Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2011 Aug 1;108(33):E488–E497. doi: 10.1073/pnas.1100898108

Human tonsil B-cell lymphoma 6 (BCL6)-expressing CD4+ T-cell subset specialized for B-cell help outside germinal centers

Salah-Eddine Bentebibel a,b, Nathalie Schmitt a, Jacques Banchereau a,c,1, Hideki Ueno a,c,2
PMCID: PMC3158181  PMID: 21808017

Abstract

T follicular helper (Tfh) cells represent a Th subset engaged in the help of B-cell responses in germinal centers (GCs). Tfh cells abundantly express the transcription repressor B-cell lymphoma 6 (Bcl6), a factor that is necessary and sufficient for their development in vivo. Whether Tfh or Tfh-committed cells are involved in the help of B cells outside GCs remains unclear, particularly in humans. In this study, we identified a previously undefined BCL6-expressing CD4+ T-cell subset in human tonsils. This subset expressed IL-7 receptor and chemokine receptor 5 (CXCR5) and inducible costimulator (ICOS) at low levels (CXCR5loICOSlo), and it was found exclusively outside GCs. CXCR5loICOSlo CD4+ T cells secreted larger amounts of IL-21 and IL-10 than CXCR5hiICOShi GC-Tfh cells upon activation, and they induced proliferation and differentiation of naïve B cells into Ig-producing cells more efficiently than GC-Tfh cells. However, this subset lacked the capacity to help GC-B cells because of the induction of apoptosis of GC-B cells through the FAS/FAS–ligand interaction. CXCR5loICOSlo CD4+ T cells expressed equivalent amounts of BCL6 transcript with CXCR5hiICOShi GC-Tfh cells, but they expressed less Bcl6 protein. Collectively, our study indicates that CXCR5loICOSlo CD4+ T cells in human tonsils represent Tfh lineage-committed cells that provide help to naïve and memory B cells outside GCs.


CD4+ T cells provide a critical help to B cells to induce antibody responses. CD4+ T cells primed by dendritic cells (DCs) loaded with antigens interact with antigen-primed naïve B cells at extrafollicular sites (1), typically at the border of the T-cell zone and primary B-cell follicles (2). This interaction initiates the B-cell differentiation process to two different paths: extrafollicular plasma cells and cells forming germinal centers (GCs) (35). Extrafollicular plasma cells contribute to the early generation of specific antibodies after antigen challenge (6). GC-B cells subsequently differentiate into either high-affinity long-lived plasma cells or memory B cells after an extensive selection step (7, 8), and thus, they are central in the generation of B-cell memory.

T follicular helper (Tfh) cells have been recently established as a Th subset specialized for the help of B cells in GCs (9, 10). Whereas no specific markers are available to distinguish Tfh cells from other canonical Th subsets such as Th1, Th2, and Th17 cells, several molecules associated with the biological properties of Tfh cells are used for their identification. The chemokine (CXC) receptor 5 (CXCR5) expressed by Tfh cells (1114) guides their migration into B-cell follicles in response to the ligand CXCL13 secreted by follicular dendritic cells (15). Tfh cells express programmed death-1 (PD-1), which was shown to play a role in the selection of high-affinity B cells in GCs (16). Inducible costimulator (ICOS) is also expressed by Tfh cells in human tonsils at high density (14). ICOS is critical for the development (17, 18) and functions (19, 20) of Tfh cells in both humans and mice. Studies in mice showed that overexpression of ICOS leads to the substantial increase of GC formation associated with exaggerated Tfh responses (21), which directly induce autoimmunity (22). Tfh cells also express other molecules required for interactions with B cells, including CD40 ligand (CD40L), CD84 (23), and signaling lymphocyte activation molecule-associated protein (9). Compared with other canonical Th subsets, Tfh cells secrete larger amounts of IL-21 (24, 25), a γc-family cytokine that strongly promotes the growth, differentiation, and class switching of B cells (2631). Tfh cells express large amounts of the transcription repressor B-cell lymphoma 6 (Bcl6) (25). Mouse studies indicate that Bcl6 positively regulates the generation of Tfh cells in vivo (3234). In contrast, B lymphocyte-induced maturation protein 1 (Blimp-1), a transcription repressor that inhibits the function of Bcl6, negatively regulates Tfh development (34). Thus, the development of Tfh cells is regulated by the balance of these two molecules.

Whereas Tfh cells are considered to help the selection and differentiation of B cells in GCs, the identity of CD4+ T cells interacting with B cells outside GCs remains largely unknown. Studies with Murphy Roths Large (MRL)/FASlpr lupus-prone mouse models showed the presence of extrafollicular CD4+ T cells that display a similar phenotype as Tfh cells, such as up-regulation of Bcl6 and IL-21 and down-regulation of P-selectin glycoprotein ligand 1 (35, 36). Whereas these studies show that extrafollicular helper cells share properties with Tfh cells under certain conditions in mice, it is unknown whether this finding holds true in humans.

Here, we identified a previously undefined BCL6-expressing CD4+ T-cell subset in human tonsils. This subset was identified by the expression of IL-7 receptor (IL-7R) and low levels of CXCR5 and ICOS (CXCR5loICOSlo), and it was exclusively localized outside GCs. CXCR5loICOSlo CD4+ T cells secreted large amounts of IL-21 and IL-10 and efficiently helped naïve and memory B cells. However, they lacked the capacity to help GC-B cells and induced the apoptosis of GC-B cells through the FAS/FAS–ligand (FAS-L) interaction. Thus, this tonsillar CD4+ T-cell subset is specialized to help B cells outside GCs.

Results

CXCR5loICOSlo CD4+ T Cells Are Localized Outside GC.

Previous studies showed that human tonsillar Tfh cells in GCs coexpress ICOS and CXCR5 at high densities (14). In our study, we obtained pediatric tonsil samples (from children ages 3–12 y), which contained 34.8 ± 0.9% of CXCR5hiICOShi GC-Tfh cells among tonsillar CD4+ T cells (Fig. 1 A and B, blue gate) (mean ± SEM, n = 20). Tonsillar CD4+ T cells included at least three other populations defined according to the expression of CXCR5 and ICOS: CXCR5ICOS (black gate; 21.4 ± 1.1%), CXCR5loICOSlo (red gate; 19.5 ± 0.9%), and CXCR5loICOShi (green gate; 10.4 ± 0.5%) cells. As observed earlier (37, 38), a majority of CXCR5hiICOShi GC-Tfh cells expressed PD-1 (Fig. 1C) (84 ± 2%, mean ± SEM, n = 3). CXCR5loICOShi CD4+ T cells also expressed PD-1, although at lower density than CXCR5hiICOShi GC-Tfh cells (41 ± 1%). Only a minor fraction of CXCR5loICOSlo CD4+ T cells expressed PD-1 (8 ± 3%) at very low density. CD57, a molecule expressed by a fraction of human GC-Tfh cells (13), was expressed at high density by a fraction of CXCR5hiICOShi GC-Tfh cells (Fig. 1C) (35 ± 3%), whereas CXCR5loICOShi CD4+ T cells and CXCR5loICOSlo CD4+ T cells contained less CD57-expressing cells (Fig. 1C) (10 ± 1% and 5 ± 1%, respectively). CXCR5ICOS CD4+ T cells did not express PD-1, CD57, or CD45RO, and thus, they were mostly composed of naïve CD4+ T cells. As reported previously (38, 39), a large fraction of CXCR5hiICOShi GC-Tfh cells did not express IL-7R (CD127) (Fig. 1C). In contrast, CXCR5loICOSlo CD4+ T cells expressed IL-7R as much as CXCR5ICOS naïve CD4+ T cells, whereas CXCR5loICOShi CD4+ T cells expressed IL-7R only at low density. A previous study reported the absence of IL-7R–expressing cells in GCs (39). We confirmed this observation by immunohistochemistry (Fig. 2A). Whereas both CXCR5ICOS and CXCR5loICOSlo CD4+ T cells expressed IL-7R, CXCR5loICOSlo CD4+ T cells coexpressed CD45RO (Fig. 1C). Thus, the localization of CXCR5loICOSlo CD4+ T cells was determined by analyzing IL-7R+CD45RO+CD4+ T cells by confocal microscopy. As shown in Fig. 2 B and C, the cells coexpressing the three molecules (green, CD45RO; red, CD4; blue, IL-7R; white, triple positive cells) were exclusively localized outside GCs. Although we cannot exclude the possibility that the identified IL-7R+CD45RO+CD4+ T cells might contain CXCR5loICOShi CD4+ T cells, our observation shows that CXCR5loICOSlo CD4+ T cells are exclusively localized outside but not within GCs.

Fig. 1.

Fig. 1.

Four Th populations in pediatric tonsil samples. (A) Four tonsillar Th populations were defined according to the expression of ICOS and CXCR5. (B) The frequency of each Th population (n = 20). One-way ANOVA Bonferonni multiple comparison test. ***P < 0.001. (C) Expression of cell surface molecules (a representative from three experiments).

