Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Humoral and circulating follicular helper T cell responses in recovered patients with COVID-19

Abstract

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has dramatically expedited global vaccine development efforts1,2,3, most targeting the viral ‘spike’ glycoprotein (S). S localizes on the virion surface and mediates recognition of cellular receptor angiotensin-converting enzyme 2 (ACE2)4,5,6. Eliciting neutralizing antibodies that block S–ACE2 interaction7,8,9, or indirectly prevent membrane fusion10, constitute an attractive modality for vaccine-elicited protection11. However, although prototypic S-based vaccines show promise in animal models12,13,14, the immunogenic properties of S in humans are poorly resolved. In this study, we characterized humoral and circulating follicular helper T cell (cTFH) immunity against spike in recovered patients with coronavirus disease 2019 (COVID-19). We found that S-specific antibodies, memory B cells and cTFH are consistently elicited after SARS-CoV-2 infection, demarking robust humoral immunity and positively associated with plasma neutralizing activity. Comparatively low frequencies of B cells or cTFH specific for the receptor binding domain of S were elicited. Notably, the phenotype of S-specific cTFH differentiated subjects with potent neutralizing responses, providing a potential biomarker of potency for S-based vaccines entering the clinic. Overall, although patients who recovered from COVID-19 displayed multiple hallmarks of effective immune recognition of S, the wide spectrum of neutralizing activity observed suggests that vaccines might require strategies to selectively target the most potent neutralizing epitopes.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Serological responses to COVID-19.
Fig. 2: Frequency and phenotype of SARS-CoV-2-specific B cells after infection.
Fig. 3: Specificity of cTFH responses to coronavirus spike proteins.
Fig. 4: Predictors of plasma neutralization activity.

Similar content being viewed by others

Data availability

The mass spectrometry data that support the findings of this study are available via ProteomeXchange with identifier PXD019163. The authors declare that all other data supporting the findings of this study are available in the paper and the Supplementary Information files.

References

  1. Amanat, F. & Krammer, F. SARS-CoV-2 vaccines: status report. Immunity 52, 583–589 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Thanh, Le,T. et al. The COVID-19 vaccine development landscape. Nat. Rev. Drug Discov. 19, 305–306 (2020).

    Article  Google Scholar 

  3. Lurie, N., Saville, M., Hatchett, R. & Halton, J. Developing Covid-19 vaccines at pandemic speed. N. Engl. J. Med. 382, 1969–1973 (2020).

    Article  CAS  PubMed  Google Scholar 

  4. Hoffmann, M. et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181, 271–280 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yan, R. et al. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 367, 1444–1448 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lan, J. et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 581, 215–220 (2020).

    Article  CAS  PubMed  Google Scholar 

  7. Rogers, T. F. et al. Rapid isolation of potent SARS-CoV-2 neutralizing antibodies and protection in a small animal model. Science https://doi.org/10.1126/science.abc7520 (2020).

  8. Wu, Y. et al. A noncompeting pair of human neutralizing antibodies block COVID-19 virus binding to its receptor ACE2. Science 368, 1274–1278 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cao, Y. et al. Potent neutralizing antibodies against SARS-CoV-2 identified by high-throughput single-cell sequencing of convalescent patients’ B cells. Cell https://doi.org/10.1016/j.cell.2020.05.025 (2020).

  10. Brouwer, P. J. M. et al. Potent neutralizing antibodies from COVID-19 patients define multiple targets of vulnerability. Science https://doi.org/10.1126/science.abc5902 (2020).

  11. Jiang, S., Hillyer, C. & Du, L. Neutralizing antibodies against SARS-CoV-2 and other human coronaviruses. Trends Immunol. 41, 355–359 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gao, Q. et al. Rapid development of an inactivated vaccine candidate for SARS-CoV-2. Science https://doi.org/10.1126/science.abc1932 (2020).

  13. Yu, J. et al. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science https://doi.org/10.1126/science.abc6284 (2020).

  14. van Doremalen, N. et al. ChAdOx1 nCoV-19 vaccination prevents SARS-CoV-2 pneumonia in rhesus macaques. Preprint at https://www.biorxiv.org/content/10.1101/2020.05.13.093195v1 (2020).

  15. Lv, H. et al. Cross-reactive antibody response between SARS-CoV-2 and SARS-CoV infections. Cell Rep. 31, 107725 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhou, W., Wang, W., Wang, H., Lu, R. & Tan, W. First infection by all four non-severe acute respiratory syndrome human coronaviruses takes place during childhood. BMC Infect. Dis. 13, 433 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Okba, N. M. A. et al. Severe acute respiratory syndrome coronavirus 2-specific antibody responses in coronavirus disease 2019 patients. Emerg. Infect. Dis. 26, 1478–1488 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Subbarao, K. et al. Prior infection and passive transfer of neutralizing antibody prevent replication of severe acute respiratory syndrome coronavirus in the respiratory tract of mice. J. Virol. 78, 3572–3577 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Houser, K. V. et al. Prophylaxis with a Middle East respiratory syndrome coronavirus (MERS-CoV)-specific human monoclonal antibody protects rabbits from MERS-CoV infection. J. Infect. Dis. 213, 1557–1561 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Robbiani, D. F. et al. Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature https://doi.org/10.1038/s41586-020-2456-9 (2020).

  21. Koutsakos, M. et al. Circulating TFH cells, serological memory, and tissue compartmentalization shape human influenza-specific B cell immunity. Sci. Trans. Med. 10, eaan8405 (2018).

    Article  Google Scholar 

  22. Lau, D. et al. Low CD21 expression defines a population of recent germinal center graduates primed for plasma cell differentiation. Sci. Immunol. 2, eaai8153 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Ou, X. et al. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 11, 1620 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Stettler, K. et al. Specificity, cross-reactivity, and function of antibodies elicited by Zika virus infection. Science 353, 823–826 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Godoy-Lozano, E. E. et al. Lower IgG somatic hypermutation rates during acute dengue virus infection is compatible with a germinal center-independent B cell response. Genome Med. 8, 23 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Liu, Y. et al. Cross-lineage protection by human antibodies binding the influenza B hemagglutinin. Nat. Commun. 10, 324 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Wu, X. et al. Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 333, 1593–1602 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Crotty, S. T follicular helper cell biology: a decade of discovery and diseases. Immunity 50, 1132–1148 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Brenna, E. et al. CD4+ T follicular helper cells in human tonsils and blood are clonally convergent but divergent from non-Tfh CD4+ cells. Cell Rep. 30, 137–152 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Morita, R. et al. Human blood CXCR5+CD4+ T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity 34, 108–121 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Dan, J. M. et al. A cytokine-independent approach to identify antigen-specific human germinal center T follicular helper cells and rare antigen-specific CD4+ T cells in blood. J. Immunol. 197, 983–993 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bentebibel, S. E. et al. Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination. Sci. Transl. Med. 5, 176ra132 (2013).

    Article  Google Scholar 

  33. Koutsakos, M., Nguyen, T. H. O. & Kedzierska, K. With a little help from T follicular helper friends: humoral immunity to influenza vaccination. J. Immunol. 202, 360–367 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Grifoni, A. et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell 181, 1489–1501 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Weiskopf, D. et al. Phenotype of SARS-CoV-2-specific T-cells in COVID-19 patients with acute respiratory distress syndrome. Sci. Immunol. 26, eabd2071 (2020).

    Article  Google Scholar 

  36. Wu, F. et al. Neutralizing antibody responses to SARS-CoV-2 in a COVID-19 recovered patient cohort and their implications. Preprint at https://www.medrxiv.org/content/10.1101/2020.03.30.20047365v2 (2020).

  37. Braun, J. et al. Presence of SARS-CoV-2 reactive T cells in COVID-19 patients and healthy donors. Preprint at https://www.medrxiv.org/content/10.1101/2020.03.30.20047365v2 (2020).

  38. Farooq, F. et al. Circulating follicular T helper cells and cytokine profile in humans following vaccination with the rVSV-ZEBOV Ebola vaccine. Sci. Rep. 6, 27944 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bentebibel, S. E. et al. ICOS+PD−1+CXCR3+ T follicular helper cells contribute to the generation of high-avidity antibodies following influenza vaccination. Sci. Rep. 6, 26494 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Niessl, J. et al. Persistent expansion and Th1-like skewing of HIV-specific circulating T follicular helper cells during antiretroviral therapy. EBioMedicine 54, 102727 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Zhang, J. et al. Circulating CXCR3+ Tfh cells positively correlate with neutralizing antibody responses in HCV-infected patients. Sci. Rep. 9, 10090 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Wrapp, D. et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367, 1260–1263 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Amanat, F. et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat. Med. https://doi.org/10.1038/s41591-020-0913-5 (2020).

  44. Hughes, C. S. et al. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments. Nat. Protoc. 14, 68–85 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Bern, M., Kil, Y. J. & Becker, C. Byonic: advanced peptide and protein identification software. Curr. Protoc. Bioinforma. 13, 13.20 (2012).

  46. Lee, L. Y. et al. Toward automated N-glycopeptide identification in glycoproteomics. J. Proteome Res. 15, 3904–3915 (2016).

    Article  CAS  PubMed  Google Scholar 

  47. Caly, L. et al. Isolation and rapid sharing of the 2019 novel coronavirus (SARS-CoV-2) from the first patient diagnosed with COVID-19 in Australia. Med. J. Aust. 212, 459–462 (2020).

    Article  PubMed  Google Scholar 

  48. Tiller, T. et al. Efficient generation of monoclonal antibodies from single human B cells by single cell RT–PCR and expression vector cloning. J. Immunol. Methods 329, 112–124 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Brochet, X., Lefranc, M. P. & Giudicelli, V. IMGT/V-QUEST: the highly customized and integrated system for IG and TR standardized V-J and V-D-J sequence analysis. Nucleic Acids Res. 36, W503–W508 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the generous participation of the trial individuals for providing samples. The SARS-CoV-2 RBD expression plasmids were kindly provided by F. Krammer (Icahn School of Medicine at Mt. Sinai). The human and mouse ACE2 expression plasmids were kindly provided by M. Thomas (Monash University). We acknowledge V. Jameson and staff at the Melbourne Cytometry Platform (Melbourne Brain Centre node) for provision of flow cytometry services. We thank the Melbourne Mass Spectrometry and Proteomics Facility of the Bio21 Molecular Science and Biotechnology Institute at the University of Melbourne for the support of mass spectrometry analysis. This study was supported by the Victorian Government and Medical Research Future Fund (MRFF) GNT2002073 (to W.-H.T., D.I.G., M.P.D., S.J.K. and A.K.W.), the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology (to S.J.K.), National Health and Medical Research Council (NHMRC) program grant APP1149990 (to S.J.K. and M.P.D.), NHMRC project grant GNT1162760 (to A.K.W), the Jack Ma Foundation (to D.I.G., N.A.G. and K.S.) and the A2 Milk Company (to K.S.). W-H.T. is a Howard Hughes Medical Institute–Wellcome Trust International Research Scholar (208693/Z/17/Z). J.A.J., D.I.G., M.P.D., W.-H.T., S.J.K. and A.K.W. are supported by NHMRC fellowships. The Melbourne WHO Collaborating Centre for Reference and Research on Influenza is supported by the Australian Government Department of Health.

Author information

Authors and Affiliations

Authors

Contributions

J.A.J., H.-X.T., W.S.L., S.J.K. and A.K.W. designed the study and experiments. J.A.J., H.-X.T., W.S.L., A.R., H.G.K., K.W., R.E., H.E.K, C.J.B., F.L.M., N.A.G., P.P., M.H.D., N.E.S. and A.K.W. performed experiments. W.-H.T., N.A.G., D.I.G. and K.S. contributed unique reagents. J.A.J., H.-X.T., W.S.L., A.R., K.S., M.P.D., S.J.K. and A.K.W. analyzed the experimental data. J.A.J., H.-X.T., W.S.L., A.R., M.P.D., S.J.K. and A.K.W. wrote the manuscript. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Stephen J. Kent or Adam K. Wheatley.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Saheli Sadanand was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1

Demographics and clinical characteristics of recruited COVID-19 patients and healthy control subjects.

Extended Data Fig. 2 Pre-screen of HCoV-HKU1 serum endpoint titres among a cohort of healthy subjects (N = 27) bled prior to the SARS-CoV-2 pandemic.

Data is shown as the median.

Extended Data Fig. 3 Correlations between antibody binding titres, ACE2/RBD binding inhibition and neutralization activity in plasma from subjects recovered from SARS-CoV-2 infection.

a, Spearman’s Correlation between endpoint titres of S-specific plasma antibody and the extent of ACE2/RBD binding inhibition, plasma neutralization titres or endpoint titres of RBD-specific plasma antibody (N = 41). b, Correlation between endpoint titres of RBD-specific plasma antibody and the extent of ACE2/RBD binding inhibition or plasma neutralziation activity (N = 41). c, Correlation between plasma ACE2/RBD binding inhibition and neutralization activity (N = 41). All correlations are two-tailed.

Extended Data Fig. 4 Representative staining of S- and RBD-specific IgD-IgG+ B cells.

3 uninfected subjects (left panels) and 6 subjects after recovery from SARS-CoV-2 infection (middle and right panels). CD19+IgD-IgG+ B cells cells were identified using gating strategy shown in Supplementary Fig. 1. Binding to SARS-CoV-2 spike and/or SARS-CoV-2 RBD probes was assessed.

Extended Data Fig. 5 Memory B cell phenotypes in subjects after SARS-CoV-2 infection.

a, b, Representative memory B cell phenotypes identified by CD21 and CD27 co-stain of probe+CD19+IgD- cells (blue) overlaid on CD19+IgD- cells (grey) (a) and the corresponding frequencies of the four populations in subjects previously infected with SARS-CoV-2 (b) (Resting memory—CD21+CD27+; activated memory—CD21CD27+; naïve/CD27lo memory—CD21+CD27; atypical B cells—CD21CD27); n.d—not detected due to absent probe+ cells. Memory B cell phenotypes were identified using gating strategy shown in Supplementary Fig. 2.

Extended Data Fig. 6 Analysis of V gene distribution, somatic mutation and CDR-H3 lengths for S- and RBD-specific BCR sequences.

a,b,c, Frequency distributions of human IGHV (a), IGKV (b) and IGLV (c) genes from BCR sequences recovered from S- (red) and RBD-binding (blue) IgG+ B cells from recovered COVID-19 patients (N = 5) with reference to distribution in cord blood (grey)49. Few IGLV sequences for RBD-binding B cells were recovered and are not shown. d, e, CDR-H3 (d) and somatic mutation (e) in S- (red) and RBD-binding (blue) IgG+ B cells compared to influenza B HA-specific IgG+ B cells recovered after immunisation23. f, Somatic mutation in S- (red) and RBD-binding (blue) IgG+ B cells for each individual subject. d,e,f, Data are shown as the median.

Extended Data Fig. 7 cTFH populations in SARS-CoV-2 positive and negative donors.

a, cTFH frequency (as a proportion of CD4+CD45RA cells) among SARS-CoV-2 negative (n = 10) and SARS-CoV-2 positive (n = 39) donors. b, Frequency of antigen-specific cTFH following stimulation with peptide pools spanning S1 (without RBD peptides), S2 or RBD regions of SARS-CoV-2 S in SARS-CoV-2 positive donors (n = 20). c, Frequency of IFNγ+, IL-17+ or TNF+ cells following SEB stimulation among cTFH or Tmem (CD4+CD45RACXCR5) populations in SARS-CoV-2 positive donors (n = 20; Wilcoxon two-tailed test). d, CCR6 and CXCR3 expression among bulk cTFH (grey), IFNγ+ (blue) or IL-17+ (red) cTFH cells following SEB stimulation in SARS-CoV-2 positive donors (n = 20). a, b, d, Data are shown as the median with interquartile range.

Extended Data Fig. 8 Peptide-specific cTFH responses in SARS-CoV-2 positive donors.

a, A 15-mer peptide derived from SARS-CoV-2 S2, LLQYGSFCTQLNRAL, was found to be immunogenic in 3 SARS-CoV-2 positive subjects (CP39, CP02, CP36). CP12 shown as representative non-responder. b, Plots indicate the CCR6/CXCR3 expression among S2 peptide pool- or peptide-specific cTFH for each donor.

Extended Data Fig. 9 CD4+ Tmem responses in COVID patients and controls.

a, Representative staining of CD134 and CD25 expression following protein or SEB stimulation among CD4 Tmem (CD4+CD45RA-CXCR5-) cells. b, Background-subtracted frequencies of SARS-CoV-2 S, SARS-CoV-2 RBD or HCoV HKU1 spike-specific CD4 Tmem cells among SARS-CoV-2 negative (n = 10) or SARS-CoV-2 positive (n = 39) donors. c, IFNγ, IL-17 and TNF responses following stimulation with overlapping peptide pools covering SARS-CoV-2 S1 or S2 domains, or SEB in SARS-CoV-2 positive donors (n = 20). d, Graphs indicate background-subtracted responses; responses not above background are indicated as 0.001% (n = 20). b, d, Data are shown as the median with interquartile range.

Extended Data Fig. 10 Association of immunological features with disease severity.

a, Analysis of the relationship between disease severity (mild, moderate or severe) and immunological parameters within the SARS-CoV-2 convalescent cohort. p < 0.05 indicated in bold (Kruskal-Wallis test, two-tailed). b, Relationship between cTFH, S-specific antibody titre, or microneutralization (MN) titre with disease severity (n = 26 mild, n = 10 moderate, n = 5 severe). Data are shown as the median with interquartile range.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Juno, J.A., Tan, HX., Lee, W.S. et al. Humoral and circulating follicular helper T cell responses in recovered patients with COVID-19. Nat Med 26, 1428–1434 (2020). https://doi.org/10.1038/s41591-020-0995-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-0995-0

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing