NK cells represent a cellular component of the mammalian innate immune system, and they mount rapid responses against viral infection, including the secretion of the potent antiviral effector cytokine IFN-γ. Following mouse CMV infection, Bhlhe40 was the most highly induced transcription factor in NK cells among the basic helix-loop-helix family. Bhlhe40 upregulation in NK cells depended upon IL-12 and IL-18 signals, with the promoter of Bhlhe40 enriched for STAT4 and the permissive histone H3K4me3, and with STAT4-deficient NK cells showing an impairment of Bhlhe40 induction and diminished H3K4me3. Transcriptomic and protein analysis of Bhlhe40-deficient NK cells revealed a defect in IFN-γ production during mouse CMV infection, resulting in diminished protective immunity following viral challenge. Finally, we provide evidence that Bhlhe40 directly promotes IFN-γ by binding throughout the Ifng loci in activated NK cells. Thus, our study reveals how STAT4-mediated control of Bhlhe40 drives protective IFN-γ secretion by NK cells during viral infection.

Natural killer cells are innate lymphocytes able to respond to stress, transformed, or infected cells without prior sensitization. However, resting NK cells can become more potent effector cells through activation by germline-encoded surface receptors and proinflammatory cytokines, including IL-12, IL-18, and type I IFN produced during viral infection (1). These activating signals will induce a robust transcriptional and epigenetic response in NK cells, leading to their effector programs (2), including the production of the potent antiviral cytokine IFN-γ.

During mouse CMV (MCMV) infection, IL-12 is produced and binds to a heterodimeric receptor, leading to a signal cascade via JAK-mediated phosphorylation and homodimerization of STAT4 in the cytoplasm. In NK cells, STAT4 activation causes it to translocate to the nucleus, where it induces the transcription of a multitude of genes by targeting both promoter and nonpromoter regions (3). Previous studies have demonstrated that an IL-12/STAT4 signaling axis in NK cells can drive a plethora of effector functions, including cytokine production and proliferation (4) through transcription factors Zbtb32, IRF8, STAT5, CBFb, and Runx factors (58), among others. However, many additional genes targeted by STAT4 in NK cells during MCMV infection have not been characterized.

In our present study, we discovered that Bhlhe40 was highly induced in activated NK cells during MCMV infection among the family of basic helix-loop-helix (bHLH) transcription factors. Bhlhe40, also known as Bhlhb2, Dec1, and Stra13, consists of a basic DNA-binding domain; a helix-loop-helix domain, which facilitates dimerization; and a protein–protein interaction Orange domain (9, 10). Members of the bHLH family (specifically factors in the E clade, including Bhlhe40) have been shown to regulate numerous physiological processes, including differentiation, proliferation, and apoptosis in multiple cell types due to its ability to act as both transcriptional activator and repressor in a context-dependent manner (11). Although Bhlhe40 was initially described as a regulator of circadian rhythm and cellular differentiation, emerging evidence suggests that it is also acting as a regulator of the innate and adaptive immune systems (11).

In T cells, Bhlhe40 is rapidly expressed following TCR activation (12) and plays a number of critical roles to support T cell identity and effector functions. In the context of autoimmunity and aging, Bhlhe40 promotes GM-CSF expression while suppressing IL-10 production (13) and maintains regulatory T cell function/identity, respectively (14, 15). Additionally, Bhlhe40 is also required for the function of tissue-resident memory CD8+ and CD4+ T cells and tumor-infiltrating lymphocytes by maintaining their mitochondrial fitness and epigenetic states (16, 17). More recently, Bhlhe40 was shown to be a cell-intrinsic negative regulator of both activated B cells and follicular helper T cells in germinal center responses (18). Within the innate immune system, Bhlhe40 controls the self-renewal and maintenance of alveolar and large peritoneal macrophages and their proliferation in response to IL-4 (19, 20). In the present study, we investigated mechanisms of Bhlhe40 induction and consequences of its deletion on NK cell activation and effector function in response to viral infection.

Mice were bred at Memorial Sloan Kettering Cancer Center in accordance with the guidelines of the institutional animal care and use committee. The following strains were used in this study: C57BL/6 (CD45.2), B6.SJL (CD45.1), Stat4−/−, Il12rb2−/−, Il18r1−/−, Bhlhe40−/−, and Klra8−/− (Ly49h−/−). Experiments were conducted using age- and sex-matched mice in accordance with approved institutional protocols. Adoptive transfer studies and generation of mixed bone marrow chimeric mice were performed as previously described (21).

MCMV (Smith) was passaged through BALB/c mice three times, followed by preparation of salivary gland viral stocks using a homogenizer to dissociate the salivary glands (of BALB/c mice 3 wk postinfection [PI]). MCMV stocks were diluted prior to injection, and 103 PFUs were delivered in adoptive transfer studies and 104 PFUs in all other experiments.

Single-cell suspensions were prepared from the indicated organs, and the indicated fluorophore-conjugated Abs (BioLegend, Tonbo, eBioscience) were used to stain lymphocytes. Flow cytometry was performed on a BD Biosciences LSR II or Cytek Biosciences Aurora, and data were analyzed with FlowJo software.

