Skip to main content
ACS AuthorChoice logoLink to ACS AuthorChoice
. 2020 Apr 7;33(4):860–879. doi: 10.1021/acs.chemrestox.9b00476

The Ah Receptor: Adaptive Metabolism, Ligand Diversity, and the Xenokine Model

Mele N Avilla , Kristen M C Malecki †,‡, Mark E Hahn §, Rachel H Wilson , Christopher A Bradfield †,∥,*
PMCID: PMC7175458  PMID: 32259433

Abstract

graphic file with name tx9b00476_0007.jpg

The Ah receptor (AHR) has been studied for almost five decades. Yet, we still have many important questions about its role in normal physiology and development. Moreover, we still do not fully understand how this protein mediates the adverse effects of a variety of environmental pollutants, such as the polycyclic aromatic hydrocarbons (PAHs), the chlorinated dibenzo-p-dioxins (“dioxins”), and many polyhalogenated biphenyls. To provide a platform for future research, we provide the historical underpinnings of our current state of knowledge about AHR signal transduction, identify a few areas of needed research, and then develop concepts such as adaptive metabolism, ligand structural diversity, and the importance of proligands in receptor activation. We finish with a discussion of the cognate physiological role of the AHR, our perspective on why this receptor is so highly conserved, and how we might think about its cognate ligands in the future.

Introduction

The role of the Ah receptor (AHR) in human health and environmental toxicology continues to be an area of considerable interest. In this review, we provide a brief history of AHR research, our interpretation of recent discoveries, and our vision for the research path forward. Owing to the thousands of publications on this topic, we have attempted to provide our own perspective on the history of AHR discovery, current state of knowledge, and opportunities for further inquiry, rather than perform a comprehensive review. This approach was taken in an effort to provide a foundation for future research and present ideas designed to stimulate new scientific directions. In an effort toward clarity, we try to emphasize reviews and examples and have not attempted to generate an exhaustive review of the primary literature. Our rationale was that these citations will represent the complicated literature, alternative interpretations of the relevant science, and can serve as primary citations when further reading is of interest.

The Ah Receptor

Historical Foundations

Polycyclic Aromatic Hydrocarbons and Discovery of the Ah Locus

Early indications for the existence of the AHR arose from studies designed to understand the metabolism of carcinogenic polycyclic aromatic hydrocarbons (PAHs), such as benzo[a]pyrene (BAP), 7,12-dimethylbenzanthracene (DMBA), and 3-methylcholanthrene (3MC) (Figure 1).1,2 Compounds like these arise from combustion processes and are common contaminants in chimney soot, charbroiled foods, diesel exhaust, outdoor burns, cigarette smoke, and coal tar.38

Figure 1.

Figure 1

Investigations of carcinogenic PAHs led to discovery of the Ah locus. Studies using BAP, DMBA, and 3-MC provided early evidence for the existence of the AHR (see text for references).

In early carcinogen metabolism studies, the prominent enzymatic activity that oxidized PAHs to hydroxylated metabolites became known as “aryl hydrocarbon hydroxylase” or simply “AHH.” From this work, three important observations arose.9,10 First, AHH activity was the product of multiple cytochrome P450-dependent monooxygenases (P450s).11 Second, AHH activity was significantly upregulated, or “induced”, by prior exposure to a broad spectrum of these same PAHs.12 Third, in mice, a single autosomal locus harbors significant control over induction sensitivity across inbred strains (i.e., some strains were more inducible than others).13 The locus became commonly known as Ah for its role in regulating AHH activity and was formally renamed the “aryl hydrocarbon receptor” or Ahr locus in later years.14,15 In addition to its importance as an early example of carcinogen metabolism regulation, the AHH system also became a widely studied model of mammalian enzyme induction and adaptive metabolism (i.e., exposure to a xenobiotic substrate inducing its metabolism).16,17

Dibenzo-p-dioxins and Discovery of the Ah Receptor (AHR)

Additional evidence for the existence of an AHR arose from experiments designed to understand the mechanism of action of chlorinated dibenzo-p-dioxins and related environmental pollutants.18 Chlorinated dioxins and the related chlorinated dibenzofurans have never seen commercial use but are commonly introduced into the environment as trace contaminants of many industrial processes, anthropogenic sources, and some natural processes (Figure 2 and Table 1). The structurally related coplanar polychlorinated biphenyls (PCBs) and coplanar polybrominated biphenyls (PBBs) have seen commercial production and are often introduced into the environment as the result of industrial accident or improper disposal. As a class, these compounds display similar environmental fates, are environmentally persistent, are lipophilic, bioaccumulate in the food chain, and they elicit similar biological responses dependent upon chlorination pattern.1921

Figure 2.

Figure 2

Further elucidation of the Ah locus arose from toxicity studies using the toxicant TCDD. TCDD, 2,3,7,8-TCDF (tetrachlorodibenzofuran), and PCB 77 (polychlorinated biphenyl) induce the same P450s and related enzymes as did BAP, DMBA, and 3-MC. TCDD has higher affinity for AHR and thus has greater potency, making TCDD a model inducer of AHR signaling (see text for details).

Table 1. Well-Known Accidents/Exposures to Dioxin and Dioxin-Like Compounds.
location contamination source route of exposure human health implications example reference
Belgium transformer oil in fat of animal feed poultry, pork, beef, milk, eggs, and various fat-containing food items 50 kg of PCBs and 1 g of “dioxin” were contaminated in 500 tons of animal feed and distributed to farms in Belgium and on some nearby countries. 20–30% of these contaminants were estimated to have been consumed by the Belgian population. [272]
Ireland fuel from drying system used during animal feed production cattle, pork meat, and pork products Significant health concerns by public. Well-documented case of source and movement of dioxin and related compounds through commerce. [273, 274]
Seveso, Italy chemical factory accident: uncontrolled exothermic reaction during the manufacturing of trichlorophenol air and soil Immediate effects from cloud deposition: nausea, headache, skin lesions, and eye irritation. Long-term effects: Chloracne, increased incidence of diabetes, cancer, and mortality from cardiovascular and respiratory disease. [275, 276]
Vietnam, Laos, and Cambodia Agent Orange, Agent Pink, Agent Green, Agent Purple, Agent White, and Agent Blue: crude phenoxyacetic acid herbicides air, soil, surrounding waters, and ingestion (legacy through hand-to-mouth contact) Found association between dioxin exposure and soft tissue sarcoma, non-Hodgkin’s Lymphoma, chronic lymphocytic leukemia, Hodgkin’s lymphoma, chloracne, hypertension, and monoclonal gammopathy. Movement of dioxins into the human food chain and human tissues and biological fluids decades after use. [277, 278]
Times Beach, Missouri waste oil used for dust control air and soil Greater than 100 ppb in community. Human, wildlife, and livestock toxicity reported. [279, 280]

The dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is widely considered as the prototype for this class of environmental pollutant. Exposure to TCDD can lead to a broad spectrum of species-specific toxic effects, often referred to as the “dioxin toxic syndrome.” This syndrome commonly includes epithelial hyperplasia/metaplasia, chloracne, porphyria, late-stage terata, lymphoid involution, intestinal damage, hepatocellular damage, and cancer.18,22 Dioxins like TCDD are remarkably potent toxicants, with a median lethal dose (LD50) that can be in the low μg/kg range in some animal models.23,24 While dioxin toxicology has its research origins from an agricultural accident in poultry in the late 1950s,25 subsequent human exposures resulting from numerous pollution sources and environmental accidents, as well as its presence in the Vietnam War defoliant known as “Agent Orange”, sparked a modern effort to understand its mechanism of toxic action (Table 1).26,27,28,29 Despite the popular, regulatory, and scientific concern that has been in place for decades, halogenated dioxins, dibenzofurans, and biphenyls can still be found in human blood samples and in ecosystems around the globe.19,3033

It was through investigations into the toxic action of TCDD that we learned that the Ahr locus encodes a receptor. This conclusion arose from four observations.18,3436 First, TCDD induced the same P450s and related enzymes as did BAP, DMBA, and 3MC but with much greater potency.37 Second, radiolabeled dioxin analogues bound to a high affinity, low-capacity soluble protein site, designated as a receptor in target tissues.35 Third, binding affinity for this “receptor” site segregated with the high and low responsiveness (inducibility) phenotype observed across the C57BL/6 (“responsive”) and DBA/2 (“nonresponsive”) mouse strains.35,38 Fourth, the rank order potency for a given ligand’s potency to induce AHH activity (or many aspects of toxicity) corresponds to its rank order potency for receptor binding affinity. In sum, the early investigations into the genetics of PAH metabolism and the mechanism of dioxin’s toxic mechanism converged to reveal a soluble receptor known as the AHR, encoded by the Ahr locus, and proved that this receptor mediates many of the biological effects of these environmentally important pollutants.

Biochemical and Molecular Characterization of the Ah Receptor (AHR)

Early Molecular and Biochemical Insights

A better understanding of AHR signal transduction arose from a long history of pharmacologic and molecular studies of the regulatory elements of the genes encoding the induced enzymes that comprised AHH activity.34,3941 Although a bit more complex, we now know that AHH and PAH metabolism can be considered the composite activity of multiple genetic loci, including, Cyp1a1, Cyp1a2, and Cyp1b1. Each of the Cyp1 gene products encodes a member of the cytochrome P450-dependent monooxygenase family with metabolic activity toward PAH substrates.9,11,4246

The genomic elements controlling the ligand-activated AHR-dependent induction were given multiple names over the years, including xenobiotic responsive elements (XREs), dioxin responsive elements (DREs), and AHR responsive elements (AHREs, which we will use here).41,42,47 The discovery and characterization of these genomic enhancers were initially based mostly on studies of Cyp1a1 regulation. Of particular importance were the observations that the enhancers controlling AHR-mediated upregulation of Cyp1 genes commonly harbored consensus sequences of 5′-T/GNGCGTGA/C-3′. For Cyp1a1 and many other inducible genes, these elements often existed in multiple copies proximal and 5′ to the transcriptional start site of the target promoter.45,4850

Ah Receptor Nuclear Translocator

A significant step in developing a basic model of AHR signal transduction came from the molecular cloning of the AHR and its dimerization partner, the Ah Receptor Nuclear Translocator (ARNT).5153 These cloning experiments revealed that both the AHR and ARNT were structurally related, heterodimeric partners, harboring both basic helix–loop–helix (bHLH) and PER-ARNT-SIM (PAS) homology domains within their N-terminal halves54,55 (Figure 3). The bHLH domain occurs in metazoan transcriptional regulators and commonly provides both a dimerization surface and an α-helix that interacts with specific sequences in the major and minor grooves of DNA.54,5660 The PAS homology domain was named based on the similarity between amino acid sequences within ARNT and the products of two regulatory loci found in Drosophila melanogaster, PER and SIM (products of the per and sim loci, respectively).54 In addition to these two fruit fly gene products, PAS domains occur in a number of important mammalian regulatory proteins, including the “hypoxia-inducible factors” (HIFs) important in physiological adaptation to low oxygen and “clock” proteins central to the maintenance of circadian rhythms.61 Importantly, PAS domains have evolutionary roots in prokaryotic and plant systems, where parallel domains also play a role in environmental adaptation to stimuli such as light and oxygen62

Figure 3.

Figure 3

Mapping of AHR and ARNT functional domains and founding bHLH-PAS family members. The bHLH domain and N-terminus provide recognition of target DNA enhancers. The PAS domains control dimerization strength and selectivity, receptor repression, chaperone interactions, and ligand binding. Approximation of those domains for AHR are depicted as lines above. The C-terminus provides possible docking sites for coactivators.5461,64,65,6770,72,74,75,77,79,80,91,131,281283 Also see text for further details.

Functional Domain Maps

The importance of the bHLH-PAS region in dimerization and DNA binding is provided by numerous functional mapping studies in both the AHR and ARNT.60,6365 Like many bHLH proteins, the AHR and ARNT employ this domain as a dimerization surface and use the basic N-terminal helix to provide recognition of target DNA enhancers, with each basic region laying within a “half-site” of the AHRE (e.g., TNGC or GTG).5759,61,6668 The functional role of the PAS domain can be thought of in the context of its two degenerate repeats or subdomains, referred to as PAS-A and PAS-B. The PAS-A domain plays a significant role in supporting the dimerization that drives DNA binding selectivity. In contrast, the PAS-B domain is important in dimerization but also harbors domains for receptor stabilization, receptor repression, chaperone interactions, and ligand binding.61,63,6973

Although the C-terminal halves of the AHR and ARNT are highly divergent at the sequence level, this region appears to harbor domains of similar function in the two proteins. Studies employing fusions of this region with heterologous DNA binding domains reveal that potent transcriptional activation domains (TADs) reside within the C-terminal halves of these proteins, overlapping with glutamine-rich or highly acidic and disordered regions.7478 In more recent years, additional docking domains for some coactivators map to the bHLH-PAS domains of both the AHR and ARNT79 (Table 2). While our understanding of coactivator associations is still nascent, the AHR–ARNT-dimer-mediated transcription of target genes such as Cyp1a1 appears to involve, at least in part, classical chromatin modifications and recruitment of members of the transcription initiation complex.79,80

Table 2. Examples of Some Coactivators That Have Been Shown to Associate with AHR or Its Complexa.
coactivator reference notes
BRCA-1 [284, 285] interaction with both AHR and ARNT
BRG-1 [286, 287] interaction with AHR, enhances complex activity
CARM-1 [288] interaction with AHR
CoCoA [289] interaction with both AHR and ARNT
COUP-TF1 [290] interaction with AHR and not ARNT
ERα [290] interaction with AHR and not ARNT
ERRα [290] interaction with AHR and not ARNT
ERAP140 [291] interaction with AHR–ARNT complex
GAC63 (GRIP1) [292] interaction with AHR
Mediator [97] interaction with AHR–ARNT complex
P160 (NcoA-1-3) [117] interaction with both AHR and ARNT
NcoA-4 [93] interaction with both AHR and ARNT
P300 [117, 293, 294] interaction with both AHR and ARNT
PGC-1 [288] interaction with AHR
RB [295] interaction with AHR
RIP140 [296, 297] interaction with AHR, cross talk with ERα
SHP [298] interaction with ARNT and not AHR
SMRT [291, 299] interaction with AHR–ARNT complex and AHR
SRC1 (NcoA-1) [283, 288] interaction with AHR Q-rich region
SRC2 (NcoA-2) [288] interaction with AHR
SRC3 (NcoA-3) [288] interaction with AHR
TAF4 [283] interaction with AHR Q-rich region
TAF6 [283] interaction with AHR Q-rich region
TBP [283] interaction with AHR Q-rich region
TIF2 [283] interaction with AHR Q-rich region
TRAP220 [288] interaction with AHR
TRIP230 [300] interaction with ARNT
SIN3A [301] enhances complex activity
a

This table of examples was generated by a cross reference of the topics, “Ah receptor” and “Coactivator” in the “Web of Science” search engine of scientific publications, apps.webofknowledge.com (8/1/2019). It was then supplemented with information found in two reports on the topic.79,117 Clear, alternative names of coactivators are given in parentheses. The table is meant to represent the diversity of known AHR–coactivator interactions and is not intended to be an exhaustive list.