Fig. 2.

Fig. 2.

CXCR5loICOSlo cells localize exclusively outside GCs. (A) Localization of IL-7R+ cells in tonsils was analyzed by immunohistochemistry using a frozen tonsil section. FM, follicular mantle. (Magnification: 4×.) (B and C) Localization of CXCR5loICOSlo CD4+ T cells was analyzed by the coexpression of CD4 (red), CD45RO (green), and IL-7R (blue) by immunofluorescence microscopy (10×). Triple positive cells illustrated by the white color include CXCR5loICOSlo CD4+ T cells. A higher magnification view (40×) is shown in C (analyzed by confocal microscopy).

Tonsillar Th Populations Differentially Help B-Cell Subsets.

We next assessed whether the four tonsillar Th populations were able to help the three major tonsillar B-cell subsets: naïve B (IgD+CD38CD19+) cells, memory B (IgDCD38CD19+) cells, and GC-B (IgDCD38+CD19+) cells (Fig. S1A). The tonsillar Th populations were carefully isolated by gating out cells expressing CD20, CD8, and CD56 during the sort. This strategy was particularly important for the isolation of CXCR5hiICOShi GC-Tfh cells, because 3–5% of CXCR5hiICOShi GC-Tfh cells coexpressed CD20 (Fig. S1B), likely representing conjugates of GC-Tfh and GC-B cells (40). The four tonsillar Th populations (2 × 104/well) were cultured with each autologous B-cell subset (2 × 104/well), and secreted Igs were measured at day 8 (Fig. 3 AC). To mimic the antigen-specific cognate interactions between B and T cells, staphylococcal enterotoxin B (SEB), a superantigen, was added to the cocultures. Previous studies showed that CXCR5hiICOShi GC-Tfh cells efficiently help tonsillar B cells compared with other tonsillar Th populations (14, 24, 41). Consistently, CXCR5hiICOShi GC-Tfh cells (blue symbols) efficiently promoted GC-B cells to produce Igs (Fig. 3A) (IgM, 1.3 ± 0.4 μg/mL; IgG, 1.0 ± 0.2 μg/mL; IgA, 0.3 ± 0.1 μg/mL; mean ± SEM, n = 11). Other tonsillar Th populations, CXCR5ICOS cells (gray symbols), CXCR5loICOSlo cells (red symbols), and CXCR5loICOShi cells (green symbols) barely induced GC-B cells to produce Igs (Fig. 3A). Indeed, only CXCR5hiICOShi GC-Tfh cells were able to maintain the survival of GC-B cells during the 8-d coculture (Fig. 3 D and E), some of which expressed surface Igs (Fig. 3D Right). Notably, whereas GC-Tfh cells are prone to apoptosis because of expression of FAS (11, 14, 42), GC-B cells were able to maintain the survival of CXCR5hiICOShi GC-Tfh cells and induce their proliferation (Fig. 3F) (142 ± 14 × 103 cells/well, mean ± SEM, n = 5; 7.1 ± 0.7-fold increase from the input). Thus, GC-Tfh and GC-B cells reciprocally help each other.

Fig. 3.

Fig. 3.

Tonsillar Th populations differentially help B-cell subsets. (A–C) Ig production by B-cell subsets cocultured with tonsillar Th populations. (A) GC-B cells (n = 11 except CXCR5loICOShi, which is n = 3), (B) naïve B cells (n = 4), and (C) memory B cells (n = 6 except CXCR5loICOShi, which is n = 3) were cocultured with each Th population in the presence of SEB for 8 d. One-way ANOVA Bonferonni multiple comparison test. ***P < 0.001, **P < 0.01, and *P < 0.05. (D and E) Recovery of GC-B cells cocultured with Th populations. GC-B cells cultured for 8 d with each Th population were stained with CD3 and CD4 mAbs. Surface IgG and IgA expression by B cells cultured with CXCR5hiICOShi GC-Tfh cells is shown in Right (a representative from four experiments). Absolute number of viable GC-B cells per well (day 8) are shown in E (n = 5). One-way ANOVA Bonferonni multiple comparison test. (F) Recovery of T cells cocultured with GC-B cells. Absolute number of viable T cells per well (day 8; n = 5). One-way ANOVA Bonferonni multiple comparison test. (G) Proliferation and CD38 expression of CFSE-labeled naïve B cells cultured for 8 d with each Th population (a representative from three experiments). (H) Recovery of naïve B cells cocultured with Th populations. Absolute number of viable CD38+ plasmablasts per well (day 8; n = 3). One-way ANOVA Bonferonni multiple comparison test. (I) Recovery of T cells cocultured with naïve B cells. Absolute number of viable T cells per well (day 8; n = 3). One-way ANOVA Bonferonni multiple comparison test.

In contrast, upon coculture with naïve B cells, CXCR5loICOSlo CD4+ T cells were the most efficient at inducing naïve B cells to produce IgM (Fig. 3B) (33.0 ± 4.9 μg/mL, mean ± SEM, n = 4) as well as IgG and IgA (IgG, 1.1 ± 0.2 μg/mL; IgA, 0.8 ± 0.2 μg/mL). This finding was not due to a contamination of CD27+ antigen-experienced subepithelial B cells (43) (3–7% in IgD+CD38CD19+ B cells), because CXCR5loICOSlo CD4+ T cells efficiently induced IgD+CD27CD38 highly purified naïve B cells to produce Igs (Fig. S1C). Furthermore, CXCR5loICOSlo CD4+ T cells induced a robust proliferation of naïve B cells (Fig. 3G) and yielded more B cells expressing CD38, a marker of plasmablasts, than other tonsillar Th populations (Fig. 3 G and H). CXCR5hiICOShi GC-Tfh cells were capable of inducing naïve B cells to produce Igs but only at low amounts (Fig. 3B) (IgM, 1.6 ± 0.5 μg/mL; IgG, 0.2 ± 0.1 μg/mL; IgA, 0.1 ± 0.1 μg/mL; mean ± SEM, n = 4). This finding might partly be due to the poor survival of CXCR5hiICOShi GC-Tfh cells, because naïve B cells did not support their survival and/or proliferation in culture (Fig. 3I) (29 ± 8 × 103 cells/well, mean ± SEM, n = 3; 1.5 ± 0.4-fold increase from the input). CXCR5ICOS cells and CXCR5loICOShi CD4+ T cells were virtually unable to induce naïve B cells to produce Igs (Fig. 3B).

Upon coculture with memory B cells, both CXCR5loICOSlo CD4+ T and CXCR5hiICOShi GC-Tfh cells were equally efficient at inducing their Ig production (Fig. 3C). CXCR5ICOS cells and CXCR5loICOShi CD4+ T cells barely induced memory B cells to produce Igs.

Thus, tonsillar Th populations differentially help B-cell subsets. CXCR5ICOS cells and CXCR5loICOShi CD4+ T cells barely help any B-cell subsets. Whereas CXCR5hiICOShi GC-Tfh cells efficiently help GC-B cells, CXCR5loICOSlo CD4+ T cells are the most potent at inducing naïve B cells to differentiate into Ig-producing cells.

Tonsillar Th Populations Differentially Secrete Cytokines/Chemokines.

Tfh cells secrete IL-4, IL-10, and IL-21, each of which differentially regulates B-cell proliferation, differentiation, and class switching (9, 10). Thus, we analyzed whether the cytokine production profiles of these tonsillar Th populations explain their differential effects on B-cell subsets. Upon coculture with naïve B cells, CXCR5loICOSlo CD4+ T cells (Fig. 4A, red symbols in IL-21 plot) secreted the largest amounts of IL-21 (3.3 ± 0.4 ng/mL, mean ± SEM, n = 6). This finding held true when tonsillar Th populations were cultured with GC-B cells (Fig. 4B, IL-21 plot) (1.8 ± 0.4 ng/mL, n = 5), or stimulated through CD3/CD28 (Fig. S2A) (6.1 ± 0.6 ng/mL, n = 3). CXCR5hiICOShi GC-Tfh cells also secreted IL-21 but at substantially lower amounts (Fig. 4 A and B and Fig. S2A) (culture with naïve B cells: 0.5 ± 0.2 ng/mL, n = 6; culture with GC-B cells: 0.4 ± 0.2 ng/mL, n = 5; CD3/CD28 stimulation: 0.7 ± 0.1 ng/mL, n = 3). The largest amounts of IL-10 were also detected in the cultures of CXCR5loICOSlo CD4+ T cells with B cells (Fig. 4 A and B) (with naïve B cells: 1.1 ± 0.2 ng/mL, n = 6; with GC-B cells: 1.4 ± 0.4 ng/mL, n = 5) as well as upon CD3/CD28 stimulation (Fig. S2A). In contrast, consistent with previous studies (14, 25, 44), CXCR5hiICOShi GC-Tfh cells produced the largest amounts of CXCL13, a chemokine secreted by Tfh cells upon culture with B cells (Fig. 4C) (with naïve B cells: 2.7 ± 0.3 ng/mL, n = 4; with GC-B cells: 2.5 ± 0.2 ng/mL, n = 3). IL-4 was detected only at low concentrations (<0.3 ng/mL) in any Th cultures with B cells (Fig. 4 A and B, IL-4 plot), although large amounts of IL-4 were secreted by CXCR5loICOSlo CD4+ T cells and CXCR5hiICOShi GC-Tfh cells upon CD3/CD28 stimulation (Fig. S2A). CXCR5loICOShi CD4+ T cells (green symbols) barely secreted IL-21 and IL-10 upon encounter with B cells (Fig. 4 A and B) (with naïve B cells: 0.2 ± 0.0 ng/mL and 0.2 ± 0.0 ng/mL, respectively, n = 6; with GC-B cells: 0.4 ± 0.2 ng/mL and 0.2 ± 0.0 ng/mL, respectively, n = 5) but secreted the largest amounts of IL-17A (with naïve B cells: 4.1 ± 1.3 ng/mL; with GC-B cells: 4.3 ± 1.6 ng/mL).