NK cells were enriched ex vivo using a no-touch bead enrichment protocol as previously described (21) and plated at 105 per well with mouse IL-2, IL-12, or IL-15 (R&D Systems) at 20 ng/ml; with IL-18 (MBL) at 10 ng/ml; or with IFN-α (R&D Systems) at 100 IU. Intracellular staining for IFN-γ was performed as previously described (21).

The following datasets and analyses were performed on NK cells as previously described by our laboratory and can be found in the GEO database as a SuperSeries under accession numbers GSE106139 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE106139) and GSE140044 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE140044): H3K4me3 chromatin immunoprecipitation sequencing (ChIP-seq) and assay for transposase-accessible chromatin with sequencing (ATAC-seq) following cytokine stimulation; RNA sequencing (RNA-seq) from MCMV-infected Il12rb2−/−, Stat4−/− mice; RNA-seq on days 0, 2, 4, 7, and 35 after MCMV infection; and RNA-seq on 3-h and overnight in vitro cytokine stimulations (2, 3, 6, 22, 23).

For new datasets generated in this study, RNA-seq was performed as previously described (2). Differential analysis was executed with DESeq2 (version 1.22.2). For all differential analyses, genes were considered differential if they showed a false discovery rate-adjusted p value (FDR) ≤0.05 by contrast using the DESeq2 model. For all sequencing counts shown, values were normalized using sizeFactors calculated by the DESeq2 software or Saccharomyces cerevisiae spike-in DNA. Paired analysis was performed between wild-type (WT) versus Bhlhe40−/− NK cells for day 0 versus day 2 PI, where calculations for generalized linear models were fitted with a design that controlled for each group. RNA-seq data comparing WT with Bhlhe40−/− NK cells before and after MCMV infection are available under GEO accession number GSE235608 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE235608) and RNA-seq data comparing WT with Il18r1−/− NK cells before and after MCMV infection are available under GEO accession number GSE244300 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE244300).

For the Stat4 and Bhlhe40 cleavage under targets and release using nuclease (CUT&RUN), 500,000 sorted NK cells were washed three times with PBS, resuspended in buffer 1 (1× eBioscience Perm/Wash Buffer, 1× Roche cOmplete EDTA-free protease inhibitor, 0.5 μM spermidine in H2O) and incubated with BHLHE40 (anti-DEC1; Novus, NB100-1800) or STAT4 (Invitrogen, 71-4500) Abs at 1:100 dilution in Ab buffer (buffer 1 + 2 μM EDTA) in a 96-well V-bottomed plate at 4°C overnight. Upon Ab incubation, cells were washed twice with buffer 1 and resuspended in 50 μl of buffer 1 + 1× pA/G-MNase (Cell Signaling Technology, 57813) and incubated on ice for 1 h and washed with buffer 2 (0.05% w/v saponin, 1× Roche cOmplete EDTA-free protease inhibitor, 0.5 μM spermidine in 1× PBS) three times. After washing, calcium buffer (buffer 2 + 2 μM CaCl2) was used to resuspend the cells for 30 min on ice to activate the pA/G-MNase reaction, and an equal volume of 2× STOP Buffer (buffer 2 + 20 μM EDTA + 4 μM EGTA) was added along with 1 pg Saccharomyces cerevisiae spike-in DNA (Cell Signaling Technology, 29987). Samples were incubated for 15 min at 37°C, and DNA was isolated and purified using the Qiagen MinElute Kit according to the manufacturer’s protocol and subjected to library amplification.

Immunoprecipitated DNA was quantified by PicoGreen, and the size was evaluated using an Agilent BioAnalyzer. When possible, fragments between 100 and 600 bp were size selected using aMPure XP beads (Beckman Coulter, A63882), and Illumina sequencing libraries were prepared using the KAPA HTP Library Preparation Kit (Kapa Biosystems, KK8234) according to the manufacturer’s instructions with 0.001–0.5 ng input DNA and 14 cycles of PCR. Barcoded libraries were run on the NovaSeq 6000 in a PE100 run using S4 kit version 1.5 with XP mode (Bhlhe40 CUT&RUN) or NovaSeq X in a PE100 run using the 10B reagent kit (200 cycles; Illumina, STAT4 CUT&RUN). An average of 23 million paired reads were generated per sample. CUT&RUN data are available under GEO accession numbers GSE242900 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE242900) and GSE242911 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE242911).

Paired reads were trimmed for adaptors and removal of low-quality reads by using Trimmomatic (version 0.39) and aligned to the mm10 reference genome using Bowtie 2 (version 2.4.1). Upon alignment, peaks were called using MACS2 (version 2.2.7.1) with input samples as a control using narrow peak parameters --cutoff-analysis -p 1e-5 --keep-dup all -B --SPMR. For each sample group, irreproducible discovery rate calculations were performed using scripts provided by the ENCODE project (https://www.encodeproject.org/software/idr/; version 2.0.4.2). Reproducible peaks showing an irreproducible discovery rate value of 0.05 or less in each condition were aggregated across the experiment and merged via union to the peak atlas, annotated with the UCSC Known Gene model. The peak atlas consisted of 316 peaks for BHLHE40 CUT&RUN and 2654 for STAT4 CUT&RUN.

Data are shown as mean ± SEM in all graphs, and statistical differences, and unless noted otherwise, were calculated using a two-tailed unpaired Student t test. A p value ≤0.05 was considered significant. In these figures, p values are denoted as *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, and ****p ≤ 0.0001. For data showing a summary of normalized counts from RNA-seq and differential analysis from various sequencing experiments, statistics were calculated using the DESeq2 method as noted above. For these data, FDR adjusted p values ≤0.05 were considered statistically significant. In these figures, *p ≤ 0.05. All statistical analyses and plots were produced in GraphPad Prism or R.

bHLH transcription factors are widely expressed in different immune cells (11). To assess the regulation of bHLH transcription factors in NK cells during MCMV infection, we performed comparative transcriptomic analysis by RNA-seq during their immune response. At day 2 PI, a time point when NK cells are robustly exhibiting their innate immune functions (1), the Bhlhe40 transcript was the most significantly upregulated among the E family bHLH transcription factors in NK cells (Fig. 1A, 1B).

FIGURE 1.

Early induction of Bhlhe40 in NK cells during MCMV infection is driven by IL-12 and IL-18. (A) Dot plot of RNA-seq log2 fold change comparing expression of select bHLH family E clade transcription factors in NK cells at day 2 (d2) PI compared with d0. Black points refer to genes with FDR ≤0.05 in d2 PI to d0 differential expression contrast. (B) RNA-seq normalized counts of Bhlhe40 from mouse Ly49H+ NK cells during MCMV infection. (C and D) RNA-seq data showing Bhlhe40 from sorted NK cells stimulated with indicated cytokines for 3 h (C) or 16 h (D) in vitro. *FDR ≤0.05 in differential expression contrast against unstimulated samples. (E and F) RNA-seq normalized counts of Bhlhe40 from WT versus IL-12R–deficient (E) or IL-18R–deficient (F) NK cells at d0 and d2 PI. *FDR ≤0.05 in differential expression contrast against both d0 PI sample groups or against the indicated deficient sample.

FIGURE 1.

Early induction of Bhlhe40 in NK cells during MCMV infection is driven by IL-12 and IL-18. (A) Dot plot of RNA-seq log2 fold change comparing expression of select bHLH family E clade transcription factors in NK cells at day 2 (d2) PI compared with d0. Black points refer to genes with FDR ≤0.05 in d2 PI to d0 differential expression contrast. (B) RNA-seq normalized counts of Bhlhe40 from mouse Ly49H+ NK cells during MCMV infection. (C and D) RNA-seq data showing Bhlhe40 from sorted NK cells stimulated with indicated cytokines for 3 h (C) or 16 h (D) in vitro. *FDR ≤0.05 in differential expression contrast against unstimulated samples. (E and F) RNA-seq normalized counts of Bhlhe40 from WT versus IL-12R–deficient (E) or IL-18R–deficient (F) NK cells at d0 and d2 PI. *FDR ≤0.05 in differential expression contrast against both d0 PI sample groups or against the indicated deficient sample.

Close modal

Given that homeostatic and proinflammatory cytokines (IL-2, IL-12, IL-15, IL-18, or type I IFN) and their downstream STAT molecules (STAT1, STAT4, or STAT5) are strongly signaling in NK cells at day 2 PI (3), we analyzed Bhlhe40 expression in NK cells stimulated with various cytokines for 3 h or overnight. We found that NK cells stimulated with IL-12 + IL-18, but not other cytokines, showed the greatest induction of Bhlhe40 (Fig. 1C, 1D). Consistent with this observation, the induction of Bhlhe40 was largely or partially ablated in NK cells deficient for the IL-12 receptor or IL-18 receptor, respectively, during MCMV infection (Fig. 1E, 1F). Together, these data suggest that Bhlhe40 induction in NK cells early after viral infection is dependent mainly on IL-12 and to a lesser extent on IL-18 signals.

Given that IL-12 signals via the transcription factor STAT4, we investigated whether STAT4 controls transcription of Bhlhe40 in NK cells during MCMV infection. Indeed, STAT4 deficiency resulted in a significant loss of Bhlhe40 induction (Fig. 2A), regardless of activating receptor Ly49H expression, suggesting that Bhlhe40 is critically dependent on STAT4 in activated NK cells. To determine whether STAT4 directly targets the Bhlhe40 loci, we performed STAT4 CUT&RUN on NK cells stimulated with IL-12 + IL-18 (2). Significantly increased differential STAT4 binding was detected at the promoter of Bhlhe40 in stimulated NK cells when contrasted with unstimulated or cytokine-stimulated Stat4−/− NK cells (log2 fold changes of 3.17 and 2.26 and FDRs of 8.95e-40 and 1.74e-17, respectively) (Fig. 2B). Although we did not observe large changes in chromatin accessibility (by ATAC-seq) at the promoter region of Bhlhe40 in NK cells stimulated with IL-12 + IL-18 (Fig. 2C), we observed a significant increase in the permissive histone mark H3K4me3 following stimulation (Fig. 2D). Furthermore, we determined that the increase in H3K4me3 at the Bhlhe40 locus in stimulated NK cells is dependent on an IL-12/STAT4 signaling axis (Fig. 2E). Altogether, we provide evidence that STAT4 regulates the expression of Bhlhe40 in activated NK cells during MCMV infection via deposition of the permissive histone mark H3K4me3.

FIGURE 2.

Bhlhe40 upregulation is controlled by Stat4 in activated NK cells. (A) RNA-seq showing Bhlhe40 from WT versus STAT4-deficient Ly49H+ or Ly49H NK cells at day 0 (d0) and d2 PI. Every permutation of contrasts for differential expression analysis show *FDR ≤0.05 except WT d0 versus STAT4-deficient d0 in both Ly49H+ and Ly49H datasets, but only WT versus STAT4-deficient at d2 is shown. (B) STAT4 CUT&RUN tracks show STAT4 binding as mean normalized fragment pileup (y-axis) plotted by genome position (x-axis) from unstimulated or IL-12 + IL-18–stimulated WT and STAT4-deficient NK cells with bin length of 10 bp. Signals normalized using spike-in DNA were averaged per bin in each sample condition group (WT groups, n = 3; knockout, n = 2). Dotted box indicates range for peak called for differential binding analysis. (C and D) ATAC-seq and ChIP-seq transcription start site (TSS)-centered metapeak plots show chromatin accessibility (C) and H3K4me3 binding signal (D), respectively, at the TSS of the Bhlhe40 locus in unstimulated NK cells (gray line) or NK cells stimulated with IL-12 + IL-18 (black line). ATAC-seq signals are normalized using size factors calculated by DESeq2 and averaged in each sample condition group (n = 3). (E) H3K4me3 ChIP-seq signal tracks at the Bhlhe40 locus in WT or STAT4-deficient NK cells stimulated with IL-12 + IL-18.

FIGURE 2.

Bhlhe40 upregulation is controlled by Stat4 in activated NK cells. (A) RNA-seq showing Bhlhe40 from WT versus STAT4-deficient Ly49H+ or Ly49H NK cells at day 0 (d0) and d2 PI. Every permutation of contrasts for differential expression analysis show *FDR ≤0.05 except WT d0 versus STAT4-deficient d0 in both Ly49H+ and Ly49H datasets, but only WT versus STAT4-deficient at d2 is shown. (B) STAT4 CUT&RUN tracks show STAT4 binding as mean normalized fragment pileup (y-axis) plotted by genome position (x-axis) from unstimulated or IL-12 + IL-18–stimulated WT and STAT4-deficient NK cells with bin length of 10 bp. Signals normalized using spike-in DNA were averaged per bin in each sample condition group (WT groups, n = 3; knockout, n = 2). Dotted box indicates range for peak called for differential binding analysis. (C and D) ATAC-seq and ChIP-seq transcription start site (TSS)-centered metapeak plots show chromatin accessibility (C) and H3K4me3 binding signal (D), respectively, at the TSS of the Bhlhe40 locus in unstimulated NK cells (gray line) or NK cells stimulated with IL-12 + IL-18 (black line). ATAC-seq signals are normalized using size factors calculated by DESeq2 and averaged in each sample condition group (n = 3). (E) H3K4me3 ChIP-seq signal tracks at the Bhlhe40 locus in WT or STAT4-deficient NK cells stimulated with IL-12 + IL-18.

Close modal

Because Bhlhe40 is strongly induced in NK cells early after viral infection, we investigated whether Bhlhe40 may be driving the specific NK cell effector functions of cytokine secretion, killing, and proliferation. To test the latter using a well-established adoptive transfer model, we injected an equal number of WT (CD45.1) and Bhlhe40−/− (CD45.2) NK cells into Ly49H-deficient recipient mice infected with MCMV and tracked the Ly49H+ NK cell responses in the recipient. No detectable difference was observed in the magnitude of NK cell expansion following MCMV infection, because WT and Bhlhe40−/− counterparts not only proliferated and contracted similarly but also generated a comparable pool of memory cells (Supplemental Fig. 1A, 1B), suggesting that NK cell proliferation and “adaptive” features are not impacted by loss of Bhlhe40. Additionally, their maturation/activation profiles were similar and commensurate with effector NK cells at day 7 PI (Supplemental Fig. 1C). Furthermore, WT and Bhlhe40−/− NK cells were indistinguishable in their ability to produce granzyme B and upregulate CD69 (Supplemental Fig. 1D), suggesting that cytolytic capacity and activation of NK cells remained intact in the absence of Bhlhe40.

In transcriptomic analysis, among the genes dysregulated in Bhlhe40−/− NK cells compared with WT during MCMV infection were Ifng, Cxcr4, and Bhlhe40 (lower in Bhlhe40−/−) and Il21r, Miga1, and Cnbd2 (elevated in Bhlhe40−/−) (Fig. 3A). Confirming that Ifng mRNA was downregulated, Bhlhe40−/− NK cells produced less IFN-γ protein than did WT NK cells following infection (Fig. 3B). Moreover, Bhlhe40−/− NK cells similarly produced diminished IFN-γ compared with their WT counterparts following in vitro stimulation with proinflammatory cytokines (Fig. 3C). Finally, we observed that Bhlhe40−/− mice were more susceptible than WT mice to MCMV challenge, and most Bhlhe40−/− mice succumbed at a similar rate and with similar kinetics to NK cell-depleted WT mice (Fig. 3D).

FIGURE 3.

Bhlhe40 control of IFN-γ in NK cell–mediated host protection. (A) Volcano plot of RNA-seq log2 fold change comparing WT versus Bhlhe40−/− NK cells at day 2 PI compared with day 0. Black dots show differentially expressed genes (FDR <0.05), and genes of note are bolded. Horizontal dashed line indicates p = 0.05, and vertical dashed lines indicate absolute log2 fold change of 0.5. (B) Representative histogram (left) shows WT versus Bhlhe40−/− NK cells from mixed bone marrow chimeric mice infected with MCMV that were assessed for IFN-γ secretion at day 2 PI and compared against NK cells from uninfected mice. Paired dot plot (right) shows IFN-γ mean fluorescence intensity (MFI) of paired WT and Bhlhe40−/− NK cells from individual mice. A paired two-tailed t test was performed for the right plot. ****p < 0.0001. (C) Representative histogram (left) of IFN-γ production between WT and Bhlhe40−/− NK cells following stimulation with IL-12 + IL-18 and percentage of NK cells producing IFN-γ measured (right). An unpaired two-tailed t test was performed for the right graph between WT and Bhlhe40−/− within each stimulation condition. ***p < 0.001. (D) Kaplan–Meier survival curves for WT versus Bhlhe40−/− mice and mice depleted of NK cells following MCMV challenge. A log-rank (Mantel-Cox) test was performed between each contrast. All data shown are representative of two or three experiments (n = 3–5 per experiment). ***p < 0.001. KO, knockout.

FIGURE 3.

Bhlhe40 control of IFN-γ in NK cell–mediated host protection. (A) Volcano plot of RNA-seq log2 fold change comparing WT versus Bhlhe40−/− NK cells at day 2 PI compared with day 0. Black dots show differentially expressed genes (FDR <0.05), and genes of note are bolded. Horizontal dashed line indicates p = 0.05, and vertical dashed lines indicate absolute log2 fold change of 0.5. (B) Representative histogram (left) shows WT versus Bhlhe40−/− NK cells from mixed bone marrow chimeric mice infected with MCMV that were assessed for IFN-γ secretion at day 2 PI and compared against NK cells from uninfected mice. Paired dot plot (right) shows IFN-γ mean fluorescence intensity (MFI) of paired WT and Bhlhe40−/− NK cells from individual mice. A paired two-tailed t test was performed for the right plot. ****p < 0.0001. (C) Representative histogram (left) of IFN-γ production between WT and Bhlhe40−/− NK cells following stimulation with IL-12 + IL-18 and percentage of NK cells producing IFN-γ measured (right). An unpaired two-tailed t test was performed for the right graph between WT and Bhlhe40−/− within each stimulation condition. ***p < 0.001. (D) Kaplan–Meier survival curves for WT versus Bhlhe40−/− mice and mice depleted of NK cells following MCMV challenge. A log-rank (Mantel-Cox) test was performed between each contrast. All data shown are representative of two or three experiments (n = 3–5 per experiment). ***p < 0.001. KO, knockout.

Close modal

To determine the direct binding sites of Bhlhe40, we performed CUT&RUN on NK cells stimulated with IL-12 + IL-18. We identified a total of 316 Bhlhe40 binding sites, where most of these peaks were called in the stimulated condition (n = 279; Fig. 4A, 4B). Through global differential binding analysis, we found that the majority of these sites were significantly enriched upon IL-12 + IL-18 stimulation (Fig. 4C). Notably, the top differential binding site of Bhlhe40 was within the Ifng locus (Fig. 4D). Among the significant differentially binding sites, most were annotated as promoter peaks (74%, 208 of 281; Fig. 4E). Motif analysis with differentially binding peaks revealed de novo motifs that suggest potential interactions with families including ATF1 and IRF6, and because the bHLH family is present within the motif database used, the top match includes TFEC, a bHLH family member (Fig. 4F). Taken together, our data indicate that Bhlhe40 is critical for host protection against viral infection by acting as a direct driver of the proinflammatory/immunoregulatory cytokine IFN-γ in NK cells.

FIGURE 4.

Bhlhe40 binds the Ifng loci of NK cells following IL-12 + IL-18 stimulation. (A) Mean normalized counts heatmap of Bhlhe40 binding in NK cells without stimulation or following IL-12 + IL-18 for 3 h, peak centered and flanked 1 kb each in 5′ and 3′ directions (n = 316) with bin length of 50 bp. Signals normalized using spike-in DNA were averaged per bin between each sample condition replicate (n = 3). (B) Venn diagram of peaks called within the peak atlas between the two sample conditions (n = 316). (C) Volcano plot of differential binding analysis between IL-12 + IL-18 stimulation and unstimulated samples. Red points represent significant differentially binding peaks (FDR <0.05). Horizontal dashed line shows guide for intercept at −log10(0.05) and vertical for −1 and 1. Select peaks and their annotated gene names are highlighted and labeled. (D) Bhlhe40 CUT&RUN tracks show normalized signal pileup for the Ifng locus. Signals normalized using spike-in DNA were averaged per bin (bin length 10 bp) between each sample condition replicate (n = 3). (E) Pie chart of annotated peak type composition for peaks that significantly increase binding upon IL-12 + IL-18 stimulation (n = 281). Two hundred eight (74.0%) peaks were assigned promoter, 41 (14.6%) peaks intergenic, 27 (9.6%) peaks intron, and 5 (1.8%) peaks exon. (F) Enriched de novo motifs found by HOMER on peaks that significantly increase binding following IL-12 + IL-18 stimulation. The top four motifs ranked by p value that exceeded HOMER motif score of 0.7 are shown. TF, transcription factor.

FIGURE 4.

Bhlhe40 binds the Ifng loci of NK cells following IL-12 + IL-18 stimulation. (A) Mean normalized counts heatmap of Bhlhe40 binding in NK cells without stimulation or following IL-12 + IL-18 for 3 h, peak centered and flanked 1 kb each in 5′ and 3′ directions (n = 316) with bin length of 50 bp. Signals normalized using spike-in DNA were averaged per bin between each sample condition replicate (n = 3). (B) Venn diagram of peaks called within the peak atlas between the two sample conditions (n = 316). (C) Volcano plot of differential binding analysis between IL-12 + IL-18 stimulation and unstimulated samples. Red points represent significant differentially binding peaks (FDR <0.05). Horizontal dashed line shows guide for intercept at −log10(0.05) and vertical for −1 and 1. Select peaks and their annotated gene names are highlighted and labeled. (D) Bhlhe40 CUT&RUN tracks show normalized signal pileup for the Ifng locus. Signals normalized using spike-in DNA were averaged per bin (bin length 10 bp) between each sample condition replicate (n = 3). (E) Pie chart of annotated peak type composition for peaks that significantly increase binding upon IL-12 + IL-18 stimulation (n = 281). Two hundred eight (74.0%) peaks were assigned promoter, 41 (14.6%) peaks intergenic, 27 (9.6%) peaks intron, and 5 (1.8%) peaks exon. (F) Enriched de novo motifs found by HOMER on peaks that significantly increase binding following IL-12 + IL-18 stimulation. The top four motifs ranked by p value that exceeded HOMER motif score of 0.7 are shown. TF, transcription factor.

Close modal

Previous studies have identified that IL-12 and STAT4 signaling can drive multiple effector functions in NK cells during MCMV infection, including the secretion of IFN-γ and the prolific expansion of antiviral “adaptive” NK cells (1). How these different processes are controlled by a single transcription factor, STAT4, and whether these specific functions can be segregated in their regulation remained incompletely understood. We previously identified that STAT4-mediated expression of Zbtb32, Runx1, Runx3, and IRF8 could specifically impact clonal proliferation but not IFN-γ secretion by antiviral NK cells (5, 6, 8), whereas STAT4-mediated upregulation of STAT5 could impact both processes (7). In the present study, we have discovered a cell-intrinsic role for the transcription factor Bhlhe40 in specifically controlling IFN-γ production but not proliferation of NK cells following viral infection, highlighting a mechanism that decouples STAT4-driven innate functions from STAT4-driven adaptive features.

Control of cytokine secretion by Bhlhe40 has also been observed in other cell types, but whether Bhlhe40 was directly regulated by STAT transcription factors in these settings was not elucidated (12, 13, 2427). Through epigenetic studies, we found that STAT4 is inducing the permissive histone H3K4me3 mark at the Bhlhe40 locus rather than mediating changes in chromatin accessibility, in contrast to many other genetic loci targeted by STAT4 (3). This distinct role for STAT4 in modulating the chromatin landscape at the Bhlhe40 locus critically drives its expression and subsequently protective IFN-γ from NK cells during viral infection. Thus, the mode of STAT4 activity can potentially show specificity dependent upon the range of cotranscription factors it induces in activated NK cells (given that STAT4 also binds to the Ifng loci [28]), and understanding such underlying mechanisms can aid in the goal of harnessing these potent effector cells in our battle against infectious diseases.

The authors have no financial conflicts of interest.

We thank members of the Sun Lab for experimental assistance and helpful discussions.

This work was supported by funding from the Ludwig Center for Cancer Immunotherapy, the American Cancer Society, the Burroughs Wellcome Fund, and the National Institutes of Health (Grants AI100874, AI130043, AI155558, and P30CA008748) to J.C.S.

The online version of this article contains supplemental material.

The RNA-seq data presented in this article have been submitted to the Gene Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/) under accession numbers GSE235608 and GSE244300. The cleavage under targets and release using nuclease (CUT&RUN) data presented in this article have been submitted to the GEO under accession numbers GSE242900 and GSE242911.

ATAC-seq

assay for transposase-accessible chromatin with sequencing

bHLH

basic helix-loop-helix

ChIP-seq

chromatin immunoprecipitation sequencing

CUT&RUN

cleavage under targets and release using nuclease

FDR

false discovery rate-adjusted p value

GEO

Gene Expression Omnibus

MCMV

mouse CMV

PI

postinfection

RNA-seq

RNA sequencing

WT

wild type

1
Mujal
,
A. M.
,
R. B.
Delconte
,
J. C.
Sun
.
2021
.
Natural killer cells: from innate to adaptive features
.
Annu. Rev. Immunol.
39
:
417
447
.
2
Lau
,
C. M.
,
N. M.
Adams
,
C. D.
Geary
,
O.-E.
Weizman
,
M.
Rapp
,
Y.
Pritykin
,
C. S.
Leslie
,
J. C.
Sun
.
2018
.
Epigenetic control of innate and adaptive immune memory
.
Nat. Immunol.
19
:
963
972
.
3
Wiedemann
,
G. M.
,
E. K.
Santosa
,
S.
Grassmann
,
S.
Sheppard
,
J.-B.
Le Luduec
,
N. M.
Adams
,
C.
Dang
,
K. C.
Hsu
,
J. C.
Sun
,
C. M.
Lau
.
2021
.
Deconvoluting global cytokine signaling networks in natural killer cells
.
Nat. Immunol.
22
:
627
638
.
4
Sun
,
J. C.
,
S.
Madera
,
N. A.
Bezman
,
J. N.
Beilke
,
M. H.
Kaplan
,
L. L.
Lanier
.
2012
.
Proinflammatory cytokine signaling required for the generation of natural killer cell memory
.
J. Exp. Med.
209
:
947
954
.
5
Beaulieu
,
A. M.
,
C. L.
Zawislak
,
T.
Nakayama
,
J. C.
Sun
.
2014
.
The transcription factor Zbtb32 controls the proliferative burst of virus-specific natural killer cells responding to infection
.
Nat. Immunol.
15
:
546
553
.
6
Rapp
,
M.
,
C. M.
Lau
,
N. M.
Adams
,
O.-E.
Weizman
,
T. E.
O’Sullivan
,
C. D.
Geary
,
J. C.
Sun
.
2017
.
Core-binding factor β and Runx transcription factors promote adaptive natural killer cell responses
.
Sci. Immunol.
2
:
eaan3796
.
7
Wiedemann
,
G. M.
,
S.
Grassmann
,
C. M.
Lau
,
M.
Rapp
,
A. V.
Villarino
,
C.
Friedrich
,
G.
Gasteiger
,
J. J.
O’Shea
,
J. C.
Sun
.
2020
.
Divergent role for STAT5 in the adaptive responses of natural killer cells
.
Cell Rep.
33
:
108498
.
8
Adams
,
N. M.
,
C. M.
Lau
,
X.
Fan
,
M.
Rapp
,
C. D.
Geary
,
O.-E.
Weizman
,
C.
Diaz-Salazar
,
J. C.
Sun
.
2018
.
Transcription factor IRF8 orchestrates the adaptive natural killer cell response
.
Immunity
48
:
1172
1182.e6
.
9
Sun
,
H.
,
S.
Ghaffari
,
R.
Taneja
.
2007
.
bHLH-Orange transcription factors in development and cancer
.
Transl. Oncogenomics
2
:
107
120
.
10
Yamada
,
K.
,
K.
Miyamoto
.
2005
.
Basic helix-loop-helix transcription factors, BHLHB2 and BHLHB3; their gene expressions are regulated by multiple extracellular stimuli
.
Front. Biosci.
10
:
3151
3171
.
11
Cook
,
M. E.
,
N. N.
Jarjour
,
C.-C.
Lin
,
B. T.
Edelson
.
2020
.
Transcription factor Bhlhe40 in immunity and autoimmunity
.
Trends Immunol.
41
:
1023
1036
.
12
Yu
,
F.
,
S.
Sharma
,
D.
Jankovic
,
R. K.
Gurram
,
P.
Su
,
G.
Hu
,
R.
Li
,
S.
Rieder
,
K.
Zhao
,
B.
Sun
,
J.
Zhu
.
2018
.
The transcription factor Bhlhe40 is a switch of inflammatory versus antiinflammatory Th1 cell fate determination
.
J. Exp. Med.
215
:
1813
1821
.
13
Lin
,
C.-C.
,
T. R.
Bradstreet
,
E. A.
Schwarzkopf
,
J.
Sim
,
J. A.
Carrero
,
C.
Chou
,
L. E.
Cook
,
T.
Egawa
,
R.
Taneja
,
T. L.
Murphy
, et al
2014
.
Bhlhe40 controls cytokine production by T cells and is essential for pathogenicity in autoimmune neuroinflammation
.
Nat. Commun.
5
:
3551
.
14
Miyazaki
,
K.
,
M.
Miyazaki
,
Y.
Guo
,
N.
Yamasaki
,
M.
Kanno
,
Z.
Honda
,
H.
Oda
,
H.
Kawamoto
,
H.
Honda
.
2010
.
The role of the basic helix-loop-helix transcription factor Dec1 in the regulatory T cells
.
J. Immunol.
185
:
7330
7339
.
15
Sun
,
H.
,
B.
Lu
,
R. Q.
Li
,
R. A.
Flavell
,
R.
Taneja
.
2001
.
Defective T cell activation and autoimmune disorder in Stra13-deficient mice
.
Nat. Immunol.
2
:
1040
1047
.
16
Li
,
C.
,
B.
Zhu
,
Y. M.
Son
,
Z.
Wang
,
L.
Jiang
,
M.
Xiang
,
Z.
Ye
,
K. E.
Beckermann
,
Y.
Wu
,
J. W.
Jenkins
, et al
2019
.
The transcription factor Bhlhe40 programs mitochondrial regulation of resident CD8+ T cell fitness and functionality
. [Published erratum appears in 2020 Immunity 52: 201–202.]
Immunity
51
:
491
507.e7
.
17
Son
,
Y. M.
,
I. S.
Cheon
,
Y.
Wu
,
C.
Li
,
Z.
Wang
,
X.
Gao
,
Y.
Chen
,
Y.
Takahashi
,
Y.-X.
Fu
,
A. L.
Dent
, et al
2021
.
Tissue-resident CD4+ T helper cells assist the development of protective respiratory B and CD8+ T cell memory responses
.
Sci. Immunol.
6
:
eabb6852
.
18
Rauschmeier
,
R.
,
A.
Reinhardt
,
C.
Gustafsson
,
V.
Glaros
,
A. V.
Artemov
,
J.
Dunst
,
R.
Taneja
,
I.
Adameyko
,
R.
Månsson
,
M.
Busslinger
,
T.
Kreslavsky
.
2022
.
Bhlhe40 function in activated B and TFH cells restrains the GC reaction and prevents lymphomagenesis
.
J. Exp. Med.
219
:
e20211406
.
19
Jarjour
,
N. N.
,
E. A.
Schwarzkopf
,
T. R.
Bradstreet
,
I.
Shchukina
,
C.-C.
Lin
,
S. C.-C.
Huang
,
C.-W.
Lai
,
M. E.
Cook
,
R.
Taneja
,
T. S.
Stappenbeck
, et al
2019
.
Bhlhe40 mediates tissue-specific control of macrophage proliferation in homeostasis and type 2 immunity
.
Nat. Immunol.
20
:
687
700
.
20
Rauschmeier
,
R.
,
C.
Gustafsson
,
A.
Reinhardt
,
N.
A-Gonzalez
,
L.
Tortola
,
D.
Cansever
,
S.
Subramanian
,
R.
Taneja
,
M. J.
Rossner
,
M. H.
Sieweke
, et al
2019
.
Bhlhe40 and Bhlhe41 transcription factors regulate alveolar macrophage self-renewal and identity
.
EMBO J.
38
:
e101233
.
21
Sun
,
J. C.
,
J. N.
Beilke
,
L. L.
Lanier
.
2009
.
Adaptive immune features of natural killer cells
. [Published erratum appears in 2009 Nature 457: 1168.]
Nature
457
:
557
561
.
22
Geary
,
C. D.
,
C.
Krishna
,
C. M.
Lau
,
N. M.
Adams
,
S. V.
Gearty
,
Y.
Pritykin
,
A. R.
Thomsen
,
C. S.
Leslie
,
J. C.
Sun
.
2018
.
Non-redundant ISGF3 components promote NK cell survival in an auto-regulatory manner during viral infection
.
Cell Rep.
24
:
1949
1957.e6
.
23
Diaz-Salazar
,
C.
,
R.
Bou-Puerto
,
A. M.
Mujal
,
C. M.
Lau
,
M.
von Hoesslin
,
D.
Zehn
,
J. C.
Sun
.
2020
.
Cell-intrinsic adrenergic signaling controls the adaptive NK cell response to viral infection
.
J. Exp. Med.
217
:
e20190549
.
24
Martínez-Llordella
,
M.
,
J. H.
Esensten
,
S. L.
Bailey-Bucktrout
,
R. H.
Lipsky
,
A.
Marini
,
J.
Chen
,
M.
Mughal
,
M. P.
Mattson
,
D. D.
Taub
,
J. A.
Bluestone
.
2013
.
CD28-inducible transcription factor DEC1 is required for efficient autoreactive CD4+ T cell response
.
J. Exp. Med.
210
:
1603
1619
.
25
Piper
,
C.
,
V.
Zhou
,
R.
Komorowski
,
A.
Szabo
,
B.
Vincent
,
J.
Serody
,
M.-L.
Alegre
,
B. T.
Edelson
,
R.
Taneja
,
W. R.
Drobyski
.
2020
.
Pathogenic Bhlhe40+ GM-CSF+ CD4+ T cells promote indirect alloantigen presentation in the GI tract during GVHD
.
Blood
135
:
568
581
.
26
Huynh
,
J. P.
,
C.-C.
Lin
,
J. M.
Kimmey
,
N. N.
Jarjour
,
E. A.
Schwarzkopf
,
T. R.
Bradstreet
,
I.
Shchukina
,
O.
Shpynov
,
C. T.
Weaver
,
R.
Taneja
, et al
2018
.
Bhlhe40 is an essential repressor of IL-10 during Mycobacterium tuberculosis infection
.
J. Exp. Med.
215
:
1823
1838
.
27
Jarjour
,
N. N.
,
T. R.
Bradstreet
,
E. A.
Schwarzkopf
,
M. E.
Cook
,
C.-W.
Lai
,
S. C.-C.
Huang
,
R.
Taneja
,
T. S.
Stappenbeck
,
S. J.
Van Dyken
,
J. F.
Urban
, Jr
., and
B. T.
Edelson
.
2020
.
BHLHE40 promotes TH2 cell-mediated antihelminth immunity and reveals cooperative CSF2RB family cytokines
.
J. Immunol.
204
:
923
932
.
28
Madera
,
S.
,
J. C.
Sun
.
2015
.
Cutting edge: stage-specific requirement of IL-18 for antiviral NK cell expansion
.
J. Immunol.
194
:
1408
1412
.

Supplementary data