Print
Aryl hydrocarbon receptors as potential therapeutic targets
expand article infoPavlina Andreeva-Gateva, Dimitar Bakalov, Zafer Sabit, Radka Tafradjiiska-Hadjiolova
‡ Medical University of Sofia, Sofia, Bulgaria
Open Access

Abstract

Aryl hydrocarbon receptors (AhR) are regulators of the expression of cytochrome P-450 isoforms, mediating a wide variety of the effects of substances from the endogenous or exogenous origin, including those produced from the microbiome. An exciting new aspect of their activity is their localization in the brain and their potential to modulate the action of the immune system. AhR is emerging as an essential toxicological and therapeutic target for neuromodulation. Further studies are needed for elucidating their utility as drug-targets.

Keywords

dioxin, indoles, toxicology, neuroprotection

Introduction

Aryl hydrocarbon receptors (AhR) are ligand-activated receptors. They form nuclear heterodimer complexes with AhR-dependent nuclear translocator protein, and this complex binds to cis-xenobiotic responsive elements in the promoter region of AhR-responsive genes (Denison et al. 2011). These receptors were initially identified as having a high binding affinity for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin), which is a highly toxic industrial toxin (Poland et al. 1976). Subsequent studies in the 1980s identified various substances used in the industry (Denison et al. 1998) and pharmacology as ligands of these receptors, such as: carbidopa (Safe 2017), omeprazole (Jin 2015), endogenous substances indole derivatives (Hubbard 2015), constituents of certain fruits and vegetables (Hooper et al. 2011) and microbiome metabolism products (Korecka et al. 2016) that have a protective effect on the gastrointestinal tract.

Many of those ligands have a much lower binding affinity for AhR than TCDD and structure-like toxic halogenated aromatic substances (Murray and Perdew 2017). Among the many xenobiotic ligands for AhR, polycyclic aromatic hydrocarbons have been the most widely studied (Mulero-Navaro and Fernandez-Salguero 2016). Many of the AhR ligands are also estrogen receptor ligands (Abdelrahim et al. 2006). AhR regulates the expression of CYP. Conjugating enzymes not only in the liver but also in the brain. Studies have demonstrated the induction of CYP1A1 by TCDD in rat brain astrocytes (Sakakibara et al. 2016), suggesting the involvement of AhR in the metabolism of xenobiotics.

After being identified as mediators of the cellular response to xenobiotics, epidemiological studies in humans were conducted. The attention was focused on the pathological conditions of the immune system, lipid metabolism, epithelial integrity, porphyria manifestations, thymus involution, and neoplasms. The involvement of AhR inspired scientific researches (Flesch-Janys et al. 1995). The creation of transgenic mouse models subsequently helps to establish the role of AhR in critical physiological and homeostatic processes. AhR-deficient mice show abnormalities in the liver, hematopoietic, cardiovascular, and immune system development (Lahvis et al. 2000). The physiological significance of AhR is further supported by the fact that they are evolutionarily conserved and exist in all multicellular animals. In some lower-order invertebrates, e.g., D. melanogaster, AhR does not have a detoxifying function but is required for the development of eyes, feet, and wings (Cespedes et al. 2010). In C. elegans, AhR is essential for neuronal differentiation and migration (Quin and Powell-Coffman 2004). According to modern understanding, the xenobiotic-dependent functions of AhR represent an adaptive mechanism that overlaps its physiologically determined features (Mulero-Navarro and Fernandez-Salguero 2016). Epigenetic mechanisms are involved both in the expression of AhR (Mulero-Navarro et al. 2006) and in regard of genes that are regulated by AhR. Exposure of the maternal organism to the action of AhR agonists is considered to be one of the possible mechanisms for the development of breast cancer in the offspring through epigenetic mechanisms (Romagnolo et al. 2016).

Table 1.

Examples of Ah ligands.

Chemical structure and name Source References Chemical structure and name Source References
microbial Hubbard (2015) endogenous Novikov et al. (2016)
Indole Kynurenine
microbial Hubbard (2015) endogenous Novikov et al. (2016)
Skatole Kynurenic acid
microbial Hubbard (2015) endogenous Novikov et al. (2016)
3-hydroxy indole Xanthurenic acid
microbial endogenous Zelante et al. (2013) endogenous Lowe et al. (2014)
Indole-3-acetaldehyde Cinnabarinic acid
microbial endogenous Jin et al. (2014) endogenous Wincent et al. (2009)
Indole-3-acetic acid FICZ
microbial Sugihara et al. (2004) endogenous Shertzer and Senft (2000)
Indigo Indolo(3,2-b) carbazole
microbial Flaveny et al. (2009) dietary Jin et al. (2014)
indirubin Tryptophan
microbial Murray and Perdew (2017) dietary Jin et al. (2014)
Truptanthrin Flavonoids

AhR as a therapeutic target

The identification of AhR ligands and their well-described positive health effect and beneficial pharmaceutical properties has stimulated studies aimed at developing drugs for various tumors, immune and inflammatory diseases, and enhancers of hematopoietic stem cell production. In the development of drugs that target AhR, the aim is mainly directed to selective AhR modulators, in which the ligand exhibits tissue-specific agonist or antagonist activity (Jin et al. 2012). Different classes of AhR ligands and different molecular types in the same class can differentially modulate AhR activity, inducing the expression of various genes. For this reason, AhRs can be considered as potentially interesting drug targets with cell-specific regulation.

Since AhRs are widely expressed in a number of tumors, molecules with antagonistic activity against AhRs could be considered as potential candidates for the treatment of such diseases. The most well-known AhR antagonist is alpha-naphtoflavone (Gasievicz and Rucci 1991). The potent AhR antagonist StemRegenin 1 has recently been developed as an inducer of human hematopoietic stem cell proliferation in vitro (Boitano et al. 2010). Interesting effects of bilirubin, as a potential AhR ligand on the immune system, have been reported (Bock and Kohle 2010) – in bilirubin-treated mice, it suppresses the development of the autoimmune disease. After endogenous bilirubin depletion, there is an increased incidence of exacerbation of autoimmune disease (Liu et al. 2008).

Throughout the many plant nutrients and chemicals of plant origin in the human diet, flavonoids are the most abundant and ubiquitous in fruits, vegetables, and wine. Quercetin, apigenin, and campherol, which are included in some foods, such as rose hips, linden flowers, honey, grapes, have been shown to exert agonist / antagonistic effects on AhR in various tissues (Hooper 2011). In addition, many flavonoids have anti-allergic and anti-inflammatory effects. Resveratrol (Papoutsis et al. 2010) has been found to inhibit CYP1A1 transcription in vitro, preventing AhR activation. Indole-glucosindolates in cruciferous vegetables is metabolized to compounds with high affinity for AhR. One of these metabolites, indole-3-carbinol, has been successfully tested in clinical trials as a dietary supplement (Reed et al. 2005). Probiotic bacteria and yeasts related to the human gut and skin microbiome also produce AhR ligands (e.g., indole-3 aldehyde, indirubin), thus enhancing the body’s barrier functions (Zelante et al. 2013).

Neuroprotective properties of AhR modulators

AhR expression in vertebrate brain has recently been demonstrated by immunohistochemistry, with the brain stem, pineal gland, and some hypothalamic nuclei (including the suprachiasmatic nucleus controlling the circadian rhythm) having significantly elevated AhR levels compared to other areas of the brain (Juricek and Coumoul 2018). AhR regulates neurogenesis, cell proliferation, differentiation, and migration (Imran et al. 2015). The neuroprotective potential of 3,3’-diindolylmethane, a selective AhR modulator, has recently been demonstrated in cellular and animal models of Parkinson’s disease, in lipopolysaccharide-induced inflammation and neuronal hypoxia (Rzemieniec et al. 2016). Activation of apoptotic signals by AhR ligands, on the other hand, can lead to NMDA (N-Methyl-d-Aspartate) receptor-mediated excitotoxicity, increased levels of calcium in the cytoplasm and oxidative stress (Wan et al. 2015). Interestingly, NMDA receptors also modulate the AhR function (Lin et al. 2008). In mouse stroke models, the kynurenine-aryl hydrocarbon receptor pathway is an essential mediator of brain neuronal damage (Cuartero et al. 2014) and represents a potential therapeutic modulation opportunity.

Conclusion

All these data put the importance of AhR as a toxicological and pharmacological target. Further evaluation of the neuropharmacological potential of substances that bind and modulate AhR is needed.

Acknowledgements

With the support of the Council of Medical Sciences of the Medical University of Sofia, grant D-62/2019.

References

  • Abdelrahim M, Ariazi E, Kim K, Khan S, Barhoumi R, Burghardt R, Liu S, Hill D, Finnell R, Wlodarczyk B, Jordan VC, Safe S (2006) 3-Methylcholanthrene and Other Aryl Hydrocarbon Receptor Agonists Directly Activate Estrogen Receptor α. Cancer Research 66(4): 2459–2467. https://doi.org/10.1158/0008-5472.CAN-05-3132
  • Bock KW, Köhle C (2010) Contributions of the Ah receptor to bilirubin homeostasis and its antioxidative and atheroprotective functions. Biological Chemistry 391(6): 645–653. https://doi.org/10.1515/bc.2010.065
  • Boitano AE, Wang J, Romeo R, Bouchez LC, Parker AE, Sutton SE, Walker JR, Flaveny CA, Perdew GH, Denison MS, Schultz PG, Cooke MP (2010) Aryl Hydrocarbon Receptor Antagonists Promote the Expansion of Human Hematopoietic Stem Cells. Science 329(5997): 1345–1348. https://doi.org/10.1126/science.1191536.
  • Céspedes MA, Galindo MI, Couso JP (2010) Dioxin Toxicity In Vivo Results from an Increase in the Dioxin-Independent Transcriptional Activity of the Aryl Hydrocarbon Receptor. PLoS ONE 5(11): e15382. https://doi.org/10.1371/journal.pone.0015382
  • Cuartero MI, Ballesteros I, de la Parra J, Harkin AL, Abautret-Daly A, Sherwin E, Fernández-Salguero P, Corbí ÁL, Lizasoain I, Moro MA (2014) L-Kynurenine/Aryl Hydrocarbon Receptor Pathway Mediates Brain Damage After Experimental Stroke. Circulation 130(23): 2040–2051. https://doi.org/10.1161/CIRCULATIONAHA.114.011394
  • Denison MS, Heath-Pagliuso S (1998) The Ah Receptor: A Regulator of the Biochemical and Toxicological Actions of Structurally Diverse Chemicals. Bulletin of Environmental Contamination and Toxicology 61(5): 557–568. https://doi.org/10.1007/PL00002973
  • Denison MS, Soshilov AA, He G, DeGroot DE, Zhao B (2011) Exactly the Same but Different: Promiscuity and Diversity in the Molecular Mechanisms of Action of the Aryl Hydrocarbon (Dioxin) Receptor. Toxicological Sciences 124(1): 1–22. https://doi.org/10.1093/toxsci/kfr218
  • Flesch-Janys D, Berger J, Gum P, Manz A, Nagel S, Waltsgott H, Dwyer JH (1995) Exposure to Polychlorinated Dioxins and Furans (PCDD/F) and Mortality in a Cohort of Workers from a Herbicide-producing Plant in Hamburg, Federal Republic of Germany. American Journal of Epidemiology 142(11): 1165–1175. https://doi.org/10.1093/oxfordjournals.aje.a117575
  • Gasiewicz TA, Rucci G (1991) Alpha-naphthoflavone acts as an antagonist of 2,3,7,8-tetrachlorodibenzo-p-dioxin by forming an inactive complex with the Ah receptor. Molecular Pharmacology 40(5): 607–612.
  • Hubbard TD, Murray IA, Perdew GH (2015) Indole and Tryptophan Metabolism: Endogenous and Dietary Routes to Ah Receptor Activation. Drug Metabolism and Disposition 43(10): 1522–1535. https://doi.org/10.1124/dmd.115.064246
  • Jin U-H, Kim S-B, Safe S (2015) Omeprazole Inhibits Pancreatic Cancer Cell Invasion through a Nongenomic Aryl Hydrocarbon Receptor Pathway. Chemical Research in Toxicology 28(5): 907–918. https://doi.org/10.1021/tx5005198
  • Jin U-H, Lee S, Safe S (2012) Aryl Hydrocarbon Receptor (AHR)-Active Pharmaceuticals Are Selective AHR Modulators in MDA-MB-468 and BT474 Breast Cancer Cells. Journal of Pharmacology and Experimental Therapeutics 343(2): 333–341. https://doi.org/10.1124/jpet.112.195339
  • Jin U-H, Lee S-O, Sridharan G, Lee K, Davidson LA, Jayaraman A, Chapkin RS, Alaniz R, Safe S (2014) Microbiome-Derived Tryptophan Metabolites and Their Aryl Hydrocarbon Receptor-Dependent Agonist and Antagonist Activities. Molecular Pharmacology 85(5): 777–788. https://doi.org/10.1124/mol.113.091165
  • Korecka A, Dona A, Lahiri S, Tett AJ, Al-Asmakh M, Braniste V, D’Arienzo R, Abbaspour A, Reichardt N, Fujii-Kuriyama Y, Rafter J, Narbad A, Holmes E, Nicholson J, Arulampalam V, Pettersson S (2016) Bidirectional communication between the Aryl hydrocarbon Receptor (AhR) and the microbiome tunes host metabolism. npj Biofilms and Microbiomes 2: e16014. https://doi.org/10.1038/npjbiofilms.2016.14
  • Lahvis GP, Lindell SL, Thomas RS, McCuskey RS, Murphy C, Glover E, Bentz M, Southard J, Bradfield CA (2000) Portosystemic shunting and persistent fetal vascular structures in aryl hydrocarbon receptor-deficient mice. Proceedings of the National Academy of Sciences of the United States of America 97(19): 10442–10447. https://doi.org/10.1073/pnas.190256997
  • Lin C-H, Juan S-H, Wang CY, Sun Y-Y, Chou C-M, Chang S-F, Hu S-Y, Lee W-S, Lee Y-H (2008) Neuronal activity enhances aryl hydrocarbon receptor-mediated gene expression and dioxin neurotoxicity in cortical neurons. Journal of Neurochemistry 104(5): 1415–1429. https://doi.org/10.1111/j.1471-4159.2007.05098.x
  • Liu Y, Li P, Lu J, Xiong W, Oger J, Tetzlaff W, Cynader M (2008) Bilirubin Possesses Powerful Immunomodulatory Activity and Suppresses Experimental Autoimmune Encephalomyelitis. The Journal of Immunology 181(3): 1887–1897. https://doi.org/10.4049/jimmunol.181.3.1887
  • Lowe MM, Mold JE, Kanwar B, Huang Y, Louie A, Pollastri MP, Wang C, Patel G, Franks DG, Schlezinger J, Sherr DH, Silverstone AE, Hahn ME, McCune JM (2014) Identification of Cinnabarinic Acid as a Novel Endogenous Aryl Hydrocarbon Receptor Ligand That Drives IL-22 Production. PLoS ONE 9: e87877. https://doi.org/10.1371/journal.pone.0087877
  • Novikov O, Wang Z, Stanford EA, Parks AJ, Ramirez-Cardenas A, Landesman E, Laklouk I, Sarita-Reyes C, Gusenleitner D, Li A, Monti S, Manteiga S, Lee K, Sherr DH (2016) An Aryl Hydrocarbon Receptor-Mediated Amplification Loop That Enforces Cell Migration in ER − /PR − /Her2 − Human Breast Cancer Cells. Molecular Pharmacology 90(5): 674–688. https://doi.org/10.1124/mol.116.105361
  • Papoutsis AJ, Lamore SD, Wondrak GT, Selmin OI, Romagnolo DF (2010) Resveratrol Prevents Epigenetic Silencing of BRCA-1 by the Aromatic Hydrocarbon Receptor in Human Breast Cancer Cells. The Journal of Nutrition 140(9): 1607–1614. https://doi.org/10.3945/jn.110.123422
  • Poland A, Glover E, Kende AS (1976) 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. The Journal of Biological Chemistry 251(16): 4936–4946.
  • Romagnolo DF, Daniels KD, Grunwald JT, Ramos SA, Propper CR, Selmin OI (2016) Epigenetics of breast cancer: Modifying role of environmental and bioactive food compounds. Molecular Nutrition & Food Research 60(6): 1310–1329. https://doi.org/10.1002/mnfr.201501063
  • Shertzer HG, Senft AP (2000) The Micronutrient Indole-3-Carbinol: Implications for Disease and Chemoprevention. Drug Metabolism and Personalized Therapy 17(1–4): 159–188. https://doi.org/10.1515/DMDI.2000.17.1-4.159
  • Sugihara K, Kitamura S, Yamada T, Okayama T, Ohta S, Yamashita K, Yasuda M, Fujii-Kuriyama Y, Saeki K, Matsui S, Matsuda T (2004) Aryl hydrocarbon receptor-mediated induction of microsomal drug-metabolizing enzyme activity by indirubin and indigo. Biochemical and Biophysical Research Communications 318(2): 571–578. https://doi.org/10.1016/j.bbrc.2004.04.066
  • Wan C, Zhang Y, Jiang J, Jiang S, Nie X, Li A, Guo A, Wu Q (2015) Critical Role of TAK1-Dependent Nuclear Factor-κB Signaling in 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced Astrocyte Activation and Subsequent Neuronal Death. Neurochemical Research 40: 1220–1231. https://doi.org/10.1007/s11064-015-1585-2
  • Wincent E, Amini N, Luecke S, Glatt H, Bergman J, Crescenzi C, Rannug A, Rannug U (2009) The Suggested Physiologic Aryl Hydrocarbon Receptor Activator and Cytochrome P4501 Substrate 6-Formylindolo[3,2- b] carbazole Is Present in Humans. Journal of Biological Chemistry 284: 2690–2696. https://doi.org/10.1074/jbc.M808321200
  • 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 (2013) Tryptophan Catabolites from Microbiota Engage Aryl Hydrocarbon Receptor and Balance Mucosal Reactivity via Interleukin-22. Immunity 39(2): 372–385. https://doi.org/10.1016/j.immuni.2013.08.003
login to comment