Fig. 4.

Fig. 4.

Cytokine secretion profiles of tonsillar Th populations. (A and B) Cytokine secretion on interaction with B-cell subsets. The four Th populations were cultured with either naïve B cells (A; n = 6) or GC-B cells (B; n = 5), and the secretions of IL-10, IL-21, IL-17A, and IL-4 were analyzed on day 2. One-way ANOVA Bonferonni multiple comparison test. ***P < 0.001, **P < 0.01, and *P < 0.05. (C) CXCL13 secretion on interaction with naïve B cells (Upper; n = 4) or GC-B cells (Lower; n = 3). One-way ANOVA Bonferonni multiple comparison test. (D) Kinetics of IL21 and CXCL13 expression. The four Th populations were analyzed for the expression of IL21 and CXCL13 by real-time RT-PCR before and after stimulation with CD3/CD28 mAb-coated beads (n = 2–3). One-way ANOVA Bonferonni multiple comparison test for n = 3 and paired t test for n = 2.

Secretion of the largest amounts of IL-21 by CXCR5loICOSlo CD4+ T cells was somewhat unexpected, because a number of previous studies showed that GC-Tfh cells expressed the highest levels of IL-21 among CD4+ T-cell subsets (14, 24, 25, 32, 38). These conclusions were based on the analysis of IL21 transcript by mRNA microarray or RT-PCR (14, 25, 38) or on the analysis of intracytoplasmic IL-21 expression after phorbol 12-myristate 13-acetate (PMA) and ionomycin stimulation (32, 38). Consistently, when isolated, CXCR5hiICOShi GC-Tfh cells expressed the most abundant IL21 transcript among tonsillar CD4+ T-cell subsets (Fig. 4D, day 0) and the highest level of intracytoplasmic IL-21 on 6-h stimulation with PMA and ionomycin (Fig. S2B). The discrepancy between these observations and the secretion of IL-21 (Fig. 4 A and B) might reflect differences in the kinetics of IL21 expression upon activation. Therefore, we analyzed IL21 expression by real-time RT-PCR at different time points after CD3/CD28 mAbs stimulation. Whereas IL21 was expressed at high levels by isolated CXCR5hiICOShi GC-Tfh cells upon isolation (day 0), CXCR5hiICOShi GC-Tfh cells rapidly lost IL21 expression after CD3/CD28 stimulation (Fig. 4D, day 1). In contrast, CXCR5loICOSlo CD4+ T cells strongly up-regulated IL21 expression in response to the stimulation. Loss of IL21 by CXCR5hiICOShi GC-Tfh cells was not because of cell death, because recovery of viable T cells at day 1 was comparable between CXCR5loICOSlo CD4+ T cells and CXCR5hiICOShi GC-Tfh cells (CXCR5loICOSlo CD4+ T-cell recovery, 68 ± 6% of input; CXCR5hiICOShi GC-Tfh cell recovery, 70 ± 9% of input, mean ± SEM, n = 3). Furthermore, expression of CXCL13 transcript by CXCR5hiICOShi GC-Tfh cells was maintained for at least 2 d after stimulation.

Thus, the four tonsillar Th populations display distinct cytokine/chemokine secretion profiles. Upon interaction with B cells, CXCR5loICOSlo CD4+ T cells secrete largest amounts of IL-10 and IL-21, whereas CXCR5hiICOShi GC-Tfh cells secrete largest amounts of CXCL13. CXCR5loICOShi CD4+ T cells secrete the largest amounts of IL-17A.

Factors Involved in the Help of Naïve B Cells by CXCR5loICOSlo CD4+ T Cells.

We next analyzed the molecules that were involved in the help of B-cell subsets by tonsillar Th populations. First, to test the role of IL-21 secreted by CD4+ T cells, a neutralizing IL-21R-Fc chimeric protein was added to the T- and B-cell cocultures. This analysis resulted in the complete inhibition of Ig production in all of the cultures of tonsillar Th populations and B-cell subsets, including those cultures of CXCR5loICOSlo CD4+ T cells and naïve B cells and CXCR5hiICOShi GC-Tfh and GC-B cells (Fig. 5A). Second, the role of ICOS was tested by blocking the ICOS/ICOS–ligand (ICOS-L) interaction with an ICOS-blocking mAb. Inhibition of the ICOS/ICOS-L interaction completely abrogated Ig production by both naïve B and GC-B cells (Fig. 5B) as well as cytokine secretion by CD4+ T cells (Fig. 5C). Third, blocking IL-10 by using a combination of blocking mAbs (i.e., anti–IL-10 and anti–IL-10 receptor) resulted in a partial inhibition of Ig production in the cultures of CXCR5loICOSlo CD4+ T cells with naïve B cells and CXCR5hiICOShi GC-Tfh cells with GC-B cells (Fig. S3). Last, blocking CD40L, a fundamental molecule for B-cell help, was analyzed. Neither CXCR5loICOSlo CD4+ T cells nor CXCR5hiICOShi GC-Tfh cells expressed CD40L when isolated, but both cells rapidly expressed CD40L upon activation (Fig. S4). Blocking CD40L also resulted in a partial inhibition of Ig production (Fig. S5). At variance with blocking the ICOS/ICOS-L interaction, blocking the CD40/CD40L interaction did not inhibit the cytokine secretion from CD4+ T cells (Fig. 5C).

Fig. 5.

Fig. 5.

Molecules associated with helper activity of tonsillar Th populations. (A) IL-21R/Fc chimera protein was added to the cultures of Th populations and the indicated B-cell subsets. Ig concentrations at day 8. A representative from four experiments. Paired t test. ***P < 0.001, **P < 0.01, and *P < 0.05. (B) ICOS mAb was added to the cultures of Th populations and the indicated B cells. Ig concentrations at day 8 (a representative from four experiments). Paired t test. (C) Cytokine secretion. CD40L mAb, ICOS mAb, or an isotype control was added to the cultures of Th populations and the indicated B cells. Cytokine concentrations at day 2 (a representative from three experiments).

Thus, both CXCR5loICOSlo CD4+ T cells and CXCR5hiICOShi GC-Tfh cells help B-cell subsets in a fashion dependent on IL-21, ICOS, IL-10, and CD40L. Whereas the ICOS/ICOS-L interaction was essential for T-cell activation, the CD40/CD40L interaction was important for B-cell differentiation.

CXCR5loICOSlo CD4+ T Cells Induce Apoptosis of GC-B Cells Through the FAS/FAS-L Interaction.

We were intrigued by the ability of CXCR5loICOSlo CD4+ T cells to help both naïve and memory B cells, while being unable to help GC-B cells. Given that the recovery of viable T cells was similar between CXCR5loICOSlo CD4+ T cells and CXCR5hiICOShi GC-Tfh cells cultured with GC-B cells (Fig. 3F), the lack of help from GC-B cells did not seem to be because of total consumption of nutrients in cultures. Furthermore, titrating down the number of CXCR5loICOSlo CD4+ T cells did not result in better help from GC-B cells (Fig. S6). Considering that cocultures of GC-B cells and CXCR5loICOSlo CD4+ T cells resulted in few remaining viable GC-B cells (Fig. 3E), we hypothesized that these T cells might induce the apoptosis of GC-B cells, which express high amounts of FAS (4547) (Fig. 6A). Indeed, soluble FAS-L was detected in cocultures of GC-B cells with CXCR5ICOS, CXCR5loICOSlo, and CXCR5loICOShi CD4+ T cells but not with CXCR5hiICOShi GC-Tfh cells (Fig. 6B). Consistently, FASLG transcript (encoding FAS-L) was expressed by the three tonsillar Th populations but not CXCR5hiICOShi GC-Tfh cells (Fig. 6C). Upon blocking the FAS/FAS-L interaction with a neutralizing anti-FAS antibody, CXCR5loICOSlo and CXCR5loICOShi CD4+ T cells were able to maintain the survival of GC-B cells (Fig. S7) and induce them to produce Igs (Fig. 6D). Thus, FAS-L is expressed by CXCR5loICOSlo and CXCR5loICOShi CD4+ T cells but not CXCR5hiICOShi GC-Tfh cells, and it induces the apoptosis of FAS-expressing GC-B cells.

Fig. 6.

Fig. 6.

CXCR5loICOSlo CD4+ T cells induce apoptosis of GC-B cells through the FAS/FAS-L interaction. (A) Expression of cell surface FAS by GC-B cells. (B) Soluble FAS-L production by Th populations cocultured with GC-B cells. FAS-L concentrations at day 2 (n = 4). One-way ANOVA Bonferonni multiple comparison test. ***P < 0.001, **P < 0.01, and *P < 0.05. (C) FASLG mRNA expression by tonsillar Th populations analyzed by real-time RT-PCR. Expression of FASLG transcript was normalized to that of HPRT1 transcript (n = 3). (D) Ig secretion by GC-B cells cocultured with tonsillar Th populations in the presence of FAS mAb. Ig concentrations were measured at day 8 (a representative from four experiments). (E) Ig secretion by naïve B cells cocultured with CXCR5ICOS CD4+ T cells, CXCR5loICOShi CD4+ T cells, or CXCR5hiICOShi GC-Tfh cells supplemented with titrated doses of recombinant IL-21. Ig secretion measured at day 8 (a representative from two experiments).

We next hypothesized that the differential capacity to help naïve B cells among tonsillar Th populations depends on the capacity to produce IL-21. Thus, we examined whether supplementation of IL-21 into the cocultures of tonsillar Th populations and naïve B cells results in the secretion of higher amounts of Igs. As shown in Fig. 6E, IL-21 dose-dependently induced naïve B cells to produce Igs when cocultured with CXCR5loICOShi CD4+ T cells and CXCR5hiICOShi GC-Tfh cells. Thus, the difference in the capacity to help naïve B cells among tonsillar Th subsets is at least partly dependent on their capacity to produce IL-21.

Thus, the superior capacity of CXCR5loICOSlo CD4+ T cells to secrete IL-21 partly explains their capacity to provide efficient help to naïve B cells.

CXCR5loICOSlo CD4+ T Cells Express Large Amounts of BCL6 Transcript.

Recent studies in mice indicate that development of Tfh cells is reciprocally regulated by the two transcription repressors, Bcl6 and Blimp-1 (3234). To determine the extent of Tfh lineage commitment, the expression of Bcl6 and Blimp-1 (encoded by BCL6 and PRDM1 genes, respectively) in tonsillar Th populations was analyzed by real-time RT-PCR. As shown in Fig. 7A, BCL6 transcript was abundantly expressed by CXCR5loICOSlo CD4+ T cells compared with CXCR5ICOS naïve CD4+ T cells, and the expression was equivalent to that of CXCR5hiICOShi GC-Tfh cells. The expression of PRDM1 transcript was equally low between CXCR5loICOSlo CD4+ T cells and CXCR5hiICOShi GC-Tfh cells. In contrast, CXCR5loICOShi expressed very high amounts of PRDM1 transcript but little BCL6 transcript.

Fig. 7.

Fig. 7.

CXCR5loICOSlo CD4+ T cells express large amounts of BCL6. (A) Expression of BCL6 and PRDM1 transcripts by tonsillar Th populations was analyzed by real-time RT-PCR. Expression of each transcript was normalized to that of HPRT1 transcript (n = 3). One-way ANOVA Bonferonni multiple comparison test. ***P < 0.001, **P < 0.01, and *P < 0.05. (B) Expression of Bcl6 and Blimp-1 proteins in tonsillar Th populations. Sorted Th populations were lysed, and equal amounts of protein were loaded per well to analyze the expression of Bcl6 and Blimp-1 by Western blotting. For the positive controls, the lysate of GC-B cells was used for Bcl6 detection, and BJAB (a B-cell line) cell lysate was used for Blimp-1 detection. Expected Blimp-1 band is indicated with an arrow. Densitometry score ratio for Bcl6 and Blimp-1 against control actin protein is indicated in number (a representative of four experiments). (C) Analysis of Bcl6 expression in tonsillar CD4+ Th populations by flow cytometry (a representative of four experiments). (D) Up-regulation of ICOS and PD-1. The four Th populations were cultured with naïve B cells, and the expression of ICOS and PD-1 on T cells was analyzed at day 5 (a representative from two experiments).

Because Bcl6 expression is controlled by many posttranscriptional regulatory mechanisms (48), the expression of Bcl6 and Blimp-1 proteins were analyzed by Western blot. As shown in Fig. 7B, CXCR5hiICOShi GC-Tfh cells expressed more abundant Bcl6 protein than CXCR5loICOSlo CD4+ T cells. Consistent results were obtained by flow cytometry (Fig. 7C). Blimp-1 protein was barely detected in any tonsillar Th populations. We assumed that the difference in the expression of Bcl6 between CXCR5hiICOShi GC-Tfh cells and CXCR5loICOSlo CD4+ T cells might reflect the difference in the maturation stages. Indeed, whereas the expression of ICOS and PD-1 was low when isolated (Fig. 1C), CXCR5loICOSlo CD4+ T cells robustly up-regulated the expression of ICOS and PD-1 on interaction with naïve B cells at even higher density than CXCR5hiICOShi GC-Tfh cells (Fig. 7D).

Thus, CXCR5loICOSlo CD4+ T cells express large amounts of BCL6 transcript but less Bcl6 protein than CXCR5hiICOShi GC-Tfh cells.

Frequency of CXCR5loICOSlo CD4+ T Cells Is Different in Adult Tonsils.

We analyzed whether CXCR5loICOSlo CD4+ T cells were also present in adult tonsils and human spleen. Whereas pediatric tonsil samples contained significantly more CXCR5hiICOShi GC-Tfh cells than CXCR5loICOSlo CD4+ T cells (Fig. 1B), the frequency of CXCR5loICOSlo CD4+ T cells was slightly higher than that of CXCR5hiICOShi GC-Tfh cells in adult tonsil samples (Fig. 8 A and B) (CXCR5loICOSlo CD4+ T cells, 32 ± 5%; CXCR5hiICOShi GC-Tfh cells, 22 ± 3%). In spleen, the majority of CD4+ T cells were CXCR5ICOSCD127+ CD4+ T cells, and CXCR5hiICOShi GC-Tfh cells were barely found. The separation between CXCR5ICOS and CXCR5loICOSlo CD4+ T-cell populations was not clear, and CXCR5loICOSlo CD4+ T cells, if there were any of these cells, expressed only low levels of CXCR5 (Fig. 8A).

Fig. 8.

Fig. 8.

CXCR5loICOSlo CD4+ T cells in adult tonsils. The four Th populations in pediatric tonsil samples, adult tonsil samples, and human spleen samples. The same number of T-enriched cells (after removing B cells with magnetic beads) was stained and analyzed in parallel in identical conditions. The four Th populations were determined based on the analysis of pediatric tonsil samples, and the same gates were applied for the adult and spleen samples. (A) A representative of at least two experiments gated to CD3+CD4+ T cells. (B) The frequency of each Th population in adult tonsils (n = 3) and spleen (n = 2). One-way ANOVA Bonferonni multiple comparison test for adult tonsil samples. *P < 0.05.

Thus, the frequency of CXCR5loICOSlo CD4+ T cells is different among human secondary lymphoid organs, and the CXCR5loICOSlo CD4+ T-cell population was not clearly identified in spleen.

Discussion

Our study characterizes a previously undefined BCL6-expressing human tonsillar CD4+ T-cell subset, which can be identified as CXCR5loICOSloIL-7R+CD45RO+ cells. The abundance of BCL6 and PRDM1 transcripts in CXCR5loICOSlo CD4+ T cells was similar to that in CXCR5hiICOShi GC-Tfh cells. A previous report showed that tonsillar CXCR5lo CD4+ T cells express lower amounts of BCL6 transcript than CXCR5hi Tfh cells (38). The discrepancy between that study and our study lies in the difference in cell isolation strategies. Our study also separated the CXCR5lo Th cell population into two populations according to the expression of ICOS. Whereas CXCR5loICOSlo CD4+ T cells expressed abundant BCL6 transcript, CXCR5loICOShi CD4+ T cells barely expressed BCL6 transcript but expressed large amounts of PRDM1 transcript. Thus, our study usefully complements this earlier study and shows that CXCR5lo CD4+ T cells contain two cell types with different functions: Tfh-committed CXCR5loICOSlo and non–Tfh-committed CXCR5loICOShi cells. Indeed, CXCR5loICOShi CD4+ T cells were poorly efficient at helping B-cell responses.