Adaptive Metabolism Pathway

Model for Adaptive Metabolism

As the result of the first 50 years of investigation into the AHR, we have a working model of the functional domains and signaling steps that regulate the expression of the xenobiotic-metabolizing enzymes such as CYP1A1 (Figure 4).12,36,47,48,58,73,81 Through the use of molecular reagents from cloned AHR and ARNT, mutant hepatoma cell lines, immunochemical tools for localization and precipitation, and high-affinity radioligands, the importance of subcellular localization, chaperones, and the ordering of signaling steps for upregulation of genes is becoming clearer.34,36 The most common description of AHR signaling as it relates to CYP1A1 gene induction is as follows: In the absence of an inducing ligand, the AHR protein resides predominantly in the cell’s cytoplasm in a complex with a number of chaperones, including a dimer of the 90 kDa heat shock protein (Hsp90) and smaller chaperones known as the AHR interacting protein (AIP, also known as ARA9 or XAP2) and the P23 protein.8289 Upon the binding of ligand to the AHR, a conformational change in the receptor leads to a reorganization of chaperones and allows presentation of the NLS in the AHR’s N-terminus. Translocation of the AHR to the nuclear compartment then allows dimerization with its nuclear partner, ARNT.58,84,87 The AHR–ARNT dimer produces a competent DNA binding dimer with specificity for AHREs within chromatin and activation of nearby target promoters such as that for Cyp1a1. While much is still to be learned about this final transcriptional step, the AHR–ARNT dimer has been shown to increase promoter accessibility and alter chromatin structure through association with numerous known coactivators9098 (see examples in Table 2).

Figure 4.

Figure 4

Classic AHR signaling pathway. Prior to ligand binding, AHR remains in cytosol bound to HSP90, P23, and ARA9. When a ligand binds, a conformational change occurs, exposing the nuclear localization sequence (NLS) in AHR’s N-terminus. Presentation of NLS permits the translocation of AHR to the nucleus and subsequent dimerization with ARNT. The AHR–ARNT heterodimer recognizes and binds to AHREs in the genome and initiates transcription of select genes. This interaction can be inhibited by AHRR, CYP1 metabolism of ligands, and post-translational modification of the receptor.12,34,36,47,48,58,73,8189,9193,96,97,291,294,297,302

Pathway Feedback Inhibition

One intriguing observation about AHR signal transduction is that several mechanisms exist to downregulate this signaling. Primary evidence for the importance of feedback inhibition comes from one of the more recently discovered targets of the ligand-activated AHR–ARNT complex, an additional bHLH-PAS protein known as the Ah receptor repressor (AHRR).99,100 The AHRR not only dimerizes with ARNT and competes for AHRE occupancy but also inhibits AHRE-mediated transcription by influencing the chromatin structure around the promoters of CYP1A1 and presumably related AHRE-driven genes.101,102 In addition to this upregulated repressor activity, the AHR also appears to be the target of multiple additional downregulators.103 Not only has the ligand-activated AHR been shown to be constitutively degraded by ubiquitination and proteasomal degradation,104106 one of its AHRE-driven target genes, Tiparp, may ADP-ribosylate the AHR, reducing its activity and half-life.107,108

The existence of AHRE regulated genes such as AHRR and TIPARP provides support for an additional perspective on AHR signaling. While we classically think of this system as a pathway to adapt to PAH molecules generated exogenously or endogenously, it is also interesting to think of the CYP1A1/CYP1A2/CYP1B1 gene targets as additional participants in a negative feedback loop. That is, activation of the AHR by PAHs (and other ligands described below) leads to the upregulation of CYP1 monooxygenases and their consequent metabolic degradation and excretion of inducing ligands. This would appear to represent a classic substrate inducing its own metabolism in a feedback loop.12,47,48 These observations lead to the question: why is so much biology directed toward AHR downregulation and attenuation of signaling? Possible answers are that the AHR is part of a biological response that must be rapidly attenuated to avoid pathological consequences or that it is part of a chronic response that must be precisely modulated over time (see below).

More to Be Learned about Functional Domains and Signaling

It is important to note that this current description of the AHR domain map and signal transduction is almost certainly an oversimplification, with issues such as the importance of receptor phosphorylation and the events dictating receptor transformation still unclear.109112 Similarly, while we have learned a great deal about the bHLH domain, much is still to be learned about the N-terminal half of the AHR. While the bHLH tail is thought to harbor both DNA recognition and nuclear localization sequences (NLS), this region also appears to play additional roles in receptor signaling. In one example of this idea, this same region harbors a nuclear export sequence (NES), which influences receptor subcellular localization, and also a motif for cellular chaperone interaction, which can influence receptor concentration and transformation.72,113115 Finally, our understanding of how this domain interacts with the genome is probably also incomplete. This conclusion is supported by the identification of noncanonical enhancer target sites that are in addition to classical AHREs as defined above.116

Similarly, our understanding of the C-terminal halves of these proteins and the transactivation events they mediate is also limited, with the role of specific coactivators in distinct cellular responses still to be determined and the importance of this domain in receptor transformation still unclear. An improved understanding of the multiple coactivator interactions and insights into their combinatorial and dynamic nature will be important if we are to explain the wide variety of cell-, species-, and ligand-specific responses induced by AHR agonists and antagonists.91,96,116,107,118 In this regard, many of the species-, tissue-, and ligand-dependent effects of AHR agonists may be due to unique consequences of specific coactivator recruitment within a given cellular environment (in addition to differences in ligand-binding affinity/specificity). Moreover, ligand-dependent recruitment of specific coactivators could underlie the unique pharmacology of distinct agonist classes.

Dioxin-Like Compound Concept

An early observation that has greatly influenced our thinking is that while most AHR agonists can achieve similar efficacy with respect to upregulation of AHH activity (i.e., CYP1A1, CYP1A2, and/or CYP1B1), only the most potent and metabolically recalcitrant (i.e., long T1/2) ligands induce the “dioxin toxic syndrome.” Weaker agonists, such as the PAHs and variety of natural ligands, which are rapidly metabolized and have lower potency, appear to upregulate the CYP 1s but do not induce chloracne and so on. The model proposed to explain this phenomena is that there is a “restricted pleiotropic response”, in addition to the upregulation of genes such as CYP1A1, which is induced by longer-lived, pharmacologically unique agonists, and this response is required for the dioxin toxic syndrome.18 In fact, certain end points like CYP1A1 induction (i.e., adaptive metabolism) can occur in response to a broad spectrum of ligands, whereas end points of the dioxin toxic syndrome appear to require receptor activation by compounds with “dioxin-like” pharmacological properties. As a heuristic, chlorinated dioxins, dibenzofurans, and biphenyls with halogens in lateral positions show the greatest potential to induce the dioxin toxic syndrome and are therefore often designated as “dioxin-like compounds” (DLCs) (Figure 5).20

Figure 5.

Figure 5

Dioxin-like compound concept and approach to measuring human exposure to mixtures. Toxic equivalency factors (TEFs) are weighted measures that reflect the relative potencies of pollutants of concern as compared to TCDD. Toxic equivalents (TEQs) are reported values used for risk characterization and management (see text for details). Left: Structures of the three classes of chlorinated DLCs. Right: Examples of three formally designated DLCs. To calculate TEQ, the mass of each chemical in a mixture is multiplied by its TEF and summed.23,119,120,122,124,209,303,305

The idea that DLCs elicit effects that are distinct from other classes of receptor ligands (like the PAHs) has regulatory implications. Agencies such as the World Health Organization (WHO) that direct global health efforts and agencies like the Environmental Protection Agency (EPA) that govern chemical releases within the United States employ the principle that compounds of environmental concern that are structurally and pharmacologically related to TCDD and that elicit toxicity through a common mechanism are formally designated as DLCs, and their exposures and release are regulated concordantly.20 The principle used is that these compounds are assessed for their relative effect potencies (REP) from dose–response assessments for a pathological end point associated with AHR activation and toxicity. This information is used to generate toxic equivalency factors (TEFs), which are weighted measures that reflect the relative potencies of a pollutant of concern as compared to TCDD (Figure 5).23,119124 Currently, TEFs are applied to 29 compounds of environmental concern, 7 polychlorinated dibenzo-p-dioxins, 10 polychlorinated dibenzofurans, and 12 polychlorinated biphenyls. The advantage of this approach is that it provides a measure of toxicity from complex mixtures of chlorinated dioxins, dibenzofurans, and biphenyls, which are common in human exposure scenarios.

Insights from Naturally Occurring Structural Diversity in the Ah Receptor

Genetic Variation/Polymorphism

Early evidence indicates that the AHR was functionally and structurally variable both within and across species. Support for this idea arose from the observation that murine Ahr polymorphisms lead to differential induction of P450s across strains.35,42 Further, examination of additional animal species, including hamster, guinea pig, rat, dog, and human, revealed significant differences in sensitivity and response to dioxins.22,125,126 This idea of receptor diversity gained further support with the development of antibodies and photoaffinity radioligands that revealed biochemical differences in AHR both across and within model species.127130 The molecular cloning of the AHR cDNAs from multiple animal species revealed important codon polymorphisms in the Ahr gene that, when paired with radioligand-binding experiments and immunochemistry, led to the identification of codons that influence receptor size and ligand-binding affinity (see below).52,53,131,132

Molecular Insights from the Structural Gene

The AHR structural gene, Ahr, resides on mouse chromosome 12 or on a highly syntenic region on human chromosome 7.133136 Comparison of the structural genes and cDNAs from mouse and human indicates that the open reading frame is encoded by 11 exons with highly conserved intron–exon boundaries across species. A comparison of these genes reveals that alternate termination codons for the open reading frame in exon 11 explain much of the receptor size differences observed within and across species. This molecular information indicates that the receptor open reading frame extends further in some species (e.g., human and rat) and much less in others (e.g., the C57BL/6J mouse). This leaves some proteins with longer C-termini than others and explains how the AHR can be as small as 97 kDa in the C57 mouse and as large as 105 kDa in the human or 124 kDa in the hamster.128,130,132,135,137

Molecular Insights from the Mouse Model

The mouse is an important animal model for the study of the AHR and its signaling pathways. The initial mouse “responsiveness” polymorphism was explained through the comparison of the AHR cDNAs derived from responsive (Ahrb allele) and the less responsive strains (Ahrd allele). These experiments revealed that there were numerous polymorphisms between the Ahrb and Ahrd alleles.138140 Among these is a polymorphism in the stop codon, resulting in an additional 43 amino acids in the carboxyl terminus of the AHRd receptor as compared to the AHRb1 (identical to the cause of the cross-species differences described above). Interestingly, two additional responsive alleles were characterized (named Ahrb2 and Ahrb3),141 one of which (Ahrb2) closely resembles the Ahrd allele in all but three amino acids and includes an identical elongated c-terminal tail. Using ligand binding of expressed polymorphic proteins, it was concluded that a primary driver of the ligand-binding affinity was residue 375, where an alanine (A) confers higher affinity ligand binding and greater responsiveness in mice harboring the b1, b2, and b3 alleles. In contrast, in Ahrd mice, a valine (V) at this position confers lower affinity binding and decreased responsiveness.139,140 Through investigations into other residues, it was also observed that the elongated C-terminal tail found in Ahrb2 and Ahrd mice may reduce ligand binding slightly as compared to Ahrb1.140 It remains unclear the extent the C-terminal half plays in ligand binding, as this region also alters receptor stability and thus perhaps cellular concentration.

Predictions of AHR structure have been modeled using receptor homology data from other PAS family proteins, such as HIF-2α. These analyses support the importance of residue 375 in ligand binding as well as the influence of alanine and valine at this position.69,142145 That is, the valine at 375 encoded by the Ahrd allele is bulkier and hypothesized to have repulsive properties toward the ligand while also altering the adjacent hydrogen bond network. Interestingly, the human harbors a valine residue at this position, and this may be better modeled by “humanized” or the AHRd models.146

Molecular Insights from the Rat Model

The rat has also served as a powerful early model of AHR biology. This utility arose from the classical use of this model as a tool in toxicology, its sensitivity to TCDD induced carcinogenicity,147 and the existence of an informative polymorphism in the receptor that influences a strain’s responsiveness to agonist.125 Similar to the mouse, some rat strains are resistant (Han–Wistar, HW, 98 kDa), while others are sensitive (e.g., Long–Evans, LE, or Sprague–Dawley, SD, 106 kDa) to the toxic and inductive effects of ligands like TCDD. Through molecular analysis of the cDNAs and structural genes of these AHR open reading frames, it is now known that the explanation for reduced signaling by the HW receptor is due to a variation at the splice junction at exon–intron 10. While multiple consequences of this altered splice junction can occur, this polymorphism commonly leads to a truncation of the C-terminal end of the HW-AHR, yielding as many as two novel protein products possible.148 Physicochemical studies indicate that this truncation reduces receptor concentration, possibly due to influences on receptor stability or the potency of the nearby transcriptionally active domains.148,149 This naturally occurring receptor polymorphism in the rat provides considerable evidence for the role of the receptor’s C-terminus in AHR signaling and dioxin toxicity. An additional note is the observation that while HW rats are resistant to many of the acute toxic effects of high-dose dioxin exposure, they display similar dose–response curve for end points such as CYP1A1 induction. Such a result would seem to be an indication that classes of AHR-mediated biological/toxicological responses exist, some of which require less receptor activation than others.150,151 Such an observation is in keeping with the restricted pleiotropic model described above.18

What Is the Normal Physiological Role of the Ah Receptor?

