Abstract
Host–microorganism encounters take place in many different ways and with different types of outcomes. Three major types of microorganisms need to be distinguished: (1) pathogens that cause harm to the host and must be controlled; (2) environmental microorganisms that can be ignored but must be controlled at higher abundance; and (3) symbiotic microbiota that require support by the host. Recent evidence indicates that the aryl hydrocarbon receptor (AHR) senses and initiates signalling and gene expression in response to a plethora of microorganisms and infectious conditions. It was originally identified as a receptor that binds xenobiotics. However, it was subsequently found to have a critical role in numerous biological processes, including immunity and inflammation and was recently classified as a pattern recognition receptor. Here we review the role of the AHR in host–pathogen interactions, focusing on AHR sensing of different microbial classes, the ligands involved, responses elicited and disease outcomes. Moreover, we explore the therapeutic potential of targeting the AHR in the context of infection.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Kewley, R. J., Whitelaw, M. L. & Chapman-Smith, A. The mammalian basic helix-loop-helix/PAS family of transcriptional regulators. Int. J. Biochem. Cell Biol. 36, 189–204 (2004).
Greenlee, W. F. & Poland, A. Nuclear uptake of 2,3,7,8-tetrachlorodibenzo-p-dioxin in C57BL/6J and DBA/2J mice. Role of the hepatic cytosol receptor protein. J. Biol. Chem. 254, 9814–9821 (1979).
Poland, A., Glover, E. & Kende, A. S. Stereospecific, high affinity binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin by hepatic cytosol. Evidence that the binding species is receptor for induction of aryl hydrocarbon hydroxylase. J. Biol. Chem. 251, 4936–4946 (1976).
Riddick, D. S. Fifty years of aryl hydrocarbon receptor research as reflected in the pages of Drug Metabolism and Disposition. Drug. Metab. Dispos. 51, 657–671 (2023).
Lin, L., Dai, Y. & Xia, Y. An overview of aryl hydrocarbon receptor ligands in the last two decades (2002–2022): a medicinal chemistry perspective. Eur. J. Med. Chem. 244, 114845 (2022).
Hahn, M. E. & Karchner, S. I. in The AH Receptor in Biology and Toxicology (ed. Pohjanvirta, R.) 387–403 (Wiley, 2011).
Powell-Coffman, J. A., Bradfield, C. A. & Wood, W. B. Caenorhabditis elegans orthologs of the aryl hydrocarbon receptor and its heterodimerization partner the aryl hydrocarbon receptor nuclear translocator. Proc. Natl Acad. Sci. USA 95, 2844–2849 (1998).
Duncan, D. M., Burgess, E. A. & Duncan, I. Control of distal antennal identity and tarsal development in Drosophila by spineless-aristapedia, a homolog of the mammalian dioxin receptor. Genes Dev. 12, 1290–1303 (1998).
Moura-Alves, P. et al. AhR sensing of bacterial pigments regulates antibacterial defence. Nature 512, 387–392 (2014). This paper shows that the AHR senses bacterial pigments and modulates the immune responses against P. aeruginosa and M. tuberculosis, proposing the AHR as a new class of PRR.
Moura-Alves, P. et al. Host monitoring of quorum sensing during Pseudomonas aeruginosa infection. Science 366, eaaw1629 (2019). This paper shows that different P. aeruginosa quorum-sensing molecules expressed at different bacterial growth stages modulate the AHR (agonists and antagonists) and AHR-elicited responses, affecting host defence against infection.
Larigot, L. et al. Identification of modulators of the C. elegans aryl hydrocarbon receptor and characterization of transcriptomic and metabolic AhR-1 profiles. Antioxidants 11, 1030 (2022). This paper shows that bacterial ligands (P. aeruginosa phenazines) activate the AHR in invertebrates (C. elegans), in opposition to dioxin.
Schwartzkopf, C. M. et al. Tripartite interactions between filamentous Pf4 bacteriophage, Pseudomonas aeruginosa, and bacterivorous nematodes. PLoS Pathog. 19, e1010925 (2023). This paper shows that the AHR in C. elegans (invertebrate) responds to bacterial ligands, namely, PYO from P. aeruginosa, and modulates host defence against infection.
Fukunaga, B. N., Probst, M. R., Reisz-Porszasz, S. & Hankinson, O. Identification of functional domains of the aryl hydrocarbon receptor. J. Biol. Chem. 270, 29270–29278 (1995).
Stockinger, B., Di Meglio, P., Gialitakis, M. & Duarte, J. H. The aryl hydrocarbon receptor: multitasking in the immune system. Annu. Rev. Immunol. 32, 403–432 (2014). This article reviews AHR functions and molecular interactions in the immune system, focusing on barrier organs.
Rothhammer, V. & Quintana, F. J. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat. Rev. Immunol. 19, 184–197 (2019). This article reviews the AHR transcriptional programs in the immune system and their role in autoimmune and neurological diseases.
Pongratz, I., Antonsson, C., Whitelaw, M. L. & Poellinger, L. Role of the PAS domain in regulation of dimerization and DNA binding specificity of the dioxin receptor. Mol. Cell Biol. 18, 4079–4088 (1998).
Chapman-Smith, A., Lutwyche, J. K. & Whitelaw, M. L. Contribution of the Per/Arnt/Sim (PAS) domains to DNA binding by the basic helix-loop-helix PAS transcriptional regulators. J. Biol. Chem. 279, 5353–5362 (2004).
Janeway, C. A. Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb. Symp. Quant. Biol. 54, 1–13 (1989).
Li, D. & Wu, M. Pattern recognition receptors in health and diseases. Signal Transduct. Target. Ther. 6, 291 (2021).
Magiatis, P. et al. Malassezia yeasts produce a collection of exceptionally potent activators of the Ah (dioxin) receptor detected in diseased human skin. J. Invest. Dermatol. 133, 2023–2030 (2013). This paper demonstrates that the AHR senses and is activated by yeast-derived molecules.
Stange, E. L. et al. Staphylococcus aureus activates the aryl hydrocarbon receptor in human keratinocytes. J. Innate Immun. 14, 582–592 (2022).
Rademacher, F. et al. Staphylococcus epidermidis activates aryl hydrocarbon receptor signaling in human keratinocytes: implications for cutaneous defense. J. Innate Immun. 11, 125–135 (2019). This paper shows that S. epidermidis produces small molecules that modulate the AHR.
Engen, S. A., Rorvik, G. H., Schreurs, O., Blix, I. J. & Schenck, K. The oral commensal Streptococcus mitis activates the aryl hydrocarbon receptor in human oral epithelial cells. Int. J. Oral Sci. 9, 145–150 (2017).
Takamura, T. et al. Lactobacillus bulgaricus OLL1181 activates the aryl hydrocarbon receptor pathway and inhibits colitis. Immunol. Cell Biol. 89, 817–822 (2011).
Ozcam, M. et al. Gut symbionts Lactobacillus reuteri R2lc and 2010 encode a polyketide synthase cluster that activates the mammalian aryl hydrocarbon receptor. Appl. Environ. Microbiol. 85, e01661-18 (2019).
Dong, F. & Perdew, G. H. The aryl hydrocarbon receptor as a mediator of host–microbiota interplay. Gut Microbes 12, 1859812 (2020).
Han, H., Safe, S., Jayaraman, A. & Chapkin, R. S. Diet–host–microbiota interactions shape aryl hydrocarbon receptor ligand production to modulate intestinal homeostasis. Annu. Rev. Nutr. 41, 455–478 (2021).
Korecka, A. et al. Bidirectional communication between the aryl hydrocarbon receptor (AhR) and the microbiome tunes host metabolism. NPJ Biofilms Microbiomes 2, 16014 (2016).
van den Bogaard, E. H., Esser, C. & Perdew, G. H. The aryl hydrocarbon receptor at the forefront of host–microbe interactions in the skin: a perspective on current knowledge gaps and directions for future research and therapeutic applications. Exp. Dermatol. 30, 1477–1483 (2021).
Safe, S., Cheng, Y. & Jin, U. H. The aryl hydrocarbon receptor (AhR) as a drug target for cancer chemotherapy. Curr. Opin. Toxicol. 2, 24–29 (2017).
Wang, X. S., Cao, F., Zhang, Y. & Pan, H. F. Therapeutic potential of aryl hydrocarbon receptor in autoimmunity. Inflammopharmacology 28, 63–81 (2020).
Shi, J. et al. Aryl hydrocarbon receptor is a proviral host factor and a candidate pan-SARS-CoV-2 therapeutic target. Sci. Adv. 9, eadf0211 (2023).
Lebwohl, M. G. et al. Phase 3 trials of tapinarof cream for plaque psoriasis. N. Engl. J. Med. 385, 2219–2229 (2021).
Strober, B. et al. One-year safety and efficacy of tapinarof cream for the treatment of plaque psoriasis: results from the PSOARING 3 trial. J. Am. Acad. Dermatol. 87, 800–806 (2022).
Smith, S. H. et al. Tapinarof is a natural AhR agonist that resolves skin inflammation in mice and humans. J. Invest. Dermatol. 137, 2110–2119 (2017).
Furue, M., Hashimoto-Hachiya, A. & Tsuji, G. Aryl hydrocarbon receptor in atopic dermatitis and psoriasis. Int. J. Mol. Sci. 20, 5424 (2019).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04200963 (2024).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04999202 (2021).
Kober, C. et al. Targeting the aryl hydrocarbon receptor (AhR) with BAY 2416964: a selective small molecule inhibitor for cancer immunotherapy. J. Immunother. Cancer 11,e007495 (2023).
Opitz, C. A. et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197–203 (2011).
DiNatale, B. C. et al. Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling. Toxicol. Sci. 115, 89–97 (2010).
Rannug, A. et al. Certain photooxidized derivatives of tryptophan bind with very high affinity to the Ah receptor and are likely to be endogenous signal substances. J. Biol. Chem. 262, 15422–15427 (1987).
Phelan, D., Winter, G. M., Rogers, W. J., Lam, J. C. & Denison, M. S. Activation of the Ah receptor signal transduction pathway by bilirubin and biliverdin. Arch. Biochem. Biophys. 357, 155–163 (1998).
Sinal, C. J. & Bend, J. R. Aryl hydrocarbon receptor-dependent induction of cyp1a1 by bilirubin in mouse hepatoma hepa 1c1c7 cells. Mol. Pharmacol. 52, 590–599 (1997).
Schaldach, C. M., Riby, J. & Bjeldanes, L. F. Lipoxin A4: a new class of ligand for the Ah receptor. Biochemistry 38, 7594–7600 (1999).
Lamas, B., Natividad, J. M. & Sokol, H. Aryl hydrocarbon receptor and intestinal immunity. Mucosal Immunol. 11, 1024–1038 (2018).
Vyhlidalova, B. et al. Gut microbial catabolites of tryptophan are ligands and agonists of the aryl hydrocarbon receptor: a detailed characterization. Int. J. Mol. Sci. 21, 2614 (2020).
Kurata, K. et al. Skatole regulates intestinal epithelial cellular functions through activating aryl hydrocarbon receptors and p38. Biochem. Biophys. Res. Commun. 510, 649–655 (2019).
Muku, G. E., Murray, I. A., Espin, J. C. & Perdew, G. H. Urolithin a is a dietary microbiota-derived human aryl hydrocarbon receptor antagonist. Metabolites 8, 86 (2018).
Singh, R. et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 10, 89 (2019).
Fukumoto, S. et al. Identification of a probiotic bacteria-derived activator of the aryl hydrocarbon receptor that inhibits colitis. Immunol. Cell Biol. 92, 460–465 (2014).
Cervantes-Barragan, L. et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 357, 806–810 (2017).
Zelante, T. et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39, 372–385 (2013).
Bansal, T., Alaniz, R. C., Wood, T. K. & Jayaraman, A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc. Natl Acad. Sci. USA 107, 228–233 (2010).
Zhao, C. et al. Dietary tryptophan-mediated aryl hydrocarbon receptor activation by the gut microbiota alleviates Escherichia coli-induced endometritis in mice. Microbiol. Spectr. 10, e0081122 (2022).
O’Mahony, S. M., Clarke, G., Borre, Y. E., Dinan, T. G. & Cryan, J. F. Serotonin, tryptophan metabolism and the brain–gut–microbiome axis. Behav. Brain Res. 277, 32–48 (2015).
Rothhammer, V. et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat. Med. 22, 586–597 (2016).
Marinelli, L. et al. Identification of the novel role of butyrate as AhR ligand in human intestinal epithelial cells. Sci. Rep. 9, 643 (2019).
Modoux, M. et al. Butyrate acts through HDAC inhibition to enhance aryl hydrocarbon receptor activation by gut microbiota-derived ligands. Gut Microbes 14, 2105637 (2022).
Pinto, C. J. G., Avila-Galvez, M. A., Lian, Y., Moura-Alves, P. & Nunes Dos Santos, C. Targeting the aryl hydrocarbon receptor by gut phenolic metabolites: a strategy towards gut inflammation. Redox Biol. 61, 102622 (2023).
Beischlag, T. V., Luis Morales, J., Hollingshead, B. D. & Perdew, G. H. The aryl hydrocarbon receptor complex and the control of gene expression. Crit. Rev. Eukaryot. Gene Expr. 18, 207–250 (2008).
Garrison, P. M. et al. Species-specific recombinant cell lines as bioassay systems for the detection of 2,3,7,8-tetrachlorodibenzo-p-dioxin-like chemicals. Fundam. Appl. Toxicol. 30, 194–203 (1996).
Xu, H. et al. Generation of Tg(cyp1a:gfp) transgenic zebrafish for development of a convenient and sensitive in vivo assay for aryl hydrocarbon receptor activity. Mar. Biotechnol. 17, 831–840 (2015).
Diny, N. L. et al. The aryl hydrocarbon receptor contributes to tissue adaptation of intestinal eosinophils in mice. J. Exp. Med. 219, e20210970 (2022).
Schiering, C. et al. Feedback control of AHR signalling regulates intestinal immunity. Nature 542, 242–245 (2017).
Wincent, E. et al. Inhibition of cytochrome P4501-dependent clearance of the endogenous agonist FICZ as a mechanism for activation of the aryl hydrocarbon receptor. Proc. Natl Acad. Sci. USA 109, 4479–4484 (2012).
Avilla, M. N., Malecki, K. M. C., Hahn, M. E., Wilson, R. H. & Bradfield, C. A. The Ah receptor: adaptive metabolism, ligand diversity, and the xenokine model. Chem. Res. Toxicol. 33, 860–879 (2020).
Bradfield, C. A. & Bjeldanes, L. F. Dietary modification of xenobiotic metabolism — contribution of indolylic compounds present in Brassica oleracea. J. Agr. Food Chem. 35, 896–900 (1987).
Bjeldanes, L. F., Kim, J. Y., Grose, K. R., Bartholomew, J. C. & Bradfield, C. A. Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Proc. Natl Acad. Sci. USA 88, 9543–9547 (1991).
Boule, L. A., Burke, C. G., Jin, G. B. & Lawrence, B. P. Aryl hydrocarbon receptor signaling modulates antiviral immune responses: ligand metabolism rather than chemical source is the stronger predictor of outcome. Sci. Rep. 8, 1826 (2018). This paper provides an example of how exposure to different AHR ligands leads to variable outcomes of FluV infection.
Hinsdill, R. D., Couch, D. L. & Speirs, R. S. Immunosuppression in mice induced by dioxin (TCDD) in feed. J. Env. Pathol. Toxicol. 4, 401–425 (1980).
Luster, M. I. et al. Examination of bone marrow, immunologic parameters and host susceptibility following pre- and postnatal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Int. J. Immunopharmacol. 2, 301–310 (1980).
Ward, E. C. et al. Immunosuppression following 7,12-dimethylbenz[a]anthracene exposure in B6C3F1 mice. I. Effects on humoral immunity and host resistance. Toxicol. Appl. Pharmacol. 75, 299–308 (1984).
Sugita-Konishi, Y., Kobayashi, K., Naito, H., Miura, K. & Suzuki, Y. Effect of lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin on the susceptibility to Listeria infection. Biosci. Biotechnol. Biochem. 67, 89–93 (2003).
Vos, J. G., Kreeftenberg, J. G., Engel, H. W., Minderhoud, A. & Van Noorle Jansen, L. M. Studies on 2,3,7,8-tetrachlorodibenzo-p-dioxin induced immune suppression and decreased resistance to infection: endotoxin hypersensitivity, serum zinc concentrations and effect of thymosin treatment. Toxicology 9, 75–86 (1978).
Kimura, A. et al. Aryl hydrocarbon receptor protects against bacterial infection by promoting macrophage survival and reactive oxygen species production. Int. Immunol. 26, 209–220 (2014).
Zhu, R. et al. Involvement of aryl hydrocarbon receptor and aryl hydrocarbon receptor repressor in Helicobacter pylori-related gastric pathogenesis. J. Cancer 9, 2757–2764 (2018).
Huang, L., Qi, W., Zuo, Y., Alias, S. A. & Xu, W. 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. Dev. Comp. Immunol. 113, 103779 (2020).
Puyskens, A. et al. Aryl hydrocarbon receptor modulation by tuberculosis drugs impairs host defense and treatment outcomes. Cell Host Microbe 27, 238–248.e7 (2020). This paper bridges the role of the AHR in infection and therapeutic efficacy, showing that the AHR senses M. tuberculosis -derived molecules and antibiotics, modulating treatment outcomes.
Memari, B. et al. Engagement of the aryl hydrocarbon receptor in Mycobacterium tuberculosis-infected macrophages has pleiotropic effects on innate immune signaling. J. Immunol. 195, 4479–4491 (2015).
van Soest, J. J. et al. Comparison of static immersion and intravenous injection systems for exposure of zebrafish embryos to the natural pathogen Edwardsiella tarda. BMC Immunol. 12, 58 (2011).
Kiss, E. A. et al. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 334, 1561–1565 (2011).
Lee, J. S. et al. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat. Immunol. 13, 144–151 (2011). This paper and the publication by Kiss et al. were published simultaneously showing the important role of the AHR in the immune response against C. rodentium associated with the crosstalk between the AHR, immune homeostasis and diet.
Qiu, J. et al. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36, 92–104 (2012).
Li, S. et al. Aryl hydrocarbon receptor signaling cell intrinsically inhibits intestinal group 2 innate lymphoid cell function. Immunity 49, 915–928.e5 (2018).
Mayer, M. et al. Dysregulation of stress erythropoiesis and enhanced susceptibility to Salmonella Typhimurium infection in AhR-deficient mice. J. Infect. Dis. https://doi.org/10.1093/infdis/jiae304 (2024).
Fueldner, C. et al. Aryl hydrocarbon receptor activation by benzo[a]pyrene prevents development of septic shock and fatal outcome in a mouse model of systemic Salmonella enterica infection. Cells 11, 737 (2022).
Kim, S. H. et al. Novel compound 2-methyl-2H-pyrazole-3-carboxylic acid (2-methyl-4-o-tolylazo-phenyl)-amide (CH-223191) prevents 2,3,7,8-TCDD-induced toxicity by antagonizing the aryl hydrocarbon receptor. Mol. Pharmacol. 69, 1871–1878 (2006).
Shi, L. Z. et al. The aryl hydrocarbon receptor is required for optimal resistance to Listeria monocytogenes infection in mice. J. Immunol. 179, 6952–6962 (2007).
Zhang, H. et al. Sustained AhR activity programs memory fate of early effector CD8+ T cells. Proc. Natl Acad. Sci. USA 121, e2317658121 (2024).
Dean, J. W. et al. The aryl hydrocarbon receptor cell intrinsically promotes resident memory CD8+ T cell differentiation and function. Cell Rep. 42, 111963 (2023).
Landemaine, L. et al. Staphylococcus epidermidis isolates from atopic or healthy skin have opposite effect on skin cells: potential implication of the AHR pathway modulation. Front. Immunol. 14, 1098160 (2023).
Bessede, A. et al. Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 511, 184–190 (2014). This paper shows a protective role for the AHR against infection with S. Typhimurium and S. agalactiae and in endotoxic tolerance.
Vorderstrasse, B. A. & Lawrence, B. P. 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 (2006).
Wang, T., Wyrick, K. L., Pecka, M. R., Wills, T. B. & Vorderstrasse, B. A. Mechanistic exploration of AhR-mediated host protection against Streptococcus pneumoniae infection. Int. Immunopharmacol. 13, 490–498 (2012).
Gaitanis, G. et al. AhR ligands, malassezin, and indolo[3,2-b]carbazole are selectively produced by Malassezia furfur strains isolated from seborrheic dermatitis. J. Invest. Dermatol. 128, 1620–1625 (2008).
Gaitanis, G., Magiatis, P., Hantschke, M., Bassukas, I. D. & Velegraki, A. The Malassezia genus in skin and systemic diseases. Clin. Microbiol. Rev. 25, 106–141 (2012).
Vlachos, C., Schulte, B. M., Magiatis, P., Adema, G. J. & Gaitanis, G. Malassezia-derived indoles activate the aryl hydrocarbon receptor and inhibit Toll-like receptor-induced maturation in monocyte-derived dendritic cells. Br. J. Dermatol. 167, 496–505 (2012).
Zhang, L. et al. Expression and role of aryl hydrocarbon receptor in Aspergillus fumigatus keratitis. Int. J. Ophthalmol. 13, 199–205 (2020).
Zelante, T. et al. Aspergillus fumigatus tryptophan metabolic route differently affects host immunity. Cell Rep. 34, 108673 (2021).
Solis, N. V., Swidergall, M., Bruno, V. M., Gaffen, S. L. & Filler, S. G. The aryl hydrocarbon receptor governs epithelial cell invasion during oropharyngeal candidiasis. mBio 8, e00025-17 (2017).
De Luca, A. et al. IL-22 and IDO1 affect immunity and tolerance to murine and human vaginal candidiasis. PLoS Pathog. 9, e1003486 (2013).
Borghi, M. et al. Targeting the aryl hydrocarbon receptor with indole-3-aldehyde protects from vulvovaginal candidiasis via the IL-22–IL-18 cross-talk. Front. Immunol. 10, 2364 (2019).
de Araujo, E. F., Preite, N. W., Veldhoen, M., Loures, F. V. & Calich, V. L. G. Pulmonary paracoccidioidomycosis in AhR deficient hosts is severe and associated with defective Treg and Th22 responses. Sci. Rep. 10, 11312 (2020).
de Araujo, E. F. et al. The IDO–AhR axis controls Th17/Treg immunity in a pulmonary model of fungal infection. Front. Immunol. 8, 880 (2017).
Maradana, M. R. et al. Dietary environmental factors shape the immune defense against Cryptosporidium infection. Cell Host Microbe 31, 2038–2050.e4 (2023).
Munck, N. A., Roth, J., Sunderkotter, C. & Ehrchen, J. Aryl hydrocarbon receptor-signaling regulates early Leishmania major-induced cytokine expression. Front. Immunol. 10, 2442 (2019).
Climaco-Arvizu, S. et al. Aryl hydrocarbon receptor influences nitric oxide and arginine production and alters M1/M2 macrophage polarization. Life Sci. 155, 76–84 (2016).
Elizondo, G., Rodriguez-Sosa, M., Estrada-Muniz, E., Gonzalez, F. J. & Vega, L. Deletion of the aryl hydrocarbon receptor enhances the inflammatory response to Leishmania major infection. Int. J. Biol. Sci. 7, 1220–1229 (2011).
Lissner, M. M. et al. Metabolic profiling during malaria reveals the role of the aryl hydrocarbon receptor in regulating kidney injury. eLife 9, e60165 (2020).
Brant, F. et al. Role of the aryl hydrocarbon receptor in the immune response profile and development of pathology during Plasmodium berghei Anka infection. Infect. Immun. 82, 3127–3140 (2014).
Sanchez, Y. et al. The unexpected role for the aryl hydrocarbon receptor on susceptibility to experimental toxoplasmosis. J. Biomed. Biotechnol. 2010, 505694 (2010).
Barroso, A. et al. The aryl hydrocarbon receptor modulates production of cytokines and reactive oxygen species and development of myocarditis during Trypanosoma cruzi infection. Infect. Immun. 84, 3071–3082 (2016).
Ambrosio, L. F. et al. Role of aryl hydrocarbon receptor (AhR) in the regulation of immunity and immunopathology during Trypanosoma cruzi infection. Front. Immunol. 10, 631 (2019). This paper highlights the importance of the threshold of AHR activation for the modulation of T. cruzi infection outcomes.
Poland, A., Palen, D. & Glover, E. Analysis of the four alleles of the murine aryl hydrocarbon receptor. Mol. Pharmacol. 46, 915–921 (1994).
Ema, M. et al. Dioxin binding activities of polymorphic forms of mouse and human arylhydrocarbon receptors. J. Biol. Chem. 269, 27337–27343 (1994).
Kashuba, E. V. et al. Regulation of transactivation function of the aryl hydrocarbon receptor by the Epstein–Barr virus-encoded EBNA-3 protein. J. Biol. Chem. 281, 1215–1223 (2006).
House, R. V. et al. Examination of immune parameters and host resistance mechanisms in B6C3F1 mice following adult exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. J. Toxicol. Env. Health 31, 203–215 (1990).
Burleson, G. R. et al. Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on influenza virus host resistance in mice. Fundam. Appl. Toxicol. 29, 40–47 (1996).
Teske, S., Bohn, A. A., Regal, J. F., Neumiller, J. J. & Lawrence, B. P. Activation of the aryl hydrocarbon receptor increases pulmonary neutrophilia and diminishes host resistance to influenza A virus. Am. J. Physiol. Lung Cell Mol. Physiol. 289, L111–L124 (2005).
Neff-LaFord, H., Teske, S., Bushnell, T. P. & Lawrence, B. P. Aryl hydrocarbon receptor activation during influenza virus infection unveils a novel pathway of IFN-γ production by phagocytic cells. J. Immunol. 179, 247–255 (2007).
Wheeler, J. L., Martin, K. C. & Lawrence, B. P. Novel cellular targets of AhR underlie alterations in neutrophilic inflammation and inducible nitric oxide synthase expression during influenza virus infection. J. Immunol. 190, 659–668 (2013).
Jin, G. B., Winans, B., Martin, K. C. & Paige Lawrence, B. 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 (2014).
Jin, G. B., Moore, A. J., Head, J. L., Neumiller, J. J. & Lawrence, B. P. Aryl hydrocarbon receptor activation reduces dendritic cell function during influenza virus infection. Toxicol. Sci. 116, 514–522 (2010).
Lawrence, B. P., Roberts, A. D., Neumiller, J. J., Cundiff, J. A. & Woodland, D. L. Aryl hydrocarbon receptor activation impairs the priming but not the recall of influenza virus-specific CD8+ T cells in the lung. J. Immunol. 177, 5819–5828 (2006).
Major, J. et al. Endothelial AHR activity prevents lung barrier disruption in viral infection. Nature 621, 813–820 (2023). This paper shows that the outcome of AHR activity upon FluV infection is tissue specific.
Giovannoni, F. et al. AHR signaling is induced by infection with coronaviruses. Nat. Commun. 12, 5148 (2021). This paper shows that infection with different coronaviruses activates the AHR as a strategy to evade immunity.
Grunewald, M. E., Shaban, M. G., Mackin, S. R., Fehr, A. R. & Perlman, S. Murine coronavirus infection activates the aryl hydrocarbon receptor in an indoleamine 2,3-dioxygenase-independent manner, contributing to cytokine modulation and proviral TCDD-inducible-PARP expression. J. Virol. 94, e01743-19 (2020).
Tang, B. S. et al. Comparative host gene transcription by microarray analysis early after infection of the Huh7 cell line by severe acute respiratory syndrome coronavirus and human coronavirus 229E. J. Virol. 79, 6180–6193 (2005).
Almeida-da-Silva, C. L. C., Matshik Dakafay, H., Liu, K. & Ojcius, D. M. Cigarette smoke stimulates SARS-CoV-2 internalization by activating AhR and increasing ACE2 expression in human gingival epithelial cells. Int. J. Mol. Sci. 22, 7669 (2021).
Tanimoto, K. et al. Inhibiting SARS-CoV-2 infection in vitro by suppressing its receptor, angiotensin-converting enzyme 2, via aryl-hydrocarbon receptor signal. Sci. Rep. 11, 16629 (2021).
Liu, Y. et al. Mucus production stimulated by IFN–AhR signaling triggers hypoxia of COVID-19. Cell Res. 30, 1078–1087 (2020).
Zhou, Y. H. et al. Tryptophan metabolism activates aryl hydrocarbon receptor-mediated pathway to promote HIV-1 infection and reactivation. mBio 10, e02591-19 (2019). This paper shows that the AHR binds to the HIV-1 5′ long terminal repeat to activate viral transcription and to the Tat viral protein to facilitate the recruitment of transcription factors to viral promotors.
Ohata, H., Tetsuka, T., Hayashi, H., Onozaki, K. & Okamoto, T. 3-Methylcholanthrene activates human immunodeficiency virus type 1 replication via aryl hydrocarbon receptor. Microbiol. Immunol. 47, 363–370 (2003).
Fiorito, F., Santamaria, R., Irace, C., De Martino, L. & Iovane, G. 2,3,7,8-Tetrachlorodibenzo-p-dioxin and the viral infection. Env. Res. 153, 27–34 (2017).
Pokrovsky, A. G., Cherykh, A. I., Yastrebova, O. N. & Tsyrlov, I. B. 2,3,7,8-Tetrachlorodibenzo-p-dioxin as a possible activator of HIV infection. Biochem. Biophys. Res. Commun. 179, 46–51 (1991).
Gollapudi, S., Kim, C. H., Patel, A., Sindhu, R. & Gupta, S. Dioxin activates human immunodeficiency virus-1 expression in chronically infected promonocytic U1 cells by enhancing NF-κB activity and production of tumor necrosis factor-α. Biochem. Biophys. Res. Commun. 226, 889–894 (1996).
Tsyrlov, I. B. & Pokrovsky, A. Stimulatory effect of the CYP1A1 inducer 2,3,7,8-tetrachlorodibenzo-p-dioxin on the reproduction of HIV-1 in human lymphoid cell culture. Xenobiotica 23, 457–467 (1993).
Rao, P. S. & Kumar, S. Polycyclic aromatic hydrocarbons and cytochrome P450 in HIV pathogenesis. Front. Microbiol. 6, 550 (2015).
Kueck, T., Cassella, E., Holler, J., Kim, B. & Bieniasz, P. D. The aryl hydrocarbon receptor and interferon γ generate antiviral states via transcriptional repression. eLife 7, e38867 (2018).
Chatterjee, D. et al. Identification of aryl hydrocarbon receptor as a barrier to HIV-1 infection and outgrowth in CD4+ T cells. Cell Rep. 42, 112634 (2023).
Ohashi, H. et al. The aryl hydrocarbon receptor-cytochrome P450 1A1 pathway controls lipid accumulation and enhances the permissiveness for hepatitis C virus assembly. J. Biol. Chem. 293, 19559–19571 (2018).
Giovannoni, F. et al. AHR is a Zika virus host factor and a candidate target for antiviral therapy. Nat. Neurosci. 23, 939–951 (2020). This paper presents the AHR as a potential target for therapy against ZIKV infection.
Vota, D. et al. Zika virus infection of first trimester trophoblast cells affects cell migration, metabolism and immune homeostasis control. J. Cell Physiol. 236, 4913–4925 (2021).
Yamada, T. et al. Constitutive aryl hydrocarbon receptor signaling constrains type I interferon-mediated antiviral innate defense. Nat. Immunol. 17, 687–694 (2016). This paper shows that AHR signalling downregulates the IFN type I response upon infection with different viruses.
Inoue, H. et al. Aryl hydrocarbon receptor-mediated induction of EBV reactivation as a risk factor for Sjogren’s syndrome. J. Immunol. 188, 4654–4662 (2012).
Jiang, Y. et al. LMP2A suppresses the role of AHR pathway through ERK signal pathway in EBV-associated gastric cancer. Virus Res. 297, 198399 (2021).
Chen, J. et al. Targeting aryl hydrocarbon receptor signaling enhances tpe I interferon-independent resistance to herpes simplex virus. Microbiol. Spectr. 9, e0047321 (2021).
Veiga-Parga, T., Suryawanshi, A. & Rouse, B. T. Controlling viral immuno-inflammatory lesions by modulating aryl hydrocarbon receptor signaling. PLoS Pathog. 7, e1002427 (2011).
McNab, F., Mayer-Barber, K., Sher, A., Wack, A. & O’Garra, A. Type I interferons in infectious disease. Nat. Rev. Immunol. 15, 87–103 (2015).
Paris, A., Tardif, N., Galibert, M. D. & Corre, S. AhR and cancer: from gene profiling to targeted therapy. Int. J. Mol. Sci. 22, 752 (2021).
Safe, S., Lee, S. O. & Jin, U. H. Role of the aryl hydrocarbon receptor in carcinogenesis and potential as a drug target. Toxicol. Sci. 135, 1–16 (2013).
Murray, I. A., Patterson, A. D. & Perdew, G. H. Aryl hydrocarbon receptor ligands in cancer: friend and foe. Nat. Rev. Cancer 14, 801–814 (2014).
Hawerkamp, H. C. et al. Vemurafenib acts as an aryl hydrocarbon receptor antagonist: implications for inflammatory cutaneous adverse events. Allergy 74, 2437–2448 (2019).
Corre, S. et al. Sustained activation of the aryl hydrocarbon receptor transcription factor promotes resistance to BRAF-inhibitors in melanoma. Nat. Commun. 9, 4775 (2018).
Stinn, A., Furkert, J., Kaufmann, S. H. E., Moura-Alves, P. & Kolbe, M. Novel method for quantifying AhR-ligand binding affinities using microscale thermophoresis. Biosensors 11, 60 (2021).
Cheng, Y. et al. Editor’s highlight: microbial-derived 1,4-dihydroxy-2-naphthoic acid and related compounds as aryl hydrocarbon receptor agonists/antagonists: structure–activity relationships and receptor modeling. Toxicol. Sci. 155, 458–473 (2017).
Song, J. et al. A ligand for the aryl hydrocarbon receptor isolated from lung. Proc. Natl Acad. Sci. USA 99, 14694–14699 (2002).
Hubbard, T. D. et al. Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Sci. Rep. 5, 12689 (2015).
Lowe, M. M. et al. Identification of cinnabarinic acid as a novel endogenous aryl hydrocarbon receptor ligand that drives IL-22 production. PLoS ONE 9, e87877 (2014).
Miller, C. A. 3rd. Expression of the human aryl hydrocarbon receptor complex in yeast. Activation of transcription by indole compounds. J. Biol. Chem. 272, 32824–32829 (1997).
Jin, U. H. et al. Microbiome-derived tryptophan metabolites and their aryl hydrocarbon receptor-dependent agonist and antagonist activities. Mol. Pharmacol. 85, 777–788 (2014).
Schroeder, J. C. et al. The uremic toxin 3-indoxyl sulfate is a potent endogenous agonist for the human aryl hydrocarbon receptor. Biochemistry 49, 393–400 (2010).
Michaudel, C. et al. Rewiring the altered tryptophan metabolism as a novel therapeutic strategy in inflammatory bowel diseases. Gut 72, 1296–1307 (2022).
Mezrich, J. D. et al. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J. Immunol. 185, 3190–3198 (2010).
Tian, W. Z. et al. PE (0:0/14:0), an endogenous metabolite of the gut microbiota, exerts protective effects against sepsis-induced intestinal injury by modulating the AHR/CYP1A1 pathway. Clin. Sci. 137, 1753–1769 (2023).
Mexia, N. et al. Pityriazepin and other potent AhR ligands isolated from Malassezia furfur yeast. Arch. Biochem. Biophys. 571, 16–20 (2015).
Seidel, S. D. et al. Activation of the Ah receptor signaling pathway by prostaglandins. J. Biochem. Mol. Toxicol. 15, 187–196 (2001).
Flegel, J. et al. The highly potent AhR agonist picoberin modulates Hh-dependent osteoblast differentiation. J. Med. Chem. 65, 16268–16289 (2022).
Ramadoss, P. & Perdew, G. H. Use of 2-azido-3-[125I]iodo-7,8-dibromodibenzo-p-dioxin as a probe to determine the relative ligand affinity of human versus mouse aryl hydrocarbon receptor in cultured cells. Mol. Pharmacol. 66, 129–136 (2004).
Chae, K., Albro, P. W., Luster, M. I. & McKinney, J. D. A screening assay for the tetrachlorodibenzo-p-dioxin receptor using the [125I]iodovaleramide derivative of trichlorodibenzo-p-dioxin as the binding ligand. Int. J. Env. Anal. Chem. 17, 267–274 (1984).
Perdew, G. H. Association of the Ah receptor with the 90-kDa heat shock protein. J. Biol. Chem. 263, 13802–13805 (1988).
Meyer, B. K., Pray-Grant, M. G., Vanden Heuvel, J. P. & Perdew, G. H. Hepatitis B virus X-associated protein 2 is a subunit of the unliganded aryl hydrocarbon receptor core complex and exhibits transcriptional enhancer activity. Mol. Cell Biol. 18, 978–988 (1998).
Kazlauskas, A., Poellinger, L. & Pongratz, I. Evidence that the co-chaperone p23 regulates ligand responsiveness of the dioxin (aryl hydrocarbon) receptor. J. Biol. Chem. 274, 13519–13524 (1999).
Enan, E. & Matsumura, F. Identification of c-Src as the integral component of the cytosolic Ah receptor complex, transducing the signal of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) through the protein phosphorylation pathway. Biochem. Pharmacol. 52, 1599–1612 (1996).
Reyes, H., Reisz-Porszasz, S. & Hankinson, O. Identification of the Ah receptor nuclear translocator protein (Arnt) as a component of the DNA binding form of the Ah receptor. Science 256, 1193–1195 (1992).
Denison, M. S., Fisher, J. M. & Whitlock, J. P. Jr. The DNA recognition site for the dioxin–Ah receptor complex. Nucleotide sequence and functional analysis. J. Biol. Chem. 263, 17221–17224 (1988).
Rijo, M. P., Diani-Moore, S., Yang, C., Zhou, P. & Rifkind, A. B. Roles of the ubiquitin ligase CUL4B and ADP-ribosyltransferase TiPARP in TCDD-induced nuclear export and proteasomal degradation of the transcription factor AHR. J. Biol. Chem. 297, 100886 (2021).
Ma, Q., Baldwin, K. T., Renzelli, A. J., McDaniel, A. & Dong, L. TCDD-inducible poly(ADP-ribose) polymerase: a novel response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Biochem. Biophys. Res. Commun. 289, 499–506 (2001).
Denison, M. S., Soshilov, A. A., He, G., DeGroot, D. E. & Zhao, B. 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 (2011).
Wright, E. J., De Castro, K. P., Joshi, A. D. & Elferink, C. J. Canonical and non-canonical aryl hydrocarbon receptor signaling pathways. Curr. Opin. Toxicol. 2, 87–92 (2017).
Ohtake, F. et al. Dioxin receptor is a ligand-dependent E3 ubiquitin ligase. Nature 446, 562–566 (2007).
Jia, Y. et al. Tetrandrine enhances the ubiquitination and degradation of Syk through an AhR–c-src–c-Cbl pathway and consequently inhibits osteoclastogenesis and bone destruction in arthritis. Cell Death Dis. 10, 38 (2019).
Zhu, J. et al. Aryl hydrocarbon receptor promotes IL-10 expression in inflammatory macrophages through Src–STAT3 signaling pathway. Front. Immunol. 9, 2033 (2018).
Acknowledgements
The authors thank their colleagues in the field whose work inspired this Review. This work was funded by the European Union’s Horizon 2020 research and innovation programme under grant agreement number 951921. Y.L. was supported by the Ludwig Institute for Cancer Research Core Award.
Author information
Authors and Affiliations
Contributions
The authors contributed to all aspects of the article.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Immunology thanks Marco Colonna and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Glossary
- AHR ligands
-
Molecules with anthropomorphic (for example, pollutants) and natural (for example, diet, host or microbiota metabolism, and microorganism derived) origin where aryl hydrocarbon receptor (AHR) binding has been confirmed.
- AHR modulators
-
Molecules that directly or indirectly activate or inhibit the aryl hydrocarbon receptor (AHR), and where binding has not been shown.
- Competitive ligand binding
-
Measurement of competition and displacement of radiolabelled known aryl hydrocarbon receptor (AHR) ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or the photoaffinity ligand 2-azido-3-[125I]iodo-7,8-dibromodibenzo-p-dioxin (125IBr2N3DpD) from the AHR.
- Danger-associated molecular patterns
-
(DAMPs). Endogenous molecules released from damaged or dying cells, such as high-mobility group box 1 and heat shock proteins, capable of eliciting an innate immune response upon recognition by a pattern recognition receptor (PRR).
- Direct ligand binding
-
Measurement of direct ligand binding, such as microscale thermophoresis, intrinsic fluorescence quenching assays or differential scanning fluorometry, to the purified aryl hydrocarbon receptor (AHR) or AHR complex (for example, with chaperones or the AHR nuclear translocator (ARNT)).
- Microorganism-associated molecular patterns
-
(MAMPs). Conserved microbial components such as lipids, polysaccharides and nucleic acids that are expressed/released by microorganisms, which are capable of eliciting an innate immune response upon recognition by a pattern recognition receptor (PRR).
- PAS
-
A protein domain named after the three proteins in which it was first discovered: period circadian protein (PER), aryl hydrocarbon receptor nuclear translocator (ARNT) and single-minded protein (SIM). The PAS domain is found in bacteria and eukaryotes and acts as a molecular sensor.
- Pattern recognition receptors
-
(PRRs). Germline-encoded receptors present in the cellular cytoplasm or membrane bound that are able to sense different molecules in health and pathological contexts, including microbial derived or components of host cells. PRRs are divided according to their localization, function, structural organization or ligand specificity.
- Quorum-sensing
-
Process of cell–cell communication between bacteria (both intraspecies and/or interspecies) through the production and detection of molecules that induce a cell density-dependent response.
- Xenobiotic
-
A chemical substance that is found in an organism but that is extrinsic to that organism and not naturally produced by it, including pharmaceutical drugs, pesticides, environmental pollutants and food additives, among others.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Barreira-Silva, P., Lian, Y., Kaufmann, S.H.E. et al. The role of the AHR in host–pathogen interactions. Nat Rev Immunol (2024). https://doi.org/10.1038/s41577-024-01088-4
Accepted:
Published:
DOI: https://doi.org/10.1038/s41577-024-01088-4