1
|
Arias-Arias JL, Monturiol-Gross L, Corrales-Aguilar E. A Live-Cell Imaging-Based Fluorescent SARS-CoV-2 Neutralization Assay by Antibody-Mediated Blockage of Receptor Binding Domain-ACE2 Interaction. BIOTECH 2025; 14:10. [PMID: 39982277 PMCID: PMC11843899 DOI: 10.3390/biotech14010010] [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: 12/12/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Neutralization assays have become an important tool since the beginning of the COVID-19 pandemic for testing vaccine responses and therapeutic antibodies as well as for monitoring humoral immunity to SARS-CoV-2 in epidemiological studies. The spike glycoprotein (S) present on the viral surface contains a receptor binding domain (RBD) that recognizes the angiotensin-converting enzyme 2 receptor (ACE2) in host cells, allowing virus entry. The gold standard for determining SARS-CoV-2 neutralizing antibodies is the plaque reduction neutralization test (PRNT), which relies on live-virus replication performed exclusively in biosafety level 3 (BSL-3) laboratories. Here, we report the development of a surrogate live-cell imaging-based fluorescent SARS-CoV-2 neutralization assay, applicable to BSL-1 or BSL-2 laboratories, by antibody-mediated blockage of the interaction between recombinant RBD with overexpressed ACE2 receptor in a genetically modified HEK 293T stable cell line. Our approach was able to detect neutralizing antibodies both in COVID-19-positive human serum samples and polyclonal equine formulations against SARS-CoV-2. This new cell-based surrogate neutralization assay represents a virus-free fluorescence imaging alternative to the reported approaches, which can be used to detect antibody-neutralizing capabilities toward SARS-CoV-2. This assay could also be extrapolated in the future to other established and emergent viral agents.
Collapse
Affiliation(s)
- Jorge L. Arias-Arias
- Centro de Investigación en Enfermedades Tropicales (CIET), Facultad de Microbiología, Universidad de Costa Rica, San José 11501-2060, Costa Rica;
- Dulbecco Lab Studio, Residencial Lisboa 2G, Alajuela 20102, Costa Rica
| | - Laura Monturiol-Gross
- Instituto Clodomiro Picado (ICP), Facultad de Microbiología, Universidad de Costa Rica, San José 11501-2060, Costa Rica;
| | - Eugenia Corrales-Aguilar
- Centro de Investigación en Enfermedades Tropicales (CIET), Facultad de Microbiología, Universidad de Costa Rica, San José 11501-2060, Costa Rica;
| |
Collapse
|
2
|
Tominaga M, Shima Y, Nozaki K, Ito Y, Someda M, Shoya Y, Hashii N, Obata C, Matsumoto-Kitano M, Suematsu K, Matsukawa T, Hosoya K, Hashiba N, Kondo A, Ishii J. Designing strong inducible synthetic promoters in yeasts. Nat Commun 2024; 15:10653. [PMID: 39702268 PMCID: PMC11659477 DOI: 10.1038/s41467-024-54865-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
Inducible promoters are essential for precise control of target gene expression in synthetic biological systems. However, engineering eukaryotic promoters is often more challenging than engineering prokaryotic promoters due to their greater mechanistic complexity. In this study, we describe a simple and reliable approach for constructing strongly inducible synthetic promoters with minimum leakiness in yeasts. The results indicate that the leakiness of yeast-inducible synthetic promoters is primarily the result of cryptic transcriptional activation of heterologous sequences that may be avoided by appropriate insulation and operator mutagenesis. Our promoter design approach has successfully generated robust, inducible promoters that achieve a > 103-fold induction in reporter gene expression. The utility of these promoters is demonstrated by using them to produce various biologics with titers up to 2 g/L, including antigens designed to raise specific antibodies against a SARS-CoV-2 omicron variant through chicken immunization.
Collapse
Affiliation(s)
- Masahiro Tominaga
- Engineering Biology Research Center, Kobe University, Kobe, Japan
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Yoko Shima
- Engineering Biology Research Center, Kobe University, Kobe, Japan
| | - Kenta Nozaki
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Yoichiro Ito
- Engineering Biology Research Center, Kobe University, Kobe, Japan
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | | | - Yuji Shoya
- Pharma Foods International Co. Ltd., Kyoto, Japan
| | - Noritaka Hashii
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Chihiro Obata
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | | | - Kohei Suematsu
- Engineering Biology Research Center, Kobe University, Kobe, Japan
| | | | - Keita Hosoya
- Engineering Biology Research Center, Kobe University, Kobe, Japan
| | - Noriko Hashiba
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Akihiko Kondo
- Engineering Biology Research Center, Kobe University, Kobe, Japan
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
- Department of Chemical Science and Engineering, Faculty of Engineering, Kobe University, Kobe, Japan
- Center for Sustainable Resource Science, RIKEN, Yokohama, Japan
| | - Jun Ishii
- Engineering Biology Research Center, Kobe University, Kobe, Japan.
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan.
| |
Collapse
|
3
|
Sookhoo JRV, Schiffman Z, Ambagala A, Kobasa D, Pardee K, Babiuk S. Protein Expression Platforms and the Challenges of Viral Antigen Production. Vaccines (Basel) 2024; 12:1344. [PMID: 39772006 PMCID: PMC11680109 DOI: 10.3390/vaccines12121344] [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: 09/20/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Several protein expression platforms exist for a wide variety of biopharmaceutical needs. A substantial proportion of research and development into protein expression platforms and their optimization since the mid-1900s is a result of the production of viral antigens for use in subunit vaccine research. This review discusses the seven most popular forms of expression systems used in the past decade-bacterial, insect, mammalian, yeast, algal, plant and cell-free systems-in terms of advantages, uses and limitations for viral antigen production in the context of subunit vaccine research. Post-translational modifications, immunogenicity, efficacy, complexity, scalability and the cost of production are major points discussed. Examples of licenced and experimental vaccines are included along with images which summarize the processes involved.
Collapse
Affiliation(s)
- Jamie R. V. Sookhoo
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3R2, Canada; (J.R.V.S.); (A.A.)
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Zachary Schiffman
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (Z.S.); (D.K.)
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Aruna Ambagala
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3R2, Canada; (J.R.V.S.); (A.A.)
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (Z.S.); (D.K.)
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Keith Pardee
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada;
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3R2, Canada; (J.R.V.S.); (A.A.)
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
4
|
Grabiński W, Karachitos A, Kicińska A. Backstage Heroes-Yeast in COVID-19 Research. Int J Mol Sci 2024; 25:12661. [PMID: 39684373 PMCID: PMC11640846 DOI: 10.3390/ijms252312661] [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: 10/01/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The extremely rapid development of understanding and technology that led to the containment of the COVID-19 pandemic resulted from collaborative efforts in the fields of Betacoronavirus pandemicum (SARS-CoV-2) biology, pharmacology, vaccinology, and medicine. Perhaps surprisingly, much of the research was conducted using simple and efficient yeast models. In this manuscript, we describe how yeast, eukaryotic microorganisms, have been used to research this global challenge, focusing on the therapeutic potential of the studies discussed herein. Thus, we outline the role of yeast in studying viral protein interactions with the host cell proteome, including the binding of the SARS-CoV-2 virus spike protein to the human ACE2 receptor and its modulation. The production and exploration of viral antigens in yeast systems, which led to the development of two approved COVID-19 vaccines, are also detailed. Moreover, yeast platforms facilitating the discovery and production of single-domain antibodies (nanobodies) against SARS-CoV-2 are described. Methods guiding modern and efficient drug discovery are explained at length. In particular, we focus on studies designed to search for inhibitors of the main protease (Mpro), a unique target for anti-coronaviral therapies. We highlight the adaptability of the techniques used, providing opportunities for rapid modification and implementation alongside the evolution of the SARS-CoV-2 virus. Approaches introduced in yeast systems that may have universal potential application in studies of emerging viral diseases are also described.
Collapse
Affiliation(s)
| | | | - Anna Kicińska
- Department of Bioenergetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznań, Poland; (W.G.); (A.K.)
| |
Collapse
|
5
|
Prabhala SV, Marshall B, Galiardi J, Fan Y, Creamer E, Wood DW. Highly selective split intein method for efficient separation and purification of recombinant therapeutic proteins from mammalian cell culture fluid. J Chromatogr A 2024; 1736:465430. [PMID: 39405639 PMCID: PMC11533640 DOI: 10.1016/j.chroma.2024.465430] [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: 06/21/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Biologics and vaccines have been successfully developed over the last few decades to treat many diseases. Each of these drugs must be highly purified for clinical use. Monoclonal antibodies (mAbs), the dominant therapeutic modality on the market, can be easily purified using the standard Protein A affinity platform. However, no generally applicable affinity platforms are available for the manufacture of other therapeutic proteins for clinical use. Thus, multicolumn chromatography processes for widely being used for product purification. These processes demand significant optimization to meet desired product quality attributes, where each step also decreases final yields. In this work, we demonstrate the novel self-removing iCapTag™ affinity tag, which provides a new platform for capturing, concentrating, and purifying recombinant proteins. Importantly, this system provides a tagless target protein, which is suitable for research and clinical use, where the only requirement for tag removal is a small change in buffer pH. No additional proteins, reagents or cofactors are required. We also present case studies demonstrating the use of iCapTag™ for highly efficient purification of untagged interferon alpha 2b, the ML39 single chain variable fragment (scFv), and the receptor binding domain (RBD) of SARS-CoV-2 spike protein. These proteins were expressed and secreted by Expi293 cells with the self-removing tag fused to their N-terminus. We were able to obtain highly pure (> 99 %) tagless protein in a single purification step with high clearance of host cell DNA, tagged precursor, higher and lower molecular weight impurities. Based on these preliminary results, we propose the iCapTag™ as a universal capture platform for diverse classes of recombinant therapeutic proteins.
Collapse
Affiliation(s)
- Sai Vivek Prabhala
- William G. Lowrie Department of Chemical and Biomolecular Engineering, Ohio State University, Columbus, OH 43210, United States
| | - Brian Marshall
- Eli Lilly and Company, Indianapolis, IN 46285, United States
| | | | - Yamin Fan
- Johnson & Johnson, 4560 Jinke Road, Shanghai 201210, China
| | - Ekaterina Creamer
- Ohio State Biochemistry Program, Ohio State University, Columbus, OH 43210, United States
| | - David W Wood
- William G. Lowrie Department of Chemical and Biomolecular Engineering, Ohio State University, Columbus, OH 43210, United States; Protein Capture Science LLC, Columbus, OH 43212, United States.
| |
Collapse
|
6
|
C L B Ferreira B, Hannard M, Lozano-Garcia M, Aston L, Tejeda G, Domena JB, Bernard B, Chen J, Bartoli M, Rech Tondin A, Zhou Y, Scorzari A, Perrone CS, Tagliaferro A, Deo S, Daunert S, Dumont CM, Leblanc RM. Investigating the Significances of Thiol Functionalities in SARS-CoV-2 Using Carbon Dots for Viral Inhibition. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58439-58451. [PMID: 39422222 DOI: 10.1021/acsami.4c14482] [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: 10/19/2024]
Abstract
While the World Health Organization has declared the end of the SARS-CoV-2 public health emergency, studies related to corona viruses are still under course. As of 2024, the severity of COVID-19 has diminished with current treatments and vaccinations. However, individuals can still face severe complications, highlighting the importance of ongoing research into innovative treatments for current and future coronavirus-related diseases. This study approaches the mechanism of viral entrance into the host cells and the current evidence on the use of sulfhydryl groups for the COVID-19 treatment. Certain thiol drugs, a key contributor to inflammatory processes, exhibit both viral inhibition properties and the potential to regulate cellular oxidative stress by scavenging free radicals. Herein, we developed biocompatible thiol-functionalized carbon dots (CDs) and investigated the correlation between the number of thiols and pseudo-SARS-CoV-2 inhibition, reactive oxygen species (ROS) scavenging, and anti-inflammatory response. The free-radical scavenging experiment and the ROS cellular assay indicate that thiolated CDs serve as effective reducing agents and potential regulators of cellular oxidative stress. The CDs also demonstrated good cell viability alongside significant antiviral capabilities, with inhibition levels up to 60.4%. Furthermore, the flow cytometry results suggest that in an inflammatory environment, the presence of thiolated CDs promotes an anti-inflammatory response. Overall, the results demonstrate a strong correlation between the number of thiols and the increased efficacy observed across experiments, presenting thiolated CDs as promising candidates to prevent and treat COVID-19 infection.
Collapse
Affiliation(s)
- Braulio C L B Ferreira
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Maxence Hannard
- Empa - Swiss Federal Laboratories for Materials Science and Technology, Dübendorf 8600, Switzerland
| | - Mercedes Lozano-Garcia
- Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Lillian Aston
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Giancarlo Tejeda
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Justin B Domena
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Brianna Bernard
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Jiuyan Chen
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Mattia Bartoli
- Center for Sustainable Future Technologies, Italian Institute of Technology, Via Livorno 60, Turin 10144, Italy
| | - Arthur Rech Tondin
- Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Yiqun Zhou
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Annalise Scorzari
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Caitlyn S Perrone
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Alberto Tagliaferro
- Department of Applied Science and Technology, Politecnico di Torino, Torino 10129 Italy
| | - Sapna Deo
- Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Sylvia Daunert
- Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| |
Collapse
|
7
|
Villada-Troncoso SM, Arévalo-Romero JA, Hernández Rivera V, Pedraza-Escalona M, Pérez-Tapia SM, Espejo-Mojica AJ, Alméciga-Díaz CJ. Study of Potential Blocking Peptides Targeting the SARS-CoV-2 RBD/hACE2 Interaction. Pharmaceuticals (Basel) 2024; 17:1240. [PMID: 39338402 PMCID: PMC11435355 DOI: 10.3390/ph17091240] [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: 09/02/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, was declared a public health emergency in early 2020. The infection initiates when the receptor-binding domain (RBD) of the viral spike protein binds to human angiotensin-converting enzyme 2 (ACE2). Despite the success of vaccination efforts, the emergence of new variants highlights the ongoing need for treatments targeting these evolving strains. In silico methods previously identified peptides BP2, BP9, and BP11 as being capable of disrupting the RBD-ACE2 interaction, though their efficacy has not been experimentally validated until now. METHODS In this study, these peptides were recombinantly produced in the yeast Komagataella phaffii, and the activity was assessed in vitro using binding assays with multiple RBD variants and the inhibition of the RBD-ACE2 interaction. RESULTS The production yield for BP2, BP9, and BP11 was 14.34, 4.01, and 1.35 mg per culture liter, respectively. Noteworthy, the three BPs interacted with the RBD of SARS-CoV-2 variants of concern, with BP2 showing higher recognition. Finally, the BPs showed an RBD/hACE2 interaction blocking capacity with IC50 values between 1.03 and 5.35 nM, with BP2 showing the lowest values among the evaluated peptides. CONCLUSIONS These results demonstrate that BP2, specifically, is a promising candidate for the development of novel therapeutic interventions targeting SARS-CoV-2 and other coronaviruses that use hACE2 for cellular entry.
Collapse
Affiliation(s)
- Sara M. Villada-Troncoso
- Institute for the Study in Inborn Errors of Metabolism—IEIM, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (S.M.V.-T.); (J.A.A.-R.); (A.J.E.-M.)
| | - Jenny Andrea Arévalo-Romero
- Institute for the Study in Inborn Errors of Metabolism—IEIM, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (S.M.V.-T.); (J.A.A.-R.); (A.J.E.-M.)
- Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud—IDCBIS, Bogotá 111611, Colombia
| | - Vanessa Hernández Rivera
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Colonia Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico; (V.H.R.); (S.M.P.-T.)
| | - Martha Pedraza-Escalona
- CONAHCyT-Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Colonia Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico;
| | - Sonia M. Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Colonia Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico; (V.H.R.); (S.M.P.-T.)
| | - Angela Johana Espejo-Mojica
- Institute for the Study in Inborn Errors of Metabolism—IEIM, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (S.M.V.-T.); (J.A.A.-R.); (A.J.E.-M.)
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study in Inborn Errors of Metabolism—IEIM, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (S.M.V.-T.); (J.A.A.-R.); (A.J.E.-M.)
| |
Collapse
|
8
|
Hieronimus K, Donauer T, Klein J, Hinkel B, Spänle JV, Probst A, Niemeyer J, Kibrom S, Kiefer AM, Schneider L, Husemann B, Bischoff E, Möhring S, Bayer N, Klein D, Engels A, Ziehmer BG, Stieβ J, Moroka P, Schroda M, Deponte M. A Modular Cloning Toolkit for the production of recombinant proteins in Leishmania tarentolae. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:128-142. [PMID: 38799406 PMCID: PMC11121976 DOI: 10.15698/mic2024.04.821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 05/29/2024]
Abstract
Modular Cloning (MoClo) is based on libraries of standardized genetic parts that can be directionally assembled via Golden Gate cloning in one-pot reactions into transcription units and multigene constructs. Here, a team of bachelor students established a MoClo toolkit for the protist Leishmania tarentolae in the frame of the international Genetically Engineered Machine (iGEM) competition. Our modular toolkit is based on a domesticated version of a commercial LEXSY expression vector and comprises 34 genetic parts encoding various affinity tags, targeting signals as well as fluorescent and luminescent proteins. We demonstrated the utility of our kit by the successful production of 16 different tagged versions of the receptor binding domain (RBD) of the SARS-CoV-2 spike protein in L. tarentolae liquid cultures. While highest yields of secreted recombinant RBD were obtained for GST-tagged fusion proteins 48 h post induction, C-terminal peptide tags were often degraded and resulted in lower yields of secreted RBD. Fusing secreted RBD to a synthetic O-glycosylation SP20 module resulted in an apparent molecular mass shift around 10 kDa. No disadvantage regarding the production of RBD was detected when the three antibiotics of the LEXSY system were omitted during the 48-h induction phase. Furthermore, the successful purification of secreted RBD from the supernatant of L. tarentolae liquid cultures was demonstrated in pilot experiments. In summary, we established a MoClo toolkit and exemplified its application for the production of recombinant proteins in L. tarentolae.
Collapse
Affiliation(s)
- Katrin Hieronimus
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
- Faculty of Chemistry, Comparative Biochemistry, RPTU
Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Tabea Donauer
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
- Faculty of Chemistry, Comparative Biochemistry, RPTU
Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Jonas Klein
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Bastian Hinkel
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Julia Vanessa Spänle
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Anna Probst
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Justus Niemeyer
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Salina Kibrom
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Anna Maria Kiefer
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Luzia Schneider
- Faculty of Chemistry, Comparative Biochemistry, RPTU
Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Britta Husemann
- Faculty of Chemistry, Comparative Biochemistry, RPTU
Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Eileen Bischoff
- Faculty of Chemistry, Comparative Biochemistry, RPTU
Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Sophie Möhring
- Faculty of Chemistry, Comparative Biochemistry, RPTU
Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Nicolas Bayer
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Dorothée Klein
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Adrian Engels
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Benjamin Gustav Ziehmer
- Faculty of Chemistry, Comparative Biochemistry, RPTU
Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Julian Stieβ
- Faculty of Computer Science, RPTU Kaiserslautern, D-67663
Kaiserslautern, Germany
| | - Pavlo Moroka
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Michael Schroda
- Faculty of Biology, Molecular Biotechnology & Systems
Biology, RPTU Kaiserslautern, D-67663 Kaiserslautern, Germany
| | - Marcel Deponte
- Faculty of Chemistry, Comparative Biochemistry, RPTU
Kaiserslautern, D-67663 Kaiserslautern, Germany
| |
Collapse
|
9
|
Idrovo-Hidalgo T, Pignataro MF, Bredeston LM, Elias F, Herrera MG, Pavan MF, Foscaldi S, Suireszcz M, Fernández NB, Wetzler DE, Paván CH, Craig PO, Roman EA, Ruberto LAM, Noseda DG, Ibañez LI, Czibener C, Ugalde JE, Nadra AD, Santos J, D'Alessio C. Deglycosylated RBD produced in Pichia pastoris as a low-cost sera COVID-19 diagnosis tool and a vaccine candidate. Glycobiology 2024; 34:cwad089. [PMID: 37944064 DOI: 10.1093/glycob/cwad089] [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: 06/26/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
During the COVID-19 outbreak, numerous tools including protein-based vaccines have been developed. The methylotrophic yeast Pichia pastoris (synonymous to Komagataella phaffii) is an eukaryotic cost-effective and scalable system for recombinant protein production, with the advantages of an efficient secretion system and the protein folding assistance of the secretory pathway of eukaryotic cells. In a previous work, we compared the expression of SARS-CoV-2 Spike Receptor Binding Domain in P. pastoris with that in human cells. Although the size and glycosylation pattern was different between them, their protein structural and conformational features were indistinguishable. Nevertheless, since high mannose glycan extensions in proteins expressed by yeast may be the cause of a nonspecific immune recognition, we deglycosylated RBD in native conditions. This resulted in a highly pure, homogenous, properly folded and monomeric stable protein. This was confirmed by circular dichroism and tryptophan fluorescence spectra and by SEC-HPLC, which were similar to those of RBD proteins produced in yeast or human cells. Deglycosylated RBD was obtained at high yields in a single step, and it was efficient in distinguishing between SARS-CoV-2-negative and positive sera from patients. Moreover, when the deglycosylated variant was used as an immunogen, it elicited a humoral immune response ten times greater than the glycosylated form, producing antibodies with enhanced neutralizing power and eliciting a more robust cellular response. The proposed approach may be used to produce at a low cost, many antigens that require glycosylation to fold and express, but do not require glycans for recognition purposes.
Collapse
Affiliation(s)
- Tommy Idrovo-Hidalgo
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - María F Pignataro
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
- Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Universidad de Buenos Aires, Junín 965 C1113AAD. Buenos Aires, Argentina
| | - Luis M Bredeston
- Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Universidad de Buenos Aires, Junín 965 C1113AAD. Buenos Aires, Argentina
- Instituto de Química y Fisicoquímica Biológicas, (IQUIFIB), CONICET-Universidad de Buenos Aires, Junín 956 C1113AAD, Buenos Aires, Argentina
| | - Fernanda Elias
- Consejo Nacional de Investigaciones Científicas y Técnicas-Fundación Pablo Cassará, Instituto de Ciencia y Tecnología Dr. César Milstein, Saladillo 2468 C1440FFX, Buenos Aires, Argentina
| | - María G Herrera
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - María F Pavan
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
| | - Sabrina Foscaldi
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - Mayra Suireszcz
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - Natalia B Fernández
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - Diana E Wetzler
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - Carlos H Paván
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
- Facultad de Farmacia y Bioquímica, LANAIS-PROEM, Instituto de Química y Fisicoquímica Biológicas, (IQUIFIB), CONICET-Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
| | - Patricio O Craig
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - Ernesto A Roman
- Instituto de Química y Fisicoquímica Biológicas, (IQUIFIB), CONICET-Universidad de Buenos Aires, Junín 956 C1113AAD, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - Lucas A M Ruberto
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 965, C1113AAD, Buenos Aires, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), CONICET-Universidad de Buenos Aires, Junín 965, C1113AAD, Buenos Aires, Argentina
- Instituto Antártico Argentino, Ministerio de Relaciones Exteriores y Culto, Av. 25 de Mayo 1147, B1650HMP, San Martín, Prov. de Buenos Aires, Argentina
| | - Diego G Noseda
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín-CONICET, Av. 25 de Mayo y Francia S/N, B1650HMP, San Martín, Prov. de Buenos Aires, Argentina
| | - Lorena I Ibañez
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), CONICET-Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
| | - Cecilia Czibener
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín-CONICET, Av. 25 de Mayo y Francia S/N, B1650HMP, San Martín, Prov. de Buenos Aires, Argentina
| | - Juan E Ugalde
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
- Instituto de Investigaciones Biotecnológicas (IIBio), Universidad Nacional de San Martín-CONICET, Av. 25 de Mayo y Francia S/N, B1650HMP, San Martín, Prov. de Buenos Aires, Argentina
| | - Alejandro D Nadra
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
| | - Javier Santos
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| | - Cecilia D'Alessio
- Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología y Biología Molecular y Celular, Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 C1425FQB, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| |
Collapse
|
10
|
Feinstein P. Rapid Degradation of the Human ACE2 Receptor Upon Binding and Internalization of SARS-Cov-2-Spike-RBD Protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583884. [PMID: 38496410 PMCID: PMC10942428 DOI: 10.1101/2024.03.07.583884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
It is widely accepted that the SARS-CoV-2 betacoronavirus infects humans through binding the human Angiotensin Receptor 2 (ACE2) that lines the nasal cavity and lungs, followed by import into a cell utilizing the Transmembrane Protease, Serine 2 (TMPRSS2) cofactor. ACE2 binding is mediated by an approximately 200-residue portion of the SARS-CoV-2 extracellular spike protein, the receptor binding domain (RBD). Robust interactions are shown using a novel cell-based assay between an RBD membrane tethered-GFP fusion protein and the membrane bound ACE2-Cherry fusion protein. Several observations were not predicted including, quick and sustained interactions leading to internalization of RBD fusion protein into the ACE2 cells and rapid downregulation of the ACE2-Cherry fluorescence. Targeted mutation in the RBD disulfide Loop 4 led to a loss of internalization for several variants tested. However, a secreted RBD did not cause ACE2 downregulation of ACE2-Cherry fluorescence. Thus, the membrane associated form of RBD found on the viral coat may have long-term system wide consequences on ACE2 expressing cells.
Collapse
Affiliation(s)
- Paul Feinstein
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065
- The Graduate Center Programs in Biochemistry, Biology and CUNY Neuroscience Collaborative, 365 5th Ave, New York, NY 10016
| |
Collapse
|
11
|
Sheng X, Wang J, Zhao L, Yan W, Qian J, Wang Z, Zhang J, Raghavan V. Inactivation mechanism of cold plasma combined with 222 nm ultraviolet for spike protein and its application in disinfecting of SARS-CoV-2. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133458. [PMID: 38215522 DOI: 10.1016/j.jhazmat.2024.133458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/14/2023] [Accepted: 01/04/2024] [Indexed: 01/14/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly transmissible virus that has precipitated a worldwide pandemic of coronavirus disease since 2019. Developing an effective disinfection strategy is crucial to prevent the risk of surface cross-contamination by SARS-CoV-2. This study employed pseudovirus and the receptor-binding domain (RBD) protein of SARS-CoV-2 as models to investigate the spike protein inactivation process and its underlying mechanisms using a novel nonthermal technology. Cold plasma combined with 222 nm ultraviolet (CP+UV) treatment was applied to accelerate the generation of reactive species and enhance sterilization efficiency. The results indicated that the binding activity of RBD protein was completely inhibited at specific concentrations (0.01-0.05 mg/cm2) with corresponding treatment times of 15-30 s. The mechanism potentially involves the reactive species generated by CP+UV, which react with the spike protein RBD of SARS-CoV-2, leading to the loss of SARS-CoV-2 infectivity by causing damage to the β-sheet structure and chemical bonds in the RBD protein. Validated by a biosafety level 3 (BSL3) laboratory, the CP+UV treatment for 30 s could completely inactivate SARS-CoV-2 with a concentration of 19054 ± 1112 TCID50/cm2. Therefore, this study potentially provides a novel disinfection strategy for the inactivation of SARS-CoV-2 on surface cross-contamination.
Collapse
Affiliation(s)
- Xiaowei Sheng
- National Center of Meat Quality and Safety Control, Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jin Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Luling Zhao
- National Center of Meat Quality and Safety Control, Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenjing Yan
- National Center of Meat Quality and Safety Control, Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jing Qian
- National Center of Meat Quality and Safety Control, Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaobin Wang
- National Center of Meat Quality and Safety Control, Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jianhao Zhang
- National Center of Meat Quality and Safety Control, Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Vijaya Raghavan
- Department of Bioresource Engineering, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, QC H9X3V9, Canada
| |
Collapse
|
12
|
Ábrahám E, Bajusz C, Marton A, Borics A, Mdluli T, Pardi N, Lipinszki Z. Expression and purification of the receptor-binding domain of SARS-CoV-2 spike protein in mammalian cells for immunological assays. FEBS Open Bio 2024; 14:380-389. [PMID: 38129177 PMCID: PMC10909970 DOI: 10.1002/2211-5463.13754] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023] Open
Abstract
The receptor-binding domain (RBD) of the spike glycoprotein of SARS-CoV-2 virus mediates the interaction with the host cell and is required for virus internalization. It is, therefore, the primary target of neutralizing antibodies. The receptor-binding domain soon became the major target for COVID-19 research and the development of diagnostic tools and new-generation vaccines. Here, we provide a detailed protocol for high-yield expression and one-step affinity purification of recombinant RBD from transiently transfected Expi293F cells. Expi293F mammalian cells can be grown to extremely high densities in a specially formulated serum-free medium in suspension cultures, which makes them an excellent tool for secreted protein production. The highly purified RBD is glycosylated, structurally intact, and forms homomeric complexes. With this quick and easy method, we are able to produce large quantities of RBD (80 mg·L-1 culture) that we have successfully used in immunological assays to examine antibody titers and seroconversion after mRNA-based vaccination of mice.
Collapse
Affiliation(s)
- Edit Ábrahám
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of BiochemistryHUN‐REN Biological Research CentreSzegedHungary
- National Laboratory for Biotechnology, Institute of GeneticsHUN‐REN Biological Research CentreSzegedHungary
| | - Csaba Bajusz
- National Laboratory for Biotechnology, Institute of GeneticsHUN‐REN Biological Research CentreSzegedHungary
| | - Annamária Marton
- National Laboratory for Biotechnology, Institute of GeneticsHUN‐REN Biological Research CentreSzegedHungary
| | - Attila Borics
- Laboratory of Chemical Biology, Institute of BiochemistryHUN‐REN Biological Research CentreSzegedHungary
| | - Thandiswa Mdluli
- Department of MicrobiologyUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Norbert Pardi
- Department of MicrobiologyUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Zoltán Lipinszki
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of BiochemistryHUN‐REN Biological Research CentreSzegedHungary
- National Laboratory for Biotechnology, Institute of GeneticsHUN‐REN Biological Research CentreSzegedHungary
| |
Collapse
|
13
|
Rajak N, Dey T, Sharma Y, Bellad V, Rangarajan PN. Unlocking Nature's Toolbox: glutamate-inducible recombinant protein production from the Komagatella phaffii PEPCK promoter. Microb Cell Fact 2024; 23:66. [PMID: 38402195 PMCID: PMC10893637 DOI: 10.1186/s12934-024-02340-1] [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: 11/30/2023] [Accepted: 02/17/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Komagataella phaffii (a.k.a. Pichia pastoris) harbors a glutamate utilization pathway in which synthesis of glutamate dehydrogenase 2 and phosphoenolpyruvate carboxykinase (PEPCK) is induced by glutamate. Glutamate-inducible synthesis of these enzymes is regulated by Rtg1p, a cytosolic, basic helix-loop-helix protein. Here, we report food-grade monosodium glutamate (MSG)-inducible recombinant protein production from K. phaffii PEPCK promoter (PPEPCK) using green fluorescent protein (GFP) and receptor binding domain of SARS-CoV-2 virus (RBD) as model proteins. RESULTS PPEPCK-RBD/GFP expression cassette was integrated at two different sites in the genome to improve recombinant protein yield from PPEPCK. The traditional, methanol-inducible alcohol oxidase 1 promoter (PAOX1) was used as the benchmark. Initial studies carried out with MSG as the inducer resulted in low recombinant protein yield. A new strategy employing MSG/ethanol mixed feeding improved biomass generation as well as recombinant protein yield. Cell density of 100-120 A600 units/ml was achieved after 72 h of induction in shake flask cultivations, resulting in recombinant protein yield from PPEPCK that is comparable or even higher than that from PAOX1. CONCLUSIONS We have designed an induction medium for recombinant protein production from K. phaffii PPEPCK in shake flask cultivations. It consists of 1.0% yeast extract, 2.0% peptone, 0.17% yeast nitrogen base with ammonium sulfate, 100 mM potassium phosphate (pH 6.0), 0.4 mg/L biotin, 2.0% MSG, and 2% ethanol. Substitution of ammonium sulphate with 0.5% urea is optional. Carbon source was replenished every 24 h during 72 h induction period. Under these conditions, GFP and RBD yields from PPEPCK equaled and even surpassed those from PAOX1. Compared to the traditional methanol-inducible expression system, the inducers of glutamate-inducible expression system are non-toxic and their metabolism does not generate toxic metabolites such as formaldehyde and hydrogen peroxide. This study sets the stage for MSG-inducible, industrial scale recombinant protein production from K. phaffii PPEPCK in bioreactors.
Collapse
Affiliation(s)
- Neetu Rajak
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Trishna Dey
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Yash Sharma
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Vedanth Bellad
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Pundi N Rangarajan
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
14
|
Fernández NB, Herrera MG, Blaustein M, Pignataro MF. Policy options and practical recommendations for determining priorities in public health research agendas in peripheral countries: insights from a collaborative work initiative in Argentina during the COVID-19 pandemic. Front Med (Lausanne) 2024; 10:1334194. [PMID: 38274453 PMCID: PMC10808489 DOI: 10.3389/fmed.2023.1334194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Affiliation(s)
- Natalia Brenda Fernández
- Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - María Georgina Herrera
- Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Matías Blaustein
- Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Florencia Pignataro
- Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
15
|
Boggiano-Ayo T, Palacios-Oliva J, Lozada-Chang S, Relova-Hernandez E, Gomez-Perez J, Oliva G, Hernandez L, Bueno-Soler A, Montes de Oca D, Mora O, Machado-Santisteban R, Perez-Martinez D, Perez-Masson B, Cabrera Infante Y, Calzadilla-Rosado L, Ramirez Y, Aymed-Garcia J, Ruiz-Ramirez I, Romero Y, Gomez T, Espinosa LA, Gonzalez LJ, Cabrales A, Guirola O, de la Luz KR, Pi-Estopiñan F, Sanchez-Ramirez B, Garcia-Rivera D, Valdes-Balbin Y, Rojas G, Leon-Monzon K, Ojito-Magaz E, Hardy E. Development of a scalable single process for producing SARS-CoV-2 RBD monomer and dimer vaccine antigens. Front Bioeng Biotechnol 2023; 11:1287551. [PMID: 38050488 PMCID: PMC10693982 DOI: 10.3389/fbioe.2023.1287551] [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: 09/01/2023] [Accepted: 10/30/2023] [Indexed: 12/06/2023] Open
Abstract
We have developed a single process for producing two key COVID-19 vaccine antigens: SARS-CoV-2 receptor binding domain (RBD) monomer and dimer. These antigens are featured in various COVID-19 vaccine formats, including SOBERANA 01 and the licensed SOBERANA 02, and SOBERANA Plus. Our approach involves expressing RBD (319-541)-His6 in Chinese hamster ovary (CHO)-K1 cells, generating and characterizing oligoclones, and selecting the best RBD-producing clones. Critical parameters such as copper supplementation in the culture medium and cell viability influenced the yield of RBD dimer. The purification of RBD involved standard immobilized metal ion affinity chromatography (IMAC), ion exchange chromatography, and size exclusion chromatography. Our findings suggest that copper can improve IMAC performance. Efficient RBD production was achieved using small-scale bioreactor cell culture (2 L). The two RBD forms - monomeric and dimeric RBD - were also produced on a large scale (500 L). This study represents the first large-scale application of perfusion culture for the production of RBD antigens. We conducted a thorough analysis of the purified RBD antigens, which encompassed primary structure, protein integrity, N-glycosylation, size, purity, secondary and tertiary structures, isoform composition, hydrophobicity, and long-term stability. Additionally, we investigated RBD-ACE2 interactions, in vitro ACE2 recognition of RBD, and the immunogenicity of RBD antigens in mice. We have determined that both the monomeric and dimeric RBD antigens possess the necessary quality attributes for vaccine production. By enabling the customizable production of both RBD forms, this unified manufacturing process provides the required flexibility to adapt rapidly to the ever-changing demands of emerging SARS-CoV-2 variants and different COVID-19 vaccine platforms.
Collapse
Affiliation(s)
- Tammy Boggiano-Ayo
- Process Development Direction, Center of Molecular Immunology, Havana, Cuba
| | | | | | | | | | - Gonzalo Oliva
- Process Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Alexi Bueno-Soler
- Process Development Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Osvaldo Mora
- Process Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Dayana Perez-Martinez
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | - Beatriz Perez-Masson
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | | | | | - Yaima Ramirez
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | - Judey Aymed-Garcia
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Yamile Romero
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | - Tania Gomez
- Quality Direction, Center of Molecular Immunology, Havana, Cuba
| | | | | | - Annia Cabrales
- Center for Genetic Engineering and Biotechnology, Playa, Cuba
| | - Osmany Guirola
- Center for Genetic Engineering and Biotechnology, Playa, Cuba
| | | | | | | | | | | | - Gertrudis Rojas
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | - Kalet Leon-Monzon
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Eugenio Hardy
- Process Development Direction, Center of Molecular Immunology, Havana, Cuba
| |
Collapse
|
16
|
Moatasim Y, Kutkat O, Osman AM, Gomaa MR, Okda F, El Sayes M, Kamel MN, Gaballah M, Mostafa A, El-Shesheny R, Kayali G, Ali MA, Kandeil A. Potent Antiviral Activity of Vitamin B12 against Severe Acute Respiratory Syndrome Coronavirus 2, Middle East Respiratory Syndrome Coronavirus, and Human Coronavirus 229E. Microorganisms 2023; 11:2777. [PMID: 38004788 PMCID: PMC10673013 DOI: 10.3390/microorganisms11112777] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Repurposing vitamins as antiviral supporting agents is a rapid approach used to control emerging viral infections. Although there is considerable evidence supporting the use of vitamin supplementation in viral infections, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the specific role of each vitamin in defending against coronaviruses remains unclear. Antiviral activities of available vitamins on the infectivity and replication of human coronaviruses, namely, SARS-CoV-2, Middle East respiratory syndrome coronavirus (MERS-CoV), and human coronavirus 229E (HCoV-229E), were investigated using in silico and in vitro studies. We identified potential broad-spectrum inhibitor effects of Hydroxocobalamin and Methylcobalamin against the three tested CoVs. Cyanocobalamin could selectively affect SARS-CoV-2 but not MERS-CoV and HCoV-229E. Methylcobalamin showed significantly higher inhibition values on SARS-CoV-2 compared with Hydroxocobalamin and Cyanocobalamin, while Hydroxocobalamin showed the highest potent antiviral activity against MERS-CoV and Cyanocobalamin against HCoV-229E. Furthermore, in silico studies were performed for these promising vitamins to investigate their interaction with SARS-CoV-2, MERS-CoV, and HCoV-229E viral-specific cell receptors (ACE2, DPP4, and hAPN protein, respectively) and viral proteins (S-RBD, 3CL pro, RdRp), suggesting that Hydroxocobalamin, Methylcobalamin, and Cyanocobalamin may have significant binding affinity to these proteins. These results show that Methylcobalamin may have potential benefits for coronavirus-infected patients.
Collapse
Affiliation(s)
- Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | - Omnia Kutkat
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | - Ahmed M. Osman
- Biochemistry Department, Faculty of Science, Cairo University, Cairo 12613, Egypt;
| | - Mokhtar R. Gomaa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | - Faten Okda
- Veterinary Research Institute, National Research Centre, Giza 12622, Egypt;
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Mohamed El Sayes
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | - Mina Nabil Kamel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | - Mohamed Gaballah
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | - Rabeh El-Shesheny
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | | | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt; (Y.M.); (O.K.); (M.R.G.); (M.E.S.); (M.N.K.); (M.G.); (A.M.); (R.E.-S.)
| |
Collapse
|
17
|
Parthiban S, Vijeesh T, Gayathri T, Shanmugaraj B, Sharma A, Sathishkumar R. Artificial intelligence-driven systems engineering for next-generation plant-derived biopharmaceuticals. FRONTIERS IN PLANT SCIENCE 2023; 14:1252166. [PMID: 38034587 PMCID: PMC10684705 DOI: 10.3389/fpls.2023.1252166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023]
Abstract
Recombinant biopharmaceuticals including antigens, antibodies, hormones, cytokines, single-chain variable fragments, and peptides have been used as vaccines, diagnostics and therapeutics. Plant molecular pharming is a robust platform that uses plants as an expression system to produce simple and complex recombinant biopharmaceuticals on a large scale. Plant system has several advantages over other host systems such as humanized expression, glycosylation, scalability, reduced risk of human or animal pathogenic contaminants, rapid and cost-effective production. Despite many advantages, the expression of recombinant proteins in plant system is hindered by some factors such as non-human post-translational modifications, protein misfolding, conformation changes and instability. Artificial intelligence (AI) plays a vital role in various fields of biotechnology and in the aspect of plant molecular pharming, a significant increase in yield and stability can be achieved with the intervention of AI-based multi-approach to overcome the hindrance factors. Current limitations of plant-based recombinant biopharmaceutical production can be circumvented with the aid of synthetic biology tools and AI algorithms in plant-based glycan engineering for protein folding, stability, viability, catalytic activity and organelle targeting. The AI models, including but not limited to, neural network, support vector machines, linear regression, Gaussian process and regressor ensemble, work by predicting the training and experimental data sets to design and validate the protein structures thereby optimizing properties such as thermostability, catalytic activity, antibody affinity, and protein folding. This review focuses on, integrating systems engineering approaches and AI-based machine learning and deep learning algorithms in protein engineering and host engineering to augment protein production in plant systems to meet the ever-expanding therapeutics market.
Collapse
Affiliation(s)
- Subramanian Parthiban
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Thandarvalli Vijeesh
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Thashanamoorthi Gayathri
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Balamurugan Shanmugaraj
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Ashutosh Sharma
- Tecnologico de Monterrey, School of Engineering and Sciences, Centre of Bioengineering, Queretaro, Mexico
| | - Ramalingam Sathishkumar
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| |
Collapse
|
18
|
Wongnak R, Brindha S, Yoshizue T, Onchaiya S, Mizutani K, Kuroda Y. E. coli production of a multi-disulfide bonded SARS-CoV-2 Omicron BA.5 RBD exhibiting native-like biochemical and biophysical properties. Biophys Physicobiol 2023; 20:e200036. [PMID: 38344033 PMCID: PMC10850476 DOI: 10.2142/biophysico.bppb-v20.0036] [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: 07/27/2023] [Accepted: 09/19/2023] [Indexed: 03/27/2024] Open
Abstract
Low-cost bacterial production of the receptor binding domain (RBD) of the SARS-CoV-2 Omicron spike protein holds significant potential in expediting the development of therapeutics against COVID-19. However, RBD contains eight cysteines forming four disulfide bonds, and expression in E. coli using standard protocols produces insoluble RBD forming non-native disulfide bonds. Here, we expressed RBD in E. coli T7 SHuffle with high aeration, which enhanced disulfide formation in the cytoplasm and reshuffling of non-native disulfide bonds, and at a low temperature of 16°C, which stabilized the native conformation and thus the formation of the native disulfide bonds. The yield of RBD was as high as 3 mg per 200 mL culture. We analyzed the conformational and biophysical properties of our E. coli-expressed RBD. First, the RP-HPLC elution profile indicated a single peak, suggesting that RBD was folded with a single disulfide bond pairing pattern. Next, circular dichroism analysis indicated a secondary structure content very close to that computed from the crystal structure. RBD's thermal denaturation monitored by CD was cooperative, strongly indicating a well-folded protein structure. Moreover, limited proteolysis showed that RBD was nearly as stable as RNase A, and the formation of native disulfide bonds was confirmed by LC-MS analysis. Furthermore, BLI analysis indicated a strong binding of RBD with the hACE2 with a dissociation constant of 0.83 nM, confirming the folded nature of RBD. Altogether, these results demonstrate that our E. coli-expression system can provide a large amount of highly purified RBD with correct disulfide bonds and native-like biochemical and biophysical properties.
Collapse
Affiliation(s)
- Rawiwan Wongnak
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
| | - Subbaian Brindha
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8538, Japan
| | - Takahiro Yoshizue
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
| | - Sawaros Onchaiya
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
| | - Kenji Mizutani
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa 230-0045, Japan
| | - Yutaka Kuroda
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8538, Japan
| |
Collapse
|
19
|
Zhou LY, Zhang S, Li LY, Yang GY, Zeng L. Optimization of mammalian expression vector by cis-regulatory element combinations. Mol Genet Genomics 2023:10.1007/s00438-023-02042-0. [PMID: 37318628 DOI: 10.1007/s00438-023-02042-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023]
Abstract
The regulation of gene expression in mammalian cells by combining various cis-regulatory features has rarely been discussed. In this study, we constructed expression vectors containing various combinations of regulatory elements to examine the regulation of gene expression by different combinations of cis-regulatory elements. The effects of four promoters (CMV promoter, PGK promoter, Polr2a promoter, and EF-1α core promoter), two enhancers (CMV enhancer and SV40 enhancer), two introns (EF-1α intron A and hybrid intron), two terminators (CYC1 terminator and TEF terminator), and their different combinations on downstream gene expression were compared in various mammalian cells using fluorescence microscopy to observe fluorescence, quantitative real-time PCR (qRT-PCR), and western blot. The receptor binding domain (RBD) sequence from severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike protein was used to replace the eGFP sequence in the expression vector and the RBD expression was detected by qRT-PCR and western blot. The results showed that protein expression can be regulated by optimizing the combination of cis-acting elements. The vector with the CMV enhancer, EF-1α core promoter, and TEF terminator was found to express approximately threefold higher eGFP than the unmodified vector in different animal cells, as well as 2.63-fold higher recombinant RBD protein than the original vector in HEK-293T cells. Moreover, we suggest that combinations of multiple regulatory elements capable of regulating gene expression do not necessarily exhibit synergistic effects to enhance expression further. Overall, our findings provide insights into biological applications that require the regulation of gene expression and will help to optimize expression vectors for biosynthesis and other fields. Additionally, we provide valuable insights into the production of RBD proteins, which may aid in developing reagents for diagnosis and treatment during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Lu-Yu Zhou
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, People's Republic of China
- Key Laboratory of Animal Biochemistry and Nutrition, Henan Agricultural University, Ministry of Agriculture and Rural Affairs, Zhengzhou, 450046, Henan, People's Republic of China
- Key Laboratory of Animal Growth and Development, The Education Department of Henan Province, Zhengzhou, 450046, Henan, People's Republic of China
| | - Shuang Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, People's Republic of China
- Key Laboratory of Animal Biochemistry and Nutrition, Henan Agricultural University, Ministry of Agriculture and Rural Affairs, Zhengzhou, 450046, Henan, People's Republic of China
- Key Laboratory of Animal Growth and Development, The Education Department of Henan Province, Zhengzhou, 450046, Henan, People's Republic of China
| | - Li-Yun Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, People's Republic of China
- Key Laboratory of Animal Biochemistry and Nutrition, Henan Agricultural University, Ministry of Agriculture and Rural Affairs, Zhengzhou, 450046, Henan, People's Republic of China
- Key Laboratory of Animal Growth and Development, The Education Department of Henan Province, Zhengzhou, 450046, Henan, People's Republic of China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Henan Agricultural University, Ministry of Agriculture and Rural Affairs, Zhengzhou, 450046, Henan, People's Republic of China
- Key Laboratory of Animal Growth and Development, The Education Department of Henan Province, Zhengzhou, 450046, Henan, People's Republic of China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, People's Republic of China.
- Key Laboratory of Animal Biochemistry and Nutrition, Henan Agricultural University, Ministry of Agriculture and Rural Affairs, Zhengzhou, 450046, Henan, People's Republic of China.
- Key Laboratory of Animal Growth and Development, The Education Department of Henan Province, Zhengzhou, 450046, Henan, People's Republic of China.
| |
Collapse
|
20
|
Soriano JB, Peláez A, Busquets X, Rodrigo-García M, Pérez-Urría EÁ, Alonso T, Girón R, Valenzuela C, Marcos C, García-Castillo E, Ancochea J. ABO blood group as a determinant of COVID-19 and Long COVID: An observational, longitudinal, large study. PLoS One 2023; 18:e0286769. [PMID: 37267401 DOI: 10.1371/journal.pone.0286769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/23/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND An association of ABO blood group and COVID-19 remains controversial. METHODS Following STROBE guidance for observational research, we explored the distribution of ABO blood group in patients hospitalized for acute COVID-19 and in those with Long COVID. Contingency tables were made and risk factors were explored using crude and adjusted Mantle-Haentzel odds ratios (OR and 95% CI). RESULTS Up to September 2022, there were a total of 5,832 acute COVID-19 hospitalizations in our hospital, corresponding to 5,503 individual patients, of whom blood group determination was available for 1,513 (27.5%). Their distribution by ABO was: 653 (43.2%) group 0, 690 (45.6%) A, 113 (7.5%) B, and 57 (3.8%) AB, which corresponds to the expected frequencies in the general population. In parallel, of 676 patients with Long COVID, blood group determination was available for 135 (20.0%). Their distribution was: 60 (44.4%) from group 0, 61 (45.2%) A, 9 (6.7%) B, and 5 (3.7%) AB. The distribution of the ABO system of Long COVID patients did not show significant differences with respect to that of the total group (p ≥ 0.843). In a multivariate analysis adjusting for age, sex, ethnicity, and severity of acute COVID-19 infection, subgroups A, AB, and B were not significantly associated with developing Long COVID with an OR of 1.015 [0.669-1.541], 1.327 [0.490-3.594] and 0.965 [0.453-2.058], respectively. The effect of the Rh+ factor was also not significant 1,423 [0.772-2,622] regarding Long COVID. CONCLUSIONS No association of any ABO blood subgroup with COVID-19 or developing Long COVID was identified.
Collapse
Affiliation(s)
- Joan B Soriano
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Adrián Peláez
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Busquets
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, Palma, Spain
| | | | - Elena Ávalos Pérez-Urría
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Tamara Alonso
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Girón
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Claudia Valenzuela
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Celeste Marcos
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elena García-Castillo
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Julio Ancochea
- Servicio de Neumología, Hospital Universitario de la Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
21
|
Cardoso NP, Rivero C, Castillo M, Mansilla FC, Pastorino F, Piccirilli G, Alonso L, Martínez G, Lullo DD, Bentancor LV, Capozzo AV. Serological screening of SARS-CoV-2 infection in companion animals of Buenos Aires suburbs. Front Vet Sci 2023; 10:1161820. [PMID: 37323839 PMCID: PMC10266215 DOI: 10.3389/fvets.2023.1161820] [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: 02/08/2023] [Accepted: 05/04/2023] [Indexed: 06/17/2023] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) is a zoonotic disease caused by the pandemic virus SARS-CoV-2. Domestic and wild animals are susceptible to infection and are potential reservoirs for virus variants. To date, there is no information about the exposure of companion animals in Buenos Aires Suburbs, the area with the largest population in Argentina where the highest number of COVID-19 human cases occurred during the first infection wave. Here we developed a multi-species indirect ELISA to measure antibodies reactive to the SARS-CoV-2 receptor-binding domain (RBD) from several vertebrates constituting the class Mammalia, making it a valuable tool for field serosurveillance. The ELISA cut-off value was estimated by sera from dogs, cats, cattle, and pigs sampled before 2019 (n = 170), considering a 98% percentile and a grey zone to completely exclude any false positive result. Specificity was confirmed by measuring levels of neutralizing antibodies against canine coronavirus, the avidity of specific antibodies, and their capacity to impede the binding of a recombinant RBD protein to VERO cells in an In-Cell ELISA. Sera from 464 cats and dogs sampled in 2020 and 2021 ("pandemic" samples) were assessed using the RBD-ELISA. Information on COVID-19 disease in the household and the animals' lifestyles was collected. In Buenos Aires Suburbs cats were infected at a higher proportion than dogs, seroprevalence was 7.1 and 1.68%, respectively. Confirmed COVID-19 in the caregivers and outdoor lifestyle were statistically associated with seropositivity in cats. The risk of cats getting infected living indoors in COVID-19-negative households was null. The susceptibility of mammals to SARS-CoV-2, the possibility of transmission between animals themselves and humans, together with the free-roaming lifestyle typical of Buenos Aires suburban companion animals, urge pursuing responsible animal care and avoiding human interaction with animals during the disease course. The multi-species RBD-ELISA we developed can be used as a tool for serosurveillance of SARS-CoV-2 infection in mammalians (domestic and wild), guiding further targeted virological analyses to encounter susceptible species, interspecies transmission, and potential virus reservoirs in our region.
Collapse
Affiliation(s)
- Nancy Patricia Cardoso
- Instituto de Virología e Innovaciones Tecnológicas “IVIT”, CONICET-INTA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
| | - Carla Rivero
- Instituto de Estudios para el Desarrollo Productivo y la Innovación, Universidad Nacional de José Clemente Paz, Buenos Aires, Argentina
| | - Mariangeles Castillo
- Instituto de Virología e Innovaciones Tecnológicas “IVIT”, CONICET-INTA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
| | | | - Florencia Pastorino
- Departamento Zoonosis Urbanas, Ministerio de Salud de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Guadalupe Piccirilli
- Departamento Zoonosis Urbanas, Ministerio de Salud de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Laura Alonso
- Departamento Zoonosis Urbanas, Ministerio de Salud de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Gustavo Martínez
- Departamento Zoonosis Urbanas, Ministerio de Salud de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - David Di Lullo
- Instituto Multidisciplinario de Salud, Tecnología y Desarrollo “IMSaTeD”, CONICET-UNSE, Santiago del Estero, Argentina
| | - Leticia Veronica Bentancor
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
- Instituto de Estudios para el Desarrollo Productivo y la Innovación, Universidad Nacional de José Clemente Paz, Buenos Aires, Argentina
| | - Alejandra Victoria Capozzo
- Instituto de Virología e Innovaciones Tecnológicas “IVIT”, CONICET-INTA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
| |
Collapse
|
22
|
Moro-Pérez L, Boggiano-Ayo T, Lozada-Chang SL, Fernández-Saiz OL, de la Luz KR, Gómez-Pérez JA. Conformational characterization of the mammalian-expressed SARS-CoV-2 recombinant receptor binding domain, a COVID-19 vaccine. Biol Res 2023; 56:22. [PMID: 37150832 PMCID: PMC10164616 DOI: 10.1186/s40659-023-00434-5] [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: 10/09/2022] [Accepted: 04/20/2023] [Indexed: 05/09/2023] Open
Abstract
The COVID-19 pandemic has caused a large number of diseases worldwide. There are few vaccines to constrain this disease and the value of them is high. In this sense, the antigens of the vaccine platform Soberana, the receptor binding domain from SARS-CoV-2 Spike protein, both the monomeric (mRBD) and dimeric (dRBD) forms, have been developed. This study encompassed several analyses by different techniques like circular dichroism (CD), fluorescence spectroscopy (FS) and Gel Filtration- High Performance Liquid ChLC of mRBD and dRBD. Monomer and dimer exhibited similar far-UV CD spectral characteristics with 54% of β-sheet content. Similar conformational features according to near-UV CD and FS studies were observed in both RBD. Stress stability studies by far-UV CD, FS, biological activity and GF-HPLC at 37 °C showed that mRBD is very stable. On the other hand, dRBD fluorescent emission showed a shift towards higher wavelengths as the incubation time increases, suggesting exposition of tryptophan residues, unlike what happens with mRBD. Biological activity outcome confirms these results. GF-HPLC profiles showed that in mRBD, the product of molecular stress are dimers and does not increase over time. However, dRBD showed dimer fragmentation as the main degradation species. This study reveals the usefulness of CD techniques for the analysis of degradation of RBD molecules as well as showed the difference in stability of both RBD molecules. Besides, our work provides useful insights into the production of a key protein used in diagnosis and therapeutics to fight COVID-19 pandemia.
Collapse
Affiliation(s)
- Leina Moro-Pérez
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba.
| | - Tammy Boggiano-Ayo
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba.
| | - Sum Lai Lozada-Chang
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba
| | - Olga Lidia Fernández-Saiz
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba
| | - Kathya Rashida de la Luz
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba
| | - Jose Alberto Gómez-Pérez
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba
| |
Collapse
|
23
|
Cavic M, Nesic A, Mirjacic Martinovic K, Vuletic A, Besu Zizak I, Tisma Miletic N, Krivokuca A, Jankovic R, Gavrovic-Jankulovic M. Detection of humoral and cellular immune response to anti-SARS-CoV-2 BNT162b2 vaccine in breastfeeding women and naïve and previously infected individuals. Sci Rep 2023; 13:6271. [PMID: 37069315 PMCID: PMC10109231 DOI: 10.1038/s41598-023-33516-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 04/13/2023] [Indexed: 04/19/2023] Open
Abstract
This study explored humoral and cellular responses to anti-SARS-CoV-2 BNT162b2 mRNA vaccine in breastfeeding women and naïve and seropositive individuals in the first six months after vaccination.Sixty-one volunteers vaccinated with two doses of the BNT162b2 mRNA vaccine were enrolled in the study. In-house developed ELISA was used for the quantification of SARS-CoV-2 RBD-specific antibodies. Cell surface marker expression and intracellular IFN-γ analysis were carried out by flow cytometry. The concentrations of IFN-γ, IL-6 and TNF were determined by ELISA. A significant rise in anti-RBD IgG antibody levels was observed 14 days after the first vaccine dose (p < 0.0001) in serum and milk. The expression of CD28 on CD4+ T cells was significantly higher compared to baseline (p < 0.05). There was a significant increase (p ≤ 0.05) in B cell lymphocyte subset after revaccination, and increased percentage of CD80+ B cells. The expression of IFN-γ in peripheral blood lymphocytes, CD3+ T cells and serum was significantly increased (p < 0.05). No significant difference in immune response was observed between breastfeeding women and other study participants. The anti-SARS-CoV-2 BNT162b2 mRNA vaccine-induced measurable and durable immune response in breastfeeding women and in naïve and previously infected individuals.
Collapse
Affiliation(s)
- Milena Cavic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia.
| | - Andrijana Nesic
- Department of Biochemistry, Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Katarina Mirjacic Martinovic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia
| | - Ana Vuletic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia
| | - Irina Besu Zizak
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia
| | - Nevena Tisma Miletic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia
| | - Ana Krivokuca
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia
| | - Radmila Jankovic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia
| | | |
Collapse
|
24
|
Production of neutralizing antibody fragment variants in the cytoplasm of E. coli for rapid screening: SARS-CoV-2 a case study. Sci Rep 2023; 13:4408. [PMID: 36927743 PMCID: PMC10019796 DOI: 10.1038/s41598-023-31369-2] [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: 12/18/2022] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Global health challenges such as the coronavirus pandemic warrant the urgent need for a system that allows efficient production of diagnostic and therapeutic interventions. Antibody treatments against SARS-CoV-2 were developed with an unprecedented pace and this enormous progress was achieved mainly through recombinant protein production technologies combined with expeditious screening approaches. A heterologous protein production system that allows efficient soluble production of therapeutic antibody candidates against rapidly evolving variants of deadly pathogens is an important step in preparedness towards future pandemic challenges. Here, we report cost and time-effective soluble production of SARS-CoV-2 receptor binding domain (RBD) variants as well as an array of neutralizing antibody fragments (Fabs) based on Casirivimab and Imdevimab using the CyDisCo system in the cytoplasm of E. coli. We also report variants of the two Fabs with higher binding affinity against SARS-CoV-2 RBD and suggest this cytoplasmic production of disulfide containing antigens and antibodies can be broadly applied towards addressing future global public health threats.
Collapse
|
25
|
Pavan MF, Bok M, Juan RBS, Malito JP, Marcoppido GA, Franco DR, Militello DA, Schammas JM, Bari S, Stone WB, López K, Porier DL, Muller J, Auguste AJ, Yuan L, Wigdorovitz A, Parreño V, Ibañez LI. Nanobodies against SARS-CoV-2 reduced virus load in the brain of challenged mice and neutralized Wuhan, Delta and Omicron Variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532528. [PMID: 36993215 PMCID: PMC10054972 DOI: 10.1101/2023.03.14.532528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
In this work, we developed llama-derived nanobodies (Nbs) directed to the receptor binding domain (RBD) and other domains of the Spike (S) protein of SARS-CoV-2. Nanobodies were selected after the biopanning of two VHH-libraries, one of which was generated after the immunization of a llama (lama glama) with the bovine coronavirus (BCoV) Mebus, and another with the full-length pre-fused locked S protein (S-2P) and the RBD from the SARS-CoV-2 Wuhan strain (WT). Most of the neutralizing Nbs selected with either RBD or S-2P from SARS-CoV-2 were directed to RBD and were able to block S-2P/ACE2 interaction. Three Nbs recognized the N-terminal domain (NTD) of the S-2P protein as measured by competition with biliverdin, while some non-neutralizing Nbs recognize epitopes in the S2 domain. One Nb from the BCoV immune library was directed to RBD but was non-neutralizing. Intranasal administration of Nbs induced protection ranging from 40% to 80% against COVID-19 death in k18-hACE2 mice challenged with the WT strain. Interestingly, protection was not only associated with a significant reduction of virus replication in nasal turbinates and lungs, but also with a reduction of virus load in the brain. Employing pseudovirus neutralization assays, we were able to identify Nbs with neutralizing capacity against the Alpha, Beta, Delta and Omicron variants. Furthermore, cocktails of different Nbs performed better than individual Nbs to neutralize two Omicron variants (B.1.529 and BA.2). Altogether, the data suggest these Nbs can potentially be used as a cocktail for intranasal treatment to prevent or treat COVID-19 encephalitis, or modified for prophylactic administration to fight this disease.
Collapse
Affiliation(s)
- María Florencia Pavan
- CONICET Universidad de Buenos Aires, Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE)
| | - Marina Bok
- Incuinta, Instituto Nacional de Tecnología Agropecuaria (INTA)
- Instituto de Virología e Innovaciones Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (IVIT-CONICET)
| | - Rafael Betanzos San Juan
- Departamento de Química Biológica, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Ciudad Universitaria, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Juan Pablo Malito
- Incuinta, Instituto Nacional de Tecnología Agropecuaria (INTA)
- Instituto de Virología e Innovaciones Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (IVIT-CONICET)
| | - Gisela Ariana Marcoppido
- Instituto de Investigación Patobiología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas (CICVyA), Instituto Nacional de Tecnología Agropecuaria (INTA)
| | - Diego Rafael Franco
- Centro de Investigaciones en Ciencias Veterinarias y Agronómicas (CICVyA), Instituto Nacional de Tecnología Agropecuaria (INTA)
| | - Daniela Ayelen Militello
- CONICET Universidad de Buenos Aires, Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE)
| | - Juan Manuel Schammas
- Instituto de Virología e Innovaciones Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (IVIT-CONICET)
| | - Sara Bari
- CONICET Universidad de Buenos Aires, Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE)
| | - William B Stone
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Krisangel López
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Danielle L Porier
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - John Muller
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Albert J Auguste
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Lijuan Yuan
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Andrés Wigdorovitz
- Incuinta, Instituto Nacional de Tecnología Agropecuaria (INTA)
- Instituto de Virología e Innovaciones Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (IVIT-CONICET)
| | - Viviana Parreño
- Incuinta, Instituto Nacional de Tecnología Agropecuaria (INTA)
- Instituto de Virología e Innovaciones Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (IVIT-CONICET)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Lorena Itatí Ibañez
- CONICET Universidad de Buenos Aires, Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE)
| |
Collapse
|
26
|
Green EA, Hamaker NK, Lee KH. Comparison of vector elements and process conditions in transient and stable suspension HEK293 platforms using SARS-CoV-2 receptor binding domain as a model protein. BMC Biotechnol 2023; 23:7. [PMID: 36882740 PMCID: PMC9990576 DOI: 10.1186/s12896-023-00777-7] [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: 10/09/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Mammalian cell lines are frequently used as protein expression hosts because of their ability to correctly fold and assemble complex proteins, produce them at high titers, and confer post-translational modifications (PTMs) critical to proper function. Increasing demand for proteins with human-like PTMs, particularly viral proteins and vectors, have made human embryonic kidney 293 (HEK293) cells an increasingly popular host. The need to engineer more productive HEK293 platforms and the ongoing nature of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic presented an opportunity to study strategies to improve viral protein expression in transient and stable HEK293 platforms. RESULTS Initial process development was done at 24 deep well plate (DWP) -scale to screen transient processes and stable clonal cell lines for recombinant SARS-CoV-2 receptor binding domain (rRBD) titer. Nine DNA vectors that drove rRBD production under different promoters and optionally contained Epstein-Barr virus (EBV) elements to promote episomal expression were screened for transient rRBD production at 37 °C or 32 °C. Use of the cytomegalovirus (CMV) promoter to drive expression at 32 °C led to the highest transient protein titers, but inclusion of episomal expression elements did not augment titer. In parallel, four clonal cell lines with titers higher than that of the selected stable pool were identified in a batch screen. Flask-scale transient transfection and stable fed-batch processes were then established that produced rRBD up to 100 mg/L and 140 mg/L, respectively. While a bio-layer interferometry (BLI) assay was crucial for efficiently screening DWP batch titers, an enzyme-linked immunosorbent assay (ELISA) was used to compare titers from the flask-scale batches due to varying matrix effects from different cell culture media compositions. CONCLUSION Comparing yields from the flask-scale batches revealed that stable fed-batch cultures produced up to 2.1x more rRBD than transient processes. The stable cell lines developed in this work are the first reported clonal, HEK293-derived rRBD producers and have titers up to 140 mg/L. As stable production platforms are more economically favorable for long-term protein production at large scales, investigation of strategies to increase the efficiency of high-titer stable cell line generation in Expi293F or other HEK293 hosts is warranted.
Collapse
Affiliation(s)
- Erica A Green
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, Delaware, 19713, USA
| | - Nathaniel K Hamaker
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, Delaware, 19713, USA
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, Delaware, 19713, USA.
| |
Collapse
|
27
|
Raoufi E, Hosseini F, Onagh B, Salehi-Shadkami M, Mehrali M, Mohsenzadegan M, Ho JQ, Bigdelou B, Sepand MR, Webster TJ, Zanganeh S, Farajollahi MM. Designing and developing a sensitive and specific SARS-CoV-2 RBD IgG detection kit for identifying positive human samples. Clin Chim Acta 2023; 542:117279. [PMID: 36871661 PMCID: PMC9985519 DOI: 10.1016/j.cca.2023.117279] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/07/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND More than 3 y into the coronavirus 2019 (COVID-19) pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to undergo mutations. In this context, the Receptor Binding Domain (RBD) is the most antigenic region among the SARS-CoV-2 Spike protein and has emerged as a promising candidate for immunological development. We designed an IgG-based indirect enzyme-linked immunoassay (ELISA) kit based on recombinant RBD, which was produced from the laboratory to 10 L industry scales in Pichia pastoris. METHODS A recombinant-RBD comprising 283 residues (31 kDa) was constructed after epitope analyses. The target gene was initially cloned into an Escherichia coli TOP10 genotype and transformed into Pichia pastoris CBS7435 muts for protein production. Production was scaled up in a 10 L fermenter after a 1 L shake-flask cultivation. The product was ultrafiltered and purified using ion-exchange chromatography. IgG-positive human sera for SARS-CoV-2 were employed by an ELISA test to evaluate the antigenicity and specific binding of the produced protein. RESULTS Bioreactor cultivation yielded 4 g/l of the target protein after 160 h of fermentation, and ion-exchange chromatography indicated a purity > 95%. A human serum ELISA test was performed in 4 parts, and the ROC area under the curve (AUC) was > 0.96 for each part. The mean specificity and sensitivity of each part was 100% and 91.5%, respectively. CONCLUSION A highly specific and sensitive IgG-based serologic kit was developed for improved diagnostic purposes in patients with COVID-19 after generating an RBD antigen in Pichia pastoris at laboratory and 10 L fermentation scales.
Collapse
Affiliation(s)
- Ehsan Raoufi
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Hosseini
- Department of Medical Biotechnology, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Onagh
- Department of Biochemistry, School of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Marjan Mehrali
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Monireh Mohsenzadegan
- Department of Medical Laboratory Science, Iran University of Medical Sciences, Tehran, Iran
| | - Jim Q Ho
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Banafsheh Bigdelou
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States
| | - Mohammad Reza Sepand
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, China; School of Engineering, Saveetha University, Chennai, India; Program in Materials Science, UFPI, Teresina, Brazil
| | - Steven Zanganeh
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States.
| | | |
Collapse
|
28
|
Germanó MJ, Giai C, Cargnelutti DE, Colombo MI, Blanco S, Konigheim B, Spinsanti L, Aguilar J, Gallego S, Valdez HA, Mackern-Oberti JP, Sanchez MV. Receptor-binding domain-based SARS-CoV-2 vaccine adjuvanted with cyclic di-adenosine monophosphate enhances humoral and cellular immunity in mice. J Med Virol 2023; 95:e28584. [PMID: 36794675 DOI: 10.1002/jmv.28584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/25/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
Novel adjuvants are highly desired to improve immune responses of SARS-CoV-2 vaccines. This work reports the potential of the stimulator of interferon genes (STING) agonist adjuvant, the cyclic di-adenosine monophosphate (c-di-AMP), in a SARS-CoV-2 vaccine based on the receptor binding domain (RBD). Here, mice immunized with two doses of monomeric RBD adjuvanted with c-di-AMP intramuscularly were found to exhibit stronger immune responses compared to mice vaccinated with RBD adjuvanted with aluminum hydroxide (Al(OH)3 ) or without adjuvant. After two immunizations, consistent enhancements in the magnitude of RBD-specific immunoglobulin G (IgG) antibody response were observed by RBD + c-di-AMP (mean: 15360) compared to RBD + Al(OH)3 (mean: 3280) and RBD alone (n.d.). Analysis of IgG subtypes indicated a predominantly Th1-biased immune response (IgG2c, mean: 14480; IgG2b, mean: 1040, IgG1, mean: 470) in mice vaccinated with RBD + c-di-AMP compared to a Th2-biased response in those vaccinated with RBD + Al(OH)3 (IgG2c, mean: 60; IgG2b: n.d.; IgG1, mean: 16660). In addition, the RBD + c-di-AMP group showed better neutralizing antibody responses as determined by pseudovirus neutralization assay and by plaque reduction neutralization assay with SARS-CoV-2 wild type. Moreover, the RBD + c-di-AMP vaccine promoted interferon-γ secretion of spleen cell cultures after RBD stimulation. Furthermore, evaluation of IgG-antibody titers in aged mice showed that di-AMP was able to improve RBD-immunogenicity at old age after 3 doses (mean: 4000). These data suggest that c-di-AMP improves immune responses of a SARS-CoV-2 vaccine based on RBD, and would be considered a promising option for future COVID-19 vaccines.
Collapse
Affiliation(s)
- María José Germanó
- Instituto de Medicina y Biología Experimental de Cuyo, Centro Científico Tecnológico, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Mendoza, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Constanza Giai
- Instituto de Histología y Embriología de Mendoza, CONICET-Mendoza, Universidad Nacional de Cuyo-(UNCuyo) CONICET, Mendoza, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego Esteban Cargnelutti
- Instituto de Medicina y Biología Experimental de Cuyo, Centro Científico Tecnológico, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Mendoza, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Isabel Colombo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Histología y Embriología de Mendoza, CONICET-Mendoza, Universidad Nacional de Cuyo-(UNCuyo) CONICET, Mendoza, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Sebastián Blanco
- Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Brenda Konigheim
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lorena Spinsanti
- Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Javier Aguilar
- Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sandra Gallego
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Virología "Dr. J. M. Vanella" (InViV), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Hugo Alberto Valdez
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Juan Pablo Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo, Centro Científico Tecnológico, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Mendoza, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Maria Victoria Sanchez
- Instituto de Medicina y Biología Experimental de Cuyo, Centro Científico Tecnológico, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Mendoza, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
29
|
Tantiwiwat T, Thaiprayoon A, Siriatcharanon AK, Tachaapaikoon C, Plongthongkum N, Waraho-Zhmayev D. Utilization of Receptor-Binding Domain of SARS-CoV-2 Spike Protein Expressed in Escherichia coli for the Development of Neutralizing Antibody Assay. Mol Biotechnol 2023; 65:598-611. [PMID: 36103078 PMCID: PMC9472194 DOI: 10.1007/s12033-022-00563-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/05/2022] [Indexed: 12/26/2022]
Abstract
The ongoing COVID-19 pandemic has resulted from widespread infection by the SARS-CoV-2 virus. As new variants of concern continue to emerge, understanding the correlation between the level of neutralizing antibodies (NAb) and clinical protection from SAR-CoV-2 infection could be critical in planning the next steps in COVID-19 vaccine programs. This study explored the potential usefulness of E. coli as an alternative expression system that can be used to produce a SARS-CoV-2 receptor-binding domain (RBD) for the development of an affordable and flexible NAb detection assay. We expressed the RBD of Beta, Delta, and Omicron variants in the E. coli BL21(DE3) strain and purified them from whole bacterial cells using His-tag-mediated affinity chromatography and urea-assisted refolding. Next, we conducted a head-to-head comparison of the binding activity of our E. coli-produced RBD (E-RBD) with commercial HEK293-produced RBD (H-RBD). The results of a direct binding assay revealed E-RBD and H-RBD binding with ACE2-hFc in similar signal strengths. Furthermore, in the NAb detection assay, % inhibition obtained from both E-RBD and H-RBD demonstrated comparable results in all the investigated assays, suggesting that non-glycosylated RBD produced from E. coli may offer a cost-effective alternative to the use of more expensive glycosylated RBD produced from human cells in the development of such an assay.
Collapse
Affiliation(s)
- Termsak Tantiwiwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, 10140 Thailand
| | - Apisitt Thaiprayoon
- Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, 10140 Thailand
| | - Ake-kavitch Siriatcharanon
- School of Bioresources and Technology, King Mongkut’s University of Technology Thonburi, Bangkok, 10150 Thailand
| | - Chakrit Tachaapaikoon
- School of Bioresources and Technology, King Mongkut’s University of Technology Thonburi, Bangkok, 10150 Thailand ,Excellent Center of Enzyme Technology and Microbial Utilization, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bangkok, 10150 Thailand
| | - Nongluk Plongthongkum
- Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, 10140 Thailand
| | - Dujduan Waraho-Zhmayev
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand.
| |
Collapse
|
30
|
Guan X, Yang Y, Du L. Advances in SARS-CoV-2 receptor-binding domain-based COVID-19 vaccines. Expert Rev Vaccines 2023; 22:422-439. [PMID: 37161869 PMCID: PMC10355161 DOI: 10.1080/14760584.2023.2211153] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
INTRODUCTION The Coronavirus Disease 2019 (COVID-19) pandemic has caused devastating human and economic costs. Vaccination is an important step in controlling the pandemic. Severe acute respiratory coronavirus-2 (SARS-CoV-2), the causative agent of COVID-19, infects cells by binding a cellular receptor through the receptor-binding domain (RBD) within the S1 subunit of the spike (S) protein. Viral entry and membrane fusion are mediated by the S2 subunit. AREAS COVERED SARS-CoV-2 S protein, particularly RBD, serves as an important target for vaccines. Here we review the structure and function of SARS-CoV-2 S protein and its RBD, summarize current COVID-19 vaccines targeting the RBD, and outline potential strategies for improving RBD-based vaccines. Overall, this review provides important information that will facilitate rational design and development of safer and more effective COVID-19 vaccines. EXPERT OPINION The S protein of SARS-CoV-2 harbors numerous mutations, mostly in the RBD, resulting in multiple variant strains. Although many COVID-19 vaccines targeting the RBD of original virus strain (and previous variants) can prevent infection of these strains, their ability against recent dominant variants, particularly Omicron and its offspring, is significantly reduced. Collective efforts are needed to develop effective broad-spectrum vaccines to control current and future variants that have pandemic potential.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yang Yang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
31
|
Arias-Arias JL, Molina-Castro SE, Monturiol-Gross L, Lomonte B, Corrales-Aguilar E. Stable production of recombinant SARS-CoV-2 receptor-binding domain in mammalian cells with co-expression of a fluorescent reporter and its validation as antigenic target for COVID-19 serology testing. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2022; 37:e00780. [PMID: 36619904 PMCID: PMC9805376 DOI: 10.1016/j.btre.2022.e00780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/08/2022] [Accepted: 12/30/2022] [Indexed: 01/02/2023]
Abstract
SARS-CoV-2 receptor binding domain (RBD) recognizes the angiotensin converting enzyme 2 (ACE2) receptor in host cells that enables infection. Due to its antigenic specificity, RBD production is important for development of serological assays. Here we have established a system for stable RBD production in HEK 293T mammalian cells that simultaneously express the recombinant fluorescent protein dTomato, which enables kinetic monitoring of RBD expression by fluorescence microscopy. In addition, we have validated the use of this recombinant RBD in an ELISA assay for the detection of anti-RBD antibodies in serum samples of COVID-19 convalescent patients. Recombinant RBD generated using this approach can be useful for generation of antibody-based therapeutics against SARS-CoV-2, as well serological assays aimed to test antibody responses to this relevant virus.
Collapse
Affiliation(s)
- Jorge L. Arias-Arias
- Centro de Investigación en Enfermedades Tropicales (CIET), Facultad de Microbiología Universidad de Costa Rica, San José, 11501-2060, Costa Rica,Dulbecco Lab Studio, Residencial Lisboa 2G, Alajuela, 20102, Costa Rica
| | - Silvia E. Molina-Castro
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, San José, 11501-2060, Costa Rica
| | - Laura Monturiol-Gross
- Instituto Clodomiro Picado (ICP), Facultad de Microbiología, Universidad de Costa Rica, San José, 11501-2060, Costa Rica,Corresponding author.
| | - Bruno Lomonte
- Instituto Clodomiro Picado (ICP), Facultad de Microbiología, Universidad de Costa Rica, San José, 11501-2060, Costa Rica
| | - Eugenia Corrales-Aguilar
- Centro de Investigación en Enfermedades Tropicales (CIET), Facultad de Microbiología Universidad de Costa Rica, San José, 11501-2060, Costa Rica
| |
Collapse
|
32
|
An engineered SARS-CoV-2 receptor-binding domain produced in Pichia pastoris as a candidate vaccine antigen. N Biotechnol 2022; 72:11-21. [PMID: 35953030 PMCID: PMC9359770 DOI: 10.1016/j.nbt.2022.08.002] [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: 07/12/2021] [Revised: 06/26/2022] [Accepted: 08/07/2022] [Indexed: 01/07/2023]
Abstract
Developing affordable and easily manufactured SARS-CoV-2 vaccines will be essential to achieve worldwide vaccine coverage and long-term control of the COVID-19 pandemic. Here the development is reported of a vaccine based on the SARS-CoV-2 receptor-binding domain (RBD), produced in the yeast Pichia pastoris. The RBD was modified by adding flexible N- and C-terminal amino acid extensions that modulate protein/protein interactions and facilitate protein purification. A fed-batch methanol fermentation with a yeast extract-based culture medium in a 50 L fermenter and an immobilized metal ion affinity chromatography-based downstream purification process yielded 30-40 mg/L of RBD. Correct folding of the purified protein was demonstrated by mass spectrometry, circular dichroism, and determinations of binding affinity to the angiotensin-converting enzyme 2 (ACE2) receptor. The RBD antigen also exhibited high reactivity with sera from convalescent individuals and Pfizer-BioNTech or Sputnik V vaccinees. Immunization of mice and non-human primates with 50 µg of the recombinant RBD adjuvanted with alum induced high levels of binding antibodies as assessed by ELISA with RBD produced in HEK293T cells, and which inhibited RBD binding to ACE2 and neutralized infection of VeroE6 cells by SARS-CoV-2. Additionally, the RBD protein stimulated IFNγ, IL-2, IL-6, IL-4 and TNFα secretion in splenocytes and lung CD3+-enriched cells of immunized mice. The data suggest that the RBD recombinant protein produced in yeast P. pastoris is suitable as a vaccine candidate against COVID-19.
Collapse
|
33
|
Brindha S, Yoshizue T, Wongnak R, Takemae H, Oba M, Mizutani T, Kuroda Y. An Escherichia coli Expressed Multi-Disulfide Bonded SARS-CoV-2 RBD Shows Native-like Biophysical Properties and Elicits Neutralizing Antisera in a Mouse Model. Int J Mol Sci 2022; 23:15744. [PMID: 36555383 PMCID: PMC9779815 DOI: 10.3390/ijms232415744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
A large-scale Escherichia coli (E. coli) production of the receptor-binding domain (RBD) of the SARS-CoV-2 could yield a versatile and low-cost antigen for a subunit vaccine. Appropriately folded antigens can potentially elicit the production of neutralizing antisera providing immune protection against the virus. However, E. coli expression using a standard protocol produces RBDs with aberrant disulfide bonds among the RBD's eight cysteines resulting in the expression of insoluble and non-native RBDs. Here, we evaluate whether E. coli expressing RBD can be used as an antigen candidate for a subunit vaccine. The expressed RBD exhibited native-like structural and biophysical properties as demonstrated by analytical RP-HPLC, circular dichroism, fluorescence, and light scattering. In addition, our E. coli expressed RBD binds to hACE2, the host cell's receptor, with a binding constant of 7.9 × 10-9 M, as indicated by biolayer interferometry analysis. Our E. coli-produced RBD elicited a high IgG titer in Jcl:ICR mice, and the RBD antisera inhibited viral growth, as demonstrated by a pseudovirus-based neutralization assay. Moreover, the increased antibody level was sustained for over 15 weeks after immunization, and a high percentage of effector and central memory T cells were generated. Overall, these results show that E. coli-expressed RBDs can elicit the production of neutralizing antisera and could potentially serve as an antigen for developing an anti-SARS-CoV-2 subunit vaccine.
Collapse
Affiliation(s)
- Subbaian Brindha
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi 184-8588, Tokyo, Japan
| | - Takahiro Yoshizue
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi 184-8588, Tokyo, Japan
| | - Rawiwan Wongnak
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi 184-8588, Tokyo, Japan
| | - Hitoshi Takemae
- Center for Infectious Disease Epidemiology and Prevention Research, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Mami Oba
- Center for Infectious Disease Epidemiology and Prevention Research, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Tetsuya Mizutani
- Center for Infectious Disease Epidemiology and Prevention Research, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Yutaka Kuroda
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi 184-8588, Tokyo, Japan
| |
Collapse
|
34
|
Liu Y, Zhao D, Wang Y, Chen Z, Yang L, Li W, Gong Y, Gan C, Tang J, Zhang T, Tang D, Dong X, Yang Q, Valencia CA, Dai L, Qi S, Dong B, Chow HY, Li Y. A vaccine based on the yeast-expressed receptor-binding domain (RBD) elicits broad immune responses against SARS-CoV-2 variants. Front Immunol 2022; 13:1011484. [PMID: 36439096 PMCID: PMC9682237 DOI: 10.3389/fimmu.2022.1011484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/24/2022] [Indexed: 06/23/2024] Open
Abstract
Development of safe and efficient vaccines is still necessary to deal with the COVID-19 pandemic. Herein, we reported that yeast-expressed recombinant RBD proteins either from wild-type or Delta SARS-CoV-2 were able to elicit immune responses against SARS-CoV-2 and its variants. The wild-type RBD (wtRBD) protein was overexpressed in Pichia pastoris, and the purified protein was used as the antigen to immunize mice after formulating an aluminium hydroxide (Alum) adjuvant. Three immunization programs with different intervals were compared. It was found that the immunization with an interval of 28 days exhibited the strongest immune response to SARS-CoV-2 than the one with an interval of 14 or 42 days based on binding antibody and the neutralizing antibody (NAb) analyses. The antisera from the mice immunized with wtRBD were able to neutralize the Beta variant with a similar efficiency but the Delta variant with 2~2.5-fold decreased efficiency. However, more NAbs to the Delta variant were produced when the Delta RBD protein was used to immunize mice. Interestingly, the NAbs may cross react with the Omicron variant. To increase the production of NAbs, the adjuvant combination of Alum and CpG oligonucleotides was used. Compared with the Alum adjuvant alone, the NAbs elicited by the combined adjuvants exhibited an approximate 10-fold increase for the Delta and a more than 53-fold increase for the Omicron variant. This study suggested that yeast-derived Delta RBD is a scalable and an effective vaccine candidate for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Yu Liu
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Danhua Zhao
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yichang Wang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhian Chen
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wenjuan Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunmei Gan
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jieshi Tang
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Tizhong Zhang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Tang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuju Dong
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qingzhe Yang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - C. Alexander Valencia
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shiqian Qi
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Real & Best Biotech Co., Ltd., Chengdu, China
| | - Hoi Yee Chow
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhua Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
35
|
Advances in Komagataella phaffii Engineering for the Production of Renewable Chemicals and Proteins. FERMENTATION 2022. [DOI: 10.3390/fermentation8110575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The need for a more sustainable society has prompted the development of bio-based processes to produce fuels, chemicals, and materials in substitution for fossil-based ones. In this context, microorganisms have been employed to convert renewable carbon sources into various products. The methylotrophic yeast Komagataella phaffii has been extensively used in the production of heterologous proteins. More recently, it has been explored as a host organism to produce various chemicals through new metabolic engineering and synthetic biology tools. This review first summarizes Komagataella taxonomy and diversity and then highlights the recent approaches in cell engineering to produce renewable chemicals and proteins. Finally, strategies to optimize and develop new fermentative processes using K. phaffii as a cell factory are presented and discussed. The yeast K. phaffii shows an outstanding performance for renewable chemicals and protein production due to its ability to metabolize different carbon sources and the availability of engineering tools. Indeed, it has been employed in producing alcohols, carboxylic acids, proteins, and other compounds using different carbon sources, including glycerol, glucose, xylose, methanol, and even CO2.
Collapse
|
36
|
Dehghani J, Movafeghi A, Mathieu-Rivet E, Mati-Baouche N, Calbo S, Lerouge P, Bardor M. Microalgae as an Efficient Vehicle for the Production and Targeted Delivery of Therapeutic Glycoproteins against SARS-CoV-2 Variants. Mar Drugs 2022; 20:md20110657. [PMID: 36354980 PMCID: PMC9698596 DOI: 10.3390/md20110657] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022] Open
Abstract
Severe acute respiratory syndrome–Coronavirus 2 (SARS-CoV-2) can infect various human organs, including the respiratory, circulatory, nervous, and gastrointestinal ones. The virus is internalized into human cells by binding to the human angiotensin-converting enzyme 2 (ACE2) receptor through its spike protein (S-glycoprotein). As S-glycoprotein is required for the attachment and entry into the human target cells, it is the primary mediator of SARS-CoV-2 infectivity. Currently, this glycoprotein has received considerable attention as a key component for the development of antiviral vaccines or biologics against SARS-CoV-2. Moreover, since the ACE2 receptor constitutes the main entry route for the SARS-CoV-2 virus, its soluble form could be considered as a promising approach for the treatment of coronavirus disease 2019 infection (COVID-19). Both S-glycoprotein and ACE2 are highly glycosylated molecules containing 22 and 7 consensus N-glycosylation sites, respectively. The N-glycan structures attached to these specific sites are required for the folding, conformation, recycling, and biological activity of both glycoproteins. Thus far, recombinant S-glycoprotein and ACE2 have been produced primarily in mammalian cells, which is an expensive process. Therefore, benefiting from a cheaper cell-based biofactory would be a good value added to the development of cost-effective recombinant vaccines and biopharmaceuticals directed against COVID-19. To this end, efficient protein synthesis machinery and the ability to properly impose post-translational modifications make microalgae an eco-friendly platform for the production of pharmaceutical glycoproteins. Notably, several microalgae (e.g., Chlamydomonas reinhardtii, Dunaliella bardawil, and Chlorella species) are already approved by the U.S. Food and Drug Administration (FDA) as safe human food. Because microalgal cells contain a rigid cell wall that could act as a natural encapsulation to protect the recombinant proteins from the aggressive environment of the stomach, this feature could be used for the rapid production and edible targeted delivery of S-glycoprotein and soluble ACE2 for the treatment/inhibition of SARS-CoV-2. Herein, we have reviewed the pathogenesis mechanism of SARS-CoV-2 and then highlighted the potential of microalgae for the treatment/inhibition of COVID-19 infection.
Collapse
Affiliation(s)
- Jaber Dehghani
- Université de Rouen Normandie, Laboratoire GlycoMEV UR 4358, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, F-76000 Rouen, France
| | - Ali Movafeghi
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 5166616471, Iran
| | - Elodie Mathieu-Rivet
- Université de Rouen Normandie, Laboratoire GlycoMEV UR 4358, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, F-76000 Rouen, France
| | - Narimane Mati-Baouche
- Université de Rouen Normandie, Laboratoire GlycoMEV UR 4358, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, F-76000 Rouen, France
| | - Sébastien Calbo
- Université de Rouen Normandie, Inserm U1234, F-76000 Rouen, France
| | - Patrice Lerouge
- Université de Rouen Normandie, Laboratoire GlycoMEV UR 4358, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, F-76000 Rouen, France
| | - Muriel Bardor
- Université de Rouen Normandie, Laboratoire GlycoMEV UR 4358, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, F-76000 Rouen, France
- Correspondence: ; Tel.: +33-2-35-14-67-51
| |
Collapse
|
37
|
He Y, Yu W, Shen L, Yan W, Xiao L, Qi J, Hu T. A SARS-CoV-2 vaccine based on conjugation of SARS-CoV-2 RBD with IC28 peptide and mannan. Int J Biol Macromol 2022; 222:661-670. [PMID: 36152702 PMCID: PMC9490959 DOI: 10.1016/j.ijbiomac.2022.09.180] [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: 03/11/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022]
Abstract
SARS-CoV-2 is a particularly transmissible virus that causes a severe respiratory disease known as COVID-19. Safe and effective vaccines are urgently needed to combat the COVID-19 pandemic. The receptor-binding domain (RBD) of SARS-CoV-2 spike protein elicits most neutralizing antibodies during viral infection and is an ideal antigen for vaccine development. In particular, RBD expressed by E. coli is amenable to low cost and high-yield manufacturability. The adjuvant is necessitated to improve the immunogenicity of RBD. IC28, a TLR5-dependent adjuvant, is a peptide from bacterial flagellin. Mannan is a ligand of TLR-4 or TLR-2 and a polysaccharide adjuvant. Here, IC28 and mannan were both covalently conjugated with RBD from E. coli. The conjugate (RBD-IC28-M) elicited high RBD-specific IgG titers, and a neutralization antibody titer of 201.4. It induced high levels of Th1-type cytokines (IFN-γ) and Th2-type cytokines (IL-5 and IL-10), along with high antigenicity and no apparent toxicity to the organs. The mouse sera of the RBD-IC28-M group competitively interfered with the interaction of RBD and ACE2. Thus, conjugation with IC28 and mannan additively enhanced the humoral and cellular immunity. Our study was expected to provide the feasibility to develop an affordable, easily scalable, effective vaccine SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Yunxia He
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Weili Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lijuan Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Wenying Yan
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Lucheng Xiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Jinming Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| | - Tao Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
38
|
Candido KL, Eich CR, de Fariña LO, Kadowaki MK, da Conceição Silva JL, Maller A, Simão RDCG. Spike protein of SARS-CoV-2 variants: a brief review and practical implications. Braz J Microbiol 2022; 53:1133-1157. [PMID: 35397075 PMCID: PMC8994061 DOI: 10.1007/s42770-022-00743-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/21/2022] [Indexed: 12/24/2022] Open
Abstract
The scientific community has been alarmed by the possible immunological evasion, higher infectivity, and severity of disease caused by the newest variants of SARS-CoV-2. The spike protein has an important role in the cellular invasion of viruses and is the target of several vaccines and therapeutic resources, such as monoclonal antibodies. In addition, some of the most relevant mutations in the different variants are on the spike (S) protein gene sequence that leads to structural alterations in the predicted protein, thus causing concern about the protection mediated by vaccines against these new strains. The present review highlights the most recent knowledge about COVID-19 and vaccines, emphasizing the different spike protein structures of SARS-CoV-2 and updating the reader about the emerging viral variants and their classifications, the more common viral mutations described and their distribution in Brazil. It also compiles a table with the most recent knowledge about all of the Omicron spike mutations.
Collapse
Affiliation(s)
- Kattlyn Laryssa Candido
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Caio Ricardo Eich
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Luciana Oliveira de Fariña
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Marina Kimiko Kadowaki
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - José Luis da Conceição Silva
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Alexandre Maller
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| | - Rita de Cássia Garcia Simão
- Present Address: Laboratório de Bioquímica Molecular (LaBioqMol), Centro de Ciências Médicas e Farmacêuticas, Unioeste, Cascavel, PR Brazil
| |
Collapse
|
39
|
Connelly GG, Kirkland OO, Bohannon S, Lim DC, Wilson RM, Richards EJ, Tay DM, Jee H, Hellinger RD, Hoang NK, Hao L, Chhabra A, Martin-Alonso C, Tan EK, Koehler AN, Yaffe MB, London WB, Lee PY, Krammer F, Bohannon RC, Bhatia SN, Sikes HD, Li H. Direct capture of neutralized RBD enables rapid point-of-care assessment of SARS-CoV-2 neutralizing antibody titer. CELL REPORTS METHODS 2022; 2:100273. [PMID: 35942328 PMCID: PMC9350670 DOI: 10.1016/j.crmeth.2022.100273] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/13/2022] [Accepted: 07/22/2022] [Indexed: 11/26/2022]
Abstract
Neutralizing antibody (NAb) titer is a key biomarker of protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, but point-of-care methods for assessing NAb titer are not widely available. Here, we present a lateral flow assay that captures SARS-CoV-2 receptor-binding domain (RBD) that has been neutralized from binding angiotensin-converting enzyme 2 (ACE2). Quantification of neutralized RBD in this assay correlates with NAb titer from vaccinated and convalescent patients. This methodology demonstrated superior performance in assessing NAb titer compared with either measurement of total anti-spike immunoglobulin G titer or quantification of the absolute reduction in binding between ACE2 and RBD. Our testing platform has the potential for mass deployment to aid in determining at population scale the degree of protective immunity individuals may have following SARS-CoV-2 vaccination or infection and can enable simple at-home assessment of NAb titer.
Collapse
Affiliation(s)
- Guinevere G. Connelly
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Orville O. Kirkland
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Daniel C. Lim
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert M. Wilson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Edward J. Richards
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Dragonfly Therapeutics, Waltham, MA 02451, USA
| | - Dousabel M. Tay
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hyuk Jee
- Division of Rheumatology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Riley D. Hellinger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ngoc K. Hoang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Liang Hao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arnav Chhabra
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Satellite Bio, Cambridge, MA 02139, USA
| | - Carmen Martin-Alonso
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Edward K.W. Tan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Angela N. Koehler
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael B. Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wendy B. London
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Pui Y. Lee
- Division of Rheumatology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Florian Krammer
- Department of Microbiology, and Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Sangeeta N. Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, and Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA
- Wyss Institute at Harvard, Boston, MA 02115, USA
| | - Hadley D. Sikes
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hojun Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
40
|
Chattopadhyay G, Bhowmick J, Manjunath K, Ahmed S, Goyal P, Varadarajan R. Mechanistic insights into global suppressors of protein folding defects. PLoS Genet 2022; 18:e1010334. [PMID: 36037221 PMCID: PMC9491731 DOI: 10.1371/journal.pgen.1010334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/09/2022] [Accepted: 07/11/2022] [Indexed: 01/14/2023] Open
Abstract
Most amino acid substitutions in a protein either lead to partial loss-of-function or are near neutral. Several studies have shown the existence of second-site mutations that can rescue defects caused by diverse loss-of-function mutations. Such global suppressor mutations are key drivers of protein evolution. However, the mechanisms responsible for such suppression remain poorly understood. To address this, we characterized multiple suppressor mutations both in isolation and in combination with inactive mutants. We examined six global suppressors of the bacterial toxin CcdB, the known M182T global suppressor of TEM-1 β-lactamase, the N239Y global suppressor of p53-DBD and three suppressors of the SARS-CoV-2 spike Receptor Binding Domain. When coupled to inactive mutants, they promote increased in-vivo solubilities as well as regain-of-function phenotypes. In the case of CcdB, where novel suppressors were isolated, we determined the crystal structures of three such suppressors to obtain insight into the specific molecular interactions responsible for the observed effects. While most individual suppressors result in small stability enhancements relative to wildtype, which can be combined to yield significant stability increments, thermodynamic stabilisation is neither necessary nor sufficient for suppressor action. Instead, in diverse systems, we observe that individual global suppressors greatly enhance the foldability of buried site mutants, primarily through increase in refolding rate parameters measured in vitro. In the crowded intracellular environment, mutations that slow down folding likely facilitate off-pathway aggregation. We suggest that suppressor mutations that accelerate refolding can counteract this, enhancing the yield of properly folded, functional protein in vivo.
Collapse
Affiliation(s)
| | - Jayantika Bhowmick
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore,
India
| | - Kavyashree Manjunath
- Centre for Chemical Biology and Therapeutics, Institute For Stem Cell
Science and Regenerative Medicine, Bangalore, India
| | - Shahbaz Ahmed
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore,
India
| | - Parveen Goyal
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore,
India
| | | |
Collapse
|
41
|
Gattinger P, Kratzer B, Tulaeva I, Niespodziana K, Ohradanova‐Repic A, Gebetsberger L, Borochova K, Garner‐Spitzer E, Trapin D, Hofer G, Keller W, Baumgartner I, Tancevski I, Khaitov M, Karaulov A, Stockinger H, Wiedermann U, Pickl W, Valenta R. Vaccine based on folded receptor binding domain-PreS fusion protein with potential to induce sterilizing immunity to SARS-CoV-2 variants. Allergy 2022; 77:2431-2445. [PMID: 35357709 PMCID: PMC9111473 DOI: 10.1111/all.15305] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the ongoing global COVID-19 pandemic. One possibility to control the pandemic is to induce sterilizing immunity through the induction and maintenance of neutralizing antibodies preventing SARS-CoV-2 from entering human cells to replicate in. METHODS We report the construction and in vitro and in vivo characterization of a SARS-CoV-2 subunit vaccine (PreS-RBD) based on a structurally folded recombinant fusion protein consisting of two SARS-CoV-2 Spike protein receptor-binding domains (RBD) fused to the N- and C-terminus of hepatitis B virus (HBV) surface antigen PreS to enable the two unrelated proteins serving as immunologic carriers for each other. RESULTS PreS-RBD, but not RBD alone, induced a robust and uniform RBD-specific IgG response in rabbits. Currently available genetic SARS-CoV-2 vaccines induce mainly transient IgG1 responses in vaccinated subjects whereas the PreS-RBD vaccine induced RBD-specific IgG antibodies consisting of an early IgG1 and sustained IgG4 antibody response in a SARS-CoV-2 naive subject. PreS-RBD-specific IgG antibodies were detected in serum and mucosal secretions, reacted with SARS-CoV-2 variants, including the omicron variant of concern and the HBV receptor-binding sites on PreS of currently known HBV genotypes. PreS-RBD-specific antibodies of the immunized subject more potently inhibited the interaction of RBD with its human receptor ACE2 and their virus-neutralizing titers (VNTs) were higher than median VNTs in a random sample of healthy subjects fully immunized with registered SARS-CoV-2 vaccines or in COVID-19 convalescent subjects. CONCLUSION The PreS-RBD vaccine has the potential to serve as a combination vaccine for inducing sterilizing immunity against SARS-CoV-2 and HBV by stopping viral replication through the inhibition of cellular virus entry.
Collapse
Affiliation(s)
- Pia Gattinger
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Bernhard Kratzer
- Center for Pathophysiology, Infectiology and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| | - Inna Tulaeva
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- Laboratory for ImmunopathologyDepartment of Clinical Immunology and AllergologySechenov First Moscow State Medical UniversityMoscowRussia
| | - Katarzyna Niespodziana
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- Karl Landsteiner University of Health SciencesKremsAustria
| | - Anna Ohradanova‐Repic
- Center for Pathophysiology, Infectiology and ImmunologyInstitute for Hygiene and Applied ImmunologyMedical University of ViennaViennaAustria
| | - Laura Gebetsberger
- Center for Pathophysiology, Infectiology and ImmunologyInstitute for Hygiene and Applied ImmunologyMedical University of ViennaViennaAustria
| | - Kristina Borochova
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Erika Garner‐Spitzer
- Institute of Specific Prophylaxis and Tropical MedicineMedical University of ViennaViennaAustria
| | - Doris Trapin
- Center for Pathophysiology, Infectiology and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| | - Gerhard Hofer
- Department of Materials and Environmental ChemistryUniversity of StockholmStockholmSweden
| | - Walter Keller
- Institute of Molecular Biosciences, BioTechMed GrazUniversity of GrazGrazAustria
| | | | - Ivan Tancevski
- Department of Internal Medicine IIMedical University of InnsbruckInnsbruckAustria
| | - Musa Khaitov
- NRC Institute of Immunology FMBA of RussiaMoscowRussia
- Pirogov Russian National Research Medical UniversityMoscowRussia
| | - Alexander Karaulov
- Laboratory for ImmunopathologyDepartment of Clinical Immunology and AllergologySechenov First Moscow State Medical UniversityMoscowRussia
| | - Hannes Stockinger
- Center for Pathophysiology, Infectiology and ImmunologyInstitute for Hygiene and Applied ImmunologyMedical University of ViennaViennaAustria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical MedicineMedical University of ViennaViennaAustria
| | - Winfried F. Pickl
- Center for Pathophysiology, Infectiology and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
- Karl Landsteiner University of Health SciencesKremsAustria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- Laboratory for ImmunopathologyDepartment of Clinical Immunology and AllergologySechenov First Moscow State Medical UniversityMoscowRussia
- Karl Landsteiner University of Health SciencesKremsAustria
- NRC Institute of Immunology FMBA of RussiaMoscowRussia
| |
Collapse
|
42
|
Patrick C, Upadhyay V, Lucas A, Mallela KM. Biophysical Fitness Landscape of the SARS-CoV-2 Delta Variant Receptor Binding Domain. J Mol Biol 2022; 434:167622. [PMID: 35533762 PMCID: PMC9076029 DOI: 10.1016/j.jmb.2022.167622] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 12/16/2022]
Abstract
Among the five known SARS-CoV-2 variants of concern, Delta is the most virulent leading to severe symptoms and increased mortality among infected people. Our study seeks to examine how the biophysical parameters of the Delta variant correlate to the clinical observations. Receptor binding domain (RBD) is the first point of contact with the human host cells and is the immunodominant form of the spike protein. Delta variant RBD contains two novel mutations L452R and T478K. We examined the effect of single as well as the double mutations on RBD expression in human Expi293 cells, RBD stability using urea and thermal denaturation, and RBD binding to angiotensin converting enzyme 2 (ACE2) receptor and to neutralizing antibodies using isothermal titration calorimetry. Delta variant RBD showed significantly higher expression compared to the wild-type RBD, and the increased expression is due to L452R mutation. Despite their non-conservative nature, none of the mutations significantly affected RBD structure and stability. All mutants showed similar binding affinity to ACE2 and to Class 1 antibodies (CC12.1 and LY-CoV016) as that of the wild-type. Delta double mutant L452R/T478K showed no binding to Class 2 antibodies (P2B-2F6 and LY-CoV555) and a hundred-fold weaker binding to a Class 3 antibody (REGN10987), and the decreased antibody binding is determined by the L452R mutation. These results indicate that the immune escape from neutralizing antibodies, rather than increased receptor binding, is the main biophysical parameter that determined the fitness landscape of the Delta variant RBD.
Collapse
Affiliation(s)
| | | | | | - Krishna M.G. Mallela
- Corresponding author at: Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Blvd, MS C238-V20, Aurora, CO 80045, USA
| |
Collapse
|
43
|
Designing and characterization of a SARS-CoV-2 immunogen with receptor binding motif grafted on a protein scaffold: An epitope-focused vaccine approach. Int J Biol Macromol 2022; 209:1359-1367. [PMID: 35469951 PMCID: PMC9033297 DOI: 10.1016/j.ijbiomac.2022.04.148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 12/24/2022]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has a significant burden on the economy and healthcare around the world. Vaccines are the most effective tools to fight infectious diseases by containing the spread of the disease. The current vaccines against SARS-CoV-2 are mostly based on the spike protein of SARS-CoV-2, which is large and has many immune-dominant non-neutralizing epitopes that may effectively skew the antibody response towards non-neutralizing antibodies. Here, we have explored the possibility of immune-focusing the receptor binding motif (RBM) of the spike protein of SARS-CoV-2 that induces mostly neutralizing antibodies in natural infection or in vacinees. The result shows that the scaffolded RBM can bind to Angiotensin Converting Enzyme 2 (ACE2) although with low affinity and induces a strong antibody response in mice. The immunized sera can bind both, the receptor binding domain (RBD) and the spike protein, which holds the RBM in its natural context. Sera from the immunized mice showed robust interferon γ response but poor neutralization of SARS-CoV-2 suggesting presence of a predominant T cell epitope on scaffolded RBM. Together, we provide a strategy for inducing strong antigenic T cell response which could be exploited further for future vaccine designing and development against SARS-CoV-2 infection.
Collapse
|
44
|
The STING Ligand and Delivery System Synergistically Enhance the Immunogenicity of an Intranasal Spike SARS-CoV-2 Vaccine Candidate. Biomedicines 2022; 10:biomedicines10051142. [PMID: 35625879 PMCID: PMC9138454 DOI: 10.3390/biomedicines10051142] [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: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 11/23/2022] Open
Abstract
The respiratory organ serves as a primary target site for SARS-CoV-2. Thus, the vaccine-stimulating immune response of the respiratory tract is significant in controlling SARS-CoV-2 transmission and disease development. In this study, mucoadhesive nanoparticles were used to deliver SARS-CoV-2 spike proteins (S-NPs) into the nasal tracts of mice. The responses in the respiratory organ and the systemic responses were monitored. The administration of S-NPs along with cGAMP conferred a robust stimulation of antibody responses in the respiratory tract, as demonstrated by an increase of IgA and IgG antibodies toward the spike proteins in bronchoalveolar lavages (BALs) and the lungs. Interestingly, the elicited antibodies were able to neutralize both the wild-type and Delta variant strains of SARS-CoV-2. Significantly, the intranasal immunization also stimulated systemic responses. This is evidenced by the increased production of circulating IgG and IgA, which were able to neutralize and bind specifically to the SARS-CoV-2 virion and spike protein. Additionally, this intranasal administration potently activated a splenic T cell response and the production of Th-1 cytokines, suggesting that this vaccine may well activate a cellular response in the respiratory tract. The results demonstrate that STING agonist strongly acts as an adjuvant to the immunogenicity of S-NPs. This platform may be an ideal vaccine against SARS-CoV-2.
Collapse
|
45
|
Prates JWO, Xisto MF, Rodrigues JVDS, Colombari JPC, Meira JMA, Dias RS, da Silva CC, de Paula ESO. Zika Virus Envelope Protein Domain III Produced in K. phaffii Has the Potential for Diagnostic Applications. Diagnostics (Basel) 2022; 12:diagnostics12051198. [PMID: 35626353 PMCID: PMC9139701 DOI: 10.3390/diagnostics12051198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV) represents a global human health threat and it is related to severe diseases such as congenital Zika syndrome (CZS) and Guillain-Barré syndrome (GBS). There is no vaccine available nor specific antiviral treatment, so developing sensitive, specific, and low-cost diagnostic tests is necessary. Thus, the objective of this work was to produce the Zika virus envelope protein domain III (ZIKV-EDIII) in Komagataella phaffii KM71H and evaluate its potential for diagnostic applications. After the K. phaffii had been transformed with the pPICZαA-ZIKV-EDIII vector, an SDS-PAGE and Western Blot were performed to characterize the recombinant protein and an ELISA to evaluate the antigenic potential. The results show that ZIKV-EDIII was produced in the expected size, with a good purity grade and yield of 2.58 mg/L. The receiver operating characteristic (ROC) curve showed 90% sensitivity and 87.5% specificity for IgM, and 93.33% sensitivity and 82.76% specificity for IgG. The ZIKV-EDIII protein was efficiently produced in K. phaffi, and it has the potential for diagnostic applications.
Collapse
Affiliation(s)
- John Willians Oliveira Prates
- Department of Microbiology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; (J.W.O.P.); (J.V.d.S.R.); (C.C.d.S.)
| | - Mariana Fonseca Xisto
- Laboratory of Molecular Immunovirology, Department of General Biology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; (M.F.X.); (R.S.D.)
| | - João Vitor da Silva Rodrigues
- Department of Microbiology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; (J.W.O.P.); (J.V.d.S.R.); (C.C.d.S.)
| | - João Pedro Cruz Colombari
- Department of Medicine and Nursing, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; (J.P.C.C.); (J.M.A.M.)
| | - Júlia Maria Alves Meira
- Department of Medicine and Nursing, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; (J.P.C.C.); (J.M.A.M.)
| | - Roberto Sousa Dias
- Laboratory of Molecular Immunovirology, Department of General Biology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; (M.F.X.); (R.S.D.)
| | - Cynthia Canedo da Silva
- Department of Microbiology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; (J.W.O.P.); (J.V.d.S.R.); (C.C.d.S.)
| | - e Sérgio Oliveira de Paula
- Laboratory of Molecular Immunovirology, Department of General Biology, Federal University of Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; (M.F.X.); (R.S.D.)
- Correspondence: ; Tel.: +55-31-36125015
| |
Collapse
|
46
|
M.D OC, Solomon BA, Tauseef A, Haroon H, R. E. E. AG, K.C. SM. EMERGENCE OF NEW STRAINS OF SARS-COV-2: AFRICA'S FATE AND ITS PREPAREDNESS AGAINST COVID-19 INFECTION WAVES. Afr J Infect Dis 2022; 16:1-12. [PMID: 35582064 PMCID: PMC9097310 DOI: 10.21010/ajid.v16i2.1] [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: 07/24/2021] [Revised: 11/13/2021] [Accepted: 11/17/2021] [Indexed: 11/29/2022] Open
Abstract
Background Severe acute respiratory syndrome coronavirus-2(SARS-CoV-2) has infected over 100million individuals worldwide with diverse impacts on nations. The rising cases of new strains and resultant infection waves create an urgent need to assess the readiness of countries especially in Africa to mitigate the impact on community transmission. This paper delivers a brief synopsis of the novel SARS-CoV-2, emerging cases of new variants reported worldwide, and implications for genetic surveillance of disease transmission in low- and middle-income countries (LMICs) especially Africa. Materials and Methods Literature search used keywords like SARS-CoV-2; COVID-19 epidemiology; pandemic waves; corona outbreak, clinical syndromes, treatments, prevention and control. Cross-sectional and observational studies published on COVID-19 from 2019 till date of study provided main information sources. Databases such as Web of Science, Embase, PubMed and Google Scholar were utilised. Main findings Over 220 countries have documented COVID-19 cases with varied severity till date. Before the spikes in resurgence, a highly virulent mutated (>90% fatality rate) novel strain of COVID-19 had been documented. There is very little data to ascertain the impact of the COVID-19 infection waves in LMICs. Discussion LMICs especially African countries still grapple with significant challenges like inefficient surveillance mechanisms, inadequate vaccination coverage, inadequate enforcement of environmental health strategies, poor health systems etc. Hence, Africa's fate remains dicey in the face of the dynamic evolution of the SARS-CoV-2 and other identified challenges. Conclusion The adoption of a multidisciplinary approach to mitigate the impact of emergence of mutant SARS-CoV-2 variants and resurgence of infection spike is recommended.
Collapse
Affiliation(s)
- Ohia Chinenyenwa M.D
- Department of Environmental Health Sciences, Faculty of Public Health, College of Medicine, University of Ibadan, Nigeria
| | - Bakarey Adeleye Solomon
- Institute for Advanced Medical Research and Training (IAMRAT), College of Medicine, University of Ibadan, Nigeria
| | - Ahmad Tauseef
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing 210096, China
| | - Haroon Haroon
- College of Life Sciences, Northwest University, No. 229, North Taibai Road, Xian, Shaanxi Province, 710069, China
| | - Ana Godson R. E. E.
- Department of Environmental Health Sciences, Faculty of Public Health, College of Medicine, University of Ibadan, Nigeria
| | - Sridhar Mynepalli K.C.
- Department of Environmental Health Sciences, Faculty of Public Health, College of Medicine, University of Ibadan, Nigeria
| |
Collapse
|
47
|
Boulton S, Poutou J, Martin NT, Azad T, Singaravelu R, Crupi MJF, Jamieson T, He X, Marius R, Petryk J, Tanese de Souza C, Austin B, Taha Z, Whelan J, Khan ST, Pelin A, Rezaei R, Surendran A, Tucker S, Fekete EEF, Dave J, Diallo JS, Auer R, Angel JB, Cameron DW, Cailhier JF, Lapointe R, Potts K, Mahoney DJ, Bell JC, Ilkow CS. Single-dose replicating poxvirus vector-based RBD vaccine drives robust humoral and T cell immune response against SARS-CoV-2 infection. Mol Ther 2022; 30:1885-1896. [PMID: 34687845 PMCID: PMC8527104 DOI: 10.1016/j.ymthe.2021.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/24/2021] [Accepted: 10/10/2021] [Indexed: 02/01/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic requires the continued development of safe, long-lasting, and efficacious vaccines for preventive responses to major outbreaks around the world, and especially in isolated and developing countries. To combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we characterize a temperature-stable vaccine candidate (TOH-Vac1) that uses a replication-competent, attenuated vaccinia virus as a vector to express a membrane-tethered spike receptor binding domain (RBD) antigen. We evaluate the effects of dose escalation and administration routes on vaccine safety, efficacy, and immunogenicity in animal models. Our vaccine induces high levels of SARS-CoV-2 neutralizing antibodies and favorable T cell responses, while maintaining an optimal safety profile in mice and cynomolgus macaques. We demonstrate robust immune responses and protective immunity against SARS-CoV-2 variants after only a single dose. Together, these findings support further development of our novel and versatile vaccine platform as an alternative or complementary approach to current vaccines.
Collapse
Affiliation(s)
- Stephen Boulton
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Joanna Poutou
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nikolas T Martin
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Taha Azad
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ragunath Singaravelu
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mathieu J F Crupi
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Taylor Jamieson
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Xiaohong He
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ricardo Marius
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Julia Petryk
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Christiano Tanese de Souza
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Bradley Austin
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Zaid Taha
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jack Whelan
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Sarwat T Khan
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Adrian Pelin
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Reza Rezaei
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Abera Surendran
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Sarah Tucker
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Emily E F Fekete
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jaahnavi Dave
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jean-Simon Diallo
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Rebecca Auer
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jonathan B Angel
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - D William Cameron
- Division of Infectious Disease, Department of Medicine, University of Ottawa at The Ottawa Hospital/ Research Institute, Ottawa, ON K1H 8L6, Canada
| | | | - Réjean Lapointe
- Institut du Cancer de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Kyle Potts
- Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4Z6, Canada; Alberta Children's Hospital Research Institute, Calgary, AB T2N 6A8, Canada; Department of Microbiology, Immunology and Infectious Disease, Cumming School of Medicine, University of Calgary, Calgary, AB T2T 1N4, Canada
| | - Douglas J Mahoney
- Arnie Charbonneau Cancer Institute, Calgary, AB T2N 4Z6, Canada; Alberta Children's Hospital Research Institute, Calgary, AB T2N 6A8, Canada; Department of Microbiology, Immunology and Infectious Disease, Cumming School of Medicine, University of Calgary, Calgary, AB T2T 1N4, Canada
| | - John C Bell
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| | - Carolina S Ilkow
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
48
|
Velappan N, Nguyen HB, Micheva-Viteva S, Bedinger D, Ye C, Mangadu B, Watts AJ, Meagher R, Bradfute S, Hu B, Waldo GS, Lillo AM. Healthy humans can be a source of antibodies countering COVID-19. Bioengineered 2022; 13:12598-12624. [PMID: 35599623 PMCID: PMC9275966 DOI: 10.1080/21655979.2022.2076390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/13/2022] [Accepted: 05/06/2022] [Indexed: 11/05/2022] Open
Abstract
Here, we describe the isolation of 18 unique anti SARS-CoV-2 human single-chain antibodies from an antibody library derived from healthy donors. The selection used a combination of phage and yeast display technologies and included counter-selection strategies meant to direct the selection of the receptor-binding motif (RBM) of SARS-CoV-2 spike protein's receptor binding domain (RBD2). Selected antibodies were characterized in various formats including IgG, using flow cytometry, ELISA, high throughput SPR, and fluorescence microscopy. We report antibodies' RBD2 recognition specificity, binding affinity, and epitope diversity, as well as ability to block RBD2 binding to the human receptor angiotensin-converting enzyme 2 (ACE2) and to neutralize authentic SARS-CoV-2 virus infection in vitro. We present evidence supporting that: 1) most of our antibodies (16 out of 18) selectively recognize RBD2; 2) the best performing 8 antibodies target eight different epitopes of RBD2; 3) one of the pairs tested in sandwich assays detects RBD2 with sub-picomolar sensitivity; and 4) two antibody pairs inhibit SARS-CoV-2 infection at low nanomolar half neutralization titers. Based on these results, we conclude that our antibodies have high potential for therapeutic and diagnostic applications. Importantly, our results indicate that readily available non immune (naïve) antibody libraries obtained from healthy donors can be used to select high-quality monoclonal antibodies, bypassing the need for blood of infected patients, and offering a widely accessible and low-cost alternative to more sophisticated and expensive antibody selection approaches (e.g. single B cell analysis and natural evolution in humanized mice).
Collapse
Affiliation(s)
- Nileena Velappan
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM87547, USA
| | - Hau B. Nguyen
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM87547, USA
| | | | - Daniel Bedinger
- Experimental division, Carterra Inc, Walnut Creek, CA, 94568, USA
| | - Chunyan Ye
- Center for Global Health and Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Betty Mangadu
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, 94551, USA
| | - Austin J. Watts
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM87547, USA
- Experimental division, Carterra Inc, Walnut Creek, CA, 94568, USA
- Center for Global Health and Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, 94551, USA
| | - Robert Meagher
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, 94551, USA
| | - Steven Bradfute
- Center for Global Health and Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Bin Hu
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM87547, USA
| | - Geoffrey S. Waldo
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM87547, USA
| | - Antonietta M. Lillo
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM87547, USA
| |
Collapse
|
49
|
Slattery SS, Giguere DJ, Stuckless EE, Shrestha A, Briere LAK, Galbraith A, Reaume S, Boyko X, Say HH, Browne TS, Frederick MI, Lant JT, Heinemann IU, O'Donoghue P, Dsouza L, Martin S, Howard P, Jedeszko C, Ali K, Styba G, Flatley M, Karas BJ, Gloor GB, Edgell DR. Phosphate-regulated expression of the SARS-CoV-2 receptor-binding domain in the diatom Phaeodactylum tricornutum for pandemic diagnostics. Sci Rep 2022; 12:7010. [PMID: 35487958 PMCID: PMC9051505 DOI: 10.1038/s41598-022-11053-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/18/2022] [Indexed: 12/22/2022] Open
Abstract
The worldwide COVID-19 pandemic caused by the SARS-CoV-2 betacoronavirus has highlighted the need for a synthetic biology approach to create reliable and scalable sources of viral antigen for uses in diagnostics, therapeutics and basic biomedical research. Here, we adapt plasmid-based systems in the eukaryotic microalgae Phaeodactylum tricornutum to develop an inducible overexpression system for SARS-CoV-2 proteins. Limiting phosphate and iron in growth media induced expression of the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein from the P. tricornutum HASP1 promoter in the wild-type strain and in a histidine auxotrophic strain that alleviates the requirement for antibiotic selection of expression plasmids. The RBD was purified from whole cell extracts (algae-RBD) with yield compromised by the finding that 90-95% of expressed RBD lacked the genetically encoded C-terminal 6X-histidine tag. Constructs that lacked the TEV protease site between the RBD and C-terminal 6X-histidine tag retained the tag, increasing yield. Purified algae-RBD was found to be N-linked glycosylated by treatment with endoglycosidases, was cross-reactive with anti-RBD polyclonal antibodies, and inhibited binding of recombinant RBD purified from mammalian cell lines to the human ACE2 receptor. We also show that the algae-RBD can be used in a lateral flow assay device to detect SARS-CoV-2 specific IgG antibodies from donor serum at sensitivity equivalent to assays performed with RBD made in mammalian cell lines. Our study shows that P. tricornutum is a scalable system with minimal biocontainment requirements for the inducible production of SARS-CoV-2 or other coronavirus antigens for pandemic diagnostics.
Collapse
Affiliation(s)
- Samuel S Slattery
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Daniel J Giguere
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Emily E Stuckless
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Arina Shrestha
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Lee-Ann K Briere
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Alexa Galbraith
- Lambton College, 1457 London Rd, Sarnia, ON, N7S 6K4, Canada
| | - Stephen Reaume
- Lambton College, 1457 London Rd, Sarnia, ON, N7S 6K4, Canada
| | - Xenia Boyko
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Henry H Say
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Tyler S Browne
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Mallory I Frederick
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Jeremy T Lant
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Ilka U Heinemann
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Patrick O'Donoghue
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
- Department of Chemistry, Western University, London, ON, N6A 3K7, Canada
| | - Liann Dsouza
- Pond Technologies Inc., Markham, ON, L3R 9W7, Canada
| | - Steven Martin
- Pond Technologies Inc., Markham, ON, L3R 9W7, Canada
| | - Peter Howard
- Pond Technologies Inc., Markham, ON, L3R 9W7, Canada
| | - Christopher Jedeszko
- International Point of Care Inc., 135 The West Mall Unit 9, Toronto, ON, M9C 1C2, Canada
| | - Kinza Ali
- International Point of Care Inc., 135 The West Mall Unit 9, Toronto, ON, M9C 1C2, Canada
| | - Garth Styba
- International Point of Care Inc., 135 The West Mall Unit 9, Toronto, ON, M9C 1C2, Canada
| | - Martin Flatley
- Suncor Energy Inc., Sarnia Refinery, 1900 River Road, Sarnia, ON, N7T 7J3, Canada
| | - Bogumil J Karas
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Gregory B Gloor
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada.
| | - David R Edgell
- Department of Biochemistry, Schlich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada.
| |
Collapse
|
50
|
Conzentino MS, Gonçalves ACA, Paula NM, Rego FGM, Zanette DL, Aoki MN, Nardin JM, Huergo LF. A magnetic bead immunoassay to detect high affinity human IgG reactive to SARS-CoV-2 Spike S1 RBD produced in Escherichia coli. Braz J Microbiol 2022; 53:1263-1269. [PMID: 35426068 PMCID: PMC9009495 DOI: 10.1007/s42770-022-00753-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/08/2022] [Indexed: 12/03/2022] Open
Abstract
Immunological assays to detect SARS-CoV-2 Spike Receptor Binding Domain (RBD) antigen seroconversion in humans are important tools to monitor the levels of protecting antibodies in the population in response to infection and/or immunization. Here we describe a simple, low cost, and high throughput Ni2+ magnetic bead immunoassay to detect human IgG reactive to Spike S1 RBD Receptor Binding Domain produced in Escherichia coli. A 6xHis-tagged Spike S1 RBD was expressed in E. coli and purified by affinity chromatography. The protein was mobilized on the surface of Ni2+ magnetic beads and used to investigate the presence of reactive IgG in the serum obtained from pre-pandemic and COVID-19 confirmed cases. The method was validated with a cohort of 290 samples and an area under the receiver operating characteristic curve of 0.94 was obtained. The method was operated with > 82% sensitivity at 98% specificity and was also able to track human IgG raised in response to vaccination with Comirnaty at > 85% sensitivity. The IgG signal obtained with the described method was well-correlated with the signal obtained when pre fusion Spike produced in HEK cell lines was used as antigen. This novel low-cost and high throughput immunoassay may act as an important tool to investigate protecting IgG antibodies against SARS-CoV-2 in the human population.
Collapse
Affiliation(s)
- Marcelo S Conzentino
- Setor Litoral, UFPR Matinhos, Rua Jaguariaíva, Tv. Caiobá, 512, Matinhos, 83260-000, PR, Brazil
| | - Ana C A Gonçalves
- Setor Litoral, UFPR Matinhos, Rua Jaguariaíva, Tv. Caiobá, 512, Matinhos, 83260-000, PR, Brazil
| | - Nigella M Paula
- Setor Litoral, UFPR Matinhos, Rua Jaguariaíva, Tv. Caiobá, 512, Matinhos, 83260-000, PR, Brazil
| | - Fabiane G M Rego
- Post-Graduation Program in Pharmaceutical Sciences, UFPR, Curitiba, PR, Brazil
| | | | - Mateus N Aoki
- Instituto Carlos Chagas - FioCruz, Curitiba, PR, Brazil
| | | | - Luciano Fernandes Huergo
- Setor Litoral, UFPR Matinhos, Rua Jaguariaíva, Tv. Caiobá, 512, Matinhos, 83260-000, PR, Brazil.
| |
Collapse
|