Open access peer-reviewed chapter

The Role of the Aryl Hydrocarbon Receptor (AhR) in the Immune Response against Microbial Infections

Written By

Lixing Huang, Rongchao He, Youyu Zhang and Qingpi Yan

Submitted: 07 December 2020 Reviewed: 09 February 2021 Published: 01 March 2021

DOI: 10.5772/intechopen.96526

From the Edited Volume

Antimicrobial Immune Response

Edited by Maria del Mar Ortega-Villaizan and Veronica Chico

Chapter metrics overview

571 Chapter Downloads

View Full Metrics

Abstract

Aryl hydrocarbon receptor (AhR), an important nuclear receptor, regulates the cellular response to environmental stressors. It is well known for its critical functions in toxicology, but is currently considered an essential regulator of diseases, with specific modulatory effects on immune, antimicrobial and inflammatory responses. The present chapter discusses AhR’s function and mechanism in the immune response against microbial infections.

Keywords

  • aryl hydrocarbon receptor (AhR)
  • functional mechanism
  • antimicrobial
  • immunity
  • gut immunity

1. Introduction

The ligand-activated transcription factor aryl hydrocarbon receptor (AhR) is structurally similar to other members of Pern-Arnt-Sim (PAS) superfamily [1, 2], which consists of a conserved signaling network that regulates signal exchange between host and environment [3, 4]. It was originally found to play a role in regulating the reactions of exogenous chemicals such as 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD). However, AhR has been recently recognized as an essential regulator of host-pathogen interactions [5, 6, 7, 8, 9], especially affecting immunity, inflammatory response and antibacterial activity [5, 9, 10, 11, 12, 13, 14, 15]. The current chapter focuses on AhR’s function in regulating immunity, inflammatory response and antibacterial activity.

Advertisement

2. Mechanism of AhR action

As a highly conserved nuclear receptor [10], AhR can regulate gene expression after binding to a ligand. AhR binds to its co-chaperones and maintains cytoplasmic localization [16, 17]. Ligand binding by AhR results in its release by co-chaperones and translocation into the nucleus, where it forms a heterodimer with the aryl hydrocarbon receptor nuclear translocator (ARNT) [18, 19]. Via binding to the genomic DNA—usually interacting with AhR response elements (AhREs, 5’-GCGTG-3′) [20, 21], also referred to as dioxin (DREs) or xenobiotic (XREs) response elements [9, 10], the AhR-ARNT heterodimer regulates multiple target genes such as Cytochrome P450 Family 1 Subfamily A Member 1 (CYP1A1), CYP1A2, CYP1B1, TCDD Inducible Poly (ADP-Ribose) Polymerase (TIP ARP), and aryl hydrocarbon receptor repressor (AhRR), which can inhibit AhR via a negative feedback circuit [22]. Target gene regulation is considered to be ligand dependent [21].

As a highly heterogeneous nuclear receptor, AhR binds to many ligands, including exogenous synthetic aromatic hydrocarbons [10, 23], exogenous natural chemicals [5, 6, 10, 14, 24] and endogenous ligands [25, 26, 27, 28, 29]. Tryptophan, an essential amino acid in humans, constitutes the precursor of many important components in the human body. Interestingly, the tryptophan (TRP) pathway has a critical function in immune and inflammatory responses through providing many ligands for AhR. In addition, AhR controls the expression and activation of tryptophan 2,3-dioxygenase (TDO2), indoleamine 2,3-dioxygenase (IDO), kynureninase (KYNU) and kynurenine 3-monooxygenase (KMO). The aforementioned enzymes catalyze the synthesis of kynurenine (KYN), which is a product of TRP metabolism, thus enabling feedback inhibition because KYN and AhR are agonists [30, 31].

Advertisement

3. AhR expression modulation

The interactions of AhR and its ligands, including polycyclic aromatic hydrocarbons (PAHs), can be used as a cytoplasmic signal sensor. The conformation of AhR changes, and it is transferred from the cytoplasm to the nucleus. The high-affinity ligand TCDD can exert toxic effects by binding with and activating AhR [32, 33]. Structural analysis of AhR revealed three domains: 1) The amino-terminal DNA binding domain (DBD) comprises the basic helix–loop–helix (bHLH) region and the nuclear localization signal (NLS); 2) The central PAS region encompasses two degenerate repeats; 3) The carboxy-terminal region features the transactivation domain (TAD) [34]. In addition, phylogenetic data showed that AhR constitutes an ancient protein whose functional orthologues are found in reptiles, amphibians, birds and mammals. However, there are many structural differences between human and murine AhR genes. Sequence analysis revealed approximately 85% structural similarity in the amino-terminal sequence, while the C-terminal region shows a low homology. The TAD or N-terminal domain is the least conservative [34]. The C-terminal domain is a highly unstructured sequence containing a transcriptionally active region and contributes to receptor transformation [35, 36].

AhR, heat shock protein 90 and X-associated protein 2 form multiple protein complexes in the cytoplasm. In the presence of ligands or agonists, AhR complexes undergo nuclear translocation and form heterodimers with ARNT. With a core sequence of 5′-GCGTG-3′, the AhR/ARNT complex interacts with DREs in the proximal site of promoters of target genes. Both AhR and ARNT recruit additional transcription co-activators for gene regulation, e.g., CYP and AhRR. Once transferred into the nucleus, AhR undergoes proteasome-induced degradation [37]. AhR function is modulated and weakened by AhRR, another member of the PAS family. After AhR activation, the level of AhRR increases rapidly [38]. Meanwhile, AhRR has a transcriptional repressor domain and can dimerize with ARNT even without an agonist, to fulfill its function [39].

Advertisement

4. AhR response to bacterial pathogens

It is known that AhR has a critical function in controlling responses to a variety of microbial pathogens. For example, it is required to effectively clear the Gram-positive pathogenic bacteria Listeria monocytogenes (LM). In mice, AhR inhibits LM by inducing ROS production via upregulation of the anti-inflammatory cytokine IL-10 and macrophage apoptosis inhibitor, resulting in suppressed macrophage apoptosis, reduced amounts of pro-inflammatory cytokines (e.g., Interleukin 6 (IL-6) and Tumor Necrosis Factor alpha (TNF-α)), and decreased the nuclear factor kappaB (NF-κB) activation. In addition, AhR ligands can enhance the response of AhR WT mice to LM, but not of AhR−/−mice [27].

When inoculated with log-phase LM intravenously, AhR deficient C57BL/6 J mice (AhR−/−) showed higher susceptibility compared with AhR heterozygous (AhR+/−) littermates. In comparison with AhR+/− animals, AhR−/− counterparts showed more colony forming units (CFUs) of LM in the spleen and liver, and more pronounced alterations in liver histopathology. Serum monocyte chemotactic protein 1 (MCP-1), IL-6, TNF-α and Interferon γ (IFN-γ) amounts were similar in AhR−/−and AhR+/−mice infected with LM. Elevated IL-12 and IL-10 amounts were detected in AhR−/−mice infected with LM. In terms of capacity of uptake and inhibition of intracellular growth of LM, AhR+/−and AhR−/−macrophages were comparable in vitro. In addition, T cell-dependent response was similar in AhR−/−and AhR+/−mice, as determined by intracellularly labelling cluster of differentiation 4 and 8 (CD4+ and CD8+) splenocytes for IFN-γ and TNF-α. AhR−/−and AhR+/−mice with prior infection showed increased resistance to re-infection by LM. The above evidence suggests that AhR is necessary to build an effective resistance, but not required for adaptive immune reactions following LM infection [40].

Streptococcus pneumonia, a common respiratory pathogen, represents a major cause of morbidity and death in humans, especially the elderly and children. The immune response after S. pneumoniae infection begins quickly in the lung, and the innate immune response can contain bacterial colonization in the ideal situation. Death, and bacterial load, cytokine/chemokine amounts, and immune cell infiltration in the lung have been assessed at different times in TCDD treated mice after S. pneumoniae infection. The survival rate of mice administered TCDD was significantly increased, while bacterial load in the lung was reduced. However, intriguingly, no evidence suggested that the protective effect was caused by increased inflammatory response. In fact, neutrophil amounts and inflammatory chemokine/cytokine levels in TCDD treated mice were lower than those of control animals. These findings suggest that AhR induction does not protect the animals by immune modulation, but likely by directly affecting lung cells upon infection [41].

Pseudomonas plecoglossicida represents the bacterial pathogen of fish visceral white spot disease with temperature dependent virulence [42]. AhR is also required for resistance to P. plecoglossicida. It was shown that ahr1a, ahr1b, ahr2 and cyp1a amounts in various organs of Danio rerio and Epinephelus coioides infected with P. plecoglossicida have similar trends. It should be noted that the intestine, liver, heart and spleen are the most affected organs, while ahr2 specifically shows a sharp increase in the spleen. After P. plecoglossicida infection, ahr1a amounts in macrophages are markedly reduced, while ahr1b, ahr2 and cyp1a are overtly upregulated. The cell viability and immune escape rates of P. plecoglossicida were significantly increased in macrophages with ahr1b and ahr2 knockdown. In conclusion, ahr1a, ahr1b, ahr2 and cyp1a are involved in immune reactions to P. plecoglossicida in various fish organs, while ahr1b and ahr2 might play a key role in splenic and macrophage immune reactions [43].

