CD4+ T Follicular Helper Cells in Human Tonsils and Blood Are Clonally Convergent but Divergent from Non-Tfh CD4+ Cells

Elena Brenna, Alexey N Davydov, Kristin Ladell, James E McLaren, Paolo Bonaiuti, Maria Metsger, James D Ramsden, Sarah C Gilbert, Teresa Lambe, David A Price, Suzanne L Campion, Dmitriy M Chudakov, Persephone Borrow, Andrew J McMichael, Elena Brenna, Alexey N Davydov, Kristin Ladell, James E McLaren, Paolo Bonaiuti, Maria Metsger, James D Ramsden, Sarah C Gilbert, Teresa Lambe, David A Price, Suzanne L Campion, Dmitriy M Chudakov, Persephone Borrow, Andrew J McMichael

Abstract

T follicular helper (Tfh) cells are fundamental for B cell selection and antibody maturation in germinal centers. Circulating Tfh (cTfh) cells constitute a minor proportion of the CD4+ T cells in peripheral blood, but their clonotypic relationship to Tfh populations resident in lymph nodes and the extent to which they differ from non-Tfh CD4+ cells have been unclear. Using donor-matched blood and tonsil samples, we investigate T cell receptor (TCR) sharing between tonsillar Tfh cells and peripheral Tfh and non-Tfh cell populations. TCR transcript sequencing reveals considerable clonal overlap between peripheral and tonsillar Tfh cell subsets as well as a clear distinction between Tfh and non-Tfh cells. Furthermore, influenza-specific cTfh cell clones derived from blood can be found in the repertoire of tonsillar Tfh cells. Therefore, human blood samples can be used to gain insight into the specificity of Tfh responses occurring in lymphoid tissues, provided that cTfh subsets are studied.

Keywords: T follicular helper cells; TCR repertoire; blood; influenza; tonsil.

Conflict of interest statement

DECLARATION OF INTERESTS

The authors declare no competing interests.

Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.. Identification of Tonsillar and Circulating…
Figure 1.. Identification of Tonsillar and Circulating Tfh Populations in Donor-Matched Samples
Matched PBMC and MNC-T from adult donors were stained ex vivo. (A) Dot plot illustrating the expression of CXCR5, PD-1, CXCR3, and ICOS within the total CD4+CD45RA− cells in a representative matched blood and tonsil sample pair. (B) Heatmaps showing the mean percentage of CXCR3+ and ICOS+ cells within populations expressing different combinations of CXCR5 and PD-1 in 12 donors. The white crossed boxes indicate the near-absence of the PD-1hiCXCR5hi population in peripheral blood. (C) Dot plot illustrating the gating strategy used to define populations of circulating CXCR5+PD-1+Tfh (cTfh CXCR3+, cTfh CXCR3−) and CXCR5− non-Tfh memory cells (cNon-Tfh) and tonsillar CXCR5hiPD-1hi GC Tfh (tTfh GC), CXCR5intPD-1int (tTfh CXCR3+, tTfh CXCR3−) Tfh and CXCR5−PD-1− non-Tfh memory cells (tNon-Tfh) within the total CD4+CD45RA− T cell pool in one representative donor. (D) The percentage of each Tfh/non-Tfh subset within the CD4+CD45RA− memory T cell population in blood and tonsils from the 12 donors. (E–G) Production of IL-21 (E), IL-4 (F), and IFN-γ (G) after PMA and ionomycin stimulation by Tfh and non-Tfh populations in 10 donors. Non-analogous subsets were compared within tonsil and peripheral blood using the Kruskal-Wallis test. *p

Figure 2.. Clonotypic Overlap between Peripheral and…

Figure 2.. Clonotypic Overlap between Peripheral and Tonsillar Tfh Cell Populations

The top2000 TCR CDR3β…

Figure 2.. Clonotypic Overlap between Peripheral and Tonsillar Tfh Cell Populations
The top2000 TCR CDR3β nucleotide sequences in each population were analyzed using Cytoscape networking. (A, C, and E) As an example, illustrated is clonotype sharing between peripheral blood cTfh CXCR3− (A), cTfh CXCR3+ (C), and cNon-Tfh (E) and 4 tonsillar populations: tTfh CXCR3+ in yellow, tTfh CXCR3− in blue, tTfh GC in red, and tNon-Tfh in gray in one representative donor (donor 1). Each square represents a different clonotype; the size of the square is proportional to the clonotype’s normalized frequency within the parent population (for shared clonotypes, the highest normalized frequency is shown). The number of clonotypes shared between different populations is shown in bold, and the respective percentage is indicated below. (B, D, and F) To compare the overlap between blood cTfh CXCR3− (B), cTfh CXCR3+ (D), and cNon-Tfh (F) subsets and tonsillar populations, the percentage of the top2000 most frequent clonotypes shared between populations was calculated in each of the 4 donors (mean ± SEM). Non-parametric Kruskal-Wallis and Dunn’s multiple comparison tests: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. (G) A heatmap depicting the average number of clonotypes shared.

Figure 3.. Size Similarities in Clonotypes Shared…

Figure 3.. Size Similarities in Clonotypes Shared between cTfh and tTfh

(A–C) Overlap between TCR…

Figure 3.. Size Similarities in Clonotypes Shared between cTfh and tTfh
(A–C) Overlap between TCR CDR3β nucleotide sequences of the top2000 most frequent clonotypes of blood cTfh CXCR3− (A), cTfh CXCR3+ (B), and cNon-Tfh (C) populations with each tonsil population, as represented by the normalized F2 metric (mean ± SEM). Non-parametric Kruskal-Wallis and Dunn’s multiple comparison test: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. (D–H) Linear regression analysis of the correlation between the within-population frequency of the shared clonotypes in the two parent populations where clonotype sharing between populations was observed: cTfh CXCR3− with tTfh GC (D), tTfh CXCR3− with cTfh CXCR3− (E), cTfh CXCR3− with tTfh CXCR3+ (F), cTfh CXCR3+ with tTfh CXCR3+ (G), and cNon-Tfh with tNon-Tfh (H). Each graph illustrates a representative donor, and the tables show the number of shared clonotypes, the square of the Pearson correlation coefficient (R2), the slope value for each donor, and the p value of the significance of the difference in the slope from zero (ns, not significant).

Figure 4.. Repertoire Diversity Correlates with the…

Figure 4.. Repertoire Diversity Correlates with the Degree of Clonotypic Overlap from the In Silico…

Figure 4.. Repertoire Diversity Correlates with the Degree of Clonotypic Overlap from the In Silico Resampling Analysis
(A and B) Chao1 index (A) and normalized Shannon-Wiener index (B) of the top2000 frequencies for each population of cells analyzed in blood and tonsils from the 4 donors (mean ± SEM). Clonotypes of each subset were resampled using the bootstrap method. (C and D) The percentage of the top2000 shared clonotypes and the normalized F2 metric (mean ± SEM) of the overlap between the two replicates of blood (C) cTfh CXCR3+, cTfh CXCR3−, and cNon-Tfh as well as tonsil (D) tTfh GC, tTfh CXCR3+, tTfh CXCR3−, and tNon-Tfh populations is shown for all 4 donors (mean ± SEM). Non-parametric Kruskal-Wallis and Dunn’s multiple comparison test: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. (E and F) Unweighted (E) and weighted (F) analysis of the replicate overlap at increasing numbers of clonotypes considered from a representative population (cTfh CXCR3+ from donor 1). (G) The graphs show the linear regression correlation between the repertoire overlap, expressed either as percentage of shared clonotypes or normalized F2 metric, with both diversity indexes (Chao1 and normalized Shannon-Wiener) in all blood and tonsil populations from the 4 donors analyzed. See also Figure S2.

Figure 5.. Tfh Cell Subsets Are Clonotypically…

Figure 5.. Tfh Cell Subsets Are Clonotypically Distinct from Non-Tfh Cells

The top2000 TCR Vβ…

Figure 5.. Tfh Cell Subsets Are Clonotypically Distinct from Non-Tfh Cells
The top2000 TCR Vβ CDR3 nucleotide sequences of each population were analyzed by Cytoscape networking. (A and B) Examples of (A) clonotype sharing between peripheral blood cTfh CXCR3+ (yellow), cTfh CXCR3− (blue), and cNon-Tfh (gray) populations and (B) clonotype sharing between tonsillar populations tTfh CXCR3+ (yellow), tTfh CXCR3− (blue), tTfh GC (red), and tNon-Tfh (gray) populations in one representative donor (donor 1). Results are expressed as in Figure 2. (C) The percentage of the top2000 total shared clonotypes in addition to the uniquely shared clonotypes between tonsil populations was calculated in each of the 4 donors (mean ± SEM). (D) The normalized F2 metric of the top2000 frequencies was also calculated for the overlap between tTfh CXCR3+ and tTfh CXCR3− subsets and each of the other populations analyzed in the 4 donors (mean ± SEM). Non-parametric Kruskal-Wallis and Dunn’s multiple comparison test: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. (E) The table shows the number of shared clonotypes, the square of R2 in their size within populations, the slope value, and the p value of the difference in the slope from zero. See also Figure S3.

Figure 6.. Influenza Virus HA-Specific Clones Are…

Figure 6.. Influenza Virus HA-Specific Clones Are Predominantly Found in cTfh CXCR3 + and cNon-Tfh…

Figure 6.. Influenza Virus HA-Specific Clones Are Predominantly Found in cTfh CXCR3+ and cNon-Tfh Populations
(A) T cell libraries generated from Tfh and non-Tfh subsets sorted from three healthy blood donors from the National Health Service (NHS) Blood Transfusion Service were tested against the influenza H3/Wisconsin HA protein. The proliferation of each individual cell line (increase in counts above background, delta cpm) is shown (mean ± SEM). The red dotted line represents the cutoff used for identification of a positive response (defined as >3000 cpm, with a stimulation index >5). (B) Calculated precursor frequency of HA-specific cells per 106 cells in each subset (mean ± SEM). (C) Bar graph showing the number of influenza H1/California HA-specific T cell clones generated from each cell subset in the 4 further donors from whommatched tonsil samples were available. Non-parametric Kruskal-Wallis and Dunn’s multiple comparison test: *p +, cTfh CXCR3−, and cNon-Tfh clones were mapped, and the frequency of clones recognizing each peptide, normalized to the total frequency of HA-specific clones in all donors (D1, D2, D3, and D4), is plotted in the heatmap. The solid arrows highlight the peptides recognized by 3 subsets and the dashed arrows the peptides recognized by 3 donors. See also Figure S4.