While the toxicology of PAHs and dioxins led to the discovery of the AHR as well as the discovery of the AHR’s roles in regulating xenobiotic metabolism, many significant questions remain regarding the role of this receptor in normal physiology. Perhaps one of the most important questions is why this receptor exists in such a wide range of animal species and in such a broad array of tissues and cell types? Early research focused on the concept that the receptor was part of a system that evolved to allow metabolic adaptation to xenobiotics, especially PAHs, which have existed on the earth for millennia due to natural processes such as fires and volcanic activity.152 Parallel thinking suggests that the AHR evolved as an allelopathic defense system, similar to those systems reducing exposures to lipophilic natural products that display toxicity when levels rise in an organism.153,154 While these ideas are all probably correct in some form, it is also probable that this is not the only physiological role of the AHR nor are they the primary reason for its evolutionary conservation (see below).

Lessons from Tissue and Cellular Expression

One common approach used to deduce the physiological role of a gene product is to determine where and when the protein is expressed in an organism. This method relies on the premise that tissue-specific or developmental expression will highlight the relevant biological system. This approach has been used to understand AHR biology and includes studies based upon ligand binding, antibodies, and RNA analysis to report receptor expression at the organ and tissue level.128,131,155157 These early studies are now complemented by high-throughput gene expression resources such as BioGPS, ENCODE, and The Human Protein Atlas.158160

While the interpretation of the collective data from the above sources is complex, a few important observations are noteworthy. At the organ level, the AHR is expressed at many sites, with the placenta expressing the highest levels of the AHR mRNA in the human.131 The human lung is also a highly expressing tissue in almost all reported studies and databases, with levels in liver and bladder/urinary tract also reproducibly high. In contrast to humans, in the mouse and rat, the lung is typically the highest expressing organ and the placenta is much lower. While issues such as gestation day may play a role in this reported cross-species difference, it is notable that the human placenta is physiologically distinct from rodent placentas.161 We draw two conclusions from these observations. The first is that the AHR is most highly expressed at tissues that represent important oxygen interfaces (lung and placenta). The second is that if the AHR is important in human placental biology, current animal models may significantly misrepresent this important physiology.

Predictions about endogenous function based on higher resolution and temporal expression data (i.e., immunohistochemistry and in situ hybridization techniques) are difficult to simplify, because these studies describe AHR expression in a remarkable array of cellular compartments and developmental times. For example, in the E13.5 day embryo, the AHR is highly expressed in the primitive pituitary, nasal septal cartilage, dorsal surface of the tongue, developing thymus lung parenchyma, liver, mucosa of the developing gut, urogenital sinus, and genital tubercle.162 A parallel analysis of the CNS indicates that AHR and ARNT are coexpressed in regions of the hypothalamus and brainstem associated with appetite and circadian regulation, and it is also highly expressed in cardiac and skeletal muscle and epithelial regions associated with epithelial to mesenchymal transitions.163165 Adding to this diversity, are reports of AHR expression in rabbit morula and blastocysts, human pancreatic ductal and acinar cells, immune cells of the intestinal stroma and ovarian granulosa cells.166169 Given that this is only a small list of unique sites of AHR expression, it seems likely that this receptor will be shown to have more than one significant role in normal animal physiology and development.

Lessons from Ahr Null Rodents

Another method used to identify putative physiological roles for the AHR is to create mammalian models that are null for the Ahr gene product and assess the consequences of that null allele on the host’s biology. Generation of the null allele has been performed by at least three independent laboratories for the mouse model and at least once in the rat model.170,171,171,172,173 In rodent models, the Ahr null allele has again provided evidence that the AHR regulates multiple developmental and physiological processes. In this regard, Ahr null mice have been reported to display a number of phenotypes, including patent ductus venosus, hepatic atrophy, altered immunity, vascular defects, decreased barrier integrity of the skin and gut, and reduced reproductive capacity.12,162,166,174183 While initial reports of variously generated mouse null alleles appear to display some discordance, there is little evidence to indicate that any differences are allelic; it is more likely they are due to genetic background issues, unique pathogen loads, and different dietary regimens.176

Interestingly, the rat null model displays a phenotype that is distinct from the mouse, with pathological alterations primarily in the urinary tract and kidney and no reported hepatovascular pathology (i.e., patent ductus venosus) which is a hallmark mouse phenotype studied in our laboratory.173 Moreover, while immune effects in the mouse have predominantly been studied for adaptive immunity and T-lymphocyte biology, the effects in the rat have been reported primarily for B-lymphocyte function.184186 Taken in sum, these data strongly support a role for the AHR in normal biology, with initial indications of an important role for this receptor in barrier integrity, immunity, reproduction, vascular development, as well as hepatic and renal biology.166,184,187189 Additionally, these cross-laboratory and cross-species studies indicate that experimental environment and genetic background are likely to have a marked influence on AHR null phenotypes and AHR biology writ large.

Lessons from Evolution

Another strategy to elucidate the physiological role of the AHR is to study its evolution. Such an approach anticipates that certain correlates might explain the selective pressures that led to the receptor’s emergence and maintenance in biological systems. The AHR, ARNT, and AHRR are members of the bHLH-PAS family of transcription factors, which arose early in evolution, with PAS domains having been found in plants, animals, and bacteria.62,190 Domains reminiscent of PAS domains are found in prokaryotes, where they play roles in phototropism and oxygen sensing, and in plants, where they are involved in photoreception and phototransduction.191

Diversification of the PAS gene family occurred early in evolution. All of the major bHLH-PAS gene subfamilies (e.g., AHR, ARNT, HIF, SIM, CLOCK, TRH, BMAL, NCOA, NPAS4) are shared by protostomes and deuterostomes and thus must have been present already in the ancestral bilaterian animal, which lived ∼570 million years ago.192,193 Metazoan PAS domain-containing proteins play roles in a variety of signal transduction pathways, many of which are involved in developmental processes and environmental adaptation.194196 Additional diversification of bHLH-PAS genes occurred early in the vertebrate lineage as a result of two whole-genome duplications,197 leading to the multiple paralogues (ohnologues) within each subfamily that exist in most vertebrates, including mammals (e.g., three HIF genes, two CLOCK genes).

The AHR genes have undergone duplication and diversification like other bHLH-PAS genes, involving both whole-genome duplications as well as a tandem duplication event. The presence of multiple AHR genes in both bony and cartilaginous fishes suggests that the AHR gene duplications occurred early in vertebrate evolution, well before the emergence of mammals.198,199 A tandem duplication produced the genes now known as AHR1 and AHR2, which occur in fish, birds, reptiles, and some early diverging mammals but have been lost from most later mammalian groups. Another duplication event produced AHRR, which evolved as a transcriptional repressor of AHR function.100,200 In mammals, AHRR may exhibit additional regulatory interactions besides repressing AHR activity, consistent with data demonstrating that human AHRR can have multiple effects on cell growth and differentiation.200 Overall, phylogenetic and comparative genomic analyses suggest that there are five groups (clades) in the AHR subfamily: AHR, AHR1, AHR2, AHR3, and AHRR, which exhibit gene- and taxon-specific functional specialization.199

Functional analyses of AHRs from extant vertebrate and invertebrate species suggest that the ability to bind to planar aromatic compounds such as PAHs and dioxins evolved in early vertebrates. It has been hypothesized that one selective force may have been the need to detoxify halogenated aromatic natural products, which are prominent in the marine environment, where early vertebrates arose.201,202 Although AHR homologues from invertebrate species appear to lack the ability to bind PAHs and dioxins, it is unknown if they can be activated by other types of ligands.

While evolutionary information does not point us toward a clear physiological role for the AHR, some intriguing observations stand out from this analytical approach. First, PAS domains have a propensity to exist in sensor proteins of environmental stimuli such as light and oxygen tension.61,191,203 This role seems to have evolved early (prokaryotes and plants) and been maintained throughout millions of years of evolution. The AHR’s role as a chemical sensor is consistent with this idea. Second, the AHR, as defined by phylogenetic analysis (orthology) within the bHLH-PAS family, has been found in almost all eumetazoan groups.199 This suggests that whatever evolutionary pressures have led to the maintenance of this gene, they have existed for millennia. Third, there may be a common thread that unites AHR function across the metazoan: such as a role in controlling cell fate during the development of neural systems and, in particular, sensory structures. For example, in the cnidarian Nematostella, AHR is expressed in the apical tuft (a sensory structure).193 In arthropods (e.g., Drosophila), AHR controls the development of the distal segment of the antenna (a chemosensory structure), mechanosensory bristles, and photoreceptors.204,205 In nematodes (e.g., C. elegans), AHR controls the development of touch receptor neurons and sensory neurons that contact the pseudocoelomic fluid.206 Emerging evidence for a role of AHR in neural development in mammals suggests this could be one possible conserved role shared by all animals.199,207 Despite these intriguing findings, it will remain a challenge to identify conserved physiological roles of AHRs and to distinguish them from novel functions that evolved in specific taxonomic groups.

Diversity of AHR Ligands

Structure–Activity Relationships

Early structure–activity relationship (SAR) analysis based on various halogenated aromatic hydrocarbons (HAHs), PAHs, and related compounds, suggested that the AHR ligand-binding pocket binds near-planar ligands with dimensions that approximated a 3 × 10 Å (Å) rectangle.18,208 More recent analyses based, in part, on structure–activity studies and on structural similarity to crystallized domains of other PAS proteins, support the idea that absolute planarity is not a requirement for receptor binding and that maximal dimensions of the ligand-binding pocket may be more closely approximated by a pocket of 14 × 12 × 5 Å (reviewed in ref (154)). It has also been observed that both the hydrophobicity and the polarizability of a compound’s substituents add an additional layer of complexity in regards to affinity for AHR.154,209213 While current structural models are useful, a solved binding pocket structure through X-ray crystallography or NMR is needed if we are to confidently predict chemical binding to the AHR and anticipate the biological effects that emerging environmental pollutants and therapeutics will induce. In the meantime, those of us with limited expertise in physical chemistry are left with a preliminary “flat hydrophobic rectangle” (FHR) model as a predictor of AHR ligand-binding activity (Figure 6).

Figure 6.

Figure 6

The flat hydrophobic rectangle model of AHR ligands. Molecular and ball and stick models of some AHR ligands discussed in this review that conform to the FHR concept of ligand structure. The three-dimensional structures are only provided as approximations, as some subtle bending and puckering of structure may occur that is not predicted by common algorithms. Figure was generated with ChemDraw software.

Several reviews have provided a comprehensive description of the structural diversity of AHR ligands and sources.24,154,209,214 While structural classification of AHR ligands based upon chemical backbone is useful (dioxins, biphenyls, PAHs, flavonoids, etc.), it is also useful to think of these compounds based upon nonstructural properties, such as source, risk for human exposure, receptor binding affinity, and biological half-life. In this regard, ligands coming from anthropogenic sources such as diesel exhaust, commercial production, or industrial contamination (PAHs, PCBs, and dioxins), are produced as natural products, or they are generated endogenously in human tissues (indigoids, indolocarbazoles, etc.). For many of these source classes, member ligands display EC50 values or binding affinities for the AHR that differ by multiple orders of magnitude. Ligands from these source classes can also harbor markedly different biological half-lives that span from hours to months.215217

Importance of Proligands

An important concept to consider is that many compounds that are thought to activate the AHR are not actual ligands of the AHR but are proligands. Proligands are precursors that are chemically transformed to the ultimate ligand, which strongly binds to the AHR pocket. Proligands typically form the ultimate ligands via condensation reactions of precursor molecules into larger planar, more stable, polycyclic aromatics.218,219 Such reactions can often be spontaneous or nonenzymatic. The first discovered and perhaps clearest example of a proligand is indole 3-carbinol (I3C) produced in broccoli, Brussels sprouts, and kale. This naturally occurring 3-substituted indole is produced from enzymatic breakdown in the plant tissue from a glucosinolate known as glucobrassicin.219221 Indole-3-carbinol was originally studied as an anticarcinogenic substance by virtue of its activity as an inducer of carcinogen metabolism.222,223 The premise was that dietary I3C protected against coadministered carcinogens such as BAP and DMBA by “blocking” their action through a reduction in their relative metabolic flux to ultimate electrophiles that damage DNA.224 Interestingly, we now know that I3C itself is not a ligand of the AHR, but when I3C is ingested, it hits the low pH environment of the stomach and spontaneously undergoes an acid-catalyzed condensation reaction, converting it to a variety of AHR ligands including the potent agonist indolo[3,2-b]carbazole (ICZ).219,225,226 Condensation products of I3C, such as ICZ, are high-affinity binders of the AHR and can be found in the bloodstream after exposure to I3C in the diet.219

One important lesson to be learned from the proligand idea is that when a compound does not fit the FHR model described above, some caution should be ascribed to any inclusion of the compound into a list of bona fide endobiotic or xenobiotic ligands. A list of “nonclassical” compounds that activate CYP1A1 expression but that do not obviously fit the FHR model is included in a recent review and includes SKF71739, thiabendazole, omeprazole, and 1,5-diaminonapthalene.154 We propose that, often, such ligands may actually be proligands. In addition to I3C described above, a number of other examples support the concept that proligands are a common source of receptor activation, including the identification of alanine serine aminotransferase (AST) and D-amino acid oxidase (DAO) as enzymes capable of activating the AHR in cell culture.227,228 The biochemical explanation for receptor activation by these enzymes is the generation of indole-3-pyruvic acid (I3P) from tryptophan (TRP) through deamination.227,229 Like I3C, I3P is a reactive indole, and this α-keto acid spontaneously condenses to a number of di-indol structures and possibly related backbones that are the ultimate AHR ligands or a more proximal precursor to them.229 Similarly, in recent studies of the immune system, numerous laboratories made the observation that small relatively polar immunomodulators produced from TRP by the enzyme indoleamine 2,3-dioxygenase (IDO) can activate the AHR.230 Given the lack of fit of many of these IDO products to the FHR model, it was again shown that IDO products such as kynurenine (KYN) or 3-hydroxyanthranilic acid (3HAA) are also proligands that are converted to a series of “trace extended aromatic condensation products” (TEACOPs).231,232 It is probable that molecules such as these are high-affinity ligands and potent AHR agonists in vivo.

The idea that proligands may be more common than is currently appreciated may explain how AHR ligands display so much reported structural diversity. In our simplistic view, it may be that all AHR ligands must fit the FHR model, and when a structure does not fit, it is more likely a proligand rather than a true ligand. Either it is being converted to a TEACOP or the TEACOP is a trace contaminant of the material being used in the experiment. In this regard, if one examines a potential ligand with a high EC50 for induction of an AHRE-mediated response, some consideration of the possibility that the ligand is contaminated with, or is generating TEACOPs, should be considered. In this regard, I3C has an EC50 that is approximately 5 orders of magnitude higher than ICZ for competition with TCDD for AHR occupancy (i.e., 5 orders of magnitude lower affinity).219 In the absence of acid condensation conditions, the I3C response may be explained by the contamination of ICZ equivalents at 1 part in 100 000 (0.001%). We argue that many compounds that are activators of the AHR at high concentrations may be contaminated with or generate a series of TEACOPs, thereby confusing structure–activity relationships.

Classifying Ligands: Xenobiotic, Endobiotic, and Cognate

We often think of AHR ligands as existing in two physiologic classes: “xenobiotic” and “endobiotic.” We employ the term xenobiotic for those compounds found in an organism that are not produced within that organism. Their presence in the organism is “foreign” or from a foreign source (“xeno”).233 Common sources of xenobiotic ligands include diesel exhaust (e.g., PAHs), chlorophenol manufacturing (e.g., dioxins), or pharmaceutics (e.g., omeprazole). Xenobiotic ligands can also be “natural” and include normal constituents of fruits and vegetables (e.g., chrysin, quercetin, and galanin).234236 In contrast, we reserve the term endobiotic ligand to denote any AHR ligand that is produced readily in a given biological system, including within the gastrointestinal tract. A few widely studied endogenous ligands are 6-formylindolo[3,2-b]carbazoles (FICZ), 2-(1′H-indole-3-carbonyl)-thiazole-carboxylic acid methyl ester (ITE), indigo, indirubin, and bilirubin.237240

A final definition that may also be useful going forward is the term “cognate.” We use this term to refer to those ligands that have provided the selective pressure for the evolutionary conservation of the AHR. This class of ligand has also been referred to as “the endogenous ligand”, “the physiological ligand”, or even “the ancient ligand.” We define cognate ligands as those ligands that correspond to the evolutionary pressure that has led to the emergence and maintenance of this receptor through evolution. Put another way, these are the ligands that have the most important consequences on normal physiology. In the absence of these ligands, the organism cannot thrive under all developmental and physiological stresses. This concept of the cognate ligand is important, because it implies that the AHR has evolved in parallel with a ligand (or set of ligands) as its evolutionary pressure. In turn, this implies that the AHR has a physiological role that is separate from and in addition to the adaptive metabolism of xenobiotics.

Xenokine Model of Ah Receptor Signaling

In a previous review, rudimentary models of AHR signaling have been put forth that might explain how potent DLCs lead to the dioxin toxic syndrome.18,36 Given that many of those models are still untested or unrefuted, we have turned our attention toward an understanding of the AHR’s cognate signaling with the underlying idea that such insight will explain many of the pathological consequences of DLCs. Based upon this history, the ideas presented above, and the models of toxicity described previously, we close this manuscript with an attempt to provide testable models of AHR cognate signaling. While our initial plan was to summarize all the ideas that have been put forth in the peer-reviewed literature, we found that there were so many ideas and so much intriguing evidence, that we could not objectively integrate them all. In this regard, the remarkable breadth of phenotypes influenced by AHR biology is provided in a recent review and reflects a potential role for this receptor in almost every major organ system and an influence on processes as diverse as cell cycle progression, immunity, DNA biochemistry, reproduction, and circadian rhythmicity.12 Therefore, we chose to articulate a parsimonious model that employs ideas borrowed from the works of many AHR scientists but that is interpreted through the prism of our own laboratory’s research experience over the last 30 years.12,18,170,187,241246

This integrated model of cognate signaling has its roots in the adaptive metabolism of PAHs, dioxin toxicology, and recent results from AHR null mouse models.12,18,170,187,242246 We refer to this way of thinking about AHR biology as the “xenokine model.” The xenokine model is based on the idea that there is parallelism between AHR’s role in the adaptive metabolism of xenobiotic environmental ligands and a similar class of endobiotic ligands that we collectively think of as “xenokines.” These xenokines are generated within the organism but commonly outside of cells, such as in interstitial spaces, the lumens of organs, or regions of cellular disruption. We propose that these xenokines are generated by endogenous chemical reactions that generate agonists in a manner similar to the generation of agonists found in the environment. In turn, these xenokines are sensed by the AHR, which stimulates a transcriptional response that is linked to a new physiological state better adapted to the new challenge they represent. Like the adaptive response to xenobiotics, it follows that the pathway is under feedback regulation and xenokine action is rapidly attenuated through CYP1 induction, AHRR upregulation, and so on.

We predict that xenokines may be as structurally varied as the spectrum of known environmental ligands but will fit the FHR model described above. We anticipate that the identification of all cognate ligands may be difficult to achieve due to the possibility that each tissue or organ system may have its own unique variety of xenokines that arise from the distinctive chemistries of each specific tissue and environmental stimuli. While the exact identity of the cognate ligands are still to be elucidated, evidence from the literature suggests they could arise from products of polyunsaturated fatty acids or heme metabolites, or they could be produced from aromatic amino acids like TRP through enzymatic reactions, nonenzymatic condensation reactions, free radical reactions, microbial metabolism, inflammation, and UV irradiation.12,154,209,247252

Aromatic amino acids such as TRP and phenylalanine are potentially important proligands and sources of AHR cognate ligands.227,253255 In fact, the molecule that has the most experimental support for this definition of xenokine is the TRP photoproduct and TEACOP known as FICZ.247 Evidence that this indolocarbazole is an important cognate ligand includes the observations that FICZ harbors an AHR binding affinity among the highest ever observed, is produced endogenously at epithelial barriers in response to UV irradiation, and appears to play a role in AHR-mediated immune and epithelial response to environmental stressors including bacterial invasion, oxygen stress, and UV damage.187,247 Moreover, FICZ is rapidly metabolized by the CYP1 monooxygenases, implying its levels are tightly regulated by the feedback loop described above.215 Lesser but provocative evidence exists for the physiological importance of additional TRP-related xenokines at other tissues. For the intestinal barrier, evidence supports the idea that TRP metabolites arising from gut microflora play important roles in activating AHR signaling to influence gut barrier integrity through influence on intestinal lymphocyte populations (e.g. refs (256258)). Even more speculative is the idea that products of the enzyme DAO, which harbors metabolic activity toward d-amino acids such as d-TRP found in bacteria, or AST, which harbors metabolic activity toward TRP, can both generate the AHR proligand I3P.227278,229,253 Thus, oxidases, deaminases, and transaminases like these have potential to generate proligands and ultimately xenokines in vivo. Enzymatic mechanisms such as these also have the potential to generate xenokines not only at environmental interfaces but also internally under conditions of tissue damage, inflammation, or remodeling.

In its simplest form, the above model can be summarized as follows. Tissues experience alterations in their external environment or their neighboring cellular environment, through inflammation, tissue damage, UV exposure, developmental remodeling, and changes in microbial populations or oxygen concentration. Each of these changes yields a unique chemistry that produces xenokines through reactive proligand intermediates that activate an AHR-mediated physiological response at the tissue level. In the gut and skin, the response is exemplified by increased barrier integrity, possibly through an influence on resident lymphocyte populations. In the lungs, hyperoxia may be eliciting its own unique chemistry and subsequent xenokine production to adapt to a new higher oxygen tension or microbiological challenges presented by the ambient air at parturition.249,259 In the vascular system, tone and vascular remodeling may respond to systemic release of xenokines or through internal production consequent to cellular remodeling, changes in oxygen tension, or shear stress.260,261

Outputs of Xenokine Signaling

This perspective has emphasized the AHR signaling pathway as an adaptive metabolic system with the plan to dedicate a future perspective on the identities of those target or output genes that might facilitate the physiological and toxicological consequences of receptor activation. While evidence for the significance of this pathway in adaptive metabolic ligand clearance is recounted above, two pieces of evidence show how tightly this adaptive response must be regulated in vivo. In one example, competitive inhibition of CYP1 activity by ligands was shown to influence the signaling of the putative cognate ligand FICZ, presumably by reducing its clearance and increasing its steady state.215 In another example, the global/constitutive expression of the Cyp1a1 gene in the mouse induced a partial phenocopy of the AHR null phenotype, presumably by reducing levels of an essential cognate ligand and/or CYP1A1 substrate.262

It is also important to note here that there is evidence both for and against the centrality of CYP 1s as outputs essential for the physiological or toxicological effects of this receptor. Arguing for their importance as outputs is evidence that such monooxygenases influence the levels of lipid mediators (LMs) derived from polyunsaturated fatty acids or arachidonic acid.12,250 Such LMs could have broad vaso- and immune activities that may ultimately explain aspects of DLC toxicology or phenotypes observed in AHR null models. A separate idea is that some of DLC toxicity or cognate physiology may be mediated through AHR’s role as a sensor of reactive oxygen species or even mediator of an oxidative stress response.263266 While many related ideas have been proposed, one of the longest-standing is that the upregulation of CYP1-dependent monooxygenases leads to an increase in reactive oxygen species, which in turn can influence cellular physiology.267269 Arguing against the importance of CYP 1s as important output genes are observations from our own laboratory, in which the CYP1A1/CYP1A2 upregulation can be genetically dissociated from hallmark phenotypes of the AHR null model (e.g., patent ductus venosus) or classical toxic end points from TCDD exposure.270,271

Finally, the AHR field has been heavily focused on the idea that a cognate ligand exists. While we have argued for the importance of xenokine ligands as well as the adaptive response for xenobiotic and xenokine ligands, the AHR may also function constitutively in some situations. Such a possibility is supported by the expansive evolutionary data described above, where AHR orthologues exist that do not appear to recognize any ligand and appear to signal constitutively and the observation that ligand recognition of PAHs seems to be a vertebrate receptor characteristic. Despite all the data on the hundreds of xenobiotic ligands and the preliminary data related to cognate ligands like FICZ, we must be accepting of the formal possibility that the AHR is a bifunctional transcription factor, a transcription factor with both intrinsic activity and ligand-inducible transactivating properties.

In Closing

The AHR field is abundant with evidence for its role in biological processes as disparate as immunity, vascular biology, stemness, neurosensory signaling, reproduction, cell cycle regulation, and nucleic acid biochemistry. We began this perspective with the objective of developing a comprehensive review of modern thought related to the role of the AHR in normal human physiology. The review evolved into a discussion of the adaptive metabolism paradigm and the promotion of the xenokine model. We conclude recognizing that we have only touched the surface, only having discussed a small portion of the provocative ideas that have been put forth over the past 50 years. As we move forward into the next half-century of AHR research we must continue asking: How can all these AHR-mediated biological processes be true? How can dioxins cause so many distinctive effects? As this effort unfolds, we suspect that the simpler answer will be the correct one and look forward to the development of an understanding of AHR signal transduction that unifies the many scientific disciplines that have been touched by this enigmatic signaling molecule.

Acknowledgments

This review is dedicated in memory of the career of Alan Poland, one of the truly great minds in pharmacology and toxicology. This work was supported by the National Institutes of Health Grants R35-ES028377, T32-ES007015, P30-CA014520, P42-ES007381, and U01-ES1026127, The UW SciMed GRS Program, and The Morgridge Foundation. The authors would like to thank Catherine Stanley of UW Media Solutions for her artwork.

Biographies

Mele Avilla holds a bachelor’s degree in biology and is currently completing her Ph.D. in Molecular and Environmental Toxicology at the University of Wisconsin at Madison. Early on, both in her academic and scientific career, Mele has participated in mammalian signal transduction research related to her interests in human health and disease prevention. Mele is currently employing molecular genetic approaches to understand the role of the Ah receptor (AHR) in murine fertility. Her research focuses on the idea that the AHR is more important to human physiology than just its role in pollutant toxicology and is currently testing the idea that the AHR has an endogenous role in many stages of female reproductive physiology.

Kristen Malecki, PhD, MPH is trained in environmental epidemiology and health policy at Johns Hopkins Bloomberg School of Public Health. The goal of her research is to discover and explain persistent health disparities and their biological underpinnings using multiomic biomarkers of exposure and response (epigenomic, transcriptomic, and microbiomic). As a member of the Molecular Environmental Toxicology Center, she conducts translational research around Ah receptor signaling and immune response to explain environmental and host susceptibility to chronic diseases including cancer. She is also Director for the Survey of the Health of Wisconsin program.

Rachel Wilson graduated from the University of Colorado-Boulder where she majored in Molecular, Cellular, and Developmental Biology. She is currently enrolled in the Molecular and Environmental Toxicology Program at the University of Wisconsin-Madison in the lab of Dr. Christopher Bradfield. Her research interests include genetic mouse model development, endogenous Ah receptor signaling, and PAS protein biology.

Mark Hahn is a Senior Scientist in the Biology Department at the Woods Hole Oceanographic Institution in Woods Hole, MA. He also is a Project Leader in the Boston University Superfund Research Program and the Woods Hole Center for Oceans and Human Health. Hahn received his PhD in Environmental Toxicology (1988) from the University of Rochester School of Medicine. Dr. Hahn’s research interests include the molecular mechanisms and evolution of ligand-activated transcription factors (AHR, NRF2), mechanisms of adaptation to chemicals, developmental toxicity of harmful algal bloom toxins, and the impact of microplastics in the marine environment.

Chris Bradfield received his BSc. in Environmental Toxicology from the University of California at Davis and his Ph.D. in Nutrition from the University of California at Berkeley. After a postdoctoral fellowship in the Laboratory of Alan Poland, he joined Northwestern University and then the University of Wisconsin where he is currently Professor of Oncology and Director of the Biotechnology Center. His awards include a RIVER and a MERIT award from the NIEHS, a Pew Scholar Award in the Biomedical Sciences, a Burroughs Wellcome Foundation Scholar Award in Toxicology, and The Achievement Award from The Society of Toxicology.

The authors declare no competing financial interest.

References

  1. Conney A. H.; et al. (1960) Adaptive increases in drug-metabolizing enzymes induced by phenobarbital and other drugs. Journal of Pharmacology and Experimental Therapeutics 130 (1), 1–8. [PubMed] [Google Scholar]
  2. Nebert D. W.; Gelboin H. V. (1969) In vivo and in vitro induction of aryl hydrocarbon hydroxylase in mammalian cells of different species, tissues, strains, and developmental and hormonal states. Arch. Biochem. Biophys. 134 (1), 76–89. 10.1016/0003-9861(69)90253-7. [DOI] [PubMed] [Google Scholar]
  3. Schmeltz I.; Schlotzhauer W. S. (1968) Benzo[a]pyrene, phenols and other products from pyrolysis of the cigarette additive, (d, l)-menthol. Nature 219 (5152), 370–371. 10.1038/219370b0. [DOI] [PubMed] [Google Scholar]
  4. Hoffman D.; et al. (1972) Chemical studies on tobacco-smoke. 16. Fluoranthenes - quantitative determination in cigarette-smoke, formation by pyrolysis, and tumor-initiating activity. Journal of the National Cancer Institute 49 (4), 1165–1175. [PubMed] [Google Scholar]
  5. Kennaway E. (1955) The identification of a carcinogenic compound in coal-tar. Bmj-British Medical Journal 2 (SEP27), 749–752. 10.1136/bmj.2.4942.749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Tye R.; Stemmer K. L. (1967) Experimental carcinogenesis of the lung. Ii. Influence of phenols in the production of carcinoma. J. Natl. Cancer Inst 39 (2), 175–186. [PubMed] [Google Scholar]
  7. Dat N. D.; Chang M. B. (2017) Review on characteristics of pahs in atmosphere, anthropogenic sources and control technologies. Sci. Total Environ. 609, 682–693. 10.1016/j.scitotenv.2017.07.204. [DOI] [PubMed] [Google Scholar]
  8. Lijinsky W. (1991) The formation and occurrence of polynuclear aromatic-hydrocarbons associated with food. Mutat. Res., Genet. Toxicol. Test. 259 (3–4), 251–261. 10.1016/0165-1218(91)90121-2. [DOI] [PubMed] [Google Scholar]
  9. Nebert D. W.; Gonzalez F. J. (1987) P450 genes - structure, evolution, and regulation. Annu. Rev. Biochem. 56, 945–993. 10.1146/annurev.bi.56.070187.004501. [DOI] [PubMed] [Google Scholar]
  10. Nebert D. W.; et al. (1975) Genetic expression of aryl hydrocarbon hydroxylase activity in the mouse. J. Cell. Physiol. 85 (S1), 393–414. 10.1002/jcp.1040850407. [DOI] [PubMed] [Google Scholar]
  11. Lang M. A.; Nebert D. W. (1981) Structural gene products of the Ah locus. Evidence for many unique p-450-mediated monooxygenase activities reconstituted from 3-methylcholanthrene-treated C57BL/6N mouse liver microsomes. J. Biol. Chem. 256 (23), 12058–12067. [PubMed] [Google Scholar]
  12. Nebert D. W. (2017) Aryl hydrocarbon receptor (ahr): “Pioneer member” of the basic-helix/loop/helix per-arnt-sim (bHLH/PAS) family of “sensors” of foreign and endogenous signals. Prog. Lipid Res. 67, 38–57. 10.1016/j.plipres.2017.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Thomas P. E.; Kouri R. E.; Hutton J. J. (1972) The genetics of aryl hydrocarbon hydroxylase induction in mice: A single gene difference between C57BL/6J and DBA/2J. Biochem. Genet. 6 (2–3), 157–168. 10.1007/BF00486400. [DOI] [PubMed] [Google Scholar]
  14. Nebert D. W.; et al. (1982) The ah locus, a multigene family necessary for survival in a chemically adverse environment - comparison with the immune-system. Adv. Genet. 21, 1–52. 10.1016/S0065-2660(08)60296-5. [DOI] [PubMed] [Google Scholar]
  15. Bult C. J.; et al. (2019) Mouse genome database (MGD) 2019. Nucleic Acids Res. 47 (D1), D801–D806. 10.1093/nar/gky1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Conney A. H. (1967) Pharmacological implications of microsomal enzyme induction. Pharmacological reviews. 19 (3), 317–366. [PubMed] [Google Scholar]
  17. Okey A. B. (1990) Enzyme-induction in the cytochrome-p-450 system. Pharmacol. Ther. 45 (2), 241–298. 10.1016/0163-7258(90)90030-6. [DOI] [PubMed] [Google Scholar]
  18. Poland A.; Knutson J. C. (1982) 2,3,7,8-tetrachlorodibenzo-para-dioxin and related halogenated aromatic-hydrocarbons - examination of the mechanism of toxicity. Annu. Rev. Pharmacol. Toxicol. 22, 517–554. 10.1146/annurev.pa.22.040182.002505. [DOI] [PubMed] [Google Scholar]
  19. Safe S. (1991) Polychlorinated dibenzo-p-dioxins and related compounds: Sources, environmental distribution and risk assessment. Journal of Environmental Science and Health, Part C 9 (2), 261–302. 10.1080/10590509109373409. [DOI] [Google Scholar]
  20. Van Ede K. I.; Van Duursen M. B. M.; Van Den Berg M. (2016) Evaluation of relative effect potencies (reps) for dioxin-like compounds to derive systemic or human-specific tefs to improve human risk assessment. Arch. Toxicol. 90 (6), 1293–1305. 10.1007/s00204-016-1724-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Birnbaum L. S. (1993) EPAs reassessment of dioxin risk - directed health research. Chemosphere 27 (1–3), 469–475. 10.1016/0045-6535(93)90328-3. [DOI] [Google Scholar]
  22. Pohjanvirta R.; Tuomisto J. (1994) Short-term toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in laboratory-animals - effects, mechanisms, and animal-models. Pharmacol. Rev. 46 (4), 483–549. [PubMed] [Google Scholar]
  23. Vandenberg M.; et al. (1994) The toxicokinetics and metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs) and their relevance for toxicity. Crit. Rev. Toxicol. 24 (1), 1–74. 10.3109/10408449409017919. [DOI] [PubMed] [Google Scholar]
  24. Denison M. S.; et al. (2011) Exactly the same but different: Promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol. Sci. 124 (1), 1–22. 10.1093/toxsci/kfr218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Firestone D. (1973) Etiology of chick edema disease. Environ. Health Perspect. 5, 59–66. 10.1289/ehp.730559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Weber R.; et al. (2008) Dioxin- and POP-contaminated sites-contemporary and future relevance and challenges. Environ. Sci. Pollut. Res. 15 (5), 363–393. 10.1007/s11356-008-0024-1. [DOI] [PubMed] [Google Scholar]
  27. White S. S.; Birnbaum L. S. (2009) An overview of the effects of dioxins and dioxin-like compounds on vertebrates, as documented in human and ecological epidemiology. Journal of Environmental Science and Health Part C-Environmental Carcinogenesis & Ecotoxicology Reviews 27 (4), 197–211. 10.1080/10590500903310047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Schecter A.; et al. (2006) Dioxins: An overview. Environ. Res. 101 (3), 419–428. 10.1016/j.envres.2005.12.003. [DOI] [PubMed] [Google Scholar]
  29. Campbell T. C.; Friedman L. (1966) Chick edema factor - some tissue distribution data and toxicologic effects in rat and chick. Exp. Biol. Med. 121 (4), 1283–1287. 10.3181/00379727-121-31029. [DOI] [PubMed] [Google Scholar]
  30. Huang R.; et al. (2019) The human body burden of polychlorinated dibenzo-p-dioxins/furans (PCDD/Fs) and dioxin-like polychlorinated biphenyls (DL-PCBs) in residents’ human milk from guangdong province, china. Toxicol. Res. 8 (4), 552–559. 10.1039/C8TX00337H. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Yang E.; et al. (2018) Exposure of dioxin-like chemicals in participants of the anniston community health survey follow-up. Sci. Total Environ. 637–638, 881–891. 10.1016/j.scitotenv.2018.05.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Muzembo B. A.; et al. (2019) Dioxins levels in human blood after implementation of measures against dioxin exposure in japan. Environ. Health Prev. Med. 24 (1), 6. 10.1186/s12199-018-0755-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Srogi K. (2008) Levels and congener distributions of PCDDs, PCDFs and dioxin-like PCBs in environmental and human samples: A review. Environ. Chem. Lett. 6 (1), 1–28. 10.1007/s10311-007-0105-2. [DOI] [Google Scholar]
  34. Okey A. B. (2007) Special contribution - an aryl hydrocarbon receptor odyssey to the shores of toxicology: The Deichmann Lecture, International Congress of Toxicology-XI. Toxicol. Sci. 98 (1), 5–38. 10.1093/toxsci/kfm096. [DOI] [PubMed] [Google Scholar]
  35. Poland A.; Glover E.; Kende A. S. (1976) Stereospecific, high affinity binding of 2,3,7,8-tetrachlorodibenzo-para-dioxin by hepatic cytosol - evidence that binding species is receptor for induction of aryl-hydrocarbon hydroxylase. J. Biol. Chem. 251 (16), 4936–4946. [PubMed] [Google Scholar]
  36. Schmidt J. V.; Bradfield C. A. (1996) Ah receptor signaling pathways. Annu. Rev. Cell Dev. Biol. 12, 55–89. 10.1146/annurev.cellbio.12.1.55. [DOI] [PubMed] [Google Scholar]
  37. Poland A.; Glover E. (1973) Studies on the mechanism of toxicity of the chlorinated dibenzo-p-dioxins. Environ. Health Perspect. 5, 245–251. 10.1289/ehp.7305245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Poland A.; Glover E. (1980) 2,3,7,8-tetrachlorodibenzo-p-dioxin - segregation of toxicity with the ah locus. Mol. Pharmacol. 17 (1), 86–94. [PubMed] [Google Scholar]
  39. Swanson H. I.; Bradfield C. A. (1993) The Ah-receptor - genetics, structure and function. Pharmacogenetics 3 (5), 213–230. 10.1097/00008571-199310000-00001. [DOI] [PubMed] [Google Scholar]
  40. Gasiewicz T. A.; Henry E.C. (2011) History of research on the AHR. The AH Receptor in Biology and Toxicology 1–32. 10.1002/9781118140574.ch1. [DOI] [Google Scholar]
  41. Whitlock J. P.; et al. (1989) Induction of hepatic cytochrome-p450 gene-expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Molecular Biology & Medicine 6 (2), 169–178. [PubMed] [Google Scholar]
  42. Nebert D. W.; et al. (2004) Role of aryl hydrocarbon receptor-mediated induction of the Cyp1 enzymes in environmental toxicity and cancer. J. Biol. Chem. 279 (23), 23847–23850. 10.1074/jbc.R400004200. [DOI] [PubMed] [Google Scholar]
  43. Genter M. B.; et al. (2006) Comparison of mouse hepatic mitochondrial versus microsomal cytochromes p450 following TCDD treatment. Biochem. Biophys. Res. Commun. 342 (4), 1375–1381. 10.1016/j.bbrc.2006.02.121. [DOI] [PubMed] [Google Scholar]
  44. Kawajiri K.; Fujii-Kuriyama Y. (2007) Cytochrome p450 gene regulation and physiological functions mediated by the aryl hydrocarbon receptor. Arch. Biochem. Biophys. 464 (2), 207–212. 10.1016/j.abb.2007.03.038. [DOI] [PubMed] [Google Scholar]
  45. Nebert D. W.; Dalton T. P. (2006) The role of cytochrome p450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat. Rev. Cancer 6 (12), 947–960. 10.1038/nrc2015. [DOI] [PubMed] [Google Scholar]
  46. Kawajiri K.; Fujii-Kuriyama Y. (2017) The aryl hydrocarbon receptor: A multifunctional chemical sensor for host defense and homeostatic maintenance. Exp. Anim. 66 (2), 75–89. 10.1538/expanim.16-0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Whitlock J. P. (1999) Induction of cytochrome p4501a1. Annu. Rev. Pharmacol. Toxicol. 39, 103–125. 10.1146/annurev.pharmtox.39.1.103. [DOI] [PubMed] [Google Scholar]
  48. Denison M. S.; Fisher J. M.; Whitlock J. P. (1988) The DNA recognition site for the dioxin-Ah receptor complex - nucleotide-sequence and functional-analysis. J. Biol. Chem. 263 (33), 17221–17224. [PubMed] [Google Scholar]
  49. Neuhold L. A.; et al. (1986) Dioxin-inducible enhancer region upstream from the mouse p1450 gene and interaction with a heterologous sv40 promoter. DNA 5 (5), 403–411. 10.1089/dna.1986.5.403. [DOI] [PubMed] [Google Scholar]
  50. Hoffman E. C.; et al. (1991) Cloning of a factor required for activity of the Ah (dioxin) receptor. Science 252 (5008), 954–958. 10.1126/science.1852076. [DOI] [PubMed] [Google Scholar]
  51. Ema M.; et al. (1992) Cdna cloning and structure of mouse putative Ah receptor. Biochem. Biophys. Res. Commun. 184 (1), 246–253. 10.1016/0006-291X(92)91185-S. [DOI] [PubMed] [Google Scholar]
  52. Burbach K. M.; Poland A.; Bradfield C. A. (1992) Cloning of the Ah-receptor cDNA reveals a distinctive ligand-activated transcription factor. Proc. Natl. Acad. Sci. U. S. A. 89 (17), 8185–8189. 10.1073/pnas.89.17.8185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Crews S. T. (1998) Control of cell lineage-specific development and transcription by bHLH-PAS proteins. Genes Dev. 12 (5), 607–620. 10.1101/gad.12.5.607. [DOI] [PubMed] [Google Scholar]
  54. Gu Y. Z.; Hogenesch J. B.; Bradfield C. A. (2000) The PAS superfamily: Sensors of environmental and developmental signals. Annu. Rev. Pharmacol. Toxicol. 40, 519–561. 10.1146/annurev.pharmtox.40.1.519. [DOI] [PubMed] [Google Scholar]
  55. Reyes H.; Reiszporszasz S.; Hankinson O. (1992) Identification of the Ah receptor nuclear translocator protein (ARNT) as a component of the DNA-binding form of the Ah receptor. Science 256 (5060), 1193–1195. 10.1126/science.256.5060.1193. [DOI] [PubMed] [Google Scholar]
  56. Jones S. (2004) An overview of the basic helix-loop-helix proteins. Genome Biology 5 (6), 226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hankinson O. (1995) The aryl-hydrocarbon receptor complex. Annu. Rev. Pharmacol. Toxicol. 35, 307–340. 10.1146/annurev.pa.35.040195.001515. [DOI] [PubMed] [Google Scholar]
  58. Swanson H. I.; Chan W. K.; Bradfield C. A. (1995) DNA-binding specificities and pairing rules of the Ah receptor, ARNT, and SIM proteins. J. Biol. Chem. 270 (44), 26292–26302. 10.1074/jbc.270.44.26292. [DOI] [PubMed] [Google Scholar]
  59. Reiszporszasz S.; et al. (1994) Identification of functional domains of the aryl-hydrocarbon receptor nuclear translocator protein (ARNT). Mol. Cell. Biol. 14 (9), 6075–6086. 10.1128/MCB.14.9.6075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. McIntosh B. E.; Hogenesch J. B.; Bradfield C. A. (2010) Mammalian PER-ARNT-SIM proteins in environmental adaptation. Annu. Rev. Physiol. 72, 625–45. 10.1146/annurev-physiol-021909-135922. [DOI] [PubMed] [Google Scholar]
  61. Henry J. T.; Crosson S. (2011) Ligand-binding PAS domains in a genomic, cellular, and structural context. Annu. Rev. Microbiol. 65, 261–286. 10.1146/annurev-micro-121809-151631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Dolwick K. M.; Swanson H. I.; Bradfield C. A. (1993) In-vitro analysis of Ah receptor domains involved in ligand-activated DNA recognition. Proc. Natl. Acad. Sci. U. S. A. 90 (18), 8566–8570. 10.1073/pnas.90.18.8566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Fukunaga B. N.; et al. (1995) Identification of functional domains of the aryl-hydrocarbon receptor. J. Biol. Chem. 270 (49), 29270–29278. 10.1074/jbc.270.49.29270. [DOI] [PubMed] [Google Scholar]
  64. Antonsson C.; et al. (1995) Distinct roles of the molecular chaperone hsp90 in modulating dioxin receptor function via the basic helix-loop-helix and PAS domains. Mol. Cell. Biol. 15 (2), 756–765. 10.1128/MCB.15.2.756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Schulte K. W.; et al. (2017) Structural basis for aryl hydrocarbon receptor-mediated gene activation. Structure 25 (7), 1025. 10.1016/j.str.2017.05.008. [DOI] [PubMed] [Google Scholar]
  66. Swanson H. I. (2002) DNA binding and protein interactions of the ahr/arnt heterodimer that facilitate gene activation. Chem.-Biol. Interact. 141 (1–2), 63–76. 10.1016/S0009-2797(02)00066-2. [DOI] [PubMed] [Google Scholar]
  67. Seok S. H.; et al. (2017) Structural hierarchy controlling dimerization and target DNA recognition in the ahr transcriptional complex. Proc. Natl. Acad. Sci. U. S. A. 114 (21), 5431–5436. 10.1073/pnas.1617035114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Scheuermann T. H.; et al. (2009) Artificial ligand binding within the hif2α pas-b domain of the hif2 transcription factor. Proc. Natl. Acad. Sci. U. S. A. 106 (2), 450–455. 10.1073/pnas.0808092106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Perdew G. H.; Bradfield C. A. (1996) Mapping the 90 kda heat shock protein binding region of the Ah receptor. IUBMB Life 39 (3), 589–593. 10.1080/15216549600201651. [DOI] [PubMed] [Google Scholar]
  70. McGuire J.; et al. (2001) Definition of a dioxin receptor mutant that is a constitutive activator of transcription - delineation of overlapping repression and ligand binding functions within the pas domain. J. Biol. Chem. 276 (45), 41841–41849. 10.1074/jbc.M105607200. [DOI] [PubMed] [Google Scholar]
  71. Berg P.; Pongratz I. (2001) Differential usage of nuclear export sequences regulates intracellular localization of the dioxin (aryl hydrocarbon) receptor. J. Biol. Chem. 276 (46), 43231–43238. 10.1074/jbc.M105261200. [DOI] [PubMed] [Google Scholar]
  72. Beischlag T. V.; et al. (2008) The aryl hydrocarbon receptor complex and the control of gene expression. Crit. Rev. Eukaryotic Gene Expression 18 (3), 207–250. 10.1615/CritRevEukarGeneExpr.v18.i3.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Jain S.; et al. (1994) Potent transactivation domains of the Ah receptor and the Ah receptor nuclear translocator map to their carboxyl termini. J. Biol. Chem. 269 (50), 31518–31524. [PubMed] [Google Scholar]
  74. Pohjanvirta R. (2009) The structure of the Ah receptor transactivation domain as a determinant of dioxin sensitivity. Toxicol. Lett. 189, S54–S54. 10.1016/j.toxlet.2009.06.077. [DOI] [Google Scholar]
  75. Watt K.; et al. (2005) Induced alpha-helix structure in the aryl hydrocarbon receptor transactivation domain modulates protein-protein interactions. Biochemistry 44 (2), 734–743. 10.1021/bi0487701. [DOI] [PubMed] [Google Scholar]
  76. Kumar M. B.; et al. (2001) The q-rich subdomain of the human ah receptor transactivation domain is required for dioxin-mediated transcriptional activity. J. Biol. Chem. 276 (45), 42302–42310. 10.1074/jbc.M104798200. [DOI] [PubMed] [Google Scholar]
  77. Jones L. C.; Whitlock J. P. (2001) Dioxin-inducible transactivation in a chromosomal setting. J. Biol. Chem. 276 (27), 25037–25042. 10.1074/jbc.M102910200. [DOI] [PubMed] [Google Scholar]
  78. Hankinson O. (2011) The ahr/arnt dimer and transcriptional coactivators. The AH Receptor in Biology and Toxicology 93–100. 10.1002/9781118140574.ch6. [DOI] [Google Scholar]
  79. Ko H. P.; et al. (1996) Dioxin-induced cyp1a1 transcription in vivo: The aromatic hydrocarbon receptor mediates transactivation, enhancer-promoter communication, and changes in chromatin structure. Mol. Cell. Biol. 16 (1), 430–436. 10.1128/MCB.16.1.430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Ma Q. (2001) Induction of Cyp1a1. The AHR/DRE paradigm: Transcription, receptor regulation, and expanding biological roles. Curr. Drug Metab. 2 (2), 149–164. 10.2174/1389200013338603. [DOI] [PubMed] [Google Scholar]
  81. Carver L. A.; Bradfield C. A. (1997) Ligand-dependent interaction of the aryl hydrocarbon receptor with a novel immunophilin homologin vivo. J. Biol. Chem. 272 (17), 11452–11456. 10.1074/jbc.272.17.11452. [DOI] [PubMed] [Google Scholar]
  82. Petrulis J. R.; Perdew G. H. (2002) The role of chaperone proteins in the aryl hydrocarbon receptor core complex. Chem.-Biol. Interact. 141 (1), 25–40. 10.1016/S0009-2797(02)00064-9. [DOI] [PubMed] [Google Scholar]
  83. Carlson D. B.; Perdew G. H. (2002) A dynamic role for the ah receptor in cell signaling? Insights from a diverse group of Ah receptor interacting proteins. J. Biochem. Mol. Toxicol. 16 (6), 317–325. 10.1002/jbt.10051. [DOI] [PubMed] [Google Scholar]
  84. Kazlauskas A.; Poellinger L.; Pongratz I. (1999) Evidence that the co-chaperone p23 regulates ligand responsiveness of the dioxin (aryl hydrocarbon) receptor. J. Biol. Chem. 274 (19), 13519–13524. 10.1074/jbc.274.19.13519. [DOI] [PubMed] [Google Scholar]
  85. Ma Q.; Whitlock J. P. (1997) A novel cytoplasmic protein that interacts with the Ah receptor, contains tetratricopeptide repeat motifs, and augments the transcriptional response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. J. Biol. Chem. 272 (14), 8878–8884. 10.1074/jbc.272.14.8878. [DOI] [PubMed] [Google Scholar]
  86. Kazlauskas A.; et al. (2001) The hsp90 chaperone complex regulates intracellular localization of the dioxin receptor. Mol. Cell. Biol. 21 (7), 2594–2607. 10.1128/MCB.21.7.2594-2607.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Carver L. A.; et al. (1998) Characterization of the Ah receptor-associated protein, Ara9. J. Biol. Chem. 273 (50), 33580–33587. 10.1074/jbc.273.50.33580. [DOI] [PubMed] [Google Scholar]
  88. Hollingshead B. D.; Petrulis J. R.; Perdew G. H. (2004) The aryl hydrocarbon (Ah) receptor transcriptional regulator hepatitis b virus x-associated protein 2 antagonizes p23 binding to Ah receptor-hsp90 complexes and is dispensable for receptor function. J. Biol. Chem. 279 (44), 45652–45661. 10.1074/jbc.M407840200. [DOI] [PubMed] [Google Scholar]
  89. Ko H. P.; et al. (1997) Transactivation domains facilitate promoter occupancy for the dioxin-inducible Cyp1a1 gene in vivo. Mol. Cell. Biol. 17 (7), 3497–3507. 10.1128/MCB.17.7.3497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Hankinson O. (2005) Role of coactivators in transcriptional activation by the aryl hydrocarbon receptor. Arch. Biochem. Biophys. 433 (2), 379–386. 10.1016/j.abb.2004.09.031. [DOI] [PubMed] [Google Scholar]
  91. Taylor R. T.; et al. (2009) Roles of coactivator proteins in dioxin induction of Cyp1a1 and Cyp1b1 in human breast cancer cells. Toxicol. Sci. 107 (1), 1–8. 10.1093/toxsci/kfn217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Kollara A.; Brown T. J. (2006) Functional interaction of nuclear receptor coactivator 4 with aryl hydrocarbon receptor. Biochem. Biophys. Res. Commun. 346 (2), 526–534. 10.1016/j.bbrc.2006.05.148. [DOI] [PubMed] [Google Scholar]
  93. Kumar M. B.; Tarpey R. W.; Perdew G. H. (1999) Differential recruitment of coactivator rip140 by Ah and estrogen receptors - absence of a role for LXXL motifs. J. Biol. Chem. 274 (32), 22155–22164. 10.1074/jbc.274.32.22155. [DOI] [PubMed] [Google Scholar]
  94. Kobayashi A.; et al. (1997) Cbp/p300 functions as a possible transcriptional coactivator of Ah receptor nuclear translocator (ARNT). J. Biochem. 122 (4), 703–710. 10.1093/oxfordjournals.jbchem.a021812. [DOI] [PubMed] [Google Scholar]
  95. Flaveny C. A.; Murray I. A.; Perdew G. H. (2010) Differential gene regulation by the human and mouse aryl hydrocarbon receptor. Toxicol. Sci. 114 (2), 217–225. 10.1093/toxsci/kfp308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Wang S.; et al. (2004) Role of mediator in transcriptional activation by the aryl hydrocarbon receptor. J. Biol. Chem. 279 (14), 13593–13600. 10.1074/jbc.M312274200. [DOI] [PubMed] [Google Scholar]
  97. Nguyen T. A.; et al. (1999) Interactions of nuclear receptor coactivator/corepressor proteins with the aryl hydrocarbon receptor complex. Arch. Biochem. Biophys. 367 (2), 250–257. 10.1006/abbi.1999.1282. [DOI] [PubMed] [Google Scholar]
  98. Baba T.; et al. (2001) Structure and expression of the Ah receptor repressor gene. J. Biol. Chem. 276 (35), 33101–33110. 10.1074/jbc.M011497200. [DOI] [PubMed] [Google Scholar]
  99. Hahn M. E.; Allan L. L.; Sherr D. H. (2009) Regulation of constitutive and inducible AHR signaling: Complex interactions involving the AHR repressor. Biochem. Pharmacol. 77 (4), 485–497. 10.1016/j.bcp.2008.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Oshima M.; et al. (2007) Molecular mechanism of transcriptional repression of ahr repressor involving ankra2, hdac4, and hdac5. Biochem. Biophys. Res. Commun. 364 (2), 276–282. 10.1016/j.bbrc.2007.09.131. [DOI] [PubMed] [Google Scholar]
  101. Evans B. R.; et al. (2008) Repression of aryl hydrocarbon receptor (ahr) signaling by ahr repressor: Role of DNA binding and competition for ahr nuclear translocator. Mol. Pharmacol. 73 (2), 387–398. 10.1124/mol.107.040204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Giannone J. V.; et al. (1998) Prolonged depletion of Ah receptor without alteration of receptor mRNA levels after treatment of cells in culture with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Biochem. Pharmacol. 55 (4), 489–497. 10.1016/S0006-2952(97)00493-0. [DOI] [PubMed] [Google Scholar]
  103. Roberts B. J.; Whitelaw M. L. (1999) Degradation of the basic helix-loop-helix/per-arnt-sim homology domain dioxin receptor via the ubiquitin/proteasome pathway. J. Biol. Chem. 274 (51), 36351–36356. 10.1074/jbc.274.51.36351. [DOI] [PubMed] [Google Scholar]
  104. Pollenz R. S. (2002) The mechanism of Ah receptor protein down-regulation (degradation) and its impact on Ah receptor-mediated gene regulation. Chem.-Biol. Interact. 141 (1–2), 41–61. 10.1016/S0009-2797(02)00065-0. [DOI] [PubMed] [Google Scholar]
  105. Santiago-Josefat B.; et al. (2001) Proteasome inhibition induces nuclear translocation and transcriptional activation of the dioxin receptor in mouse embryo primary fibroblasts in the absence of xenobiotics. Mol. Cell. Biol. 21 (5), 1700–1709. 10.1128/MCB.21.5.1700-1709.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Grimaldi G.; Rajendra S.; Matthews J. (2018) The aryl hydrocarbon receptor regulates the expression of tiparp and its cis long non-coding RNA, tiparp-as1. Biochem. Biophys. Res. Commun. 495 (3), 2356–2362. 10.1016/j.bbrc.2017.12.113. [DOI] [PubMed] [Google Scholar]
  107. MacPherson L.; et al. (2014) Aryl hydrocarbon receptor repressor and tiparp (artd14) use similar, but also distinct mechanisms to repress aryl hydrocarbon receptor signaling. Int. J. Mol. Sci. 15 (5), 7939–7957. 10.3390/ijms15057939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Perdew G. H.; Hollenback C. E. (1990) Analysis of photoaffinity-labeled aryl-hydrocarbon receptor heterogeneity by 2-dimensional gel-electrophoresis. Biochemistry 29 (26), 6210–6214. 10.1021/bi00478a014. [DOI] [PubMed] [Google Scholar]
  109. Mahon M. J.; Gasiewicz T. A. (1995) Ah receptor phosphorylation - localization of phosphorylation sites to the c-terminal half of the protein. Arch. Biochem. Biophys. 318 (1), 166–174. 10.1006/abbi.1995.1217. [DOI] [PubMed] [Google Scholar]
  110. Pongratz I.; et al. (1991) Inhibition of the specific DNA-binding activity of the dioxin receptor by phosphatase treatment. J. Biol. Chem. 266 (25), 16813–16817. [PubMed] [Google Scholar]
  111. Chen Y. H.; Tukey R. H. (1996) Protein kinase c modulates regulation of the Cyp1a1 gene by the aryl hydrocarbon receptor. J. Biol. Chem. 271 (42), 26261–26266. 10.1074/jbc.271.42.26261. [DOI] [PubMed] [Google Scholar]
  112. Tkachenko A.; et al. (2016) The q-rich/pst domain of the AHR regulates both ligand-induced nuclear transport and nucleocytoplasmic shuttling. Sci. Rep. 6, 32009. 10.1038/srep32009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Pollenz R. S.; Barbour E. R. (2000) Analysis of the complex relationship between nuclear export and aryl hydrocarbon receptor-mediated gene regulation. Mol. Cell. Biol. 20 (16), 6095–6104. 10.1128/MCB.20.16.6095-6104.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Levine S. L.; et al. (2000) A tetratricopeptide repeat half-site in the aryl hydrocarbon receptor is important for DNA binding and trans-activation potential. Mol. Pharmacol. 58 (6), 1517–1524. 10.1124/mol.58.6.1517. [DOI] [PubMed] [Google Scholar]
  115. Jackson D. P.; Joshi A. D.; Elferink C. J. (2015) Ah receptor pathway intricacies; signaling through diverse protein partners and DNA-motifs. Toxicol. Res. 4 (5), 1143–1158. 10.1039/C4TX00236A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Hestermann E. V.; Brown M. (2003) Agonist and chemopreventative ligands induce differential transcriptional cofactor recruitment by aryl hydrocarbon receptor. Mol. Cell. Biol. 23 (21), 7920–7925. 10.1128/MCB.23.21.7920-7925.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Powis M.; Celius T.; Matthews J. (2011) Differential ligand-dependent activation and a role for y322 in aryl hydrocarbon receptor-mediated regulation of gene expression. Biochem. Biophys. Res. Commun. 410 (4), 859–865. 10.1016/j.bbrc.2011.06.079. [DOI] [PubMed] [Google Scholar]
  118. Van den Berg M., and Birnbaum L. S.; et al. (2006) The 2005 World Health Organization reevaluation of human and mammailan toxic equivalency factors for dioxins and dioxin-like compounds. Toxicol. Sci. 223–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Barnes D.; et al. (1991) Toxicity equivalency factors for PCBs?. Qual Assur 1 (1), 70–81. [PubMed] [Google Scholar]
  120. Barnes D.; et al. (1991) Toxicity equivalents and EPA’s risk assessment of 2,3,7,8-TCDD. Sci. Total Environ. 104, 73–86. 10.1016/0048-9697(91)90008-3. [DOI] [PubMed] [Google Scholar]
  121. Safe S. (1997) Limitations of the toxic equivalency factor approach for risk assessment of TCDD and related compounds. Teratog., Carcinog., Mutagen. 17 (4–5), 285–304. . [DOI] [PubMed] [Google Scholar]
  122. Safe S. H. (1998) Development validation and problems with the toxic equivalency factor approach for risk assessment of dioxins and related compounds. J. Anim. Sci. 76 (1), 134–141. 10.2527/1998.761134x. [DOI] [PubMed] [Google Scholar]
  123. Birnbaum L. S.; DeVito M. J. (1995) Use of toxic equivalency factors for risk assessment for dioxins and related compounds. Toxicology 105 (2–3), 391–401. 10.1016/0300-483X(95)03237-A. [DOI] [PubMed] [Google Scholar]
  124. Pohjanvirta R.; et al. (1999) Physicochemical differences in the Ah receptors of the most TCDD-susceptible and the most TCDD-resistant rat strains. Toxicol. Appl. Pharmacol. 155 (1), 82–95. 10.1006/taap.1998.8565. [DOI] [PubMed] [Google Scholar]
  125. Gasiewicz T. A.; Rucci G. (1984) Cytosolic receptor for 2,3,7,8-tetrachlorodibenzo-para-dioxin - evidence for a homologous nature among various mammalian-species. Mol. Pharmacol. 26 (1), 90–98. [PubMed] [Google Scholar]
  126. Bank P. A.; et al. (1992) Species-specific binding of transformed Ah receptor to a dioxin responsive transcriptional enhancer. Eur. J. Pharmacol., Environ. Toxicol. Pharmacol. Sect. 228 (2–3), 85–94. 10.1016/0926-6917(92)90016-6. [DOI] [PubMed] [Google Scholar]
  127. Poland A.; Glover E.; Bradfield C. A. (1991) Characterization of polyclonal antibodies to the Ah receptor prepared by immunization with a synthetic peptide hapten. Mol. Pharmacol. 39 (1), 20–26. [PubMed] [Google Scholar]
  128. Hahn M. E.; et al. (1994) Photoaffinity-labeling of the Ah receptor - phylogenetic survey of diverse vertebrate and invertebrate species. Arch. Biochem. Biophys. 310 (1), 218–228. 10.1006/abbi.1994.1160. [DOI] [PubMed] [Google Scholar]
  129. Poland A.; et al. (1986) Photoaffinity-labeling of the Ah receptor. J. Biol. Chem. 261 (14), 6352–6365. [PubMed] [Google Scholar]
  130. Dolwick K. M.; et al. (1993) Cloning and expression of a human Ah receptor cDNA. Mol. Pharmacol. 44 (5), 911–917. [PubMed] [Google Scholar]
  131. Hahn M. E. (2002) Aryl hydrocarbon receptors: Diversity and evolution. Chem.-Biol. Interact. 141 (1–2), 131–160. 10.1016/S0009-2797(02)00070-4. [DOI] [PubMed] [Google Scholar]
  132. Poland A.; Glover E.; Taylor B. A. (1987) The murine Ah locus - a new allele and mapping to chromosome-12. Mol. Pharmacol. 32 (4), 471–478. [PubMed] [Google Scholar]
  133. Lebeau M. M.; et al. (1994) Chromosomal localization of the human AHR locus encoding the structural gene for the Ah receptor to 7p21- p15. Cytogenet. Genome Res. 66 (3), 172–176. 10.1159/000133694. [DOI] [PubMed] [Google Scholar]
  134. Schmidt J. V.; Carver L. A.; Bradfield C. A. (1993) Molecular characterization of the murine AHR gene - organization, promoter analysis, and chromosomal assignment. J. Biol. Chem. 268 (29), 22203–22209. [PubMed] [Google Scholar]