The two BCL6-expressing tonsillar Th subsets, CXCR5loICOSlo CD4+ T cells and CXCR5hiICOShi GC-Tfh cells, were found to differentially help B cells. Consistent with previous studies showing efficient help of B cells by GC-Tfh cells (14, 24, 41), CXCR5hiICOShi GC-Tfh cells were efficient at helping GC-B cells. Reciprocally, GC-B cells were able to maintain the survival of CXCR5hiICOShi GC-Tfh cells, because the recovery of viable CXCR5hiICOShi GC-Tfh cells was significantly better when cultured with GC-B cells than with naïve B cells (Fig. 3 F and I) (P < 0.05, n = 3, paired t test). Equivalent concentrations of cytokines were detected when CXCR5hiICOShi GC-Tfh cells were cultured with either GC-B or naïve B cells (Fig. 4 A and B), suggesting that the better survival of GC-Tfh cells does not depend on cytokines derived from GC-Tfh cells but rather, on intrinsic factors of GC-B cells that remain to be determined.

We found that CXCR5loICOSlo CD4+ T cells were far more efficient than GC-Tfh cells at inducing naïve B cells to proliferate and differentiate into Ig-producing cells. The difference between the two Th populations in the ability to help naïve B cells was at least partly explained by the differences in the secretion of cytokines, particularly IL-21. Our study shows that the two Th populations display intrinsically distinct cytokine production properties. Upon activation, GC-Tfh cells secreted substantially higher levels of CXCL13 protein and maintained higher levels of CXCL13 transcript expression than CXCR5loICOSlo CD4+ T cells, showing that GC-Tfh cells display a machinery to produce large amounts of CXCL13. In contrast, CXCR5loICOSlo CD4+ T cells secreted larger amounts of IL-10 and IL-21 than GC-Tfh cells upon stimulation with CD3/CD28 stimulation and even upon coculture with B cells. Whereas GC-Tfh cells express highest levels of IL21 transcript upon isolation, GC-Tfh cells rapidly lost IL21 expression upon activation. This finding was in contrast to the up-regulation of IL21 transcript by CXCR5loICOSlo CD4+ T cells after activation, which leads to the secretion of large amounts of IL-21.

Our study suggests that CXCR5loICOSloCD4+ T cells are committed to the Tfh lineage, because (i) they showed similar profiles of BCL6 and PRDM1 expression with GC-Tfh cells, (ii) they helped B cells in a manner dependent on IL-21, IL-10, ICOS, and CD40L, thus sharing functions with GC-Tfh cells, and (iii) they robustly up-regulated ICOS and PD-1 expression upon activation, thus sharing phenotype with GC-Tfh cells. CXCR5loICOSloCD4+ T cells were exclusively localized outside GCs and therefore, might represent extrafollicular helper cells engaged in inducing the differentiation of B cells into extrafollicular plasma cells. Consistently, recent studies using lupus-prone mice showed that extrafollicular helper cells share the property with GC-Tfh cells, including Bcl-6 expression (35, 36). We cannot exclude, however, the possibility that CXCR5loICOSlo CD4+ T cells represent precursors of GC-Tfh cells. CXCR5loICOSlo CD4+ T cells expressed lower amounts of Bcl6 protein than CXCR5hiICOShi GC-Tfh cells, which might reflect the fact that CXCR5loICOSlo CD4+ T cells are in a transition stage of differentiating into mature GC-Tfh cells. Robust up-regulation of ICOS and PD-1 by CXCR5loICOSlo CD4+ T cells upon interaction with B cells supports this hypothesis. Alternatively, CXCR5loICOSlo CD4+ T cells might constitute both extrafollicular helper cells and GC-Tfh precursors and modify their balance according the microenvironment of secondary lymphoid organs.

The specialized role of CXCR5loICOSloCD4+ T cells for the help of B cells outside GCs was also supported by the observation that they lacked the capacity to help GC-B cells. Instead, they induced the apoptosis of GC-B cells through the interaction of FAS/FAS-L. Whereas activated naïve B cells and memory B cells also express FAS (45), CXCR5loICOSlo CD4+ T cells did not induce their apoptosis, likely because naïve and memory B cells express abundant Bcl2, which protects them from apoptosis through the FAS/FAS-L interaction (45). Induction of apoptosis of GC-B cells by CXCR5loICOSlo CD4+ T cells might represent a fail-safe system to eliminate GC-B cells that prematurely migrate out of GCs. Alternatively, because the FAS/FAS-L interaction is essential for the depletion of low-affinity GC-B cells during GC reactions (49), CXCR5loICOSlo CD4+ T cells that have just migrated into GCs, provided that they have an ability to do so, might be involved in this type of negative selection process of B cells in GCs. In addition, CXCR5loICOShi CD4+ T cells, which largely lacked an ability to help B cells, were also able to help GC-B cells when the FAS/FAS-L interaction was blocked. Thus, CXCR5loICOShi CD4+ T cells might also be involved in this negative selection process. In line with this finding, CXCR5loICOShi CD4+ T cells contained CD57+ cells, which were reported to express FAS-L (42).

Identification of BCL6-expressing Tfh-committed cells outside GCs suggests that the differentiation of human Tfh cells initiates outside B-cell follicles. Therefore, similar to other conventional Th subsets, the Th differentiation program to the Tfh lineage might initiate when naïve CD4+ T cells interact with antigen-presenting DCs. This observation supports our previous proposal that a particular type of DC subset is specialized for the development of Tfh cells (50). In humans, IL-12 secreted by DCs seems to be a major cytokine in the development of IL-21–producing Tfh-like cells (51, 52). Furthermore, IL-12 was shown to induce Bcl6 expression in human CD4+ T cells (38, 53). Therefore, IL-12–producing DC subsets, such as CD14+ dermal DCs (50), likely represent an important human DC subset that initiates the development of Tfh cells.

In conclusion, tonsillar CXCR5loICOSlo CD4+ T cells represent Tfh-committed cells helping B-cell responses outside GCs. Determining whether circulating CXCR5+ CD4+ T cells, a cell population proposed to be a memory component of Tfh cells (54, 55), can be derived from CXCR5loICOSlo CD4+ T cells will be of great interest. Additional characterization of CXCR5loICOSlo CD4+ T cells together with determination of their developmental mechanisms will bring significant insights to the understanding of human autoimmune disease pathogenesis, in which extrafollicular plasma cell differentiation might be involved (36). Furthermore, studies on CXCR5loICOSloCD4+ T cells will be also beneficial for the design of vaccines against infectious diseases.

Materials and Methods

Cell Isolation.

All of the studies described here were approved by the Institutional Review Board of Baylor Research Institute. Tonsil samples were obtained from young patients (3–12 y) or adults undergoing tonsillectomy, and single cells were collected by mechanical disruption of tonsil samples. Spleen samples were obtained from a cadaveric organ donor or the tissue bank at Baylor University Medical Center. B cells were first positively selected with CD19 MACS Microbeads (Miltenyi Biotech). For the isolation of Th populations, the CD19-negative fraction was stained with biotin-conjugated anti-ICOS (ISA-3; eBioscience) /streptavidin PE-Cy7 (BD), anti-CD20 FITC (2H7; eBioscience), anti-CXCR5 phycoerythrin (PE) (51505.111; R&D Systems), anti-CD4 Pacific Blue (RPA-T4; BD), anti-CD8 allophycocyanin (APC) (RPA-8; eBioscience), and anti-CD56 APC (MEM 188; eBioscience). The four Th populations were sorted with FACSAria (BD Biosciences) according to the expression of CXCR5 and ICOS within the CD4+CD20CD8CD56 cell population. For the isolation of B-cell subsets, CD19+ B cells were stained with anti-CD20 FITC (2H7; eBioscience), anti-CD3 PE-Cy5 (UCHTI; eBioscience), anti-IgD PE (IA6-2; BD), and anti-CD38 APC (HIT2; eBioscience). Naïve B cells (CD20+IgD+CD38), memory B cells (CD20+IgDCD38), and GC-B cells (CD20+IgDCD38+) were sorted within the CD3-negative cell population.

Phenotype Analysis.

The phenotypes of tonsillar Th and B-cell subsets were analyzed with the antibodies anti-PD1 PE (J116; eBioscience), CD57 PE (NK-1; SouthernBiotech), CD45RO PE-Cy5 (UCHL1; eBioscience), CD127 PE (hIL-7R-M21; BD), and CD95 Pacific blue (DX2; eBioscience) after gating to each population, which was described above. For the analysis of Bcl6 protein expression, isolated tonsillar Th cells were stained with anti-Bcl6 AlexaFluor 647 (K112-91; BD) after cell permeabilization.

Immunohistochemistry and Immunofluorescence.

Tonsils were placed in Tissue-Tek OCT (Sakura), frozen in liquid nitrogen, and stored at −80 °C. For immunohistochemistry, 6-μm cryostat sections were fixed in acetone and air-dried. After treatment with FcReceptor Block and CytoQ Background Buster (Innovex Biosciences) for 20 min each, slides were incubated for 40 min at room temperature with the primary antibody against CD127 (eBioRDR5, purified; eBioscience) diluted in Cyto Q Immuno Diluent. The Stat-Q Peroxidase Staining System with AEC (Innovex Biosciences) was used according to the manufacturer's protocol. Sections were counterstained with Aqua Hematoxylin (Innovex Biosciences), rinsed in tap water, and prepared on coverslips using Advantage Mounting media from Innovex. Slides images were acquired using a Nikon DXM1200C digital camera and an Olympus BX60 microscope with Plan4×/0.13 objective.

