Generation of Ag-specific humoral responses requires the orchestrated development and function of highly specialized immune cells in secondary lymphoid organs. We used a multiparametric approach combining flow cytometry, confocal microscopy, and histocytometry to analyze, for the first time to our knowledge in children, tonsils from seasonal influenza–vaccinated children. We used these novel imaging assays to address the mucosal immune dynamics in tonsils investigating the spatial positioning, frequency, and phenotype of immune cells after vaccination. Vaccination was associated with a significantly higher frequency of follicular helper CD4 T cells compared with the unvaccinated control group. The imaging analysis revealed that potential suppressor (FOXP3hi) CD4 T cells are mainly located in extrafollicular areas. Furthermore, a significantly reduced frequency of both follicular and extrafollicular FOXP3hi CD4 T cells was found in the vaccine group compared with the control group. Levels of circulating CXCL13 were higher in those vaccinated compared with controls, mirroring an increased germinal center reactivity in the tonsils. Notably, a strong correlation was found between the frequency of tonsillar T follicular helper cells and tonsillar Ag-specific Ab-secreting cells. These data demonstrate that influenza vaccination promotes the prevalence of relevant immune cells in tonsillar follicles and support the use of tonsils as lymphoid sites for the study of germinal center reactions after vaccination in children.

Vaccine efficacy is strictly dependent on the generation of Ag-specific Abs and linked to the differentiation of long-lived memory B cells able to respond to rechallenge. Follicular helper CD4 T (TFH) cells represent a subset of highly specialized lymphoid organ CD4 T cells essential for helping B cells and able to regulate the germinal center (GC) reaction (13). TFH cells express a unique phenotypic profile characterized by high expression of surface receptors like PD-1, ICOS, CXCR4, and CD95 (4, 5). Subpopulations of this heterogeneous CD4 T cell compartment have been previously described based on the expression of CD57 (6). Furthermore, TFH cells express a unique molecular signature compared with other CD4 T cell populations (4, 7, 8). The trafficking of CD4 and B cells within the lymphoid organ is mediated by the interaction between chemokines (mainly CCL19/CCL21 and CXCL13) and their ligands (CCR7 and CXCR5) (9), whereas the interaction between TFH cells and GC B cells relies on a complex network made of soluble mediators (i.e., IL-4, IL-21) and surface receptors (i.e., CD40, PD-1, ICOS) (3). Besides the TFH cells, other CD4 subsets have been recently described in the follicle including the CD4 follicular regulatory CD4 T (TFR) cells, a population likely originated from FOXP3hi regulatory CD4 T (TREG) cells (3). These cells are capable of controlling the magnitude of the GC reactivity (10).

Given the difficulty to acquire secondary lymphoid organs, particularly in pediatric settings, many studies have focused on the investigation of circulating memory CXCR5hi CD4 T cells as counterparts of the GC TFH cells (11). However, their origin and relationship to bona fide GC TFH cells is not well understood (1214). More recently, the use of the levels of circulating CXCL13 as a surrogate for GC reactivity after vaccination has been shown (15).

Tonsils are chronically exposed to foreign Ags, provide protection against respiratory pathogens such as influenza, and their crypt epithelium is rich in lymphocytes, thus behaving as a lymphoid compartment (16). The access to secondary lymphoid organs is extremely challenging in humans, especially in children. By extensions, tonsils could represent a valuable and approachable secondary lymphoid organ. Investigation of the cell dynamics and immune reactions in such anatomical sites would provide valuable information regarding the cellular and molecular mechanisms governing the generation of these responses and further fuel the development of novel vaccine strategies.

All the patients were enrolled at the Children’s Hospital Bambino Gesù in Rome between October 2015 and October 2016. It was a prospective observational study involving pediatric patients aged 3–15 y scheduled for elective tonsillectomy. Apart from fulfilling the criteria for tonsillectomy, our patients are otherwise healthy, showing no sign of immune compromise. They had not been vaccinated against influenza during the previous years. Children in the vaccine arm had been immunized with the quadrivalent vaccine (Fluarix Tetra; GlaxoSmithKline Biologicals) consisting of 60 μg hemagglutinin per 0.5 ml dose, in the recommended ratio of 15 μg of hemagglutinin in each of the following virus strains: A/California/7/2009 (H1N1), A/Switzerland/9715293/2013 (H3N2), B/Phuket/3073/2013, and B/Brisbane/60/2008.

Tonsils were obtained from children scheduled for elective tonsillectomy. Tonsils from vaccinated children were collected 9 ± 2 d after vaccination. Part of the tonsil specimen was formalin-fixed and then embedded in paraffin blocks. Tonsillar mononuclear cells were isolated from the remaining specimen by mechanical disruption followed by Ficoll-Paque density gradient centrifugation. Plasma samples were collected from whole blood before and after vaccination in the vaccinated group and at the day of the surgery for the nonvaccinated group.

Flow cytometry.

Polychromatic flow cytometry was performed using the following directly conjugated Abs: 1) BD Biosciences: CD3-H7APC (SK7), BCL6-PE (K112-91), CD134 (OX40)-BV650 (ACT35), IgM-Cy5PE (G20-127), IgG-Cy5PE (G18-145), CD210 (IL-10R)-PE (3F9), IgG-APC (G18-145), CD19-APC (HIB19), CD95-BV421 (DX2); 2) eBioscience: HELIOS-FITC (22F6); 3) BioLegend: CD185 (CXCR5)-FITC (J252D4), CD150 (SLAM)-PE (A12-7D4), CD279 (PD-1)-BV711 (EH12.2H7), CD20-BV570 (2H7), CD278 (ICOS)-PB (C398.4A), Ki-67–Alexa Fluor 700 (Ki-67), FOXP3-PB (206D), CD27-BV605 (O323), CD38-BV785 (HIT2), CD124 (IL-4R)-Cy7PE (G077F6), CCR7-BV605 (G043H7); 4) Invitrogen: CD4-Cy5.5PE (S3.5); 5) Beckman Coulter: CD19-ECD, CD45-ECD, CD27-PC5; 6) R&D Systems: TGF-β RII-FLUORESCIN; 7) SouthernBiotech: IgD-PE. CD57 (NK1) Ab was conjugated in-house.

Confocal imaging.

Tissue sections were stained with titrated amounts of the following Abs; 1) primary: anti-HELIOS (N3C3; GeneTex), anti–PD-1 (goat polyclonal; R&D Systems), anti-BCL6 (PG-B6p; Dako), anti-IgD (EPR6146; Abcam), anti-CXCL13 (goat polyclonal; Thermo Fisher Scientific); 2) secondary: anti-mouse IgG1 Alexa Fluor 546 and anti-goat IgG Alexa Fluor 546 (Life Technologies) and anti-rabbit IgG BV421 (BioLegend); 3) primary conjugated: Ki-67-BV510 (B56; BD Biosciences), CD4–Alexa Fluor 488 (goat polyclonal; R&D Systems), FOXP3 Alexa Fluor 647 (206D; BioLegend), JOJO-1 iodide (for nuclear staining; Life Technologies). The CD20-PB (L26) Ab was conjugated in-house.

Tonsillar mononuclear cell suspensions were analyzed by flow cytometry using the aforementioned Abs. Briefly, 2–3 × 106 cells were thawed and rested for 2 h at 37°C. Following incubation with Live/Dead Fixable Aqua (Invitrogen) cells were surface stained with a titrated amount of Abs. FOXP3 and BCL6 staining was performed using the Transcription Factor Buffer Set (BD Pharmingen) following the manufacturer’s instructions. Intracellular staining for Ki-67 was performed using the Fixation/Permeabilization Solution Kit (BD Biosciences) following the manufacturer’s instructions. After washing, cells were fixed with 1% paraformaldehyde and 0.5–1 × 106 events were acquired on a modified LSR II flow cytometer (Becton Dickinson, San Jose, CA). Electronic compensation was performed with Ab capture beads (BD Biosciences). Data were analyzed using FlowJo version 9.9 (Tree Star).

Tissue imaging analysis was performed as previously described (17). Tissue blocks were sliced into 10-μm sections and tissue sections were mounted on glass slides. Following a deparaffinization/rehydration step, Ag retrieval was performed in Borg Decloaker RTU (Biocare Medical) in a decloaking chamber heating slides to 110°C for 15 min. After blocking and permeabilization, slides were incubated (overnight at 4°C) with titrated primary Abs. Next, slides were washed in PBS and incubated for 2 h in the dark with Alexa Fluor dye-conjugated secondary Abs prior to staining with titrated amounts of primary-conjugated Abs. Finally, JOJO-1 (Life Technologies) was applied and slides were mounted with Fluoromount-G solution (SouthernBiotech). Confocal images were obtained on a NIKON (C2si) confocal system operating NIS-Elements Advanced Research with 40× (numerical aperture 1.3) and 20× (numerical aperture 0.75) objectives and analyzed using the Imaris software version 8.4 (Bitplane). Spectral spillover between optical detection channels was corrected through live spectral unmixing using data acquired from samples singly stained with the respective fluorochromes. Images for histocytometry were acquired at a 512 × 512-pixel density and the method was applied as previously published (17, 18). Briefly, imaging datasets were segmented postacquisition based on their nuclear staining signal and average voxel intensities for all channels were extrapolated in Imaris after iso-surface generation. Data were then exported to Microsoft Excel, concatenated into a single comma-separated values format, and imported into FlowJo version 10 for further analysis.

