Register      Login
Microbiology Australia Microbiology Australia Society
Microbiology Australia, bringing Microbiologists together
RESEARCH ARTICLE (Open Access)

Long-term and short-term immunity to SARS-CoV-2: why it matters

John Zaunders A C and Chansavath Phetsouphanh B
+ Author Affiliations
- Author Affiliations

A Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia

B Kirby Institute, UNSW Sydney, Australia

C Email: j.zaunders@amr.org.au

Microbiology Australia 42(1) 34-38 https://doi.org/10.1071/MA21010
Submitted: 29 January 2021  Accepted: 10 March 2021   Published: 13 April 2021

Journal Compilation © The Authors 2021 Open Access CC BY, published (by CSIRO Publishing) on behalf of the ASM

Abstract

The adaptive immune system, regulated by CD4 T cells, is essential for control of many viral infections. Endemic coronavirus infections generally occur as short-term upper respiratory tract infections which in many cases appear to be cleared before adaptive immunity is fully involved, since adaptive immunity takes approximately 1.5–2 weeks to ramp up the response to a primary infection, or approximately 1 week for a recurrent infection. However, the adaptive immune response to SARS-CoV-2 infection will be critical to full recovery with minimal long-lasting effects, and to either prevention of recurrence of infection or at least reduced severity of symptoms. The detailed kinetics of this infection versus the dynamics of the immune response, including in vaccinated individuals, will largely determine these outcomes.


References

[1]  Clayton, E. and Munir, M. (2020) Fundamental characteristics of bat interferon systems. Front. Cell. Infect. Microbiol. 10, 527921.
Fundamental characteristics of bat interferon systems.Crossref | GoogleScholarGoogle Scholar | 33363045PubMed |

[2]  Cevik, M. et al. (2020) Virology, transmission, and pathogenesis of SARS-CoV-2. BMJ 371, m3862.
Virology, transmission, and pathogenesis of SARS-CoV-2.Crossref | GoogleScholarGoogle Scholar | 33097561PubMed |

[3]  Andreakos, E. et al. (2019) Lambda interferons come to light: dual function cytokines mediating antiviral immunity and damage control. Curr. Opin. Immunol. 56, 67–75.
Lambda interferons come to light: dual function cytokines mediating antiviral immunity and damage control.Crossref | GoogleScholarGoogle Scholar | 30399529PubMed |

[4]  Zhang, Q. et al. (2020) Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 370, eabd4570.
Inborn errors of type I IFN immunity in patients with life-threatening COVID-19.Crossref | GoogleScholarGoogle Scholar | 33243877PubMed |

[5]  Bastard, P. et al. (2020) Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 370, eabd4585.
Autoantibodies against type I IFNs in patients with life-threatening COVID-19.Crossref | GoogleScholarGoogle Scholar | 32972996PubMed |

[6]  Hadjadj, J. et al. (2020) Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369, 718–724.
Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients.Crossref | GoogleScholarGoogle Scholar | 32661059PubMed |

[7]  Blanco-Melo, D. et al. (2020) Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181, 1036–1045.e9.
Imbalanced host response to SARS-CoV-2 drives development of COVID-19.Crossref | GoogleScholarGoogle Scholar | 32416070PubMed |

[8]  Galani, I.E. et al. (2021) Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison. Nat. Immunol. 22, 32–40.
Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison.Crossref | GoogleScholarGoogle Scholar | 33277638PubMed |

[9]  Lei, X. et al. (2020) Activation and evasion of type I interferon responses by SARS-CoV-2. Nat. Commun. 11, 3810.
Activation and evasion of type I interferon responses by SARS-CoV-2.Crossref | GoogleScholarGoogle Scholar | 32733001PubMed |

[10]  Kikkert, M. (2020) Innate immune evasion by human respiratory RNA viruses. J. Innate Immun. 12, 4–20.
Innate immune evasion by human respiratory RNA viruses.Crossref | GoogleScholarGoogle Scholar | 31610541PubMed |