For immunofluorescence, the following antibodies were used: anti-CD4 (RPA-T4, purified; BD) conjugated with Molecular Probes Alexa 568 monoclonal antibody kit, anti-CD127 Alexa647 (eBioRDR5; eBioscience), and anti-CD45RO FITC (UCHL1; eBioscience). Anti-FITC Alexa 488 goat IgG fraction (Molecular Probes) was used as a secondary to enhance the FITC signal. Sections were blocked with both FcReceptor Block and Background Buster, and then, they were incubated for 1 h in the antibodies diluted in CytoQ immunodiluent, rinsed, and incubated in anti-FITC. DAPI (Molecular Probes) was used to stain the nuclei. Slides were mounted with Fluoromount-G (Southern Biotech). They were observed using Metamorph software version 6.2 (Universal Imaging Corporation), a Roper coolsnap HQ camera, and an Olympus BCXCR51 microscope with Plan10×/0.40 objective. Slides were also observed under a Leica SP5 confocal microscope with a 40×/1.25 Planapo objective.

Cell Culture.

Sorted Th populations were cocultured with B-cell subsets (2 × 104 cells/well each for 8 d for Ig measurements and 5 × 104 cells/well each for 2 d for cytokine measurements) in RPMI medium 1640 (GIBCO) supplemented with 1% l-glutamine (Sigma), 1% penicillin/streptomycin (Sigma), 1% sodium pyruvate (Sigma), 1% nonessential amino acids (Sigma), 50 μM β-mercaptoethanol (Sigma), 50 μg/mL gentamycin (GIBCO), and 10% heat-inactivated FCS (ATCC) in the presence of SEB (1 μg/mL, Sigma-Aldrich) in U-bottomed 96-well plates. The concentrations of Igs (IgM, IgG, and IgA) were determined by ELISA as described previously (51). Indicated cytokines were measured with Luminex (51). The concentrations of CXCL13 and soluble FAS-L (TNFSF6; both from R&D Systems) were determined by ELISA. Sorted Th populations (5 × 104 cells/well) were also stimulated for 2 d with CD3/CD28 beads (0.2 μL/well) for cytokine measurement. In blocking experiments, the following reagents were added to the cocultures: anti-ICOS blocking mAb (10 μg/mL; Ancell), IL-21R/Fc (20 μg/mL; R&D Systems), anti-Fas blocking antibody (20 μg/mL; Millipore), and isotype-matched controls. In some experiments, titrated amounts of IL-21 (R&D Systems) were added to the cocultures. For the analysis of plasma cell differentiation, naïve B cells labeled with 5-(and-6)-carboxyfluorescein diacetate succinimidyl este (Molecular Probes) were cultured for 8 d with Th populations. The cultured cells were stained with anti-CD3 PE-Cy5, anti-CD4 Pacific Blue, and anti-CD38 APC, and the frequency of CFSECD38hi plasmablast cells within CD3CD4 cells was analyzed by FACS Canto II. In some experiments, Th populations were cultured with naïve B cells for 5 d in the presence of SEB and then assessed for their expression of ICOS and PD1.

Real-Time PCR.

Total RNA was extracted from tonsil Th populations before or after culture with CD3/CD28 mAb-coated beads (5 × 104 cells/0.2 μL beads per well; Dynal) using the RNeasy Mini kit (QIAGEN) and reverse-transcribed into cDNA in a 96-well plate using the High Capacity cDNA Archive kit (Applied Biosystems). The primer pairs (Integrated DNA Technology) used in this study were designed using the Roche Primer Design Program. Primer sequences were as follows: BCL6 (accession number NM_001706.2) forward primer: 5′-ttccgctacaagggcaac-3′, reverse primer: 5′-tgcaacgatagggtttctca-3′; PRDM1 (accession number NM_001198.2) forward primer: 5′-gtggtgggttaatcggtttg-3′, reverse primer: 5′-gaagctcccctctggaataga-3′; FASLG (accession number NM_000639.1) forward primer: 5′-tggggatgtttcagctcttc-3′, reverse primer: 5′-tgtgcatctggctggtagac-3′; IL-21 (accession number NM_021803.2) forward primer: 5′-aggaaaccaccttccacaaa-3′, reverse primer: 5′-gaatcacatgaagggcatgtt-3′; CXCL13 (accession number NM_0006419.2) forward primer: 5′-tctctgcttctcatgctgct-3′, reverse primer: 5′-tcaagcttgtgtaatagacctcca-3′.

Real-time PCR was set up with Roche Probes Master reagents and Universal Probe Library hydrolysis probes. PCR reactions were performed on the LightCycler 480 (Roche Applied Science) followed these conditions: step 1 (denaturation) at 95 °C for 5 min, step 2 (amplification for 45 cycles) at 95 °C for 10 s and 60 °C for 30 s, and step 3 (cooling) at 40 °C for 30 s. The expression of each gene was normalized to that of a housekeeping gene HRPT1.

Western Blotting.

Total proteins were extracted from sorted tonsil Th populations using radioimmunoprecipitation assay buffer (Sigma-Aldrich) supplemented with 1% protease inhibitor mixture (Sigma-Aldrich). Equal amounts of protein per sample were separated on NuPAGE (Invitrogen) 4–12% Bis⋅Tris gradient gels and transferred to PVDF membranes (Invitrogen). Membranes were incubated with Abs against Bcl6 (clone D8) and Blimp-1 (clone 6D3) antibody (Santa Cruz Biotechnology, Inc.) followed by HRP-conjugated anti-mouse or anti-rat, respectively (Santa Cruz Biotechnology). Equal protein loading was confirmed using goat antiactin (clone I-19; Santa Cruz Biotechnology, Inc). Densitometry score was determined with ImageJ software.

Supplementary Material

Supporting Information

Acknowledgments

We thank E. Kowalski and S. Coquery for cell sorting, S. Zurawski for IL-21 multiplex assay, I. Munagala for RT-PCR, and C. Harrod for proofreading the manuscript. We thank Dr. Robert Coffman for reading the manuscript and providing comments. We thank M. Ramsay for continuous help. This study was supported by National Institutes of Health (NIH) Grants R01-CA84512, R01-CA078846, AR054083-01, U19-AI057234 (to J.B.), and U19-AI082715-01 (to H.U.; Program principal investigator: Virginia Pascual); and the Baylor Health Care System (to J.B. and H.U.).

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

See Author Summary on page 13371.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1100898108/-/DCSupplemental.