Figure 7.. HA-Specific cTfh and cNon-Tfh Clones…

Figure 7.. HA-Specific cTfh and cNon-Tfh Clones Do Not Overlap, and HA-Specific cTfh CXCR3 +…

Figure 7.. HA-Specific cTfh and cNon-Tfh Clones Do Not Overlap, and HA-Specific cTfh CXCR3+ T Cell Clonotypes Can Be Detected in the tTfh CXCR3+ Subset
(A) TCR CDR3α and CDR3β sequencing was performed on clones specific for the same HA H1/California peptide that had been generated from either the cTfh CXCR3+, cTfh CXCR3−, or cNon-Tfh subset in 3 donors. For each clone, the peptide specificity, the CDR3α and CDR3β amino acid (AA) sequences, and TRAV/TRAJ and TRBV/TRBJ gene usage are shown. Clones are grouped based on the peptide recognized. (B and C) The TCR CDR3α and CDR3β region sequences of 75 HA-specific clones generated from the blood populations were determined, and the ex vivo Vα and Vβ repertoires of tonsil and blood Tfh and non-Tfh populations from the same subject were searched for matching sequences. Matches were detected only in donor 2. The frequency distributions of each of the individual clonotypes detected in the tonsil tTfh CXCR3+ (B) and peripheral blood cTfh CXCR3+ (C) repertoires are shown, with yellow circles indicating the sequences that matched those of the HA-specific cTfh CXCR3+ clones generated from the peripheral blood of the same donor. See also Figure S5.
All figures (7)
Figure 2.. Clonotypic Overlap between Peripheral and…
Figure 2.. Clonotypic Overlap between Peripheral and Tonsillar Tfh Cell Populations
The top2000 TCR CDR3β nucleotide sequences in each population were analyzed using Cytoscape networking. (A, C, and E) As an example, illustrated is clonotype sharing between peripheral blood cTfh CXCR3− (A), cTfh CXCR3+ (C), and cNon-Tfh (E) and 4 tonsillar populations: tTfh CXCR3+ in yellow, tTfh CXCR3− in blue, tTfh GC in red, and tNon-Tfh in gray in one representative donor (donor 1). Each square represents a different clonotype; the size of the square is proportional to the clonotype’s normalized frequency within the parent population (for shared clonotypes, the highest normalized frequency is shown). The number of clonotypes shared between different populations is shown in bold, and the respective percentage is indicated below. (B, D, and F) To compare the overlap between blood cTfh CXCR3− (B), cTfh CXCR3+ (D), and cNon-Tfh (F) subsets and tonsillar populations, the percentage of the top2000 most frequent clonotypes shared between populations was calculated in each of the 4 donors (mean ± SEM). Non-parametric Kruskal-Wallis and Dunn’s multiple comparison tests: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. (G) A heatmap depicting the average number of clonotypes shared.
Figure 3.. Size Similarities in Clonotypes Shared…
Figure 3.. Size Similarities in Clonotypes Shared between cTfh and tTfh
(A–C) Overlap between TCR CDR3β nucleotide sequences of the top2000 most frequent clonotypes of blood cTfh CXCR3− (A), cTfh CXCR3+ (B), and cNon-Tfh (C) populations with each tonsil population, as represented by the normalized F2 metric (mean ± SEM). Non-parametric Kruskal-Wallis and Dunn’s multiple comparison test: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. (D–H) Linear regression analysis of the correlation between the within-population frequency of the shared clonotypes in the two parent populations where clonotype sharing between populations was observed: cTfh CXCR3− with tTfh GC (D), tTfh CXCR3− with cTfh CXCR3− (E), cTfh CXCR3− with tTfh CXCR3+ (F), cTfh CXCR3+ with tTfh CXCR3+ (G), and cNon-Tfh with tNon-Tfh (H). Each graph illustrates a representative donor, and the tables show the number of shared clonotypes, the square of the Pearson correlation coefficient (R2), the slope value for each donor, and the p value of the significance of the difference in the slope from zero (ns, not significant).
Figure 4.. Repertoire Diversity Correlates with the…
Figure 4.. Repertoire Diversity Correlates with the Degree of Clonotypic Overlap from the In Silico Resampling Analysis
