In response to an intracellular infectious agent, the immune system produces a specific cellular response as well as a T cell–dependent Ab response. Precursor T cells differentiate into effector T cells, including Th1 cells, and T follicular helper (TFH) cells. The latter cooperate with B cells to form germinal centers and induce the formation of Ab-forming plasmacytes. One major focal point for control of T cell differentiation is the transcription factor BCL6. In this study, we demonstrated that the Bcl6 gene is regulated by FOXO1-binding, cis-acting sequences located in a highly conserved region of the first Bcl6 intron. In both mouse and human T cells, deletion of the tandem FOXO1 binding sites increased the expression of BCL6 and enhanced the proportion of TFH cells. These results reveal a fundamental control point for cellular versus humoral immunity.

The Bcl6 gene encodes a member of the broad complex, tramtrack, and bric-à-brac–zinc finger family of proteins. As a transcriptional repressor able to recruit corepressors (1), it plays an essential role in the germinal center (GC) reaction for both B and T cells (25), and thus constitutes a pivotal control point in adaptive immunity. Consistent with this, Bcl6 is expressed at increased levels in T follicular helper (TFH) cells as compared with T effector (TEF) cells, and this is manifest in the enhancement of H3K27 acetylation in a nonpromoter gene element located at the extreme upstream region of the first Bcl6 intron (6).

In a previous study, we identified tandem FOXO1 binding sites in the first intron of the Bcl6 gene, and we further showed that deletion of the Foxo1 gene led to enhanced expression of BCL6. This correlated with enhanced TFH cell differentiation (CXCR5hiBCL6lo), but a profound loss of GC TFH cells (CXCR5hiBCL6hi) (7). Analysis of the Bcl6 intronic region revealed two 9-base consensus forkhead sites located within 28 bases of one another at the point of FOXO1 binding. In our previous study (7), we proposed that FOXO1 acts as a transcriptional corepressor, an activity previously described for FOXO transcription factors despite their strong transcriptional activation domains (8, 9). The notion of FOXO1 mediating repression of Bcl6 is consistent with the observation that a PI3Kδ gain of function in humans and mice, as well as the resulting effects on FOXO1, strongly affects TFH cell abundance and the GC reaction (1013). In B cells, FOXO1 was shown to be necessary for GC dark zone development (14).

To determine whether the effects of FOXO1 regulation of TFH cell differentiation were all or in part due to its direct effects on Bcl6 gene expression, we sought to carry out deletions of the FOXO1 binding sites in the Bcl6 gene in mouse embryos. In this study, we describe the differentiation of TFH and GC-TFH cells from two strains of mice harboring selective deletions of the two forkhead binding sites and show that BCL6 expression is increased, resulting in an enhanced GC response. This mechanism of control was found to be conserved because deletion of the very same sequence in cultured human T cells similarly enhanced TFH cell differentiation.

To delete FOXO1 binding sites in the first intron of the Bcl6 gene in mice, CRISPR/Cas9 technology was carried out in C57BL/6J zygotes. Briefly, two single guide RNAs (sgRNAs), sgRNA6 (5′-ATAATGATCATGAGCAGCGG-3′) and sgRNA11 (5′-GCAGCAACAGCAATAATCACC-3′), were designed to target at the flanking sequences of FOXO1 binding sites in the Bcl6 intronic region. Pronuclear microinjection was carried out using sgRNA6, sgRNA11, and Cas9 mRNA with or without a DNA oligonucleotide to mediate homology-directed repair that would selectively delete the two FOXO1 binding sites. The preparation of sgRNA for pronuclei injection was carried out using in vitro transcription synthesis. The T7 promoter sgRNA scaffold template was produced by PCR (15). The sgRNAs were synthesized and purified by a MEGAshortscript kit (Ambion) and MEGAclear kit purification for large-scale transcription reactions (Ambion), respectively.

All procedures were carried out by the University of California, San Diego (UCSD) transgenic shared resource at the UCSD Moores Cancer Center. Two independent founders, IR18 and IR4, collectively referred to as Bcl6ΔFKHD, were selected for further study. The differential length and sequence of PCR product confirmed deletion of the Bcl6 locus. IR18 and IR4 mice were crossed to include the P14 TCR transgene, SMARTA TCR transgene, or OT-II TCR transgene. Congenic CD45.1, CD45.1/CD45.2, Foxo1AAA (7), and CD4Cre-ERT2 (16) mice were bred in our animal facility. CD4Cre-ERT2–mediated activation of Foxo1AAA alleles was induced by i.p. injection of 2 mg of tamoxifen (Cayman Chemical) emulsified in 200 μl of sunflower seed oil (Sigma-Aldrich) for 5 constitutive days, followed by 3 d of rest. Bcl6ΔFKHD mice were crossed with TCRα−/− mice to generate Bcl6ΔFKHD TCRα−/− mice. All mouse studies were carried out in a specific pathogen-free facility at the UCSD vivarium and the experiments were performed according to the Institutional Animal Care and Use Committee of the UCSD.

Bcl6 mRNA amounts were determined by quantitative RT-PCR using forward primer (5′-TGAGCAGTTTAGAGCCCATAAG-3′) and reverse primer (5′-GTACATGAAGTCCAGGAGGATG-3′).

The desired amount of citrated sheep blood (Colorado Serum) was washed three times with PBS by centrifugation at 930 × g (2000 rpm) for 10 min at 4°C. The SRBC pellet was resuspended at the concentration of 1010 cells/ml. Then, 2 × 109 SRBCs per mouse were injected i.p.

Splenocytes from OT-II TCR mice were obtained by mashing total spleens through a 70-μm nylon cell strainer (BD Biosciences), and the OT-II T cells were purified by an EasySep mouse naive CD4+ T cell isolation kit (STEMCELL Technologies). The purity of OT-II T cells was verified by FACS, and 105 OT-II cells/mouse were adoptively transferred. The following day, mice were infected i.v. with 105 PFU of vesicular stomatitis virus (VSV)–OVA (gift from Dr. Ananda Goldrath). For lymphocytic choriomeningitis virus (LCMV) infection experiments, the P14 or SMARTA T cells were purified by an EasySep mouse naive CD8+ T cell isolation kit or naive CD4+ T cell isolation kit (STEMCELL Technologies), respectively. The purity of P14 or SMARTA cells was verified by FACS, and these cells were adoptively transferred into congenic mice. Frozen stocks of LCMV Armstrong and clone-13 were diluted in PBS, and 2 × 105 PFU of LCMV Armstrong was injected i.p. or 2 × 106 PFU of LCMV clone-13 was injected i.v.

Spleens were homogenized and passed through a nylon cell strainer in HBSS (Life Technologies) supplemented with 2% FBS. Surface staining was performed for 30 min at 4°C in PBS supplemented with 2% FCS (FACS buffer) using the following Abs: CD4 (GK1.5), CD8α (53-6.7), CD38 (90), FAS (15A7), GL-7, SLAM (TC15-12F12.2), CD62L (MEL-14), CD8 (53-5.8), CD45.1 (A20), CD45.2 (104), CD44 (IM7), and PD-1 (J43) (BioLegend, San Diego, CA). CXCR5 staining was carried out as described (17) using anti-CXCR5 (2G8; BD Biosciences, San Jose, CA). Each cell-staining reaction was preceded by a 15-min incubation with a purified anti-mouse CD16/32 Ab. Gp33 and Gp66 tetramers were made and provided by the National Institutes of Health tetramer core facilities (Atlanta, GA). Gp66 tetramer staining was performed at 37°C for 90 min. Intracellular transcription factor staining was performed with a FOXP3 staining kit (eBioscience) and stained with Abs specific for TCF7 (C63D9; Cell Signaling) or BCL6 (K112-91; BD Biosciences).

