegneria Genetica-Biotecnologie Avanzate s.c.a rl, Naples 80131, Italy.
| | - Walter Sanseverino
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy.
| | - Giuseppe Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| |
Collapse
|
9
|
Chatterjee S, Zaia J. Proteomics-based mass spectrometry profiling of SARS-CoV-2 infection from human nasopharyngeal samples. MASS SPECTROMETRY REVIEWS 2024; 43:193-229. [PMID: 36177493 PMCID: PMC9538640 DOI: 10.1002/mas.21813] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 05/12/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the on-going global pandemic of coronavirus disease 2019 (COVID-19) that continues to pose a significant threat to public health worldwide. SARS-CoV-2 encodes four structural proteins namely membrane, nucleocapsid, spike, and envelope proteins that play essential roles in viral entry, fusion, and attachment to the host cell. Extensively glycosylated spike protein efficiently binds to the host angiotensin-converting enzyme 2 initiating viral entry and pathogenesis. Reverse transcriptase polymerase chain reaction on nasopharyngeal swab is the preferred method of sample collection and viral detection because it is a rapid, specific, and high-throughput technique. Alternate strategies such as proteomics and glycoproteomics-based mass spectrometry enable a more detailed and holistic view of the viral proteins and host-pathogen interactions and help in detection of potential disease markers. In this review, we highlight the use of mass spectrometry methods to profile the SARS-CoV-2 proteome from clinical nasopharyngeal swab samples. We also highlight the necessity for a comprehensive glycoproteomics mapping of SARS-CoV-2 from biological complex matrices to identify potential COVID-19 markers.
Collapse
Affiliation(s)
- Sayantani Chatterjee
- Department of Biochemistry, Center for Biomedical Mass SpectrometryBoston University School of MedicineBostonMassachusettsUSA
| | - Joseph Zaia
- Department of Biochemistry, Center for Biomedical Mass SpectrometryBoston University School of MedicineBostonMassachusettsUSA
- Bioinformatics ProgramBoston University School of MedicineBostonMassachusettsUSA
| |
Collapse
|
10
|
Samrat SK, Bashir Q, Zhang R, Huang Y, Liu Y, Wu X, Brown T, Wang W, Zheng YG, Zhang QY, Chen Y, Li Z, Li H. A universal fluorescence polarization high throughput screening assay to target the SAM-binding sites of SARS-CoV-2 and other viral methyltransferases. Emerg Microbes Infect 2023; 12:2204164. [PMID: 37060263 PMCID: PMC10165934 DOI: 10.1080/22221751.2023.2204164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/16/2023]
Abstract
SARS-CoV-2 has caused a global pandemic with significant humanity and economic loss since 2020. Currently, only limited options are available to treat SARS-CoV-2 infections for vulnerable populations. In this study, we report a universal fluorescence polarization (FP)-based high throughput screening (HTS) assay for SAM-dependent viral methyltransferases (MTases), using a fluorescent SAM-analogue, FL-NAH. We performed the assay against a reference MTase, NSP14, an essential enzyme for SARS-CoV-2 to methylate the N7 position of viral 5'-RNA guanine cap. The assay is universal and suitable for any SAM-dependent viral MTases such as the SARS-CoV-2 NSP16/NSP10 MTase complex and the NS5 MTase of Zika virus (ZIKV). Pilot screening demonstrated that the HTS assay was very robust and identified two candidate inhibitors, NSC 111552 and 288387. The two compounds inhibited the FL-NAH binding to the NSP14 MTase with low micromolar IC50. We used three functional MTase assays to unambiguously verified the inhibitory potency of these molecules for the NSP14 N7-MTase function. Binding studies indicated that these molecules are bound directly to the NSP14 MTase with similar low micromolar affinity. Moreover, we further demonstrated that these molecules significantly inhibited the SARS-CoV-2 replication in cell-based assays at concentrations not causing cytotoxicity. Furthermore, NSC111552 significantly synergized with known SARS-CoV-2 drugs including nirmatrelvir and remdesivir. Finally, docking suggested that these molecules bind specifically to the SAM-binding site on the NSP14 MTase. Overall, these molecules represent novel and promising candidates to further develop broad-spectrum inhibitors for the management of viral infections.
Collapse
Affiliation(s)
- Subodh Kumar Samrat
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Qamar Bashir
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Ran Zhang
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Yiding Huang
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Yuchen Liu
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Xiangmeng Wu
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Tyler Brown
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Wei Wang
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Y. George Zheng
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Qing-Yu Zhang
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Yin Chen
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Zhong Li
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Hongmin Li
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
- Department of Chemistry and Biochemistry, College of Science & College of Medicine, The University of Arizona, Tucson, AZ, USA
- The BIO5 Institute, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
11
|
Le K, Kannappan S, Kim T, Lee JH, Lee HR, Kim KK. Structural understanding of SARS-CoV-2 virus entry to host cells. Front Mol Biosci 2023; 10:1288686. [PMID: 38033388 PMCID: PMC10683510 DOI: 10.3389/fmolb.2023.1288686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major global health concern associated with millions of fatalities worldwide. Mutant variants of the virus have further exacerbated COVID-19 mortality and infection rates, emphasizing the urgent need for effective preventive strategies. Understanding the viral infection mechanism is crucial for developing therapeutics and vaccines. The entry of SARS-CoV-2 into host cells is a key step in the infection pathway and has been targeted for drug development. Despite numerous reviews of COVID-19 and the virus, there is a lack of comprehensive reviews focusing on the structural aspects of viral entry. In this review, we analyze structural changes in Spike proteins during the entry process, dividing the entry process into prebinding, receptor binding, proteolytic cleavage, and membrane fusion steps. By understanding the atomic-scale details of viral entry, we can better target the entry step for intervention strategies. We also examine the impacts of mutations in Spike proteins, including the Omicron variant, on viral entry. Structural information provides insights into the effects of mutations and can guide the development of therapeutics and vaccines. Finally, we discuss available structure-based approaches for the development of therapeutics and vaccines. Overall, this review provides a detailed analysis of the structural aspects of SARS-CoV-2 viral entry, highlighting its significance in the development of therapeutics and vaccines against COVID-19. Therefore, our review emphasizes the importance of structural information in combating SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Kim Le
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Shrute Kannappan
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Sungkyunkwan University, Suwon, Republic of Korea
- Research Center for Advanced Materials Technology Core Research Institute, Suwon, Republic of Korea
| | - Truc Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jung Heon Lee
- Research Center for Advanced Materials Technology Core Research Institute, Suwon, Republic of Korea
- School of Advanced Materials and Science Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hye-Ra Lee
- Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong, Republic of Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
12
|
Wu HB, Wang CH, Chung YD, Shan YS, Lin YJ, Tsai HP, Lee GB. Highly-specific aptamer targeting SARS-CoV-2 S1 protein screened on an automatic integrated microfluidic system for COVID-19 diagnosis. Anal Chim Acta 2023; 1274:341531. [PMID: 37455073 DOI: 10.1016/j.aca.2023.341531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
Variants of the severe acute respiratory syndrome coronavirus (SARS-CoV-2) have evolved such that it may be challenging for diagnosis and clinical treatment of the pandemic coronavirus disease-19 (COVID-19). Compared with developed SARS-CoV-2 diagnostic tools recently, aptamers may exhibit some advantages, including high specificity/affinity, longer shelf life (vs. antibodies), and could be easily prepared. Herein an integrated microfluidic system was developed to automatically carry out one novel screening process based on the systematic evolution of ligands by exponential enrichment (SELEX) for screening aptamers specific with SARS-CoV-2. The new screening process started with five rounds of positive selection (with the S1 protein of SARS-CoV-2). In addition, including non-target viruses (influenza A and B), human respiratory tract-related cancer cells (adenocarcinoma human alveolar basal epithelial cells and dysplastic oral keratinocytes), and upper respiratory tract-related infectious bacteria (including methicillin-resistant Staphylococcus aureus, Pseudomonas aeruginosa, Acinetobacter baumannii, and Klebsiella pneumoniae), and human saliva were involved to increase the specificity of the screened aptamer during the negative selection. Totally, all 10 rounds could be completed within 20 h. The dissociation constant of the selected aptamer was determined to be 63.0 nM with S1 protein. Limits of detection for Wuhan and Omicron clinical strains were found to be satisfactory for clinical applications (i.e. 4.80 × 101 and 1.95 × 102 copies/mL, respectively). Moreover, the developed aptamer was verified to be capable of capturing inactivated SARS-CoV-2 viruses, eight SARS-CoV-2 pseudo-viruses, and clinical isolates of SARS-CoV-2 viruses. For high-variable emerging viruses, this developed integrated microfluidic system can be used to rapidly select highly-specific aptamers based on the novel SELEX methods to deal with infectious diseases in the future.
Collapse
Affiliation(s)
- Hung-Bin Wu
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Hung Wang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Da Chung
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan; Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Jun Lin
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Huey-Pin Tsai
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Gwo-Bin Lee
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan; Institute of NanoEngineering and Microsystems, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
13
|
Bostanghadiri N, Ziaeefar P, Mofrad MG, Yousefzadeh P, Hashemi A, Darban-Sarokhalil D. COVID-19: An Overview of SARS-CoV-2 Variants-The Current Vaccines and Drug Development. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1879554. [PMID: 37674935 PMCID: PMC10480030 DOI: 10.1155/2023/1879554] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/07/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023]
Abstract
The world is presently in crisis facing an outbreak of a health-threatening microorganism known as COVID-19, responsible for causing uncommon viral pneumonia in humans. The virus was first reported in Wuhan, China, in early December 2019, and it quickly became a global concern due to the pandemic. Challenges in this regard have been compounded by the emergence of several variants such as B.1.1.7, B.1.351, P1, and B.1.617, which show an increase in transmission power and resistance to therapies and vaccines. Ongoing researches are focused on developing and manufacturing standard treatment strategies and effective vaccines to control the pandemic. Despite developing several vaccines such as Pfizer/BioNTech and Moderna approved by the U.S. Food and Drug Administration (FDA) and other vaccines in phase 4 clinical trials, preventive measures are mandatory to control the COVID-19 pandemic. In this review, based on the latest findings, we will discuss different types of drugs as therapeutic options and confirmed or developing vaccine candidates against SARS-CoV-2. We also discuss in detail the challenges posed by the variants and their effect on therapeutic and preventive interventions.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pardis Ziaeefar
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morvarid Golrokh Mofrad
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Parsa Yousefzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Patil S, Vijayanand S, Menon I, Gomes KB, Kale A, Bagwe P, Yacoub S, Uddin MN, D’Souza MJ. Adjuvanted-SARS-CoV-2 Spike Protein-Based Microparticulate Vaccine Delivered by Dissolving Microneedles Induces Humoral, Mucosal, and Cellular Immune Responses in Mice. Pharmaceuticals (Basel) 2023; 16:1131. [PMID: 37631046 PMCID: PMC10457992 DOI: 10.3390/ph16081131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19 continues to cause an increase in the number of cases and deaths worldwide. Due to the ever-mutating nature of the virus, frequent vaccination against COVID-19 is anticipated. Most of the approved SARS-CoV-2 vaccines are administered using the conventional intramuscular route, causing vaccine hesitancy. Thus, there is a need for an effective, non-invasive vaccination strategy against COVID-19. This study evaluated the synergistic effects of a subunit microparticulate vaccine delivered using microneedles. The microparticles encapsulated a highly immunogenic subunit protein of the SARS-CoV-2 virus, such as the spike protein's receptor binding domain (RBD). Adjuvants were also incorporated to enhance the spike RBD-specific immune response. Our vaccination study reveals that a microneedle-based vaccine delivering these microparticles induced spike RBD-specific IgM, IgG, IgG1, IgG2a, and IgA antibodies. The vaccine also generated high levels of CD4+ and CD8a+ molecules in the secondary lymphoid organs. Overall, dissolving microneedles delivery spike RBD antigen in microparticulate form induced a robust immune response, paving the way for an alternative self-administrable, non-invasive vaccination strategy against COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mohammad N. Uddin
- Center for Drug Delivery and Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (S.P.); (S.V.); (I.M.); (K.B.G.); (A.K.); (P.B.); (S.Y.)
| | - Martin J. D’Souza
- Center for Drug Delivery and Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (S.P.); (S.V.); (I.M.); (K.B.G.); (A.K.); (P.B.); (S.Y.)
| |
Collapse
|
15
|
Palma M. Epitopes and Mimotopes Identification Using Phage Display for Vaccine Development against Infectious Pathogens. Vaccines (Basel) 2023; 11:1176. [PMID: 37514992 PMCID: PMC10384025 DOI: 10.3390/vaccines11071176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Traditional vaccines use inactivated or weakened forms of pathogens which could have side effects and inadequate immune responses. To overcome these challenges, phage display has emerged as a valuable tool for identifying specific epitopes that could be used in vaccines. This review emphasizes the direct connection between epitope identification and vaccine development, filling a crucial gap in the field. This technique allows vaccines to be engineered to effectively stimulate the immune system by presenting carefully selected epitopes. Phage display involves screening libraries of random peptides or gene/genome fragments using serum samples from infected, convalescent, or vaccinated individuals. This method has been used to identify epitopes from various pathogens including SARS-CoV-2, Mycobacterium tuberculosis, hepatitis viruses, H5N1, HIV-1, Human T-lymphotropic virus 1, Plasmodium falciparum, Trypanosoma cruzi, and Dirofilaria repens. Bacteriophages offer advantages such as being immunogenic carriers, low production costs, and customization options, making them a promising alternative to traditional vaccines. The purpose of this study has been to highlight an approach that encompasses the entire process from epitope identification to vaccine production using a single technique, without requiring additional manipulation. Unlike conventional methods, phage display demonstrates exceptional efficiency and speed, which could provide significant advantages in critical scenarios such as pandemics.
Collapse
Affiliation(s)
- Marco Palma
- Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain
- Protheragen Inc., Ronkonkoma, NY 11779, USA
| |
Collapse
|
16
|
Lorenzo MM, Marín-López A, Chiem K, Jimenez-Cabello L, Ullah I, Utrilla-Trigo S, Calvo-Pinilla E, Lorenzo G, Moreno S, Ye C, Park JG, Matía A, Brun A, Sánchez-Puig JM, Nogales A, Mothes W, Uchil PD, Kumar P, Ortego J, Fikrig E, Martinez-Sobrido L, Blasco R. Vaccinia Virus Strain MVA Expressing a Prefusion-Stabilized SARS-CoV-2 Spike Glycoprotein Induces Robust Protection and Prevents Brain Infection in Mouse and Hamster Models. Vaccines (Basel) 2023; 11:1006. [PMID: 37243110 PMCID: PMC10220993 DOI: 10.3390/vaccines11051006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The COVID-19 pandemic has underscored the importance of swift responses and the necessity of dependable technologies for vaccine development. Our team previously developed a fast cloning system for the modified vaccinia virus Ankara (MVA) vaccine platform. In this study, we reported on the construction and preclinical testing of a recombinant MVA vaccine obtained using this system. We obtained recombinant MVA expressing the unmodified full-length SARS-CoV-2 spike (S) protein containing the D614G amino-acid substitution (MVA-Sdg) and a version expressing a modified S protein containing amino-acid substitutions designed to stabilize the protein a in a pre-fusion conformation (MVA-Spf). S protein expressed by MVA-Sdg was found to be expressed and was correctly processed and transported to the cell surface, where it efficiently produced cell-cell fusion. Version Spf, however, was not proteolytically processed, and despite being transported to the plasma membrane, it failed to induce cell-cell fusion. We assessed both vaccine candidates in prime-boost regimens in the susceptible transgenic K18-human angiotensin-converting enzyme 2 (K18-hACE2) in mice and in golden Syrian hamsters. Robust immunity and protection from disease was induced with either vaccine in both animal models. Remarkably, the MVA-Spf vaccine candidate produced higher levels of antibodies, a stronger T cell response, and a higher degree of protection from challenge. In addition, the level of SARS-CoV-2 in the brain of MVA-Spf inoculated mice was decreased to undetectable levels. Those results add to our current experience and range of vaccine vectors and technologies for developing a safe and effective COVID-19 vaccine.
Collapse
Affiliation(s)
- María M. Lorenzo
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Alejandro Marín-López
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Kevin Chiem
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Luis Jimenez-Cabello
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Irfan Ullah
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Sergio Utrilla-Trigo
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Gema Lorenzo
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Sandra Moreno
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Jun-Gyu Park
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Alejandro Matía
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Alejandro Brun
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Juana M. Sánchez-Puig
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| | - Aitor Nogales
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA; (W.M.); (P.D.U.)
| | - Pradeep D. Uchil
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA; (W.M.); (P.D.U.)
| | - Priti Kumar
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal, INIA CSIC, Carretera Valdeolmos a El Casar, Valdeolmos, E-28130 Madrid, Spain; (L.J.-C.); (S.U.-T.); (E.C.-P.); (G.L.); (A.B.); (A.N.); (J.O.)
| | - Erol Fikrig
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA; (A.M.-L.); (I.U.); (E.F.)
| | - Luis Martinez-Sobrido
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA; (K.C.); (C.Y.); (J.-G.P.); (P.K.)
| | - Rafael Blasco
- Departamento de Biotecnología, INIA CSIC, Carretera La Coruña km 7.5, E-28040 Madrid, Spain; (M.M.L.); (S.M.); (A.M.); (J.M.S.-P.)
| |
Collapse
|
17
|
Sarnelli G, Del Re A, Pesce M, Lu J, Esposito G, Sanseverino W, Corpetti C, Basili Franzin S, Seguella L, Palenca I, Rurgo S, De Palma FDE, Zilli A, Esposito G. Oral Immunization with Escherichia coli Nissle 1917 Expressing SARS-CoV-2 Spike Protein Induces Mucosal and Systemic Antibody Responses in Mice. Biomolecules 2023; 13:biom13030569. [PMID: 36979504 PMCID: PMC10046078 DOI: 10.3390/biom13030569] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/04/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
As of October 2022, the COVID-19 pandemic continues to pose a major public health conundrum, with increased rates of symptomatic infections in vaccinated individuals. An ideal vaccine candidate for the prevention of outbreaks should be rapidly scalable, easy to administer, and able to elicit a potent mucosal immunity. Towards this aim, we proposed an engineered Escherichia coli (E. coli) Nissle 1917 (EcN) strain with SARS-CoV-2 spike protein (SP)-coding plasmid, which was able to expose SP on its cellular surface by a hybridization with the adhesin involved in diffuse adherence 1 (AIDA1). In this study, we presented the effectiveness of a 16-week intragastrically administered, engineered EcN in producing specific systemic and mucosal immunoglobulins against SARS-CoV-2 SP in mice. We observed a time-dependent increase in anti-SARS-CoV-2 SP IgG antibodies in the sera at week 4, with a titre that more than doubled by week 12 and a stable circulating titre by week 16 (+309% and +325% vs. control; both p < 0.001). A parallel rise in mucosal IgA antibody titre in stools, measured via intestinal and bronchoalveolar lavage fluids of the treated mice, reached a plateau by week 12 and until the end of the immunization protocol (+300, +47, and +150%, at week 16; all p < 0.001 vs. controls). If confirmed in animal models of infection, our data indicated that the engineered EcN may be a potential candidate as an oral vaccine against COVID-19. It is safe, inexpensive, and, most importantly, able to stimulate the production of both systemic and mucosal anti-SARS-CoV-2 spike-protein antibodies.
Collapse
Affiliation(s)
- Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, 80138 Naples, Italy
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
| | - Alessandro Del Re
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, 80138 Naples, Italy
| | - Jie Lu
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
- Department of Anatomy and Cell Biology, China Medical University, Shenyang 110122, China
| | - Giovanni Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, Centro Ingegneria Genetica-Biotecnologie Avanzate s.c.a rl, 80131 Naples, Italy
| | - Walter Sanseverino
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
| | - Chiara Corpetti
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Silvia Basili Franzin
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Irene Palenca
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Sara Rurgo
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, 80138 Naples, Italy
| | - Fatima Domenica Elisa De Palma
- Department of Molecular Medicine and Medical Biotechnologies, Centro Ingegneria Genetica-Biotecnologie Avanzate s.c.a rl, 80131 Naples, Italy
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Giuseppe Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), 80100 Naples, Italy
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
18
|
Pacheco-García U, Serafín-López J. Indirect Dispersion of SARS-CoV-2 Live-Attenuated Vaccine and Its Contribution to Herd Immunity. Vaccines (Basel) 2023; 11:655. [PMID: 36992239 PMCID: PMC10055900 DOI: 10.3390/vaccines11030655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
It has been 34 months since the beginning of the SARS-CoV-2 coronavirus pandemic, which causes the COVID-19 disease. In several countries, immunization has reached a proportion near what is required to reach herd immunity. Nevertheless, infections and re-infections have been observed even in vaccinated persons. That is because protection conferred by vaccines is not entirely effective against new virus variants. It is unknown how often booster vaccines will be necessary to maintain a good level of protective immunity. Furthermore, many individuals refuse vaccination, and in developing countries, a large proportion of the population has not yet been vaccinated. Some live-attenuated vaccines against SARS-CoV-2 are being developed. Here, we analyze the indirect dispersion of a live-attenuated virus from vaccinated individuals to their contacts and the contribution that this phenomenon could have to reaching Herd Immunity.