(A and B) Chao1 index (A) and normalized Shannon-Wiener index (B) of the top2000 frequencies for each population of cells analyzed in blood and tonsils from the 4 donors (mean ± SEM). Clonotypes of each subset were resampled using the bootstrap method. (C and D) The percentage of the top2000 shared clonotypes and the normalized F2 metric (mean ± SEM) of the overlap between the two replicates of blood (C) cTfh CXCR3+, cTfh CXCR3−, and cNon-Tfh as well as tonsil (D) tTfh GC, tTfh CXCR3+, tTfh CXCR3−, and tNon-Tfh populations is shown for all 4 donors (mean ± SEM). Non-parametric Kruskal-Wallis and Dunn’s multiple comparison test: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. (E and F) Unweighted (E) and weighted (F) analysis of the replicate overlap at increasing numbers of clonotypes considered from a representative population (cTfh CXCR3+ from donor 1). (G) The graphs show the linear regression correlation between the repertoire overlap, expressed either as percentage of shared clonotypes or normalized F2 metric, with both diversity indexes (Chao1 and normalized Shannon-Wiener) in all blood and tonsil populations from the 4 donors analyzed. See also Figure S2.
Figure 5.. Tfh Cell Subsets Are Clonotypically…
Figure 5.. Tfh Cell Subsets Are Clonotypically Distinct from Non-Tfh Cells
The top2000 TCR Vβ CDR3 nucleotide sequences of each population were analyzed by Cytoscape networking. (A and B) Examples of (A) clonotype sharing between peripheral blood cTfh CXCR3+ (yellow), cTfh CXCR3− (blue), and cNon-Tfh (gray) populations and (B) clonotype sharing between tonsillar populations tTfh CXCR3+ (yellow), tTfh CXCR3− (blue), tTfh GC (red), and tNon-Tfh (gray) populations in one representative donor (donor 1). Results are expressed as in Figure 2. (C) The percentage of the top2000 total shared clonotypes in addition to the uniquely shared clonotypes between tonsil populations was calculated in each of the 4 donors (mean ± SEM). (D) The normalized F2 metric of the top2000 frequencies was also calculated for the overlap between tTfh CXCR3+ and tTfh CXCR3− subsets and each of the other populations analyzed in the 4 donors (mean ± SEM). Non-parametric Kruskal-Wallis and Dunn’s multiple comparison test: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. (E) The table shows the number of shared clonotypes, the square of R2 in their size within populations, the slope value, and the p value of the difference in the slope from zero. See also Figure S3.
Figure 6.. Influenza Virus HA-Specific Clones Are…
Figure 6.. Influenza Virus HA-Specific Clones Are Predominantly Found in cTfh CXCR3+ and cNon-Tfh Populations
(A) T cell libraries generated from Tfh and non-Tfh subsets sorted from three healthy blood donors from the National Health Service (NHS) Blood Transfusion Service were tested against the influenza H3/Wisconsin HA protein. The proliferation of each individual cell line (increase in counts above background, delta cpm) is shown (mean ± SEM). The red dotted line represents the cutoff used for identification of a positive response (defined as >3000 cpm, with a stimulation index >5). (B) Calculated precursor frequency of HA-specific cells per 106 cells in each subset (mean ± SEM). (C) Bar graph showing the number of influenza H1/California HA-specific T cell clones generated from each cell subset in the 4 further donors from whommatched tonsil samples were available. Non-parametric Kruskal-Wallis and Dunn’s multiple comparison test: *p +, cTfh CXCR3−, and cNon-Tfh clones were mapped, and the frequency of clones recognizing each peptide, normalized to the total frequency of HA-specific clones in all donors (D1, D2, D3, and D4), is plotted in the heatmap. The solid arrows highlight the peptides recognized by 3 subsets and the dashed arrows the peptides recognized by 3 donors. See also Figure S4.
Figure 7.. HA-Specific cTfh and cNon-Tfh Clones…
Figure 7.. HA-Specific cTfh and cNon-Tfh Clones Do Not Overlap, and HA-Specific cTfh CXCR3+ T Cell Clonotypes Can Be Detected in the tTfh CXCR3+ Subset
(A) TCR CDR3α and CDR3β sequencing was performed on clones specific for the same HA H1/California peptide that had been generated from either the cTfh CXCR3+, cTfh CXCR3−, or cNon-Tfh subset in 3 donors. For each clone, the peptide specificity, the CDR3α and CDR3β amino acid (AA) sequences, and TRAV/TRAJ and TRBV/TRBJ gene usage are shown. Clones are grouped based on the peptide recognized. (B and C) The TCR CDR3α and CDR3β region sequences of 75 HA-specific clones generated from the blood populations were determined, and the ex vivo Vα and Vβ repertoires of tonsil and blood Tfh and non-Tfh populations from the same subject were searched for matching sequences. Matches were detected only in donor 2. The frequency distributions of each of the individual clonotypes detected in the tonsil tTfh CXCR3+ (B) and peripheral blood cTfh CXCR3+ (C) repertoires are shown, with yellow circles indicating the sequences that matched those of the HA-specific cTfh CXCR3+ clones generated from the peripheral blood of the same donor. See also Figure S5.

References

    1. Alam S, and Sant AJ (2011). Infection with seasonal influenza virus elicits CD4 T cells specific for genetically conserved epitopes that can be rapidly mobilized for protective immunity to pandemic H1N1 influenza virus. J. Virol 85, 13310–13321.
    1. Ansel KM, Ngo VN, Hyman PL, Luther SA, Förster R, Sedgwick JD, Browning JL, Lipp M, and Cyster JG (2000). A chemokine-driven positive feedback loop organizes lymphoid follicles. Nature 406, 309–314.
    1. Aschenbrenner D, Foglierini M, Jarrossay D, Hu D, Weiner HL, Kuchroo VK, Lanzavecchia A, Notarbartolo S, and Sallusto F (2018). An immuno-regulatory and tissue-residency program modulated by c-MAF in human TH17 cells. Nat. Immunol 19, 1126–1136.
    1. Avery DT, Bryant VL, Ma CS, de Waal Malefyt R, and Tangye SG (2008). IL-21-induced isotype switching to IgG and IgA by human naive B cells is differentially regulated by IL-4. J. Immunol 181, 1767–1779.
    1. Baumjohann D, Preite S, Reboldi A, Ronchi F, Ansel KM, Lanzavecchia A, and Sallusto F (2013). Persistent antigen and germinal center B cells sustain T follicular helper cell responses and phenotype. Immunity 38, 596–605.
    1. Becattini S, Latorre D, Mele F, Foglierini M, De Gregorio C, Cassotta A, Fernandez B, Kelderman S, Schumacher TN, Corti D, et al. (2014). Functional heterogeneity of human memory CD4+ T cell clones primed by pathogens or vaccines. Science 347, 400–406.
    1. Bentebibel S, Lopez S, Obermoser G, Schmitt N, Mueller C, Harrod C, Flano E, Mejias A, Albrecht RA, Blankenship D, et al. (2013). Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination. Sci. Transl. Med 5, 176ra32.
    1. Bolotin DA, Poslavsky S, Mitrophanov I, Shugay M, Mamedov IZ, Putintseva EV, and Chudakov DM (2015). MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381.
    1. Boswell KL, Paris R, Boritz E, Ambrozak D, Yamamoto T, Darko S, Wloka K, Wheatley A, Narpala S, McDermott A, et al. (2014). Loss of circulating CD4 T cells with B cell helper function during chronic HIV infection. PLoS Pathog. 10, e1003853.
    1. Bowyer G, Grobbelaar A, Rampling T, Venkatraman N, Morelle D, Ballou RW, Hill AVS, and Ewer KJ (2018). CXCR3+ T follicular helper cells induced by co-administration of RTS,S/AS01B and viral-vectored vaccines are associated with reduced immunogenicity and efficacy against malaria. Front. Immunol 9, 1660.
    1. Boyden AW, Legge KL, and Waldschmidt TJ (2012). Pulmonary infection with influenza A virus induces site-specific germinal center and T follicular helper cell responses. PLoS ONE 7, e40733.
    1. Breitfeld D, Ohl L, Kremmer E, Ellwart J, Sallusto F, Lipp M, and Förster R (2000). Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J. Exp. Med 192, 1545–1552.
    1. Bryant VL, Ma CS, Avery DT, Li Y, Good KL, Corcoran LM, deWaal Malefyt R, and Tangye SG (2007). Cytokine-mediated regulation of human B cell differentiation into Ig-secreting cells: predominant role of IL-21 produced by CXCR5+ T follicular helper cells. J. Immunol 179, 8180–8190.
    1. Campion SL, Brodie TM, Fischer W, Korber BT, Rossetti A, Goonetilleke N, McMichael AJ, and Sallusto F (2014). Proteome-wide analysis of HIV-specific naive and memory CD4(+) T cells in unexposed blood donors. J. Exp. Med 211, 1273–1280.
    1. Casamayor-Palleja M, Khan M, and MacLennan IC (1995). A subset of CD4+ memory T cells contains preformed CD40 ligand that is rapidly but transiently expressed on their surface after activation through the T cell receptor complex. J. Exp. Med 181, 1293–1301.
    1. Caza T, and Landas S (2015). Functional and phenotypic plasticity of CD4(+) T cell subsets. BioMed Res. Int 2015, 521957.
    1. Chevalier N, Jarrossay D, Ho E, Avery DT, Ma CS, Yu D, Sallusto F, Tangye SG, and Mackay CR (2011). CXCR5 expressing human central memory CD4 T cells and their relevance for humoral immune responses. J. Immunol 185, 5556–5568.
    1. Choi YS, Kageyama R, Eto D, Escobar TC, Johnston RJ, Monticelli L, Lao C, and Crotty S (2011). ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity 34, 932–946.
    1. Colwell RK, Chao A, Gotelli NJ, Lin S, Mao CX, Chazdon RL, and Longino JT (2012). Modelsand estimators linking individual-based and sample-based rarefaction, extrapolation and comparison of assemblages. J. Plant Ecol 5, 3–21.
    1. Crotty S (2014). T follicular helper cell differentiation, function, and roles in disease. Immunity 41, 529–542.
    1. Crotty S (2018). Do Memory CD4 T Cells Keep Their Cell-Type Programming: Plasticity versus Fate Commitment? Complexities of interpretation due to the heterogeneity of memory CD4 T Cells, including T follicular helper cells. Cold Spring Harb. Perspect. Biol 10, a032102.
    1. Cubas R, van Grevenynghe J, Wills S, Kardava L, Santich BH, Buckner CM, Muir R, Tardif V, Nichols C, Procopio F, et al. (2015). Reversible reprogramming of circulating memory T follicular helper cell function during chronic HIV infection. J. Immunol 195, 5625–5636.
    1. Dan JM, Havenar-Daughton C, Kendric K, Al-Kolla R, Kaushik K, Rosales SL, Anderson EL, LaRock CN, Vijayanand P, Seumois G, et al. (2019). Recurrent group A Streptococcus tonsillitis is an immunosusceptibility disease involving antibody deficiency and aberrant TFH cells. Sci. Transl. Med 11, 1–11.
    1. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C, et al. (2006). PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443, 350–354.
    1. Djuretic IM, Levanon D, Negreanu V, Groner Y, Rao A, and Ansel KM (2007). Transcription factors T-bet and Runx3 cooperate to activate Ifng and silence Il4 in T helper type 1 cells. Nat. Immunol 8, 145–153.
    1. Dorfman DM, Brown JA, Shahsafaei A, and Freeman GJ (2006). Programmed death-1 (PD-1) is a marker of germinal center-associated T cells and angioimmunoblastic T-cell lymphoma. Am. J. Surg. Pathol 30, 802–810.
    1. Durand M, Walter T, Pirnay T, Naessens T, Gueguen P, Goudot C, Lameiras S, Chang Q, Talaei N, Ornatsky O, et al. (2019). Human lymphoid organ cDC2 and macrophages play complementary roles in T follicular helper responses. J. Exp. Med 216, 1561–1581.
    1. Eren MI, Chao A, Hwang WH, and Colwell RK (2012). Estimating the richness of a population when the maximum number of classes is fixed: a nonparametric solution to an archaeological problem. PLoS ONE 7, e34179.
    1. Fazilleau N, Eisenbraun MD, Malherbe L, Ebright JN, Pogue-Caley RR, McHeyzer-Williams LJ, and McHeyzer-Williams MG (2007). Lymphoid reservoirs of antigen-specific memory T helper cells. Nat. Immunol 8, 753–761.
    1. Förster R, Mattis AE, Kremmer E, Wolf E, Brem G, and Lipp M (1996). A putative chemokine receptor, BLR1, directs B cell migration to defined lymphoid organs and specific anatomic compartments of the spleen. Cell 87, 1037–1047.
    1. Geiger R, Duhen T, Lanzavecchia A, and Sallusto F (2009). Human naive and memory CD4+ T cell repertoires specific for naturally processed antigens analyzed using libraries of amplified T cells. J. Exp. Med 206, 1525–1534.
    1. Grivel JC, and Margolis L (2009). Use of human tissue explantsto study human infectious agents. Nat. Protoc 4, 256–269.
    1. Groom JR, and Luster AD (2011). CXCR3 in T cell function. Exp. Cell Res 317, 620–631.
    1. Hale JS, and Ahmed R (2015). Memory T follicular helper CD4 T cells. Front. Immunol 6, 16.
    1. Hansen DS, Obeng-Adjei N, Ly A, Ioannidis LJ, and Crompton PD (2017). Emerging concepts in T follicular helper cell responses to malaria. Int. J. Parasitol 47, 105–110.
    1. Harrell FE (2001). Regression Modeling Strategies With Applications To Linear Models, Logistic Regression, and Survival Analysis (Springer; ).
    1. Havenar-Daughton C, Carnathan DG, Torrents de la Peña A, Pauthner M, Briney B, Reiss SM, Wood JS, Kaushik K, van Gils MJ, Rosales SL, et al. (2016). Direct probing of germinal center responses reveals immunological features and bottlenecks for neutralizing antibody responses to HIV Env trimer. Cell Rep. 17, 2195–2209.
    1. Haynes NM, Allen CDC, Lesley R, Ansel KM, Killeen N, and Cyster JG (2007). Role of CXCR5 and CCR7 in follicular Th cell positioning and appearance of a programmed cell death gene-1high germinal center-associated subpopulation. J. Immunol 179, 5099–5108.
    1. He J, Tsai LM, Leong YA, Hu X, Ma CS, Chevalier N, Sun X, Vandenberg K, Rockman S, Ding Y, et al. (2013). Circulating precursor CCR7(lo) PD-1(hi) CXCR5+ CD4+ T cells indicate Tfh cell activity and promote antibody responses upon antigen reexposure. Immunity 39, 770–781.
    1. Heather JM, Ismail M, Oakes T, and Chain B (2018). High-throughput sequencing of the T-cell receptor repertoire: pitfalls and opportunities. Brief. Bioinform 19, 554–565.
    1. Hegazy AN, Peine M, Helmstetter C, Panse I, Fröhlich A, Bergthaler A, Flatz L, Pinschewer DD, Radbruch A, and Löhning M (2010). Interferons direct Th2 cell reprogramming to generate a stable GATA-3(+)T-bet(+) cell subset with combined Th2 and Th1 cell functions. Immunity 32, 116–128.
    1. Heit A, Schmitz F, Gerdts S, Flach B, Moore MS, Perkins JA, Robins HS, Aderem A, Spearman P, Tomaras GD, et al. (2017). Vaccination establishes clonal relatives of germinal center T cells in the blood of humans. J. Exp. Med 214, 2139–2152.
    1. Herati RS, Muselman A, Vella L, Bengsch B, Parkhouse K, Alcazar D, Del, Kotzin J, Doyle SA, Tebas P, Hensley SE, et al. (2017). Successive annual influenza vaccination induces a recurrent oligoclonotypic memory response in circulating T follicular helper cells. Sci. Immunol 2, eaag2152.
    1. Hill DL, Pierson W, Bolland DJ, Mkindi C, Carr EJ, Wang J, Houard S, Wingett SW, Audran R, Wallin EF, et al. (2019). The adjuvant GLA-SE promotes human Tfh cell expansion and emergence of public TCRβ clonotypes. J. Exp. Med 216, 1857–1873.
    1. Izraelson M, Nakonechnaya TO, Moltedo B, Egorov ES, Kasatskaya SA, Putintseva EV, Mamedov IZ, Staroverov DB, Shemiakina II, Zakharova MY, et al. (2018). Comparative analysis of murine T-cell receptor repertoires. Immunology 153, 133–144.
    1. Kim CH, Rott LS, Clark-Lewis I, Campbell DJ, Wu L, and Butcher EC (2001). Subspecialization of CXCR5+ T cells: B helper activity is focused in a germinal center-localized subset of CXCR5+ T cells. J. Exp. Med 193, 1373–1381.
    1. Lefranc MP (2003). IMGT, the international ImMunoGeneTics database. Nucleic Acids Res. 31, 307–310.
    1. Lindestam Arlehamn CS, Gerasimova A, Mele F, Henderson R, Swann J, Greenbaum JA, Kim Y, Sidney J, James EA, Taplitz R, et al. (2013). Memory T cells in latent Mycobacterium tuberculosis infection are directed against three antigenic islands and largely contained in a CXCR3+CCR6+ Th1 subset. PLoS Pathog. 9, e1003130.
    1. Liu YJ, Joshua DE, Williams GT, Smith CA, Gordon J, and MacLennan ICM (1989). Mechanism of antigen-driven selection in germinal centres. Nature 342, 929–931.
    1. Locci M, Havenar-Daughton C, Landais E, Wu J, Kroenke MA, Arlehamn CL, Su LF, Cubas R, Davis MM, Sette A, et al.; International AIDSVaccine Initiative Protocol C Principal Investigators (2013). Human circulating PD-1+CXCR3−CXCR5+ memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody responses. Immunity 39, 758–769.
    1. Locci M, Wu JE, Arumemi F, Mikulski Z, Dahlberg C, Miller AT, and Crotty S (2016). Activin A programs the differentiation of human TFH cells. Nat. Immunol 17, 976–984.
    1. Ma CS, Suryani S, Avery DT, Chan A, Nanan R, Santner-Nanan B, Deenick EK, and Tangye SG (2009). Early commitment of naïve human CD4(+) T cells to the T follicular helper (T(FH)) cell lineage is induced by IL-12. Immunol. Cell Biol 87, 590–600.
    1. Mele F, Fornara C, Jarrossay D, Furione M, Arossa A, Spinillo A, Lanzavecchia A, Gerna G, Sallusto F, and Lilleri D (2017). Phenotype and specificity of T cells in primary human cytomegalovirus infection during pregnancy: IL-7Rpos long-term memory phenotype is associated with protection from vertical transmission. PLoS ONE 12, e0187731.
    1. Miura K, Orcutt AC, Muratova OV, Miller LH, Saul A, and Long CA (2008). Development and characterization of a standardized ELISA including a reference serum on each plate to detect antibodies induced by experimental malaria vaccines. Vaccine 26, 193–200.
    1. Morita R, Schmitt N, Bentebibel SE, Ranganathan R, Bourdery L, Zurawski G, Foucat E, Dullaers M, Oh S, Sabzghabaei N, et al. (2011). 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.
    1. Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L, Wang YH, Watowich SS, Jetten AM, Tian Q, and Dong C (2008). Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity 29, 138–149.
    1. O’Shea JJO, Cells CDT, and Paul WE (2010). Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science 327, 1098–1102.
    1. Obeng-Adjei N, Portugal S, Tran TM, Yazew TB, Skinner J, Li S, Jain A, Felgner PL, Doumbo OK, Kayentao K, et al. (2015). Circulating Th1-Cell-type Tfh cells that exhibit impaired B cell help are preferentially activated during acute malaria in children. Cell Rep. 13, 425–439.
    1. Palladino G, Scherle PA, and Gerhard W (1991). Activity of CD4+ T-cell clones of type 1 and type 2 in generation of influenza virus-specific cytotoxic responses in vitro. J. Virol 65, 6071–6076.
    1. Pallikkuth S, Parmigiani A, Silva SY, George VK, Fischl M, Pahwa R, and Pahwa S (2012). Impaired peripheral blood T-follicular helper cell function in HIV-infected nonresponders to the 2009 H1N1/09 vaccine. Blood 120, 985–993.
    1. Pedersen GK, Sjursen H, Nøstbakken JK, Jul-Larsen Å, Hoschler K, and Cox RJ (2014). Matrix M(™) adjuvanted virosomal H5N1 vaccine induces balanced Th1/Th2 CD4(+) T cell responses in man. Hum. Vaccin. Immunother 10, 2408–2416.
    1. Pepper M, Pagán AJ, Igyártó BZ, Taylor JJ, and Jenkins MK (2011). Opposing signals from the Bcl6 transcription factor and the interleukin-2 receptor generate T helper 1 central and effector memory cells. Immunity 35, 583–595.
    1. Ploquin MJY, Eksmond U, and Kassiotis G (2011). B cells and TCR avidity determine distinct functions of CD4+ T cells in retroviral infection. J. Immunol 187, 3321–3330.
    1. Precopio ML, Butterfield TR, Casazza JP, Little SJ, Richman DD, Koup RA, and Roederer M (2008). Optimizing peptide matrices for identifying T-cell antigens. Cytometry A 73, 1071–1078.
    1. Price DA, Brenchley JM, Ruff LE, Betts MR, Hill BJ, Roederer M, Koup RA, Migueles SA, Gostick E, Wooldridge L, et al. (2005). Avidity for antigen shapes clonal dominance in CD8+ T cell populations specific for persistent DNA viruses. J. Exp. Med 202, 1349–1361.
    1. Quigley MF, Almeida JR, Price DA, and Douek DC (2011). Unbiased molecular analysis of T Cell receptor expression using template-switch anchored RT-PCR. Curr. Protoc. Immunol Chapter 10, Unit 10.33.
    1. Reinhardt RL, Liang HE, and Locksley RM (2009). Cytokine-secreting follicular T cells shape the antibody repertoire. Nat. Immunol 10, 385–393.
    1. Rosati E, Dowds CM, Liaskou E, Henriksen EKK, Karlsen TH, and Franke A (2017). Overview of methodologies for T-cell receptor repertoire analysis. BMC Biotechnol. 17, 61.
    1. Ryg-Cornejo V, Ioannidis LJ, Ly A, Chiu CY, Tellier J, Hill DL, Preston SP, Pellegrini M, Yu D, Nutt SL, et al. (2016). Severe malaria infections impair germinal center responses by inhibiting T follicular helper cell differentiation. Cell Rep. 14, 68–81.
    1. Sallusto F, Cassotta A, Hoces D, Foglierini M, and Lanzavecchia A (2018). Do Memory CD4 T Cells Keep Their Cell-Type Programming: Plasticity versus Fate Commitment? T-Cell heterogeneity, plasticity, and selection in humans. Cold Spring Harb. Perspect. Biol 10, a029421.
    1. Sant AJ, DiPiazza AT, Nayak JL, Rattan A, and Richards KA (2018). CD4 T cells in protection from influenza virus: Viral antigen specificity and functional potential. Immunol. Rev 284, 91–105.
    1. Schaerli P, Willimann K, Lang AB, Lipp M, Loetscher P, and Moser B (2000). CXC chemokine receptor 5 expression defines follicular homing T cells with B cell helper function. J. Exp. Med 192, 1553–1562.
    1. Shi J, Hou S, Fang Q, Liu X, Liu X, and Qi H (2018). PD-1 controls follicular T helper cell positioning and function. Immunity 49, 264–274. e4.
    1. Shugay M, Bolotin DA, Putintseva EV, Pogorelyy MV, Mamedov IZ, and Chudakov DM (2013). Huge overlap of individual TCR beta repertoires. Front. Immunol 4, 466.
    1. Shugay M, Britanova OV, Merzlyak EM, Turchaninova MA, Mamedov IZ, Tuganbaev TR, Bolotin DA, Staroverov DB, Putintseva EV, Plevova K, et al. (2014). Towards error-free profiling of immune repertoires. Nat. Methods 11, 653–655.
    1. Shugay M, Bagaev DV,Turchaninova MA, Bolotin DA, Britanova OV, Putintseva EV, Pogorelyy MV, Nazarov VI, Zvyagin IV, Kirgizova VI, et al. (2015). VDJtools: unifying post-analysis of T cell receptor repertoires. PLoS Comput. Biol 11, e1004503.
    1. Tew JG, and Mandel TE (1979). Prolonged antigen half-life inthe lymphoid follicles of specifically immunized mice. Immunology 37, 69–76.
    1. Tew JG, Mandel TE, Phipps RP, and Szakal AK (1984). Tissue localization and retention of antigen in relation to the immune response. Am. J. Anat 170, 407–420.
    1. Valkenburg SA, Li OTW, Li A, Bull M, Waldmann TA, Perera LP, Peiris M, and Poon LLM (2018). Protection by universal influenza vaccine is mediated by memory CD4 T cells. Vaccine 36, 4198–4206.
    1. Vella LA, Buggert M, Manne S, Herati RS, Sayin I, Kuri-Cervantes L, Bukh Brody I, O’Boyle KC, Kaprielian H, Giles JR, et al. (2019). T follicular helper cells in human efferent lymph retain lymphoid characteristics.J. Clin. Invest 129, 3185–3200.
    1. Velu V, Mylvaganam GH, Gangadhara S, Hong JJ, Iyer SS, Gumber S, Ibegbu CC, Villinger F, and Amara RR (2016). Induction of Th1-biased T follicular helper (Tfh) cells in lymphoid tissues during chronic simian immune-deficiency virus infection defines functionally distinct germinal center Tfh cells. J. Immunol 197, 1832–1842.
    1. Vinuesa CG, Tangye SG, Moser B, and Mackay CR (2005). Follicular B helper T cells in antibody responses and autoimmunity. Nat. Rev. Immunol 5, 853–865.
    1. Vinuesa CG, Linterman MA, Yu D, and MacLennan ICM (2016). Follicular helper T cells. Annu. Rev. Immunol 34, 335–368.
    1. Weber JP, Fuhrmann F, Feist RK, Lahmann A, Al Baz MS, Gentz LJ, Vu Van D, Mages HW, Haftmann C, Riedel R, et al. (2015). ICOS maintains the T follicular helper cell phenotype by down-regulating Krüppel-like factor 2. J. Exp. Med 212, 217–233.
    1. Wong MT, Ong DEH, Lim FSH, Teng KWW, McGovern N, Narayanan S, Ho WQ, Cerny D, Tan HKK, Anicete R, et al. (2016). A High-dimensional atlas of human T cell diversity reveals tissue-specific trafficking and cytokine signatures. Immunity 45, 442–456.
    1. Xu H, Li X, Liu D, Li J, Zhang X, Chen X, Hou S, Peng L, Xu C, Liu W, et al. (2013). Follicular T-helper cell recruitment governed by bystander B cells and ICOS-driven motility. Nature 496, 523–527.
    1. Yang J, James E, Gates TJ, DeLong JH, LaFond RE, Malhotra U, and Kwok WW (2013). CD4+ T cells recognize unique and conserved 2009 H1N1 influenza hemagglutinin epitopes after natural infection and vaccination. Int. Immunol 25, 447–457.
    1. Yoon SW, Wong SS, Zhu H, Chen R, Li L, Zhang Y, Guan Y, and Webby RJ (2018). Dysregulated T-helper type 1 (Th1):Th2 cytokine profile and poor immune response in pregnant ferrets infected with 2009 pandemic influenza A(H1N1) virus. J. Infect. Dis 217, 438–442.
    1. Yusuf I, Kageyama R, Monticelli L, Johnston RJ, Ditoro D, Hansen K, Barnett B, and Crotty S (2010). Germinal center T follicular helper cell IL-4 production is dependent on signaling lymphocytic activation molecule receptor (CD150). J. Immunol 185, 190–202.
    1. Zielinski CE, Corti D, Mele F, Pinto D, Lanzavecchia A, and Sallusto F (2011). Dissecting the human immunologic memory for pathogens. Immunol. Rev 240, 40–51.

Source: PubMed

3
Suscribir