Memory T cell responses targeting the SARS coronavirus persist up to 11 years post-infection

Oi-Wing Ng, Adeline Chia, Anthony T Tan, Ramesh S Jadi, Hoe Nam Leong, Antonio Bertoletti, Yee-Joo Tan, Oi-Wing Ng, Adeline Chia, Anthony T Tan, Ramesh S Jadi, Hoe Nam Leong, Antonio Bertoletti, Yee-Joo Tan

Abstract

Severe acute respiratory syndrome (SARS) is a highly contagious infectious disease which first emerged in late 2002, caused by a then novel human coronavirus, SARS coronavirus (SARS-CoV). The virus is believed to have originated from bats and transmitted to human through intermediate animals such as civet cats. The re-emergence of SARS-CoV remains a valid concern due to the continual persistence of zoonotic SARS-CoVs and SARS-like CoVs (SL-CoVs) in bat reservoirs. In this study, the screening for the presence of SARS-specific T cells in a cohort of three SARS-recovered individuals at 9 and 11 years post-infection was carried out, and all memory T cell responses detected target the SARS-CoV structural proteins. Two CD8(+) T cell responses targeting the SARS-CoV membrane (M) and nucleocapsid (N) proteins were characterized by determining their HLA restriction and minimal T cell epitope regions. Furthermore, these responses were found to persist up to 11 years post-infection. An absence of cross-reactivity of these CD8(+) T cell responses against the newly-emerged Middle East respiratory syndrome coronavirus (MERS-CoV) was also demonstrated. The knowledge of the persistence of SARS-specific celullar immunity targeting the viral structural proteins in SARS-recovered individuals is important in the design and development of SARS vaccines, which are currently unavailable.

Keywords: Epitope; Immunity; SARS-CoV; T cell.

Copyright © 2016 Elsevier Ltd. All rights reserved.

Figures

Fig. 1
Fig. 1
IFNγ ELISpot results for SARS-specific memory T cell screening. PBMCs from (A) a healthy individual and (B) SARS-recovered individual (SARS subject 1) were expanded in vitro using a mixture of SARS-CoV peptides, followed by IFNγ ELISpot assay using SARS peptide matrix pools of the structural (top panels) and accessory proteins (lower panels). Each bar represents the IFNγ-producing response to an individual peptide matrix pool (numeric or alphabetic) in SFU per 5 × 104 cells. The threshold for a positive response was set as two times above the mean SFU of unstimulated cells (Neg), as indicated by the dotted line in the right panels. Cells stimulated with PMA/ionomycin were included as positive control (Pos).
Fig. 2
Fig. 2
ICS and flow cytometry analysis of unstimulated and M29-stimulated T cells after restimulation using M29 peptide. The percentages of CD8+ IFNγ+ and CD8+CD107a+ T cells shown represent the percentage of the T cells in total T cell population (after gating the CD3+ cells) present in the short-term T cell line obtained by restimulation using M29 peptide from SARS subject 1 at 9 years post-infection.
Fig. 3
Fig. 3
ICS and flow cytometry analysis of restimulated T cells from SARS subject 1 at 11 years post-infection. Percentages of CD8+IFNγ+ responses (left panels) and CD8+CD107a+ responses (right panels) of (A) unstimulated, (B) M29147–155-stimulated, (C) N53266–275-stimulated T cells are as indicated in the upper right quadrant of each dot plot. Percentage CD8+ IFNγ+ cells shown represent the percentage of IFNγ-producing cells in the total T cell population (after gating the CD3+ cells) which were in vitro expanded in the presence of M29147–155 and N53266–275 peptides.
Fig. 4
Fig. 4
Cross-reactivity of SARS-specific M29 and N53 CD8+ T cells. Sequence alignments of (A) M29 and (B) N53 regions of human SARS-CoV (HKU39849), civet SARS-CoV (SZ3), bat SL-CoVs (Rp3, Rf1 and Rs3367) and MERS-CoV. (C) Percentages of CD8+IFNγ+ T cell responses induced by SARS-CoV and MERS-CoV M29 (left) and N53 (right) minimal peptides. Percentage CD8+ IFNγ+ cells shown represents the percentage of IFNγ-producing cells in the total T cell population (after gating the CD3+ cells) present in the short-term T cell line obtained by restimulation using SARS-CoV M29 and N53 minimal peptides (M29147–155 and N53266–275) from SARS subject 1 at 9 years post-infection.

References

    1. Ksiazek T.G., Erdman D., Goldsmith C.S., Zaki S.R., Peret T., Emery S. A novel coronavirus associated with severe acute respiratory syndrome. N Engl J Med. 2003;348(20 (May)):1953–1966.
    1. Peiris J.S., Guan Y., Yuen K.Y. Severe acute respiratory syndrome. Nat Med. 2004;10(12 Suppl. (December)):S88–S97.
    1. Li W., Shi Z., Yu M., Ren W., Smith C., Epstein J.H. Bats are natural reservoirs of SARS-like coronaviruses. Science. 2005;310(5748 (October)):676–679.
    1. Lau S.K., Woo P.C., Li K.S., Huang Y., Tsoi H.W., Wong B.H. Severe acute respiratory syndrome coronavirus-like virus in Chinese horseshoe bats. Proc Natl Acad Sci U S A. 2005;102(39 (September)):14040–14045.
    1. Ge X.Y., Li J.L., Yang X.L., Chmura A.A., Zhu G., Epstein J.H. Isolation and characterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature. 2013;503(7477 (November)):535–538.
    1. Guan Y., Zheng B.J., He Y.Q., Liu X.L., Zhuang Z.X., Cheung C.L. Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China. Science. 2003;302(5643 (October)):276–278.
    1. Rota P.A., Oberste M.S., Monroe S.S., Nix W.A., Campagnoli R., Icenogle J.P. Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science. 2003;300(5624 (May)):1394–1399.
    1. Chen J., Lau Y.F., Lamirande E.W., Paddock C.D., Bartlett J.H., Zaki S.R. Cellular immune responses to severe acute respiratory syndrome coronavirus (SARS-CoV) infection in senescent BALB/c mice: CD4+ T cells are important in control of SARS-CoV infection. J Virol. 2010;84(3 (February)):1289–1301.
    1. Zhao J., Perlman S. T cell responses are required for protection from clinical disease and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. J Virol. 2010;84(18 (September)):9318–9325.
    1. Channappanavar R., Fett C., Zhao J., Meyerholz D.K., Perlman S. Virus-specific memory CD8T cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection. J Virol. 2014;88(19 (October)):11034–11044.
    1. Li T., Qiu Z., Zhang L., Han Y., He W., Liu Z. Significant changes of peripheral T lymphocyte subsets in patients with severe acute respiratory syndrome. J Infect Dis. 2004;189(4 (February)):648–651.
    1. Cameron M.J., Bermejo-Martin J.F., Danesh A., Muller M.P., Kelvin D.J. Human immunopathogenesis of severe acute respiratory syndrome (SARS) Virus Res. 2008;133(1 (April)):13–19.
    1. Oh H.L., Chia A., Chang C.X., Leong H.N., Ling K.L., Grotenbreg G.M. Engineering T cells specific for a dominant severe acute respiratory syndrome coronavirus CD8T cell epitope. J Virol. 2011;85(20 (October)):10464–10471.
    1. Case Definitions for Surveillance of Severe Acute Respiratory Syndrome (SARS). . 2003 [Available from: ].
    1. Li T., Xie J., He Y., Fan H., Baril L., Qiu Z. Long-term persistence of robust antibody and cytotoxic T cell responses in recovered patients infected with SARS coronavirus. PLoS ONE. 2006;1:e24.
    1. Li C.K., Wu H., Yan H., Ma S., Wang L., Zhang M. T cell responses to whole SARS coronavirus in humans. J Immunol. 2008;181(8 (October)):5490–5500.
    1. Yang L.T., Peng H., Zhu Z.L., Li G., Huang Z.T., Zhao Z.X. Long-lived effector/central memory T-cell responses to severe acute respiratory syndrome coronavirus (SARS-CoV) S antigen in recovered SARS patients. Clin Immunol. 2006;120(2 (August)):171–178.
    1. Zhou M., Xu D., Li X., Li H., Shan M., Tang J. Screening and identification of severe acute respiratory syndrome-associated coronavirus-specific CTL epitopes. J Immunol. 2006;177(4 (August)):2138–2145.
    1. Betts M.R., Brenchley J.M., Price D.A., De Rosa S.C., Douek D.C., Roederer M. Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. J Immunol Methods. 2003;281(1–2 (October)):65–78.
    1. Harding C.V., Geuze H.J. Antigen processing and intracellular traffic of antigens and MHC molecules. Curr Opin Cell Biol. 1993;5(4 (August)):596–605.
    1. Liu J., Sun Y., Qi J., Chu F., Wu H., Gao F. The membrane protein of severe acute respiratory syndrome coronavirus acts as a dominant immunogen revealed by a clustering region of novel functionally and structurally defined cytotoxic T-lymphocyte epitopes. J Infect Dis. 2010;202(8 (October)):1171–1180.
    1. Fan Y.Y., Huang Z.T., Li L., Wu M.H., Yu T., Koup R.A. Characterization of SARS-CoV-specific memory T cells from recovered individuals 4 years after infection. Arch Virol. 2009;154(7):1093–1099.
    1. Tsao Y.P., Lin J.Y., Jan J.T., Leng C.H., Chu C.C., Yang Y.C. HLA-A*0201 T-cell epitopes in severe acute respiratory syndrome (SARS) coronavirus nucleocapsid and spike proteins. Biochem Biophys Res Commun. 2006;344(1 (May)):63–71.
    1. Kim T.W., Lee J.H., Hung C.F., Peng S., Roden R., Wang M.C. Generation and characterization of DNA vaccines targeting the nucleocapsid protein of severe acute respiratory syndrome coronavirus. J Virol. 2004;78(9 (May)):4638–4645.
    1. Rivino L., Tan A.T., Chia A., Kumaran E.A., Grotenbreg G.M., MacAry P.A. Defining CD8+ T cell determinants during human viral infection in populations of Asian ethnicity. J Immunol. 2013;191(8 (October)):4010–4019.
    1. Zaki A.M., van Boheemen S., Bestebroer T.M., Osterhaus A.D., Fouchier R.A. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med. 2012;367(19 (November)):1814–1820.
    1. de Groot R.J., Baker S.C., Baric R.S., Brown C.S., Drosten C., Enjuanes L. Middle East respiratory syndrome coronavirus (MERS-CoV): announcement of the Coronavirus Study Group. J Virol. 2013;87(14 (July)):7790–7792.
    1. Chan J.F., Lau S.K., To K.K., Cheng V.C., Woo P.C., Yuen K.Y. Middle East respiratory syndrome coronavirus: another zoonotic betacoronavirus causing SARS-like disease. Clin Microbiol Rev. 2015;28(2 (April)):465–522.
    1. Cheng V.C., Lau S.K., Woo P.C., Yuen K.Y. Severe acute respiratory syndrome coronavirus as an agent of emerging and reemerging infection. Clin Microbiol Rev. 2007;20(4 (October)):660–694.
    1. van Boheemen S., de Graaf M., Lauber C., Bestebroer T.M., Raj V.S., Zaki A.M. Genomic characterization of a newly discovered coronavirus associated with acute respiratory distress syndrome in humans. MBio. 2012;3(6)
    1. Martin J.E., Louder M.K., Holman L.A., Gordon I.J., Enama M.E., Larkin B.D. A SARS DNA vaccine induces neutralizing antibody and cellular immune responses in healthy adults in a Phase I clinical trial. Vaccine. 2008;26(50 (November)):6338–6343.

Source: PubMed

3
Předplatit