Bone marrow cells were isolated from TCRα−/−, Bcl6ΔFKHD TCRα−/− and wild-type (WT) CD45.1/CD45.2 mice and processed under sterile conditions. A single-cell suspension in PBS was obtained with a 1:1 ratio of TCRα−/− and WT or Bcl6ΔFKHD TCRα−/− and WT mice bone marrow cells. Cells (5 × 106) were injected i.v. into lethally irradiated (10 Gy) CD45.1 hosts in a volume of 200 μl. A second model of bone marrow transplantation was obtained by injecting a 1:1 ratio of WT (CD45.1) and IR18 (CD45.2) bone marrow cells into lethally irradiated (10 Gy) TCRα−/− hosts in a volume of 200 μl. Radiation chimeras received autoclaved water treated with antibiotics (trimethoprim-sulfamethoxazole) until 4 wk after injection.

Human blood was obtained from the Stanford Blood Center. The CD3+ human T cells were isolated using a RosetteSep human T cell enrichment cocktail kit (STEMCELL Technologies) according to the vendor’s manual. Surface staining was performed on enriched samples for 15 min at room temperature in HBSS supplemented with 2% FBS using the following Abs: CD4 (RPA-T4), CD8 (RPA-T8), PD-1 (EH12.2H7), CD45RO (UCHL1), and CD45RA (HI100) (BioLegend). For live/dead discrimination, Zombie NIR dye was used (BioLegend). The purity of CD3 T cells was checked by FACS and reached to 90–98%. T cells were activated with Ab-coated beads specific for CD3, CD28, and CD2 (STEMCELL Technologies) with the addition of IL-2. At 2 d postactivation cells were electroporated using a Neon transfection system under the following conditions (1600 V, 10 ms, and three pulses) to incorporate the empty vector (pSpCas9(BB)-2A-GFP [PX458], Addgene) or two PX458 vectors, with one expressing sgRNA6 and the other expressing shRNA11. The cells were then cultured for 5 d under conditions that promote TFH cell differentiation (IL-12 at 5 ng/ml, TGF-β at 1 ng/ml, and activin A at 50 ng/ml) (18). CD4+ GFP+ T cells were FACS sorted and stained for CD4, CXCR5, PD-1, and BCL6.

Flow cytometry measurements of cells were performed on a BD LSRFortessa or BD LSRFortessa X-20 cytometer, and cell sorting was performed on BD FACSAria III. All FACS data were analyzed using FlowJo 10 (Tree Star). Prism 6 software (GraphPad Software) was used to make and to analyze data by a two-tailed unpaired or paired Student t test. Significance was defined as follows: *p < 0.05, ** p < 0.01, ***p < 0.001, and ****p < 0.0001. Data are presented as means ± SEM.

The Bcl6 gene consists of 10 exons with the first 2 noncoding exons separated by a long intron (Fig. 1A). The first exon and the upstream 270 bp of the first intron of Bcl6 constitute a region that is notable for sequence conservation in placental mammals (Supplemental Fig. 1). It is often a site of translocations, deletions, and mutations (kataegis) resulting from somatic hypermutation found in normal GC B cells and GC-derived B cell lymphomas (1922). This region also constitutes a superenhancer with convergent anti-sense transcription (human LOC106146153) and dense H3K27 modifications that attract activation-induced cytidine deaminase–mediated mutations and translocations (2227) (Fig. 1B, Supplemental Fig. 1).

FIGURE 1.

Generation of Bcl6ΔFKHD mice. (A) Putative Foxo1 binding sites and sequence located in Bcl6 intron 1. (B) The binding pattern of FOXO1 in the Bcl6 intron 1 region based on previous result of FOXO1-specific chromatin immunoprecipitation sequencing data. The bottom track depicts the sequence conservation of FOXO1 binding sites. The upstream nucleotide sequence of the first Bcl6 exon in: C, chimpanzee; D, dog; H, horse; Hu, human; M, mouse; R, rat; Rh, rhesus macaque. (C) The DNA sequences from two strains resulting form CAS9-sgRNA–mediated genetic manipulation of C57BL/6 mouse embryos are depicted. Strain IR4 was produced with CAS9 and two sgRNAs (mit-6 and mit-11), whereas IR18 was produced by injecting the same sgRNAs along with a template designed to precisely excise the forkhead binding sites. The arrowheads point to the sites adjacent to the PAM sites.

FIGURE 1.

Generation of Bcl6ΔFKHD mice. (A) Putative Foxo1 binding sites and sequence located in Bcl6 intron 1. (B) The binding pattern of FOXO1 in the Bcl6 intron 1 region based on previous result of FOXO1-specific chromatin immunoprecipitation sequencing data. The bottom track depicts the sequence conservation of FOXO1 binding sites. The upstream nucleotide sequence of the first Bcl6 exon in: C, chimpanzee; D, dog; H, horse; Hu, human; M, mouse; R, rat; Rh, rhesus macaque. (C) The DNA sequences from two strains resulting form CAS9-sgRNA–mediated genetic manipulation of C57BL/6 mouse embryos are depicted. Strain IR4 was produced with CAS9 and two sgRNAs (mit-6 and mit-11), whereas IR18 was produced by injecting the same sgRNAs along with a template designed to precisely excise the forkhead binding sites. The arrowheads point to the sites adjacent to the PAM sites.

Close modal

To target the forkhead sites bound by FOXO1 (Fig. 1B), candidate flanking Cas9-sgRNAs were validated in fibroblasts in culture followed by DNA sequencing. Two of the sgRNAs flanking the FOXO1 binding sites (sgRNA mit-6 and sgRNA mit-11) were effective in producing indels when used separately and ∼72-bp deletions when used in conjunction (data not shown). These two sgRNA sequences were transcribed in vitro for injection into C57BL/6J single-cell mouse embryos along with mRNA encoding CAS9. Some embryos were also injected with a ssDNA template homologous to the region separating the sgRNA cut sites but with the 9-bp deletions corresponding precisely with the two forkhead consensus sequences.

From a number of mouse strains derived from this procedure (Bcl6ΔFKHD), two were chosen for analysis. Bcl6IR4 has a 78-bp deletion corresponding with imprecise nonhomologous end-joining of the two sgRNA cut sites, and Bcl6IR18 represents a homology-directed repair event similar but not identical to the ssDNA template (Fig. 1C). These alleles were transmitted at a Mendelian frequency and the mice showed no obvious phenotypic effects. For most experiments, heterozygous strains were analyzed.

To test the phenotype of Bcl6ΔFKHD strains, heterozygous mice were immunized with SRBCs, and the splenic T and B cells were monitored after 7 d. Spleen cells were gated for CD4+CD44hi cells and analyzed for a TFH phenotype based on the expression of PD-1 or BCL6 versus CXCR5. As shown, both strains displayed a 2-fold increase in the percentage and total number of GC TFH cells (CXCR5hiPD-1hi or CXCR5hiBCL6hi) (Fig. 2A, Supplemental Fig. 2A). In addition, BCL6 expression (mean fluorescence intensity [MFI]) in T cells from the Bcl6ΔFKHD strains was increased by ∼30% over the WT (Fig. 2B). A similar phenotype was seen for the number GC B cells characterized by expression of either FAS+GL-7+ or CD38loBCL6+ (Fig. 2C, Supplemental Fig. 2B), and the amount of BCL6 expressed by GC B cells (peak MFI) was increased by ∼22% (Fig. 2D). These increases were not seen preimmunization (Supplemental Fig. 2C, 2D). In initial experiments, the Bcl6IR18 mice homozygous displayed a phenotype that was further exaggerated compared with heterozygous Bcl6ΔFKHD mice (Supplemental Fig. 2D, left); however, upon testing more mice, this difference was not consistently found (Supplemental Fig. 2D, right). The increase in BCL6 protein expression was reflected in an increase in Bcl6 mRNA expression (Supplemental Fig. 2E).

FIGURE 2.

The GC reaction is enhanced in Bcl6ΔFKHD mice after SRBC immunization. WT or Bcl6ΔFKHD mice (Bcl6IR18 and Bcl6IR4) were immunized with SRBCs and the splenocytes were harvested and analyzed at day 7 postimmunization. (A) Example of flow cytometric analysis of TFH cell differentiation based on the percentages of CXCR5+PD1+ (top) or CXCR5lo, CXCR5int, and CXCR5hi BCL6+ cells (bottom) gated on CD4+CD44hi cells. Individual mice were plotted for the percentage of TFH (CXCR5+PD-1+) or GC TFH cells (CXCR5hiBCL6+) (right). (B) Histograms of BCL6 expression from WT and Bcl6ΔFKHD mouse lines. (C) The percentages of splenic B cells expressing GL7+Fas+ (top) or CD38loBCL6+ (bottom) were analyzed by flow cytometry, and individual mice were plotted for the percentage of each population (right). (D) A flow cytometry histogram plot showing the MFI of the BCL6+ splenic B cells was determined for GL7+Fas+B220+ from WT or Bcl6IR18 and Bcl6IR4 B cells at 7 d postimmunization. Follicular B cells were also analyzed as BCL6+CD38 cells. Data are from one experiment, representative of three individual experiments. ***p < 0.001 by Student t test.

FIGURE 2.

The GC reaction is enhanced in Bcl6ΔFKHD mice after SRBC immunization. WT or Bcl6ΔFKHD mice (Bcl6IR18 and Bcl6IR4) were immunized with SRBCs and the splenocytes were harvested and analyzed at day 7 postimmunization. (A) Example of flow cytometric analysis of TFH cell differentiation based on the percentages of CXCR5+PD1+ (top) or CXCR5lo, CXCR5int, and CXCR5hi BCL6+ cells (bottom) gated on CD4+CD44hi cells. Individual mice were plotted for the percentage of TFH (CXCR5+PD-1+) or GC TFH cells (CXCR5hiBCL6+) (right). (B) Histograms of BCL6 expression from WT and Bcl6ΔFKHD mouse lines. (C) The percentages of splenic B cells expressing GL7+Fas+ (top) or CD38loBCL6+ (bottom) were analyzed by flow cytometry, and individual mice were plotted for the percentage of each population (right). (D) A flow cytometry histogram plot showing the MFI of the BCL6+ splenic B cells was determined for GL7+Fas+B220+ from WT or Bcl6IR18 and Bcl6IR4 B cells at 7 d postimmunization. Follicular B cells were also analyzed as BCL6+CD38 cells. Data are from one experiment, representative of three individual experiments. ***p < 0.001 by Student t test.

Close modal

To characterize the phenotype of a Bcl6ΔFKHD deletion in the response to a pathogen, mixed bone marrow chimeras were set up in which bone marrow cells from WT and Bcl6IR18 mice were used to reconstitute lethally irradiated TCRα−/− mice. Eight weeks following reconstitution, mice were infected with LCMV clone 13 virus and analyzed for tetramer-positive (GP66-specific) T cells 8, 29, and 56 d later (Fig. 3A). As shown, surprisingly, there was no difference in the proportion of WT compared with Bcl6IR18 GP66-specific GC TFH cells at day 8, and yet, at later times there was a consistent difference comparing WT and Bcl6IR18 GC TFH cells within individual animals (Fig. 3B). Further analysis at day 56 revealed that within the same animal, the numbers of GC TFH (GP66+CXCR5+BCL6+) cells and total GP66+ T cells were significantly increased, and the GC TFH cells present expressed a higher amount of BCL6 as revealed by the MFI (Fig. 3C).

FIGURE 3.

Deletion of FOXO1 binding sites in Bcl6 intron 1 causes cell-intrinsic enhancement in TFH cell differentiation in the late phase of LCMV-c13 infection. (A) WT and Bcl6IR18 bone marrow cells were cotransferred into irradiated TCRα−/− hosts, reconstituted for 8 wk, and infected with LCMV-c13. The spleen cells were harvested and analyzed at the indicated time points cpostinfection. (B) Plots show the percentage of CXCR5hiBCL6+gp66+ T cells originating from WT and Bcl6IR18 bone marrow at various days postinfection. (C) Representative plot of BCL6 versus CXCR5 at day 56 along with the number of gp66+CXCR5+BCL6+T cells, the number of total gp66+ T cells, or the MFI of BCL6 for each paired cell population from each mouse. Data from one experiment representative of two individual experiments. *p < 0.05, **p < 0.01, ***p < 0.001 by Student t test.

FIGURE 3.

Deletion of FOXO1 binding sites in Bcl6 intron 1 causes cell-intrinsic enhancement in TFH cell differentiation in the late phase of LCMV-c13 infection. (A) WT and Bcl6IR18 bone marrow cells were cotransferred into irradiated TCRα−/− hosts, reconstituted for 8 wk, and infected with LCMV-c13. The spleen cells were harvested and analyzed at the indicated time points cpostinfection. (B) Plots show the percentage of CXCR5hiBCL6+gp66+ T cells originating from WT and Bcl6IR18 bone marrow at various days postinfection. (C) Representative plot of BCL6 versus CXCR5 at day 56 along with the number of gp66+CXCR5+BCL6+T cells, the number of total gp66+ T cells, or the MFI of BCL6 for each paired cell population from each mouse. Data from one experiment representative of two individual experiments. *p < 0.05, **p < 0.01, ***p < 0.001 by Student t test.

Close modal

The process of GC formation involves the comigration of T cells and B cells to the cortical/follicular boundary where B cells present Ag to developing TFH cells in an exchange that promotes further differentiation of both cell subsets. Thus, an enhancement of either cell type might be predicted to affect the GC reaction and the expansion of both GC T and B cells. In order to determine whether the effects are T cell specific, WT SMARTA or Bcl6IR18 SMARTA T cells were transferred into C57BL/6 hosts that were then infected with an acutely infectious (LCMV Armstrong) virus (Fig. 4A). Mice were then analyzed at day 9, a time when the virus has been cleared (28).

FIGURE 4.

Bcl6IR18 CD4 T cells specific for gp66 are enhanced upon acute or chronic infection. (A) WT or Bcl6IR18 SMARTA cells were transferred into CD45.1 hosts, infected with LCMV Armstrong, and the spleen cells were analyzed at day 9 postimmunization. (B) Flow cytometric analysis of TFH and GC TFH cell differentiation based on CXCR5loBCL6 and CXCR5hiBCL6+ (top) or CXCR5+SLAM (bottom) at day 9 gated on CD4+CD44hi cells and graphed for the percentage of GC TFH cell (CXCR5hiBCL6+), TFH cell (CXCR5loBCL6), and CXCR5+SLAM expression. The MFI of BCL6 for CXCR5hiBCL6+ SMARTA cells is shown (right). Data from one experiment are representative of two individual experiments. (C) Model of adoptive cotransfer experiment and infection with the persistent LCMV-c13. WT (CD45.2) and Bcl6IR18 SMARTA (CD45.1/45.2) cells were transferred into CD45.1 hosts, infected with LCMV-c13, and spleen cells were analyzed on days 9 and 29 postimmunization. (D) Splenocytes were harvested at indicated time points after infection. Cytometric analysis similar to (A) comparing WT and Bcl6IR18 SMARTA cells in the same host at day 9 (left) and day 29 postinfection (right). The percentage of CXCR5hiBCL6+ cells, the MFI of BCL6 expression, and the percentage of SMARTA cells are graphed (bottom). Data are from one experiment representative of two individual experiments. *p < 0.05, **p < 0.01, ***p < 0.001 by Student t test.

FIGURE 4.

Bcl6IR18 CD4 T cells specific for gp66 are enhanced upon acute or chronic infection. (A) WT or Bcl6IR18 SMARTA cells were transferred into CD45.1 hosts, infected with LCMV Armstrong, and the spleen cells were analyzed at day 9 postimmunization. (B) Flow cytometric analysis of TFH and GC TFH cell differentiation based on CXCR5loBCL6 and CXCR5hiBCL6+ (top) or CXCR5+SLAM (bottom) at day 9 gated on CD4+CD44hi cells and graphed for the percentage of GC TFH cell (CXCR5hiBCL6+), TFH cell (CXCR5loBCL6), and CXCR5+SLAM expression. The MFI of BCL6 for CXCR5hiBCL6+ SMARTA cells is shown (right). Data from one experiment are representative of two individual experiments. (C) Model of adoptive cotransfer experiment and infection with the persistent LCMV-c13. WT (CD45.2) and Bcl6IR18 SMARTA (CD45.1/45.2) cells were transferred into CD45.1 hosts, infected with LCMV-c13, and spleen cells were analyzed on days 9 and 29 postimmunization. (D) Splenocytes were harvested at indicated time points after infection. Cytometric analysis similar to (A) comparing WT and Bcl6IR18 SMARTA cells in the same host at day 9 (left) and day 29 postinfection (right). The percentage of CXCR5hiBCL6+ cells, the MFI of BCL6 expression, and the percentage of SMARTA cells are graphed (bottom). Data are from one experiment representative of two individual experiments. *p < 0.05, **p < 0.01, ***p < 0.001 by Student t test.

Close modal

The analysis of TFH cells by means of BCL6 or SLAM expression versus CXCR5 revealed an increased proportion of Bcl6IR18 TFH cells as compared with WT at day 9 postinfection (Fig. 4B). In this experiment, there was a trend toward increased proportions of GC TFH cells (CXCR5hiBCL6+) when comparing WT and Bcl6IR18 T cells, whereas the increase in the TFH cell compartment as measured by either CXCR5+ (upper panel) or CXCR5+SLAM expression (lower panel) achieved significance. Furthermore, the amount of BCL6 expressed per cell was increased in T cells from Bcl6IR18 mice. These results are consistent with an intrinsic effect of the Bcl6ΔFKHD allele on the transcription of the Bcl6 locus and the differentiation of TFH cells.

Similarly, the T cell–intrinsic phenotypic effects of a Bcl6ΔFKHD allele were analyzed in response to a chronic viral infection (Fig. 4C). In this experiment, genetically distinct WT and Bcl6IR18 T cells were transferred followed by infection with LCMV clone 13. At days 9 and 29 postinfection, there was a significant increase in the proportion of GC TFH cells among SMARTA T cells from Bcl6IR18 mice compared with T cells from WT mice (Fig. 4D). Again, the amount of BCL6 expressed by cells within this subset was also increased. In contrast, the proportions of total transferred SMARTA T cells did not differ between Bcl6IR18 and WT T cells at either day 9 or 29; why this result at day 29 differs from the expansion found in bone marrow chimeras at day 56 (Fig. 3) is discussed. These results are consistent with an enhanced T cell–intrinsic TFH cell differentiation in Bcl6ΔFKHD mice under conditions of equivalent expansion.

To determine whether there was also a B cell–intrinsic component to the Bcl6ΔFKHD allele, mixed bone marrow radiation chimeras were set up such that the expansion of WT or Bcl6IR18 B cells could be compared in the presence of WT T cells (Fig. 5A). After 8 wk, the mice were infected with VSV-OVA and GC B cells were analyzed as above. Perhaps surprisingly, there was no difference in the proportion of GC B cells (out of total B cells) when comparing WT to Bcl6IR18 B cells, and this was found at 8 and 15 d postinfection (Fig. 5B, 5C). We conclude that, although GC B cells from immunized Bcl6ΔFKHD mice express an increased amount of BCL6, this increase may originate from enhanced TFH cell maturation and the signals thereby provided.

FIGURE 5.

Effect of Bcl6ΔFKHD allele is not B cell intrinsic. (A) Mixed bone marrow chimeras were produced such that WT or Bcl6IR18 B cells could be compared in the presence of WT T cells. (B and C) Mice were infected with VSV-OVA and the GC B cells were compared. An exemplary flow cytometric plot is shown along with the percentage and total number of WT or Bcl6IR18 B cells on day 8 (B) or day 15 (C). Data are from one experiment representative of two individual experiments. None of the comparisons showed significance as measured by a Student t test.

FIGURE 5.

Effect of Bcl6ΔFKHD allele is not B cell intrinsic. (A) Mixed bone marrow chimeras were produced such that WT or Bcl6IR18 B cells could be compared in the presence of WT T cells. (B and C) Mice were infected with VSV-OVA and the GC B cells were compared. An exemplary flow cytometric plot is shown along with the percentage and total number of WT or Bcl6IR18 B cells on day 8 (B) or day 15 (C). Data are from one experiment representative of two individual experiments. None of the comparisons showed significance as measured by a Student t test.

Close modal

The deletion of Foxo1 results in increased Bcl6 mRNA and BCL6 protein expression in TFH cells (7), and deletion of the only FOXO1 binding sites in the Bcl6 gene also enhances BCL6 expression (Figs. 3B, 4B, 4D). We also previously showed that expression of a Foxo1 triple phosphorylation mutant (Foxo1AAA) that results in the expression of a constitutively nuclear form of FOXO1 prevents TFH cell differentiation (7). A prediction is that the loss of TFH cells resulting from Foxo1AAA expression might be rescued by the Bcl6ΔFKHD allele; if the effect of constitutively active FOXO1 is through suppression of Bcl6 expression, then the Bcl6ΔFKHD allele should be resistant to the suppression.

To test this possibility, T cells from OT-II, OT-II Foxo1AAA, or OT-II Bcl6IR18Foxo1AAA mice were transferred into naive C57BL/6 recipients that were subsequently infected with VSV-OVA. The Ag-specific CD4 T cells were analyzed 8 d later (Fig. 6A). WT OVA-specific T cells were predominantly TFH cells (CXCR5+BCL6low) (Fig. 6B, left panel, 6C, middle panel, average 68%). With the addition of constitutively nuclear FOXO1 (Foxo1AAA allele), most T cells instead assumed an effector phenotype (CXCR5) (Fig. 6B, middle panel, 6C, left panel, average 79%). With the further addition of a hemizygous Bcl6IR18 allele, the differentiation to a TFH cells phenotype was partially rescued such that the proportions of TEF and TFH cells were nearly equivalent (Fig. 6B, right panel, 6C). This result is consistent with the notion that FOXO1 mediates Bcl6 repression; however, in the presence of even a haploid Bcl6ΔFKHD allele, TFH cell differentiation was partially restored.

FIGURE 6.

The blockage of TFH cell differentiation caused by Foxo1AAA can be partially restored by addition of a Bcl6IR18 allele. (A) The strategy used to measure the TFH cell differentiation of WT or Bcl6IR18 OT-II cells with or without enforced nuclear expression of FOXO1 after infection with VSV-OVA. OT-II cells isolated from CD4CreER Foxo1AAA WT or Bcl6IR18 mice, which were pretreated by tamoxifen for 5 d, were transferred into congenic mice and infected with VSV-OVA. The splenocytes were harvested and analyzed at day 8 postinfection. (B and C) Plots show the percentage of CXCR5BCL6, CXCR5loBCL6, and CXCR5hiBCL6+ populations of OT-II cells from CD4creERFoxo1AAA WT, CD4creER+ Foxo1AAA WT, and CD4creER+ Foxo1AAABcl6IR18 mice at day 8 postinfection (top). The percentage of indicated splenic populations (CXCR5BCL6, CXCR5loBCL6, and CXCR5hiBCL6+) are depicted for each mouse. Data are from one experiment representative of two individual experiments. ***p < 0.001, ****p < 0.0001 by Student t test.

FIGURE 6.

The blockage of TFH cell differentiation caused by Foxo1AAA can be partially restored by addition of a Bcl6IR18 allele. (A) The strategy used to measure the TFH cell differentiation of WT or Bcl6IR18 OT-II cells with or without enforced nuclear expression of FOXO1 after infection with VSV-OVA. OT-II cells isolated from CD4CreER Foxo1AAA WT or Bcl6IR18 mice, which were pretreated by tamoxifen for 5 d, were transferred into congenic mice and infected with VSV-OVA. The splenocytes were harvested and analyzed at day 8 postinfection. (B and C) Plots show the percentage of CXCR5BCL6, CXCR5loBCL6, and CXCR5hiBCL6+ populations of OT-II cells from CD4creERFoxo1AAA WT, CD4creER+ Foxo1AAA WT, and CD4creER+ Foxo1AAABcl6IR18 mice at day 8 postinfection (top). The percentage of indicated splenic populations (CXCR5BCL6, CXCR5loBCL6, and CXCR5hiBCL6+) are depicted for each mouse. Data are from one experiment representative of two individual experiments. ***p < 0.001, ****p < 0.0001 by Student t test.

Close modal

To further study BCL6 regulation, we sought to determine whether human TFH cells are also regulated by these conserved forkhead consensus sites found in the human BCL6 locus. As shown in (Fig. 1B, the mouse and human regions of interest are identical such that the same sgRNA sequences, that is, sgRNA mit-6 and sgRNA mit-11, that were used for deletion of the mouse locus in fertilized embryos could be used for human T cells. To carry out the deletion, we cloned each of the 21-bp CRISPR RNA (crRNA) targeting sequences individually into the PX458 vector such that the human U6 promoter will transcribe a composite scaffold-guide RNA targeting sequence flanking the forkhead binding sites. The PX458 vector also encodes CAS9 and enhanced GFP separated by a T2A self-splicing sequence and driven by a hybrid CMV-chicken β-actin promoter (29). CD3+ T cells from fresh human blood were stimulated for 2 d with Abs specific for CD3, CD28, and CD2 and subjected to electroporation to deliver guide RNA and Cas9-coding plasmids (Fig. 7A). The experimental cells were electroporated with two vectors, one expressing sgRNA mit6-11 and the other sgRNA mit11-10. As a control, T cells were electroporated with the PX458 plasmid expressing the trans-activating crRNA without a crRNA recognition sequence. The cells were further cultured for 5 d in the presence of IL-12, TGF-β, and activin A (18) and sorted for enhanced GFP fluorescence (Fig. 7A).

FIGURE 7.

Deletion of FOXO1 binding sites within the Bcl6 intron in human primary T cells results in an increased CXCR5hiBcl6+ population. (A) Strategy for purifying human primary T cells and transfecting with a plasmid with or without the crRNA sequences flanking the human/mouse tandem forkhead sites. (B) The PCR products from human T cells transfected with or without sgRNA6/11 analyzed by electrophoresis. (C) The results of sequencing the PCR products are plotted as a histogram showing the number of sequences with specific nucleotide deletions. (D) The phenotype of T cells following differentiation in culture. The plot represents that the percentage of CXCR5hiBCL6+ or CXCR5hiPD-1+ populations within GFP+CD4+ T cells. The BCL6 and CXCR5 profiles are also shown as histograms (bottom). These profiles depict the results of one experiment out of a total of three.

FIGURE 7.

Deletion of FOXO1 binding sites within the Bcl6 intron in human primary T cells results in an increased CXCR5hiBcl6+ population. (A) Strategy for purifying human primary T cells and transfecting with a plasmid with or without the crRNA sequences flanking the human/mouse tandem forkhead sites. (B) The PCR products from human T cells transfected with or without sgRNA6/11 analyzed by electrophoresis. (C) The results of sequencing the PCR products are plotted as a histogram showing the number of sequences with specific nucleotide deletions. (D) The phenotype of T cells following differentiation in culture. The plot represents that the percentage of CXCR5hiBCL6+ or CXCR5hiPD-1+ populations within GFP+CD4+ T cells. The BCL6 and CXCR5 profiles are also shown as histograms (bottom). These profiles depict the results of one experiment out of a total of three.

Close modal

The analysis of these T cells showed that, for a typical experiment, 32% of the BCL6 alleles were deleted (Fig. 7B). The PCR products were further subjected to sequencing, and this revealed substantial deletions around each of the target sequences (Fig. 7C). The analysis of phenotype further revealed a strong increase in the percentage of CXCR5hiBCL6hi or CXCR5hiPD-1hi T cells (Fig. 7D).

Large-scale projects to decode the cis-acting sequences that control gene expression have revealed some of the complexity and redundancy of gene elements involved in, for example, tissue specification during development (3032) or housekeeping functions in mammalian cell lines (33). One of the overriding themes in much of this work is robustness of gene expression and its resistance to perturbation based on short indels or even tiled deletions that encompass up to 1000 bases. More precisely, analyses indicate the existence of widespread enhancer redundancy or the concept of shadow enhancers, that is, two or more enhancers that promote activation of gene transcription in a way that renders deletion of any one enhancer phenotypically silent. Upon this background, deletion of a factor binding site downstream of the transcription start site might not be anticipated to affect expression or cellular differentiation; however, when deletion does result in a change in gene expression, it may identify such an element as being of central importance.

One question is how this type of redundancy extends to enhancers marked by abundant H3K27 acetylation and bound by p300, BRD4, and mediator, defined as superenhancers (34, 35). Such enhancers are essential for the expression of genes that are often lineage defining or key to lineage commitment (25). They attract a diverse array of transcription factors, and recent work links their activity with the formation of phase-separated condensates produced by interaction between the intrinsically disordered domains of BRD4, mediator (MED1), and transcription factor trans-activation domains. A notion is that superenhancers may be excluded from the concept of redundancy, as they appear to be highly sensitive to perturbation (36), and, at least for GC B cells, this applies to the regulatory region located at the first Bcl6 exon and the 5′ region of the first intron (22, 37). In fact, based on the density of H3K27Ac marks listed in Encode, the region surrounding the first noncoding exon of Bcl6 appears to be a superenhancer in several unrelated cell types. A second question is whether there is redundancy of repressive elements, and exactly how repressive elements act mechanistically.

Although we have not attempted to address these complex questions directly, we extended our observations on the role of FOXO1 in TFH cell differentiation, affirming that the FOXO1 tandem binding sites, mapped by chromatin immunoprecipitation sequencing and unique within a megabase of the Bcl6 gene, mediate nonredundant repressive activity that impacts the differentiation of T cells. Thus, loss of FOXO1 binding to the Bcl6 gene enhanced GC TFH cell differentiation, whereas constitutively nuclear FOXO1 blocked TFH and GC TFH cell differentiation. For example, following infection with VSV-OVA, the average proportion of TEF cells averaged ∼18%, but with the addition of the Foxo1AAA allele this number jumped to almost 80%. With the additional Bcl6IR18 allele the TEF cells dropped back to 50%. Likewise, the average proportion of TFH cells was 68% for WT that was reduced to 20% for Foxo1AAA T cells, and this deficiency was partially rescued to 43% with the further addition of a hemizygous Bcl6ΔFKHD allele, an addition that should alleviate FOXO1 binding on one chromosome. A possibility is that Foxo1AAABcl6ΔFKHD/ΔFKHD OT-II mice (with a homozygous Bcl6ΔFKHD allele) might have been completely rescued from the effects of the Foxo1AAA inhibition, but we were not able to test such mice. We conclude that nuclear FOXO1, bound to these specific sites, is an important determining factor in Bcl6 expression and the differentiation of TFH cells, although we cannot conclude that this is the only effect exerted by FOXO1 on TFH or GC TFH cell differentiation.

A complication to understanding this process is that although TFH cell differentiation is inhibited by FOXO1, the differentiation from TFH to the GC TFH cell phenotype actually requires FOXO1 (7). This, in turn, implies that mechanisms of FOXO1 regulation may vary within the time frame of the initial activation of naive T cells through to the establishment of a T cell–replete GC. Consistent with this notion, studies on the activation of T cells showed that FOXO1 is initially sequestered from the nucleus via triple, AKT-mediated phosphorylation, and we deduce that this is required to relieve cell cycle inhibition via p27kip1 (38). As this inhibition subsides, it is replaced by posttranscriptional inhibition mediated by miRNA-182, part of the miR-183–miR-96–miR-182 cluster (39). In the differentiation phase, transition to effector Th17 cells was shown to depend on miR-183 as a third means of reducing Foxo1 mRNA (40).

An unanticipated result was that the increases in GC B cells appeared to be cell extrinsic. In the experiments presented, irradiated mice were reconstituted such that the Bcl6ΔFKHD allele was present in cells other than T cells and then infected with VSV-OVA. Subsequently, we analyzed the total number of GC B cells similar to our initial experiments using SRBCs as an immunogen. We saw neither an increase in the proportion of GC B cells nor an increase in B cell–expressed BCL6. Why there was found to be no difference in BCL6 expression under this protocol is unknown, but the direct implication is that BCL6 expression in GC B cells is affected by the strength of T cell help, and the increased BCL6 expression noted in immunized Bcl6ΔFKHD mice resulted from the enhanced T cell help provided by Bcl6ΔFKHD T cells. T cell help includes CD40L–CD40 interactions and the production cytokines such as IL-4 and IL-21, and these signals can have important effects on the dynamics and magnitude of GC B cell differentiation that can include the expression of BCL6 (41, 42).

Another facet of the requirement for FOXO1 in GC TFH cell differentiation may derive from a requirement for TCF7 and LEF1 acting to repress expression of Prdm1 (Blimp-1) (4346). In CD8+ T cells, TCF7 expression is entirely dependent on FOXO1 postactivation (47, 48). If TCF7 is likewise dependent on FOXO1 in CD4+ T cells, GC TFH cell differentiation would be expected to be FOXO1-dependent. As FOXO1 can have opposing effects at different stages of the process, and it responds to many different extrinsic signals, it is likely to affect T cell differentiation through multiple mechanisms (49).

The Bcl6 gene is a central control point for many aspects of T and B cell differentiation and function (50). It is known to be regulated in T cells by several signaling pathways, including those that result in the activation of STAT3, STAT4, STAT5, TCF7, LEF1, TBET, BCL6, AIOLOS, IKAROS, BOB1, and others (5, 45, 5153). In addition, the locus is regulated by the organization of its genomic architecture (37). STAT5-inducing cytokines, IL-2 and IL-7, have been found to repress Bcl6 and the differentiation of TFH cells (54, 55), whereas STAT3/4-inducing cytokines promote Bcl6 expression and the appearance of TFH cells (51, 5658). In addition, for an individual naive T cell, the proportion of daughter cells that differentiate to become TFH cells versus Th1 or Th17 effector cells in response to infection appears to be determined by the T cell Ag receptor expressed, and experiments show that the strength of signal generated by recognition of dendritic cell–peptide–MHC complexes is the determining factor (59). However, the correlation between dwell time (or affinity) and effector versus TFH cell differentiation appeared to differ depending on the exact interval of affinities being tested (60). In addition, different TCRs gave rise to different amounts of IL-2Rα-chain expression, implying that IL-2 signals constitute a control point for the differentiation of individual clones of T cells. Because FOXO1 responds to TCR ligation and cytokine signals that activate STAT5- and STAT3/4-inducing cytokines, it clearly constitutes at least one integrating principle in TFH versus effector cell differentiation.

This publication is dedicated to A.D. in memoriam.

This work was supported by The Chancellor’s Associates Chair in the Biological Sciences and by National Institutes of Health/National Institute of Allergy and Infectious Diseases Grant R01AI103440 to S.M.H.

Stephen M. Hedrick is a Distinguished Fellow of AAI.

The online version of this article contains supplemental material.

Abbreviations used in this article:

crRNA

CRISPR RNA

GC

germinal center

LCMV

lymphocytic choriomeningitis virus

MFI

mean fluorescence intensity

sgRNA

single guide RNA

TEF

T effector

TFH

T follicular helper

UCSD

University of California, San Diego

VSV

vesicular stomatitis virus

WT

wild-type

1.
Huynh
K. D.
,
V. J.
Bardwell
.
1998
.
The BCL-6 POZ domain and other POZ domains interact with the co-repressors N-CoR and SMRT.
Oncogene
17
:
2473
2484
.
2.
Hale
J. S.
,
R.
Ahmed
.
2015
.
Memory T follicular helper CD4 T cells.
Front. Immunol.
6
:
16
.
3.
Vinuesa
C. G.
,
M. A.
Linterman
,
D.
Yu
,
I. C.
MacLennan
.
2016
.
Follicular helper T cells.
Annu. Rev. Immunol.
34
:
335
368
.
4.
Zhang
Y.
,
L.
Garcia-Ibanez
,
K. M.
Toellner
.
2016
.
Regulation of germinal center B-cell differentiation.
Immunol. Rev.
270
:
8
19
.
5.
Crotty
S.
2019
.
T follicular helper cell biology: a decade of discovery and diseases.
Immunity
50
:
1132
1148
.
6.
Weinstein
J. S.
,
K.
Lezon-Geyda
,
Y.
Maksimova
,
S.
Craft
,
Y.
Zhang
,
M.
Su
,
V. P.
Schulz
,
J.
Craft
,
P. G.
Gallagher
.
2014
.
Global transcriptome analysis and enhancer landscape of human primary T follicular helper and T effector lymphocytes.
Blood
124
:
3719
3729
.
7.
Stone
E. L.
,
M.
Pepper
,
C. D.
Katayama
,
Y. M.
Kerdiles
,
C. Y.
Lai
,
E.
Emslie
,
Y. C.
Lin
,
E.
Yang
,
A. W.
Goldrath
,
M. O.
Li
, et al
2015
.
ICOS coreceptor signaling inactivates the transcription factor FOXO1 to promote Tfh cell differentiation.
Immunity
42
:
239
251
.
8.
van der Vos
K. E.
,
P. J.
Coffer
.
2008
.
FOXO-binding partners: it takes two to tango.
Oncogene
27
:
2289
2299
.
9.
Langlet
F.
,
R. A.
Haeusler
,
D.
Lindén
,
E.
Ericson
,
T.
Norris
,
A.
Johansson
,
J. R.
Cook
,
K.
Aizawa
,
L.
Wang
,
C.
Buettner
,
D.
Accili
.
2017
.
Selective Inhibition of FOXO1 activator/repressor balance modulates hepatic glucose handling.
Cell
171
:
824
835.e18
.
10.
Angulo
I.
,
O.
Vadas
,
F.
Garçon
,
E.
Banham-Hall
,
V.
Plagnol
,
T. R.
Leahy
,
H.
Baxendale
,
T.
Coulter
,
J.
Curtis
,
C.
Wu
, et al
2013
.
Phosphoinositide 3-kinase δ gene mutation predisposes to respiratory infection and airway damage.
Science
342
:
866
871
.
11.
Lucas
C. L.
,
H. S.
Kuehn
,
F.
Zhao
,
J. E.
Niemela
,
E. K.
Deenick
,
U.
Palendira
,
D. T.
Avery
,
L.
Moens
,
J. L.
Cannons
,
M.
Biancalana
, et al
2014
.
Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110δ result in T cell senescence and human immunodeficiency.
Nat. Immunol.
15
:
88
97
.
12.
Crank
M. C.
,
J. K.
Grossman
,
S.
Moir
,
S.
Pittaluga
,
C. M.
Buckner
,
L.
Kardava
,
A.
Agharahimi
,
H.
Meuwissen
,
J.
Stoddard
,
J.
Niemela
, et al
2014
.
Mutations in PIK3CD can cause hyper IgM syndrome (HIGM) associated with increased cancer susceptibility.
J. Clin. Immunol.
34
:
272
276
.
13.
Preite
S.
,
B.
Huang
,
J. L.
Cannons
,
D. B.
McGavern
,
P. L.
Schwartzberg
.
2019
.
PI3K orchestrates T follicular helper cell differentiation in a context dependent manner: implications for autoimmunity.
Front. Immunol.
9
:
3079
.
14.
Dominguez-Sola
D.
,
J.
Kung
,
A. B.
Holmes
,
V. A.
Wells
,
T.
Mo
,
K.
Basso
,
R.
Dalla-Favera
.
2015
.
The FOXO1 transcription factor instructs the germinal center dark zone program.
Immunity
43
:
1064
1074
.
15.
Shao
Y.
,
Y.
Guan
,
L.
Wang
,
Z.
Qiu
,
M.
Liu
,
Y.
Chen
,
L.
Wu
,
Y.
Li
,
X.
Ma
,
M.
Liu
,
D.
Li
.
2014
.
CRISPR/Cas-mediated genome editing in the rat via direct injection of one-cell embryos.
Nat. Protoc.
9
:
2493
2512
.
16.
Aghajani
K.
,
S.
Keerthivasan
,
Y.
Yu
,
F.
Gounari
.
2012
.
Generation of CD4CreER(T2) transgenic mice to study development of peripheral CD4-T-cells.
Genesis
50
:
908
913
.
17.
Poholek
A. C.
,
K.
Hansen
,
S. G.
Hernandez
,
D.
Eto
,
A.
Chandele
,
J. S.
Weinstein
,
X.
Dong
,
J. M.
Odegard
,
S. M.
Kaech
,
A. L.
Dent
, et al
2010
.
In vivo regulation of Bcl6 and T follicular helper cell development.
J. Immunol.
185
:
313
326
.
18.
Locci
M.
,
J. E.
Wu
,
F.
Arumemi
,
Z.
Mikulski
,
C.
Dahlberg
,
A. T.
Miller
,
S.
Crotty
.
2016
.
Activin A programs the differentiation of human TFH cells. [Published erratum appears in 2016 Nat. Immunol. 17: 1235.]
Nat. Immunol.
17
:
976
984
.
19.
Iqbal
J.
,
T. C.
Greiner
,
K.
Patel
,
B. J.
Dave
,
L.
Smith
,
J.
Ji
,
G.
Wright
,
W. G.
Sanger
,
D. L.
Pickering
,
S.
Jain
, et al
Leukemia/Lymphoma Molecular Profiling Project
.
2007
.
Distinctive patterns of BCL6 molecular alterations and their functional consequences in different subgroups of diffuse large B-cell lymphoma.
Leukemia
21
:
2332
2343
.
20.
Saito
M.
,
J.
Gao
,
K.
Basso
,
Y.
Kitagawa
,
P. M.
Smith
,
G.
Bhagat
,
A.
Pernis
,
L.
Pasqualucci
,
R.
Dalla-Favera
.
2007
.
A signaling pathway mediating downregulation of BCL6 in germinal center B cells is blocked by BCL6 gene alterations in B cell lymphoma.
Cancer Cell
12
:
280
292
.
21.
Khodabakhshi
A. H.
,
R. D.
Morin
,
A. P.
Fejes
,
A. J.
Mungall
,
K. L.
Mungall
,
M.
Bolger-Munro
,
N. A.
Johnson
,
J. M.
Connors
,
R. D.
Gascoyne
,
M. A.
Marra
, et al
2012
.
Recurrent targets of aberrant somatic hypermutation in lymphoma.
Oncotarget
3
:
1308
1319
.
22.
Shen
J. C.
,
A. S.
Kamath-Loeb
,
B. F.
Kohrn
,
K. R.
Loeb
,
B. D.
Preston
,
L. A.
Loeb
.
2019
.
A high-resolution landscape of mutations in the BCL6 super-enhancer in normal human B cells.
Proc. Natl. Acad. Sci. USA
116
:
24779
24785
.
23.
Meng
F. L.
,
Z.
Du
,
A.
Federation
,
J.
Hu
,
Q.
Wang
,
K. R.
Kieffer-Kwon
,
R. M.
Meyers
,
C.
Amor
,
C. R.
Wasserman
,
D.
Neuberg
, et al
2014
.
Convergent transcription at intragenic super-enhancers targets AID-initiated genomic instability.
Cell
159
:
1538
1548
.
24.
Qian
J.
,
Q.
Wang
,
M.
Dose
,
N.
Pruett
,
K. R.
Kieffer-Kwon
,
W.
Resch
,
G.
Liang
,
Z.
Tang
,
E.
Mathé
,
C.
Benner
, et al
2014
.
B cell super-enhancers and regulatory clusters recruit AID tumorigenic activity.
Cell
159
:
1524
1537
.
25.
Hnisz
D.
,
B. J.
Abraham
,
T. I.
Lee
,
A.
Lau
,
V.
Saint-André
,
A. A.
Sigova
,
H. A.
Hoke
,
R. A.
Young
.
2013
.
Super-enhancers in the control of cell identity and disease.
Cell
155
:
934
947
.
26.
Heinz
S.
,
C. E.
Romanoski
,
C.
Benner
,
C. K.
Glass
.
2015
.
The selection and function of cell type-specific enhancers.
Nat. Rev. Mol. Cell Biol.
16
:
144
154
.
27.
Saint-André
V.
,
A. J.
Federation
,
C. Y.
Lin
,
B. J.
Abraham
,
J.
Reddy
,
T. I.
Lee
,
J. E.
Bradner
,
R. A.
Young
.
2016
.
Models of human core transcriptional regulatory circuitries.
Genome Res.
26
:
385
396
.
28.
Dutko
F. J.
,
M. B.
Oldstone
.
1983
.
Genomic and biological variation among commonly used lymphocytic choriomeningitis virus strains.
J. Gen. Virol.
64
:
1689
1698
.
29.
Ran
F. A.
,
P. D.
Hsu
,
J.
Wright
,
V.
Agarwala
,
D. A.
Scott
,
F.
Zhang
.
2013
.
Genome engineering using the CRISPR-Cas9 system.
Nat. Protoc.
8
:
2281
2308
.
30.
Cannavò
E.
,
P.
Khoueiry
,
D. A.
Garfield
,
P.
Geeleher
,
T.
Zichner
,
E. H.
Gustafson
,
L.
Ciglar
,
J. O.
Korbel
,
E. E.
Furlong
.
2016
.
Shadow enhancers are pervasive features of developmental regulatory networks.
Curr. Biol.
26
:
38
51
.
31.
Farley
E. K.
,
K. M.
Olson
,
W.
Zhang
,
D. S.
Rokhsar
,
M. S.
Levine
.
2016
.
Syntax compensates for poor binding sites to encode tissue specificity of developmental enhancers.
Proc. Natl. Acad. Sci. USA
113
:
6508
6513
.
32.
Osterwalder
M.
,
I.
Barozzi
,
V.
Tissières
,
Y.
Fukuda-Yuzawa
,
B. J.
Mannion
,
S. Y.
Afzal
,
E. A.
Lee
,
Y.
Zhu
,
I.
Plajzer-Frick
,
C. S.
Pickle
, et al
2018
.
Enhancer redundancy provides phenotypic robustness in mammalian development.
Nature
554
:
239
243
.
33.
Gasperini
M.
,
G. M.
Findlay
,
A.
McKenna
,
J. H.
Milbank
,
C.
Lee
,
M. D.
Zhang
,
D. A.
Cusanovich
,
J.
Shendure
.
2017
.
CRISPR/Cas9-mediated scanning for regulatory elements required for HPRT1 expression via thousands of large, programmed genomic deletions.
Am. J. Hum. Genet.
101
:
192
205
.
34.
Witte
S.
,
A.
Bradley
,
A. J.
Enright
,
S. A.
Muljo
.
2015
.
High-density P300 enhancers control cell state transitions.
BMC Genomics
16
:
903
.
35.
Sabari
B. R.
,
A.
Dall’Agnese
,
A.
Boija
,
I. A.
Klein
,
E. L.
Coffey
,
K.
Shrinivas
,
B. J.
Abraham
,
N. M.
Hannett
,
A. V.
Zamudio
,
J. C.
Manteiga
, et al
2018
.
Coactivator condensation at super-enhancers links phase separation and gene control.
Science
361
:
eaar3958
.
36.
Hnisz
D.
,
K.
Shrinivas
,
R. A.
Young
,
A. K.
Chakraborty
,
P. A.
Sharp
.
2017
.
A phase separation model for transcriptional control.
Cell
169
:
13
23
.
37.
Bunting
K. L.
,
T. D.
Soong
,
R.
Singh
,
Y.
Jiang
,
W.
Béguelin
,
D. W.
Poloway
,
B. L.
Swed
,
K.
Hatzi
,
W.
Reisacher
,
M.
Teater
, et al
2016
.
Multi-tiered reorganization of the genome during B cell affinity maturation anchored by a germinal center-specific locus control region.
Immunity
45
:
497
512
.
38.
Charvet
C.
,
A. J.
Canonigo
,
S.
Bécart
,
U.
Maurer
,
A. V.
Miletic
,
W.
Swat
,
M.
Deckert
,
A.
Altman
.
2006
.
Vav1 promotes T cell cycle progression by linking TCR/CD28 costimulation to FOXO1 and p27kip1 expression.
J. Immunol.
177
:
5024
5031
.
39.
Stittrich
A. B.
,
C.
Haftmann
,
E.
Sgouroudis
,
A. A.
Kühl
,
A. N.
Hegazy
,
I.
Panse
,
R.
Riedel
,
M.
Flossdorf
,
J.
Dong
,
F.
Fuhrmann
, et al
2010
.
The microRNA miR-182 is induced by IL-2 and promotes clonal expansion of activated helper T lymphocytes.
Nat. Immunol.
11
:
1057
1062
.
40.
Ichiyama
K.
,
A.
Gonzalez-Martin
,
B. S.
Kim
,
H. Y.
Jin
,
W.
Jin
,
W.
Xu
,
M.
Sabouri-Ghomi
,
S.
Xu
,
P.
Zheng
,
C.
Xiao
,
C.
Dong
.
2016
.
The microRNA-183-96-182 cluster promotes T helper 17 cell pathogenicity by negatively regulating transcription factor Foxo1 expression.
Immunity
44
:
1284
1298
.
41.
Laman
J. D.
,
E.
Claassen
,
R. J.
Noelle
.
2017
.
Functions of CD40 and its ligand, gp39 (CD40L).
Crit. Rev. Immunol.
37
:
371
420
.
42.
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.
95
:
925
932
.
43.
Wu
T.
,
H. M.
Shin
,
E. A.
Moseman
,
Y.
Ji
,
B.
Huang
,
C.
Harly
,
J. M.
Sen
,
L. J.
Berg
,
L.
Gattinoni
,
D. B.
McGavern
,
P. L.
Schwartzberg
.
2015
.
TCF1 is required for the T follicular helper cell response to viral infection.
Cell Rep.
12
:
2099
2110
.
44.
Choi
Y. S.
,
J. A.
Gullicksrud
,
S.
Xing
,
Z.
Zeng
,
Q.
Shan
,
F.
Li
,
P. E.
Love
,
W.
Peng
,
H. H.
Xue
,
S.
Crotty
.
2015
.
LEF-1 and TCF-1 orchestrate TFH differentiation by regulating differentiation circuits upstream of the transcriptional repressor Bcl6.
Nat. Immunol.
16
:
980
990
.
45.
Xu
L.
,
Y.
Cao
,
Z.
Xie
,
Q.
Huang
,
Q.
Bai
,
X.
Yang
,
R.
He
,
Y.
Hao
,
H.
Wang
,
T.
Zhao
, et al
2015
.
The transcription factor TCF-1 initiates the differentiation of TFH cells during acute viral infection.
Nat. Immunol.
16
:
991
999
.
46.
Shao
P.
,
F.
Li
,
J.
Wang
,
X.
Chen
,
C.
Liu
,
H. H.
Xue
.
2019
.
Cutting edge: Tcf1 instructs T follicular helper cell differentiation by repressing Blimp1 in response to acute viral infection.
J. Immunol.
203
:
801
806
.
47.
Hess Michelini
R.
,
A. L.
Doedens
,
A. W.
Goldrath
,
S. M.
Hedrick
.
2013
.
Differentiation of CD8 memory T cells depends on Foxo1.
J. Exp. Med.
210
:
1189
1200
.
48.
Delpoux
A.
,
C. Y.
Lai
,
S. M.
Hedrick
,
A. L.
Doedens
.
2017
.
FOXO1 opposition of CD8+ T cell effector programming confers early memory properties and phenotypic diversity.
Proc. Natl. Acad. Sci. USA
114
:
E8865
E8874
.
49.
Hedrick
S. M.
,
R.
Hess Michelini
,
A. L.
Doedens
,
A. W.
Goldrath
,
E. L.
Stone
.
2012
.
FOXO transcription factors throughout T cell biology.
Nat. Rev. Immunol.
12
:
649
661
.
50.
Song
W.
,
J.
Craft
.
2019
.
T follicular helper cell heterogeneity: time, space, and function.
Immunol. Rev.
288
:
85
96
.
51.
Nakayamada
S.
,
Y.
Kanno
,
H.
Takahashi
,
D.
Jankovic
,
K. T.
Lu
,
T. A.
Johnson
,
H. W.
Sun
,
G.
Vahedi
,
O.
Hakim
,
R.
Handon
, et al
2011
.
Early Th1 cell differentiation is marked by a Tfh cell-like transition.
Immunity
35
:
919
931
.
52.
Read
K. A.
,
M. D.
Powell
,
C. E.
Baker
,
B. K.
Sreekumar
,
V. M.
Ringel-Scaia
,
H.
Bachus
,
R. E.
Martin
,
I. D.
Cooley
,
I. C.
Allen
,
A.
Ballesteros-Tato
,
K. J.
Oestreich
.
2017
.
Integrated STAT3 and Ikaros zinc finger transcription factor activities regulate Bcl-6 expression in CD4+ Th cells.
J. Immunol.
199
:
2377
2387
.
53.
Stauss
D.
,
C.
Brunner
,
F.
Berberich-Siebelt
,
U. E.
Höpken
,
M.
Lipp
,
G.
Müller
.
2016
.
The transcriptional coactivator Bob1 promotes the development of follicular T helper cells via Bcl6.
EMBO J.
35
:
881
898
.
54.
Nurieva
R. I.
,
A.
Podd
,
Y.
Chen
,
A. M.
Alekseev
,
M.
Yu
,
X.
Qi
,
H.
Huang
,
R.
Wen
,
J.
Wang
,
H. S.
Li
, et al
2012
.
STAT5 protein negatively regulates T follicular helper (Tfh) cell generation and function.
J. Biol. Chem.
287
:
11234
11239
.
55.
McDonald
P. W.
,
K. A.
Read
,
C. E.
Baker
,
A. E.
Anderson
,
M. D.
Powell
,
A.
Ballesteros-Tato
,
K. J.
Oestreich
.
2016
.
IL-7 signalling represses Bcl-6 and the TFH gene program.
Nat. Commun.
7
:
10285
.
56.
Schmitt
N.
,
Y.
Liu
,
S. E.
Bentebibel
,
I.
Munagala
,
L.
Bourdery
,
K.
Venuprasad
,
J.
Banchereau
,
H.
Ueno
.
2014
.
The cytokine TGF-β co-opts signaling via STAT3-STAT4 to promote the differentiation of human TFH cells.
Nat. Immunol.
15
:
856
865
.
57.
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 (TFH) cell lineage is induced by IL-12.
Immunol. Cell Biol.
87
:
590
600
.
58.
Ray
J. P.
,
H. D.
Marshall
,
B. J.
Laidlaw
,
M. M.
Staron
,
S. M.
Kaech
,
J.
Craft
.
2014
.
Transcription factor STAT3 and type I interferons are corepressive insulators for differentiation of follicular helper and T helper 1 cells.
Immunity
40
:
367
377
.
59.
Tubo
N. J.
,
A. J.
Pagán
,
J. J.
Taylor
,
R. W.
Nelson
,
J. L.
Linehan
,
J. M.
Ertelt
,
E. S.
Huseby
,
S. S.
Way
,
M. K.
Jenkins
.
2013
.
Single naive CD4+ T cells from a diverse repertoire produce different effector cell types during infection.
Cell
153
:
785
796
.
60.
Kotov
D. I.
,
J. S.
Mitchell
,
T.
Pengo
,
C.
Ruedl
,
S. S.
Way
,
R. A.
Langlois
,
B. T.
Fife
,
M. K.
Jenkins
.
2019
.
TCR affinity biases Th cell differentiation by regulating CD25, Eef1e1, and Gbp2.
J. Immunol.
202
:
2535
2545
.

The authors have no financial conflicts of interest.

Supplementary data