Huang et al. described the first pathogenic Aeromonas salmonicida (SRW-OG1) obtained from the warm water fish E. coioides, and studied AhR’s role in the immune response to SRW-OG1 infection [44]. They found that AhR is induced by unknown ligands in the intestine, spleen and macrophages. At the same time, ahr1a and ahr1b amounts were markedly elevated in the intestine, spleen and macrophages, while ahr2 only showed an increase in the intestine, suggesting ahr2 may contribute less to immune reactions compared with ahr1a and ahr1b. In SRW-OG1 infected E. coioides, major genes contributing to bacterial recognition, macrophage inflammatory response and gut immunity were overtly upregulated. However, decreased ROS amounts and the downregulation of other associated genes were equally detected, which indicated that SRW-OG1 could prevent ROS production by macrophages through its virulence mechanism. In addition, repression of AhR with an inhibitor or by gene silencing rescued the increases of IL-1 β and IL-8 associated with SRW-OG1 infection, clearly demonstrating that induction of E. coioides macrophages by IL-1 β and IL-8 is controlled by AhR. Nevertheless, AhR exerted no effects on bactericidal permeability-increasing protein/lipopolysaccharide-binding protein (BPI/LBP), reactive oxygen species (ROS) biosynthesis and associated genes. Compared with wild-type macrophages, survival and immune escape rates after SRW-OG1 infection were significantly increased in ahr1a/ahr1b-knockdown and 3′, 4’-DMF treated macrophages. Taken together, ahr1a and ahr1b are necessary for the immune response to SRW-OG1 [44].

Lipopolysaccharide (LPS) stimulation is often utilized to model Gram-negative bacteria-induced sepsis for assessing AhR’s functions in infection resistance and septic shock regulation. AhR and TDO2 are required for survival after the initial exposure to LPS [14, 20], while subsequent exposures are dependent on AhR and IDO1/2. LPS up-regulates TDO2 and IDO1/2, the rate-limiting enzymes of TRP transformation into KYN, and further induces AhR, thus downregulating pro-inflammatory cytokines and regulating long-term systemic inflammation [20]. In addition, compared with AhR wild type mice or immune cells, LPS challenged AhR−/−mice or immune cells produce higher concentrations of pro-inflammatory cytokines, including IL-1 β, IL-6, IL-18, IL-12, TNF-α and IFN-γ, as well as NLR Family Pyrin Domain Containing 3 (NLRP3) that regulates multiple pro-inflammatory cytokines. The AhR agonists 3-methylcholine (3-Mc), 6-Formylindolo[3,2-b]carbazole (FICZ), KYN and TCDD could protect AhR WT mice, but conferred no protection to AhR−/− animals, from extremely high amounts of pro-inflammatory cytokines and septic shock [45]. Thus, the immune response to bacterial pathogens requires AhR, and the underlying mechanisms are vital in identifying novel therapeutic agents to combat bacterial pathogens.

Advertisement

5. AhR response to viral pathogens

AhR is also associated with response to viral pathogens. For example, herpes simplex virus (HSV)-associated eye infection can lead to chronic immune-inflammatory response, causing blindness. However, in a mouse model, a single dose of TCDD could alleviate herpetic keratitis lesions, reduce viral load and decrease pro-inflammatory cytokine levels. However, similar effects were not obtained with FICZ, thus indicating a difference between both AhR ligands [46]. Therefore, response to viral pathogens requires AhR, and nontoxic AhR agonists could be used in the treatment of HSV-induced eye infections.

In influenza virus infection, activation of AhR doubles the number of neutrophils in the airway and interstitium of the lung, which reduces the survival rate from an otherwise sub-lethal infection [47, 48]. Interestingly, no increase in neutrophil inflammation or decreased survival was observed in AhR deficient mice treated with TCDD and influenza virus [37]. Innate immune reactions, including excessive pulmonary neutrophilia, can lead to severer pathological conditions and poor clinical outcomes after influenza virus infection [49, 50, 51]. Meanwhile, epidemiological reports have shown that exposure to environmental AhR ligands is associated with elevated respiratory tract infection, pulmonary congestion and exacerbation of inflammatory lung disease [52, 53, 54]. Therefore, there is parallel evidence in rodent animal models and humans that AhR regulates neutrophil inflow during infection. Overall, these data suggest that AhR regulates a new pathway to regulate neutrophil migration during influenza virus infection. A possible new target gene of AhR is inducible nitric oxide synthase (iNOS). Meanwhile, activation of AhR can increase the expression of iNOS in the mouse lung upon infection with influenza virus [55].

Advertisement

6. AhR response to parasitic pathogens

The immune response to parasites also requires AhR. For example, immune response to Toxoplasma gondii, a pathogenic parasite causing toxoplasmosis, requires increased AhR-dependent production of IL-10. Indeed, AhR−/− mice have reduced response to T. gondii and a less pronounced IL-10 increase [56].

After intraperitoneal infection with T. gondii, the death rate of AhR−/− mice was significantly higher than that of WT mice. Moreover, AhR−/− mice showed greater liver injury, and higher levels of NO, IgE and TNF-α, but lower IL-10 secretion in the serum. Interestingly, fewer cysts were found in the brain. The increased mortality was related to reduced IL-10, 5-LOX and GATA-3 expression levels, but increased IFN-γ expression in the spleen. In addition, AhR−/− mice had increased IL-12 and IFN-γ amounts, but decreased TLR2 levels compared with wild-type mice in peritoneal exudate cells. These findings suggest that AhR is vital for limiting inflammation during toxoplasmosis [57].

Therefore, AhR is necessary for parasitic pathogen response. This provides information on a response pathway and can be used to design new treatments.

Advertisement

7. AhR and the intestinal microbiota

AhR is found at high levels in the epithelial barrier [58], and the intestinal barrier of AhR−/−mice is inadequate, suggesting AhR might be important in maintaining or generating a healthy intestinal barrier [19]. In addition, low levels of AhR and AhR’s target genes are found in sterile mice [9], and AhR is needed for maintaining the RORγt+ innate lymphoblastoid cell (ILC) balance in the intestine [18]. In addition, the TRP metabolizing indole biosynthesized by select bacterial components of the intestinal microbiota is an AhR ligand [59, 60]. Diet without indole or antibiotic treatment can lead to the differentiation of mononuclear phagocytes, dependent on AhR, into dendritic cells (DCs) [48], which are more susceptible to gut pathogens in mice [17]. Overall, the above findings suggest AhR might be important in host gut-microbiota interactions.

AhR also plays a role in the reciprocal relationship among intestinal bacteria, bacterial metabolites and the intestinal immune system. AhR-deficient RORγt+ ILCs (the main producers of gut IL-22) with lower IL-22 amounts make mice easily die upon Citrobacter rodentium infection. It was pointed out that treatment with FICZ markedly enhances RORγt+ ILC accumulation in AhR+/− and AhR+/+ mice, but not in AhR−/− animals [61]. Lactobacillus species (nonpathogenic intestinal bacteria) are capable of producing AhR ligands, including indole-3-aldehydes, from tryptophan in the gut, thus enhancing the production of AhR dependent IL-22 [62]. Indole-3-aldehydes induces AhR-associated transcription, but exclusively at elevated concentrations, indicating its low affinity. However, indole-3-acetaldehyde (a product of indole-3-aldehydes) produces the high-affinity ligand FICZ [63], which may be related to the effect reported by Zelante et al. IL-22 affects epithelial cells and causes them to produce antimicrobial peptides, such as type III Reg (regenerating gene product) gamma (RegIIIg), and to stimulate tissue regeneration. Meanwhile, symbiotic bacteria may outperform bacterial pathogens and inhibit Candida albicans colonization [51]. Similar to keratinocyte and skin immune cell levels, AhR amounts are high in IECs and intestinal immune system cells [64].

In AhR-null mice, the number of intraepithelial lymphocytes (IELs) in the small intestine is significantly reduced [6, 64, 65], which is related to lower IL-22 amounts, and therefore to downregulated ileal antimicrobial peptides, including RegIIIb and RegIIIg. The microbial loads of the small and large intestines are also elevated. Loss of IELs is cell-intrinsic since AhR-deficient bone marrow cells do not reconstruct the gut in Rag−/− mice [51]. Over time after birth, intestinal Group 3 Innate Lymphoid Cells (ILC3s) [66], ILC22 and CD32NKp46+ lymphoid tissue inducer cells are lost in AhR-deficient mice. Similarly, ILC3’s inability to multiply in AhR-deficient mice constitutes an intrinsic function since AhR is required for the transcription of the cell-specific proliferator c-kit [67, 68]. As a result, secondary lymphoid structures, including cryptopatches and innate lymphoid follicles, are absent from the gut of AhR-deficient mice, which show susceptibility to C. rodentium. ILC3s feature the secretion of IL-17 and IL-22 [69]. AhR-deficient mice have elevated susceptibility to infection by C. rodentium, as well as dextran sulfate sodium (DSS)-associated colitis. DSS can damage the intestinal epithelium and induce inflammatory reactions and microbial dissemination. AhR-deficient mice containing wild-type IELs are resistant to DSS colitis, indicating IEL role in injury reduction.

AhR-deficient mice have lower amounts of skin and intestinal IELs and intestinal ILCs, thereby increasing susceptibility to C. rodentium infection. These cell types, and the generation of normal gut lymphoid follicles, are regulated by AhR ligands in the diet. In addition, activation of AhR by microbial products equally regulates the production of DP IELs, which constitute another critical group that controls intestinal immunity [70]. It may also be due to the lack of IL-22 that affects the commensal flora [71]. In fact, ID2, a transcription factor, regulates the expression of IL-22 in ILCs via AhR- and IL-23-dependent mechanisms, thereby modulating the intestinal colonization of C. rodentium [72]. In addition, AhR also controls the production of IL-22 by Th22 cells, which protect against intestinal pathogens [73, 74]. All these data suggest AhR has a critical function in controlling the interaction at environmental interfaces with microorganisms by regulating IL-22 and other cellular factors. Interestingly, cyp1a1 overexpression leads to the exhaustion of physiological AhR ligands and also increases susceptibility to intestinal bacterial infections [75], highlighting that AhR ligand availability and metabolism are important in controlling AhR-dependent immune effects.

Advertisement

8. AhR and T cells

AhR plays an important role in controlling adaptive immunity, and regulating T cell differentiation and direct or indirect functions by affecting antigen presenting cells. It was found that TCDD-activated AhR could inhibit the immune response [76], which is subsequently associated with CD4+ T cell induction [77, 78, 79]. In addition, the role of AhR in Th17 function and T cell-induced IL-22 biosynthesis have also been determined [74, 80, 81, 82, 83].

8.1 AhR and regulatory T cells (Tregs)

AhR shows high expression in Th17 cells, undetectable amounts in Th1 and Th2 cells, and low expression in Tregs. Tregs constitute a T cell subgroup, which helps maintain tolerance to autoantigens, preventing autoimmune pathologies. FoxP3+ Tregs [84, 85] and IL-10-producing type 1 regulatory T cells (Tr1 cells) [86] are the most typical Treg entities. Foxp3+ Tregs and Tr1 cells are associated with AhR.

TCDD, ITE, KYN and laquinimod derivatives activate AhR, thus increasing FoxP3+ Treg amounts via various mechanisms, e.g., by directly activating epigenetic modifications that regulate Foxp3 transcriptionally and via DC regulation [80, 87, 88, 89, 90, 91, 92]. In the presence of TGF - β 1, activating AhR with TCDD can also upregulate SMAD1 in human Tregs, resulting in stable expression of FoxP3 [93]. It was shown in mice with AhR-deficient T cells that AhR could also inhibit the activation of STAT1, which in turn inhibits FoxP3+ Treg differentiation [94]. In addition, AhR regulates the epigenetic modifier Aiolos, which downregulates genes associated with T cell’s effector function, such as IL-2 [87]. However, the effect of AhR on FoxP3+ Tregs may be affected by the applied experimental model, which may reflect the different effects of tissue-specific action and/or AhR agonist provided by the symbiotic flora [95].

Tr1 cells participate in controlling tissue inflammation via IL-10 secretion. IL-27 promotes the differentiation of Tr1 cells [96, 97, 98], while IL-21 plays an autocrine role in their stabilization [98, 99]. IL-27 upregulates AhR in Tr1 cells via STAT3. Then, AhR amounts are maintained by transactivation of the AhR promoter by AhR itself [100, 101, 102]. The important role of AhR in Tr1 cells in vivo is reflected by insufficient Tr1 cell differentiation induced by long-term anti-CD3 treatment of AhR-mutant mice.

AhR triggered CD39 equally affects Tr1 cell differentiation. After induction, T cells secrete eATP [103], which then interferes with the differentiation of Tr1 cells through hypoxia inducible factor-1α (HIF1-α). HIF1-α binding is superior to the interaction between AhR and ARNT, and promotes the degradation of AhR through the immune proteasome, thus inhibiting the differentiation of AhR dependent Tr1 cells [101]. The expression of CD39 driven by AhR can deplete eATP and promote the differentiation of Tr1 cells. Therefore, AhR regulates central genes in the Tr1 cell transcription program, while limiting the inhibitory effect of eATP-dependent HIF1-α induction on Tr1 cell differentiation. Overall, the above findings confirm AhR as a potential therapeutic target for immunomodulation.

8.2 AhR and T helper 17 (Th17) cells

Th17 cells, forming a unique CD4+ T cell subgroup, can biosynthesize Th17 cytokines and play key roles in the pathogenetic mechanisms of multiple inflammatory ailments. Their differentiation is triggered by IL-6 and transforming growth factor-beta (TGF-β). AhR can modulate Th17 cells by binding to the DRE site in the IL-17 promoter. In addition, AhR and STAT3 can synergistically upregulate Aiolos (IKZF3), an Ikaros family member, which can decrease the expression of IL-2, thus increasing Th17 cell amounts [64].

Th17 cells, producing IL-17A and expressing ROR-γt, are involved in immune responses to extracellular bacterial and fungal pathogens, and participate in the pathological mechanisms of multiple autoimmune diseases [104, 105]. Their differentiation involves joint effects of TGF-β and IL-6 or IL-21 [106, 107, 108]. AhR shows high expression in Th17 cells and is activated by FICZ, which can enhance Th17 cell differentiation and promote IL-22 expression. On the contrary, AhR deficiency can cause Th17 cells to produce IL-22, which may reflect AhR’s function in promoting RORγt recruitment to the IL-22 promoter.

8.3 AhR and other T cells

Th22 cells are a CD4+ T cell subpopulation. They produce IL-22 without IL-17’s intervention and their differentiation is induced by IL-6, IL-21 or IL-23. AhR controls the production of IL-22 in Th22 cells, and other cellular factors are essential for their mucosal immune functions [73, 109, 110, 111].

The AhR pathway also significantly affects CD8+ T cells. Activation of AhR by TCDD indirectly inhibits the primary response of CD8+ T cells to influenza virus through the regulatory mechanism of DC function [112]. In addition, CD8+ T cells of mouse models administered the AhR agonist TCDD in the developmental stage show a weak response to influenza virus infection later in life [113]. The above data indicate epigenetic alterations that can lead to prolonged functional defects in CD8+ T cells detectable after viral attack. Compared with other CD8+ T cell subsets, AhR expression is much higher in tissue resident CD8+ memory cells (TRMs). Taken together, these findings indicate that, similar to previously reported CD4+ T cell data, the AhR pathway plays a major role in regulating specific CD8+ T cell subgroups, such as TRMs and DP IELs.

AhR equally regulates γδ T cells, which are tissue resident lymphocytes. It regulates first-line immune response at epithelial sites and controls tissue homeostasis [114]. Despite AhR expression in the totality of γδ T-cell subgroups, AhR-deficiency significantly reduces the amounts of skin intraepithelial lymphocytes, mostly composed of Vγ3 and Vγ5 γδ T cells in the intestine and CD8αα αβ T cells [115]. AhR also regulates IL-22 expression by γδ T cells that produce IL-17 [116, 117]. The above data indicate that AhR has a significant effect on T cells residing in tissues, which supports further investigation of AhR’s function in non-CD4+ T cells.

In conclusion, AhR controls T cell responses at many levels and regulates transcription factors, enzymes, epigenetic modifiers and effector molecules that modulate T cell stability and metabolism. Lineage-specific responses to AhR induction may lead to ligand-specific effects, which are combined with cytokine-driven activities on the genome, thereby regulating AhR-interacting chaperones and controlling the accessibility of AhR’s direct and indirect transcription targets [118]. Comprehensive studies of these interactions should provide insights into the design of immune-modulators against AhR.

Advertisement

9. AhR and B cells

The B lymphocyte is an important part of humoral immunity, which has high specificity against a variety of pathogens. After stimulation via an antigen receptor, activation of immature B cells leads to clonal expansion, antibody isotype conversion and differentiation into antibody-secreting plasma cells, thus producing strong immune reactions [119]. In the process of infection, mature B cells in the lymph nodes and secondary lymphoid organs undergo somatic hypermutation and produce plasma cells featuring elevated antigen affinity and unique effector function [120].

It seems that all B cells produce AhR, but specific subsets, e.g., marginal B cell and B1 B cell subsets, have higher levels than the others. Li and collaborators demonstrated that AhR contributes to the development of B lymphocytes, based on cord blood CD34 and feeder cells, which promote B cell development. Meanwhile, AhR induction inhibits the formation of early B cells and pro-B cells. AhR controls B cell differentiation by transcriptionally suppressing the early B cell genes EBF1 and PAX5 [121].

AhR, overtly induced after activation of B cells, has a critical function in regulating the fate of activated cells. Vaidyanathan and colleagues revealed AhR suppresses switch-like recombination by changing the amounts of activated cytidine deaminase. These authors showed that AhR suppresses B cell transformation into plasmablasts and plasma cells that secrete antibodies [122]. In addition, Villa et al. provided evidence of a role for AhR in B cells, revealing that AhR expression is increased after administration of IL-4 as well as B cell receptor engagement. Nevertheless, the proliferation of AhR-deficient B cells is decreased, and cells could not progress to the S-phase. Furthermore, AhR-deficient B cells could not compete with the decreased AhR+/+ B cell capability of reconstructing the empty host, and could not induce antigen-dependent proliferation in mice. Gene expression profile analysis showed that AhR excision downregulates cyclin O, an important gene controlling the cell cycle [123].

Advertisement

10. AhR and dendritic cells (DCs)

DCs are essential in controlling T cell response and regulating immune tolerance [124]. AhR regulates DC differentiation and function, thereby profoundly affecting T cell-dependent immune reactions. AhR also affects antigen presentation by DCs. Bone marrow derived DCs (BMDCs) exposed to TCDD show decreased CD11c amounts, but increased production of MHC-II, CD86, IL6 and TNFα [125]. Similar findings were reported in TCDD treated splenic DCs [126]. However, different results were observed by using the AhR agonist ITE. The expression of MHC-II and co-stimulatory molecules and the production of Th1 and Th17 polarization cytokines in splenic DCs were decreased by ITE stimulation of AhR.

Recent experiments in ovalbumin-induced asthma models provide additional evidence for the physiological regulation of AhR in DCs, with AhR-deficient mice exhibiting enhanced inflammatory reactions, elevated Th2 differentiation and higher DC MHC-II and CD86 amounts [127]. In addition, AhR signaling has been reported to regulate the activity of CD103+/CD11b+ DCs during influenza virus infection, thereby reducing induction in protective CD8+ T cells [128]. Overall, this evidence confirms that AhR is a potential therapeutic target for regulating T cell responses in DC.

Multiple mechanisms are involved in AhR-associated regulation of DC function. AhR upregulates IDO 1 and 2 [129, 130], which catalyze the production of KYN, thus promoting the differentiation of FoxP3+ Tregs [131]. Indeed, AhR-deficient DCs could not induce Treg differentiation and Th17 cell proliferation in culture. It is consistent with the immunosuppressive effect of AhR in DCs. Recently, it was reported that IDO expression is maintained by an autocrine loop involving AhR and KYN in tumor infiltrating tolerogenic DCs [132]. Additionally, AhR induction in DCs induces a retinoic acid-dependent enzymatic mechanism, thus promoting FoxP3+ Treg differentiation and inhibiting effector T cells [133, 134, 135, 136, 137].

11. Conclusions

Studies evaluating AhR’s functions in immune cell development, immune response modulation and immune tolerance have aroused great interest. Originally, AhR was considered a protein sensing environmental substances and regulating drug metabolism. Recently, the role of AhR in regulating normal physiological processes has attracted increasing attention. The organism must perceive and mount substantial responses to environmental changes. Indeed, AhR senses biochemical, chemical and physical environments. Combined with a small amount of high-affinity physiological ligands, including FICZ and ICZ, AhR plays a role in cell proliferation, differentiation and function.

Current evidence indicates that AhR has a critical function in host response to bacterial pathogens. It also overtly influences resistance to infections by extracellular and intracellular bacteria. AhR is considered the best resistance factor for LM. It may have a new function in the innate immunity of LM infection, and AhR-deficient mice have increased sensitivity to LM. Activation of AhR can protect mice from the deadly attack of S. pneumoniae, inhibit bacterial growth and fight infection. AhR can also react with viral pathogens and parasitic infections. After infection by viruses and parasites, lack of AhR aggravates the host’s inflammatory response. AhR regulates host’s immune cells, confirming that AhR is a regulatory molecule with essential functions in the activation and induction of immune cells, e.g., T cells and inflammatory factors. Barrier organs are critical in immunity; specifically, large amounts of ahr are expressed in the intestine, which has a high potential for preventive and treatment interventions. AhR has a critical function in controlling the degree of inflammation in response to symbiotic microbiota and tissue destruction. Progress is being made in determining the molecular mechanisms by which AhR affects different cell types. To understand the complex process of AhR in immunity and antibacterial, to mitigate risks, and to develop novel treatment and prevention tools, more research is needed.

Abbreviations

AhRaryl hydrocarbon receptor
PASpern-arnt-sim
TCDD2,3,7,8-tetrachlorodibenzo-p-dioxin
ARNTaryl hydrocarbon receptor nuclear Translocator
AhREsAhR response elements
DREsdioxin
XREsxenobiotic
CYP1A1cytochrome P450 family 1 subfamily A member 1
AhRRaryl hydrocarbon receptor repressor
TDO22,3-dioxygenase
IDO2,3-dioxygenase
KYNUkynureninase
KMOkynurenine 3-monooxygenase
KYNkynurenine
DBDthe amino-terminal DNA binding domain
Bhlhbasic helix–loop–helix
NLSthe nuclear localization signal
TADthe transactivation domain
LMListeria monocytogenes
IL-6interleukin 6
TNF-αtumor necrosis factor alpha
NF-κBthe nuclear factor kappaB
CFUscolony forming units
MCP-1monocyte chemotactic protein 1
IFN-γinterferon γ
ROSreactive oxygen species
BPI/LBPbactericidal/permeability-increasing protein / lipopolysaccharide-binding protein
LPSlipopolysaccharide
NLRP3NLR family pyrin domain containing 3
3-Mc3-methylcholine
FICZ6-formylindolo[3,2-b]carbazole
HSVherpes simplex virus
iNOSinducible nitric oxide synthase
DCsdendritic cells
ILClymphoblastoid cell
RegIIIgType III Reg gamma
IELsintraepithelial lymphocytes
ILC3sgroup 3 Innate lymphoid cells
DSSdextran sulfate sodium
Tr1 cellstype 1 regulatory T cells
HIF1-αhypoxia inducible factor-1α
Th17T helper 17
TGF-βtransforming growth factor-beta
TRMstissue resident CD8+ memory cells
DCsdendritic cells
BMDCsbone marrow derived DCs

References

  1. 1. Y. Zhang, L. Huang, Z. Zuo, Y. Chen, C. Wang, Phenanthrene exposure causes cardiac arrhythmia in embryonic zebrafish via perturbing calcium handling, Aquatic toxicology 142 (2013) 26-32
  2. 2. L. Huang, Z. Xi, C. Wang, Y. Zhang, Z. Yang, S. Zhang, Y. Chen, Z. Zuo, Phenanthrene exposure induces cardiac hypertrophy via reducing miR-133a expression by DNA methylation, Scientific reports 6 (2016) 20105
  3. 3. L. Huang, Z. Zuo, Y. Zhang, C. Wang, Toxicogenomic analysis in the combined effect of tributyltin and benzo [a] pyrene on the development of zebrafish embryos, Aquatic Toxicology 158 (2015) 157-164
  4. 4. L. Huang, D. Gao, Y. Zhang, C. Wang, Z. Zuo, Exposure to low dose benzo [a] pyrene during early life stages causes symptoms similar to cardiac hypertrophy in adult zebrafish, Journal of hazardous materials 276 (2014) 377-382
  5. 5. T.V. Beischlag, J.L. Morales, B.D. Hollingshead, G.H. Perdew, The aryl hydrocarbon receptor complex and the control of gene expression, Critical Reviews™ in Eukaryotic Gene Expression 18(3) (2008)
  6. 6. T. Nakahama, A. Kimura, N.T. Nguyen, I. Chinen, H. Hanieh, K. Nohara, Y. Fujii-Kuriyama, T. Kishimoto, Aryl hydrocarbon receptor deficiency in T cells suppresses the development of collagen-induced arthritis, Proceedings of the National Academy of Sciences 108(34) (2011) 14222-14227
  7. 7. H. Liu, I. Ramachandran, D.I. Gabrilovich, Regulation of plasmacytoid dendritic cell development in mice by aryl hydrocarbon receptor, Immunology and cell biology 92(2) (2014) 200-203
  8. 8. F.J. Quintana, LeA (H) Rning self-control, Cell research 24(10) (2014) 1155-1156
  9. 9. B. Stockinger, P.D. Meglio, M. Gialitakis, J.H. Duarte, The aryl hydrocarbon receptor: multitasking in the immune system, Annual review of immunology 32 (2014) 403-432
  10. 10. P.B. Busbee, M. Rouse, M. Nagarkatti, P.S. Nagarkatti, Use of natural AhR ligands as potential therapeutic modalities against inflammatory disorders, Nutrition reviews 71(6) (2013) 353-369
  11. 11. M. Colonna, AHR: making the keratinocytes thick skinned, Immunity 40(6) (2014) 863-864
  12. 12. M.A. Wheeler, V. Rothhammer, F.J. Quintana, Control of immune-mediated pathology via the aryl hydrocarbon receptor, Journal of Biological Chemistry 292(30) (2017) 12383-12389
  13. 13. C.F. Vogel, E.M. Khan, P.S. Leung, M.E. Gershwin, W.W. Chang, D. Wu, T. Haarmann-Stemmann, A. Hoffmann, M.S. Denison, Cross-talk between Aryl hydrocarbon receptor and the inflammatory response a role for nuclear factor-κB, Journal of Biological Chemistry 289(3) (2014) 1866-1875
  14. 14. A. Korecka, A. Dona, S. Lahiri, A.J. Tett, M. Al-Asmakh, V. Braniste, R. D’Arienzo, A. Abbaspour, N. Reichardt, Y. Fujii-Kuriyama, Bidirectional communication between the Aryl hydrocarbon Receptor (AhR) and the microbiome tunes host metabolism, npj Biofilms and Microbiomes 2(1) (2016) 1-10
  15. 15. J. Fu, S.V. Nogueira, V. van Drongelen, P. Coit, S. Ling, E.F. Rosloniec, A.H. Sawalha, J. Holoshitz, Shared epitope–aryl hydrocarbon receptor crosstalk underlies the mechanism of gene–environment interaction in autoimmune arthritis, Proceedings of the National Academy of Sciences 115(18) (2018) 4755-4760
  16. 16. L. Huang, C. Wang, Y. Zhang, J. Li, Y. Zhong, Y. Zhou, Y. Chen, Z. Zuo, Benzo [a] pyrene exposure influences the cardiac development and the expression of cardiovascular relative genes in zebrafish (Danio rerio) embryos, Chemosphere 87(4) (2012) 369-375
  17. 17. Y. Zhang, C. Wang, L. Huang, R. Chen, Y. Chen, Z. Zuo, Low-level pyrene exposure causes cardiac toxicity in zebrafish (Danio rerio) embryos, Aquatic toxicology 114 (2012) 119-124
  18. 18. L. Huang, C. Wang, Y. Zhang, M. Wu, Z. Zuo, Phenanthrene causes ocular developmental toxicity in zebrafish embryos and the possible mechanisms involved, Journal of hazardous materials 261 (2013) 172-180
  19. 19. Y. Zhang, L. Huang, C. Wang, D. Gao, Z. Zuo, Phenanthrene exposure produces cardiac defects during embryo development of zebrafish (Danio rerio) through activation of MMP-9, Chemosphere 93(6) (2013) 1168-1175
  20. 20. F.L. Casado, K.P. Singh, T.A. Gasiewicz, The aryl hydrocarbon receptor: regulation of hematopoiesis and involvement in the progression of blood diseases, Blood Cells, Molecules, and Diseases 44(4) (2010) 199-206
  21. 21. F.J. Quintana, Regulation of central nervous system autoimmunity by the aryl hydrocarbon receptor, Seminars in immunopathology, Springer, 2013, pp. 627-635
  22. 22. Y. Zhang, L. Huang, Y. Zhao, T. Hu, Musk xylene induces malignant transformation of human liver cell line L02 via repressing the TGF-β signaling pathway, Chemosphere 168 (2017) 1506-1514
  23. 23. H. Sekine, J. Mimura, M. Oshima, H. Okawa, J. Kanno, K. Igarashi, F.J. Gonzalez, T. Ikuta, K. Kawajiri, Y. Fujii-Kuriyama, Hypersensitivity of aryl hydrocarbon receptor-deficient mice to lipopolysaccharide-induced septic shock, Molecular and cellular biology 29(24) (2009) 6391-6400
  24. 24. S. Mohammadi, F.S. Seyedhosseini, N. Behnampour, Y. Yazdani, Indole-3-carbinol induces G1 cell cycle arrest and apoptosis through aryl hydrocarbon receptor in THP-1 monocytic cell line, Journal of receptors and signal transduction 37(5) (2017) 506-514
  25. 25. B. Stockinger, K. Hirota, J. Duarte, M. Veldhoen, External influences on the immune system via activation of the aryl hydrocarbon receptor, Seminars in immunology, Elsevier, 2011, pp. 99-105
  26. 26. H. Hanieh, Toward understanding the role of aryl hydrocarbon receptor in the immune system: current progress and future trends, BioMed research international 2014 (2014)
  27. 27. A. Kimura, H. Abe, S. Tsuruta, S. Chiba, Y. Fujii-Kuriyama, T. Sekiya, R. Morita, A. Yoshimura, Aryl hydrocarbon receptor protects against bacterial infection by promoting macrophage survival and reactive oxygen species production, International immunology 26(4) (2014) 209-220
  28. 28. P. Di Meglio, J.H. Duarte, H. Ahlfors, N.D. Owens, Y. Li, F. Villanova, I. Tosi, K. Hirota, F.O. Nestle, U. Mrowietz, Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions, Immunity 40(6) (2014) 989-1001
  29. 29. A. Bessede, M. Gargaro, M.T. Pallotta, D. Matino, G. Servillo, C. Brunacci, S. Bicciato, E.M. Mazza, A. Macchiarulo, C. Vacca, Aryl hydrocarbon receptor control of a disease tolerance defence pathway, Nature 511(7508) (2014) 184-190
  30. 30. T.D. Hubbard, I.A. Murray, G.H. Perdew, Indole and tryptophan metabolism: endogenous and dietary routes to Ah receptor activation, Drug Metabolism and Disposition 43(10) (2015) 1522-1535
  31. 31. D. Liu, B. Ray, D.R. Neavin, J. Zhang, A.P. Athreya, J.M. Biernacka, W.V. Bobo, D.K. Hall-Flavin, M.K. Skime, H. Zhu, Beta-defensin 1, aryl hydrocarbon receptor and plasma kynurenine in major depressive disorder: metabolomics-informed genomics, Translational psychiatry 8(1) (2018) 1-13
  32. 32. D.W. Nebert, J.R. Robinson, A. Niwa, K. Kumari, A.P. Poland, Genetic expression of aryl hydrocarbon hydroxylase activity in the mouse, Journal of cellular physiology 85(S1) (1975) 393-414
  33. 33. W.F. Greenlee, A. Poland, Nuclear uptake of 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin in C57BL/6J and DBA/2J mice. Role of the hepatic cytosol receptor protein, Journal of Biological Chemistry 254(19) (1979) 9814-9821
  34. 34. M.B. Black, R.A. Budinsky, A. Dombkowski, D. Cukovic, E.L. LeCluyse, S.S. Ferguson, R.S. Thomas, J.C. Rowlands, Cross-species comparisons of transcriptomic alterations in human and rat primary hepatocytes exposed to 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin, Toxicological Sciences 127(1) (2012) 199-215
  35. 35. K.W. Schulte, E. Green, A. Wilz, M. Platten, O. Daumke, Structural basis for aryl hydrocarbon receptor-mediated gene activation, Structure 25(7) (2017) 1025-1033. e3
  36. 36. M.S. Denison, S.R. Nagy, Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals, Annual review of pharmacology and toxicology 43(1) (2003) 309-334
  37. 37. S. Luecke-Johansson, M. Gralla, H. Rundqvist, J.C. Ho, R.S. Johnson, K. Gradin, L. Poellinger, A molecular mechanism to switch the aryl hydrocarbon receptor from a transcription factor to an E3 ubiquitin ligase, Molecular and cellular biology 37(13) (2017)
  38. 38. J. Mimura, M. Ema, K. Sogawa, Y. Fujii-Kuriyama, Identification of a novel mechanism of regulation of Ah (dioxin) receptor function, Genes & development 13(1) (1999) 20-25
  39. 39. T. Baba, J. Mimura, K. Gradin, A. Kuroiwa, T. Watanabe, Y. Matsuda, J. Inazawa, K. Sogawa, Y. Fujii-Kuriyama, Structure and expression of the Ah receptor repressor gene, Journal of Biological Chemistry 276(35) (2001) 33101-33110
  40. 40. L.Z. Shi, N.G. Faith, Y. Nakayama, M. Suresh, H. Steinberg, C.J. Czuprynski, The aryl hydrocarbon receptor is required for optimal resistance to Listeria monocytogenes infection in mice, The Journal of Immunology 179(10) (2007) 6952-6962
  41. 41. B.A. Vorderstrasse, B.P. Lawrence, Protection against lethal challenge with Streptococcus pneumoniae is conferred by aryl hydrocarbon receptor activation but is not associated with an enhanced inflammatory response, Infection and immunity 74(10) (2006) 5679-5686
  42. 42. L. Huang, Y. Zuo, Q . Jiang, Y. Su, Y. Qin, X. Xu, L. Zhao, Q . Yan, A metabolomic investigation into the temperature-dependent virulence of Pseudomonas plecoglossicida from large yellow croaker (Pseudosciaena crocea), Journal of fish diseases 42(3) (2019) 431-446
  43. 43. R. He, L. Zhao, X. Xu, W. Zheng, J. Zhang, J. Zhang, Q . Yan, L. Huang, Aryl hydrocarbon receptor is required for immune response in Epinephelus coioides and Danio rerio infected by Pseudomonas plecoglossicida, Fish & Shellfish Immunology 97 (2020) 564-570
  44. 44. L. Huang, W. Qi, Y. Zuo, S.A. Alias, W. Xu, The immune response of a warm water fish orange-spotted grouper (Epinephelus coioides) infected with a typical cold water bacterial pathogen Aeromonas salmonicida is AhR dependent, Developmental & Comparative Immunology 113 (2020) 103779
  45. 45. W. Huai, R. Zhao, H. Song, J. Zhao, L. Zhang, L. Zhang, C. Gao, L. Han, W. Zhao, Aryl hydrocarbon receptor negatively regulates NLRP3 inflammasome activity by inhibiting NLRP3 transcription, Nature communications 5(1) (2014) 1-9
  46. 46. T. Veiga-Parga, A. Suryawanshi, B.T. Rouse, Controlling viral immuno-inflammatory lesions by modulating aryl hydrocarbon receptor signaling, PLoS Pathog 7(12) (2011) e1002427
  47. 47. T.K. Warren, K.A. Mitchell, B.P. Lawrence, 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, Toxicological Sciences 56(1) (2000) 114-123
  48. 48. S. Teske, A. Bohn, J. Regal, J. Neumiller, B. Lawrence, Exploring mechanisms that underlie aryl hydrocarbon receptor-mediated increases in pulmonary neutrophilia and diminished host resistance to influenza A virus, Am J Physiol Lung Cell Mol Physiol 289 (2005) 111-124
  49. 49. T. Mauad, L.A. Hajjar, G.D. Callegari, L.F. da Silva, D. Schout, F.R. Galas, V.A. Alves, D.M. Malheiros, J.O. Auler Jr, A.F. Ferreira, Lung pathology in fatal novel human influenza A (H1N1) infection, American journal of respiratory and critical care medicine 181(1) (2010) 72-79
  50. 50. L.A. Perrone, J.K. Plowden, A. García-Sastre, J.M. Katz, T.M. Tumpey, H5N1 and 1918 pandemic influenza virus infection results in early and excessive infiltration of macrophages and neutrophils in the lungs of mice, PLoS Pathog 4(8) (2008) e1000115
  51. 51. Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D’Angelo C, Massi-Benedetti C, Fallarino F, Carvalho A, Puccetti P, Romani L. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22, Immunity 39(2) 372-85
  52. 52. F. Dallaire, É. Dewailly, C. Vézina, G. Muckle, J.-P. Weber, S. Bruneau, P. Ayotte, Effect of prenatal exposure to polychlorinated biphenyls on incidence of acute respiratory infections in preschool Inuit children, Environmental health perspectives 114(8) (2006) 1301-1305
  53. 53. S.B. Stølevik, U.C. Nygaard, E. Namork, M. Haugen, H.E. Kvalem, H.M. Meltzer, J. Alexander, J.H. van Delft, H. van Loveren, M. Løvik, Prenatal exposure to polychlorinated biphenyls and dioxins is associated with increased risk of wheeze and infections in infants, Food and chemical toxicology 49(8) (2011) 1843-1848
  54. 54. R.L. Van Den Heuvel, G. Koppen, J.A. Staessen, E.D. Hond, G. Verheyen, T.S. Nawrot, H.A. Roels, R. Vlietinck, G.E. Schoeters, Immunologic biomarkers in relation to exposure markers of PCBs and dioxins in Flemish adolescents (Belgium), Environmental Health Perspectives 110(6) (2002) 595-600
  55. 55. H. Neff-LaFord, S. Teske, T.P. Bushnell, B.P. Lawrence, Aryl hydrocarbon receptor activation during influenza virus infection unveils a novel pathway of IFN-γ production by phagocytic cells, The Journal of Immunology 179(1) (2007) 247-255
  56. 56. S. Wagage, B. John, B.L. Krock, A.O.H. Hall, L.M. Randall, C.L. Karp, M.C. Simon, C.A. Hunter, The aryl hydrocarbon receptor promotes IL-10 production by NK cells, The Journal of Immunology 192(4) (2014) 1661-1670
  57. 57. Y. Sanchez, J. de Dios Rosado, L. Vega, G. Elizondo, E. Estrada-Muñiz, R. Saavedra, I. Juárez, M. Rodríguez-Sosa, The unexpected role for the aryl hydrocarbon receptor on susceptibility to experimental toxoplasmosis, Journal of Biomedicine and Biotechnology 2010 (2010)
  58. 58. J. Stange, M. Veldhoen, The aryl hydrocarbon receptor in innate T cell immunity, Seminars in immunopathology, Springer, 2013, pp. 645-655
  59. 59. H.U. Lee, Z.E. McPherson, B. Tan, A. Korecka, S. Pettersson, Host-microbiome interactions: the aryl hydrocarbon receptor and the central nervous system, Journal of molecular medicine 95(1) (2017) 29-39
  60. 60. C. Goudot, A. Coillard, A.-C. Villani, P. Gueguen, A. Cros, S. Sarkizova, T.-L. Tang-Huau, M. Bohec, S. Baulande, N. Hacohen, Aryl hydrocarbon receptor controls monocyte differentiation into dendritic cells versus macrophages, Immunity 47(3) (2017) 582-596. e6
  61. 61. J. Qiu, X. Guo, E.C. Zong-ming, L. He, G.F. Sonnenberg, D. Artis, Y.-X. Fu, L. Zhou, Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon receptor signaling and regulation of microflora, Immunity 39(2) (2013) 386-399
  62. 62. T. Zelante, R.G. Iannitti, C. Cunha, A. De Luca, G. Giovannini, G. Pieraccini, R. Zecchi, C. D’Angelo, C. Massi-Benedetti, F. Fallarino, Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22, Immunity 39(2) (2013) 372-385
  63. 63. A. Rannug, U. Rannug, H. Rosenkranz, L. Winqvist, R. Westerholm, E. Agurell, A. Grafström, Certain photooxidized derivatives of tryptophan bind with very high affinity to the Ah receptor and are likely to be endogenous signal substances, Journal of Biological Chemistry 262(32) (1987) 15422-15427
  64. 64. S. Chmill, S. Kadow, M. Winter, H. Weighardt, C. Esser, 2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin impairs stable establishment of oral tolerance in mice, Toxicological Sciences 118(1) (2010) 98-107
  65. 65. Y. Li, S. Innocentin, D.R. Withers, N.A. Roberts, A.R. Gallagher, E.F. Grigorieva, C. Wilhelm, M. Veldhoen, Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation, Cell 147(3) (2011) 629-640
  66. 66. H. Spits, D. Artis, M. Colonna, A. Diefenbach, J.P. Di Santo, G. Eberl, S. Koyasu, R.M. Locksley, A.N. McKenzie, R.E. Mebius, Innate lymphoid cells—a proposal for uniform nomenclature, Nature reviews immunology 13(2) (2013) 145-149
  67. 67. E.A. Kiss, C. Vonarbourg, S. Kopfmann, E. Hobeika, D. Finke, C. Esser, A. Diefenbach, Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles, Science 334(6062) (2011) 1561-1565
  68. 68. J.S. Lee, M. Cella, K.G. McDonald, C. Garlanda, G.D. Kennedy, M. Nukaya, A. Mantovani, R. Kopan, C.A. Bradfield, R.D. Newberry, AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch, Nature immunology 13(2) (2012) 144-151
  69. 69. A. Diefenbach, Innate lymphoid cells in the defense against infections, European Journal of Microbiology and Immunology 3(3) (2013) 143-151
  70. 70. L. Cervantes-Barragan, J.N. Chai, M.D. Tianero, B. Di Luccia, P.P. Ahern, J. Merriman, V.S. Cortez, M.G. Caparon, M.S. Donia, S. Gilfillan, Lactobacillus reuteri induces gut intraepithelial CD4+ CD8αα+ T cells, Science 357(6353) (2017) 806-810
  71. 71. L.A. Zenewicz, X. Yin, G. Wang, E. Elinav, L. Hao, L. Zhao, R.A. Flavell, IL-22 deficiency alters colonic microbiota to be transmissible and colitogenic, The Journal of Immunology 190(10) (2013) 5306-5312
  72. 72. X. Guo, Y. Liang, Y. Zhang, A. Lasorella, B.L. Kee, Y.-X. Fu, Innate lymphoid cells control early colonization resistance against intestinal pathogens through ID2-dependent regulation of the microbiota, Immunity 42(4) (2015) 731-743
  73. 73. R. Basu, D.B. O’Quinn, D.J. Silberger, T.R. Schoeb, L. Fouser, W. Ouyang, R.D. Hatton, C.T. Weaver, Th22 cells are an important source of IL-22 for host protection against enteropathogenic bacteria, Immunity 37(6) (2012) 1061-1075
  74. 74. A. Yeste, I.D. Mascanfroni, M. Nadeau, E.J. Burns, A.-M. Tukpah, A. Santiago, C. Wu, B. Patel, D. Kumar, F.J. Quintana, IL-21 induces IL-22 production in CD4+ T cells, Nature communications 5(1) (2014) 1-13
  75. 75. C. Schiering, E. Wincent, A. Metidji, A. Iseppon, Y. Li, A.J. Potocnik, S. Omenetti, C.J. Henderson, C.R. Wolf, D.W. Nebert, Feedback control of AHR signalling regulates intestinal immunity, Nature 542(7640) (2017) 242-245
  76. 76. N.I. Kerkvliet, B. Smith, L.B. STEPPAN, J. Youngberg, M. Henderson, D. Buhler, Role of the Ah locus in suppression of cytotoxic T lymphocyte activity by halogenated aromatic hydrocarbons (PCBs and TCDD): structure-activity relationships and effects in C57BI/6 mice congenic at the Ah locus, Toxicological Sciences 14(3) (1990) 532-541
  77. 77. C.J. Funatake, N.B. Marshall, L.B. Steppan, D.V. Mourich, N.I. Kerkvliet, 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, The Journal of Immunology 175(7) (2005) 4184-4188
  78. 78. N.I. Kerkvliet, D.M. Shepherd, L. Baecher-Steppan, 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, Toxicology and applied pharmacology 185(2) (2002) 146-152
  79. 79. N.B. Marshall, W.R. Vorachek, L.B. Steppan, D.V. Mourich, N.I. Kerkvliet, Functional characterization and gene expression analysis of CD4+ CD25+ regulatory T cells generated in mice treated with 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin, The Journal of Immunology 181(4) (2008) 2382-2391
  80. 80. F.J. Quintana, A.S. Basso, A.H. Iglesias, T. Korn, M.F. Farez, E. Bettelli, M. Caccamo, M. Oukka, H.L. Weiner, Control of T reg and TH 17 cell differentiation by the aryl hydrocarbon receptor, Nature 453(7191) (2008) 65-71
  81. 81. S. Rutz, R. Noubade, C. Eidenschenk, N. Ota, W. Zeng, Y. Zheng, J. Hackney, J. Ding, H. Singh, W. Ouyang, Transcription factor c-Maf mediates the TGF-β-dependent suppression of IL-22 production in TH 17 cells, Nature immunology 12(12) (2011) 1238-1245
  82. 82. M. Veldhoen, K. Hirota, A.M. Westendorf, J. Buer, L. Dumoutier, J.-C. Renauld, B. Stockinger, The aryl hydrocarbon receptor links TH 17-cell-mediated autoimmunity to environmental toxins, Nature 453(7191) (2008) 106-109
  83. 83. M. Veldhoen, K. Hirota, J. Christensen, A. O’Garra, B. Stockinger, Natural agonists for aryl hydrocarbon receptor in culture medium are essential for optimal differentiation of Th17 T cells, Journal of Experimental Medicine 206(1) (2009) 43-49
  84. 84. J.D. Fontenot, M.A. Gavin, A.Y. Rudensky, Foxp3 programs the development and function of CD4+ CD25+ regulatory T cells, Nature immunology 4(4) (2003) 330-336
  85. 85. S. Hori, T. Nomura, S. Sakaguchi, Control of regulatory T cell development by the transcription factor Foxp3, Science 299(5609) (2003) 1057-1061
  86. 86. H. Groux, A. O’Garra, M. Bigler, M. Rouleau, S. Antonenko, J.E. De Vries, M.G. Roncarolo, A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis, Nature 389(6652) (1997) 737-742
  87. 87. J.A. Goettel, R. Gandhi, J.E. Kenison, A. Yeste, G. Murugaiyan, S. Sambanthamoorthy, A.E. Griffith, B. Patel, D.S. Shouval, H.L. Weiner, AHR activation is protective against colitis driven by T cells in humanized mice, Cell reports 17(5) (2016) 1318-1329
  88. 88. J. Kaye, V. Piryatinsky, T. Birnberg, T. Hingaly, E. Raymond, R. Kashi, E. Amit-Romach, I.S. Caballero, F. Towfic, M.A. Ator, Laquinimod arrests experimental autoimmune encephalomyelitis by activating the aryl hydrocarbon receptor, Proceedings of the National Academy of Sciences 113(41) (2016) E6145-E6152
  89. 89. N.I. Kerkvliet, L.B. Steppan, W. Vorachek, S. Oda, D. Farrer, C.P. Wong, D. Pham, D.V. Mourich, Activation of aryl hydrocarbon receptor by TCDD prevents diabetes in NOD mice and increases Foxp3+ T cells in pancreatic lymph nodes, Immunotherapy 1(4) (2009) 539-547
  90. 90. J.D. Mezrich, J.H. Fechner, X. Zhang, B.P. Johnson, W.J. Burlingham, C.A. Bradfield, An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells, The Journal of Immunology 185(6) (2010) 3190-3198
  91. 91. N.P. Singh, U.P. Singh, M. Rouse, J. Zhang, S. Chatterjee, P.S. Nagarkatti, M. Nagarkatti, Dietary indoles suppress delayed-type hypersensitivity by inducing a switch from proinflammatory Th17 cells to anti-inflammatory regulatory T cells through regulation of microRNA, The Journal of Immunology 196(3) (2016) 1108-1122
  92. 92. N.P. Singh, U.P. Singh, B. Singh, R.L. Price, M. Nagarkatti, P.S. Nagarkatti, 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(8) (2011) e23522
  93. 93. R. Gandhi, D. Kumar, E.J. Burns, M. Nadeau, B. Dake, A. Laroni, D. Kozoriz, H.L. Weiner, F.J. Quintana, Activation of the aryl hydrocarbon receptor induces human type 1 regulatory T cell–like and Foxp3+ regulatory T cells, Nature immunology 11(9) (2010) 846-853
  94. 94. A. Kimura, T. Naka, K. Nohara, Y. Fujii-Kuriyama, T. Kishimoto, Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells, Proceedings of the National Academy of Sciences 105(28) (2008) 9721-9726
  95. 95. T.A. Dant, K.L. Lin, D.W. Bruce, S.A. Montgomery, O.V. Kolupaev, H. Bommiasamy, L.M. Bixby, J.T. Woosley, K.P. McKinnon, F.J. Gonzalez, T-cell expression of AhR inhibits the maintenance of pTreg cells in the gastrointestinal tract in acute GVHD, Blood, The Journal of the American Society of Hematology 130(3) (2017) 348-359
  96. 96. A. Awasthi, Y. Carrier, J.P. Peron, E. Bettelli, M. Kamanaka, R.A. Flavell, V.K. Kuchroo, M. Oukka, H.L. Weiner, A dominant function for interleukin 27 in generating interleukin 10–producing anti-inflammatory T cells, Nature immunology 8(12) (2007) 1380-1389
  97. 97. D.C. Fitzgerald, B. Ciric, T. Touil, H. Harle, J. Grammatikopolou, J.D. Sarma, B. Gran, G.-X. Zhang, A. Rostami, Suppressive effect of IL-27 on encephalitogenic Th17 cells and the effector phase of experimental autoimmune encephalomyelitis, The Journal of Immunology 179(5) (2007) 3268-3275
  98. 98. J.S. Stumhofer, A. Laurence, E.H. Wilson, E. Huang, C.M. Tato, L.M. Johnson, A.V. Villarino, Q . Huang, A. Yoshimura, D. Sehy, Interleukin 27 negatively regulates the development of interleukin 17–producing T helper cells during chronic inflammation of the central nervous system, Nature immunology 7(9) (2006) 937-945
  99. 99. R. Spolski, H.-P. Kim, W. Zhu, D.E. Levy, W.J. Leonard, IL-21 mediates suppressive effects via its induction of IL-10, The Journal of Immunology 182(5) (2009) 2859-2867
  100. 100. L. Apetoh, F.J. Quintana, C. Pot, N. Joller, S. Xiao, D. Kumar, E.J. Burns, D.H. Sherr, H.L. Weiner, V.K. Kuchroo, The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27, Nature immunology 11(9) (2010) 854-861
  101. 101. I.D. Mascanfroni, M.C. Takenaka, A. Yeste, B. Patel, Y. Wu, J.E. Kenison, S. Siddiqui, A.S. Basso, L.E. Otterbein, D.M. Pardoll, Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1-α, Nature medicine 21(6) (2015) 638-646
  102. 102. H.Y. Wu, F.J. Quintana, A.P. Da Cunha, B.T. Dake, T. Koeglsperger, S.C. Starossom, H.L. Weiner, In vivo induction of Tr1 cells via mucosal dendritic cells and AHR signaling, PloS one 6(8) (2011) e23618
  103. 103. M.C. Takenaka, S. Robson, F.J. Quintana, Regulation of the T cell response by CD39, Trends in immunology 37(7) (2016) 427-439
  104. 104. I.I. Ivanov, B.S. McKenzie, L. Zhou, C.E. Tadokoro, A. Lepelley, J.J. Lafaille, D.J. Cua, D.R. Littman, The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells, Cell 126(6) (2006) 1121-1133
  105. 105. T. Korn, E. Bettelli, M. Oukka, V.K. Kuchroo, IL-17 and Th17 Cells, Annual review of immunology 27 (2009) 485-517
  106. 106. T. Korn, J. Reddy, W. Gao, E. Bettelli, A. Awasthi, T.R. Petersen, B.T. Bäckström, R.A. Sobel, K.W. Wucherpfennig, T.B. Strom, Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation, Nature medicine 13(4) (2007) 423-431
  107. 107. R. Nurieva, X.O. Yang, G. Martinez, Y. Zhang, A.D. Panopoulos, L. Ma, K. Schluns, Q . Tian, S.S. Watowich, A.M. Jetten, Essential autocrine regulation by IL-21 in the generation of inflammatory T cells, Nature 448(7152) (2007) 480-483
  108. 108. L. Vikström Bergander, W. Cai, B. Klocke, M. Seifert, I. Pongratz, Tryptamine serves as a proligand of the AhR transcriptional pathway whose activation is dependent of monoamine oxidases, Molecular Endocrinology 26(9) (2012) 1542-1551
  109. 109. T. Duhen, R. Geiger, D. Jarrossay, A. Lanzavecchia, F. Sallusto, Production of interleukin 22 but not interleukin 17 by a subset of human skin-homing memory T cells, Nature immunology 10(8) (2009) 857-863
  110. 110. J.M. Ramirez, N.C. Brembilla, O. Sorg, R. Chicheportiche, T. Matthes, J.M. Dayer, J.H. Saurat, E. Roosnek, C. Chizzolini, Activation of the aryl hydrocarbon receptor reveals distinct requirements for IL-22 and IL-17 production by human T helper cells, European journal of immunology 40(9) (2010) 2450-2459
  111. 111. S. Trifari, C.D. Kaplan, E.H. Tran, N.K. Crellin, H. Spits, Identification of a human helper T cell population that has abundant production of interleukin 22 and is distinct from TH-17, TH 1 and TH 2 cells, Nature immunology 10(8) (2009) 864-871
  112. 112. B.P. Lawrence, A.D. Roberts, J.J. Neumiller, J.A. Cundiff, D.L. Woodland, Aryl hydrocarbon receptor activation impairs the priming but not the recall of influenza virus-specific CD8+ T cells in the lung, The Journal of Immunology 177(9) (2006) 5819-5828
  113. 113. B. Winans, A. Nagari, M. Chae, C.M. Post, C.-I. Ko, A. Puga, W.L. Kraus, B.P. Lawrence, Linking the aryl hydrocarbon receptor with altered DNA methylation patterns and developmentally induced aberrant antiviral CD8+ T cell responses, The Journal of Immunology 194(9) (2015) 4446-4457
  114. 114. A.C. Hayday, γδ T cells and the lymphoid stress-surveillance response, Immunity 31(2) (2009) 184-196
  115. 115. S. Kadow, B. Jux, S.P. Zahner, B. Wingerath, S. Chmill, B.E. Clausen, J. Hengstler, C. Esser, Aryl hydrocarbon receptor is critical for homeostasis of invariant γδ T cells in the murine epidermis, The Journal of Immunology 187(6) (2011) 3104-3110
  116. 116. D. Cibrian, M.L. Saiz, H. de la Fuente, R. Sánchez-Díaz, O. Moreno-Gonzalo, I. Jorge, A. Ferrarini, J. Vázquez, C. Punzón, M. Fresno, CD69 controls the uptake of L-tryptophan through LAT1-CD98 and AhR-dependent secretion of IL-22 in psoriasis, Nature immunology 17(8) (2016) 985-996
  117. 117. B. Martin, K. Hirota, D.J. Cua, B. Stockinger, M. Veldhoen, Interleukin-17-producing γδ T cells selectively expand in response to pathogen products and environmental signals, Immunity 31(2) (2009) 321-330
  118. 118. N. Yosef, A. Regev, Writ large: genomic dissection of the effect of cellular environment on immune response, Science 354(6308) (2016) 64-68
  119. 119. J.J. Taylor, M.K. Jenkins, K.A. Pape, Heterogeneity in the differentiation and function of memory B cells, Trends in immunology 33(12) (2012) 590-597
  120. 120. C. Boboila, F.W. Alt, B. Schwer, Classical and alternative end-joining pathways for repair of lymphocyte-specific and general DNA double-strand breaks, Advances in immunology, Elsevier2012, pp. 1-49
  121. 121. J. Li, S. Bhattacharya, J. Zhou, A.S. Phadnis-Moghe, R.B. Crawford, N.E. Kaminski, Aryl hydrocarbon receptor activation suppresses EBF1 and PAX5 and impairs human B lymphopoiesis, The Journal of Immunology 199(10) (2017) 3504-3515
  122. 122. B. Vaidyanathan, A. Chaudhry, W.T. Yewdell, D. Angeletti, W.-F. Yen, A.K. Wheatley, C.A. Bradfield, A.B. McDermott, J.W. Yewdell, A.Y. Rudensky, The aryl hydrocarbon receptor controls cell-fate decisions in B cells, Journal of Experimental Medicine 214(1) (2017) 197-208
  123. 123. M. Villa, M. Gialitakis, M. Tolaini, H. Ahlfors, C.J. Henderson, C.R. Wolf, R. Brink, B. Stockinger, Aryl hydrocarbon receptor is required for optimal B-cell proliferation, The EMBO journal 36(1) (2017) 116-128
  124. 124. P. Guermonprez, J. Valladeau, L. Zitvogel, C. Théry, S. Amigorena, Antigen presentation and T cell stimulation by dendritic cells, Annual review of immunology 20(1) (2002) 621-667
  125. 125. J. Bankoti, B. Rase, T. Simones, D.M. Shepherd, Functional and phenotypic effects of AhR activation in inflammatory dendritic cells, Toxicology and applied pharmacology 246(1-2) (2010) 18-28
  126. 126. J. Bankoti, A. Burnett, S. Navarro, A.K. Miller, B. Rase, D.M. Shepherd, Effects of TCDD on the fate of naive dendritic cells, Toxicological Sciences 115(2) (2010) 422-434
  127. 127. T.H. Thatcher, M.A. Williams, S.J. Pollock, C.E. McCarthy, S.H. Lacy, R.P. Phipps, P.J. Sime, Endogenous ligands of the aryl hydrocarbon receptor regulate lung dendritic cell function, Immunology 147(1) (2016) 41-54
  128. 128. G.B. Jin, B. Winans, K.C. Martin, B.P. Lawrence, New insights into the role of the aryl hydrocarbon receptor in the function of CD11c+ cells during respiratory viral infection, European journal of immunology 44(6) (2014) 1685-1698
  129. 129. N.T. Nguyen, A. Kimura, T. Nakahama, I. Chinen, K. Masuda, K. Nohara, Y. Fujii-Kuriyama, T. Kishimoto, Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism, Proceedings of the National Academy of Sciences 107(46) (2010) 19961-19966
  130. 130. C.F. Vogel, S.R. Goth, B. Dong, I.N. Pessah, F. Matsumura, Aryl hydrocarbon receptor signaling mediates expression of indoleamine 2, 3-dioxygenase, Biochemical and biophysical research communications 375(3) (2008) 331-335
  131. 131. F. Fallarino, U. Grohmann, S. You, B.C. McGrath, D.R. Cavener, C. Vacca, C. Orabona, R. Bianchi, M.L. Belladonna, C. Volpi, The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor ζ-chain and induce a regulatory phenotype in naive T cells, The Journal of Immunology 176(11) (2006) 6752-6761
  132. 132. Q . Li, J.L. Harden, C.D. Anderson, N.K. Egilmez, Tolerogenic Phenotype of IFN-γ–Induced IDO+ Dendritic Cells Is Maintained via an Autocrine IDO–Kynurenine/AhR–IDO Loop, The Journal of Immunology 197(3) (2016) 962-970
  133. 133. J.L. Coombes, K.R. Siddiqui, C.V. Arancibia-Cárcamo, J. Hall, C.-M. Sun, Y. Belkaid, F. Powrie, A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-β–and retinoic acid–dependent mechanism, Journal of Experimental Medicine 204(8) (2007) 1757-1764
  134. 134. D. Mucida, Y. Park, G. Kim, O. Turovskaya, I. Scott, M. Kronenberg, H. Cheroutre, Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid, science 317(5835) (2007) 256-260
  135. 135. J. Nolting, C. Daniel, S. Reuter, C. Stuelten, P. Li, H. Sucov, B.-G. Kim, J.J. Letterio, K. Kretschmer, H.-J. Kim, Retinoic acid can enhance conversion of naive into regulatory T cells independently of secreted cytokines, Journal of Experimental Medicine 206(10) (2009) 2131-2139
  136. 136. K. Pino-Lagos, Y. Guo, C. Brown, M.P. Alexander, R. Elgueta, K.A. Bennett, V. De Vries, E. Nowak, R. Blomhoff, S. Sockanathan, A retinoic acid–dependent checkpoint in the development of CD4+ T cell–mediated immunity, Journal of Experimental Medicine 208(9) (2011) 1767-1775
  137. 137. C.-M. Sun, J.A. Hall, R.B. Blank, N. Bouladoux, M. Oukka, J.R. Mora, Y. Belkaid, Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid, The Journal of experimental medicine 204(8) (2007) 1775-1785

Written By

Lixing Huang, Rongchao He, Youyu Zhang and Qingpi Yan

Submitted: 07 December 2020 Reviewed: 09 February 2021 Published: 01 March 2021