key: cord-0883875-r4fitfos authors: Yang, Jihyun; Kim, Eunjin; Lee, Jong-Soo; Poo, Haryoung title: A Murine CD8(+) T Cell Epitope Identified in the Receptor-Binding Domain of the SARS-CoV-2 Spike Protein date: 2021-06-11 journal: Vaccines (Basel) DOI: 10.3390/vaccines9060641 sha: e96451938591aec7c7a0394cd3f2c48fe60b539d doc_id: 883875 cord_uid: r4fitfos The ongoing COVID-19 pandemic caused by SARS-CoV-2 has posed a devastating threat worldwide. The receptor-binding domain (RBD) of the spike protein is one of the most important antigens for SARS-CoV-2 vaccines, while the analysis of CD8 cytotoxic T lymphocyte activity in preclinical studies using mouse models is critical for evaluating vaccine efficacy. Here, we immunized C57BL/6 wild-type mice and transgenic mice expressing human angiotensin-converting enzyme 2 (ACE2) with the SARS-CoV-2 RBD protein to evaluate the IFN-γ-producing T cells in the splenocytes of the immunized mice using an overlapping peptide pool by an enzyme-linked immunospot assay and flow cytometry. We identified SARS-CoV-2 S(395–404) as a major histocompatibility complex (MHC) class I-restricted epitope for the RBD-specific CD8 T cell responses in C57BL/6 mice. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative pathogen of human coronavirus disease 2019 (COVID-19) [1, 2] . COVID-19 was declared as a pandemic by the WHO in 2020 and has inflicted tremendous negative impacts on global health, society, and economic institutions. As vaccination is considered to be one of the most effective methods for preventing the rapid global spread of this disease, many researchers and companies in the world are attempting to develop safe and effective COVID-19 vaccines [3] . Most COVID-19 vaccine candidates have focused on the SARS-CoV-2 spike protein or the receptor-binding domain (RBD) of the spike protein as the vaccine antigen [4] . The SARS-CoV-2 RBD primarily binds to angiotensin-converting enzyme 2 (ACE2) and facilitates the entry of the virus into host cells [5] . The protective efficacy of the vaccine is conferred by the induction of humoral immunity such as neutralizing antibodies and cellmediated immunity. CD8 cytotoxic T lymphocytes (CTLs) play a critical role in eliminating virus-infected cells, thereby leading to the effective clearance of viral infection. In particular, CTL activity is important to provide long-term protection from reinfection of the virus [6] . Recently, a clinical report also showed the importance of T cell immunity in COVID-19 recovery [7] . Of patients with severe COVID-19, a significantly higher expression of surface markers for T cell activation was observed in the recovering group than the severe group, whereas no significant differences were observed for neutralizing antibodies in both groups. To analyze the CTL activity induced by SARS-CoV-2 vaccines, the overlapping peptide pool for T cell stimulation was used due to the CTL epitopes of SARS-CoV-2 presently being unidentified [8] [9] [10] . A recent study reported SARS-CoV-2 S 526-533 as a CTL epitope in BALB/c mice, through the experimental evaluation of programming-predicted peptides [11] . C57BL/6 mice have been widely used for the evaluation of SARS-CoV-2 vaccines [8, 9] . Transgenic mice with a C57BL/6 genetic background expressing human ACE2 (hACE2) are currently being used as an animal model of SARS-CoV-2 study [12] [13] [14] [15] ; however, the CTL epitope in C57BL/6 mice has not yet been identified. Additionally, many experimental approaches aimed at quantifying SARS-CoV-2-specific CTL activity have used RBD peptide pools, which are associated with limitations such as high cost and time-consuming synthesis of peptides [8, 10] . Thus, finding a murine CTL epitope is important to resolve the limitations and to identify a peptide that can be used for exactly analyzing SARS-CoV-2-specific CD8 T cell activity when evaluating SARS-CoV-2 vaccine candidates in an animal model. In this study, we identified a murine CD8 T cell epitope of the SARS-CoV-2 spike RBD using C57BL/6 wild-type and hACE2-expressing transgenic (K18-hACE2) mice. To our knowledge, this is the first report to indicate that SARS-CoV-2 S 395-404 is a CD8 T cell epitope in the RBD of the SARS-CoV-2 spike protein in a C57BL/6 mouse model. C57BL/6 wild-type (OrientBio, Gyeonggi-do, Korea) and K18-hACE2 transgenic mice (Jackson Laboratory) were housed in a specific pathogen-free facility at the Korea Research Institute of Bioscience and Biotechnology (KRIBB). Mouse care and experiments were reviewed and conducted under guidelines of the Institutional Animal Care and Use Committee of KRIBB (KRIBB-AEC-20279). Recombinant SARS-CoV-2 spike RBD protein was purchased from Sino Biologics (Shanghai, China) and produced in HEK293 mammalian cells using a codon-optimized plasmid encoding SARS-CoV-2 RBD (amino acid residues (aa) 319-541 of SARS-CoV-2 S (accession no. YP_009724390.1)). The Sigma Adjuvant System (SAS), composed of monophosphoryl lipid A and squalene, was obtained from Sigma-Aldrich. Synthetic RBD peptides (8-11-mer and 15-mer peptides) were chemically synthesized by AnyGen (Gwangju, South Korea) and Peptron (Daejeon, South Korea) (summarized in Tables 1 and 2 ). The endotoxinfree EndoFit OVA protein and OVA 257-264 peptide were obtained from InvivoGen (San Diego, CA, USA) and AnaSpec (San Jose, CA, USA), respectively. Mouse IgG2a isotype control monoclonal antibody (mAb) was obtained from Bio X Cell (West Lebanon, NH, USA), and fluorescent dye-conjugated antibodies, anti-H2-K b and anti-H2-D b mAbs, were obtained from BD Biosciences (San Jose, CA, USA). 1 321 335 QPTESIVRFPNITNL 2 326 340 IVRFPNITNLCPFGE 3 331 345 NITNLCPFGEVFNAT 4 336 350 CPFGEVFNATRFASV 5 341 355 VFNATRFASVYAWNR 6 346 360 RFASVYAWNRKRISN 7 351 365 YAWNRKRISNCVADY 8 356 370 KRISNCVADYSVLYN 9 361 375 CVADYSVLYNSASFS 10 366 380 SVLYNSASFSTFKCY 11 371 385 SASFSTFKCYGVSPT 12 376 390 TFKCYGVSPTKLNDL 13 381 395 GVSPTKLNDLCFTNV 14 386 400 KLNDLCFTNVYADSF 15 391 405 CFTNVYADSFVIRGD Six-to 10-week-old female mice were randomly distributed in groups of 3 to 5 mice and intramuscularly (i.m.) immunized with 5 µg of RBD recombinant protein combined with SAS on days 0 and 14. In a separate experiment, wild-type mice were immunized i.m. with 10 µg of OVA protein plus SAS twice, with a two-week interval. One week after the last immunization, mononuclear splenocytes were generated by mincing whole mouse spleens, followed by the lysis of red blood cells and 70-µm filtration. Splenocytes were cultured in RPMI 1640 medium plus 10% heat-inactivated FBS, 100 U/mL penicillin, and 100 mg/mL streptomycin (Gibco, Dublin, Ireland). Antigen-specific IFN-γ-producing spot forming units (SFUs) were evaluated using mouse IFN-γ ELISPOT kits (BD Biosciences). Splenocytes were plated at 1 × 10 6 cells/well onto purified IFN-γ antibody-coated PVDF-backed plates and stimulated with an RBD peptide pool (2 µg/mL each), 5 µg/mL individual peptide, or 5 µg/mL OVA 257-264 at 37 • C for 2 days. In a separate experiment, splenocytes were stimulated with 10-mer peptides (5 µg/mL) in the presence of mouse IgG2a isotype as a control, anti-H2K b and/or anti-H2D b mAbs (20 µg/mL) at 37 • C for 2 days. The IFN-γ-positive SFUs were enumerated using an ELISPOT plate reader (Cellular Technology Ltd., Cleveland, OH, USA). Splenocytes at 1 × 10 6 cells/well were exposed to an RBD peptide pool (2 µg/mL for each peptide) or 5 µg/mL of each peptide in the presence of a protein transport inhibitor containing brefeldin A (BD Biosciences) for 12 h at 37 • C. Intracellular cytokine staining was performed according to a protocol modified from a previous paper [8] . The treated cells were incubated with LIVE/DEAD Fixable Red Dead Cell stain (Invitrogen) for 20 min at room temperature. The cells were washed, incubated with an anti-CD16/32 antibody (2.4G2) solution to block the Fc receptor, and stained for surface markers (CD3ε, CD4, and CD8 molecules), followed by intracellular IFN-γ cytokine staining using the Foxp3/transcription factor buffer set (eBioscience). The antibodies used were anti-CD3ε BUV737 (17A2), anti-CD4 APC (RM4-5), anti-CD8α PE-Cy7 (53-6.7), and anti-IFN-γ BV650 (XMG1.2). All the antibodies were obtained from BD Biosciences. The stained cells were acquired on a FACS Aria Fusion (BD Biosciences) and were analyzed via FlowJo TM V10 (TreeStar, Ashland, OR, USA). All the analyses were performed