References

  • 1.Garside P, et al. Visualization of specific B and T lymphocyte interactions in the lymph node. Science. 1998;281:96–99. doi: 10.1126/science.281.5373.96. [DOI] [PubMed] [Google Scholar]
  • 2.Okada T, et al. Antigen-engaged B cells undergo chemotaxis toward the T zone and form motile conjugates with helper T cells. PLoS Biol. 2005;3:e150. doi: 10.1371/journal.pbio.0030150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.MacLennan IC, Gray D. Antigen-driven selection of virgin and memory B cells. Immunol Rev. 1986;91:61–85. doi: 10.1111/j.1600-065x.1986.tb01484.x. [DOI] [PubMed] [Google Scholar]
  • 4.McHeyzer-Williams MG, McLean MJ, Lalor PA, Nossal GJ. Antigen-driven B cell differentiation in vivo. J Exp Med. 1993;178:295–307. doi: 10.1084/jem.178.1.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Jacob J, Kassir R, Kelsoe G. In situ studies of the primary immune response to (4-hydroxy-3-nitrophenyl)acetyl. I. The architecture and dynamics of responding cell populations. J Exp Med. 1991;173:1165–1175. doi: 10.1084/jem.173.5.1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.MacLennan IC, et al. Extrafollicular antibody responses. Immunol Rev. 2003;194:8–18. doi: 10.1034/j.1600-065x.2003.00058.x. [DOI] [PubMed] [Google Scholar]
  • 7.Tarlinton DM. Evolution in miniature: Selection, survival and distribution of antigen reactive cells in the germinal centre. Immunol Cell Biol. 2008;86:133–138. doi: 10.1038/sj.icb.7100148. [DOI] [PubMed] [Google Scholar]
  • 8.MacLennan IC. Germinal centers. Annu Rev Immunol. 1994;12:117–139. doi: 10.1146/annurev.iy.12.040194.001001. [DOI] [PubMed] [Google Scholar]
  • 9.King C, Tangye SG, Mackay CR. T follicular helper (TFH) cells in normal and dysregulated immune responses. Annu Rev Immunol. 2008;26:741–766. doi: 10.1146/annurev.immunol.26.021607.090344. [DOI] [PubMed] [Google Scholar]
  • 10.Fazilleau N, Mark L, McHeyzer-Williams LJ, McHeyzer-Williams MG. Follicular helper T cells: Lineage and location. Immunity. 2009;30:324–335. doi: 10.1016/j.immuni.2009.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Breitfeld D, et al. Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med. 2000;192:1545–1552. doi: 10.1084/jem.192.11.1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Schaerli P, et al. CXC chemokine receptor 5 expression defines follicular homing T cells with B cell helper function. J Exp Med. 2000;192:1553–1562. doi: 10.1084/jem.192.11.1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kim CH, et al. Subspecialization of CXCR5+ T cells: B helper activity is focused in a germinal center-localized subset of CXCR5+ T cells. J Exp Med. 2001;193:1373–1381. doi: 10.1084/jem.193.12.1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rasheed AU, Rahn HP, Sallusto F, Lipp M, Müller G. 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. 2006;36:1892–1903. doi: 10.1002/eji.200636136. [DOI] [PubMed] [Google Scholar]
  • 15.Ansel KM, McHeyzer-Williams LJ, Ngo VN, McHeyzer-Williams MG, Cyster JG. In vivo-activated CD4 T cells upregulate CXC chemokine receptor 5 and reprogram their response to lymphoid chemokines. J Exp Med. 1999;190:1123–1134. doi: 10.1084/jem.190.8.1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Good-Jacobson KL, et al. PD-1 regulates germinal center B cell survival and the formation and affinity of long-lived plasma cells. Nat Immunol. 2010;11:535–542. doi: 10.1038/ni.1877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Akiba H, et al. The role of ICOS in the CXCR5+ follicular B helper T cell maintenance in vivo. J Immunol. 2005;175:2340–2348. doi: 10.4049/jimmunol.175.4.2340. [DOI] [PubMed] [Google Scholar]
  • 18.Bossaller L, et al. ICOS deficiency is associated with a severe reduction of CXCR5+CD4 germinal center Th cells. J Immunol. 2006;177:4927–4932. doi: 10.4049/jimmunol.177.7.4927. [DOI] [PubMed] [Google Scholar]
  • 19.Hutloff A, et al. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature. 1999;397:263–266. doi: 10.1038/16717. [DOI] [PubMed] [Google Scholar]
  • 20.Vogelzang A, et al. A fundamental role for interleukin-21 in the generation of T follicular helper cells. Immunity. 2008;29:127–137. doi: 10.1016/j.immuni.2008.06.001. [DOI] [PubMed] [Google Scholar]
  • 21.Vinuesa CG, et al. A RING-type ubiquitin ligase family member required to repress follicular helper T cells and autoimmunity. Nature. 2005;435:452–458. doi: 10.1038/nature03555. [DOI] [PubMed] [Google Scholar]
  • 22.Linterman MA, et al. Follicular helper T cells are required for systemic autoimmunity. J Exp Med. 2009;206:561–576. doi: 10.1084/jem.20081886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cannons JL, et al. Optimal germinal center responses require a multistage T cell:B cell adhesion process involving integrins, SLAM-associated protein, and CD84. Immunity. 2010;32:253–265. doi: 10.1016/j.immuni.2010.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bryant VL, et al. 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. 2007;179:8180–8190. doi: 10.4049/jimmunol.179.12.8180. [DOI] [PubMed] [Google Scholar]
  • 25.Chtanova T, et al. 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. 2004;173:68–78. doi: 10.4049/jimmunol.173.1.68. [DOI] [PubMed] [Google Scholar]
  • 26.Good KL, Bryant VL, Tangye SG. Kinetics of human B cell behavior and amplification of proliferative responses following stimulation with IL-21. J Immunol. 2006;177:5236–5247. doi: 10.4049/jimmunol.177.8.5236. [DOI] [PubMed] [Google Scholar]
  • 27.Spolski R, Leonard WJ. Interleukin-21: Basic biology and implications for cancer and autoimmunity. Annu Rev Immunol. 2008;26:57–79. doi: 10.1146/annurev.immunol.26.021607.090316. [DOI] [PubMed] [Google Scholar]
  • 28.Ozaki K, et al. A critical role for IL-21 in regulating immunoglobulin production. Science. 2002;298:1630–1634. doi: 10.1126/science.1077002. [DOI] [PubMed] [Google Scholar]
  • 29.Kuchen S, et al. Essential role of IL-21 in B cell activation, expansion, and plasma cell generation during CD4+ T cell-B cell collaboration. J Immunol. 2007;179:5886–5896. doi: 10.4049/jimmunol.179.9.5886. [DOI] [PubMed] [Google Scholar]
  • 30.Pène J, et al. Cutting edge: IL-21 is a switch factor for the production of IgG1 and IgG3 by human B cells. J Immunol. 2004;172:5154–5157. doi: 10.4049/jimmunol.172.9.5154. [DOI] [PubMed] [Google Scholar]
  • 31.Ettinger R, et al. IL-21 induces differentiation of human naive and memory B cells into antibody-secreting plasma cells. J Immunol. 2005;175:7867–7879. doi: 10.4049/jimmunol.175.12.7867. [DOI] [PubMed] [Google Scholar]
  • 32.Yu D, et al. The transcriptional repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity. 2009;31:457–468. doi: 10.1016/j.immuni.2009.07.002. [DOI] [PubMed] [Google Scholar]
  • 33.Nurieva RI, et al. Bcl6 mediates the development of T follicular helper cells. Science. 2009;325:1001–1005. doi: 10.1126/science.1176676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Johnston RJ, et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science. 2009;325:1006–1010. doi: 10.1126/science.1175870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Poholek AC, et al. In vivo regulation of Bcl6 and T follicular helper cell development. J Immunol. 2010;185:313–326. doi: 10.4049/jimmunol.0904023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Odegard JM, et al. ICOS-dependent extrafollicular helper T cells elicit IgG production via IL-21 in systemic autoimmunity. J Exp Med. 2008;205:2873–2886. doi: 10.1084/jem.20080840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Haynes NM, et al. 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. 2007;179:5099–5108. doi: 10.4049/jimmunol.179.8.5099. [DOI] [PubMed] [Google Scholar]
  • 38.Ma CS, et al. 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. 2009;87:590–600. doi: 10.1038/icb.2009.64. [DOI] [PubMed] [Google Scholar]
  • 39.Lim HW, Kim CH. Loss of IL-7 receptor alpha on CD4+ T cells defines terminally differentiated B cell-helping effector T cells in a B cell-rich lymphoid tissue. J Immunol. 2007;179:7448–7456. doi: 10.4049/jimmunol.179.11.7448. [DOI] [PubMed] [Google Scholar]
  • 40.Reinhardt RL, Liang HE, Locksley RM. Cytokine-secreting follicular T cells shape the antibody repertoire. Nat Immunol. 2009;10:385–393. doi: 10.1038/ni.1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kim JR, Lim HW, Kang SG, Hillsamer P, Kim CH. Human CD57+ germinal center-T cells are the major helpers for GC-B cells and induce class switch recombination. BMC Immunol. 2005;6:3. doi: 10.1186/1471-2172-6-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Marinova E, Han S, Zheng B. Human germinal center T cells are unique Th cells with high propensity for apoptosis induction. Int Immunol. 2006;18:1337–1345. doi: 10.1093/intimm/dxl066. [DOI] [PubMed] [Google Scholar]
  • 43.Dono M, et al. Heterogeneity of tonsillar subepithelial B lymphocytes, the splenic marginal zone equivalents. J Immunol. 2000;164:5596–5604. doi: 10.4049/jimmunol.164.11.5596. [DOI] [PubMed] [Google Scholar]
  • 44.Kim CH, et al. Unique gene expression program of human germinal center T helper cells. Blood. 2004;104:1952–1960. doi: 10.1182/blood-2004-03-1206. [DOI] [PubMed] [Google Scholar]
  • 45.Martinez-Valdez H, et al. Human germinal center B cells express the apoptosis-inducing genes Fas, c-myc, P53, and Bax but not the survival gene bcl-2. J Exp Med. 1996;183:971–977. doi: 10.1084/jem.183.3.971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Watanabe D, Suda T, Nagata S. Expression of Fas in B cells of the mouse germinal center and Fas-dependent killing of activated B cells. Int Immunol. 1995;7:1949–1956. doi: 10.1093/intimm/7.12.1949. [DOI] [PubMed] [Google Scholar]
  • 47.Smith KG, Nossal GJ, Tarlinton DM. FAS is highly expressed in the germinal center but is not required for regulation of the B-cell response to antigen. Proc Natl Acad Sci USA. 1995;92:11628–11632. doi: 10.1073/pnas.92.25.11628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Crotty S, Johnston RJ, Schoenberger SP. Effectors and memories: Bcl-6 and Blimp-1 in T and B lymphocyte differentiation. Nat Immunol. 2010;11:114–120. doi: 10.1038/ni.1837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hao Z, et al. Fas receptor expression in germinal-center B cells is essential for T and B lymphocyte homeostasis. Immunity. 2008;29:615–627. doi: 10.1016/j.immuni.2008.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Klechevsky E, et al. Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity. 2008;29:497–510. doi: 10.1016/j.immuni.2008.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Schmitt N, et al. Human dendritic cells induce the differentiation of interleukin-21-producing T follicular helper-like cells through interleukin-12. Immunity. 2009;31:158–169. doi: 10.1016/j.immuni.2009.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ueno H, Schmitt N, Palucka AK, Banchereau J. Dendritic cells and humoral immunity in humans. Immunol Cell Biol. 2010;88:376–380. doi: 10.1038/icb.2010.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lund R, et al. Identification of genes involved in the initiation of human Th1 or Th2 cell commitment. Eur J Immunol. 2005;35:3307–3319. doi: 10.1002/eji.200526079. [DOI] [PubMed] [Google Scholar]
  • 54.Morita R, et al. Human blood CXCR5(+)CD4(+) T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity. 2011;34:108–121. doi: 10.1016/j.immuni.2010.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chevalier N, et al. CXCR5 expressing human central memory CD4 T cells and their relevance for humoral immune responses. J Immunol. 2011;186:5556–5568. doi: 10.4049/jimmunol.1002828. [DOI] [PubMed] [Google Scholar]
Proc Natl Acad Sci U S A. 2011 Aug 16;108(33):13371–13372.

Author Summary

Author Summary

CD4+ helper T cells derived from the thymus play a fundamental role in the generation of antibodies against specific antigens, an important function in immune response. Among CD4+ helper T cells, a subset called T follicular helper (Tfh) cells provides help to antibody-producing B cells in germinal centers, structures found in secondary lymphoid organs such as lymph nodes and spleen. In germinal centers, B cells are selected through their interaction with Tfh cells and follicular dendritic cells. Selected B cells then differentiate into either long-lived, antibody-producing plasma cells or memory B cells, which produce large amounts of antibodies upon a secondary antigen challenge (1). Therefore, germinal center response is central to the generation of high-affinity antibodies. CD4+ T cells also provide help to B cells outside germinal centers and induce the differentiation of B cells into either short-lived plasma cells (extrafollicular plasma cells) or B cells that participate in germinal center responses. However, the identity of such CD4+ T cells involved in the assistance of B cells outside germinal centers remains unclear, particularly in humans. In this study, we identified a CD4+ T-cell subset in human tonsils. This CD4+ T-cell subset shares properties with Tfh cells but exclusively localizes outside germinal centers.

We analyzed the phenotype and functions of distinct CD4+ T-cell subsets in human tonsils. Human tonsillar Tfh cells coexpress the chemokine receptor 5 (CXCR5) and the inducible costimulator (ICOS) at high density (2) but not a receptor protein called IL-7 receptor. The tonsillar CD4+ T-cell subset specialized for the help of B cells outside germinal centers expressed CXCR5 and ICOS only at low levels (here called CXCR5loICOSlo CD4+ T cells) but expressed the marker IL-7 receptor abundantly. Furthermore, although a majority of Tfh cells express the protein marker programmed death-1 (PD-1), only a minor fraction of CXCR5loICOSlo CD4+ T cells expressed PD-1 at very low density.

To determine their functions, the isolated tonsillar CD4+ T-cell subsets were cocultured with B-cell subsets, including naïve (IgD+CD38CD19+), memory (IgDCD38CD19+), and germinal center B (IgDCD38+CD19+) cells. T- and B-cell cocultures revealed that CXCR5loICOSlo CD4+ T cells and Tfh cells (i) differentially secrete cytokines and chemokines and (ii) differentially help B-cell subsets (Fig. P1). Thus, Tfh cells secrete large amounts of the chemokine CXCL13 among tonsillar CD4+ T-cell populations, whereas CXCR5loICOSlo CD4+ T cells secrete large amounts of cytokines IL-10 and IL-21. Tfh cells are efficient at helping germinal center B cells, and they promote their survival and Ig production. In contrast, CXCR5loICOSlo CD4+ T cells are far more efficient than Tfh cells at inducing naïve B cells to proliferate and differentiate into antibody secreting cells. Blocking experiments showed that CXCR5loICOSlo CD4+ T cells help naïve B cells in a manner dependent on multiple cytokines and surface molecules, including IL-21, IL-10, ICOS, and CD40L. The difference between the CXCR5loICOSlo CD4+ T cells and Tfh cells in the capacity to help naïve B cells was, at least partly, explained by the differences in IL-21 secretion.

Fig. P1.

Fig. P1.

Two distinct Tfh-committed cells in human tonsils differentially help B-cell subsets. CXCR5loICOSlo CD4+ T cells localize exclusively outside germinal centers (GC) and induce naïve and memory B cells to become antibody-secreting cells. CXCR5hiICOShi GC-Tfh cells are specialized to help B cells in GCs. Whereas CXCR5loICOSlo CD4+ T cells produce higher levels of IL-21 upon interaction with B cells, GC-Tfh cells produce higher levels of CXCL13. CXCR5loICOSlo CD4+ T cells and GC-Tfh cells show similar expression profiles of BCL6 and PRDM1 transcripts. Our study suggests that tonsillar CXCR5loICOSlo CD4+ T cells represent Tfh-committed extrafollicular helper cells and/or precursors of GC-Tfh cells.

Notably, CXCR5loICOSloCD4+ T cells failed to help germinal center B cells and did not maintain their survival. We hypothesize that CXCR5loICOSloCD4+ T cells might induce the death of germinal center B cells, which express high amounts of surface molecules delivering a death signal called FAS. Indeed, soluble FAS ligand was detected in cocultures of germinal center B cells with CXCR5loICOSlo CD4+ T cells but not with Tfh cells. Consistently, FASLG transcript (encoding FAS-L) was expressed by CXCR5loICOSlo CD4+ T cells but was expressed very little by Tfh cells. Upon blocking the FAS/FAS–ligand interaction with a neutralizing anti-FAS antibody, CXCR5loICOSlo CD4+ T cells were able to maintain the survival of germinal center B cells and induce them to produce antibodies. These observations indicate that CXCR5loICOSlo CD4+ T cells induce apoptosis of germinal center B cells through FAS/FAS–ligand interaction.

We also analyzed the expression of two transcriptional repressors, B-cell lymphoma 6 (Bcl6) and B lymphocyte-induced maturation protein 1 (encoded by PRDM1 gene), by tonsillar CD4+ T-cell populations. Bcl6 is expressed at high levels by tonsillar Tfh cells. Mouse studies indicate that Tfh cell generation in vivo is positively regulated by Bcl6 and negatively regulated by B lymphocyte-induced maturation protein 1 (3). We found that the CXCR5loICOSlo CD4+ T cells express BCL6 and PRDM1 transcripts at equivalent levels with Tfh cells (thus, abundant BCL6 and few PRDM1 transcripts).

Collectively, our study suggests that CXCR5loICOSloCD4+ T cells are committed to the Tfh lineage; they showed similar profiles of BCL6 and PRDM1 expression with Tfh cells, and they helped B cells in a manner dependent on IL-21, IL-10, ICOS, and CD40L, thus sharing functions with Tfh cells. Furthermore, CXCR5loICOSloCD4+ T cells robustly up-regulated ICOS and PD-1 expression upon activation, thus sharing a phenotype with Tfh cells. At variance with Tfh cells, CXCR5loICOSloCD4+ T cells were exclusively localized outside germinal centers. Therefore, CXCR5loICOSloCD4+ T cells might represent extrafollicular helper cells engaged in inducing the differentiation of B cells into extrafollicular plasma cells. This hypothesis is supported by the demonstration in lupus-prone mice models of the presence of extrafollicular helper cells that share the property with Tfh cells, including Bcl-6 expression (4). We cannot exclude, however, the possibility that CXCR5loICOSlo CD4+ T cells represent precursors of Tfh cells.

Our study shows that CXCR5loICOSlo CD4+ T cells represent Tfh-committed cells helping B-cell responses outside germinal centers. Additional characterization of CXCR5loICOSlo CD4+ T cells together with determination of their developmental mechanisms will bring significant insights to the pathogenesis of human autoimmune diseases, where extrafollicular B-cell responses are involved (4). Such studies might also benefit the design of vaccines against infectious diseases.

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

See full research article on page E488 of www.pnas.org.

Cite this Author Summary as: PNAS 10.1073/pnas.1100898108.

References

  • 1.King C, Tangye SG, Mackay CR. T follicular helper (TFH) cells in normal and dysregulated immune responses. Annu Rev Immunol. 2008;26:741–766. doi: 10.1146/annurev.immunol.26.021607.090344. [DOI] [PubMed] [Google Scholar]
  • 2.Rasheed AU, Rahn HP, Sallusto F, Lipp M, Müller G. 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. 2006;36:1892–1903. doi: 10.1002/eji.200636136. [DOI] [PubMed] [Google Scholar]
  • 3.Johnston RJ, et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science. 2009;325:1006–1010. doi: 10.1126/science.1175870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Poholek AC, et al. In vivo regulation of Bcl6 and T follicular helper cell development. J Immunol. 2010;185:313–326. doi: 10.4049/jimmunol.0904023. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES