Antibodies Produced Toward Recombinant RBD and Nucleocapsid Neutralize SARS-COV-2


PDF - Export to EndNote - PubMed Central XML format - PubMed Central XML format - PubMed Central XML format
PMID: 36504571 (PubMed) - PMCID: PMC9706246 - View online: PubReader
Volume 14, Issue 4, October-December , Page 270 to 277
Wednesday, February 16, 2022 :Received , Monday, May 30, 2022 :Accepted



  • - Department of Biology, Shahed University, Tehran, Iran
  • Corresponding author Molecular Biotechnology Research Center and Department of Biology, Imam Hussein University, Tehran, Iran, Tel: +98 21 51212232; Fax: +98 21 51212232; E-mail: nazarian56@gmail.com
  • Corresponding author Department of Biology, Shahed University, Tehran, Iran, Tel: +98 21 51212232; Fax: +98 21 51212232; E-mail: slmousavi@shahed.ac.ir

Abstract:

Background: The highly contagious SARS-COV-2 virus spread rapidly from China and formed a global pandemic. The virus has infected over 509 million people worldwide and killed about 6.32 million up to date. Up on invasion, the Receptor Binding Domain (RBD) of Spike protein plays a crucial role in the entry of the virus into the host cell. The virus N protein is another protein that has a critical role for genome packaging.

Methods: As bioinformatics approaches, the cassette design, codon adaptation, and protein stability were investigated in this study. Synthetic genes of RBD and N were cloned separately in pET28a + expression vector. They were transferred into Escherichia coli (E. coli) BL21 DE3 host cell, and expression of recombinant proteins was induced with IPTG. The recombinant proteins were purified by column chromatography and approved by Western blotting. Animal immunization was performed with each of the recombinant proteins individually and in combination of the two. The antibody titer of the blood serum from control and immunized mice groups was determined by ELISA technique. Finally, the anti-spike neutralization test was performed.

Results: The expression and purification of RBD protein were monitored on SDS-PAGE, two bands of about 28 and 45 kDa for RBD and N appeared on gel distinctly, which were further validated by Western blotting. According to ELISA results, related antibodies were traced to a dilution of 1/64000 in immunized sera. The neutralization test exhibited produced antibodies' potency to bind the virus proteins. Using SPSS software, statistical analysis was performed by Duncan's test and T-test.

Conclusion: According to the present study, recombinant proteins, either RBD alone or in combination with N adequately stimulated the immune response, and the raised antibodies could neutralize the virus in in vitro test.


 

 


Introduction :

In 1960, the first human coronaviruses were discovered during research into cold-causing viruses in the United States and Britain 1,2. On December 29, 2019, hospital doctors in Wuhan, China, noticed unusual cases of patients with pneumonia 3. The ongoing coronavirus disease 2019 (COVID-19) pandemic became a serious public health crisis, caused by Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) 4. The primary host for the virus appears to be bats, however other organisms like pigs and snakes, can serve as a host 5,6. To date, many research groups have tried to find effective ways to fight the virus 7. According to the World Health Organization (WHO) report, until August 2021, the number of infected people was about 200 million and the number of dead was about 2.4 million (https://covid19.who.int); while the number of infected and dead population is increasing daily. Accordingly, developing efficient vaccines against the virus was considered as a priority, yet.

Coronavirus is a spherical virus that looks like a crown due to the large and small proteins on its surface 2,8. It is a small RNA-positive virus with a length of 29736 nucleotides; its genome has 79.5% similarity with SARS virus. The genome encodes four main structural proteins of the virus: Spike (S), Envelope (E), Membrane (M), and Nucleocapsid (N). Among them, Spike is more crucial and prepares the virus for invadind and attaching to the host cell 9,10. The N protein with 419 amino acids has more expression compared to other viral proteins  and its immunogenic antibodies have been observed in infected patients 11-13. This protein surrounds the virus genome, and following the virus entrance in to host cell, the protein is separated and RNA is released into the cytoplasm 14. The N protein also acts as a chaperone and is essential for viral assembly 15,16. On average, there are 74 spike proteins on the surface of coronavirus 17. It divides into two parts; one part is responsible for attachment to the host cell and entrance. Another part is related to the placement and attachment to the virus membrane. The new coronavirus (COVID-19) uses the Angiotensin-Converting Enzyme 2 (ACE2) receptor to enter the host cell similar to that of  SARS virus 18. The virus enters through the Receptor-Binding Domain (RBD) in the S1 subunit. The SPIKE protein detects Lys 31 in ACE2 through Gln 394 in the RBD domain 18,19. After binding to the ACE2, the spike is cleaved  by acid-dependent proteolysis of Human Airway Trypsin-like protease (HAT) and cathepsin and transmembrane protease Ser 2 (TMPRSS2), into two subunits of S1 and S2 20. The S1 fragment consists the RBD section and the S2 fragment contains the C-terminal with the HR1 and HR2 (Heptad Repeat) sequences 21,22. The RBD is the target of many primary neutralizing antibodies, for preventing the virus from binding to the receptor (ACE2/DPP4). Therefore, RBD of spike can be a proper candidate for vaccine development against coronavirus. It has been reported that co-administration of protein subunits increases the immunogenicity compared to single administrations 23. Therefore, in the present study, RBD and N proteins were expressed in Escherichia coli (E. coli), and purified discretely. The recombinant RBD, alone or in RBD-N combinations were compared as immunogenic candidates against SARS-COVID-2 in mice model. The antibodies produced were used in virus neutralization tests.

 


Materials and Methods :

In silico design of the construct: Bioinformatics tools have a great impact on the design analysis of immunogens. RBD and N protein sequences with accession numbers NC_045512.2 and sp-P0DTC9 were obtained from the GeneBank and UNIPROT databases in bioinformatics approaches. The sequences were aligned through the BLAST tools at the GeneBank site for confidence. PROTPARAM tools of EXPASY analyzed structural and physical information of the proteins, and the I-TASSER software was utilized to predict the three dimensional structure of proteins. The Ramachandran plot tool in the RAMPAGE server was utilized to confirm protein structures by measuring the statistical distribution of the combinations of the backbone dihedral angles ϕ and ψ. BCEpred online software was employed to predict the induction of humoral immunity, and VaxiJen online software was used to estimate the overall antigenicity of the sequences.

Expression and purification of the recombinant proteins: The synthetic genes were obtained on pUC18 and subcloned into the pET28a vector with Nedl and BamHl restriction sites at Imam Hussein University, Tehran, Iran. The constructs were transformed into E. coli Bl21 (DE3) and cultured on an LB agar plate containing 70 µg/mL kanamycin.

Cells were induced with 1 mM Isopropyl β-d-1- thiogalactopyranoside (IPTG) for five hours. Cells were harvested and lysed with sonication. The recombinant proteins as inclusion bodies were dissolved in phosphate buffer (100 mM NaH2PO4, 10 mM Tris-HCl) containing 8 M urea and were analyzed on 15% SDS-PAGE.

Recombinant proteins were purified using Nickel-nitrilotriacetic acid (Ni-NTA) affinity chromatography. Each column was equilibrated with phosphate buffer and loaded with approximately 500 µg of protein. The column for protein N was washed five times with a 20 mM concentration of imidazole, and the recombinant protein was eluted with a 250 mM concentration of imidazole. To purify the RBD protein, the column was washed seven times with phosphate buffers: pH=6.3 (C buffer), pH=5.5 (D buffer), and pH=4 (E buffer). The recombinant protein was eluted with 250 µl of E buffer. All collected fractions were analyzed on the SDS-PAGE, and concentration proteins was assessed with Bradford method.

Western blot analysis: The purified recombinant proteins were transformed onto the nitrocellulose membrane with transfer buffer (39 mM glycine, 48 mM Tris-base, 0.037% SDS, and 20% methanol). The membrane was blocked with blocking buffer (PBST+5% Skimmed Milk Powder) for 15 hr at 4°C and washed three times with PBST (137 mM NaCl, 2.7 mM KCl, 4.3 mM Na2HPO4+0.05% Tween). Nitrocellulose membranes were soaked in mice anti his-tag IgG (1:10000), and incubated for one hour at 37°C. Membranes were washed and added with DAB (3, 3´-diaminobenzidine) as a chromogenic substrate. The reaction was stopped by distilled water.

Animal immunization: Twenty-four female BALB/c mice (20-25 gr, 5-week old) were obtained from Tehran University, Tehran, Iran. The mice were divided into four groups. Two groups each with nine mice for immunization, were named as RBD and RBD-N groups. Two other groups each with three mice were kept as control groups. Mice were kept in the animal care facility of Shahed University under standard and ventilated conditions. The principles in the guide for the care and use of laboratory animals were followed. All animals were kept in compliance with the Welfare act (Helsinki and its later amendments). The animal care rule was ethically certified by Shahed University with the ethical code of IR.SHAHED.REC.1400.018.

Mice in RBD-N group were subcutaneously injected with 100 µl of recombinant proteins mixed with equal volume of Allum adjuvant on three boosters on days 0, 15, and 30. The control group received Phosphate-Buffered Saline (PBS) with Allum adjuvant (Table 1). Blood samples were collected from the eye corners of mice on days 14, 29, and 44 after the first injection. The sera were collected and stored at -20°C.

Antibody response: Enzyme-linked Immunosorbent Assay (ELISA) technique was applied to determine antibody response. 5 µg of each recombinant protein (in 100 µl of coating buffer) was coated in ELISA wells and incubated at 4°C overnight. The wells were blocked with skimmed milk, and then the serial dilution (1:100 to 1:64000 in PBST) of serum antibodies was added to the wells and incubated at 37°C for 2 hr. Anti-mouse IgG HRP conjugate (1/2000 dilution in PBST) was added to each well and incubated at 37°C for 90 min. 100 µl Tetramethylbenzidine (TMB) was added to each well and incubated at room temperature for 10 min. Wells were washed after each incubation. The reactions were stopped with 3N H2SO4, and the OD450 was recorded.

Neutralization tests: The neutralization of the SARS-CoV-2 was performed using a laboratory kit supplied by PishtazTeb Zaman Diagnostics. This kit contains the virus spike protein and ACE2 receptors. The ACE2 receptors in the kit will not be able to bind to the spike proteins in the presence of anti-RBD antibodies. Tests were performed according to the protocol provided by the supplier.

Statistical analysis: The data in each figure was a representative of three independent experiments expressed as the mean± standard deviation (SD). All statistical analyses were performed using a SPSS 12.0 statistical program. Student t-test was used to analyze the data for antibody responses between immunized and non-immunized groups. A value of p<0.05 was considered statistically significant.

 


Results :

Bioinformatics analysis: Validated sequences were obtained from the GenBank database. The three-dimensional structures of proteins were predicted by the I-TASSER software (Figure 1). According to this tool's description, a Confidence score (C-score) is utilized for estimating the quality of predicted models by the server. Its algorithm is based on the significance of threading template alignments and the convergence parameters of the structure assembly simulations. C-score is typically in the range of 2-5, where a C-score of higher value signifies a model with high confidence and vice-versa. RBD protein structure was predicted with a C-score of 1.05, and for the N protein, it was -3.56. The Ramachandran plot quality assessment analysis showed that most RBD protein residues, i.e., more than 98%, are in the favored + allowed region. Most N protein residues, i.e., more than 99%, are in the favored area as well and hence these structures can be reliable. The B cell epitopes were also predicted as shown in tables 2 and 3.

 Expression and purification of the recombinant proteins: Expression and purification of recombinant RBD (28 kDa) and N (45 kDa) proteins was analysed on SDS-PAGE. The SDS-PAGE and Western blotting method verified the recombinant proteins (Figures 2-5).

Determination of serum IgG titer: Indirect-ELISA was performed to evaluate the raised IgG antibody titers of sera. After second blood sampling, antibody titers measurement exhibited a significant increase in the test groups compared to the control ones. High antibody titers were observed in both experimental groups until day 87 following the first injection. The RBD-N group developed more robust and more persistent immunogenicity than the RBD alone (Figure 6).

Neutralization tests: According to the supplier's instructions, if the average concentration of antibodies from the vaccine is more than 2.5 µg/ml, the neutralization test is considered positive. The mean light absorption of antibodies from the RBD single vaccine in virus neutralization was 0.6186, corresponding to 19.48 µg/ml of RBD antigen alone. The mean light absorption of antibodies from the combination of RBD-N antigens in virus neutralization was 0.3949, corresponding to 25.17 µg/ml of RBD-N mixture.

 


Discussion :

Since June 2020, about 157 vaccine candidates have been tested for the COVID-19 in laboratories worldwide. Several vaccines, including inactivated viruses, adenovirus vectors containing the spike protein, and mRNA vaccines against the virus have been used in clinical and commercial development 24-26. Because of their safety concerns and high efficacy, about 53% of researches have focused on recombinant vaccines 27. Available research data from the Mers virus indicates that inactivated viruses and recombinant vaccines efficiently neutralize viruse activity 28. Today, inactivated viruses are produced by UV and formaldehyde treatment. However, viruses such as corona cannot be cultivated at any laboratory and require a level three laboratory, which is challenging to establish and maintain due to the high cost. At high-level production of attenuated virus, checking each batch's inactivation is another problem concerned with these vaccines 29,30. Furthermore, inactive microbes cannot multiply in the host, and they must be used with adjuvants and administered in multiple doses of the vaccine 31. Some antibodies are produced against parts of the pathogen but having no role in causing the disease. In this case, cytotoxic T cells are not produced and can not be used to stop infections by intracellular pathogens 32. Animals vaccinated with inactivated bacteria developed fever at the end of the experiment, whereas animals immunized with the recombinant protein showed no such side effects 33. S1, S2, and RBD were the first recombinant vaccine candidates, of which RBD remains an essential candidate for primary vaccines and boosters due to the high neutralizing effect and CD4+ T cell epitopes 34-37. Interestingly, the RBD effectively boost immune responses against emerging variants 38. The RBD domain is the most concentrated area of epitopes in most articles and has been regarded as an attractive immunogen 39-41. In contrast, the S1 protein induced a lower neutralization titer than the RBD protein 42 and hence the RBD is a better candidate than S1 for SARS-CoV-2 vaccine design. The toxicity of recombinant RBD protein is very low and has no effect on weight, appetite, and behavior. Also, it has no adverse pathological effects on the brain, liver, kidneys, or heart and does not reduce the number of blood cells 42. In these experiments, aluminum adjuvants were selected as one of the best immunogenic adjuvants, and unlike Freund's adjuvant, this adjuvant can also be used for humans 42,43.

Recombinant RBD and N proteins were expressed in the bacterial host at approximately 40 mg/L for RBD and 50 mg/L for N protein. The average expression of RBD and N in eukaryotic hosts are 45 mg/L and 50 mg/L, respectively 44-48. Despite the glycosylation in eukaryotic cells and the sugar removal process, it seems appropriate for expressing these proteins in the bacterial host. In addition, these proteins maintain their stability at -70°C and can be stored for a long time.

In our study, the average percentage of neutralization by the serum induced toward RBD alone was 48.7%, and increased to about 63% with serum of mice immunized both with RBD and N proteins. The increased percentage of neutralization in the presence of N can be attributed to the adjuvant role of N rather than its role as an immunogen. In a similar work, antibodies from the RBD vaccine prevented the protein from binding to its receptor by 20 to 40% 49.

 


Conclusion :

Combination vaccines are a new way to make vaccines safer, more durable, and more effective. Recombinant measles viruses containing proteins S and N on their surface provided better immunity 50. The immunogenicity of the single RBD vaccine has been compared with the RBD-dimer. The RBD monomer acts as a poor immunogen and triggers low levels of IgG and neutralization results 51. Data presented here and the results obtained by other researchers, shows that RBD as a recombinant vaccine candidate, with its simple, rapid, and economical preparation, might be an effective and convenient strategy to control COVID-19 epidemics.

 


Acknowledgement :

The authors are thankful to Shahed University and Imam Hussein University for supporting this work.

 


Conflict of Interest :

The authors declare no conflict of interest.

 



<p>Figure 1. A) Three-dimensional structure of RBD. B) three-dimen-sional structure of N protein.</p>

Figure 1. A) Three-dimensional structure of RBD. B) three-dimen-sional structure of N protein.





<p>Figure 2. A) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of RBD expressions: Lane 1: RBD clone uninduced with IPTG as control extract in B buffer, Lane 2: RBD clone induced with 0/5 <em>mM</em> IPTG, 4 <em>hr</em> and extract in B buffer, Lane 3: RBD clone induced with 1 <em>mM</em> IPTG, 4 <em>hr</em> and extract in PBS buffer, Lane 4: RBD clone induced with 1 <em>mM</em> IPTG, 4 <em>hr</em> and extract in B buffer, Lane 5: Molecular weight marker, L ane 6: RBD clone induced with 0/5 <em>mM</em> IPTG, over night and extract in B buffer, Lane 7: RBD clone induced with 1 <em>mM</em> IPTG, over night and extract in B buffer. B) Western Blot analysis of the RBD protein, Lane 1: Molecular weight marker, Lane 2: RBD protein.</p>

Figure 2. A) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of RBD expressions: Lane 1: RBD clone uninduced with IPTG as control extract in B buffer, Lane 2: RBD clone induced with 0/5 mM IPTG, 4 hr and extract in B buffer, Lane 3: RBD clone induced with 1 mM IPTG, 4 hr and extract in PBS buffer, Lane 4: RBD clone induced with 1 mM IPTG, 4 hr and extract in B buffer, Lane 5: Molecular weight marker, L ane 6: RBD clone induced with 0/5 mM IPTG, over night and extract in B buffer, Lane 7: RBD clone induced with 1 mM IPTG, over night and extract in B buffer. B) Western Blot analysis of the RBD protein, Lane 1: Molecular weight marker, Lane 2: RBD protein.





<p>Figure 3. A) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of N expressions: lane1: N clone uninduced with IPTG as control. Lane 2: N clone induced with Isopropyl &beta;-d-1-thiogalactopyranoside (IPTG), lane 3: Molecular weight marker. B) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of N purification: lane 1: Pre Flu, lane 2: Flu, lane 3: I40 (1 <em>ml</em>), lane 4: I100 (1 <em>ml</em>), lane 5: I150 (1 <em>ml</em>), lane 6: I250 (500 <em>&mu;</em><em>l</em>).</p>

Figure 3. A) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of N expressions: lane1: N clone uninduced with IPTG as control. Lane 2: N clone induced with Isopropyl β-d-1-thiogalactopyranoside (IPTG), lane 3: Molecular weight marker. B) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of N purification: lane 1: Pre Flu, lane 2: Flu, lane 3: I40 (1 ml), lane 4: I100 (1 ml), lane 5: I150 (1 ml), lane 6: I250 (500 μl).





<p>Figure 4. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of RBD purification. Lane 1: sample before purification, lane 2: Flu, lane 3: Molecular weight marker, lane 4: C(2 <em>ml</em>), lane 5: D(2 <em>ml</em>), lane 6: E1(500 <em>&mu;l</em>), lane 7: E2(500 <em>&mu;l</em>), lane 8: E3(500 <em>&mu;l</em>), lane 9: E4(500 <em>&mu;l</em>), lane 10: Cu Buffer(1 <em>ml</em>).</p>

Figure 4. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of RBD purification. Lane 1: sample before purification, lane 2: Flu, lane 3: Molecular weight marker, lane 4: C(2 ml), lane 5: D(2 ml), lane 6: E1(500 μl), lane 7: E2(500 μl), lane 8: E3(500 μl), lane 9: E4(500 μl), lane 10: Cu Buffer(1 ml).





<p>Figure 5. Western Blot analysis of the N protein, Lane 1: Molecular weight marker, lane 2: N protein.</p>

Figure 5. Western Blot analysis of the N protein, Lane 1: Molecular weight marker, lane 2: N protein.





<p>Figure 6. Linear and bar charts compare ELISA antibody titers in three blood samples in safe and control mice. A-1) Line chart of ELISA antibody titer obtained from single RBD vaccine in three blood samples 2 weeks apart and final blood sample 45 days after the last immunization. A-2) Column chart of ELISA antibody titer obtained from single RBD vaccine in three blood samples 2 weeks apart and final blood sample 45 days after the last immunization. B-1) Line chart of ELISA antibody titer from RBD &amp; N combination vaccine in three blood samples 2 weeks apart and final blood sample 45 days after the last immunization. B-2) Column chart of ELISA antibody titer from RBD &amp; N combination vaccine in three blood samples 2 weeks apart and final blood sample 45 days after the last immunization. C-1) Comparison of linear graphs of ELISA antibody titers from RBD single vaccine and RBD &amp; N combination vaccine in the third blood sample. C-2) Comparison of Column graphs of ELISA antibody titers from RBD single vaccine and RBD &amp; N combination vaccine in the third blood sample.</p>

Figure 6. Linear and bar charts compare ELISA antibody titers in three blood samples in safe and control mice. A-1) Line chart of ELISA antibody titer obtained from single RBD vaccine in three blood samples 2 weeks apart and final blood sample 45 days after the last immunization. A-2) Column chart of ELISA antibody titer obtained from single RBD vaccine in three blood samples 2 weeks apart and final blood sample 45 days after the last immunization. B-1) Line chart of ELISA antibody titer from RBD & N combination vaccine in three blood samples 2 weeks apart and final blood sample 45 days after the last immunization. B-2) Column chart of ELISA antibody titer from RBD & N combination vaccine in three blood samples 2 weeks apart and final blood sample 45 days after the last immunization. C-1) Comparison of linear graphs of ELISA antibody titers from RBD single vaccine and RBD & N combination vaccine in the third blood sample. C-2) Comparison of Column graphs of ELISA antibody titers from RBD single vaccine and RBD & N combination vaccine in the third blood sample.





<p>Table 1. Mouse immunization protocol</p>

Table 1. Mouse immunization protocol





<p>Table 2. B-cell epitopes from RBD protein; using BCEPred</p>
<p>Antigenicity of RBD protein; using VaxiJen</p>

Table 2. B-cell epitopes from RBD protein; using BCEPred

Antigenicity of RBD protein; using VaxiJen





<p>Table 3. B-cell epitopes from N protein; using BCPred</p>
<p>Antigenicity of N protein; using VaxiJen.</p>

Table 3. B-cell epitopes from N protein; using BCPred

Antigenicity of N protein; using VaxiJen.






References :
  1. Kahn JS, McIntosh K. History and recent advances in coronavirus discovery. Pediatr Infect Dis J 2005;24(11 Suppl):S223-7, discussion S6.   [PubMed]
  2. Mahase E. Covid-19: Coronavirus was first described in The BMJ in 1965. BMJ 2020;369:m1547.   [PubMed]
  3. Smith TRF, Patel A, Ramos S, Elwood D, Zhu X, Yan J, et al. Immunogenicity of a DNA vaccine candidate for COVID-19. Nat Commun 2020;11(1):2601.   [PubMed]
  4. Zang J, Gu C, Zhou B, Zhang C, Yang Y, Xu S, et al. Immunization with the receptor-binding domain of SARS-CoV-2 elicits antibodies cross-neutralizing SARS-CoV-2 and SARS-CoV without antibody-dependent enhancement. Cell Discov 2020;6:61.   [PubMed]
  5. Ji W, Wang W, Zhao X, Zai J, Li X. Cross‐species transmission of the newly identified coronavirus 2019‐nCoV. J Med Virol 2020;92(4):433-40.   [PubMed]
  6. Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020;579(7798):270-3.   [PubMed]
  7. Hamre D, Procknow JJ. A new virus isolated from the human respiratory tract. Proc Soc Exp Biol Med 1966;121(1):190-3.   [PubMed]
  8. Goldsmith CS, Tatti KM, Ksiazek TG, Rollin PE, Comer JA, Lee WW, et al. Ultrastructural characterization of SARS coronavirus. Emerg Infect Dis 2004;10(2):320-6.   [PubMed]
  9. Weiss SR, Navas-Martin SJM, reviews mb. Coronavirus pathogenesis and the emerging pathogen severe acute respiratory syndrome coronavirus. Microbiol Mol Biol Rev 2005;69(4):635-64.   [PubMed]
  10. Hussain A, Kaler J, Tabrez E, Tabrez S, Tabrez SSM. Novel COVID-19: A comprehensive review of transmission, manifestation, and pathogenesis. Cureus 2020;12(5):e8184.   [PubMed]
  11. Chang CK, Hou MH, Chang CF, Hsiao CD, Huang TH. The SARS coronavirus nucleocapsid protein–forms and functions. Antiviral Res 2014;103:39-50.   [PubMed]
  12. Li D, Li JJJocm. Immunologic testing for SARS-CoV-2 infection from the antigen perspective. J Clin Microbiol 2020;59(5):e02160-20.   [PubMed]
  13. Ye Q, West AM, Silletti S, Corbett KDJPS. Architecture and self‐assembly of the SARS‐CoV‐2 nucleocapsid protein. Protein Sci 2020;29(9):1890-901.   [PubMed]
  14. Glowacka I, Bertram S,Müller MA, Allen P, Soilleux E, Pfefferle S, et al. Evidence that TMPRSS2 activates the severe acute respiratory syndrome coronavirus spike protein for membrane fusion and reduces viral control by the humoral immune response. J Virol 2011; 85(9):4122–34.   [PubMed]
  15. McBride R, Van Zyl M, Fielding BCJV. The coronavirus nucleocapsid is a multifunctional protein. Viruses 2014;6(8):2991-3018.
  16. Chang CK, Lo SC, Wang YS, Hou MH. Recent insights into the development of therapeutics against coronavirus diseases by targeting N protein. Drug Discov Today 2016;21(4):562-72.   [PubMed]
  17. De Groot RJ, Baker SC, Baric RS, Brown CS, Drosten C, Enjuanes L, et al. Commentary: Middle east respiratory syndrome coronavirus (mers-cov): announcement of the coronavirus study group. J Virol 2013;87(14):7790-2.   [PubMed]
  18. Zhang J, Zeng H, Gu J, Li H, Zheng L, Zou Q. Progress and prospects on vaccine development against SARS-CoV-2. Vaccines (Basel) 2020;8(2):153.   [PubMed]
  19. Shereen MA, Khan S, Kazmi A, Bashir N, Siddique R. COVID-19 infection: Origin, transmission, and characteristics of human coronaviruses. J Adv Res 2020;24:91-8.   [PubMed]
  20. Walls AC, Park Y-J, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell 2020;181(2): 281-92.e6.   [PubMed]
  21. Weiss SR, Navas-Martin S. Coronavirus pathogenesis and the emerging pathogen severe acute respiratory syndrome coronavirus. Microbiol Mol Biol Rev 2005;69(4):635-64.   [PubMed]
  22. Lalchhandama K. The chronicles of coronaviruses: the electron microscope, the doughnut, and the spike. Science Vision 2020;20(2):78-92.
  23. Vance DJ, Rong Y, Brey RN 3rd, Mantis NJ. Combination of two candidate subunit vaccine antigens elicits protective immunity to ricin and anthrax toxin in mice. Vaccine 2015;33(3):417-21.   [PubMed]
  24. Mulligan MJ, Lyke KE, Kitchin N, Absalon J, Gurtman A, Lockhart S, et al. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature 2020;586(7830):589-93.   [PubMed]
  25. Gao Q, Bao L, Mao H, Wang L, Xu K, Yang M, et al. Development of an inactivated vaccine candidate for SARS-CoV-2. Science 2020;369(6499):77-81.   [PubMed]
  26. Zhu FC, Li YH, Guan XH, Hou LH, Wang WJ, Li JX, et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. Lancet 2020;395(10240):1845-54.   [PubMed]
  27. Shin MD, Shukla S, Chung YH, Beiss V, Chan SK, Ortega-Rivera OA, et al. COVID-19 vaccine development and a potential nanomaterial path forward. Nat Nanotechnol 2020;15(8):646-55.   [PubMed]
  28. Deng Y, Lan J, Bao L, Huang B, Ye F, Chen Y, et al. Enhanced protection in mice induced by immunization with inactivated whole viruses compare to spike protein of middle east respiratory syndrome coronavirus. Emerg Microbes Infect 2018;7(1):60.   [PubMed]
  29. Zhou J, Wang W, Zhong Q, Hou W, Yang Z, Xiao SY, et al. Immunogenicity, safety, and protective efficacy of an inactivated SARS-associated coronavirus vaccine in rhesus monkeys. Vaccine 2005;23(24):3202-9.   [PubMed]
  30. Tsunetsugu-Yokota Y. Large-scale preparation of UV-inactivated SARS coronavirus virions for vaccine antigen. Methods Mol Biol 2008;454:119-26.   [PubMed]
  31. Zhao J, Zhao S, Ou J, Zhang J, Lan W, Guan W, et al. COVID-19: vaccine development updates. Front Immunol 2020;11:3435.   [PubMed]
  32. Baxter DJOm. Active and passive immunity, vaccine types, excipients and licensing. Occup Med (Lond) 2007;57(8):552-6.   [PubMed]
  33. Tamer C, Albayrak H, Gumusova SJAR. Comparison of immune response between Escherichia coli‐derived recombinant subunit vaccine and formol‐inactivated whole particle vaccine against viral haemorrhagic septicaemia virus (VHSV) in rainbow trout. Aquaculture Research 2021;52(6):2706-14.
  34. Dejnirattisai W, Zhou D, Ginn HM, Duyvesteyn HM, Supasa P, Case JB, et al. The antigenic anatomy of SARS-CoV-2 receptor binding domain. Cell 2021;184(8):2183-200. e22.   [PubMed]
  35. Starr TN, Greaney AJ, Dingens AS, Bloom JDJCRM. Complete map of SARS-CoV-2 RBD mutations that escape the monoclonal antibody LY-CoV555 and its cocktail with LY-CoV016. Cell Rep Med 2021;2(4):100255.   [PubMed]
  36. Liu C, Ginn HM, Dejnirattisai W, Supasa P, Wang B, Tuekprakhon A, et al. Reduced neutralization of SARS-CoV-2 B. 1.617 by vaccine and convalescent serum. Cell 2021;184(16):4220-36. e13.   [PubMed]
  37. Low JS, Vaqueirinho D, Mele F, Foglierini M, Jerak J, Perotti M, et al. Clonal analysis of immunodominance and cross-reactivity of the CD4 T cell response to SARS-CoV-2. Science 2021;372(6548):1336-41.   [PubMed]
  38. Tan H-X, Juno JA, Lee WS, Barber-Axthelm I, Kelly HG, Wragg KM, et al. Immunogenicity of prime-boost protein subunit vaccine strategies against SARS-CoV-2 in mice and macaques. Nat Commun 2021;12(1):1403.   [PubMed]
  39. Zumla A, Chan JFW, Azhar EI, Hui DSC, Yuen KY. Coronaviruses—drug discovery and therapeutic options. Nat Rev Drug Discov 2016;15(5):327-47.   [PubMed]
  40. Li Y, Lai DY, Zhang HN, Jiang HW, Tian X, Ma Ml, et al. Linear epitopes of SARS-CoV-2 spike protein elicit neutralizing antibodies in COVID-19 patients. Cell Mol Immunol 2020;17(10):1095-7.   [PubMed]
  41. Li Y, Ma Ml, Lei Q, Wang F, Hong W, Lai DY, et al. Linear epitope landscape of the SARS-CoV-2 spike protein constructed from 1,051 COVID-19 patients. Cell Rep 2021;34(13):108915.   [PubMed]
  42. Yang J, Wang W, Chen Z, Lu S, Yang F, Bi Z, et al. A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunity. Nature 2020;586(7830):572-7.   [PubMed]
  43. Hotez PJ, Corry DB, Strych U, Bottazzi MEJNRI. COVID-19 vaccines: neutralizing antibodies and the alum advantage. Nat Rev Immunol 2020;20(7):399-400.   [PubMed]
  44. Arbeitman CR, Auge G, Blaustein M, Bredeston L, Corapi ES, Craig PO, et al. Structural and functional comparison of SARS-CoV-2-spike receptor binding domain produced in Pichia pastoris and mammalian cells. Sci Rep 2020;10(1):21779.   [PubMed]
  45. Limonta-Fernández M, Chinea-Santiago G, Martín-Dunn AM, Gonzalez-Roche D, Bequet-Romero M, Marquez-Perera G, et al. The SARS-CoV-2 receptor-binding domain expressed in Pichia pastoris as a candidate vaccine antigen. medRxiv 2021.06.29.21259605.
  46. Liu B, Yin Y, Liu Y, Wang T, Sun P, Ou Y, et al. A vaccine based on the receptor-binding domain of the spike protein expressed in glycoengineered Pichia pastoris targeting SARS-CoV-2 stimulates neutralizing and protective antibody responses. Engineering (Beijing) 2021.   [PubMed]
  47. Li G, Li W, Fang X, Song X, Teng S, Ren Z, et al. Expression and purification of recombinant SARS-CoV-2 nucleocapsid protein in inclusion bodies and its application in serological detection. Protein Expr Purif 2021;186:105908.   [PubMed]
  48. García-Cordero J, Mendoza-Ramírez J, Fernández-Benavides D, Roa-Velazquez D, Filisola-Villaseñor J, Martínez-Frías SP, et al. Recombinant protein expression and purification of N, S1, and RBD of SARS-CoV-2 from mammalian cells and their potential applications. Diagnostics (Basel) 2021;11(10):1808.   [PubMed]
  49. Huang WC, Zhou S, He X, Chiem K, Mabrouk MT, Nissly RH, et al. SARS‐CoV‐2 RBD neutralizing antibody induction is enhanced by particulate vaccination. Adv Mater 2020;32(50):2005637.
  50. Liniger M, Zuniga A, Tamin A, Azzouz-Morin TN, Knuchel M, Marty RR, et al. Induction of neutralising antibodies and cellular immune responses against SARS coronavirus by recombinant measles viruses.Vaccine 2008;26(17):2164-74.   [PubMed]
  51. Dai L, Zheng T, Xu K, Han Y, Xu L, Huang E, et al. A universal design of betacoronavirus vaccines against COVID-19, MERS, and SARS. Cell 2020;182(3):722-33. e11.   [PubMed]