Plasma samples were analyzed with Luminex bead-based multiplexed assay according to the manufacturer’s instructions.

Hemagglutination inhibition (HI) Ab titer for each strain of influenza was determined before and after vaccination as previously described (19).

A FluoroSpot assay was used for the quantification of Ag-specific Ab-secreting cells (ASCs) present in the tonsil-derived cells. Ag-specific responses were examined by wells coated with H1N1 A/California/07/2009, H3N2 A/Switzerland/9715293/2013, or B Phuket/3073/2013 Yamagata lineage Ags diluted 1:20. These Ags were provided by Italian National Institute of Health, Rome. Wells coated with keyhole limpet hemocyanin (0.5 μg per well) (Sigma-Aldrich) were used as negative control, whereas wells coated with capture mAb MT91/145 (MabTech), recognizing total IgG, were used as positive control. After an overnight incubation at 4°C with the Ags, wells were blocked with 200 μl RPMI 1640 medium (Life Technologies) supplemented with 10% FBS (Life Technologies) for 2 h at 37°C. Thawed cells from 20 patients and 12 controls were rested for 2 h and then 300,000 (Ag-coated wells) or 50,000 (capture mAb-coated wells) tonsil cells were added on the FluoroSpot plates for 20 h at 37°C. After washing the plates, detection mAb anti-human IgG (MT78/145) (MabTech) was added on the plates for 2 h at room temperature. The plates were developed using the fluorescence enhancer (MabTech). Spots were counted using an AID FluoroSpot reader. Spots in the well with negative control were subtracted from the number of spots in the wells with Ag. Experiments were performed in triplicate and the number of spots were normalized for millions per cell.

Statistical significance was assessed using the two-tailed Student t test paired or unpaired according to the analysis for normal distributed data. Analyses of nonnormal distributed data were carried out with Mann–Whitney U test for unpaired data and with Wilcoxon matched-pairs signed rank test for comparison of two time points. Correlations were determined by using Pearson and Spearman analysis. Results were considered to be significant for p < 0.05. Statistical analysis was performed with Prism software (version 6; GraphPad Software, La Jolla, CA).

This study was approved by the local ethics committee of the Bambino Gesù Children’s Hospital-Research Institute and written informed consent was obtained in accordance with the Declaration of Helsinki from parents or guardians of each child participant on the child’s behalf.

The dynamics of relevant immune cell types in lymphoid organs after vaccination, especially in children, are not well understood. We sought to investigate the dynamics of B and T cell subsets in tonsils after i.m. influenza vaccination from a cohort of 36 children (Table I). Previous history of any vaccination during the last year before study entry was considered as an exclusion criterion. The percentage of total lymphocytes as well as the amount of IgM, IgG, and IgA Igs, and the age and gender did not differ among the groups at the baseline (Table I). We applied an imaging assay allowing for the simultaneous detection of CD4, CD20, Ki-67, IgD, and JOJO-1 (nucleus) and the identification of follicular areas (Fig. 1A). The obtained imaging data were further analyzed with histocytometry (17, 18). Our imaging analysis showed that GCs are enriched with B cells characterized by increased expression, judged by the mean fluorescence intensity (MFI), of CD20 compared with mantle zone B cells where naive (IgDhi) and memory non-GC B cells locate (Fig. 1B). Afterwards, the combination of CD20 and Ki-67 was used as a surrogate for the detection of GCs. In line with previous reports, our imaging analysis showed that tonsils are highly populated with B cell follicles, most of them expressing a polarized profile judged by the localization of Ki-67hi proliferating B cells (20) (Fig. 1C). Furthermore, the majority of follicular CD4 T cells express a PD-1hi phenotype and among them many were also characterized by a PD-1hiCD57hi phenotype (Fig. 1C) in agreement with previous studies (6, 20).

Table I.
Demographic and immunologic characteristics of the patients enrolled
TotalVaccinatedControl
Number 36 24 12 
Sex, M/F 22/14 17/7 5/7 
Age y, mean (SD) 6.3 (2.5) 5.6 (1.4) 7.7 (3.4) 
WBC 103 cells/μl, median (IQR) 9.2 (6.5–11.7) 7.96 (6.2–11.0) 10.6 (9.0–13.0) 
Lymphocytes 103 cells/μl, median (IQR) 3.2 (2.5–4.0) 3.2 (2.5–3.9) 3.2 (2.7–5.0) 
IgA mg/dl, median (IQR) 139 (93.0–194.5) 139 (93–196.5) 132 (85.25–187.8) 
IgG mg/dl, median (IQR) 1049 (892–1164) 1074 (900-1164) 1025 (852–1289) 
IgM mg/dl, median (IQR) 98.5 (77.25–226.0) 98.5 (77.25–116.3) 101 (77.25–129.8) 
TotalVaccinatedControl
Number 36 24 12 
Sex, M/F 22/14 17/7 5/7 
Age y, mean (SD) 6.3 (2.5) 5.6 (1.4) 7.7 (3.4) 
WBC 103 cells/μl, median (IQR) 9.2 (6.5–11.7) 7.96 (6.2–11.0) 10.6 (9.0–13.0) 
Lymphocytes 103 cells/μl, median (IQR) 3.2 (2.5–4.0) 3.2 (2.5–3.9) 3.2 (2.7–5.0) 
IgA mg/dl, median (IQR) 139 (93.0–194.5) 139 (93–196.5) 132 (85.25–187.8) 
IgG mg/dl, median (IQR) 1049 (892–1164) 1074 (900-1164) 1025 (852–1289) 
IgM mg/dl, median (IQR) 98.5 (77.25–226.0) 98.5 (77.25–116.3) 101 (77.25–129.8) 

Previous history of any vaccination during the last year before study entry was considered as an exclusion criterion.

F, female; IQR, interquartile range; M, male.

FIGURE 1.

Accumulation of tonsillar TFH CD4 T cells after influenza vaccination. (A) Confocal images (original magnification ×40) showing follicular areas from a vaccinated tonsil defined by the expression of CD4, CD20, Ki-67, IgD, and JOJO-1. A zoomed area (red box) with individual markers used is shown in the lower panel. (B) Histocytometry gating strategy used to define GC coordinates based on IgD and Ki-67 expression. The histograms show the CD20 expression (MFI) in the GCs (IgDloKi-67hi) compared with the mantle zone (IgDhiKi-67lo). (C) Confocal image (original magnification ×40) showing the expression of CD4, CD57, PD-1, CD20, and Ki-67 in a tonsil from a nonvaccinated individual. A zoomed area (red box) is shown too. (D) Flow cytometry gating scheme for identification of tonsillar CD4 T cell subsets. Specific CD4 T cell populations are shown in different colors. (E) Flow cytometry pooled data showing the increased relative frequency of PD-1hiCD57lo CD4 T cells in vaccinated (n = 24) and control (n = 12) individuals. (F) Histocytometry gating scheme used for the identification of follicles (CD20, Ki-67) and the detection of CD4 T cell populations. T cells areas were identified based on the CD4 density, whereas GC coordinates were detected within the CD20hiKi-67hi cells. The simultaneous expression of PD-1 and Ki-67 on CD4 T cells from GC and T cell areas is shown on the right panel. (G) Histocytometry pooled data showing the PD-1hiCD57lo TFH cells expressed as frequency of either total or GC CD4 T cells in control (n = 6) and vaccinated (n = 6) individuals. Each symbol represents a different GC. Symbols with the same shape represent GCs from the same donor. Mann–Whitney U test was used for statistical analysis. Mean values (horizontal lines) and SD error bars are shown.

FIGURE 1.

Accumulation of tonsillar TFH CD4 T cells after influenza vaccination. (A) Confocal images (original magnification ×40) showing follicular areas from a vaccinated tonsil defined by the expression of CD4, CD20, Ki-67, IgD, and JOJO-1. A zoomed area (red box) with individual markers used is shown in the lower panel. (B) Histocytometry gating strategy used to define GC coordinates based on IgD and Ki-67 expression. The histograms show the CD20 expression (MFI) in the GCs (IgDloKi-67hi) compared with the mantle zone (IgDhiKi-67lo). (C) Confocal image (original magnification ×40) showing the expression of CD4, CD57, PD-1, CD20, and Ki-67 in a tonsil from a nonvaccinated individual. A zoomed area (red box) is shown too. (D) Flow cytometry gating scheme for identification of tonsillar CD4 T cell subsets. Specific CD4 T cell populations are shown in different colors. (E) Flow cytometry pooled data showing the increased relative frequency of PD-1hiCD57lo CD4 T cells in vaccinated (n = 24) and control (n = 12) individuals. (F) Histocytometry gating scheme used for the identification of follicles (CD20, Ki-67) and the detection of CD4 T cell populations. T cells areas were identified based on the CD4 density, whereas GC coordinates were detected within the CD20hiKi-67hi cells. The simultaneous expression of PD-1 and Ki-67 on CD4 T cells from GC and T cell areas is shown on the right panel. (G) Histocytometry pooled data showing the PD-1hiCD57lo TFH cells expressed as frequency of either total or GC CD4 T cells in control (n = 6) and vaccinated (n = 6) individuals. Each symbol represents a different GC. Symbols with the same shape represent GCs from the same donor. Mann–Whitney U test was used for statistical analysis. Mean values (horizontal lines) and SD error bars are shown.

Close modal

Tonsil-derived cells were analyzed with polychromatic flow cytometry (Fig. 1D). CD27 and CD45RO markers were used for defining naive and memory subsets and in combination with CCR7 and CXCR5 (Fig. 1D), chemokine receptors critical for the intrafollicular trafficking of CD4 T cells (1). Our flow cytometry analysis revealed higher relative frequency of TFH cells, reaching significant levels for the PD-1hiCD57lo cells, in tonsils from vaccinated compared with nonvaccinated donors (Fig. 1E).

Next, tonsil-derived tissues were investigated by a multiplexed confocal imaging assay and histocytometry. The gating scheme for identification of follicles/GCs and detection of TFH cells is shown in Fig. 1F. Our quantitative imaging analysis showed a significantly higher frequency, expressed as a percentage of either total or GC CD4 T cells, of PD-1hiCD57lo CD4 T cells in vaccinated group (n = 6) compared with control group tonsils (n = 6) (Fig. 1G), in line with our flow cytometry derived data (Fig. 1E).

Tonsil-derived cells were further investigated for the expression of Ox40, ICOS, and SLAM (CD150) (Fig. 2A), surface receptors associated with the development and function of TFH cells (1, 4, 2123). No difference was found between the vaccine and control groups when the expression of Ox40 and ICOS on TFH cells was investigated (Fig. 2B, left panel, Supplemental Fig. 1, left panel). However, a significantly higher relative frequency of CD150dim TFH cells, specifically in the PD-1hiCD57lo TFH compartment, was found (Fig. 2B, central and right panels, Supplemental Fig. 1, central and right panels). Altogether, our data show that influenza vaccination is associated with a significantly higher prevalence of TFH cell subsets in tonsils.

FIGURE 2.

Preferential accumulation of tonsillar CD150lo TFH CD4 T cells after influenza vaccination. (A) Flow cytometry plots showing the expression of OX40, ICOS, and CD150 (SLAM) on TFH cell subpopulations (upper panel) and naive CD4 T cells (lower panel). (B) Flow cytometry pooled data showing the relative frequency of PD-1hiCD57lo TFH cells expressing a OX40hiICOShi (left panel), CD150hi (middle panel) or CD150lo (right panel) phenotype. Tonsil-derived cells from control (n = 12) and vaccinated (n = 24) donors were analyzed. An unpaired t test for normally distributed data was used for statistical analysis. Mean values (horizontal lines) and SD error bars are shown.

FIGURE 2.

Preferential accumulation of tonsillar CD150lo TFH CD4 T cells after influenza vaccination. (A) Flow cytometry plots showing the expression of OX40, ICOS, and CD150 (SLAM) on TFH cell subpopulations (upper panel) and naive CD4 T cells (lower panel). (B) Flow cytometry pooled data showing the relative frequency of PD-1hiCD57lo TFH cells expressing a OX40hiICOShi (left panel), CD150hi (middle panel) or CD150lo (right panel) phenotype. Tonsil-derived cells from control (n = 12) and vaccinated (n = 24) donors were analyzed. An unpaired t test for normally distributed data was used for statistical analysis. Mean values (horizontal lines) and SD error bars are shown.

Close modal

Previous studies have shown a possible role for CD4 TREG cells as regulators of follicular helper T cell activity (24, 25). Therefore, we sought to investigate the dynamics of FOXP3hi CD4 T cells, a population with potential suppressor activity, in our cohort. First, the localization of FOXP3hi CD4 T cells was investigated by a multiplexed confocal imaging assay allowing for the simultaneous detection of CD20, Ki-67, CD4, PD-1, and FOXP3, and also HELIOS, another transcription factor found in CD4 suppressor subsets (26, 27) (Fig. 3A). Further analysis using histocytometry (Fig. 3B) confirmed a preferential localization of FOXP3hi CD4 T cells in the extrafollicular areas compared with follicles/GCs both in control (Fig. 3C, upper panels) and vaccine groups (data not shown). Moreover, histocytometry data revealed a reduction of TREG cells upon vaccination, especially in the T cell area (Fig. 3C, lower panels). Next, tonsil-derived cells were analyzed with flow cytometry allowing for the simultaneous expression of FOXP3, HELIOS, and BCL6, a master regulator of TFH cells (1) (Supplemental Fig. 2A). We found a significantly lower frequency of FOXP3hi cells in the vaccine compared with control group for all CD4 populations tested (Fig. 3D, Supplemental Fig. 2B). Furthermore, we found a dichotomy regarding the expression of FOXP3 and BCL6 (Supplemental Fig. 2A). Again, the frequency of FOXP3+BCL6 and FOXP3hiHELIOSlo/hi CD4 T cells was found consistently lower in the tonsils from vaccinated children compared with the control group (Fig. 3E, Supplemental Fig. 2C). Our data indicate that influenza vaccination is associated with reduced frequency of potential suppressor CD4 T cells within a secondary lymphoid organ.

FIGURE 3.

Influenza vaccination is associated with reduced frequency of tonsillar FOXP3hi CD4 T cells. (A) Confocal images (original magnification ×40) showing the detection of FOXP3, HELIOS, CD4, and PD-1 in tonsils from a nonvaccinated (upper panel) and a vaccinated donor (lower panel). Zoomed areas (yellow boxes) are also shown. (B) Representative histocytometry derived plots showing the simultaneous expression of HELIOS and FOXP3 in CD4 T cells located in GC and T cell area. (C) Pooled histocytometry data showing the frequency of FOXP3hi (HELIOShi or lo) CD4 T cells in GC compared with T cell area (upper panel) or between control and vaccinated samples in T and GC areas (lower panel). (D) Flow cytometry pooled data showing the relative frequency of tonsillar FOXP3hi CD4 T cells in CD27hiCD45ROhi, PD-1hiCD57lo, and PD-1hiCD57hi from vaccinated (n = 24) and control (n = 12) donors. (E) Flow cytometry pooled data showing the relative frequency of tonsillar FOXP3hiPD-1hiCD57lo TFH cells expressing BCL6lo, HELIOSlo, or HELIOShi phenotypes from vaccinated (n = 24) and control (n = 12) donors. Mann–Whitney U test was used for the statistical analysis. n = sample size. Mean values (horizontal lines) and SD error bars are shown.

FIGURE 3.

Influenza vaccination is associated with reduced frequency of tonsillar FOXP3hi CD4 T cells. (A) Confocal images (original magnification ×40) showing the detection of FOXP3, HELIOS, CD4, and PD-1 in tonsils from a nonvaccinated (upper panel) and a vaccinated donor (lower panel). Zoomed areas (yellow boxes) are also shown. (B) Representative histocytometry derived plots showing the simultaneous expression of HELIOS and FOXP3 in CD4 T cells located in GC and T cell area. (C) Pooled histocytometry data showing the frequency of FOXP3hi (HELIOShi or lo) CD4 T cells in GC compared with T cell area (upper panel) or between control and vaccinated samples in T and GC areas (lower panel). (D) Flow cytometry pooled data showing the relative frequency of tonsillar FOXP3hi CD4 T cells in CD27hiCD45ROhi, PD-1hiCD57lo, and PD-1hiCD57hi from vaccinated (n = 24) and control (n = 12) donors. (E) Flow cytometry pooled data showing the relative frequency of tonsillar FOXP3hiPD-1hiCD57lo TFH cells expressing BCL6lo, HELIOSlo, or HELIOShi phenotypes from vaccinated (n = 24) and control (n = 12) donors. Mann–Whitney U test was used for the statistical analysis. n = sample size. Mean values (horizontal lines) and SD error bars are shown.

Close modal

Next the dynamics of tonsillar B cell subsets were investigated by applying a polychromatic flow cytometry assay (Fig. 4A). No differences were found when total or memory subsets, defined by the expression of IgD and CD27, of CD19hi B cells were analyzed (Fig. 4B, Supplemental Fig. 3A, upper panels). We focused our analysis in the CD19hiIgDloCD27hi B cell memory population that can be further divided into subsets based on the expression of CD20 and CD38 with CD20dimCD38hi representing enriched plasma cells and CD20dimCD38lo and CD20hiCD38dim representing enriched memory non-GC and GC B cell compartments respectively (Fig. 4A). No difference between control group and vaccinated group tonsils was found for the B cell subsets investigated (Supplemental Fig. 3A, lower panels). Furthermore, similar frequency of CD20hiCD38dimCD95hi GC B cells were found between the two groups (Fig. 4B, upper panel). However, a significantly lower frequency of CD20hiCD38dimIgGhi B cells was found in tonsils from the vaccinated group compared with controls (Fig. 4B, lower panel). The expression of cytokine receptors (IL-4R, IL-10R, and TGFβ RII) critical for the development of B cell responses (28) was analyzed by flow cytometry. To facilitate the comparison of MFI values obtained from different experiments, we considered the naive B cell population as our reference and the ratio of the MFI on a given B cell subset to the MFI on naive B cells was calculated. A significantly higher expression of IL-4R on plasma cells (CD20dimCD38hi) was found in the vaccinated group compared with control group (Fig. 4B, right panel). No differences were found for the other cytokine receptors tested (data not shown). We further analyzed local B cell populations with histocytometry (Supplemental Fig. 3B). Ki-67hi/Ki-67hiBCL6hi were almost exclusively found in the GC area (Supplemental Fig. 3C). A trend, although not significant, for higher frequencies of B cells expressing a BCL6hiKi-67hi phenotype was found in the tonsils from the vaccinated group compared with control group (Supplemental Fig. 3C, right panel).

FIGURE 4.

Influenza vaccination is associated with increased levels of circulating CXCL13 and altered distribution of follicular CXCL13. (A) Flow cytometry gating scheme showing the identification of tonsillar B cells subsets. Memory B cell subsets were defined based on the expression of CD27 and IgD, whereas further analysis of the CD27hiIgDlo population was carried out using the markers CD20 and CD38. (B) Flow cytometry pooled data showing the relative frequency of tonsillar total (CD19hi) B cells and subsets of GC-enriched B cells (CD27hiIgDloCD20hiCD38dim) from vaccinated (n = 24) and control (n = 12) tonsils. The expression level of IL-4R on plasma cells (PC), normalized to the level of matched naive (PC/naive ratio) between the two groups (right panel). (C) Pooled data showing the circulating levels (picograms per milliliter) of CXCL13 in control (n = 17) and vaccinated (n = 22) donors, pre- and postvaccination. (D) Confocal images (original magnification ×40) showing the distribution of CXCL13 (in red) in tonsils from one vaccinated (upper panel) and one control (lower panel) donor. One zoomed area from each donor is also shown (right panel). (E) Dot plots showing the average frequency (%) of CXCL13hi GC B (upper) and T (lower) cells. Tissues from vaccinated individuals (n = 3, in red) with high circulating CXCL13 titers and controls (n = 3, in black) with low CXCL13 titers were analyzed. Each dot represents the average of all GCs per donor analyzed. An unpaired t test for normally distributed data was used for statistical analysis for (B) and a Wilcoxon matched-pairs signed rank test was used for comparison between prevaccine and postvaccine samples. Mean values (horizontal lines) and SD error bars are shown.

FIGURE 4.

Influenza vaccination is associated with increased levels of circulating CXCL13 and altered distribution of follicular CXCL13. (A) Flow cytometry gating scheme showing the identification of tonsillar B cells subsets. Memory B cell subsets were defined based on the expression of CD27 and IgD, whereas further analysis of the CD27hiIgDlo population was carried out using the markers CD20 and CD38. (B) Flow cytometry pooled data showing the relative frequency of tonsillar total (CD19hi) B cells and subsets of GC-enriched B cells (CD27hiIgDloCD20hiCD38dim) from vaccinated (n = 24) and control (n = 12) tonsils. The expression level of IL-4R on plasma cells (PC), normalized to the level of matched naive (PC/naive ratio) between the two groups (right panel). (C) Pooled data showing the circulating levels (picograms per milliliter) of CXCL13 in control (n = 17) and vaccinated (n = 22) donors, pre- and postvaccination. (D) Confocal images (original magnification ×40) showing the distribution of CXCL13 (in red) in tonsils from one vaccinated (upper panel) and one control (lower panel) donor. One zoomed area from each donor is also shown (right panel). (E) Dot plots showing the average frequency (%) of CXCL13hi GC B (upper) and T (lower) cells. Tissues from vaccinated individuals (n = 3, in red) with high circulating CXCL13 titers and controls (n = 3, in black) with low CXCL13 titers were analyzed. Each dot represents the average of all GCs per donor analyzed. An unpaired t test for normally distributed data was used for statistical analysis for (B) and a Wilcoxon matched-pairs signed rank test was used for comparison between prevaccine and postvaccine samples. Mean values (horizontal lines) and SD error bars are shown.

Close modal

Next, the blood levels of chemokines (CXCL13) and cytokines (IL-6, BAFF, IL-10, IL-21) with critical role for the GC reactivity (1) were measured. Significantly higher levels of CXCL13, a surrogate for GC reactivity (15), was found in the vaccinated group compared with control group (Fig. 4C), whereas no difference was found for the rest of the cytokines tested (Supplemental Fig. 3D). The tissue expression of CXCL13 was investigated in tonsils from vaccinated donors with high levels of circulating CXCL13 (n = 3) and controls with low levels of circulating CXCL13 (n = 3). Our imaging analysis showed that CXCL13 was mainly localized in the follicular area (Fig. 4D, Supplemental Fig. 4). However, a different follicular distribution of CXCL13 was observed in the follicles from vaccinated compared with control subjects (Fig. 4D, Supplemental Fig. 4). A higher spread of CXCL13 across the follicle/GC was found in most of the follicles (42/61 follicles with homogeneous-spread pattern) from vaccinated individuals compared with controls (23/62 follicles with homogeneous-spread pattern) where a polarized distribution of CXCL13 was found in the majority of follicles analyzed. Furthermore, histocytometry analysis revealed a higher number of follicles harboring increased frequencies of CXCL13hi B and TFH cells in tonsils in the vaccinated compared with nonvaccinated groups (Fig. 4E). Our data point to a higher in situ production of CXCL13.

We further investigated whether the described tonsil immune dynamics have any impact on the development of the vaccine-specific B cell responses. To this end, the possible association between tonsil-derived Ag-specific ASCs and particular tonsillar CD4 T cell subsets was investigated. The specific HI titer against each strain of the influenza vaccine was measured in the entire cohort at the baseline and after vaccination and showed as titer and fold increase (Fig. 5A, 5B). Although none of the donors had previously received any vaccination for influenza, most of them were seroprotected (19) (≥1:40) against each strain of influenza virus especially against H3N2 and H1N1 strains at the baseline due to the previous exposure to influenza Ags (Fig. 5B). However, vaccination was able to boost Ag-specific responses especially against H1N1, H3N2, and B Phuket strains (Fig. 5A, 5B). Tonsil-derived cells were analyzed ex vivo for Ag-specific ASCs against H1N1, H3N2, and B Phuket strains using a B cell FluoroSpot assay (29). Firstly, we found a difference in the frequency for Ag-specific ASCs between vaccinated donors and control, confirming the validity of our assay (Fig. 5C). Moreover, among the populations analyzed, a significant positive correlation was found between the frequency of PD-1hiCD57lo tonsillar TFH cells and IgG ASCs specific for each strain of influenza virus (Fig. 5D). Furthermore, the PD-1hiCD57loSLAMdim TFH cell subset strongly correlated with ASCs specific for H1N1 and H3N2 but not for B Phuket (Fig. 5D, right panels). Our data suggest that vaccination-induced tonsillar TFH cell dynamics reflect on the development of vaccine-specific B cell responses.

FIGURE 5.

Significant correlation between tonsillar PD-1hiCD57lo TFH cells and tonsillar Ag-specific ASCs. (A) Circos plot showing the differential fold increase in an HI assay for the four different Ags contained in the influenza vaccine. Bow thickness defines level of fold increase. Cumulative and single patient fold increase for every Ag is shown in the bandage below the Ag labels and patient labels, respectively. Circos plot was generated by circos.ca. (B) Bar graph showing the HI titer before (T0) and after (T1) vaccination for all the 24 patients vaccinated. The horizontal line represents the protection rate (1:40). (C) Plots showing the difference in the frequency of tonsil flu-specific ASCs between vaccinated donors and control. (D) Plots showing the correlation between the absolute counts of PD-1hiCD57lo (left panel) or PD-1hiCD57loCD150lo (right panel) TFH subsets and the number of tonsil IgG-producing ASCs specific for three out of the four strains included in the 2015/2016 seasonal influenza vaccine. Spearman rank test was used for the correlations. Mean values (horizontal lines) and SD error bars are shown.

FIGURE 5.

Significant correlation between tonsillar PD-1hiCD57lo TFH cells and tonsillar Ag-specific ASCs. (A) Circos plot showing the differential fold increase in an HI assay for the four different Ags contained in the influenza vaccine. Bow thickness defines level of fold increase. Cumulative and single patient fold increase for every Ag is shown in the bandage below the Ag labels and patient labels, respectively. Circos plot was generated by circos.ca. (B) Bar graph showing the HI titer before (T0) and after (T1) vaccination for all the 24 patients vaccinated. The horizontal line represents the protection rate (1:40). (C) Plots showing the difference in the frequency of tonsil flu-specific ASCs between vaccinated donors and control. (D) Plots showing the correlation between the absolute counts of PD-1hiCD57lo (left panel) or PD-1hiCD57loCD150lo (right panel) TFH subsets and the number of tonsil IgG-producing ASCs specific for three out of the four strains included in the 2015/2016 seasonal influenza vaccine. Spearman rank test was used for the correlations. Mean values (horizontal lines) and SD error bars are shown.

Close modal

Advances made in the recent past have emphasized the importance of specialized locations in secondary lymphoid organs that specifically support the variety of cell types and functions. Studies on mice model and human tissue highlight the crucial role of microanatomy and spatial organization for the proper control and initiation of innate and adaptive immune responses facilitating the interaction between lymphocytes and APCs (30). It is indeed increasingly evident that rather than in a stable, local, integrated, and stereotypical manner, immune cells work in a flexible and largely dynamic system (31). In this study, we sought to investigate the dynamics of different lymphocyte subsets in tonsils after seasonal influenza vaccine describing the effect of vaccination on the phenotype and spatial segregation of B and T cells at tissue level.

Tonsil-derived cells and tissues were investigated by cutting-edge polychromatic flow cytometry, confocal imaging assays, and, to our knowledge for the first time in children, by histocytometry. Tonsils constitute a secondary lymphoid organ associated with nasopharyngeal mucosa with unique location and structure, providing protection against respiratory pathogens (16). Although the majority of the tonsillar tissue is covered by follicular B cell areas, distinct follicular immune cell types (i.e., TFH cell subsets) can be observed similar to lymph nodes (LNs) (20). Considering that children are continuously scheduled for tonsillectomy, tonsils could represent a valuable and approachable secondary lymphoid organ compared with LNs, which are not easily accessible in this population.

Different molecular signatures and spatial localization within particular follicular areas, presumably associated with the exposure to different local signals, could contribute to the heterogeneity of the TFH cell pool. Our imaging analysis was able to define follicular areas and facilitate the quantitative analysis of the relevant T and B cell populations in these areas. Despite the uniform expression of some of the receptors (i.e., PD-1, CXCR5, ICOS) used for TFH cell characterization, a divergent expression was found for others like OX40, CD150, and CD57 in agreement with previous studies (4, 20). The relationship between phenotype and function of particular TFH cell subpopulations is not well understood (20, 32, 33). In line with previous data (20, 34), we found the majority of PD-1hiCD57hi TFH cells had been localized within the GC area, whereas those expressing a PD-1hiCD57lo profile were found within and mainly around the GC area. Differentiation of TFH cells starts in the T cell zones and interfollicular areas (35) and gradually translocate to the follicle. Although a comprehensive analysis of factors mediating the spatial localization of TFH subsets in human lymphoid organs is lacking, studies in mouse models have shown the contribution of several cellular/molecular mechanisms in this process, including the following: 1) The gradient of relevant chemokines like CCL19/CCL21 (ligands for CCR7) and CXCL13 (ligand for CXCR5). Downregulation of CCR7 with concomitant upregulation of CXCR5 has been shown to be a major regulator for the trafficking of CD4 into the follicle (9). Within the follicle, positioning of TFH cells could be further affected by the local expression of SDF1 (ligand for CXCR4, a receptor highly expressed on TFH and B cells) (3638). Furthermore, compartmentalization of other chemokines, like CXCL10 and CXCL9, produced by several cell types, including monocytes (39, 40), could affect not only the differentiation of TFH cells but their trafficking within the LN areas, too (41). 2) The relative expression of S1PR family receptors; transcription of S1PR1, a dominant receptor facilitating the egression of T cells into bloodstream (42), is significantly reduced in TFH cells (4). The concomitant expression of high levels of CD69, a negative regulator of S1PR1 (43), on TFH cells may further contribute to a reduced capacity of TFH cells for egression into the bloodstream. On the other hand, expression of S1PR2, which exerts cellular functions opposite to these of S1PR1 (44), has been shown to be critical for the positioning of TFH cells in the follicle (45). 3) The relative expression of a complex network of signaling transducers like STAT3 (a potent regulator of both BCL6 and Blimp-1) (46, 47) and transcriptional factors involved in the differentiation and trafficking of TFH cells like BCL6 (promotes the downregulation of CCR7 and upregulation of CXCR5) (48), KLF2 (increases Blimp-1 and S1PR1, negative regulators of TFH cells) (49), and EB12 (its downregulation mediates the restriction of the primary TFH cell subpopulation in the GC) (50). 4) The presence of Ag (persistent Ag in the follicle sustains TFH cell responses) (51, 52). Vaccination was associated with a significant induction of all TFH cells, particularly the PD-1hiCD57lo ones. This was confirmed by both flow cytometry and tissue histocytometry analysis. These findings suggest that an induction of specific TFH subsets with potential helper function occurs early after an antigenic stimulation. Although no differences were found between the vaccinated and control groups when the expression of OX40 and ICOS was investigated, a significantly higher frequency of CD150lo TFH cells was found in the vaccinated group. Previous studies have shown that CD150-SAP axis is critical for the development of TFH cells (23, 53). A reduced in vivo cycling of CD150lo TFH cells was previously described (4) indicating that these cells are possibly more differentiated compared with CD150hi TFH cells. Interestingly, CD150lo TFH cells were able to secrete, after in vitro stimulation, the highest amounts of IL-4, a critical cytokine for the development of GC B cell responses (54), among LN CD4 T cell subsets tested (4). Our data indicate that vaccination was associated with the induction of potent helper CD4 T cells in the follicle. However, the role of such TFH cell subpopulations in the development of Ag-specific B cell responses needs further investigation.

The presence of suppressor TFR cells (FOXP3hi) that can control the GC responses has been previously described (10, 55). Although it is still controversial whether TFH and TFR cell profiles represent different CD4 T cell lineages or cells with the same origin but in various stages of differentiation, the concept of T helper commitment has recently been challenged by several data suggesting plasticity between different T follicular cell populations (3, 56). Several subsets with suppressor activity have been described like conventional TREG, CD25hiTFR, and CD25loTFR cells (57, 58). Imaging analysis showed that the majority of potential suppressor FOXP3hi CD4 T cells were located outside the follicle. Our data are in line with previous reports showing TFR cells, especially the FOXP3hiCD25hi ones, represent a small fraction of follicular CD4 T cells (4, 25). Our flow cytometry analysis showed that vaccination was associated with a significantly lower frequency of both extrafollicular and follicular (PD-1hi) regulatory CD4 T cells. The lower local suppressing activity could represent one of the cellular mechanisms leading to the relative expansion of TFH cells we found in the vaccinated individuals. Many of the FOXP3hi CD4 T cells were found positive for the transcription factor HELIOS, too, both inside and outside the follicle. Flow cytometry analysis showed that contrary to HELIOS, most of the follicular FOXP3hi CD4 T cells expressed a BCL6lo phenotype, implying that the origin or maturation of regulatory follicular T cells and TFH cells may differ (3). Further analysis of these potent suppressor CD4 T cells with respect to their in vivo function (i.e., production of IL-10) and follicular location is needed.

No significant differences were found between vaccinated and nonvaccinated donors for most of the main tonsillar B cell populations analyzed. Among the biological factors tested, a significantly higher expression per cell, judged by MFI, of the IL-4R on plasma cells from vaccinated donors was found, potentially providing survival signals. Among the circulating cytokines/chemokines tested, CXCL13 was induced after vaccination, in line with the increased frequency of tonsillar TFH cells found. Follicular dendritic cells (59, 60), TFH cells (4, 15), and potentially monocytes (61, 62) represent cellular sources of CXCL13 production. The direct correlation between circulating CXCL13 levels and GC reactivity was recently described (15). Potential cellular sources of CXCL13, located in lymphoid organs other than tonsils, could contribute to the elevated levels of CXCL13 found in our study. However, our imaging analysis revealed an altered distribution in vaccinated individuals with high levels of circulating CXCL13 compared with control subjects, with CXCL13 occupying a broader area of the follicle/GC indicating a possibly higher GC reactivity in these donors. Although a direct comparison between follicular and circulating CXCL13 was technically challenging, quantitative imaging analysis showed that higher frequencies of follicular T and B cells were positive for CXCL13 (presumably bound on CXCR5 that is highly expressed on these cells) indicating a higher in situ production of CXCL13 in vaccinated individuals. Importantly, a correlation was found between the frequency of tonsillar PD-1hiCD57lo TFH cells and the tonsil-derived influenza-specific ASCs. Despite the i.m. vaccine delivery, this finding is in line with recent data showing increased vaccine-specific ASCs in blood and tonsils from children vaccinated with an intranasal live attenuated influenza vaccine (63). We should mention, however, that our study cannot address directly whether these lymphocytes are activated in the tonsils or are retained from circulating influenza-specific ASCs that originated from the draining lymphoid tissue. Indeed, despite no history of seasonal influenza vaccine, high basal level of HI titers in the cohort studied suggest previous exposure to different influenza virus strains.

Our data demonstrate that vaccination impacts on the tonsillar relevant CD4 T cell subpopulations and their investigation could be used to shed light on the cellular and molecular mechanisms governing the development of Ag-specific B cell responses. Vaccine was delivered i.m. in the arm; therefore, the tonsil does not represent a classic draining LN site. Whether immunogen was able to drain the tonsils during the 9 d before their biopsy is not known. Our study cannot address directly whether the described dynamics is due to trafficking-redistribution of the relevant populations or their de novo, in situ expansion. Therefore, the driven force for the described tonsil immune dynamics needs further investigation. Given the anatomical location of tonsils, it is possible that low level contemporaneous environmental exposure to influenza virus could sensitize the tonsillar responses to immunogen. Alternatively, previous exposure to influenza, documented by the prevaccination titers, could result in the generation of memory follicular CD4 and B cells, residing in peripheral tissues (64), including tonsils. This is supported by a higher H1N1 seroprotection rate before vaccination compared with the other strains (Fig. 5B) and reinforced by the significant difference of H1N1-specific ASCs detected a few days postvaccination in the peripheral blood of vaccinees compared with controls (data not shown). Vaccination could lead to restimulation of tonsil resident memory TFH and memory B cells, trafficking of effector TFH from LNs, tissue redistribution of peripheral TFH (11), or in situ generation/expansion of new TFH and B cell responses. As is mentioned above, the capacity of primary effector TFH for egression into the bloodstream is compromised. Although the tissue memory TFH cells likely express a different molecular profile compared with effector TFH cells (64), whether this profile differs between primary effector and memory restimulated TFH cells is not known. The tonsil could represent a unique mucosal site for better characterization of primary and memory reactivated follicular immune cells with respect to their blood counterparts. Our data, however, point to coordinated cellular dynamics characterized by lower potential follicular suppressor cells concomitant with higher TFH cell numbers after vaccination, supporting the idea of locally developed immune dynamics, too.

The quantification of cell populations using imaging analysis of tissue-derived sections is subject to the so-called sampling error, where the representative value of data obtained from a single section is not certain. Moreover, tonsils are immune-activated tissues with abundant, mature GCs creating a noisy local environment for the detection of changes of local populations after vaccination. Obviously, the tissues investigated cannot come from longitudinal samples. We would like to highlight, however, the similar profiles obtained for the CD4 T cell populations under investigation by both flow cytometry analysis of tonsil-derived cells and the tissue imaging histocytometry analysis. Furthermore, our analysis of different biological parameters consistently points to a higher GC reactivity in tonsils from vaccinated individuals. Our data support the value of tissue quantitative imaging for studies aiming to understand local immune dynamics.

Understanding the dynamics of GC-immune populations will further inform on the mechanistic base of the development of Ag-specific B cell responses. This is of great importance not only for the designing of novel vaccines but also for our understanding of the humoral responses in viral infections and autoimmunity. Furthermore, this type of study could identify new biomarkers for vaccine monitoring. Tonsillectomy is routinely performed in the pediatric population (65). To validate tonsils as valuable secondary lymphoid organs in children could open the way to performing a new set of immunological studies in a population with its own biological characteristics. We propose that tonsils represent an anatomical site where such dynamics can be investigated, especially in children where access to lymph nodes is challenging.

We thank the members of the Tissue Analysis Core at the Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health for helpful discussions and suggestions and Dr. Margaret Beddall of the ImmunoTechnology Section (also at the Vaccine Research Center) for preparation of the in-house–conjugated Abs. We thank Giovanni De Vincentiis for surgical procedures and Rocco Roma for assistance in coordinating patient enrollment and sample collection. We are grateful to the patients and the patients’ families for being part of this study.

This work was supported by the Intramural Research Program of the Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health and by the Collaboration for AIDS Vaccine Discovery (Grant OP1032325) from the Bill and Melinda Gates Foundation (to R.A.K.). This study was also supported by the Bambino Gesù Children’s Hospital-Research Institute through the European Union–funded project Global Research in Pediatrics Network of Excellence (http://www.grip-network.org) and a 2016 annual research grant on the search of novel vaccine biomarkers in children with acquired immunodeficiencies.

The online version of this article contains supplemental material.

Abbreviations used in this article:

ASC

Ab-secreting cell

GC

germinal center

HI

hemagglutination inhibition

LN

lymph node

MFI

mean fluorescence intensity

TFH

follicular helper CD4 T

TFR

follicular regulatory CD4 T

TREG

regulatory CD4 T.

1
Crotty
,
S.
2011
.
Follicular helper CD4 T cells (TFH).
Annu. Rev. Immunol.
29
:
621
663
.
2
Butler
,
N. S.
,
D. I.
Kulu
.
2015
.
The regulation of T follicular helper responses during infection.
Curr. Opin. Immunol.
34
:
68
74
.
3
Vinuesa
,
C. G.
,
M. A.
Linterman
,
D.
Yu
,
I. C.
MacLennan
.
2016
.
Follicular helper T cells.
Annu. Rev. Immunol.
34
:
335
368
.
4
Petrovas
,
C.
,
T.
Yamamoto
,
M. Y.
Gerner
,
K. L.
Boswell
,
K.
Wloka
,
E. C.
Smith
,
D. R.
Ambrozak
,
N. G.
Sandler
,
K. J.
Timmer
,
X.
Sun
, et al
.
2012
.
CD4 T follicular helper cell dynamics during SIV infection.
J. Clin. Invest.
122
:
3281
3294
.
5
Ma
,
C. S.
,
S.
Suryani
,
D. T.
Avery
,
A.
Chan
,
R.
Nanan
,
B.
Santner-Nanan
,
E. K.
Deenick
,
S. G.
Tangye
.
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
.
6
Kim
,
C. H.
,
L. S.
Rott
,
I.
Clark-Lewis
,
D. J.
Campbell
,
L.
Wu
,
E. C.
Butcher
.
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
.
7
Chtanova
,
T.
,
S. G.
Tangye
,
R.
Newton
,
N.
Frank
,
M. R.
Hodge
,
M. S.
Rolph
,
C. R.
Mackay
.
2004
.
T follicular helper cells express a distinctive transcriptional profile, reflecting their role as non-Th1/Th2 effector cells that provide help for B cells.
J. Immunol.
173
:
68
78
.
8
Kim
,
C. H.
,
H. W.
Lim
,
J. R.
Kim
,
L.
Rott
,
P.
Hillsamer
,
E. C.
Butcher
.
2004
.
Unique gene expression program of human germinal center T helper cells.
Blood
104
:
1952
1960
.
9
Haynes
,
N. M.
,
C. D.
Allen
,
R.
Lesley
,
K. M.
Ansel
,
N.
Killeen
,
J. G.
Cyster
.
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
.
10
Chung
,
Y.
,
S.
Tanaka
,
F.
Chu
,
R. I.
Nurieva
,
G. J.
Martinez
,
S.
Rawal
,
Y. H.
Wang
,
H.
Lim
,
J. M.
Reynolds
,
X. H.
Zhou
, et al
.
2011
.
Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions.
Nat. Med.
17
:
983
988
.
11
Morita
,
R.
,
N.
Schmitt
,
S. E.
Bentebibel
,
R.
Ranganathan
,
L.
Bourdery
,
G.
Zurawski
,
E.
Foucat
,
M.
Dullaers
,
S.
Oh
,
N.
Sabzghabaei
, 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
.
12
Bentebibel
,
S. E.
,
S.
Lopez
,
G.
Obermoser
,
N.
Schmitt
,
C.
Mueller
,
C.
Harrod
,
E.
Flano
,
A.
Mejias
,
R. A.
Albrecht
,
D.
Blankenship
, et al
.
2013
.
Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination.
Sci. Transl. Med.
5
:
176ra32
.
13
He
,
J.
,
L. M.
Tsai
,
Y. A.
Leong
,
X.
Hu
,
C. S.
Ma
,
N.
Chevalier
,
X.
Sun
,
K.
Vandenberg
,
S.
Rockman
,
Y.
Ding
, 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
.
14
Boswell
,
K. L.
,
R.
Paris
,
E.
Boritz
,
D.
Ambrozak
,
T.
Yamamoto
,
S.
Darko
,
K.
Wloka
,
A.
Wheatley
,
S.
Narpala
,
A.
McDermott
, et al
.
2014
.
Loss of circulating CD4 T cells with B cell helper function during chronic HIV infection.
PLoS Pathog.
10
:
e1003853
.
15
Havenar-Daughton
,
C.
,
M.
Lindqvist
,
A.
Heit
,
J. E.
Wu
,
S. M.
Reiss
,
K.
Kendric
,
S.
Bélanger
,
S. P.
Kasturi
,
E.
Landais
,
R. S.
Akondy
, et al
IAVI Protocol C Principal Investigators
.
2016
.
CXCL13 is a plasma biomarker of germinal center activity.
Proc. Natl. Acad. Sci. USA
113
:
2702
2707
.
16
Perry
,
M.
,
A.
Whyte
.
1998
.
Immunology of the tonsils.
Immunol. Today
19
:
414
421
.
17
Petrovas
,
C.
,
S.
Ferrando-Martinez
,
M. Y.
Gerner
,
J. P.
Casazza
,
A.
Pegu
,
C.
Deleage
,
A.
Cooper
,
J.
Hataye
,
S.
Andrews
,
D.
Ambrozak
, et al
.
2017
.
Follicular CD8 T cells accumulate in HIV infection and can kill infected cells in vitro via bispecific antibodies.
Sci. Transl. Med.
9
:
eaag2285
.
18
Gerner
,
M. Y.
,
W.
Kastenmuller
,
I.
Ifrim
,
J.
Kabat
,
R. N.
Germain
.
2012
.
Histo-cytometry: a method for highly multiplex quantitative tissue imaging analysis applied to dendritic cell subset microanatomy in lymph nodes.
Immunity
37
:
364
376
.
19
Rinaldi
,
S.
,
P.
Zangari
,
N.
Cotugno
,
E. C.
Manno
,
N.
Brolatti
,
M. R.
Castrucci
,
I.
Donatelli
,
P.
Rossi
,
P.
Palma
,
A.
Cagigi
.
2014
.
Antibody but not memory B-cell responses are tuned-down in vertically HIV-1 infected children and young individuals being vaccinated yearly against influenza.
Vaccine
32
:
657
663
.
20
Kim
,
J. R.
,
H. W.
Lim
,
S. G.
Kang
,
P.
Hillsamer
,
C. H.
Kim
.
2005
.
Human CD57+ germinal center-T cells are the major helpers for GC-B cells and induce class switch recombination.
BMC Immunol.
6
:
3
.
21
Weber
,
J. P.
,
F.
Fuhrmann
,
R. K.
Feist
,
A.
Lahmann
,
M. S.
Al Baz
,
L. J.
Gentz
,
D.
Vu Van
,
H. W.
Mages
,
C.
Haftmann
,
R.
Riedel
, et al
.
2015
.
ICOS maintains the T follicular helper cell phenotype by down-regulating Krüppel-like factor 2.
J. Exp. Med.
212
:
217
233
.
22
Jacquemin
,
C.
,
N.
Schmitt
,
C.
Contin-Bordes
,
Y.
Liu
,
P.
Narayanan
,
J.
Seneschal
,
T.
Maurouard
,
D.
Dougall
,
E. S.
Davizon
,
H.
Dumortier
, et al
.
2015
.
OX40 ligand contributes to human lupus pathogenesis by promoting T follicular helper response.
Immunity
42
:
1159
1170
.
23
Cannons
,
J. L.
,
H.
Qi
,
K. T.
Lu
,
M.
Dutta
,
J.
Gomez-Rodriguez
,
J.
Cheng
,
E. K.
Wakeland
,
R. N.
Germain
,
P. L.
Schwartzberg
.
2010
.
Optimal germinal center responses require a multistage T cell:B cell adhesion process involving integrins, SLAM-associated protein, and CD84.
Immunity
32
:
253
265
.
24
Fields
,
M. L.
,
B. D.
Hondowicz
,
M. H.
Metzgar
,
S. A.
Nish
,
G. N.
Wharton
,
C. C.
Picca
,
A. J.
Caton
,
J.
Erikson
.
2005
.
CD4+ CD25+ regulatory T cells inhibit the maturation but not the initiation of an autoantibody response.
J. Immunol.
175
:
4255
4264
.
25
Linterman
,
M. A.
,
W.
Pierson
,
S. K.
Lee
,
A.
Kallies
,
S.
Kawamoto
,
T. F.
Rayner
,
M.
Srivastava
,
D. P.
Divekar
,
L.
Beaton
,
J. J.
Hogan
, et al
.
2011
.
Foxp3+ follicular regulatory T cells control the germinal center response.
Nat. Med.
17
:
975
982
.
26
Himmel
,
M. E.
,
K. G.
MacDonald
,
R. V.
Garcia
,
T. S.
Steiner
,
M. K.
Levings
.
2013
.
Helios+ and Helios- cells coexist within the natural FOXP3+ T regulatory cell subset in humans.
J. Immunol.
190
:
2001
2008
.
27
Fu
,
W.
,
A.
Ergun
,
T.
Lu
,
J. A.
Hill
,
S.
Haxhinasto
,
M. S.
Fassett
,
R.
Gazit
,
S.
Adoro
,
L.
Glimcher
,
S.
Chan
, et al
.
2012
.
A multiply redundant genetic switch ‘locks in’ the transcriptional signature of regulatory T cells.
Nat. Immunol.
13
:
972
980
.
28
Vinuesa
,
C. G.
,
M. A.
Linterman
,
C. C.
Goodnow
,
K. L.
Randall
.
2010
.
T cells and follicular dendritic cells in germinal center B-cell formation and selection.
Immunol. Rev.
237
:
72
89
.
29
Ellebedy
,
A. H.
,
K. J.
Jackson
,
H. T.
Kissick
,
H. I.
Nakaya
,
C. W.
Davis
,
K. M.
Roskin
,
A. K.
McElroy
,
C. M.
Oshansky
,
R.
Elbein
,
S.
Thomas
, 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
.
30
Bajénoff
,
M.
,
J. G.
Egen
,
L. Y.
Koo
,
J. P.
Laugier
,
F.
Brau
,
N.
Glaichenhaus
,
R. N.
Germain
.
2006
.
Stromal cell networks regulate lymphocyte entry, migration, and territoriality in lymph nodes.
Immunity
25
:
989
1001
.
31
Qi
,
H.
,
W.
Kastenmüller
,
R. N.
Germain
.
2014
.
Spatiotemporal basis of innate and adaptive immunity in secondary lymphoid tissue.
Annu. Rev. Cell Dev. Biol.
30
:
141
167
.
32
Bouzahzah
,
F.
,
A.
Bosseloir
,
E.
Heinen
,
L. J.
Simar
.
1995
.
Human germinal center CD4+CD57+ T cells act differently on B cells than do classical T-helper cells.
Dev. Immunol.
4
:
189
197
.
33
Rasheed
,
A. U.
,
H. P.
Rahn
,
F.
Sallusto
,
M.
Lipp
,
G.
Müller
.
2006
.
Follicular B helper T cell activity is confined to CXCR5(hi)ICOS(hi) CD4 T cells and is independent of CD57 expression.
Eur. J. Immunol.
36
:
1892
1903
.
34
Wallin
,
E. F.
,
E. C.
Jolly
,
O.
Suchánek
,
J. A.
Bradley
,
M.
Espéli
,
D. R.
Jayne
,
M. A.
Linterman
,
K. G.
Smith
.
2014
.
Human T-follicular helper and T-follicular regulatory cell maintenance is independent of germinal centers.
Blood
124
:
2666
2674
.
35
Ballesteros-Tato
,
A.
,
T. D.
Randall
.
2014
.
Priming of T follicular helper cells by dendritic cells.
Immunol. Cell Biol.
92
:
22
27
.
36
Estes
,
J. D.
,
T. C.
Thacker
,
D. L.
Hampton
,
S. A.
Kell
,
B. F.
Keele
,
E. A.
Palenske
,
K. M.
Druey
,
G. F.
Burton
.
2004
.
Follicular dendritic cell regulation of CXCR4-mediated germinal center CD4 T cell migration.
J. Immunol.
173
:
6169
6178
.
37
Elsner
,
R. A.
,
D. N.
Ernst
,
N.
Baumgarth
.
2012
.
Single and coexpression of CXCR4 and CXCR5 identifies CD4 T helper cells in distinct lymph node niches during influenza virus infection.
J. Virol.
86
:
7146
7157
.
38
Rodda
,
L. B.
,
O.
Bannard
,
B.
Ludewig
,
T.
Nagasawa
,
J. G.
Cyster
.
2015
.
Phenotypic and Morphological properties of germinal center dark zone Cxcl12-expressing reticular cells.
J. Immunol.
195
:
4781
4791
.
39
Ioannidis
,
L. J.
,
C. Q.
Nie
,
A.
Ly
,
V.
Ryg-Cornejo
,
C. Y.
Chiu
,
D. S.
Hansen
.
2016
.
Monocyte- and neutrophil-derived CXCL10 impairs efficient control of blood-stage malaria infection and promotes severe disease.
J. Immunol.
196
:
1227
1238
.
40
Rodriguez-Pla
,
A.
,
P.
Patel
,
H. T.
Maecker
,
J.
Rossello-Urgell
,
N.
Baldwin
,
L.
Bennett
,
V.
Cantrell
,
J.
Baisch
,
M.
Punaro
,
A.
Gotte
, et al
.
2014
.
IFN priming is necessary but not sufficient to turn on a migratory dendritic cell program in lupus monocytes.
J. Immunol.
192
:
5586
5598
.
41
Groom
,
J. R.
,
J.
Richmond
,
T. T.
Murooka
,
E. W.
Sorensen
,
J. H.
Sung
,
K.
Bankert
,
U. H.
von Andrian
,
J. J.
Moon
,
T. R.
Mempel
,
A. D.
Luster
.
2012
.
CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T helper 1 cell differentiation.
Immunity
37
:
1091
1103
.
42
Spiegel
,
S.
,
S.
Milstien
.
2011
.
The outs and the ins of sphingosine-1-phosphate in immunity.
Nat. Rev. Immunol.
11
:
403
415
.
43
Bankovich
,
A. J.
,
L. R.
Shiow
,
J. G.
Cyster
.
2010
.
CD69 suppresses sphingosine 1-phosophate receptor-1 (S1P1) function through interaction with membrane helix 4.
J. Biol. Chem.
285
:
22328
22337
.
44
Blankenbach
,
K. V.
,
S.
Schwalm
,
J.
Pfeilschifter
,
D.
Meyer Zu Heringdorf
.
2016
.
Sphingosine-1-phosphate receptor-2 antagonists: therapeutic potential and potential risks.
Front. Pharmacol.
7
:
167
.
45
Moriyama
,
S.
,
N.
Takahashi
,
J. A.
Green
,
S.
Hori
,
M.
Kubo
,
J. G.
Cyster
,
T.
Okada
.
2014
.
Sphingosine-1-phosphate receptor 2 is critical for follicular helper T cell retention in germinal centers.
J. Exp. Med.
211
:
1297
1305
.
46
Diehl
,
S. A.
,
H.
Schmidlin
,
M.
Nagasawa
,
S. D.
van Haren
,
M. J.
Kwakkenbos
,
E.
Yasuda
,
T.
Beaumont
,
F. A.
Scheeren
,
H.
Spits
.
2008
.
STAT3-mediated up-regulation of BLIMP1 is coordinated with BCL6 down-regulation to control human plasma cell differentiation.
J. Immunol.
180
:
4805
4815
.
47
Wu
,
H.
,
L. L.
Xu
,
P.
Teuscher
,
H.
Liu
,
M. H.
Kaplan
,
A. L.
Dent
.
2015
.
An inhibitory role for the transcription factor Stat3 in controlling IL-4 and Bcl6 expression in follicular helper T cells.
J. Immunol.
195
:
2080
2089
.
48
Kroenke
,
M. A.
,
D.
Eto
,
M.
Locci
,
M.
Cho
,
T.
Davidson
,
E. K.
Haddad
,
S.
Crotty
.
2012
.
Bcl6 and Maf cooperate to instruct human follicular helper CD4 T cell differentiation.
J. Immunol.
188
:
3734
3744
.
49
Lee
,
J. Y.
,
C. N.
Skon
,
Y. J.
Lee
,
S.
Oh
,
J. J.
Taylor
,
D.
Malhotra
,
M. K.
Jenkins
,
M. G.
Rosenfeld
,
K. A.
Hogquist
,
S. C.
Jameson
.
2015
.
The transcription factor KLF2 restrains CD4+ T follicular helper cell differentiation.
Immunity
42
:
252
264
.
50
Suan
,
D.
,
A.
Nguyen
,
I.
Moran
,
K.
Bourne
,
J. R.
Hermes
,
M.
Arshi
,
H. R.
Hampton
,
M.
Tomura
,
Y.
Miwa
,
A. D.
Kelleher
, et al
.
2015
.
T follicular helper cells have distinct modes of migration and molecular signatures in naive and memory immune responses.
Immunity
42
:
704
718
.
51
Baumjohann
,
D.
,
S.
Preite
,
A.
Reboldi
,
F.
Ronchi
,
K. M.
Ansel
,
A.
Lanzavecchia
,
F.
Sallusto
.
2013
.
Persistent antigen and germinal center B cells sustain T follicular helper cell responses and phenotype.
Immunity
38
:
596
605
.
52
Deenick
,
E. K.
,
A.
Chan
,
C. S.
Ma
,
D.
Gatto
,
P. L.
Schwartzberg
,
R.
Brink
,
S. G.
Tangye
.
2010
.
Follicular helper T cell differentiation requires continuous antigen presentation that is independent of unique B cell signaling.
Immunity
33
:
241
253
.
53
Schwartzberg
,
P. L.
,
K. L.
Mueller
,
H.
Qi
,
J. L.
Cannons
.
2009
.
SLAM receptors and SAP influence lymphocyte interactions, development and function.
Nat. Rev. Immunol.
9
:
39
46
.
54
Chevrier
,
S.
,
T.
Kratina
,
D.
Emslie
,
D. M.
Tarlinton
,
L. M.
Corcoran
.
2017
.
IL4 and IL21 cooperate to induce the high Bcl6 protein level required for germinal center formation.
Immunol. Cell Biol.
DOI: 10.1038/icb.2017.71.
55
Vaeth
,
M.
,
G.
Müller
,
D.
Stauss
,
L.
Dietz
,
S.
Klein-Hessling
,
E.
Serfling
,
M.
Lipp
,
I.
Berberich
,
F.
Berberich-Siebelt
.
2014
.
Follicular regulatory T cells control humoral autoimmunity via NFAT2-regulated CXCR5 expression.
J. Exp. Med.
211
:
545
561
.
56
Ichimiya
,
S.
,
R.
Kamekura
,
K.
Kawata
,
M.
Kamei
,
T.
Himi
.
2017
.
Functional RNAs control T follicular helper cells.
J. Hum. Genet.
62
:
81
86
.
57
Wing
,
J. B.
,
Y.
Kitagawa
,
M.
Locci
,
H.
Hume
,
C.
Tay
,
T.
Morita
,
Y.
Kidani
,
K.
Matsuda
,
T.
Inoue
,
T.
Kurosaki
, et al
.
2017
.
A distinct subpopulation of CD25(-) T-follicular regulatory cells localizes in the germinal centers.
Proc. Natl. Acad. Sci. USA
114
:
E6400
E6409
.
58
Panduro
,
M.
,
C.
Benoist
,
D.
Mathis
.
2016
.
Tissue Tregs.
Annu. Rev. Immunol.
34
:
609
633
.
59
Wang
,
X.
,
B.
Cho
,
K.
Suzuki
,
Y.
Xu
,
J. A.
Green
,
J.
An
,
J. G.
Cyster
.
2011
.
Follicular dendritic cells help establish follicle identity and promote B cell retention in germinal centers.
J. Exp. Med.
208
:
2497
2510
.
60
Vermi
,
W.
,
S.
Lonardi
,
D.
Bosisio
,
M.
Uguccioni
,
G.
Danelon
,
S.
Pileri
,
C.
Fletcher
,
S.
Sozzani
,
F.
Zorzi
,
G.
Arrigoni
, et al
.
2008
.
Identification of CXCL13 as a new marker for follicular dendritic cell sarcoma.
J. Pathol.
216
:
356
364
.
61
Cohen
,
K. W.
,
A. S.
Dugast
,
G.
Alter
,
M. J.
McElrath
,
L.
Stamatatos
.
2015
.
HIV-1 single-stranded RNA induces CXCL13 secretion in human monocytes via TLR7 activation and plasmacytoid dendritic cell-derived type I IFN.
J. Immunol.
194
:
2769
2775
.
62
Carlsen
,
H. S.
,
E. S.
Baekkevold
,
H. C.
Morton
,
G.
Haraldsen
,
P.
Brandtzaeg
.
2004
.
Monocyte-like and mature macrophages produce CXCL13 (B cell-attracting chemokine 1) in inflammatory lesions with lymphoid neogenesis.
Blood
104
:
3021
3027
.
63
Mohn
,
K. G.
,
K. A.
Brokstad
,
R. D.
Pathirana
,
G.
Bredholt
,
Å.
Jul-Larsen
,
M. C.
Trieu
,
S. L.
Lartey
,
E.
Montemoli
,
C.
Tøndel
,
H. J.
Aarstad
,
R. J.
Cox
.
2016
.
Live attenuated influenza vaccine in children induces B-cell responses in tonsils.
J. Infect. Dis.
214
:
722
731
.
64
Hale
,
J. S.
,
B.
Youngblood
,
D. R.
Latner
,
A. U.
Mohammed
,
L.
Ye
,
R. S.
Akondy
,
T.
Wu
,
S. S.
Iyer
,
R.
Ahmed
.
2013
.
Distinct memory CD4+ T cells with commitment to T follicular helper- and T helper 1-cell lineages are generated after acute viral infection.
Immunity
38
:
805
817
.
65
Mahadevan
,
M.
,
G.
van der Meer
,
M.
Gruber
,
P.
Reed
,
C.
Jackson
,
C.
Brown
,
N.
Mills
,
L. J.
Salkeld
,
M.
Neeff
,
J.
Evans
, et al
.
2016
.
The starship children’s hospital tonsillectomy: a further 10 years of experience.
Laryngoscope
126
:
E416
E420
.

The authors have no financial conflicts of interest.

Supplementary data