Abstract
T follicular helper (Tfh) cells support antibody responses and are a critical component of adaptive immune responses to respiratory viral infections. Tfh cells are regulated by a network of signaling pathways that are controlled, in part, by transcription factors. The aryl hydrocarbon receptor (AHR) is an environment-sensing transcription factor that modulates many aspects of adaptive immunity by binding of a range of small molecules. However, the contribution of AHR signaling to Tfh cell differentiation and function is not known. Herein, we report that AHR activation by three different agonists reduced the frequency of Tfh cells during primary infection of C57Bl/6 mice with influenza A virus (IAV). Further, using the high affinity and AHR-specific agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), we show that AHR activation reduced Tfh cell differentiation and T-cell dependent B cell responses. Using conditional AHR knockout mice, we demonstrated that alterations of Tfh cells and T-cell dependent B cell responses after AHR activation required the AHR in T cells. AHR activation reduced the number of T follicular regulatory (Tfr) cells; however, the ratio of Tfr to Tfh cells was amplified. These alterations to Tfh and Tfr cells during IAV infection corresponded with differences in expression of BCL6 and FOXP3 in CD4+ T cells and required AHR to have a functional DNA-binding domain. Overall, these novel findings support that the AHR modulates Tfh cells during viral infection, which has broad reaching consequences for understanding how environmental factors contribute to variation in immune defenses against infectious pathogens, such as influenza and SARS-CoV-2.
Introduction
During the past several decades, there have been major advances in the knowledge of immune responses to viral infection and ways to reduce the impact of pathogenic viruses on human health. In spite of these developments, respiratory viral infections remain among the major causes of illness and death (1). The SARS-CoV-2 pandemic has brought respiratory viruses to the forefront; yet, even prior to the emergence of this new threat, other viruses such as influenza viruses, have been persistent causes of epidemics and pandemics (2). Among key unanswered questions about pathogenic viruses are uncertainties about the drivers of disparities in responses to infection at the individual and population level (3, 4). It is largely acknowledged that environment factors, such as contaminated water, affect the emergence and spread of communicable diseases. Yet, the impact of chemicals from our environment on immune responses to infection is often overlooked. In part, this reflects the complex connections between the environment, host, and pathogens (3). Environmental factors, ranging from pollutants to nutritional sources, can modify immune responses in a manner that can be protective or detrimental (4). However, the causal relationships and cellular mechanisms are poorly understood. Despite these uncertainties, there is mounting evidence that environmental exposures are important contributors to differences in antiviral immune responses (3, 5).
Ultimately, the adaptive immune response is responsible for resolving infection and building long-term immunity (6). Thus, environmental agents that influence the function of the adaptive immune system have the potential to modulate disease severity. Several epidemiological studies have reported associations between exposure to pollutants that bind the aryl hydrocarbon receptor (AHR) and increased severity of respiratory infections, as well as dampened responses to vaccination (7–9). The AHR is an environmental-sensing transcription factor that was initially discovered as an intracellular receptor for a broad class of persistent pollutants, which includes dioxins and polychlorinated biphenyls (10). In addition to pollutants, AHR ligands include small molecules derived from commonly consumed foods, metabolites from microorganisms, and several pharmaceuticals (10, 11). Thus, the AHR is a conduit for environmental signals to modulate the magnitude and nature of immune responses (3, 11). Indeed, in animal models, changes to adaptive immunity are among the most consistently observed consequences of AHR activation (10–12). CD4+ T cells, for example, play a pivotal role in adaptive immune responses to influenza and other respiratory viruses (13, 14). During acute infection with influenza A virus (IAV), CD4+ T cells clonally expand and differentiate into functionally distinct effector subsets, which include T helper 1 (Th1), Th17, T follicular helper (Tfh), and regulatory T (Tregs) cells (14). During primary IAV infection in mice, AHR activation reduces the frequency of Th1 cells and increases the frequency of Tregs (15). In other disease models, AHR activation alters the frequency of Th1, Th2, Th17, and Treg cells, although the affected subset(s) and direction of change varies depending on experimental system (10, 11). Moreover, AHR modulation of CD4+ T cells has been associated with changes in disease progression and outcome (16–22).
In contrast, relatively little research has examined the impact of AHR activation on Tfh cells. A recent report of IAV infection in mice, suggested that in addition to modulating other T cell populations, AHR activation likely also affects Tfh cells (15). This is intriguing for two reasons. First, Tfh cells are essential to establish and support germinal centers, which are the main sites in which B cells produce high-affinity, isotype switched antibodies and generate antibody-secreting plasma cells (23). Second, activation of the AHR perturbs antibody responses to immune challenges, including IAV infection (15, 24, 25). However, the impact of AHR activation on Tfh cells and the contribution of AHR signaling in CD4+ T cells on the germinal center response during viral infection is unclear. To address this knowledge gap, we determined the impact of AHR activation on Tfh cell differentiation and proliferation over the course of acute IAV infection. We also evaluated the consequences of AHR activation on key Tfh cell functions. Using conditional AHR knockout mice, we determined whether the AHR within the CD4+ cell lineage directly contributes to observed changes in Tfh cells, germinal center B cells, plasma cells, and virus-specific antibodies.
Methods and Methods
Mice and Treatments.
C57Bl/6 (B6) mice (age 5–6 weeks) were purchased from the Jackson Laboratory (Bar Harbor, ME). Initial breeding stocks of B6 AHRfx/fx and AHRdbd/dbd (26, 27) mice were provided by Christopher Bradfield (University of Wisconsin, Madison, WI), and B6.Cg-Tg(CD4-cre)1Cwi (CD4Cre) mice were purchased from the Jackson Laboratory (28). DNA isolated from tail biopsies was used to genotype AHRfx/fx, AHRdbd/dbd, CD4Cre, and CD4CreAHRfx/fx mice by PCR (26, 27, 29). All primers were purchased from Integrated DNA Technologies (San Diego, CA). The AHRfx allele was detected using the primers OL4064 (5′-CAGTGGGAATAAGGCAAGAGTGA-3′) and OL4088 (5′-GGTACAAGTGCACATGCCTGC-3′) (26). The following primers were used to detect the AHRdbd allele: OL941 (5′-CTGAGGGGACGTTTTAATG-3′) and OL942 (5′-AACATTTGCACTCATGGATAG-3′) (27). For some experiments, CD4Cre mice were crossed with AHRfx/fx mice. To verify Ahr excision from CD4+ T cells in CD4CreAHRfx/fx mice, DNA was isolated from purified CD4+ T cells using DirectPCR Lysis Reagent (Viagen Biotech, Los Angeles, CA) with 150 μg of Proteinase K (Invitrogen), and PCR was performed using the primers OL4062 (5′-GTCACTCAGCATTACACTTTCTA-3′), OL4064 (5′-CAGTGGGAATAAGGCAAGAGTGA-3′), and OL4088 (5′-GGTACAAGTGCACATGCCTGC-3′) (26). The CD4Cre transgene was detected using the primers cd4cremu (5′-TTAGGGTGGGGCTCAGAAGG-3′), cd4creco (5′-AACTTCACAGC TCAGATGC-3′), and cd4crewt (5′-ACCTGGAT CC ACAAA CTTG-3′) (29). Mice were maintained at the University of Rochester Medical Center (URMC). All mice were housed in microisolator cages in a specific-pathogen free facility, and were provided food (LabDiet 5010, St. Louis MO) and water ad libitum. The animals were housed on a 12 hr light/dark cycle, and ambient temperatures were between 20–22°C. Commercially obtained mice were randomly housed with 3–5 mice per cage. Mice bred on-site were housed with same sex litter mates at weaning, and then randomly assigned to treatment groups at 6–12 weeks of age, with 3–5 mice per cage. All data presented are from female mice that were 6–12 weeks of age at the time of experiments, and each point in time relative to infection included 3–8 mice in each treatment group.
To activate the AHR, mice were administered the prototype AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; ≥99% purity, Cambridge Isotope Laboratories, Tewksbury, MA). TCDD was dissolved in anisole and diluted in peanut oil. B6 mice were randomly assigned to a group and administered a single dose orally of either TCDD (10 μg/kg body weight) or the vehicle control (peanut oil-anisole). AHRdbd/dbd mice express the Ahrd/d allele, which encodes a protein with 10-fold lower binding affinity for TCDD compared to the Ahrb/b allele expressed by B6 mice (30). Thus, AHRdbd/dbd mice were treated with 100 μg TCDD/kg body weight (31). For some experiments, mice were administered a different AHR agonist. Specifically, either 2-(1H-Indol-3-ylcarbonyl)-4-thiazolcarboxylic acid methyl ester (ITE; Tocris Bioscience, Minneapolis, MN) or Kynurenic acid (KYNA; Tocris Bioscience, Minneapolis, MN). ITE was dissolved in acetone and diluted in peanut oil. Mice were injected i.p. with 10 mg ITE/kg body weight/day, or the peanut oil acetone vehicle control, starting one day prior to infection (15). KYNA was dissolved in autoclaved drinking water at 250 mg/L, and provided to mice via the drinking water at a concentration of 25 mg/L (32). Mice were given the KYNA-containing water starting 14 days prior to infection and maintained on this water until they were sacrificed. The KYNA solution was made and replenished every 3–5 days. Another group of mice was given sterile, vivarium drinking water as controls for KYNA. For some experiments, mice were injected i.p. with 100 μg of BrdU 3 h prior to sacrifice (33, 34). All animal treatments had prior approval of the Institutional Animal Care and Use Committee of the University of Rochester. The University is accredited through the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC). Animals were treated humanely and with due consideration to alleviation of any distress and discomfort. All U.S. Public Health Service Policy on Human Care and Use of Laboratory Animals guidelines for the handling of vertebrate animals were followed.
Influenza A virus.
Influenza A virus (IAV) strain A/HKx31 (HKx31; H3N2) was propagated according to previously described methods (25, 35). Briefly, fertilized chicken eggs (Charles River, Wilmington, MA) were inoculated with 0.05 hemagglutinating units (HAU) influenza virus in 100 μL of Hank’s balanced salt solution (HBSS), containing 10 mM HEPES. Inoculated eggs were incubated for 48 h at 37°C, followed by refrigeration overnight at 4°C. Allantoic fluid was harvested under aseptic conditions, centrifuged, and frozen at −80°C. The titer of the allantoic fluid was determined by hemagglutination of avian erythrocytes (35). Mice were anesthetized by i.p. injection of avertin (2,2,2-tribromoethanol; Sigma Aldrich, St. Louis, MO) and infected intranasally (i.n.) with a sublethal dose (120 HAU virus in 25 μL) diluted in endotoxin-tested PBS (15, 25). Infections were performed in the morning (between 8:00 and 10:00 AM). All work with infectious agents was conducted with prior approval of the Institutional Biosafety Committee of the University of Rochester, following guidelines of the NIH/CDC.
Tissue collection and preparation of immune cells.
Tissue collection was initiated in the morning. Mice were sacrificed using lethal dose of anesthetic (e.g., Euthasol, Virbac, Greeley, CO), and either cervical dislocation or exsanguination. Blood was collected by cardiac puncture, with syringes containing 100 μL of 250 U/mL of heparin sodium salt (Grade I-A, ≥180 USP units/mg; Sigma Aldrich), and plasma was stored at −80 °C. Immune cells were isolated from the mediastinal lymph nodes (MLN), as previously described (36, 37). Briefly, single-cell suspensions were prepared by pressing lymph nodes from a single mouse between the frosted ends of two microscope slides. MLN cells were suspended in cold HBSS containing 2.5% FBS (Hyclone, Logan, UT). Erythrocytes were removed by incubating pelleted cells in a solution containing 0.15 M NH4Cl, 10 mM NaHCO3, and 1 mM EDTA for 5 min at room temperature. Erythrocyte lysis was terminated by adding equal volume of cold HBSS containing 2.5% FBS. Cell suspensions were then washed and passed through a 70μM cell strainer. After centrifugation (200 × g; 6 min), cell pellets were re-suspended in cold HBSS, 2.5% FBS for enumeration using a TC10 automated cell counter (BioRad, Hercules, CA), or a hemocytometer and Trypan blue exclusion. Cells were maintained at 4°C until labeling with fluorescent-conjugated antibodies, which was performed the same day.
Flow cytometry.
Single-cell suspensions containing 2 × 106 lymph node cells were incubated with anti-mouse CD16/32 monoclonal antibody (mAb; clone93) for 10 min at 4° C to block non-specific staining. Cells were then incubated for 20 min at 4° C with previously determined optimal concentrations of fluorochrome-conjugated monoclonal antibodies against extracellular antigens (Supplemental Table I). After cell surface labeling, cells were fixed using 2% formaldehyde in PBS for 20 min, and analyzed directly by flow cytometry, or permeabilized for labeling of intracellular constituents (Supplemental Table I). Specifically, to detect BCL6 and FOXP3, cells were fixed and permeabilized using the FOXP3 Staining Kit (eBioscience, San Diego, CA) followed by incubation with anti-mouse CD16/32 mAb to block intracellular non-specific staining for 10 min at room temperature. Cells were then incubated with antibodies to BCL6 and FOXP3 for 45 min at room temperature. The BrdU Flow Kit (BD Biosciences, Franklin Lakes, NJ) was used to detect BrdU, and staining was performed following the manufacturer’s instructions. Cells were then stained with intracellular antibodies to BrdU for 20 min at room temperature. To identify live cells, prior to extracellular staining, cells were incubated for 30 min in PBS at 4°C with fixable viability dye eFluor 506 (eBioscience). Tfh cells were defined as CD4+ T cells that were CD44hiCXCR5hiPD1hi (15, 38, 39). Tfr cells were defined as CD4+CD44hiCXCR5hiPD1hiFOXP3+ cells (40). Germinal center (GC) B cells were defined as CD3−B220+CD95+GL7+ cells (41). Plasma cells were identified as CD3−B220intCD138hi cells (42). Fluorescence minus one (FMO) controls were used to determine non-specific fluorescence and define gating parameters. Data (500,000 events per sample) were collected using an LSRII flow cytometer (BD Biosciences). Data were analyzed using the FlowJo software program (Version 10; Tree Star, Ashland, OR).
Anti-influenza virus antibody ELISA.
Relative levels of influenza virus-specific antibodies in plasma were measured using enzyme linked immunosorbent assays (ELISA) as previously described (15, 24). In summary, 96-well ELISA microplates (MICROLON®, flat-bottom, Greiner Bio-one, Monroe, NC) were coated with purified HKx31 IAV (5 μg/mL, Charles River, Wilmington, MA) at 37°C overnight. Plates were washed with PBS containing 0.05% Tween 20 in between each step. To reduce non-specific binding, PBS containing 5% BSA was added to all wells, and plates incubated at 4°C for 2 h. Plasma samples were serially diluted in PBS 5% BSA from 1:100 to 1:25600. Diluted plasma was added to the plates and incubated overnight (16–20 h) at 4°C. Biotinylated goat anti-mouse IgG2a antibodies (Southern Biotechnology) were diluted to 1:5000 from commercial stock in PBS 5% BSA, and added to the plates, which were incubated at room temperature for 45 min. Avidin-peroxidase (1:400 dilution in PBS 5% BSA; Sigma Aldrich) was added directly to the plates and incubated at room temperature for 30 min. 2,2′-Azino-bis(3-ethylbenzothiazoline-6-sulphonic acid) with 0.03% H202 was used to induce colorimetric change. Absorbance values were read at 405 nm using a SpectraMax Plate reader (Molecular Devices) To measure antibody affinity, PBS containing 1.0M guanidine hydrochloride (GuHCL; Sigma Aldrich), 0.2%Tween20 and 10 mg/mL BSA was added to the plates for 15 min at room temperature prior to adding biotinylated isotype-specific antibodies (43).
Statistical analysis.
Data were analyzed using JMP Pro 15 software (SAS Institute Inc.). Differences between means of two treatment groups at a single point in time were evaluated using a Student’s t-test. A two-way ANOVA followed by a Tukey honest significant difference (HSD) test was used to analyze means between treatment groups across time or mouse genotype. A two-way ANOVA followed by a Dunnett’s post hoc test was used to compare means within a treatment group across time. The specific statistical test used is indicated in each figure legend. Differences were considered statistically significant when p-values were ≤ 0.05. Error bars on all graphs and tables represent standard error of mean (SEM).
Results
AHR activation modulates Tfh cells over the course of IAV infection.
The AHR can be activated by small molecule agonists derived from a range of sources (44). For example, some AHR ligands, such as kynurenic acid (KYNA), are products of tryptophan metabolism and can arise in vivo. Other small molecules that bind AHR, such as 2-(1H-Indol-3-ylcarbonyl)-4-thiazolecarboxylic acid methyl ester (ITE), are derived from natural substances and are produced synthetically (45). Xenobiotics, including industrial chemicals and pollutants, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), also activate the AHR (44). We compared the impact of these three different AHR-binding molecules on Tfh cells during primary IAV infection. Tfh cells can be identified by flow cytometry as CD4+ T cells that express high levels of CD44, PD1, and CXCR5 (Fig. 1A) (15, 23, 38, 39). Treatment of mice with KYNA (Fig. 1B), ITE (Fig. 1C), and TCDD (Fig. 1D) significantly reduced the percentage of Tfh cells compared to infected mice treated with vehicle (control). Similarly, exposure to each AHR agonist significantly reduced the number of Tfh cells in infected mice (Fig. 1E–G). To further interrogate the effects of AHR activation on Tfh cells, we selected TCDD for three key reasons: it has a well-defined in vivo distribution and metabolism (46–48), it binds the AHR with high affinity and specificity (49, 50), and there is evidence that AHR-binding dioxin-like pollutants, affect humoral immune defenses in human populations (7, 51, 52). Therefore, TCDD is an excellent compound to study the AHR because it does not act through other receptors and pathways, maintains AHR activation for the duration of acute primary infection, and is relevant to human health.
FIGURE 1. AHR activation modulates Tfh cells during IAV infection.

C57BL/6 mice (6 weeks old, female) were administered the indicated AHR ligands as follows: TCDD (10 μg/kg body weight) was administered once by gavage one day prior to infection, ITE (10 mg/kg body weight) was given i.p. daily on days −1 to +8 relative to infection, and KYNA (25 mg/L) was administered via drinking water, starting 14 days prior to infection, and continuing until termination of the experiment. Control mice received the appropriate vehicle following the same treatment route and dosing schedule. Mice were infected intranasally (i.n.) with 120 hemagglutinating units (HAU) influenza A virus (IAV, strain HKx31; H3N2). Mediastinal lymph nodes (MLN) were harvested on day 9 post-infection for analysis by flow cytometry. (A) FACS plots depict gating strategy for identifying Tfh cells (CD4+CD44hiCXCR5hiPD1hi). Prior gating excluded doublets and non-viable cells, and gated regions were defined using FMO controls. (B-D) Bar graphs indicate the percent of CD4+ T cells that were Tfh cells in mice treated with (B) KYNA, (C) ITE, and (D) TCDD on day 9 of infection. Bar graphs show the number of Tfh cells in (E) KYNA, (F) ITE, and (G) TCDD treated mice. *Denotes p≤ 0.05 compared to vehicle treatment group (two-sided Student’s t-test). All experiments had 6–8 mice in each treatment group. Experiments using ITE and TCDD were performed, independently, at least twice, whereas the effects of KYNA on Tfh cells were measured once.
The activation and differentiation of naïve CD4+ T cells to Tfh cells is a multi-factorial process that occurs over time (23). AHR activation could modulate Tfh cells at several different phases; thus, we evaluated the impact of AHR activation on Tfh cells over the course of acute primary IAV infection. In the control group, Tfh cells became clearly discernable by the 5th day of infection, peaked around the 7–9th day after infection, and then contracted (Fig. 2A). For instance, 5 days post-infection the percent of Tfh cells was over 55-fold higher than uninfected mice (Fig. 2A) and remained significantly higher than naïve mice till day 14 (Supplemental Table II). The number of Tfh cells showed a similar pattern (Fig. 2B). Compared to infected controls, AHR activation by the prototype AHR agonist, TCDD significantly reduced the percent (Fig. 2A) and number (Fig. 2B) of Tfh cells over the course of infection. For example, on day 5 post-infection, the percent of Tfh cells in TCDD treated mice was 70% less than in vehicle-treated infected mice and was not different from naïve mice (Fig. 2A and Supplemental Table II). Moreover, at the peak of the Tfh cell response, on day 9 post-infection, the number of Tfh cells was 15-fold lower in TCDD-treated mice compared to infected mice given the vehicle control (Fig. 2B). Thus, AHR activation suppressed the generation of Tfh cells over the course of infection.
FIGURE 2. AHR activation by TCDD reduces the percentage and number of Tfh cells over the course of IAV infection.

C57BL/6 mice were given TCDD or vehicle by gavage one day prior to infection with IAV. MLN were harvested on the indicated days relative to infection for analysis by flow cytometry. (A) FACS plots depict the percent of Tfh cells (CD4+CD44hiCXCR5hiPD1hi cells) on the indicated days relative to infection. The number on each plot denotes the mean percentage of CD4+ T cells (± SEM) that were defined as Tfh cells. (B) Line graph shows the number of Tfh cells over time. Closed circles indicate the vehicle group and open squares indicate the TCDD treatment group. Error bars indicate the SEM. The p-value of the 2-way ANOVA is shown on the graph. *Denotes p≤ 0.05 compared to vehicle treatment group on the same day relative to infection (2-way ANOVA, Tukey HSD). A complete list of p-values from the ANOVA and post-hoc tests is in Supplemental Table II. Data are representative of one independent time-course experiment with 48 mice in each treatment group. All mice were infected at the same time, and each point in time relative to infection represents 6–8 mice in each group.
AHR activation by TCDD suppresses the germinal center response during infection.
Given that a major function of Tfh cells is helping B cells in germinal centers (GC) (23), we determined whether reduction in Tfh cells observed when the AHR was activated correlated with diminished GC B cells. In vehicle treated mice, the percent (Fig. 3A) and number (Fig. 3B) of GC B cells were significantly greater than naive mice by the 9th day after infection (Supplemental Table II). On day 14 post-IAV infection, the number of GC B cells was over 500-fold greater than uninfected mice (Fig. 3B). However, AHR activation significantly reduced the percent (Fig. 3A) and number (Fig. 3B) of GC B cells in IAV infected mice. For instance, the percent of GC B cells in the TCDD treatment group did not significantly increase above what was observed in naïve mice until the 14th day after infection (Fig. 3A, Supplemental Table II). Moreover, the number of GC B cells in mice treated with TCDD never exceeded the number detected in uninfected mice at any point in time examined (Fig. 3B, Supplemental Table II). When integrated with examination of Tfh cells, there was a strong correlation between AHR-driven reductions in the number of GC B cells and Tfh cells (Fig. 3C).
FIGURE 3. AHR activation by TCDD suppresses the germinal center response during viral infection.

C57BL/6 mice were given TCDD or vehicle by gavage one day prior to infection with IAV. MLN and blood (plasma) were harvested on the indicated days relative to infection for analysis by flow cytometry and ELISA. (A-B) Line graphs show (A) the percentage of live cells defined as germinal center (GC) B cells (CD3−B220+CD95+GL7+), and the (B) number of GC B cells. The p-value of the 2-way ANOVA is shown on the graph. *Denotes p≤ 0.05 for treatment groups compared on the same day after infection (2-way ANOVA, Tukey HSD). (C) The scatter plot depicts the number of Tfh cells versus the number of GC B cells on days 9 and 14 post-infection. The best-fit line was determined by linear regression. Open squares denote TCDD and closed circles indicate vehicle treated group. (D) Representative plots show the mean percentage of live cells defined as plasma cells (CD3-B220intCD138hi) in the MLN on day 14 of infection. (E) The bar graph shows the number of plasma cells. (F,G) Anti-IAV antibody levels in serially-diluted plasma (1:100 to 1:25600) were determined using ELISA. (F) Graph shows the mean circulating levels of IAV-specific IgG2a (1:1600 dilution). *Denotes p≤ 0.05 compared to vehicle (two-sided Student’s t-test). (G) Line graph depicts levels IAV-specific IgG2a in serially diluted plasma that was either untreated or treated with 1M GuHCL. Samples from vehicle treated mice are denoted with circles, and samples from TCDD treated mice with squares. Open symbols denote samples that were not treated with GuHCl (control), while closed symbols indicate treatment with 1M GuHCl. Samples from uninfected (naïve) mice are denoted using triangles. (H) Bar graphs show the median area under the curve for all plasma dilutions. * p≤ 0.05 for treatment groups that are significantly different from on another (2-way ANOVA, Tukey HSD). A compilation of p-values from the 2-way ANOVA and post-hoc tests are in Supplemental Table II. All data are shown as mean ± SEM, and are representative of two independent assessments with 6–8 mice per treatment group at each point in time.
Other B cell functions that depend on Tfh cells are the generation of plasma cells and high-affinity antibodies (23). AHR activation reduced the percentage (Fig. 3D) and number (Fig. 3E) of plasma cells by >80% compared to infected mice in the control group. Consistent with the diminished number of GC B cells and plasma cells, AHR activation reduced the levels of IAV-specific IgG2a by 7-fold (Fig. 3F). Antibody avidity can be measured using a modified ELISA that incorporates a chaotropic agent, such as guanidine hydrochloride (GuHCL), which disrupts low avidity antigen-antibody interactions (43). In plasma from control-treated infected mice, treatment with GuHCL significantly reduced levels of anti-IAV specific antibody by about 30%, such that the area under the curve (AUC) declined from a mean AUC of 3.65 to 2.45 (Fig. 3G–H). In contrast, treatment with GuHCL did not cause a significant change in IAV-specific antibodies in plasma from infected mice in the TCDD treatment group (Fig. 3G–H). At first glance, this might suggest greater antibody avidity compared to vehicle; however, the level of anti-IAV antibodies in the TCDD treated group was not different from plasma of naïve animals (Fig 3G–H). When integrated together, these results show that AHR activation reduced the generation of Tfh cells and attenuated key Tfh cell functions. The generation of Tfh cells involves CD4+ T cell proliferation and the upregulation of key molecules that drive Tfh cell fate commitment. Therefore, the reduced frequency and function of Tfh cells observed could result from AHR-mediated impact on proliferation, or the expression of critical transcriptional regulators that modulate Tfh cells, such as BCL6, and these are not mutually exclusive.
AHR activation attenuates CD4+ T cell proliferation in response to viral infection
To determine whether AHR activation affects Tfh cell proliferation, we used BrdU to label dividing cells in vivo (34). In mice treated with vehicle, BrdU+ Tfh cells were discernable 3 days after infection (Fig. 4A). The percent (Fig. 4A) and number (Fig. 4B) of BrdU+ Tfh cells peaked on the 5th day and steadily declined thereafter in infected controls. This pattern was similar in mice in which the AHR was activated (Fig. 4A); however, the number of BrdU+ Tfh cells was significantly reduced (Fig. 4B). For instance, on day 5 post-infection, which was the peak day of Tfh cell proliferation in vehicle controls, there were >3 times fewer BrdU+ Tfh cells in the TCDD-treated group (Fig. 4B). When assessing the entire CD4+ T cell population, CD4+ T cells proliferated with similar kinetics as Tfh cells after infection (Fig. 4C–D). Consistent with prior reports using other antigenic challenges (53, 54), AHR activation blunted CD4+ T cell proliferation over the course of IAV infection (Fig. 4C–D). These findings suggest that one means by which AHR activation dampens the frequency of Tfh cells is, in part, by diminishing CD4+ T cell proliferation.
FIGURE 4. AHR activation attenuates CD4+ T cell proliferation in response to viral infection.

C57BL/6 mice were given TCDD or vehicle control one day prior to infection with IAV. Mice were administered (i.p.) 100 μg BrdU 3 hr prior to tissue collection. MLN were obtained on the indicated days relative to infection for analysis by flow cytometry. (A-B) The graphs indicate the (A) percent and (B) number of Tfh cells (CD4+CD44hiCXCR5hiPD1hi cells) that were BrdU+ prior to and up to 14 days following IAV infection. Closed circles indicate mean values from vehicle group and open squares indicate the TCDD treatment group. (C-D) Line graphs show the (C) percent of CD4+ T cells that were BrdU+ and (D) number of BrdU+CD4+ T cells. The p-values on graphs were derived from the 2-way ANOVA. *Denotes p≤ 0.05 compared vehicle treated mice on the same day post-infection (2-way ANOVA, followed by Tukey HSD). Data are presented as mean ± SEM, and are from time-course experiment, in which all mice were infected at the same time. Each point in time relative to infection included 6–8 mice from each treatment group. A complete list of p-values from the 2-way ANOVA and post-hoc tests are in Supplemental Table III.
Fewer Tfh cells correlates with diminished levels of BCL6 in CD4+ T cells and alterations to T follicular regulatory (Tfr) cells
The transcriptional regulator BCL6 is required for Tfh cell differentiation (55). In response to IAV infection, CD4+ T cells significantly upregulated BCL6, which was 2.6-fold greater on day 5 post-infection compared to uninfected mice (Fig. 5A). Compared to day 5 post-infection, BCL6 levels in CD4+ T cells were about 2-fold lower on day 7, but were still significantly elevated compared to naïve mice. Moreover, 9 days after infection, BCL6 levels were higher than on day 7, but not as high as on day 5. By 14 days post-infection, BCL6 levels were similar to CD4+ T cells from uninfected mice (Fig. 5A). However, this biphasic expression pattern was not observed after AHR activation (Fig. 5B). Compared to vehicle treated mice, AHR activation reduced overall levels of BCL6 in CD4+ T cells (p=0.0008; 2-way ANOVA). Similar to the vehicle group, infection triggered an initial elevation in BCL6 (e.g., 5 days after infection compared to naïve); although the level of BCL6 was significantly lower than it was in CD4+ T cells from vehicle treated mice at this point in time (Fig. 5B). Moreover, 7 and 9 days after infection, BCL6 levels in CD4+ T cells from TCDD treated mice were not different from uninfected mice (Fig. 5B). Regression analysis showed a significant correlation between the level of BCL6 expression in CD4+ T cells and percentage of Tfh cells in vehicle and TCDD treated mice on day 9 post-infection (Fig. 5C). Thus, AHR-driven reduction of BCL6 levels in CD4+ T cells correlates with decreased Tfh cells.
FIGURE 5. BCL6 expression is diminished in CD4+ T cells after AHR activation.

C57BL/6 mice were administered TCDD or vehicle control one day prior to infection. MLN were harvested on the indicated days relative to infection for analysis by flow cytometry. (A-B) Histogram shows BCL6 expression in CD4+CD44hi T cells from (A) vehicle and (B) TCDD treated mice over the course of infection. The number on each histogram denotes the mean fluorescence intensity (MFI) of BCL6 in CD4+CD44hi cells. The fluorescence minus one (FMO) control is denoted with light grey. Asterisks (*) indicate p ≤ 0.05 compared to vehicle (2-way ANOVA, Tukey HSD) at that specific point in time. The hashtags (#) specify p ≤ 0.05 compared to naïve mice (day 0) in the same treatment group (2-way ANOVA, Dunnett’s test). A complete set of p-values from 2-way ANOVA and post-hoc tests are in Supplemental Table IV. (C) Regression analysis of the percentage of Tfh cells and BCL6 MFI in CD4+CD44hi T cells on day 9 post-infection. Best-fit line was determined by linear regression. Closed circles denote vehicle and open squares denote TCDD treated groups. Data are mean ± SEM and are from a time course experiment. There were 6–8 mice/treatment group at each point in time.
Another category of CXCR5hiPD1hiCD4+ T cells are T follicular regulatory (Tfr) cells, which can be distinguished from Tfh cells based on the expression of the transcription factor FOXP3 (Fig. 6A) (40, 56). Whether AHR activation affects Tfr cells has not been investigated. During IAV infection, AHR activation significantly reduced the number of Tfr cells (Fig. 6B). However, the percentage CD4+CD44hiCXCR5hiPD1hi cells that expressed FOXP3+ was elevated after AHR activation (Fig. 6C), such that the ratio of Tfr to Tfh cells was increased by 2.6-fold compared to infected mice given vehicle control (Fig. 6D). These results suggest that AHR activation shifts the balance of immunostimulatory and immunosuppressive follicular CD4+ T cells during IAV infection.
FIGURE 6. AHR activation modifies the balance of Tfh cells and T follicular regulatory (Tfr) cells.

C57BL/6 mice were given TCDD or vehicle control one day prior to infection. MLN were harvested on 9 days after infection for analysis by flow cytometry. (A) Representative flow plots show the gating strategy to identify Tfr cells (CD4+CD44hiCXCR5hiPD1hiFOXP3+). (B) The bar graph shows the number of Tfr cells in the MLN. (C) Graph depicts the percent of follicular CD4+ T cells (CD4+CD44hiCXCR5hiPD1hi) that were FOXP3+. (D) Stacked bar graph shows the ratio of Tfr to Tfh cells. The number beside each bar indicates the percentage of Tfr cells. *Denotes p≤ 0.05 compared to vehicle (two-sided Student’s t-test). Data are mean ± SEM and are representative of two independent experiments with 3–7 mice/group in each experiment.
The AHR modulates Tfh cells via a mechanism that requires AHR’s intrinsic DNA binding domain.
After ligand binding and translocation into the nucleus, the activated AHR-ligand complex binds a highly conserved core DNA sequence called an aryl hydrocarbon response element (AHRE) to modulate gene expression (57). There is also some evidence that the AHR may influence gene expression via alternative pathways that are independent of AHR binding to AHREs (58, 59). To determine whether AHR activation affects Tfh cells through a mechanism that requires AHR’s DNA binding domain (DBD), we used AHR DBD mutant (AHRdbd/dbd) mice. These mice express an AHR protein that binds ligands, and translocates to the nucleus, but lacks its intrinsic DBD and cannot bind AHREs (27). In contrast to wild-type mice, exposure of AHRdbd/dbd mice to TCDD no longer reduced the percent (Fig. 7A) or number (Fig. 7B) of Tfh cells during infection with IAV. Correspondingly, AHR activation significantly reduced the MFI of BCL6 in CD4+ T cells of wild-type, but not mutant mice (Fig. 7C). Furthermore, unlike in wild-type animals, AHR activation did not alter the percent (Fig. 6D) and number (Fig. 6E) of Tfr cells in AHRdbd/dbd mice. In addition, the MFI of FOXP3 was significantly increased in CD4+ T cells from wild-type, but not mutant mice (Fig. 7F). Therefore, the AHR requires its ability to interact with DNA via its cognate DBD in order to alter Tfh, as well as Tfr cells during IAV infection.
FIGURE 7. The AHR modulates Tfh cells via a mechanism that requires AHR’s intrinsic DNA binding domain.

C57BL/6 (AHRwt/wt) and DNA binding mutant (AHRdbd/dbd) mice were administered vehicle or TCDD once by gavage one day prior to infection. 9 days after infection, mice were given 100 μg BrdU i.p. 3 hr prior to tissue collection. (A) Representative flow plots show the percentage of Tfh cells in the MLN. (B) Bar graph shows the number of Tfh cells. (C) Histograms show BCL6 expression in CD4+CD44hi T cells from vehicle (solid, grey line) and TCDD (dotted, black line) treated AHRwt/wt and AHRdbd/dbd mice. (D-E) Bar graphs show the (D) percent of CD4+CD44hiCXCR5hiPD1hi cells that are Tfr cells and the (E) number of Tfr cells. (F) Histograms show FOXP3 expression in CD4+CD44hi T cells from vehicle (solid, grey line) and TCDD (dotted, black line) treated AHRwt/wt and AHRdbd/dbd mice. The numbers on the histograms denote the MFI. *Signifies p≤ 0.05 compared to vehicle in treatment group (2-way ANOVA, Tukey HSD). Data are mean ± SEM and are representative of two independent experiments, each of which had 6–8 mice/group.
AHR modulation of Tfh cells and T cell-dependent B cell responses to IAV infection requires the AHR in CD4+ cells.
The AHR is broadly expressed in immune cells, including dendritic cells, B cells and CD4+ T cells, including Tregs and Tfh cells (10, 22, 60–66). To determine whether changes in Tfh cell differentiation and function observed upon AHR activation are intrinsic to CD4+ T cells, we crossed CD4Cre mice with AHRfx/fx mice to ablate AHR from CD4+ lineage cells. Similar to wild-type mice, AHR activation reduced the percent (Fig. 8A) and number (Fig. 8B) of Tfh cells AHRfx/fx mice. In addition, in AHRfx/fx mice, AHR activation reduced the number of Tfr cells (Fig. 8C), but significantly increased the percent (Fig. 8D) of Tfr cells. In contrast, infected CD4CreAHRfx/fx mice treated with TCDD no longer exhibited a change in the percent (Fig. 8A) or number (Fig. 8B) of Tfh cells. The percent (Fig. 8D) and number (Fig. 8C) of Tfr cells was also unchanged by AHR activation in CD4CreAHRfx/fx mice. To examine Tfh cell function, we assessed key B cell responses during infection that rely on Tfh cells. While there were fewer GC B cells in AHRfx/fx mice treated with TCDD (Fig 8E–F), in CD4CreAHRfx/fx mice, AHR activation did not affect the percent (Fig. 8E) or number (Fig. 8F) of GC B cells. Similarly, the percent (Fig. 8G) and number (Fig. 8H) of plasma cells in CD4CreAHRfx/fx were no longer repressed by AHR activation. In AHRfx/fx mice, TCDD treatment reduced levels of IAV-specific IgG2a compared to vehicle (Fig. 8I,8K), while vehicle and TCDD treatment groups generated comparable levels of IAV-specific IgG2a in CD4CreAHRfx/fx mice (Fig. 8J, 8K). Thus, in the absence of AHR expression in CD4+ T cells, the antibody response is protected from the suppressive effects of TCDD. Another observation from these studies was that, compared to infected AHRfx/fx mice, CD4CreAHRfx/fx mice produced significantly less IAV-specific antibodies (Fig. 8K). This suggests an intrinsic role of AHR in CD4+ T cells for optimal virus-specific antibody responses during acute primary infection. More broadly, these studies using CD4CreAHRfx/fx mice, indicate that AHR mediated events in CD4+ T cells modulate the generation and function of Tfh cells during IAV infection.
FIGURE 8. AHR modulation of Tfh cells and T cell-dependent B cell responses requires the AHR in CD4+ cells.

AHRfx/fx and CD4CreAHRfx/fx mice were administered vehicle or TCDD once by gavage one day prior to i.n infection with IAV. MLN were harvested 9 and 14 days after infection, and plasma was collected 14 days after infection. (A-D) Tfh and Tfr cells were assessed in MLN 9 days after infection by flow cytometry. (A) The percentage of all CD4+ T cells that were Tfh cells (CD4+CD44hiCXCR5hiPD1hi cells) in AHRfx/fx and CD4CreAHRfx/fx mice. (B) The number of Tfh cells in AHRfx/fx and CD4CreAHRfx/fx mice. (C-D) The (C) percent and (D) number of Tfr cells, defined as CD4+CD44hiCXCR5hiPD1hiFOXP3+ cells. GC B cells, plasma cells and IAV-specific antibody levels were measured 14 days after infection using flow cytometry and ELISA. (E) Representative flow plots show the percent of live cells defined as GC B cells (CD3−B220+CD95+GL7+) in each group day 14 post-infection. The number on each plot indicates the mean for each group of mice. (F) Bar graph shows the number of GC B cells in the MLN from AHRfx/fx and CD4CreAHRfx/fx mice. (G-H) Bar graphs show the (G) percent and (H) number of live cells defined as plasma cells (CD3−B220intCD138hi cells) in the MLN. (I-J) IAV-specific antibody levels in serially-diluted plasma were determined by ELISA. Line graphs show the relative level of IAV-specific IgG2a in (I) AHRfx/fx and (J) CD4CreAHRfx/fx mice treated with vehicle (black circles) and TCDD (open squares). The p-values of the 2-way ANOVA between vehicle and TCDD are shown on the graphs. (K) Bar graph shows the median AUC for each group. *Signifies p≤ 0.05 compared to vehicle in treatment group (2-way ANOVA, Tukey HSD). Data are mean ± SEM with 6–8 mice/treatment group, and are representative of two independent experiments.
Discussion
While there is a growing appreciation that environmental exposures contribute to the variation observed in immune responses to infection in the human population (3, 4), it is unclear how external factors influence antiviral immune defenses. The work reported expands our understanding of how the AHR influences adaptive immune responses, showing that in addition to other T cell subsets, the AHR modulates Tfh cells. Using a common respiratory virus, IAV, and the prototype agonist, TCDD, we demonstrated that triggering the AHR affected several cellular processes critical for generating Tfh cells. These included CD4+ T cell proliferation and differentiation, which directly correlated with reduced metrics of antiviral humoral immune defenses, such as fewer germinal center B cells, plasma cells, and virus-specific antibodies. Given that Tfh cells are gatekeepers of humoral immunity and play a critical role in the production of high-affinity antibodies, these new findings have broad implications for host defenses against viral pathogens. Moreover, multiple studies have reported associations between exposure to chemicals that bind the AHR and increased severity of respiratory illness and diminished immune responses to vaccination (7–9). In animal models, prior studies have linked AHR ligands to alterations in cell-mediated antiviral T cell responses (15); however, less is known about how AHR alters T cell responses related to the humoral arm of adaptive responses to viral infection. Given that antibody responses are essential for long-lasting immunity, this current study provides new insight into how AHR signaling in T cells contributes directly to antiviral humoral immune defenses.
One of the key discoveries reported herein was that AHR activation modulated the proliferation and differentiation of CD4+ T cells into Tfh cells, as well as attenuated key Tfh cell-dependent functions over the course of infection. Moreover, AHR within T cells is a central driver of AHR-mediated influence on humoral immunity to respiratory infection. Although AHR modulation of Tfh cells has not been explored in other disease contexts, this finding is consistent with studies that have pointed to CD4+ T cells as central to AHR-mediated changes in non-infectious disease models in mice (20, 67–69). For example, transfer of CD4+ T cells treated with the AHR ligand ITE into mice with colitis significantly improved disease score and reduced colonic inflammation (67). In another mouse colitis model, transfer of AHR deficient Tregs into mice worsened gut inflammation and histopathological changes (69). The AHR was also required in CD4+ T cells in order for AHR agonist treatment to increase pathogenic Th17 cells in a mouse model of rheumatoid arthritis (68). Other than autoimmune diseases, AHR activation by TCDD also dampens T cell function in a graft-vs-host model in a manner that depends on the AHR in CD4+ T cells (20). Importantly, in CD4CreAHRfx/fx mice, the AHR is also excised from other cell types that express CD4 during development, including CD8+ T cells and a small subset of dendritic cells (70, 71). Prior studies have shown that AHR activation with TCDD and ITE suppresses CD8+ T cell responses during IAV infection (15). As such, one explanation that Tfh cells are unaffected by TCDD in CD4CreAHRfx/fx mice is that CD8+ T cells are able to control infection. However, vehicle and TCDD treated mice have similar lung viral burdens over the course of IAV infection (15, 24, 25, 72); thus, it is less likely that CD8+ T cells have a role in AHR-driven alterations to Tfh cells. Yet, the observation that only the second phase of elevated BCL6 expression in CD4+ T cells was attenuated by AHR activation suggests that AHR signaling in other cell types may also contribute to modulation of Tfh cell responses to infection. For example, dendritic cells play a particularly important role in early T cell activation, and are also modulated by AHR activation (31, 37, 73, 74). As such, even though events in CD4+ T cells were clearly affected by the AHR, the overall mechanism by which the AHR regulates CD4+ T cell activation and their differentiation to Tfh cells may be multi-faceted and involve more than one immune cell type.
In addition to demonstrating that AHR in CD4+ T cells is necessary for in vivo modulation of Tfh cells by an exogenous AHR ligand, the current study strongly connects AHR in CD4+ T cells to regulation of humoral immune responses to viral infection more broadly. That is, key B cell responses facilitated by Tfh cells were no longer affected by AHR ligand when CD4+ T cells lacked the AHR. This finding suggests that AHR signaling in CD4+ T cells not only regulates CD4+ T cell differentiation, but also modulates T cell cues to B cells. Further support that AHR in CD4+ T cells provides signals to B cells comes from the overall lower level of IAV-specific antibodies when CD4+ T cells lacked the AHR. This suggests that even in the absence of an exogenously added agonist, endogenous AHR signaling regulates key CD4+ T cell functions that influence antibody production or isotype switching. Other studies have reported that AHR activation affects CD4+ T cell expression of numerous cytokines, such as IFNγ and IL-10, suggesting one possible means to influence antibody isotype switching (75–77). Thus, the AHR likely regulates multiple pathways in CD4+ T cells. These include those involved in differentiation to functionally distinct subsets, including Tfh cells, and others involved in promoting optimal antibody production and isotype switching.
Another important finding from these current studies was that AHR activation blunted Tfh cell differentiation and proliferation starting during initial stages of viral infection. Although the percentage of BrdU+ Tfh cells unaffected by AHR activation, the entire pool of proliferating Tfh cells was significantly smaller in mice given TCDD. When integrated with AHR-mediated changes to Tfh cells occurring in a CD4+ T cell intrinsic manner, these results indicate that the AHR influences multiple early events in CD4+ T cells, including activation, proliferation, and differentiation. This idea aligns well with and extends prior observations from a graft-vs-host model, which showed that AHR activation induced transcriptional and functional changes in CD4+ T cells as early as 2 days into the response (19). Other prior studies have shown that AHR activation modulates CD4+ T cell proliferation, primarily in the context of non-infectious antigenic challenges, such as ovalbumin and alloantigens (17, 53, 54). Given that AHR activation also reduced the proliferation of the entire CD4+ T cell population in IAV-infected mice, the AHR may regulate some pathways that are generalizable to CD4+ T cells, rather than specific to Tfh cells. This seems likely because during IAV infection, AHR activation also reduced the frequency of Th1 cells, while increasing the percentage of Tregs (15). Furthermore, in response to a range of immune challenges, AHR activation can elicit distinct effects on CD4+ T cell subsets, such that there are more or less of a particular subset (15, 22). For instance, in contrast to TCDD, treatment of mice with FICZ increased the frequency of Tfh and Th1 cells during IAV infection (15). TCDD and FICZ also had opposite effects on Th17 cells and Tregs in a mouse model experimental autoimmune encephalomyelitis (22). Thus, it is likely that the AHR modulates CD4+ T cell proliferation and differentiation, but the specific consequences are dependent upon the antigenic challenge, AHR ligand, and also potentially microenvironmental differences in organs and tissues.
Tfr cells are a subset of follicular CD4+ T cells that are unique among FOXP3+ Treg cells in that they gain access to germinal centers and can suppress Tfh and B cell functions (56, 78, 79). Similar to Tfh cells, the impact of AHR activation on Tfr cells is poorly characterized. Considering that AHR activation increases Tregs during IAV infection (15) and modulates Tregs in other model systems (17, 22, 75), we postulated that Tfr cells may also be affected. Our observations suggest a nuanced role of AHR in modulating regulatory T cell subsets. Similar to Tfh cells, the number of Tfr cells was significantly reduced by AHR activation during IAV infection, which is consistent with, and perhaps a consequence of, diminution of CD4+ T cell responses overall. However, the relative ratio of Tfr to Tfh cells was almost tripled in TCDD treated mice. Thus, within the pool of follicular CD4+ T cells, AHR activation affects the balance of helper vs regulatory cells. One mechanism by which the AHR may regulate the proportion of Tfr and Tfh cells is by altering expression of key signaling molecules, such as the transcription factors BCL6 and FOXP3. BCL6 expression in CD4+ T cells is required for Tfh cell differentiation (55), and both BCL6 and FOXP3 are required to generate Tfr cells (79, 80). During IAV infection, AHR activation concurrently reduced BCL6 and increased FOXP3 levels in CD4+ T cells. One way in which AHR could affect these transcriptional regulators is by regulating gene expression. Consistent with this idea, changes in FOXP3 and BCL6 levels in CD4+ T cells from infected mice treated with TCDD required the AHR to have a functional DBD. Other studies have shown that the AHR regulates FOXP3 expression in CD4+ T cells by directly binding AHREs in the Foxp3 promoter region (22, 81). In addition to IAV infection, treatment with TCDD or ITE reduced BCL6 levels in CD4+ T cells in other model systems (67, 82); however, the mechanism by which the AHR regulates BCL6 is not yet certain. The AHR may directly regulate BCL6 gene transcription, as genomic analyses show that the Bcl6 promoter region harbors AHREs (83). Importantly, another explanation for the changes in protein expression of BCL6 and FOXP3 in CD4+ T cells is that AHR activation is changing the proportion of BCL6 and FOXP3-expressing cells, which could in turn affect the percentage of cells expressing these markers, or the overall level of expression of these transcription factors detected. Nonetheless, one potential mechanism by which the AHR modulates the balance between Tfh and Tfr cells may be via divergent regulation of Bcl6 and Foxp3 expression.
Demonstrating that the AHR modulates the differentiation and expansion of Tfh cells provides deeper understanding of how AHR agonists modulate humoral immune defenses during acute primary respiratory viral infection. This is important because humans are regularly exposed to AHR ligands from a variety of anthropogenic and natural sources (10, 84, 85). Thus, exposure to AHR ligands is one source of variability in responses to infection that are observed even in populations exposed to the same or similar pathogen (3, 5, 86). Indeed, exposure to AHR ligands from the environment, such as polyaromatic hydrocarbons in air pollution and cigarette smoke, as well as dioxins and PCBs in foods and other substances, has been associated with increased incidence and severity of infection and reduced antibody responses (7–9). Thus, via binding the AHR, environmental factors may contribute to variable responses to infection, and also potentially to vaccination. Furthermore, the importance of the AHR as a modulator of Tfh cells extends beyond understanding how pollutants impact humoral immunity in two key ways. There are myriad AHR agonists, which are not pollutants, that may be useful for attenuating antibody-mediated diseases (10, 11, 87).
Indeed, there is evidence that engaging the AHR may not necessarily always result in suppression of a beneficial immune response. For instance, in some instances AHR activation, even with TCDD, leads to a favorable outcome (22, 88). Also, some AHR agonists, particularly ones that transiently activate the AHR, may be able to boost Tfh cell responses which could, in turn, bolster antibody responses (15, 89). For example, during IAV infection, mice given the short-lived AHR ligand FICZ had a greater frequency of Tfh cells compared to control mice (15). This may relate to differences in ligand metabolism; however, the molecular reasons for differential immunological effects of distinct AHR ligands is not completely understood (15, 90, 91). In addition to boosting the number of Tfh cells, another possible avenue to elevate Tfh cell responses is screening for AHR ligands that modulate Tfr cells (89). However, approaches to bolster Tfh cells need to be viewed with caution as excessive Tfh cells can promote spontaneous germinal center formation, autoantibody production, and impair affinity maturation (23, 92, 93). Hence, there are scenarios in which modulating Tfh cell responses are beneficial in one setting and deleterious in another, and the AHR provides a potential targetable means to modulate Tfh cell responses. In summary, AHR modulation of Tfh cells and humoral immunity expands our understanding of how environmental contaminants can impact human health and disease, and also provides new perspectives on how we can improve vaccine efforts to pathogenic viruses that impact humans, such as IAV and SARS-CoV-2.
Supplementary Material
Key Points.
AHR modulates Tfh cell responses during infection in a CD4+ T cell intrinsic manner
AHR alters Tfr:Tfh cell ratio and levels of BCL6 and FOXP3 in CD4+ T cells
AHR requires a functional DBD to modulate Tfh and Tfr cells during IAV infection
Acknowledgements
The authors are grateful to Dr. Timothy Bushnell and Mr. Matt Cochran at the URMC Flow Cytometry Core. We also thank Dr. Lisbeth Boule for contributions to this project, and Dr. Anthony Franchini, Catherine Donegan, and Colleen O’Dell for help maintaining the mouse colonies used in this research.
This work was supported by grants from the National Institutes of Health (R01ES030300, R01ES004862, P30ES01247, T32AI007285 and F31ES032301)
Abbreviations used in this article:
- MLN
mediastinal lymph node
- IAV
influenza A virus
- AHR
aryl hydrocarbon receptor
- Tfh
T follicular helper
- Treg
regulatory T cells
- Tfr
T follicular regulatory
- TCDD
2,3,7,8-tetrachlorodibenzo-p-dioxin
- GuHCL
guanidine hydrochloride
- GC
germinal center
- DBD
DNA-binding domain
- AHRE
aryl hydrocarbon response element
- i.n.
intranasally
- ITE
2-(1H-Indol-3-ylcarbonyl)-4-thiazolcarboxylic acid methyl ester
- KYNA
Kynurenic acid
Footnotes
The authors declare that they have no competing, personal or financial interests
References
- 1.GBD 2016 Lower Respiratory Infections Collaborators. 2018. Estimates of the global, regional, and national morbidity, mortality, and aetiologies of lower respiratory infections in 195 countries, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Infect Dis. 18: 1191–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Abdelrahman Z, Li M, and Wang X. 2020. Comparative Review of SARS-CoV-2, SARS-CoV, MERS-CoV, and Influenza A Respiratory Viruses. Front Immunol. 11: 2309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Franchini AM, and Lawrence BP. 2018. Environmental exposures are hidden modifiers of anti-viral immunity. Curr Opin Toxicol. 10: 54–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Prüss-Üstün A, Wolf J, Corvalán C, Bos R, and Neira M. 2016. Preventing disease through healthy environments : a global assessment of the burden of disease from environmental risks. World Health Organization, Geneva, Switzerland. Available from: https://www.who.int/publications/i/item/9789241565196 (9 September 2021, date last accessed). [Google Scholar]
- 5.Feingold BJ, Vegosen L, Davis M, Leibler J, Peterson A, and Silbergeld EK. 2010. A niche for infectious disease in environmental health: rethinking the toxicological paradigm. Environ Health Perspect. 118: 1165–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Newton AH, Cardani A, and Braciale TJ. 2016. The host immune response in respiratory virus infection: balancing virus clearance and immunopathology. Semin Immunopathol. 38: 471–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Heilmann C, Budtz-Jorgensen E, Nielsen F, Heinzow B, Weihe P, and Grandjean P. 2010. Serum concentrations of antibodies against vaccine toxoids in children exposed perinatally to immunotoxicants. Environ Health Perspect. 118: 1434–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Jusko TA, De Roos AJ, Lee SY, Thevenet-Morrison K, Schwartz SM, Verner MA, Murinova LP, Drobna B, Kocan A, Fabisikova A, Conka K, Trnovec T, Hertz-Picciotto I, and Lawrence BP. 2016. A Birth Cohort Study of Maternal and Infant Serum PCB-153 and DDE Concentrations and Responses to Infant Tuberculosis Vaccination. Environ Health Perspect. 124: 813–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Stolevik SB, Nygaard UC, Namork E, Haugen M, Kvalem HE, Meltzer HM, Alexander J, van Delft JH, Loveren H, Lovik M, and Granum B. 2011. Prenatal exposure to polychlorinated biphenyls and dioxins is associated with increased risk of wheeze and infections in infants. Food Chem Toxicol. 49: 1843–1848. [DOI] [PubMed] [Google Scholar]
- 10.Esser C, and Rannug A. 2015. The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol Rev. 67: 259–279. [DOI] [PubMed] [Google Scholar]
- 11.Rothhammer V, and Quintana FJ. 2019. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunology. 19: 184–197. [DOI] [PubMed] [Google Scholar]
- 12.Lawrence BP, and Vorderstrasse BA. 2013. New insights into the aryl hydrocarbon receptor as a modulator of host responses to infection. Semin Immunopathol. 35: 615–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sun B, and Zhang Y. 2014. Overview of orchestration of CD4+ T cell subsets in immune responses. Adv Exp Med Biol. 841: 1–13. [DOI] [PubMed] [Google Scholar]
- 14.Swain SL, McKinstry KK, and Strutt TM. 2012. Expanding roles for CD4(+) T cells in immunity to viruses. Nat Rev Immunol. 12: 136–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Boule LA, Burke CG, Jin GB, and Lawrence BP. 2018. Aryl hydrocarbon receptor signaling modulates antiviral immune responses: ligand metabolism rather than chemical source is the stronger predictor of outcome. Sci Rep. 8: 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Quintana FJ, Murugaiyan G, Farez MF, Mitsdoerffer M, Tukpah AM, Burns EJ, and Weiner HL. 2010. An endogenous aryl hydrocarbon receptor ligand acts on dendritic cells and T cells to suppress experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A. 107: 20768–20773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ehrlich AK, Pennington JM, Tilton S, Wang X, Marshall NB, Rohlman D, Funatake C, Punj S, O’Donnell E, Yu Z, Kolluri SK, and Kerkvliet NI. 2017. AhR activation increases IL-2 production by alloreactive CD4(+) T cells initiating the differentiation of mucosal-homing Tim3(+) Lag3(+) Tr1 cells. Eur J Immunol. 47: 1989–2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ehrlich AK, Pennington JM, Wang X, Rohlman D, Punj S, Lohr CV, Newman MT, Kolluri SK, and Kerkvliet NI. 2016. Activation of the Aryl Hydrocarbon Receptor by 10-Cl-BBQ Prevents Insulitis and Effector T Cell Development Independently of Foxp3+ Regulatory T Cells in Nonobese Diabetic Mice. J Immunol. 196: 264–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Funatake CJ, Marshall NB, Steppan LB, Mourich DV, and Kerkvliet NI. 2005. Cutting edge: activation of the aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo-p-dioxin generates a population of CD4+ CD25+ cells with characteristics of regulatory T cells. J Immunol. 175: 4184–4188. [DOI] [PubMed] [Google Scholar]
- 20.Kerkvliet NI, Shepherd DM, and Baecher-Steppan L. 2002. T lymphocytes are direct, aryl hydrocarbon receptor (AhR)-dependent targets of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD): AhR expression in both CD4+ and CD8+ T cells is necessary for full suppression of a cytotoxic T lymphocyte response by TCDD. Toxicol Appl Pharmacol. 185: 146–152. [DOI] [PubMed] [Google Scholar]
- 21.Kerkvliet NI, Steppan LB, Vorachek W, Oda S, Farrer D, Wong CP, Pham D, and Mourich DV. 2009. Activation of aryl hydrocarbon receptor by TCDD prevents diabetes in NOD mice and increases Foxp3+ T cells in pancreatic lymph nodes. Immunotherapy. 1: 539–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E, Caccamo M, Oukka M, and Weiner HL. 2008. Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature. 453: 65–71. [DOI] [PubMed] [Google Scholar]
- 23.Crotty S 2019. T Follicular Helper Cell Biology: A Decade of Discovery and Diseases. Immunity. 50: 1132–1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lawrence BP, and Vorderstrasse BA. 2004. Activation of the aryl hydrocarbon receptor diminishes the memory response to homotypic influenza virus infection but does not impair host resistance. Toxicol Sci. 79: 304–314. [DOI] [PubMed] [Google Scholar]
- 25.Warren TK, Mitchell KA, and Lawrence BP. 2000. Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) suppresses the humoral and cell-mediated immune responses to influenza A virus without affecting cytolytic activity in the lung. Toxicol Sci. 56: 114–123. [DOI] [PubMed] [Google Scholar]
- 26.Walisser JA, Glover E, Pande K, Liss AL, and Bradfield CA. 2005. Aryl hydrocarbon receptor-dependent liver development and hepatotoxicity are mediated by different cell types. Proc Natl Acad Sci U S A. 102: 17858–17863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bunger MK, Glover E, Moran SM, Walisser JA, Lahvis GP, Hsu EL, and Bradfield CA. 2008. Abnormal liver development and resistance to 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicity in mice carrying a mutation in the DNA-binding domain of the aryl hydrocarbon receptor. Toxicol Sci. 106: 83–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lee PP, Fitzpatrick DR, Beard C, Jessup HK, Lehar S, Makar KW, Perez-Melgosa M, Sweetser MT, Schlissel MS, Nguyen S, Cherry SR, Tsai JH, Tucker SM, Weaver WM, Kelso A, Jaenisch R, and Wilson CB. 2001. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity. 15: 763–774. [DOI] [PubMed] [Google Scholar]
- 29.Westendorf K, Durek P, Ayew S, Mashreghi MF, and Radbruch A. 2016. Chromosomal localisation of the CD4cre transgene in B6.Cg-Tg(Cd4-cre)1Cwi mice. J Immunol Methods. 436: 54–57. [DOI] [PubMed] [Google Scholar]
- 30.Poland A, Palen D, and Glover E. 1994. Analysis of the four alleles of the murine aryl hydrocarbon receptor. Mol Pharmacol. 46: 915–921. [PubMed] [Google Scholar]
- 31.Jin GB, Moore AJ, Head JL, Neumiller JJ, and Lawrence BP. 2010. Aryl hydrocarbon receptor activation reduces dendritic cell function during influenza virus infection. Toxicol Sci. 116: 514–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Malaczewska J, Siwicki AK, Wojcik RM, Kaczorek E, and Turski WA. 2014. Effect of oral administration of kynurenic acid on the activity of the peripheral blood leukocytes in mice. Cent Eur J Immunol. 39: 6–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wu T, Shin HM, Moseman EA, Ji Y, Huang B, Harly C, Sen JM, Berg LJ, Gattinoni L, McGavern DB, and Schwartzberg PL. 2015. TCF1 Is Required for the T Follicular Helper Cell Response to Viral Infection. Cell Rep. 12: 2099–2110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wang P, Wang Y, Xie L, Xiao M, Wu J, Xu L, Bai Q, Hao Y, Huang Q, Chen X, He R, Li B, Yang S, Chen Y, Wu Y, and Ye L. 2019. The Transcription Factor T-Bet Is Required for Optimal Type I Follicular Helper T Cell Maintenance During Acute Viral Infection. Front Immunol. 10: 606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Barrett T, Inglis SC. 1985. Growth purification and titration influenza viruses. Virology: A Practical Approach. 119: 119–150. [Google Scholar]
- 36.Boule LA, Winans B, Lambert K, Vorderstrasse BA, Topham DJ, Pavelka MS, and Lawrence BP. 2015. Activation of the aryl hydrocarbon receptor during development enhances the pulmonary CD4+ T-cell response to viral infection. Am J Physiol Lung Cell Mol Physiol. 309: L305–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Jin GB, Winans B, Martin KC, and Paige Lawrence B. 2014. New insights into the role of the aryl hydrocarbon receptor in the function of CD11c⁺ cells during respiratory viral infection. Eur J Immunol. 44: 1685–1698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Knowlden ZA, and Sant AJ. 2016. CD4 T cell epitope specificity determines follicular versus non-follicular helper differentiation in the polyclonal response to influenza infection or vaccination. Sci Rep. 6: 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yu D, and Vinuesa CG. 2010. The elusive identity of T follicular helper cells. Trends Immunol. 31: 377–383. [DOI] [PubMed] [Google Scholar]
- 40.Linterman MA, Pierson W, Lee SK, Kallies A, Kawamoto S, Rayner TF, Srivastava M, Divekar DP, Beaton L, Hogan JJ, Fagarasan S, Liston A, Smith KG, and Vinuesa CG. 2011. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. 17: 975–982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Schrock DC, Leddon SA, Hughson A, Miller J, Lacy-Hulbert A, and Fowell DJ. 2019. Pivotal role for alphaV integrins in sustained Tfh support of the germinal center response for long-lived plasma cell generation. Proc Natl Acad Sci U S A. 116: 4462–4470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Boule LA, Winans B, and Lawrence BP. 2014. Effects of developmental activation of the AhR on CD4+ T-cell responses to influenza virus infection in adult mice. Environ Health Perspect. 122: 1201–1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dauner JG, Pan Y, Hildesheim A, Kemp TJ, Porras C, and Pinto LA. 2012. Development and application of a GuHCl-modified ELISA to measure the avidity of anti-HPV L1 VLP antibodies in vaccinated individuals. Mol Cell Probes. 26: 73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Denison MS, and Nagy SR. 2003. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol. 43: 309–334. [DOI] [PubMed] [Google Scholar]
- 45.Song J, Clagett-Dame M, Peterson RE, Hahn ME, Westler WM, Sicinski RR, and DeLuca HF. 2002. A ligand for the aryl hydrocarbon receptor isolated from lung. Proc Natl Acad Sci U S A. 99: 14694–14699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gasiewicz TA, Geiger LE, Rucci G, and Neal RA. 1983. Distribution, excretion, and metabolism of 2,3,7,8-tetrachlorodibenzo-p-dioxin in C57BL/6J, DBA/2J, and B6D2F1/J mice. Drug Metab Dispos. 11: 397–403. [PubMed] [Google Scholar]
- 47.Aylward LL, Brunet RC, Carrier G, Hays SM, Cushing CA, Needham LL, Patterson DG Jr., Gerthoux PM, Brambilla P, and Mocarelli P. 2005. Concentration-dependent TCDD elimination kinetics in humans: toxicokinetic modeling for moderately to highly exposed adults from Seveso, Italy, and Vienna, Austria, and impact on dose estimates for the NIOSH cohort. J Expo Anal Environ Epidemiol. 15: 51–65. [DOI] [PubMed] [Google Scholar]
- 48.Kerger BD, Leung HW, Scott P, Paustenbach DJ, Needham LL, Patterson DG Jr., Gerthoux PM, and Mocarelli P. 2006. Age- and concentration-dependent elimination half-life of 2,3,7,8-tetrachlorodibenzo-p-dioxin in Seveso children. Environ Health Perspect. 114: 1596–1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Poland A, and Knutson JC. 1982. 2,3,7,8-tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: examination of the mechanism of toxicity. Annu Rev Pharmacol Toxicol. 22: 517–554. [DOI] [PubMed] [Google Scholar]
- 50.Fernandez-Salguero PM, Hilbert DM, Rudikoff S, Ward JM, and Gonzalez FJ. 1996. Aryl-hydrocarbon receptor-deficient mice are resistant to 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced toxicity. Toxicol Appl Pharmacol. 140: 173–179. [DOI] [PubMed] [Google Scholar]
- 51.Hochstenbach K, van Leeuwen DM, Gmuender H, Stolevik SB, Nygaard UC, Lovik M, Granum B, Namork E, van Delft JH, and van Loveren H. 2010. Transcriptomic profile indicative of immunotoxic exposure: in vitro studies in peripheral blood mononuclear cells. Toxicol Sci. 118: 19–30. [DOI] [PubMed] [Google Scholar]
- 52.Baccarelli A, Mocarelli P, Patterson DG Jr., Bonzini M, Pesatori AC, Caporaso N, and Landi MT. 2002. Immunologic effects of dioxin: new results from Seveso and comparison with other studies. Environ Health Perspect. 110: 1169–1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Funatake CJ, Dearstyne EA, Steppan LB, Shepherd DM, Spanjaard ES, Marshak-Rothstein A, and Kerkvliet NI. 2004. Early consequences of 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure on the activation and survival of antigen-specific T cells. Toxicol Sci. 82: 129–142. [DOI] [PubMed] [Google Scholar]
- 54.Shepherd DM, Dearstyne EA, and Kerkvliet NI. 2000. The effects of TCDD on the activation of ovalbumin (OVA)-specific DO11.10 transgenic CD4(+) T cells in adoptively transferred mice. Toxicol Sci. 56: 340–350. [DOI] [PubMed] [Google Scholar]
- 55.Hollister K, Kusam S, Wu H, Clegg N, Mondal A, Sawant DV, and Dent AL. 2013. Insights into the role of Bcl6 in follicular Th cells using a new conditional mutant mouse model. J Immunol. 191: 3705–3711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Fonseca VR, Ribeiro F, and Graca L. 2019. T follicular regulatory (Tfr) cells: Dissecting the complexity of Tfr-cell compartments. Immunol Rev. 288: 112–127. [DOI] [PubMed] [Google Scholar]
- 57.Larigot L, Juricek L, Dairou J, and Coumoul X. 2018. AhR signaling pathways and regulatory functions. Biochim Open. 7: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Vogel CF, Khan EM, Leung PS, Gershwin ME, Chang WL, Wu D, Haarmann-Stemmann T, Hoffmann A, and Denison MS. 2014. Cross-talk between aryl hydrocarbon receptor and the inflammatory response: a role for nuclear factor-κB. J Biol Chem. 289: 1866–1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Dere E, Lo R, Celius T, Matthews J, and Zacharewski TR. 2011. Integration of genome-wide computation DRE search, AhR ChIP-chip and gene expression analyses of TCDD-elicited responses in the mouse liver. BMC Genomics. 12: 365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Veldhoen M, Hirota K, Westendorf AM, Buer J, Dumoutier L, Renauld JC, and Stockinger B. 2008. The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature. 453: 106–109. [DOI] [PubMed] [Google Scholar]
- 61.Villa M, Gialitakis M, Tolaini M, Ahlfors H, Henderson CJ, Wolf CR, Brink R, and Stockinger B. 2017. Aryl hydrocarbon receptor is required for optimal B-cell proliferation. EMBO J 36: 116–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sherr DH, and Monti S. 2013. The role of the aryl hydrocarbon receptor in normal and malignant B cell development. Semin Immunopathol. 35: 705–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Frericks M, Temchura VV, Majora M, Stutte S, and Esser C. 2006. Transcriptional signatures of immune cells in aryl hydrocarbon receptor (AHR)-proficient and AHR-deficient mice. Biol Chem. 387: 1219–1226. [DOI] [PubMed] [Google Scholar]
- 64.Vacchio MS, Ciucci T, Gao Y, Watanabe M, Balmaceno-Criss M, McGinty MT, Huang A, Xiao Q, McConkey C, Zhao Y, Shetty J, Tran B, Pepper M, Vahedi G, Jenkins MK, McGavern DB, and Bosselut R. 2019. A Thpok-Directed Transcriptional Circuitry Promotes Bcl6 and Maf Expression to Orchestrate T Follicular Helper Differentiation. Immunity. 51: 465–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gutiérrez-Vázquez C, and Quintana FJ. 2018. Regulation of the Immune Response by the Aryl Hydrocarbon Receptor. Immunity. 48: 19–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Burke CG, Myers JR, Boule LA, Post CM, Brookes PS, and Lawrence BP. 2019. Early life exposures shape the CD4(+) T cell transcriptome, influencing proliferation, differentiation, and mitochondrial dynamics later in life. Sci Rep. 9: 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Goettel JA, Gandhi R, Kenison JE, Yeste A, Murugaiyan G, Sambanthamoorthy S, Griffith AE, Patel B, Shouval DS, Weiner HL, Snapper SB, and Quintana FJ. 2016. AHR Activation Is Protective against Colitis Driven by T Cells in Humanized Mice. Cell Rep. 17: 1318–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nakahama T, Kimura A, Nguyen NT, Chinen I, Hanieh H, Nohara K, Fujii-Kuriyama Y, and Kishimoto T. 2011. Aryl hydrocarbon receptor deficiency in T cells suppresses the development of collagen-induced arthritis. Proc Natl Acad Sci U S A. 108: 14222–14227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ye J, Qiu J, Bostick JW, Ueda A, Schjerven H, Li S, Jobin C, Chen ZE, and Zhou L. 2017. The Aryl Hydrocarbon Receptor Preferentially Marks and Promotes Gut Regulatory T Cells. Cell Rep. 21: 2277–2290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Henri S, Vremec D, Kamath A, Waithman J, Williams S, Benoist C, Burnham K, Saeland S, Handman E, and Shortman K. 2001. The dendritic cell populations of mouse lymph nodes. J Immunol. 167: 741–748. [DOI] [PubMed] [Google Scholar]
- 71.Ellmeier W, Haust L, and Tschismarov R. 2013. Transcriptional control of CD4 and CD8 coreceptor expression during T cell development. Cell Mol Life Sci. 70: 4537–4553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lawrence BP, Warren TK, and Luong H. 2000. Fewer T lymphocytes and decreased pulmonary influenza virus burden in mice exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). J Toxicol Environ Health A. 61: 39–53. [DOI] [PubMed] [Google Scholar]
- 73.Krishnaswamy JK, Alsen S, Yrlid U, Eisenbarth SC, and Williams A. 2018. Determination of T Follicular Helper Cell Fate by Dendritic Cells. Front Immunol. 9: 2169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Franchini AM, Myers JR, Jin GB, Shepherd DM, and Lawrence BP. 2019. Genome-Wide Transcriptional Analysis Reveals Novel AhR Targets That Regulate Dendritic Cell Function during Influenza A Virus Infection. Immunohorizons. 3: 219–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ambrosio LF, Insfran C, Volpini X, Acosta Rodriguez E, Serra HM, Quintana FJ, Cervi L, and Motran CC. 2019. Role of Aryl Hydrocarbon Receptor (AhR) in the Regulation of Immunity and Immunopathology During Trypanosoma cruzi Infection. Front Immunol. 10: 631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.McAleer JP, Fan J, Roar B, Primerano DA, and Denvir J. 2018. Cytokine Regulation in Human CD4 T Cells by the Aryl Hydrocarbon Receptor and Gq-Coupled Receptors. Sci Rep. 8: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Rohlman D, Pham D, Yu Z, Steppan LB, and Kerkvliet NI. 2012. Aryl Hydrocarbon Receptor-Mediated Perturbations in Gene Expression during Early Stages of CD4(+) T-cell Differentiation. Front Immunol. 3: 223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Stebegg M, Kumar SD, Silva-Cayetano A, Fonseca VR, Linterman MA, and Graca L. 2018. Regulation of the Germinal Center Response. Front Immunol. 9: 2469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Maceiras AR, Almeida SCP, Mariotti-Ferrandiz E, Chaara W, Jebbawi F, Six A, Hori S, Klatzmann D, Faro J, and Graca L. 2017. T follicular helper and T follicular regulatory cells have different TCR specificity. Nat Commun. 8: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chung Y, Tanaka S, Chu F, Nurieva RI, Martinez GJ, Rawal S, Wang YH, Lim H, Reynolds JM, Zhou XH, Fan HM, Liu ZM, Neelapu SS, and Dong C. 2011. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat Med. 17: 983–988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Singh NP, Singh UP, Singh B, Price RL, Nagarkatti M, and Nagarkatti PS. 2011. Activation of aryl hydrocarbon receptor (AhR) leads to reciprocal epigenetic regulation of FoxP3 and IL-17 expression and amelioration of experimental colitis. PLoS One. 6: e23522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Marshall NB, Vorachek WR, Steppan LB, Mourich DV, and Kerkvliet NI. 2008. Functional characterization and gene expression analysis of CD4+ CD25+ regulatory T cells generated in mice treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. J Immunol. 181: 2382–2391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sun YV, Boverhof DR, Burgoon LD, Fielden MR, and Zacharewski TR. 2004. Comparative analysis of dioxin response elements in human, mouse and rat genomic sequences. Nucleic Acids Re.s 32: 4512–4523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Esser C, Lawrence BP, Sherr DH, Perdew GH, Puga A, Barouki R, and Coumoul X. 2018. Old Receptor, New Tricks-The Ever-Expanding Universe of Aryl Hydrocarbon Receptor Functions. Report from the 4th AHR Meeting, 29(−)31 August 2018 in Paris, France. Int J Mol Sci. 19: 3603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Denison MS, Soshilov AA, He G, DeGroot DE, and Zhao B. 2011. Exactly the same but different: promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol Sci. 124: 1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Samadizadeh S, Masoudi M, Rastegar M, Salimi V, Shahbaz MB, and Tahamtan A. 2021. COVID-19: Why does disease severity vary among individuals? Respir Med. 180: 106356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Busbee PB, Rouse M, Nagarkatti M, and Nagarkatti PS. 2013. Use of natural AhR ligands as potential therapeutic modalities against inflammatory disorders. Nutr Rev. 71: 353–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Vorderstrasse BA, and Lawrence BP. 2006. Protection against lethal challenge with Streptococcus pneumoniae is conferred by aryl hydrocarbon receptor activation but is not associated with an enhanced inflammatory response. Infect Immun. 74: 5679–5686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Linterman MA, and Hill DL. 2016. Can follicular helper T cells be targeted to improve vaccine efficacy? F1000Res. 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ehrlich AK, Pennington JM, Bisson WH, Kolluri SK, and Kerkvliet NI. 2018. TCDD, FICZ, and Other High Affinity AhR Ligands Dose-Dependently Determine the Fate of CD4+ T Cell Differentiation. Toxicol Sci. 161: 310–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wheeler JL, Martin KC, Resseguie E, and Lawrence BP. 2014. Differential consequences of two distinct AhR ligands on innate and adaptive immune responses to influenza A virus. Toxicol Sci. 137: 324–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Craft JE 2012. Follicular helper T cells in immunity and systemic autoimmunity. Nat Rev Rheumatol. 8: 337–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Preite S, Baumjohann D, Foglierini M, Basso C, Ronchi F, Fernandez Rodriguez BM, Corti D, Lanzavecchia A, and Sallusto F. 2015. Somatic mutations and affinity maturation are impaired by excessive numbers of T follicular helper cells and restored by Treg cells or memory T cells. Eur J Immunol. 45: 3010–3021. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