using the PRISM software 9.0.0 (GraphPad, San Diego, CA, USA), and p values less than 0.05 (p < 0.05) were considered to be statistically significant. To identify the CTL epitope(s) of the SARS-CoV-2 RBD, C57BL/6 mice (n = 5) were i.m. immunized twice with SARS-CoV-2 RBD recombinant protein along with SAS-composed of monophosphoryl lipid A and squalene-with a two-week interval. One week after the last immunization, an IFN-γ ELISPOT assay was performed by stimulating the splenocytes obtained from immunized mice with an RBD peptide pool of 42 synthetic RBD peptides with 15-mers overlapping by 5 aa (Table 1) . As shown in Figure 1A , the number of IFN-γ SFUs was significantly increased in cells treated with 15th peptide out of 42 peptides (CFTNVYADSFVIRGD, corresponding to S 391-405 ) (202 ± 96 SFUs) compared to that in the medium control (2 ± 3 SFUs) (p < 0.0001). The level of increase provoked by treatment with the S 391-405 peptide was similar to the SFU values achieved via stimulation with the RBD peptide pool (214 ± 95 SFUs) (p < 0.0001). However, none of the other 41 individual RBD peptides induced the production of IFN-γ SFUs. As CD8 CTLs recognize foreign antigen peptides of 8-11 aa residues, we attempted to identify the minimum length of CTL epitope that could be recognized by major histocompatibility complex (MHC) class I on CD8 T cells. Splenocytes obtained from the RBD- To confirm that S 391-405 -induced IFN-γ production was dependent on CD8 T cells but not CD4 T cells, splenocytes were subjected to intracellular IFN-γ staining. The gating strategy for flow cytometry employed is depicted in Figure 1B . In line with our expectations, the percentages of IFN-γ + CD8 T cells were almost 1.7-fold higher in the S 391-405 -treated cells (1.25 ± 0.07%) than the medium-treated cells (0.80 ± 0.25%) (p = 0.0044) ( Figure 1C) . However, the percentages of IFN-γ + CD4 T cells were similar between the S 391-405 -and medium-treated cells (p = 0.5667). Next, we assessed the SARS-CoV-2 RBD-specific CTL activity using K18-hACE2 transgenic mice (C57BL/6 background), which have been reported as an animal model suitable for SARS-CoV-2 studies [14] . When stimulating splenocytes from the RBD protein-immunized K18-hACE2 transgenic mice (n = 3) with the RBD peptide pool, the IFN-γ SFU numbers were also observed to be higher in the S 391-405 peptide-stimulated cells (240 ± 64 SFUs) than in the control cells (5 ± 3 SFUs) (p = 0.0068). The extent of this finding is similar to the effect of stimulation with the RBD peptide pool (252 ± 81 SFUs) (p = 0.0053) ( Figure 1D ). These results suggest that the SARS-CoV-2 S 391-405 peptide contains CTL epitope(s) in the C57BL/6 mouse model. As CD8 CTLs recognize foreign antigen peptides of 8-11 aa residues, we attempted to identify the minimum length of CTL epitope that could be recognized by major histocompatibility complex (MHC) class I on CD8 T cells. Splenocytes obtained from the RBD-immunized mice (n = 4) were harvested and subjected to IFN-γ ELISPOT with 8-11-mer peptides derived from SARS-CoV-2 S 391-405 ( Table 2 ). As shown in Figure 2A , none of the 8-or 9-mer peptides triggered a significant generation of IFN-γ SFUs. Of the 10-mer peptides, however, a significantly higher number of IFN-γ SFUs was observed after treatment with peptide #5 only (VYADSFVIRG, corresponding to S 395-404 ; 119 ± 47 SFUs) than with the medium control (2 ± 2 SFUs) (p = 0.0023). Two 11-mer peptides, including S 395-404 , also significantly augmented IFN-γ SFU numbers (190 ± 78 SFUs for peptide #4, corresponding to S 394-404 ; 157 ± 100 SFUs for peptide #5, corresponding to S 395-405 ) (p < 0.0001). Flow cytometry also showed that the percentages of IFN-γ + CD8 T cells were significantly higher (three-fold) in the cells treated with only the S 395-404 peptide (1.3 ± 0.12%) than in those treated with other 10-mer peptides (0.36 ± 0.06, 0.37 ± 0.1, 0.37 ± 0.08, 0.41 ± 0.11, and 0.41 ± 0.07% for peptide numbers 1, 2, 3, 4, and 6, respectively) (p < 0.0001) ( Figure 2B ). These results indicate that the SARS-CoV-2 S 395-404 peptide is a minimal epitope for CTLs specific for the SARS-CoV-2 spike protein RBD. Finally, we tested MHC restriction for the recognition of the SARS-CoV-2 S 395-404 peptide using antibodies blocking MHC class I, such as anti-H-2K b and anti-H-2D b mAbs, via an IFN-γ ELISPOT assay. The S 395-404 -increasing IFN-γ SFUs (115 ± 42 SFUs) were similar to those for cells treated with the mAb isotype (111 ± 45 SFUs) (p = 0.9999) ( Figure 2C) . However, the IFN-γ SFUs were significantly blocked by treatment with anti-H-2K b mAb (21 ± 3 SFUs; p = 0.0112), anti-H-2D b mAb (24 ± 5 SFUs; p = 0.0142), or a mixture of both (17 ± 12 SFUs; p = 0.0079), as compared with treatment with the mAb isotype. Furthermore, when using splenocytes from the SARS-CoV-2 spike RBD-immunized K18-hACE2 mice (n = 3), the S 395-404 peptide significantly augmented IFN-γ SFU numbers (104 ± 20 SFUs) compared to the medium control (3 ± 5 SFUs) (p < 0.0001) ( Figure 2D ). Similar S 395-404inducing IFN-γ SFU numbers were observed in cells treated with the control mAb isotype (80 ± 14 SFUs) (p = 0.1213). However, these numbers were significantly decreased by the addition of anti-H-2K b mAb (5 ± 3 SFUs), anti-H-2D b mAb (9 ± 4 SFUs), or a mixture of both (9 ± 3 SFUs) (p < 0.0001). To confirm our results, we tested splenocytes from OVA-immunized mice (n = 3) using the well-known CTL epitope OVA 257-264 [16] . After the stimulation of splenocytes with OVA 257-264 , significant production of IFN-γ SFUs was observed in the OVA-immunized mice (54 ± 5 SFUs and 52 ± 10 SFUs, in the absence and presence of isotype mAb, respectively), as compared with that in the medium control (0 SFU) (p < 0.0001). However, treatment with both anti-H-2K b and anti-H-2D b mAbs significantly inhibited OVA 257-264 -stimulated IFN-γ SFU production (18 ± 6 SFUs after treatment with anti-H-2K b mAb, p = 0.0002; 13 ± 6 SFUs after treatment with anti-H-2D b mAb, p = 0.0004; 8 ± 9 SFUs after treatment with anti-H-2K b /D b mAbs, p = 0.0001) ( Figure 2E ). Collectively, these data indicate that SARS-CoV-2 S 395-404 is the MHC class I H-2K b /D b -restricted CTL epitope of the SARS-CoV-2 spike protein RBD in the C57BL/6 mouse model. The SARS-CoV-2 S 395-404 peptide, VYADSFVIRG, is localized upstream of the receptorbinding motif within the RBD region and is not absolutely identical to the SARS-CoV S 382-391 peptide (VYADSFVVKG, Accession no. YP_009825051.1). A previous study reported SARS-CoV S 436-443 (YNKKYRYL) and S 525-532 (VNFNFNGL) as H-2K b -restricted epitopes [17] ; however, these sequences were not found in the SARS-CoV-2 S protein. The SARS-CoV-2 S 526-533 peptide (GPKKSTNL) was recently reported to be the H-2K drestricted CTL epitope in BALB/c mice [11] . However, the S 526-533 -containing peptide #42 did not induce generation of IFN-γ + cells from immunized C57BL/6 mice, as shown in Figure 1A , suggesting that SARS-CoV-2 S 395-404 might be a unique CTL epitope of the SARS-CoV-2 S RBD in the C57BL/6 mouse model. Similarly to our results, some peptides, including glycoprotein 33-43 of lymphocytic choriomeningitis virus [18] and the ovalbumin peptide (OVA 257-264 ) [16] , have been reported to be the H-2K b /D b -binding epitope. Further studies are needed to measure the binding affinity of SARS-CoV-2 S 395-404 peptide to MHC class I molecules. Various domains within SARS-CoV S protein are considered SARS-CoV-2 vaccine targets. RBD domain is one of the important antigens for SARS-CoV-2 vaccines and several RBD-based vaccine candidates have been tested in clinical trials [19] . Of them, lipid nanoparticle-formulated RBD mRNA vaccine candidates are reported to induce both neutralizing antibody response and T cell activation in preclinical [20, 21] and clinical fields [22] . Aluminum-adjuvanted RBD vaccine candidate is reported to increase neutralizing antibody response rather than T cell immunity in mice, rabbits and non-human primates [23] . A recombinant subunit vaccine candidate containing the RBD of SARS-CoV-2 and the Fc fragment of human lgG is shown that increase neutralizing antibody response only in a mouse model [24] . Several studies have reported that SARS-CoV-specific CD8 CTLs substantially protected against SARS-CoV infection in mice [25] and were increased in mice that were vaccinated with SARS-CoV S DNA as the vaccine antigen [26] . As an increase in CTL activity specific to SARS-CoV-2 has been reported in the peripheral blood mononuclear cells from COVID-19 convalescent patients [27] , SARS-CoV-2-specific CTLs may have a protective role in SARS-CoV-2 infection. Translation of the vaccine immunity in human subjects may be difficult to accurately predict from animal models, but animal models is most useful in stratifying the vaccine candidates through careful evaluation of vaccine efficacy before clinical trials [28] . Many experimental approaches aimed at quantifying SARS-CoV-2-specific CTL activity have used RBD peptide pools having limitations such as expensive and time-consuming synthesis of peptides [8, 10] . Our findings would be useful for resolving these problems and could be beneficial for evaluating SARS-CoV-2 spike RBD-specific CTL responses and studying T cell functions. Moreover, these findings may be helpful for the generation of a tetrameric MHC-peptide complex that can be used to directly quantify and visualize SARS-CoV-2 S RBD-specific T cells [29] . Taken together, the identification of SARS-CoV-2 S 395-404 as the CTL epitope may facilitate the evaluation of CTL activity of RBD-based COVID-19 vaccine candidates in a mouse model and the investigation of CTL function in SARS-CoV-2 infection. Identification of the SARS-CoV-2 spike RBD-specific CTL epitope is necessary to exactly analyze SARS-CoV-2-specific CTL activity and to solve problems of costly and time-consuming peptide synthesis for peptide pools. Our finding of SARS-CoV-2 S 395-404 as the CTL epitope is helpful in the study of SARS-CoV-2-specific CTL functions and the evaluation of CTL activity of RBD-based COVID-19 vaccine candidates using a mouse model. Informed Consent Statement: Not applicable. The data presented in this study are available on request from the corresponding author. The authors declare no conflict of interest. New SARS-like virus in China triggers alarm COVID-19) Situation Report-62 COVID-19 vaccine development pipeline gears up Rapid COVID-19 vaccine development Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor CD8+ T cell efficacy in vaccination and disease COVID-19 Severity Correlates with Weaker T-Cell Immunity, Hypercytokinemia, and Lung Epithelium Injury SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness Immunogenicity of a DNA vaccine candidate for COVID-19 ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques Identification of a dominant CD8(+) CTL epitope in the SARS-associated coronavirus 2 spike protein Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease Animal and translational models of SARS-CoV-2 infection and COVID-19 Lethality of SARS-CoV-2 infection in K18 human angiotensin-converting enzyme 2 transgenic mice SARS-CoV-2 infection in the lungs of human ACE2 transgenic mice causes severe inflammation, immune cell infiltration, and compromised respiratory function Epitope Prediction Assays Combined with Validation Assays Strongly Narrows down Putative Cytotoxic T Lymphocyte Epitopes Screening and identification of severe acute respiratory syndrome-associated coronavirus-specific CTL epitopes The signal sequence of lymphocytic choriomeningitis virus contains an immunodominant cytotoxic T cell epitope that is restricted by both H-2D(b) and H-2K(b) molecules. Virology Antibodies and Vaccines Target RBD of SARS-CoV-2. Front. Mol. Biosci. 2021, 8, 671633 A Thermostable mRNA Vaccine against COVID-19 BNT162b vaccines protect rhesus macaques from SARS-CoV-2 COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunity RBD-Fc-based COVID-19 vaccine candidate induces highly potent SARS-CoV-2 neutralizing antibody response Virus-specific memory CD8 T cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection CD8+ T cell response in HLA-A*0201 transgenic mice is elicited by epitopes from SARS-CoV S protein Suboptimal SARS-CoV-2-specific CD8(+) T cell response associated with the prominent HLA-A*02:01 phenotype What Is the Predictive Value of Animal Models for Vaccine Efficacy in Humans? The Importance of Bridging Studies and Species-Independent Correlates of Protection MHC-peptide tetramers for the analysis of antigen-specific T cells