[11]  Liu, Y.J. (2005) IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu. Rev. Immunol. 23, 275–306.
IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors.Crossref | GoogleScholarGoogle Scholar | 15771572PubMed |

[12]  Splunter, M.V. et al. (2019) Plasmacytoid dendritic cell and myeloid dendritic cell function in ageing: a comparison between elderly and young adult women. PLoS One 14, e0225825.
Plasmacytoid dendritic cell and myeloid dendritic cell function in ageing: a comparison between elderly and young adult women.Crossref | GoogleScholarGoogle Scholar | 31830086PubMed |

[13]  Götzinger, F. et al. (2020) COVID-19 in children and adolescents in Europe: a multinational, multicentre cohort study. Lancet Child Adolesc. Health 4, 653–661.
COVID-19 in children and adolescents in Europe: a multinational, multicentre cohort study.Crossref | GoogleScholarGoogle Scholar | 32593339PubMed |

[14]  Tosif, S. et al. (2020) Immune responses to SARS-CoV-2 in three children of parents with symptomatic COVID-19. Nat. Commun. 11, 5703.
Immune responses to SARS-CoV-2 in three children of parents with symptomatic COVID-19.Crossref | GoogleScholarGoogle Scholar | 33177504PubMed |

[15]  Pantaleo, G. and Koup, R.A. (2004) Correlates of immune protection in HIV-1 infection: what we know, what we don’t know, what we should know. Nat. Med. 10, 806–810.
Correlates of immune protection in HIV-1 infection: what we know, what we don’t know, what we should know.Crossref | GoogleScholarGoogle Scholar | 15286782PubMed |

[16]  Crotty, S. (2011) Follicular helper CD4 T cells (TFH). Annu. Rev. Immunol. 29, 621–663.
Follicular helper CD4 T cells (TFH).Crossref | GoogleScholarGoogle Scholar | 21314428PubMed |

[17]  Walker, B. and McMichael, A. (2012) The T-cell response to HIV. Cold Spring Harb. Perspect. Med. 2, a007054.
The T-cell response to HIV.Crossref | GoogleScholarGoogle Scholar | 23002014PubMed |

[18]  Akkaya, M. et al. (2020) B cell memory: building two walls of protection against pathogens. Nat. Rev. Immunol. 20, 229–238.
B cell memory: building two walls of protection against pathogens.Crossref | GoogleScholarGoogle Scholar | 31836872PubMed |

[19]  Tellier, J. et al. (2016) Blimp-1 controls plasma cell function through the regulation of immunoglobulin secretion and the unfolded protein response. Nat. Immunol. 17, 323–330.
Blimp-1 controls plasma cell function through the regulation of immunoglobulin secretion and the unfolded protein response.Crossref | GoogleScholarGoogle Scholar | 26779600PubMed |

[20]  Papadatou, I. et al. (2019) The role of serotype-specific immunological memory in pneumococcal vaccination: current knowledge and future prospects. Vaccines (Basel) 7, 13.
The role of serotype-specific immunological memory in pneumococcal vaccination: current knowledge and future prospects.Crossref | GoogleScholarGoogle Scholar |

[21]  Harper, D.M. and DeMars, L.R. (2017) HPV vaccines – a review of the first decade. Gynecol. Oncol. 146, 196–204.
HPV vaccines – a review of the first decade.Crossref | GoogleScholarGoogle Scholar | 28442134PubMed |

[22]  Zaunders, J.J. et al. (2006) CD127+CCR5+CD38+++ CD4+ Th1 effector cells are an early component of the primary immune response to vaccinia virus and precede development of interleukin-2+ memory CD4+ T cells. J. Virol. 80, 10151–10161.
CD127+CCR5+CD38+++ CD4+ Th1 effector cells are an early component of the primary immune response to vaccinia virus and precede development of interleukin-2+ memory CD4+ T cells.Crossref | GoogleScholarGoogle Scholar | 17005692PubMed |

[23]  Miller, J.D. et al. (2008) Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity 28, 710–722.
Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines.Crossref | GoogleScholarGoogle Scholar | 18468462PubMed |

[24]  Hornick, E.E. et al. (2019) Kinetics and phenotype of the CD4 T cell response to influenza virus infections. Front. Immunol. 10, 2351.
Kinetics and phenotype of the CD4 T cell response to influenza virus infections.Crossref | GoogleScholarGoogle Scholar | 31632414PubMed |

[25]  Tenforde, M.W. et al. (2020) Symptom duration and risk factors for delayed return to usual health among outpatients with COVID-19 in a multistate health care systems network – United States, March-June 2020. MMWR Morb. Mortal. Wkly Rep. 69, 993–998.
Symptom duration and risk factors for delayed return to usual health among outpatients with COVID-19 in a multistate health care systems network – United States, March-June 2020.Crossref | GoogleScholarGoogle Scholar | 32730238PubMed |

[26]  Herati, R.S. et al. (2017) Successive annual influenza vaccination induces a recurrent oligoclonotypic memory response in circulating T follicular helper cells. Sci. Immunol. 2, eaag2152.
Successive annual influenza vaccination induces a recurrent oligoclonotypic memory response in circulating T follicular helper cells.Crossref | GoogleScholarGoogle Scholar | 28620653PubMed |

[27]  Baccam, P. et al. (2006) Kinetics of influenza A virus infection in humans. J. Virol. 80, 7590–7599.
Kinetics of influenza A virus infection in humans.Crossref | GoogleScholarGoogle Scholar | 16840338PubMed |

[28]  Doherty, P.C. and Christensen, J.P. (2000) Accessing complexity: the dynamics of virus-specific T cell responses. Annu. Rev. Immunol. 18, 561–592.
Accessing complexity: the dynamics of virus-specific T cell responses.Crossref | GoogleScholarGoogle Scholar | 10837069PubMed |

[29]  Seow, J. et al. (2020) Longitudinal observation and decline of neutralizing antibody responses in the three months following SARS-CoV-2 infection in humans. Nat. Microbiol. 5, 1598–1607.
Longitudinal observation and decline of neutralizing antibody responses in the three months following SARS-CoV-2 infection in humans.Crossref | GoogleScholarGoogle Scholar | 33106674PubMed |

[30]  Ibarrondo, F.J. et al. (2020) Rapid decay of anti-SARS-CoV-2 antibodies in persons with mild Covid-19. N. Engl. J. Med. 383, 1085–1087.
Rapid decay of anti-SARS-CoV-2 antibodies in persons with mild Covid-19.Crossref | GoogleScholarGoogle Scholar | 32706954PubMed |

[31]  Wajnberg, A. et al. (2020) Robust neutralizing antibodies to SARS-CoV-2 infection persist for months. Science 370, 1227–1230.
Robust neutralizing antibodies to SARS-CoV-2 infection persist for months.Crossref | GoogleScholarGoogle Scholar | 33115920PubMed |

[32]  Tea  F.et al2020 SARS-CoV-2 neutralizing antibodies; longevity, breadth, and evasion by emerging viral variants.medRxiv[Preprint].

[33]  Thevarajan, I. et al. (2020) Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat. Med. 26, 453–455.
Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19.Crossref | GoogleScholarGoogle Scholar | 32284614PubMed |

[34]  Mathew, D. et al. (2020) Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 369, eabc8511.
Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications.Crossref | GoogleScholarGoogle Scholar | 32669297PubMed |

[35]  Kuri-Cervantes, L. et al. (2020) Comprehensive mapping of immune perturbations associated with severe COVID-19. Sci. Immunol. 5, eabd7114.
Comprehensive mapping of immune perturbations associated with severe COVID-19.Crossref | GoogleScholarGoogle Scholar | 32669287PubMed |

[36]  Lee, W.S. et al. (2020) Antibody-dependent enhancement and SARS-CoV-2 vaccines and therapies. Nat. Microbiol. 5, 1185–1191.
Antibody-dependent enhancement and SARS-CoV-2 vaccines and therapies.Crossref | GoogleScholarGoogle Scholar | 32908214PubMed |

[37]  Parasa, S. et al. (2020) Prevalence of gastrointestinal symptoms and fecal viral shedding in patients with coronavirus disease 2019: a systematic review and meta-analysis. JAMA Netw. Open 3, e2011335.
Prevalence of gastrointestinal symptoms and fecal viral shedding in patients with coronavirus disease 2019: a systematic review and meta-analysis.Crossref | GoogleScholarGoogle Scholar | 32525549PubMed |

[38]  Seydoux, E. et al. (2020) Analysis of a SARS-CoV-2-infected individual reveals development of potent neutralizing antibodies with limited somatic mutation. Immunity 53, 98–105.e5.
Analysis of a SARS-CoV-2-infected individual reveals development of potent neutralizing antibodies with limited somatic mutation.Crossref | GoogleScholarGoogle Scholar | 32561270PubMed |

[39]  Kaneko, N. et al. (2020) Loss of Bcl-6-expressing T follicular helper cells and germinal centers in COVID-19. Cell 183, 143–157.e13.
Loss of Bcl-6-expressing T follicular helper cells and germinal centers in COVID-19.Crossref | GoogleScholarGoogle Scholar | 32877699PubMed |

[40]  Lax, S.F. et al. (2020) Pulmonary arterial thrombosis in COVID-19 with fatal outcome: results from a prospective, single-center, clinicopathologic case series. Ann. Intern. Med. 173, 350–361.
Pulmonary arterial thrombosis in COVID-19 with fatal outcome: results from a prospective, single-center, clinicopathologic case series.Crossref | GoogleScholarGoogle Scholar | 32422076PubMed |

[41]  Elsoukkary, S.S. et al. (2021) Autopsy findings in 32 patients with COVID-19: a single-institution experience. Pathobiology 88, 56–68.
Autopsy findings in 32 patients with COVID-19: a single-institution experience.Crossref | GoogleScholarGoogle Scholar | 32942274PubMed |

[42]  Dijkman, R. et al. (2008) Human coronavirus NL63 and 229E seroconversion in children. J. Clin. Microbiol. 46, 2368–2373.
Human coronavirus NL63 and 229E seroconversion in children.Crossref | GoogleScholarGoogle Scholar | 18495857PubMed |

[43]  Edridge, A.W.D. et al. (2020) Seasonal coronavirus protective immunity is short-lasting. Nat. Med. 26, 1691–1693.
Seasonal coronavirus protective immunity is short-lasting.Crossref | GoogleScholarGoogle Scholar |

[44]  Zaunders, J. et al. (2017) Divergent expression of CXCR5 and CCR5 on CD4+ T cells and the paradoxical accumulation of T follicular helper cells during HIV infection. Front. Immunol. 8, 495.
Divergent expression of CXCR5 and CCR5 on CD4+ T cells and the paradoxical accumulation of T follicular helper cells during HIV infection.Crossref | GoogleScholarGoogle Scholar | 28553284PubMed |

[45]  Woldemeskel, B.A. et al. (2020) Healthy donor T cell responses to common cold coronaviruses and SARS-CoV-2. J. Clin. Invest. 130, 6631–6638.
Healthy donor T cell responses to common cold coronaviruses and SARS-CoV-2.Crossref | GoogleScholarGoogle Scholar | 32966269PubMed |

[46]  Le Bert, N. et al. (2020) SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 584, 457–462.
SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls.Crossref | GoogleScholarGoogle Scholar | 32668444PubMed |

[47]  Altmann, D.M. and Boyton, R.J. (2020) SARS-CoV-2 T cell immunity: specificity, function, durability, and role in protection. Sci. Immunol. 5, eabd6160.
SARS-CoV-2 T cell immunity: specificity, function, durability, and role in protection.Crossref | GoogleScholarGoogle Scholar | 33361161PubMed |

[48]  Bertoletti, A. et al. (2021) The T cell response to SARS-CoV-2: kinetic and quantitative aspects and the case for their protective role. Oxford Open Immunology 2, iqab006.
The T cell response to SARS-CoV-2: kinetic and quantitative aspects and the case for their protective role.Crossref | GoogleScholarGoogle Scholar |

[49]  Wilk, A.J. et al. (2020) A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 26, 1070–1076.
A single-cell atlas of the peripheral immune response in patients with severe COVID-19.Crossref | GoogleScholarGoogle Scholar | 32514174PubMed |

[50]  Zaunders, J.J. et al. (2005) Early proliferation of CCR5+ CD38+++ antigen-specific CD4+ Th1 effector cells during primary HIV-1 infection. Blood 106, 1660–1667.
Early proliferation of CCR5+ CD38+++ antigen-specific CD4+ Th1 effector cells during primary HIV-1 infection.Crossref | GoogleScholarGoogle Scholar | 15905189PubMed |

[51]  Ellebedy, A.H. et al. (2016) Defining antigen-specific plasmablast and memory B cell subsets in human blood after viral infection or vaccination. Nat. Immunol. 17, 1226–1234.
Defining antigen-specific plasmablast and memory B cell subsets in human blood after viral infection or vaccination.Crossref | GoogleScholarGoogle Scholar | 27525369PubMed |

[52]  Fahey, L.M. et al. (2011) Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells. J. Exp. Med. 208, 987–999.
Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells.Crossref | GoogleScholarGoogle Scholar | 21536743PubMed |

[53]  Zaunders, J.J. et al. (2009) High levels of human antigen-specific CD4+ T cells in peripheral blood revealed by stimulated coexpression of CD25 and CD134 (OX40). J. Immunol. 183, 2827–2836.
High levels of human antigen-specific CD4+ T cells in peripheral blood revealed by stimulated coexpression of CD25 and CD134 (OX40).Crossref | GoogleScholarGoogle Scholar | 19635903PubMed |

[54]  Mateus, J. et al. (2020) Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 370, 89–94.
Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans.Crossref | GoogleScholarGoogle Scholar | 32753554PubMed |

[55]  Li  Z.et al2020 SARS-CoV-2-specific T cell memory is long-lasting in the majority of convalsecent COVID-19 individuals.bioRxiv [Preprint]. 10.1101/2020.11.15.383463

[56]  Dan, J.M. et al. (2021) Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 371, eabf4063.
Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection.Crossref | GoogleScholarGoogle Scholar | 33408181PubMed |

[57]  Orlov, M. et al. (2020) A case for targeting Th17 cells and IL-17A in SARS-CoV-2 infections. J. Immunol. 205, 892–898.
A case for targeting Th17 cells and IL-17A in SARS-CoV-2 infections.Crossref | GoogleScholarGoogle Scholar | 32651218PubMed |

[58]  Grant, R.A. et al. (2021) Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia. Nature 590, 635–641.
Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia.Crossref | GoogleScholarGoogle Scholar | 33429418PubMed |

[59]  Chen, Z. and John Wherry, E. (2020) T cell responses in patients with COVID-19. Nat. Rev. Immunol. 20, 529–536.
T cell responses in patients with COVID-19.Crossref | GoogleScholarGoogle Scholar | 32728222PubMed |

[60]  Chodick  G.et al2021 The effectiveness of the first dose of vaccine in reducing SARS-CoV-2 infection 13-24 days after immunization: real world evidence.medRxiv[Preprint].

[61]  Hall  V.et al2021 Effectiveness of BNT162b2 mRNA vaccine against infection and COVID-19 vaccine coverage in healthcare workers in England, multicentre prospective cohort study (the SIREN study).medRxiv[Preprint].