Collapse
Affiliation(s)
- Ursino Pacheco-García
- Department of Cardio-Renal Pathophysiology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico
| | - Jeanet Serafín-López
- Department of Immunology, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico
| |
Collapse
|
19
|
Synthesis, Molecular Docking, and Dynamic Simulation Targeting Main Protease (Mpro) of New, Thiazole Clubbed Pyridine Scaffolds as Potential COVID-19 Inhibitors. Curr Issues Mol Biol 2023; 45:1422-1442. [PMID: 36826038 PMCID: PMC9955078 DOI: 10.3390/cimb45020093] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Many biological activities of pyridine and thiazole derivatives have been reported, including antiviral activity and, more recently, as COVID-19 inhibitors. Thus, in this paper, we designed, synthesized, and characterized a novel series of N-aminothiazole-hydrazineethyl-pyridines, beginning with a N'-(1-(pyridine-3-yl)ethylidene)hydrazinecarbothiohydrazide derivative and various hydrazonoyl chlorides and phenacyl bromides. Their Schiff bases were prepared from the condensation of N-aminothiazole derivatives with 4-methoxybenzaldehyde. FTIR, MS, NMR, and elemental studies were used to identify new products. The binding energy for non-bonding interactions between the ligand (studied compounds) and receptor was determined using molecular docking against the SARS-CoV-2 main protease (PDB code: 6LU7). Finally, the best docked pose with highest binding energy (8a = -8.6 kcal/mol) was selected for further molecular dynamics (MD) simulation studies to verify the outcomes and comprehend the thermodynamic properties of the binding. Through additional in vitro and in vivo research on the newly synthesized chemicals, it is envisaged that the achieved results will represent a significant advancement in the fight against COVID-19.
Collapse
|
20
|
Gao X, Xia Y, Liu X, Xu Y, Lu P, dong Z, Liu J, Liang G. A perspective on SARS-CoV-2 virus-like particles vaccines. Int Immunopharmacol 2023; 115:109650. [PMID: 36649673 PMCID: PMC9832101 DOI: 10.1016/j.intimp.2022.109650] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/18/2022] [Accepted: 12/25/2022] [Indexed: 01/13/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) first appeared in Wuhan, China, in December 2019. The 2019 coronavirus disease (COVID-19) pandemic, caused by SARS-CoV-2, has spread to almost all corners of the world at an alarming rate. Vaccination is important for the prevention and control of the COVID-19 pandemic. Efforts are underway worldwide to develop an effective vaccine against COVID-19 using both traditional and innovative vaccine strategies. Compared to other vaccine platforms, SARS-CoV-2 virus-like particles (VLPs )vaccines, as a new vaccine platform, have unique advantages: they have artificial nanostructures similar to natural SARS-CoV-2, which can stimulate good cellular and humoral immune responses in the organism; they have no viral nucleic acids, have good safety and thermal stability, and can be mass-produced and stored; their surfaces can be processed and modified, such as the adjuvant addition, etc.; they can be considered as an ideal platform for COVID-19 vaccine development. This review aims to shed light on the current knowledge and progress of VLPs vaccines against COVID-19, especially those undergoing clinical trials.
Collapse
Affiliation(s)
- Xiaoyang Gao
- Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China,School of Basic Medical Sciences, Henan University of Science & Technology, Luoyang 471023, China
| | - Yeting Xia
- Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Xiaofang Liu
- The First People's Hospital of Nanyang Affiliated to Henan University, Nanyang 473000, China
| | - Yinlan Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Pengyang Lu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Zhipeng dong
- Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Jing Liu
- Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Gaofeng Liang
- School of Basic Medical Sciences, Henan University of Science & Technology, Luoyang 471023, China.
| |
Collapse
|
21
|
Fiorelli D, Francavilla B, Magrini A, Di Girolamo S, Bernardini S, Nuccetelli M. Evaluation of the accuracy in the mucosal detection of anti-SARS-CoV-2 antibodies in nasal secretions and saliva. Int Immunopharmacol 2023; 115:109615. [PMID: 36563531 PMCID: PMC9763213 DOI: 10.1016/j.intimp.2022.109615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/07/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
COVID-19 vaccination with mRNA vaccines induces immune responses capable of neutralizing SARS-CoV-2. Commercially available serological anti-SARS-CoV-2 quantitative and neutralizing assays are essential for the determination of immune responses to vaccines. Nevertheless, at present there is a lack of validated tests to assess the mucosal response to COVID-19 vaccination and standardized analytic and pre-analytic methods have not yet been defined. The aim of our study was to evaluate the accuracy of two diagnostic immunoassays for COVID-19 (ELISA for IgA-S1 and chemiluminescent assay for IgG-RBD) on serum, saliva, and nasal secretions, by the enrollment of three study populations (healthy controls, vaccinated subjects, and subjects recovered from COVID-19 infection). In order to obtain an appropriate cut-off value for the biological matrices studied, ROC curve analyses were performed. Data demonstrate that the analytical and pre-analytical method we have developed can provide accurate and reliable results for the detection of anti-SARS-CoV-2 mucosal specific antibodies (IgA-S1 and IgG-RBD) on saliva and, as a novelty, on nasal secretions, either after COVID-19 infection or in vaccinated subjects.
Collapse
Affiliation(s)
- Denise Fiorelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Beatrice Francavilla
- Department of Otorhinolaryngology, University of Rome "Tor Vergata", Rome, Italy.
| | - Andrea Magrini
- Department of Occupational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Stefano Di Girolamo
- Department of Otorhinolaryngology, University of Rome "Tor Vergata", Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
| |
Collapse
|
22
|
A Bibliometric Visualization Analysis on Vaccine Development of Coronavirus Disease 2019 (COVID-19). Vaccines (Basel) 2023; 11:vaccines11020295. [PMID: 36851173 PMCID: PMC9959778 DOI: 10.3390/vaccines11020295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), beginning in December 2019, has spread worldwide, leading to the death of millions. Owing to the absence of definitive treatment, vaccination against COVID-19 emerged as an effective strategy against the spread of the pandemic. Acceptance of the COVID-19 vaccine has advanced considerably, and vaccine-related research has significantly increased over the past three years. This study aimed to evaluate the content and external characteristics of COVID-19 vaccine-related literature for tracking research trends related to the global COVID-19 vaccine with the means of bibliometrics and visualization maps. A total of 18,285 records in 3499 journals were retrieved in the Web of Science Core Collection database and included in the final analysis. China was the first to focus on COVID-19 vaccine research, while European and American countries started late but developed rapidly. The USA and the UK are the top contributors to COVID-19 vaccine development, with the largest number of publications. The University of Washington and Harvard Medical School were the leading institutions, while Krammer, F. from Icahn School of Medicine at Mount Sinai was the author most active and influential to the topic. The New England Journal of Medicine had the highest number of citations and the highest TLS, and was the most cited and influential journal in the field of COVID-19 vaccine research. COVID-19 vaccine research topics and hotspots focused on populations' attitudes towards vaccination, immunity-related information analysis of spike proteins, the effectiveness and side effects of the COVID-19 vaccine, and the public management of epidemic transmission. The findings of this study provide the global status, research hotspots and potential trends in the field of COVID-19 vaccine research, which will assist researchers in mastering the knowledge structure, and evaluating and guiding future developmental directions of COVID-19 vaccine.
Collapse
|
23
|
Beheshti Shirazi SS, Sakhaee F, Sotoodehnejadnematalahi F, Zamani MS, Ahmadi I, Anvari E, Fateh A. rs12329760 Polymorphism in Transmembrane Serine Protease 2 Gene and Risk of Coronavirus Disease 2019 Mortality. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7841969. [PMID: 36457338 PMCID: PMC9708353 DOI: 10.1155/2022/7841969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/04/2022] [Accepted: 11/12/2022] [Indexed: 08/29/2023]
Abstract
The protease produced by the transmembrane serine protease 2 (TMPRSS2) gene enhances viral infections and has been linked to severe acute respiratory syndrome coronavirus 2 pathogenesis. Therefore, this study evaluated the association between TMPRSS2 and coronavirus disease 2019 (COVID-19) mortality. TMPRSS2 rs12329760 polymorphism was genotyped using the tetraprimer amplification refractory mutation system-polymerase chain reaction method in 592 dead and 693 improved patients. In the current study, the frequency of TMPRSS2 rs12329760 CC than TT genotypes was significantly lower in improved patients than in dead patients. According to the findings of the multivariate logistic regression test, higher levels of mean age, creatinine, erythrocyte sedimentation rate, C-reactive protein, aspartate aminotransferase, lower levels of 25-hydroxyvitamin D, uric acid, and real-time PCR Ct values and TMPRSS2 rs12329760 CC genotype were observed to be associated with increased COVID-19 mortality rates. In conclusion, the TMPRSS2 rs12329760 CC genotype was a polymorphism linked to a significantly higher incidence of severe COVID-19. Further studies are required to corroborate the obtained findings.
Collapse
Affiliation(s)
| | - Fatemeh Sakhaee
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Iraj Ahmadi
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | - Enayat Anvari
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
24
|
Kodchakorn K, Kongtawelert P. Molecular dynamics study on the strengthening behavior of Delta and Omicron SARS-CoV-2 spike RBD improved receptor-binding affinity. PLoS One 2022; 17:e0277745. [PMID: 36395151 PMCID: PMC9671323 DOI: 10.1371/journal.pone.0277745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022] Open
Abstract
The COVID-19 pandemic caused by a virus that can be transmitted from human to human via air droplets has changed the quality of life and economic systems all over the world. The viral DNA has mutated naturally over time leading to the diversity of coronavirus victims which has posed a serious threat to human security on a massive scale. The current variants have developed in a dominant way and are considered "Variants of Concern" by the World Health Organization (WHO). In this work, Kappa (B.1.617.1), Delta (B.1.617.2), and Omicron (B.1.1.529) variants were obtained to evaluate whether naturally occurring mutations have strengthened viral infectivity. We apply reliable in silico structural dynamics and energetic frameworks of the mutated S-RBD protein for ACE2-binding to analyze and compare the structural information related to the wild-type. In particular, the hotspot residues at Q493, Q498, and N501 on the S-RBD protein were determined as contributing factors to the employment stability of the relevant binding interface. The L452R mutation induces an increment of the hydrogen bonds formed by changing the Q493 environment for ACE2 binding. Moreover, the Q493K exchange in Omicron enables the formation of two additional salt bridges, leading to a strong binding affinity by increased electrostatic interaction energy. These results could be used in proposing concrete informative data for a structure-based design engaged in finding better therapeutics against novel variants.
Collapse
Affiliation(s)
- Kanchanok Kodchakorn
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- * E-mail:
| |
Collapse
|
25
|
Park JS, Kim K. Antigen Coverage Presented by MHC Class I Has a Negative Correlation with SARS-CoV-2-Induced Mortality. Vaccines (Basel) 2022; 10:1917. [PMID: 36423013 PMCID: PMC9698063 DOI: 10.3390/vaccines10111917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/29/2022] [Accepted: 11/09/2022] [Indexed: 11/11/2023] Open
Abstract
The COVID-19 pandemic has caused a health crisis worldwide; therefore, it is necessary to understand the factors related to its prognosis. In this study, we hypothesized that SARS-CoV-2-derived antigens presented by MHC class I may correlate with mortality in COVID-19 because they induce adaptive immune responses. Antigen coverage at the national level was inferred using country-specific HLA allele frequencies and relative predictions of binding antigens. We performed regression analysis between antigen coverage and the death rate due to COVID-19 across countries and found a negative correlation, although it was statistically significant only in HLA-B. This negative correlation was corroborated in multiple regression analysis with known risk factors, such as the prevalence of underlying disease. Furthermore, we analyzed antigen coverage in accordance with SARS-CoV-2 domains and identified a significant negative correlation when it was derived from the spike domain, which is reported to be favorable for COVID-19 prognosis. Taken together, the results indicate that the antigen coverage of SARS-CoV-2 specifically presented by HLA-B may act as a favorable factor when explaining COVID-19-induced mortality.
Collapse
Affiliation(s)
- Ji Soo Park
- Department of Biology, Kyung Hee University, Seoul 02447, Korea
| | - Kwoneel Kim
- Department of Biology, Kyung Hee University, Seoul 02447, Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
26
|
徐 偲, 唐 丹. [Progress on the development of the SARS-CoV-2 vaccine and antibody drugs]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2022; 39:1059-1064. [PMID: 36310496 PMCID: PMC9927717 DOI: 10.7507/1001-5515.202207063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/30/2022] [Indexed: 06/16/2023]
Abstract
The raging global epidemic of coronavirus disease 2019 (COVID-19) not only poses a major threat to public health, but also has a huge impact on the global health care system and social and economic development. Therefore, accelerating the development of vaccines and antibody drugs to provide people with effective protection and treatment measures has become the top priority of researchers and medical institutions in the field. At present, several vaccines and antibody drugs targeting SARS-Cov-2 have been in the stage of clinical research or approved for marketing around the world. In this manuscript, we summarized the vaccines and antibody drugs which apply genetic engineering technologies to target spike protein, including subunit vaccines, viral vector vaccines, DNA vaccines, mRNA vaccines, and several neutralizing antibody drugs, and discussed the trends of vaccines and antibody drugs in the future.
Collapse
Affiliation(s)
- 偲瑜 徐
- 四川大学华西第二医院 运营管理部(成都 610065)Operation Management Office, West China Second University Hospital, Chengdu 610065, P. R. China
| | - 丹 唐
- 四川大学华西第二医院 运营管理部(成都 610065)Operation Management Office, West China Second University Hospital, Chengdu 610065, P. R. China
| |
Collapse
|
27
|
Maghsood F, Amiri MM, Zarnani AH, Salimi V, Kardar GA, Khoshnoodi J, Mobini M, Ahmadi Zare H, Ghaderi A, Jeddi-Tehrani M, Schmidt S, Laumond G, Moog C, Shokri F. Epitope mapping of severe acute respiratory syndrome coronavirus 2 neutralizing receptor binding domain-specific monoclonal antibodies. Front Med (Lausanne) 2022; 9:973036. [PMID: 36148457 PMCID: PMC9485472 DOI: 10.3389/fmed.2022.973036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the outbreak led to the coronavirus disease 2019 (COVID-19) pandemic. Receptor binding domain (RBD) of spike (S) protein of SARS-CoV-2 is considered as a major target for immunotherapy and vaccine design. Here, we generated and characterized a panel of anti-RBD monoclonal antibodies (MAbs) isolated from eukaryotic recombinant RBD-immunized mice by hybridoma technology. Epitope mapping was performed using a panel of 20-mer overlapping peptides spanning the entire sequence of the RBD protein from wild-type (WT) Wuhan strain by enzyme-linked immunosorbent assay (ELISA). Several hybridomas showed reactivity toward restricted RBD peptide pools by Pepscan analysis, with more focus on peptides encompassing aa 76-110 and 136-155. However, our MAbs with potent neutralizing activity which block SARS-CoV-2 spike pseudovirus as well as the WT virus entry into angiotensin-converting enzyme-2 (ACE2) expressing HEK293T cells showed no reactivity against these peptides. These findings, largely supported by the Western blotting results suggest that the neutralizing MAbs recognize mainly conformational epitopes. Moreover, our neutralizing MAbs recognized the variants of concern (VOC) currently in circulation, including alpha, beta, gamma, and delta by ELISA, and neutralized alpha and omicron variants at different levels by conventional virus neutralization test (CVNT). While the neutralization of MAbs to the alpha variant showed no substantial difference as compared with the WT virus, their neutralizing activity was lower on omicron variant, suggesting the refractory effect of mutations in emerging variants against this group of neutralizing MAbs. Also, the binding reactivity of our MAbs to delta variant showed a modest decline by ELISA, implying that our MAbs are insensitive to the substitutions in the RBD of delta variant. Our data provide important information for understanding the immunogenicity of RBD, and the potential application of the novel neutralizing MAbs for passive immunotherapy of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Faezeh Maghsood
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholam Ali Kardar
- Immunology, Asthma and Allergy Research Institute, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalal Khoshnoodi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mobini
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hengameh Ahmadi Zare
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Sylvie Schmidt
- Laboratoire d’ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Géraldine Laumond
- Laboratoire d’ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Christiane Moog
- Laboratoire d’ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Ling Y, Dong Y, Li Q, Huang D, Li Z, Chen M. Treatment of COVID-19 by monoclonal antibodies and the traditional Chinese medicine. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022; 15:100159. [PMID: 35937968 PMCID: PMC9343068 DOI: 10.1016/j.medntd.2022.100159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/23/2022] Open
Abstract
The mortality rate of the recent global pandemic corona virus disease 2019 (COVID-19) is currently as high as 7%. The SARS-CoV-2 virus is the culprit behind COVID-19. SARS-CoV-2 is an enveloped single-stranded RNA virus, the genome encodes four types of the structural proteins: S protein, E protein (envelope protein), M protein (matrix protein) and N protein (nucleocapsid protein). In COVID-19, monoclonal antibodies have played a significant role in diagnosis and treatment. This article briefly introduced the development of monoclonal antibodies targeting on S protein and N protein, which represents the main direction of monoclonal antibody drugs used in the diagnosis and treatment of COVID-19. Meanwhile, the traditional Chinese medicine also plays important role in the fight against COVID-19 by regulating human immunity. The article introduced the use of traditional Chinese medicine in fighting against COVID-19.
Collapse
Affiliation(s)
- Yuyan Ling
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuxuan Dong
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qirui Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Doudou Huang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhihui Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Meijuan Chen
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
29
|
Samrat SK, Xu J, Xie X, Gianti E, Chen H, Zou J, Pattis JG, Elokely K, Lee H, Li Z, Klein ML, Shi PY, Zhou J, Li H. Allosteric inhibitors of the main protease of SARS-CoV-2. Antiviral Res 2022; 205:105381. [PMID: 35835291 PMCID: PMC9272661 DOI: 10.1016/j.antiviral.2022.105381] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022]
Abstract
SARS-CoV-2 has raised the alarm to search for effective therapy for this virus. To date several vaccines have been approved but few available drugs reported recently still need approval from FDA. Remdesivir was approved for emergency use only. In this report, the SARS-CoV-2 3CLpro was expressed and purified. By using a FRET-based enzymatic assay, we have screened a library consisting of more than 300 different niclosamide derivatives and identified three molecules JMX0286, JMX0301, and JMX0941 as potent allosteric inhibitors against SARS-CoV-2 3CLpro, with IC50 values similar to that of known covalent inhibitor boceprevir. In a cell-based antiviral assay, these inhibitors can inhibit the virus growth with EC50 in the range of 2-3 μM. The mechanism of action of JMX0286, JMX0301, and JMX0941 were characterized by enzyme kinetics, affinity binding and protein-based substrate digestion. Molecular docking, molecular dynamics (MD) simulations and hydration studies suggested that JMX0286, JMX0301, JMX0941 bind specifically to an allosteric pocket of the SARS-CoV-2 3CL protease. This study provides three potent compounds for further studies.
Collapse
Affiliation(s)
- Subodh Kumar Samrat
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ, 85721-0207, USA.
| | - Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Eleonora Gianti
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA, 19122, USA
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jing Zou
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jason G Pattis
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA, 19122, USA
| | - Khaled Elokely
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA, 19122, USA
| | - Hyun Lee
- Department of Pharmaceutical Sciences at College of Pharmacy and Biophysics Core at Research Resources Center, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Zhong Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ, 85721-0207, USA
| | - Michael L Klein
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA, 19122, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA; Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX, 77555, USA; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ, 85721-0207, USA; BIO5 Institute, The University of Arizona, Tucson, Tucson, AZ, 85721, USA.
| |
Collapse
|
30
|
Wilson B, Geetha KM. Lipid nanoparticles in the development of mRNA vaccines for COVID-19. J Drug Deliv Sci Technol 2022; 74:103553. [PMID: 35783677 PMCID: PMC9238147 DOI: 10.1016/j.jddst.2022.103553] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/25/2022] [Accepted: 06/26/2022] [Indexed: 12/25/2022]
Abstract
Coronavirus disease (COVID-19) was first reported in December 2019, Hubei Province, China. As on 9th December 2021, severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) has affected 266018810 people worldwide with 5265092 deaths. The outbreak of COVID-19 pandemic has caused severe public health crisis across the world. Nucleic acids have been emerging as potential drugs to treat a variety of diseases. Lipid nanoparticles (LNPs) have great potential to deliver nucleic acids including mRNAs. The two mRNA-based vaccines namely the BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna) have been given emergency use authorization (EUA) by the US-FDA to prevent SARS-CoV-2 caused COVID-19 and the vaccines were developed using LNPs. This article focuses on the potential application of LNPs in the development and delivery of mRNA vaccines for COVID-19.
Collapse
Affiliation(s)
- Barnabas Wilson
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Kumaraswamy Layout, Bangalore, Karnataka, 560078, India,Corresponding author
| | - Kannoth Mukundan Geetha
- Department of Pharmacology, College of Pharmaceutical Sciences, Dayananda Sagar University, Kumaraswamy Layout, Bangalore, Karnataka, 560078, India
| |
Collapse
|
31
|
Schindell BG, Allardice M, McBride JA, Dennehy B, Kindrachuk J. SARS-CoV-2 and the Missing Link of Intermediate Hosts in Viral Emergence - What We Can Learn From Other Betacoronaviruses. FRONTIERS IN VIROLOGY 2022; 2. [DOI: 10.3389/fviro.2022.875213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The emergence of SARS-CoV-2 in 2019 has resulted in a global pandemic with devastating human health and economic consequences. The development of multiple vaccines, antivirals and supportive care modalities have aided in our efforts to gain control of the pandemic. However, the emergence of multiple variants of concern and spillover into numerous nonhuman animal species could protract the pandemic. Further, these events also increase the difficulty in simultaneously monitoring viral evolution across multiple species and predicting future spillback potential into the human population. Here, we provide historic context regarding the roles of reservoir and intermediate hosts in coronavirus circulation and discuss current knowledge of these for SARS-CoV-2. Increased understanding of SARS-CoV-2 zoonoses are fundamental for efforts to control the global health and economic impacts of COVID-19.
Collapse
|
32
|
Azali MA, Mohamed S, Harun A, Hussain FA, Shamsuddin S, Johan MF. Application of Baculovirus Expression Vector system (BEV) for COVID-19 diagnostics and therapeutics: a review. J Genet Eng Biotechnol 2022; 20:98. [PMID: 35792966 PMCID: PMC9259773 DOI: 10.1186/s43141-022-00368-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/20/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND The baculovirus expression vector system has been developed for expressing a wide range of proteins, including enzymes, glycoproteins, recombinant viruses, and vaccines. The availability of the SARS-CoV-2 genome sequence has enabled the synthesis of SARS-CoV2 proteins in a baculovirus-insect cell platform for various applications. The most cloned SARS-CoV-2 protein is the spike protein, which plays a critical role in SARS-CoV-2 infection. It is available in its whole length or as subunits like S1 or the receptor-binding domain (RBD). Non-structural proteins (Nsps), another recombinant SARS-CoV-2 protein generated by the baculovirus expression vector system (BEV), are used in the identification of new medications or the repurposing of existing therapies for the treatment of COVID-19. Non-SARS-CoV-2 proteins generated by BEV for SARS-CoV-2 diagnosis or treatment include moloney murine leukemia virus reverse transcriptase (MMLVRT), angiotensin converting enzyme 2 (ACE2), therapeutic proteins, and recombinant antibodies. The recombinant proteins were modified to boost the yield or to stabilize the protein. CONCLUSION This review covers the wide application of the recombinant protein produced using the baculovirus expression technology for COVID-19 research. A lot of improvements have been made to produce functional proteins with high yields. However, there is still room for improvement and there are parts of this field of research that have not been investigated yet.
Collapse
Affiliation(s)
- Muhammad Azharuddin Azali
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- School of Agriculture Science and Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, 22200, Besut, Terengganu, Malaysia
| | - Salmah Mohamed
- School of Agriculture Science and Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, 22200, Besut, Terengganu, Malaysia
| | - Azian Harun
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Faezahtul Arbaeyah Hussain
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Muhammad Farid Johan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
33
|
Homogeneous surrogate virus neutralization assay to rapidly assess neutralization activity of anti-SARS-CoV-2 antibodies. Nat Commun 2022; 13:3716. [PMID: 35778399 PMCID: PMC9249905 DOI: 10.1038/s41467-022-31300-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 06/13/2022] [Indexed: 12/23/2022] Open
Abstract
The COVID-19 pandemic triggered the development of numerous diagnostic tools to monitor infection and to determine immune response. Although assays to measure binding antibodies against SARS-CoV-2 are widely available, more specific tests measuring neutralization activities of antibodies are immediately needed to quantify the extent and duration of protection that results from infection or vaccination. We previously developed a 'Serological Assay based on a Tri-part split-NanoLuc® (SATiN)' to detect antibodies that bind to the spike (S) protein of SARS-CoV-2. Here, we expand on our previous work and describe a reconfigured version of the SATiN assay, called Neutralization SATiN (Neu-SATiN), which measures neutralization activity of antibodies directly from convalescent or vaccinated sera. The results obtained with our assay and other neutralization assays are comparable but with significantly shorter preparation and run time for Neu-SATiN. As the assay is modular, we further demonstrate that Neu-SATiN enables rapid assessment of the effectiveness of vaccines and level of protection against existing SARS-CoV-2 variants of concern and can therefore be readily adapted for emerging variants.
Collapse
|
34
|
Lima Neto JX, Vieira DS, de Andrade J, Fulco UL. Exploring the Spike-hACE 2 Residue-Residue Interaction in Human Coronaviruses SARS-CoV-2, SARS-CoV, and HCoV-NL63. J Chem Inf Model 2022; 62:2857-2868. [PMID: 35617018 PMCID: PMC9159508 DOI: 10.1021/acs.jcim.1c01544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Indexed: 12/20/2022]
Abstract
Coronaviruses (CoVs) have been responsible for three major outbreaks since the beginning of the 21st century, and the emergence of the recent COVID-19 pandemic has resulted in considerable efforts to design new therapies against coronaviruses. Thus, it is crucial to understand the structural features of their major proteins related to the virus-host interaction. Several studies have shown that from the seven known CoV human pathogens, three of them use the human Angiotensin-Converting Enzyme 2 (hACE-2) to mediate their host's cell entry: SARS-CoV-2, SARS-CoV, and HCoV-NL63. Therefore, we employed quantum biochemistry techniques within the density function theory (DFT) framework and the molecular fragmentation with conjugate caps (MFCC) approach to analyze the interactions between the hACE-2 and the spike protein-RBD of the three CoVs in order to map the hot-spot residues that form the recognition surface for these complexes and define the similarities and differences in the interaction scenario. The total interaction energy evaluated showed a good agreement with the experimental binding affinity order: SARS-2 > SARS > NL63. A detailed investigation revealed the energetically most relevant regions of hACE-2 and the spike protein for each complex, as well as the key residue-residue interactions. Our results provide valuable information to deeply understand the structural behavior and binding site characteristics that could help to develop antiviral therapeutics that inhibit protein-protein interactions between CoVs S protein and hACE-2.
Collapse
Affiliation(s)
- José X. Lima Neto
- Departamento de Biofísica e Farmacologia,
Universidade Federal do Rio Grande do Norte, 59072-970
Natal-RN, Brazil
| | - Davi S. Vieira
- Instituto de Química, Universidade
Federal do Rio Grande do Norte, 59072-970 Natal-RN,
Brazil
| | - Jones de Andrade
- Department of Physical Chemistry,
Universidade Federal do Rio Grande do Sul, 91501-970 Porto
Alegre-RS, Brazil
| | - Umberto Laino Fulco
- Departamento de Biofísica e Farmacologia,
Universidade Federal do Rio Grande do Norte, 59072-970
Natal-RN, Brazil
| |
Collapse
|
35
|
Troyano-Hernáez P, Reinosa R, Holguín Á. Evolution of SARS-CoV-2 in Spain during the First Two Years of the Pandemic: Circulating Variants, Amino Acid Conservation, and Genetic Variability in Structural, Non-Structural, and Accessory Proteins. Int J Mol Sci 2022; 23:6394. [PMID: 35742840 PMCID: PMC9223475 DOI: 10.3390/ijms23126394] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Monitoring SARS-CoV-2’s genetic diversity and emerging mutations in this ongoing pandemic is crucial to understanding its evolution and ensuring the performance of COVID-19 diagnostic tests, vaccines, and therapies. Spain has been one of the main epicenters of COVID-19, reaching the highest number of cases and deaths per 100,000 population in Europe at the beginning of the pandemic. This study aims to investigate the epidemiology of SARS-CoV-2 in Spain and its 18 Autonomous Communities across the six epidemic waves established from February 2020 to January 2022. We report on the circulating SARS-CoV-2 variants in each epidemic wave and Spanish region and analyze the mutation frequency, amino acid (aa) conservation, and most frequent aa changes across each structural/non-structural/accessory viral protein among the Spanish sequences deposited in the GISAID database during the study period. The overall SARS-CoV-2 mutation frequency was 1.24 × 10−5. The aa conservation was >99% in the three types of protein, being non-structural the most conserved. Accessory proteins had more variable positions, while structural proteins presented more aa changes per sequence. Six main lineages spread successfully in Spain from 2020 to 2022. The presented data provide an insight into the SARS-CoV-2 circulation and genetic variability in Spain during the first two years of the pandemic.
Collapse
Affiliation(s)
| | | | - África Holguín
- HIV-1 Molecular Epidemiology Laboratory, Microbiology Department and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) in Hospital Universitario Ramón y Cajal, CIBER en Epidemiología y Salud Pública (CIBERESP), Red en Investigación Translacional en Infecciones Pediátricas (RITIP), 28034 Madrid, Spain; (P.T.-H.); (R.R.)
| |
Collapse
|
36
|
Feng W, Xiang Y, Wu L, Chen Z, Li Q, Chen J, Guo Y, Xia D, Chen N, Zhang L, Zhu S, Zhao K. Nucleocapsid protein of SARS-CoV-2 is a potential target for developing new generation of vaccine. J Clin Lab Anal 2022; 36:e24479. [PMID: 35527696 PMCID: PMC9169192 DOI: 10.1002/jcla.24479] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/28/2022] [Accepted: 04/23/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND SARS-CoV-2 has spread worldwide causing more than 400 million people with virus infections since early 2020. Currently, the existing vaccines targeting the spike glycoprotein (S protein) of SARS-CoV-2 are facing great challenge from the infection of SARS-CoV-2 virus and its multiple S protein variants. Thus, we need to develop a new generation of vaccines to prevent infection of the SARS-CoV-2 variants. Compared with the S protein, the nucleocapsid protein (N protein) of SARS-CoV-2 is more conservative and less mutations, which also plays a vital role in viral infection. Therefore, the N protein may have the great potential for developing new vaccines. METHODS The N protein of SARS-CoV-2 was recombinantly expressed and purified in Escherichia coli. Western Blot and ELISA assays were used to demonstrate the immunoreactivity of the recombinant N protein with the serum of 22 COVID-19 patients. We investigated further the response of the specific serum antibodies and cytokine production in BALB/c mice immunized with recombinant N protein by Western Blot and ELISA. RESULTS The N protein had good immunoreactivity and the production of IgG antibody against N protein in COVID-19 patients was tightly correlated with disease severity. Furthermore, the N protein was used to immunize BALB/c mice to have elicited strong immune responses. Not only high levels of IgG antibody, but also cytokine-IFN-γ were produced in the N protein-immunized mice. Importantly, the N protein immunization induced a high level of IgM antibody produced in the mice. CONCLUSION SARS-CoV-2 N protein shows a great big bundle of potentiality for developing a new generation of vaccines in fighting infection of SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Weixu Feng
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Yunru Xiang
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Lianpeng Wu
- Department of Laboratory MedicineThe Sixth People Hospital of WenzhouWenzhouChina
| | - Zhuo Chen
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Qingfeng Li
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Jun Chen
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Yanru Guo
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Dandan Xia
- Department of Laboratory MedicineThe Sixth People Hospital of WenzhouWenzhouChina
| | - Na Chen
- Department of Laboratory MedicineThe Sixth People Hospital of WenzhouWenzhouChina
| | - Lifang Zhang
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Shanli Zhu
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
| | - Kong‐Nan Zhao
- School of Basic Medical ScienceWenzhou Medical UniversityWenzhouChina
- Department of Obstetrics and GynaecologyThe Second Affiliated Hospital and Yuyin Children Hospital of Wenzhou Medical UniversityWenzhouChina
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt LuciaQueenslandAustralia
| |
Collapse
|
37
|
Impact of ChAdOx1 nCoV-19 (Covishield™) Vaccination: How Long Will It Persist? Int J Microbiol 2022; 2022:4729844. [PMID: 35685779 PMCID: PMC9173867 DOI: 10.1155/2022/4729844] [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: 12/09/2021] [Revised: 04/21/2022] [Accepted: 05/18/2022] [Indexed: 12/04/2022] Open
Abstract
An increase in COVID-19 immunization coverage has been linked to a decrease in the average case fatality rate. As a result, further research is needed to determine the persistence and duration of vaccine-induced protective antibodies in order to assess the effectiveness of COVID-19 vaccinations. The present study aimed to determine the COVID-19 IgG antibodies among healthcare workers (HCWs) before and after the ChAdOx1 nCoV-19 (Covishield™) vaccination. A total of 150 HCWs who had received the Covishield™ vaccine were assessed after obtaining written informed consent. Blood samples were drawn at three time points, namely, within one week prior to first dose of vaccination, prior to second dose of vaccination (28–33 days after the first dose of vaccination), and 90–95 days after the second dose of vaccination for detecting neutralizing antibodies, i.e., IgG antibodies by ELISA. The overall baseline seropositivity among the HCWs was found to be 28% (n = 42), assessed by the sample collected prior to first dose of COVID-19 vaccination. The seroconversion rate was reported to be 80% (n = 120) one month after the first dosage and increased to 92.7% (n = 139) three months later. Additionally, there was a significant gradual increase in the IgG concentrations postvaccination in majority of the study participants. In those HCWs who had prior history of SARS-CoV-2 infection, significantly higher antibody level was observed compared to antibody-naive individuals. Fever, pain or swelling at the site of injection, and headache were the most frequently reported adverse events following vaccination among the study participants. Regardless of prior SARS-CoV-2 positivity, two doses of the CovishieldTM vaccine elicited a protective neutralizing antibody response that lasted for three months after the second dose of vaccination.
Collapse
|
38
|
Farid E, Herrera-Uribe J, Stevenson NJ. The Effect of Age, Gender and Comorbidities Upon SARS-CoV-2 Spike Antibody Induction After Two Doses of Sinopharm Vaccine and the Effect of a Pfizer/BioNtech Booster Vaccine. Front Immunol 2022; 13:817597. [PMID: 35711448 PMCID: PMC9197101 DOI: 10.3389/fimmu.2022.817597] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/07/2022] [Indexed: 12/23/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 emerged in China in 2019 and has since travelled the world infecting millions. SARS-CoV-2 causes Corona Virus Disease (COVID-19), that has to date taken over 4 million lives. The Kingdom of Bahrain’s vaccine roll-out has consisted of Sinopharm’s BBIBP-CorV (Sinopharm) and Pfizer/BioNtech’s BNT162b2 (Pfizer/BioNtech). Testing for SARS-CoV-2 anti-Spike (S) antibodies is a useful technique in estimating an individual’s immune protection against the infection. In this study we evaluated S antibody levels by electro-chemiluminescence immunoassay in 379 individuals double vaccinated with Sinopharm and 15 of whom were given a booster with the Pfizer/BioNtech vaccine. Among our double vaccinated cohort, we found a spectrum of S antibody levels. Indeed, we found that a significant proportion of individuals with low S antibody levels had clinical conditions, which were mainly immune-related disorders. Furthermore, a significant proportion of individuals with low S antibody levels were above 50 years of age. Finally, we observed a significant increase in S antibody levels after the Pfizer/BioNtech booster was administered. These findings reveal that while a large proportion of Sinopharm vaccinated individuals did not develop high levels of antibodies against the S protein, a booster dose of the Pfizer/BioNtech vaccine significantly enhances S antibody levels, revealing this “triple dose” vaccination strategy as a useful method of ensuring protective immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Eman Farid
- Department of Microbiology, Immunology and Infectious Diseases, College of Medicine, Arabian Gulf University, Manama, Bahrain
- Department of Pathology, Salmanyia Medical Complex, Government Hospital, Manama, Bahrain
- Laboratory Department, Al Salam Specialist Hospital, Riffa, Bahrain
| | - Juber Herrera-Uribe
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Nigel J. Stevenson
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Viral Immunology Group, Royal College of Surgeons in Ireland (RCSI), Medical College of Bahrain, Busaiteen, Bahrain
- *Correspondence: Nigel J. Stevenson,
| |
Collapse
|
39
|
Altheaby A, Alloqmani D, AlShammari R, Alsuhaibani A, Hakeem A, Alam S, Alharbi S, Al Zunitan M, Bosaeed M, Alharbi NK. Safety and Efficacy of the COVID-19 Vaccine in Kidney Transplant Recipients. Cureus 2022; 14:e24753. [PMID: 35686249 PMCID: PMC9170370 DOI: 10.7759/cureus.24753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 12/14/2022] Open
Abstract
Background: Kidney transplant recipients appear to be at high risk for critical coronavirus disease 2019 (COVID-19) illness. They are considered a priority for COVID-19 vaccination. Only a few studies report on the safety and efficacy of the COVID-19 vaccine in these patients. Methods: In this prospective observational study, we measured anti-severe acute respiratory syndrome coronavirus 2 (anti-SARS-CoV-2) spike-specific IgG post first and second COVID-19 mRNA vaccines in 113 kidney transplant recipients and compared them to 62 healthy volunteers. Result: After the first COVID-19 vaccine, SARS-CoV-2-specific antibodies were undetectable in 38.9% of kidney transplant recipients, and after the second, it remained undetectable in 12.4%. SARS-CoV-2-specific antibodies were significantly lower in kidney transplant recipients. The average antibody titer after the first vaccine was 1243.6±4137.5 in kidney transplant recipients compared to 20012.2±44436.4 in the controls after the first dose (P=0.002), and 7965.5±12431.3 versus 82891.3±67418.7, respectively, after the second dose (P <0.001). The immune response to the COVID-19 vaccine seemed to be influenced by mycophenolate dose in kidney transplant recipients and pre-vaccination infection. Conclusion: Kidney transplant recipients are prone to have attenuated antibody responses (anti-spike IgGs) to mRNA COVID-19 vaccines. Patients on mycophenolate mofetil (2 gm daily) had significantly lower SARS-CoV-2 spike-specific IgG levels as compared to patients on no or reduced dose of mycophenolate. Hence, kidney transplant recipients need to continue all infection control precautionary measures against COVID-19 infection and should be considered a priority for a third COVID-19 vaccine.
Collapse
|
40
|
Martínez L, Malaina I, Salcines-Cuevas D, Terán-Navarro H, Zeoli A, Alonso S, M De la Fuente I, Gonzalez-Lopez E, Ocejo-Vinyals JG, Gozalo-Margüello M, Calvo-Montes J, Alvarez-Dominguez C. First computational design using lambda-superstrings and in vivo validation of SARS-CoV-2 vaccine. Sci Rep 2022; 12:6410. [PMID: 35440789 PMCID: PMC9016385 DOI: 10.1038/s41598-022-09615-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is the greatest threat to global health at the present time, and considerable public and private effort is being devoted to fighting this recently emerged disease. Despite the undoubted advances in the development of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, uncertainty remains about their future efficacy and the duration of the immunity induced. It is therefore prudent to continue designing and testing vaccines against this pathogen. In this article we computationally designed two candidate vaccines, one monopeptide and one multipeptide, using a technique involving optimizing lambda-superstrings, which was introduced and developed by our research group. We tested the monopeptide vaccine, thus establishing a proof of concept for the validity of the technique. We synthesized a peptide of 22 amino acids in length, corresponding to one of the candidate vaccines, and prepared a dendritic cell (DC) vaccine vector loaded with the 22 amino acids SARS-CoV-2 peptide (positions 50-71) contained in the NTD domain (DC-CoVPSA) of the Spike protein. Next, we tested the immunogenicity, the type of immune response elicited, and the cytokine profile induced by the vaccine, using a non-related bacterial peptide as negative control. Our results indicated that the CoVPSA peptide of the Spike protein elicits noticeable immunogenicity in vivo using a DC vaccine vector and remarkable cellular and humoral immune responses. This DC vaccine vector loaded with the NTD peptide of the Spike protein elicited a predominant Th1-Th17 cytokine profile, indicative of an effective anti-viral response. Finally, we performed a proof of concept experiment in humans that included the following groups: asymptomatic non-active COVID-19 patients, vaccinated volunteers, and control donors that tested negative for SARS-CoV-2. The positive control was the current receptor binding domain epitope of COVID-19 RNA-vaccines. We successfully developed a vaccine candidate technique involving optimizing lambda-superstrings and provided proof of concept in human subjects. We conclude that it is a valid method to decipher the best epitopes of the Spike protein of SARS-CoV-2 to prepare peptide-based vaccines for different vector platforms, including DC vaccines.
Collapse
Affiliation(s)
- Luis Martínez
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, 48940, Leioa, Spain. .,BCAM, Basque Center for Applied Mathematics, 48009, Bilbao, Spain.
| | - Iker Malaina
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, 48940, Leioa, Spain.,BioCruces Health Research Institute, Cruces University Hospital, 48903, Barakaldo, Spain
| | | | - Héctor Terán-Navarro
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Andrea Zeoli
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Santos Alonso
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, 48940, Leioa, Spain.,María Goyri Building. Animal Biotechnology Center, University of the Basque Country, UPV/EHU, 48940, Leioa, Spain
| | - Ildefonso M De la Fuente
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, 48940, Leioa, Spain.,Department of Nutrition, CEBAS-CSIC Institute, Espinardo University Campus, 30100, Murcia, Spain
| | - Elena Gonzalez-Lopez
- Servicio de Inmunología, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - J Gonzalo Ocejo-Vinyals
- Servicio de Inmunología, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Mónica Gozalo-Margüello
- Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Jorge Calvo-Montes
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain.,Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain.,CIBER Enfermedades Infecciosas, ISCIII, Madrid, Spain
| | - Carmen Alvarez-Dominguez
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain. .,Universidad Internacional de La Rioja, 26006, Logroño, Spain.
| |
Collapse
|
41
|
Zhao X, Xiong D, Luo S, Duan L. Origin of the tight binding mode to ACE2 triggered by multi-point mutations in the omicron variant: a dynamic insight. Phys Chem Chem Phys 2022; 24:8724-8737. [PMID: 35373810 DOI: 10.1039/d2cp00449f] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The continuous spread of the newly emerged SARS-CoV-2 Omicron variant (B.1.1.529) has become an important reason for the surge in COVID-19 infections. Its numerous mutated residues containing key sites on the receptor-binding domain (RBD) undoubtedly pose new challenges for epidemic control. Although the preventive measures are becoming more sophisticated, the effects of mutations on the binding of the virus to the receptor protein remain to be elucidated. Here, we used molecular dynamics (MD) simulations to investigate the differences in the binding mode between the Omicron variant and the angiotensin-converting enzyme 2 (ACE2) compared to the wild-type strain (WT). Multi-point mutations in the Omicron variant RBD could cause the conformation shift in the large Loop (where T478K and E484A are located), which makes it easier to wrap the N-terminal helix of ACE2 and form tighter contacts. The stronger electrostatic interaction was the main reason for its enhanced binding affinity as compared to WT. This was due to the large number of positively charged patches (N440K, T478K, Q493R, Q498R, and Y505H) formed by the substitution of neutral amino acids at multiple sites. The appearance of these highly polar hydrophilic amino acids may cause local perturbations and affect the electrostatic complementarity of RBD with the ACE2, and further mediate conformational changes. Thus, a more extensive interaction network was found in the mutation system and the complex interaction cluster was formed near E37@ACE2, which was essential for the stable binding of the two. In addition, we speculated that these mutations may affect the electrostatic complementarity with the four potential antibodies to reduce the sensitivity of the virus to antibodies. This study reveals the key details of the Omicron variant binding to ACE2 and provides important theoretical views for the enhanced infectivity of this variant. We hope that these observations can provide timely molecular insights for responding to the Omicron variant pandemic.
Collapse
Affiliation(s)
- Xiaoyu Zhao
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China.
| | - Danyang Xiong
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China.
| | - Song Luo
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China.
| | - Lili Duan
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
42
|
Ventura Fernandes BH, Feitosa NM, Barbosa AP, Bomfim CG, Garnique AMB, Rosa IF, Rodrigues MS, Doretto LB, Costa DF, Camargo-Dos-Santos B, Franco GA, Neto JF, Lunardi JS, Bellot MS, Alves NPC, Costa CC, Aracati MF, Rodrigues LF, Costa CC, Cirilo RH, Colagrande RM, Gomes FIF, Nakajima RT, Belo MAA, Giaquinto PC, de Oliveira SL, Eto SF, Fernandes DC, Manrique WG, Conde G, Rosales RRC, Todeschini I, Rivero I, Llontop E, Sgro GG, Oka GU, Bueno NF, Ferraris FK, de Magalhães MTQ, Medeiros RJ, Mendonça-Gomes JM, Junqueira MS, Conceição K, Pontes LGD, Condino-Neto A, Perez AC, Barcellos LJG, Júnior JDC, Dorlass EG, Camara NOS, Durigon EL, Cunha FQ, Nóbrega RH, Machado-Santelli GM, Farah CS, Veras FP, Galindo-Villegas J, Costa-Lotufo LV, Cunha TM, Chammas R, Carvalho LR, Guzzo CR, Malafaia G, Charlie-Silva I. Toxicity of spike fragments SARS-CoV-2 S protein for zebrafish: A tool to study its hazardous for human health? THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 813:152345. [PMID: 34942250 PMCID: PMC8688160 DOI: 10.1016/j.scitotenv.2021.152345] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/17/2021] [Accepted: 12/08/2021] [Indexed: 05/19/2023]
Abstract
Despite the significant increase in the generation of SARS-CoV-2 contaminated domestic and hospital wastewater, little is known about the ecotoxicological effects of the virus or its structural components in freshwater vertebrates. In this context, this study evaluated the deleterious effects caused by SARS-CoV-2 Spike protein on the health of Danio rerio, zebrafish. We demonstrated, for the first time, that zebrafish injected with fragment 16 to 165 (rSpike), which corresponds to the N-terminal portion of the protein, presented mortalities and adverse effects on liver, kidney, ovary and brain tissues. The conserved genetic homology between zebrafish and humans might be one of the reasons for the intense toxic effects followed inflammatory reaction from the immune system of zebrafish to rSpike which provoked damage to organs in a similar pattern as happen in severe cases of COVID-19 in humans, and, resulted in 78,6% of survival rate in female adults during the first seven days. The application of spike protein in zebrafish was highly toxic that is suitable for future studies to gather valuable information about ecotoxicological impacts, as well as vaccine responses and therapeutic approaches in human medicine. Therefore, besides representing an important tool to assess the harmful effects of SARS-CoV-2 in the aquatic environment, we present the zebrafish as an animal model for translational COVID-19 research.
Collapse
Affiliation(s)
- Bianca H Ventura Fernandes
- Laboratório de Controle Genético e Sanitário, Diretoria Técnica de Apoio ao Ensino e Pesquisa, Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Natália Martins Feitosa
- Laboratório Integrado de Biociências Translacionais (LIBT), Instituto de Biodiversidade e Sustentabilidade (NUPEM), Universidade Federal do Rio de Janeiro (UFRJ), Macaé, RJ, Brazil
| | - Ana Paula Barbosa
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Camila Gasque Bomfim
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Anali M B Garnique
- Department of Cell Biology, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | - Ivana F Rosa
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, Sao Paulo State University, Botucatu, São Paulo, Brazil
| | - Maira S Rodrigues
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, Sao Paulo State University, Botucatu, São Paulo, Brazil
| | - Lucas B Doretto
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, Sao Paulo State University, Botucatu, São Paulo, Brazil
| | - Daniel F Costa
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, Sao Paulo State University, Botucatu, São Paulo, Brazil
| | - Bruno Camargo-Dos-Santos
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University, SP, Brazil
| | - Gabrielli A Franco
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University, SP, Brazil
| | - João Favero Neto
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University, SP, Brazil
| | - Juliana Sartori Lunardi
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University, SP, Brazil
| | - Marina Sanson Bellot
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University, SP, Brazil
| | - Nina Pacheco Capelini Alves
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University, SP, Brazil
| | - Camila C Costa
- Department of Preventive Veterinary Medicine, São Paulo State University (UNESP), Jaboticabal, Brazil
| | - Mayumi F Aracati
- Department of Preventive Veterinary Medicine, São Paulo State University (UNESP), Jaboticabal, Brazil
| | - Letícia F Rodrigues
- Department of Preventive Veterinary Medicine, São Paulo State University (UNESP), Jaboticabal, Brazil
| | - Camila C Costa
- Department of Preventive Veterinary Medicine, São Paulo State University (UNESP), Jaboticabal, Brazil
| | - Rafaela Hemily Cirilo
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University, SP, Brazil
| | - Raul Marcelino Colagrande
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University, SP, Brazil
| | - Francisco I F Gomes
- Department of Pharmacology, Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Rafael T Nakajima
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, Sao Paulo State University, Botucatu, São Paulo, Brazil
| | | | - Percília Cardoso Giaquinto
- Universidade Estadual Paulista Júlio de Mesquita Filho, Instituto de Biociências - Departamento de Fisiologia, São Paulo, Brazil
| | | | - Silas Fernandes Eto
- Postgraduate Program in Health Sciences, PROCISA, Federal University of Roraima, Brazil
| | | | - Wilson G Manrique
- Aquaculture Health Research and Extension Group, GRUPESA, Aquaculture Health Laboratory, LABSA, Department of Veterinary Medicine, Federal University of Rondônia, Rolim de Moura campus, Rondônia, Brazil
| | - Gabriel Conde
- Department of Preventive Veterinary Medicine, São Paulo State University, Jaboticabal, Brazil
| | - Roberta R C Rosales
- Department of Cell and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Iris Todeschini
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil
| | - Ilo Rivero
- Pontifícia Universidade Católica de Minas Gerais, Brazil
| | - Edgar Llontop
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil
| | - Germán G Sgro
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil; Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Gabriel Umaji Oka
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil
| | | | - Fausto K Ferraris
- Department of Pharmacology and Toxicology, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - Mariana T Q de Magalhães
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Renata J Medeiros
- Laboratory of Physiology, INCQS/Fiocruz Zebrafish Facility, Departament of Pharmacology and Toxicology, National Institute for Quality Control in Health, Brasil
| | - Juliana M Mendonça-Gomes
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, Universidade de Sao Paulo, Brazil
| | - Mara Souza Junqueira
- Center for Translational Research in Oncology, Cancer Institute of the State of Sao Paulo, Faculty of Medicine, University of São Paulo, Sao Paulo, Brazil
| | - Kátia Conceição
- Laboratory of Peptide Biochemistry, Federal University of São Paulo, Brazil
| | - Leticia Gomes de Pontes
- Laboratory of Human Immunology, Department Immunology, Institute Biomedical Sciences, University São Paulo, Sao Paulo, Brazil
| | - Antonio Condino-Neto
- Laboratory of Human Immunology, Department Immunology, Institute Biomedical Sciences, University São Paulo, Sao Paulo, Brazil
| | - Andrea C Perez
- Department of Pharmacology, Universidade Federal de Minas Gerais, Brazil
| | - Leonardo J G Barcellos
- Graduate Program of Pharmacology, Federal University of Santa Maria, Brazil; Laboratory of Fish Physiology, Graduate Program of Bioexperimentation and of Environmental Sciences, University of Passo Fundo, Brazil
| | - José Dias Correa Júnior
- Laboratório do Estudo da Interação Químico Biológica e da Reprodução Animal, LIQBRA, Bloco O3,174, Brazil; Departamento de Morfologia Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - Erick Gustavo Dorlass
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Niels O S Camara
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, Universidade de Sao Paulo, Brazil
| | - Edison Luiz Durigon
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Center of Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rafael H Nóbrega
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, Sao Paulo State University, Botucatu, São Paulo, Brazil
| | - Glaucia M Machado-Santelli
- Department of Cell Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Chuck S Farah
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil
| | - Flavio P Veras
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil; Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | | | - Letícia V Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, Brazil
| | - Thiago M Cunha
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil; Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Luciani R Carvalho
- Disciplina de Endocrinologia do Departamento de Clinica Medica e Laboratório de Hormônios e Genética Molecular, LIM 42, Brazil
| | - Cristiane R Guzzo
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Guilherme Malafaia
- Biological Research Laboratory, Goiano Federal Institute, Urutaí Campus, Brazil.
| | - Ives Charlie-Silva
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, Brazil.
| |
Collapse
|
43
|
Bahoussi AN, Guo YY, Shi RZ, Wang PH, Li YQ, Wu CX, Xing L. Genetic Characteristics of Porcine Hemagglutinating Encephalomyelitis Coronavirus: Identification of Naturally Occurring Mutations Between 1970 and 2015. Front Microbiol 2022; 13:860851. [PMID: 35369458 PMCID: PMC8971845 DOI: 10.3389/fmicb.2022.860851] [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: 01/23/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) is a Betacoronavirus characterized by neurological symptoms and a worldwide prevalence. Although PHEV is one of the earliest discovered porcine coronaviruses, it remains poorly studied. The full-length genome of the earliest PHEV strain collected in 1970 in the United States (PHEV/67 N/US/1970) was determined in October 2020. Using this virus as a prototype, we comparatively analyzed all available PHEV full-length sequences during 1970–2015. In phylogenetic trees based on PHEV full-length or spike glycoprotein open reading frame genomic sequences, PHEV/67 N/US/1970 was sorted into a clade different from that of viruses isolated in the United States in 2015. Intriguingly, United States and Belgium viruses isolated in 2015 and 2005, respectively, revealed multiple deletion mutation patterns compared to the strain PHEV/67 N/US/1970, leading to a truncated or a non-functional NS2A coding region. In addition, the genomic similarity analysis showed a hypervariability of the spike glycoprotein coding region, which can affect at least eight potential linear B cell epitopes located in the spike glycoprotein. This report indicates that PHEVs in the United States underwent a significant genetic drift, which might influence PHEV surveillance in other countries.
Collapse
Affiliation(s)
| | - Yan-Yan Guo
- Institute of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Rui-Zhu Shi
- Institute of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Pei-Hua Wang
- Institute of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Ya-Qian Li
- Institute of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Chang-Xin Wu
- Institute of Biomedical Sciences, Shanxi University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan, China
- Shanxi Provincial Key Laboratory for Prevention and Treatment of Major Infectious Diseases, Taiyuan, China
| | - Li Xing
- Institute of Biomedical Sciences, Shanxi University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan, China
- Shanxi Provincial Key Laboratory for Prevention and Treatment of Major Infectious Diseases, Taiyuan, China
- *Correspondence: Li Xing,
| |
Collapse
|
44
|
Wu WL, Chiang CY, Lai SC, Yu CY, Huang YL, Liao HC, Liao CL, Chen HW, Liu SJ. Monoclonal antibody targeting the conserved region of the SARS-CoV-2 spike protein to overcome viral variants. JCI Insight 2022; 7:157597. [PMID: 35290246 PMCID: PMC9089791 DOI: 10.1172/jci.insight.157597] [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: 12/15/2021] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
Most therapeutic mAbs target the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2. Unfortunately, the RBD is a hot spot for mutations in SARS-CoV-2 variants, which will lead to loss of the neutralizing function of current therapeutic mAbs. Universal mAbs for different variants are necessary. We identified mAbs that recognized the S2 region of the spike protein, which is identical in different variants. The mAbs could neutralize SARS-CoV-2 infection and protect animals from SARS-CoV-2 challenge. After cloning the variable region of the light chain and heavy chain, the variable region sequences were humanized to select a high-affinity humanized mAb, hMab5.17. hMab5.17 protected animals from SARS-CoV-2 challenge and neutralized SARS-CoV-2 variant infection. We further identified the linear epitope of the mAb, which is not mutated in any variant of concern. These data suggest that a mAb recognizing the S2 region of the spike protein will be a potential universal therapeutic mAb for COVID-19.
Collapse
Affiliation(s)
- Wan-Ling Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chen-Yi Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Szu-Chia Lai
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei, Taiwan
| | - Chia-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Ling Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
45
|
Advances in purification of SARS-CoV-2 spike ectodomain protein using high-throughput screening and non-affinity methods. Sci Rep 2022; 12:4458. [PMID: 35292666 PMCID: PMC8923338 DOI: 10.1038/s41598-022-07485-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/17/2021] [Indexed: 12/23/2022] Open
Abstract
The spike (S) glycoprotein of the pandemic virus, SARS-CoV-2, is a critically important target of vaccine design and therapeutic development. A high-yield, scalable, cGMP-compliant downstream process for the stabilized, soluble, native-like S protein ectodomain is necessary to meet the extensive material requirements for ongoing research and development. As of June 2021, S proteins have exclusively been purified using difficult-to-scale, low-yield methodologies such as affinity and size-exclusion chromatography. Herein we present the first known non-affinity purification method for two S constructs, S_dF_2P and HexaPro, expressed in the mammalian cell line, CHO-DG44. A high-throughput resin screen on the Tecan Freedom EVO200 automated bioprocess workstation led to identification of ion exchange resins as viable purification steps. The chromatographic unit operations along with industry-standard methodologies for viral clearances, low pH treatment and 20 nm filtration, were assessed for feasibility. The developed process was applied to purify HexaPro from a CHO-DG44 stable pool harvest and yielded the highest yet reported amount of pure S protein. Our results demonstrate that commercially available chromatography resins are suitable for cGMP manufacturing of SARS-CoV-2 Spike protein constructs. We anticipate our results will provide a blueprint for worldwide biopharmaceutical production laboratories, as well as a starting point for process intensification.
Collapse
|
46
|
Liu J, Kan M, Zhang L, Yue Y, Wang S, Hong M, Hong X. Rapid Degradation of SARS-CoV-2 Spike S Protein by A Specific Serine Protease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061882. [PMID: 35335246 PMCID: PMC8954242 DOI: 10.3390/molecules27061882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022]
Abstract
The S protein of SARS-CoV-2 is a crucial structural and functional component for virus entry. Due to the constant mutation of the virus, there are very limited ways to prevent and control COVID-19. This experiment used a macroscopic SDS-PAGE method and proved that the S protein of wild-type SARS-CoV-2 virus, especially the S1 subunit, is very sensitive to alkaline serine protease with acidic pI (ASPNJ), NJ represents Neanthes japonica (Izuka) from which ASP is purified). ASPNJ cleaves proteins when the carbonyl group of the peptide bond is contributed by arginine or lysine. ASPNJ can degrade the S protein very quickly and effectively in vitro with relative selectivity. It can be inferred that the S, S1 and RBD of SARS-CoV-2 variants can also be easily degraded by ASPNJ. This rapid and strong degradation of the S protein by ASPNJ may become a potential new treatment strategy.
Collapse
Affiliation(s)
- Jiankai Liu
- Biochemistry Department, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (J.L.); (M.K.); (L.Z.); (M.H.)
| | - Mujie Kan
- Biochemistry Department, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (J.L.); (M.K.); (L.Z.); (M.H.)
| | - Lianzhi Zhang
- Biochemistry Department, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (J.L.); (M.K.); (L.Z.); (M.H.)
| | - Yuan Yue
- Laboratory of Medical Biology Center, College of Basic Medical Sciences, Jilin University, Changchun 130021, China;
| | - Shaohua Wang
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun 130021, China;
| | - Min Hong
- Biochemistry Department, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (J.L.); (M.K.); (L.Z.); (M.H.)
| | - Xinyu Hong
- Neuroscience Research Laboratory, Neurosurgery Department, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
- Correspondence: ; Tel.: +86-135-9640-0500
| |
Collapse
|
47
|
Vo GV, Bagyinszky E, An SSA. COVID-19 Genetic Variants and Their Potential Impact in Vaccine Development. Microorganisms 2022; 10:598. [PMID: 35336173 PMCID: PMC8954257 DOI: 10.3390/microorganisms10030598] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 01/18/2023] Open
Abstract
In the two years since the SARS-CoV-2 pandemic started, it has caused over 5 million deaths and 400 million infected cases, and the world continues to be on high alert for COVID-19. Among the variants of interest and concern of SARS-CoV-2, the current Omicron (B.1.1.529) and stealth Omicron (BA.2) raised serious concerns due to rapid rates of infection caused by numerous mutations in the spike protein, which could escape from the antibody-mediated neutralization and increase the risk of reinfections. Hence, this work aims to describe the most relevant mutations in the SARS-CoV-2 spike protein, discuss vaccine against variant of concerns, describe rare adverse events after COVID-19 vaccination, introduce the most available promising COVID-19 vaccine candidates, and provide few perspectives of the future variants.
Collapse
Affiliation(s)
- Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City 70000, Vietnam;
- Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University, Ho Chi Minh City (VNU-HCM), Ho Chi Minh City 70000, Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City 70000, Vietnam
| | - Eva Bagyinszky
- Graduate School of Environment Department of Industrial and Environmental Engineering, Gachon University, Seongnam 13120, Korea
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon University, Seongnam 13120, Korea
| |
Collapse
|
48
|
Beaudoin CA, Bartas M, Volná A, Pečinka P, Blundell TL. Are There Hidden Genes in DNA/RNA Vaccines? Front Immunol 2022; 13:801915. [PMID: 35211117 PMCID: PMC8860813 DOI: 10.3389/fimmu.2022.801915] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/14/2022] [Indexed: 02/02/2023] Open
Abstract
Due to the fast global spreading of the Severe Acute Respiratory Syndrome Coronavirus - 2 (SARS-CoV-2), prevention and treatment options are direly needed in order to control infection-related morbidity, mortality, and economic losses. Although drug and inactivated and attenuated virus vaccine development can require significant amounts of time and resources, DNA and RNA vaccines offer a quick, simple, and cheap treatment alternative, even when produced on a large scale. The spike protein, which has been shown as the most antigenic SARS-CoV-2 protein, has been widely selected as the target of choice for DNA/RNA vaccines. Vaccination campaigns have reported high vaccination rates and protection, but numerous unintended effects, ranging from muscle pain to death, have led to concerns about the safety of RNA/DNA vaccines. In parallel to these studies, several open reading frames (ORFs) have been found to be overlapping SARS-CoV-2 accessory genes, two of which, ORF2b and ORF-Sh, overlap the spike protein sequence. Thus, the presence of these, and potentially other ORFs on SARS-CoV-2 DNA/RNA vaccines, could lead to the translation of undesired proteins during vaccination. Herein, we discuss the translation of overlapping genes in connection with DNA/RNA vaccines. Two mRNA vaccine spike protein sequences, which have been made publicly-available, were compared to the wild-type sequence in order to uncover possible differences in putative overlapping ORFs. Notably, the Moderna mRNA-1273 vaccine sequence is predicted to contain no frameshifted ORFs on the positive sense strand, which highlights the utility of codon optimization in DNA/RNA vaccine design to remove undesired overlapping ORFs. Since little information is available on ORF2b or ORF-Sh, we use structural bioinformatics techniques to investigate the structure-function relationship of these proteins. The presence of putative ORFs on DNA/RNA vaccine candidates implies that overlapping genes may contribute to the translation of smaller peptides, potentially leading to unintended clinical outcomes, and that the protein-coding potential of DNA/RNA vaccines should be rigorously examined prior to administration.
Collapse
Affiliation(s)
- Christopher A. Beaudoin
- Department of Biochemistry, Sanger Building, University of Cambridge, Cambridge, United Kingdom
| | - Martin Bartas
- Department of Biology and Ecology, University of Ostrava, Ostrava, Czechia
| | - Adriana Volná
- Department of Physics, University of Ostrava, Ostrava, Czechia
| | - Petr Pečinka
- Department of Biology and Ecology, University of Ostrava, Ostrava, Czechia
| | - Tom L. Blundell
- Department of Biochemistry, Sanger Building, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
49
|
Jin Y, Hou C, Li Y, Zheng K, Wang C. mRNA Vaccine: How to Meet the Challenge of SARS-CoV-2. Front Immunol 2022; 12:821538. [PMID: 35126377 PMCID: PMC8813741 DOI: 10.3389/fimmu.2021.821538] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with high infectivity, pathogenicity, and variability, is a global pandemic that severely affected public health and the world economy. The development of safe and effective vaccines is crucial to the prevention and control of an epidemic. As an emerging technology, mRNA vaccine is widely used for infectious disease prevention and control and has significant safety, efficacy, and high production. It has received support and funding from many pharmaceutical enterprises and becomes one of the main technologies for preventing COVID-19. This review introduces the current status of SARS-CoV-2 vaccines, specifically mRNA vaccines, focusing on the challenges of developing mRNA vaccines against SARS-CoV-2, and discusses the relevant strategies.
Collapse
Affiliation(s)
- Yingqi Jin
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
| | - Chen Hou
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
| | - Yonghao Li
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
| | - Kang Zheng
- Department of Clinical Laboratory, Hengyang Central Hospital, Hengyang, China
| | - Chuan Wang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
50
|
Tariq H, Batool S, Asif S, Ali M, Abbasi BH. Virus-Like Particles: Revolutionary Platforms for Developing Vaccines Against Emerging Infectious Diseases. Front Microbiol 2022; 12:790121. [PMID: 35046918 PMCID: PMC8761975 DOI: 10.3389/fmicb.2021.790121] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Virus-like particles (VLPs) are nanostructures that possess diverse applications in therapeutics, immunization, and diagnostics. With the recent advancements in biomedical engineering technologies, commercially available VLP-based vaccines are being extensively used to combat infectious diseases, whereas many more are in different stages of development in clinical studies. Because of their desired characteristics in terms of efficacy, safety, and diversity, VLP-based approaches might become more recurrent in the years to come. However, some production and fabrication challenges must be addressed before VLP-based approaches can be widely used in therapeutics. This review offers insight into the recent VLP-based vaccines development, with an emphasis on their characteristics, expression systems, and potential applicability as ideal candidates to combat emerging virulent pathogens. Finally, the potential of VLP-based vaccine as viable and efficient immunizing agents to induce immunity against virulent infectious agents, including, SARS-CoV-2 and protein nanoparticle-based vaccines has been elaborated. Thus, VLP vaccines may serve as an effective alternative to conventional vaccine strategies in combating emerging infectious diseases.
Collapse
Affiliation(s)
- Hasnat Tariq
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sannia Batool
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saaim Asif
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Mohammad Ali
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | | |
Collapse
|