  135. Bennett P.; Ramsden D. B.; Williams A. C. (1996) Complete structural characterisation of the human aryl hydrocarbon receptor gene. Mol. Pathol. 49 (1), M12–M16. 10.1136/mp.49.1.M12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Poland A.; Glover E. (1987) Variation in the molecular mass of the Ah receptor among vertebrate species and strains of rats. Biochem. Biophys. Res. Commun. 146 (3), 1439–1449. 10.1016/0006-291X(87)90811-4. [DOI] [PubMed] [Google Scholar]
  137. Chang C. Y.; et al. (1993) 10 nucleotide differences, 5 of which cause amino-acid changes, are associated with the Ah receptor locus polymorphism of C57BL/6 and DBA/2 mice. Pharmacogenetics 3 (6), 312–321. 10.1097/00008571-199312000-00005. [DOI] [PubMed] [Google Scholar]
  138. Poland A.; Palen D.; Glover E. (1994) Analysis of the 4 alleles of the murine aryl-hydrocarbon receptor. Mol. Pharmacol. 46 (5), 915–921. [PubMed] [Google Scholar]
  139. Ema M.; et al. (1994) Dioxin binding activities of polymorphic forms of mouse and human arylhydrocarbon receptors. J. Biol. Chem. 269 (44), 27337–27343. [PubMed] [Google Scholar]
  140. Poland A.; Glover E. (1990) Characterization and strain distribution pattern of the murine Ah receptor specified by the ahd and ahb-3 alleles. Mol. Pharmacol. 38 (3), 306–312. [PubMed] [Google Scholar]
  141. Xing Y.; et al. (2012) Identification of the Ah-receptor structural determinants for ligand preferences. Toxicol. Sci. 129, 86–97. 10.1093/toxsci/kfs194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Fraccalvieri D.; et al. (2013) Comparative analysis of homology models of the Ah receptor ligand binding domain: Verification of structure-function predictions by site-directed mutagenesis of a nonfunctional receptor. Biochemistry 52 (4), 714–725. 10.1021/bi301457f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Goryo K.; et al. (2007) Identification of amino acid residues in the Ah receptor involved in ligand binding. Biochem. Biophys. Res. Commun. 354 (2), 396–402. 10.1016/j.bbrc.2006.12.227. [DOI] [PubMed] [Google Scholar]
  144. Pandini A.; et al. (2009) Detection of the TCDD binding-fingerprint within the Ah receptor ligand binding domain by structurally driven mutagenesis and functional analysis. Biochemistry 48 (25), 5972–5983. 10.1021/bi900259z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Moriguchi T.; et al. (2003) Distinct response to dioxin in an arylhydrocarbon receptor (ahr)-humanized mouse. Proc. Natl. Acad. Sci. U. S. A. 100 (10), 5652–5657. 10.1073/pnas.1037886100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Kociba R. (1978) Results of a 2-year chronic toxicity and oncogenicity study of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in rats. Toxicol. Appl. Pharmacol. 279–303. [DOI] [PubMed] [Google Scholar]
  147. Moffat I. D.; et al. (2007) Aryl hydrocarbon receptor splice variants in the dioxin-resistant rat: Tissue expression and transactivational activity. Mol. Pharmacol. 72 (4), 956–966. 10.1124/mol.107.037218. [DOI] [PubMed] [Google Scholar]
  148. Pohjanvirta R.; et al. (1998) Point mutation in intron sequence causes altered carboxyl-terminal structure in the aryl hydrocarbon receptor of the most 2,3,7,8-tetrachlorodibenzo-p-dioxin-resistant rat strain. Mol. Pharmacol. 54 (1), 86–93. 10.1124/mol.54.1.86. [DOI] [PubMed] [Google Scholar]
  149. Nukaya M.; Moran S.; Bradfield C. A. (2009) The role of the dioxin-responsive element cluster between the Cyp1a1 and Cyp1a2 loci in aryl hydrocarbon receptor biology. Proc. Natl. Acad. Sci. U. S. A. 106 (12), 4923–4928. 10.1073/pnas.0809613106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Boutros P. C.; et al. (2011) Hepatic transcriptomic responses to TCDD in dioxin-sensitive and dioxin-resistant rats during the onset of toxicity. Toxicol. Appl. Pharmacol. 251 (2), 119–129. 10.1016/j.taap.2010.12.010. [DOI] [PubMed] [Google Scholar]
  151. Wilcke W. (2000) Polycyclic aromatic hydrocarbons (PAHs) in soil - a review. J. Plant Nutr. Soil Sci. 163 (3), 229–248. . [DOI] [Google Scholar]
  152. Baerson S. R.; et al. (2005) Detoxification and transcriptome response in arabidopsis seedlings exposed to the allelochemical benzoxazolin-2(3h)-one. J. Biol. Chem. 280 (23), 21867–21881. 10.1074/jbc.M500694200. [DOI] [PubMed] [Google Scholar]
  153. Denison M. S.; Nagy S. R. (2003) Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu. Rev. Pharmacol. Toxicol. 43, 309–334. 10.1146/annurev.pharmtox.43.100901.135828. [DOI] [PubMed] [Google Scholar]
  154. Carver L. A.; Hogenesch J.B.; Bradfield C.A. (1994) Tissue specific expression of the rat Ah-receptor and ARNT mRNAs. Nucleic Acids Res. 22 (15), 3038–3044. 10.1093/nar/22.15.3038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Gasiewicz T. A.; Ness W. C.; Rucci G. (1984) Ontogeny of the cytosolic receptor for 2,3,7,8-tetrachlorodibenzo-p-dioxin in rat liver, lung, and thymus. Biochem. Biophys. Res. Commun. 118 (1), 183–190. 10.1016/0006-291X(84)91084-2. [DOI] [PubMed] [Google Scholar]
  156. Mason M. E.; Okey A. B. (1982) Cytosolic and nuclear binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin to the Ah receptor in extra-hepatic tissues of rats and mice. Eur. J. Biochem. 123 (1), 209–215. 10.1111/j.1432-1033.1982.tb06518.x. [DOI] [PubMed] [Google Scholar]
  157. Wu C. (2009) BIOGPS: An extensible and customizable portal for querying and organizing gene annotation resources. Genome Biol. 10 (11), R130. 10.1186/gb-2009-10-11-r130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Thul P. J.; Lindskog C. (2018) The human protein atlas: A spatial map of the human proteome. Protein Sci. 27 (1), 233–244. 10.1002/pro.3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Yue F.; et al. (2014) A comparative encyclopedia of DNA elements in the mouse genome. Nature 515 (7527), 355–364. 10.1038/nature13992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Schmidt A. (2015) Only humans have human placentas: Molecular differences between mice and humans. J. Reprod. Immunol. 108, 65–71. 10.1016/j.jri.2015.03.001. [DOI] [PubMed] [Google Scholar]
  161. Jain S.; et al. (1998) Expression of arnt, arnt2, hif1 alpha, hif2 alpha and Ah receptor mRNAs in the developing mouse. Mech. Dev. 73 (1), 117–123. 10.1016/S0925-4773(98)00038-0. [DOI] [PubMed] [Google Scholar]
  162. Walker M. K.; et al. (2000) Molecular characterization and developmental expression of the aryl hydrocarbon receptor from the chick embryo. Comp. Biochem. Physiol., Part C: Pharmacol., Toxicol. Endocrinol. 126 (3), 305–319. 10.1016/S0742-8413(00)00119-5. [DOI] [PubMed] [Google Scholar]
  163. Petersen S. L.; et al. (2000) Distribution of mrnas encoding the arylhydrocarbon receptor, arylhydrocarbon receptor nuclear translocator, and arylhydrocarbon receptor nuclear translocator-2 in the rat brain and brainstem. J. Comp. Neurol. 427 (3), 428–439. . [DOI] [PubMed] [Google Scholar]
  164. Kimura E.; Tohyama C. (2017) Embryonic and postnatal expression of aryl hydrocarbon receptor mrna in mouse brain. Front. Neuroanat. 11, 4. 10.3389/fnana.2017.00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Baba T.; et al. (2005) Intrinsic function of the aryl hydrocarbon (dioxin) receptor as a key factor in female reproduction. Mol. Cell. Biol. 25 (22), 10040–51. 10.1128/MCB.25.22.10040-10051.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Ikuta T.; et al. (2016) Tissue distribution of aryl hydrocarbon receptor in the intestine: Implication of putative roles in tumor suppression. Exp. Cell Res. 343 (2), 126–134. 10.1016/j.yexcr.2016.03.012. [DOI] [PubMed] [Google Scholar]
  167. Koliopanos A. (2002) Increased arylhydrocarbon receptor expression offers a potential therapeutic target for pancreatic cancer. Oncogene 21 (39), 6059–6070. 10.1038/sj.onc.1205633. [DOI] [PubMed] [Google Scholar]
  168. Tscheudschilsuren G.; et al. (1999) Induction of arylhydrocarbon receptor expression in embryoblast cells of rabbit preimplantation blastocysts upon degeneration of rauber’s polar trophoblast. Toxicol. Appl. Pharmacol. 157 (2), 125–133. 10.1006/taap.1999.8667. [DOI] [PubMed] [Google Scholar]
  169. Schmidt J. V.; et al. (1996) Characterization of a murine Ahr null allele: Involvement of the Ah receptor in hepatic growth and development. Proc. Natl. Acad. Sci. U. S. A. 93 (13), 6731–6736. 10.1073/pnas.93.13.6731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Mimura J.; et al. (1997) Loss of teratogenic response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in mice lacking the Ah (dioxin) receptor. Genes to Cells 2 (10), 645–654. 10.1046/j.1365-2443.1997.1490345.x. [DOI] [PubMed] [Google Scholar]
  171. Fernandezsalguero P.; et al. (1995) Immune-system impairment and hepatic-fibrosis in mice lacking the dioxin-binding Ah receptor. Science 268 (5211), 722–726. 10.1126/science.7732381. [DOI] [PubMed] [Google Scholar]
  172. Harrill J. A.; et al. (2013) Knockout of the aryl hydrocarbon receptor results in distinct hepatic and renal phenotypes in rats and mice. Toxicol. Appl. Pharmacol. 272 (2), 503–518. 10.1016/j.taap.2013.06.024. [DOI] [PubMed] [Google Scholar]
  173. Haas K.; et al. (2016) Aryl hydrocarbon receptor in keratinocytes is essential for murine skin barrier integrity. J. Invest. Dermatol. 136 (11), 2260–2269. 10.1016/j.jid.2016.06.627. [DOI] [PubMed] [Google Scholar]
  174. Schmidt J. V.; et al. (1996) Characterization of a murine Ahr null allele: Involvement of the Ah receptor in hepatic growth and development. Proc. Natl. Acad. Sci. U. S. A. 93 (13), 6731–6736. 10.1073/pnas.93.13.6731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Lahvis G. P.; Bradfield C. A. (1998) Ahr null alleles: Distinctive or different?. Biochem. Pharmacol. 56 (7), 781–787. 10.1016/S0006-2952(98)00134-8. [DOI] [PubMed] [Google Scholar]
  176. Gonzalez F. J.; Fernandez-Salguero P. (1998) The aryl hydrocarbon receptor - studies using the Ahr-null mice. Drug Metab. Dispos. 26 (12), 1194–1198. [PubMed] [Google Scholar]
  177. Lund A. K.; et al. (2005) Endothelin-1-mediated increase in reactive oxygen species and nadph oxidase activity in hearts of aryl hydrocarbon receptor (Ahr) null mice. Toxicol. Sci. 88 (1), 265–273. 10.1093/toxsci/kfi284. [DOI] [PubMed] [Google Scholar]
  178. Benedict J.; et al. (2000) Physiological role for the aryl hydrocarbon receptor in mouse ovary biology. Toxicol. Sci. 56, 382–388. 10.1093/toxsci/56.2.382. [DOI] [PubMed] [Google Scholar]
  179. Lew B. J.; Manickam R.; Lawrence B. P. (2011) Activation of the aryl hydrocarbon receptor during pregnancy in the mouse alters mammary development through direct effects on stromal and epithelial tissues. Biol. Reprod. 84 (6), 1094–1102. 10.1095/biolreprod.110.087544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Hushka L. J.; Williams J. S.; Greenlee W. F. (1998) Characterization of 2,3,7,8-tetrachlorodibenzofuran-dependent suppression and ah receptor pathway gene expression in the developing mouse mammary gland. Toxicol. Appl. Pharmacol. 152 (1), 200–210. 10.1006/taap.1998.8508. [DOI] [PubMed] [Google Scholar]
  181. FernandezSalguero P. M.; et al. (1997) Lesions of aryl-hydrocarbon receptor-deficient mice. Vet. Pathol. 34 (6), 605–614. 10.1177/030098589703400609. [DOI] [PubMed] [Google Scholar]
  182. Abbott B. D.; et al. (1999) Adverse reproductive outcomes in the transgenic ah receptor-deficient mouse. Toxicol. Appl. Pharmacol. 155 (1), 62–70. 10.1006/taap.1998.8601. [DOI] [PubMed] [Google Scholar]
  183. Quintana F. J.; Sherr D. H. (2013) Aryl hydrocarbon receptor control of adaptive immunity. Pharmacol. Rev. 65 (4), 1148–1161. 10.1124/pr.113.007823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Stockinger B.; et al. (2014) The aryl hydrocarbon receptor: Multitasking in the immune system. Annu. Rev. Immunol. 32, 403–432. 10.1146/annurev-immunol-032713-120245. [DOI] [PubMed] [Google Scholar]
  185. Phadnis-Moghe A. S.; et al. (2016) Immunological characterization of the aryl hydrocarbon receptor (Ahr) knockout rat in the presence and absence of 2,3,7,8-tetrachlorodibenzo- p -dioxin (TCDD). Toxicology 368–369, 172–182. 10.1016/j.tox.2016.08.019. [DOI] [PubMed] [Google Scholar]
  186. Esser C.; Rannug A. (2015) The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol. Rev. 67 (2), 259–279. 10.1124/pr.114.009001. [DOI] [PubMed] [Google Scholar]
  187. Walisser J. A.; et al. (2004) Gestational exposure of ahr and arnt hypomorphs to dioxin rescues vascular development. Proc. Natl. Acad. Sci. U. S. A. 101 (47), 16677–16682. 10.1073/pnas.0404379101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Diaz-Diaz C. J.; et al. (2016) The aryl hydrocarbon receptor is a repressor of inflammation-associated colorectal tumorigenesis in mouse. Ann. Surg. 264 (3), 429–436. 10.1097/SLA.0000000000001874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. McIntosh B. E.; Hogenesch J. B.; Bradfield C. A. (2010) Mammalian per-arnt-sim proteins in environmental adaptation. Annu. Rev. Physiol. 72, 625–645. 10.1146/annurev-physiol-021909-135922. [DOI] [PubMed] [Google Scholar]
  190. Taylor B. L.; Zhulin I. B. (1999) Pas domains: Internal sensors of oxygen, redox potential, and light. Microbiol. Mol. Biol. Rev. 63 (2), 479–506. 10.1128/MMBR.63.2.479-506.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Hahn M. E.; et al. (2006) Unexpected diversity of aryl hydrocarbon receptors in non-mammalian vertebrates: Insights from comparative genomics. Journal of Experimental Zoology Part A: Comparative Experimental Biology 305A (9), 693–706. 10.1002/jez.a.323. [DOI] [PubMed] [Google Scholar]
  192. Reitzel A. M.; et al. (2014) Aryl hydrocarbon receptor (AHR) in the cnidarian nematostella vectensis: Comparative expression, protein interactions, and ligand binding. Dev. Genes Evol. 224 (1), 13–24. 10.1007/s00427-013-0458-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Ema M.; et al. (1997) A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1 alpha regulates the VEGF expression and is potentially involved in lung and vascular development. Proc. Natl. Acad. Sci. U. S. A. 94 (9), 4273–4278. 10.1073/pnas.94.9.4273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Tian H.; McKnight S. L.; Russell D. W. (1997) Endothelial pas domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes Dev. 11 (1), 72–82. 10.1101/gad.11.1.72. [DOI] [PubMed] [Google Scholar]
  195. Wang G. L.; et al. (1995) Hypoxia-inducible factor-1 is a basic-helix-loop-helix-pas heterodimer regulated by cellular o-2 tension. Proc. Natl. Acad. Sci. U. S. A. 92 (12), 5510–5514. 10.1073/pnas.92.12.5510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Ohno S. (1970) Evolution by gene duplication, Springer, Berlin. [Google Scholar]
  197. Hahn M. E.; et al. (1997) Molecular evolution of two vertebrate aryl hydrocarbon (dioxin) receptors (ahr1 and ahr2) and the pas family. Proc. Natl. Acad. Sci. U. S. A. 94 (25), 13743–13748. 10.1073/pnas.94.25.13743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Hahn M. E.; Karchner S. I.; Merson R. R. (2017) Diversity as opportunity: Insights from 600 million years of ahr evolution. Current Opinion in Toxicology 2, 58–71. 10.1016/j.cotox.2017.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Karchner S. I.; et al. (2009) The active form of human aryl hydrocarbon receptor (AHR) repressor lacks exon 8, and its pro(185) and ala(185) variants repress both AHR and hypoxia-inducible factor. Mol. Cell. Biol. 29 (13), 3465–3477. 10.1128/MCB.00206-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Hahn M. E. (1998) The aryl hydrocarbon receptor: A comparative perspective. Comp. Biochem. Physiol., Part C: Pharmacol., Toxicol. Endocrinol. 121 (1–3), 23–53. 10.1016/S0742-8413(98)10028-2. [DOI] [PubMed] [Google Scholar]
  201. Stegeman J. J., and Hahn M. E. (1994) Biochemistry and molecular biology of monooxygenases: Current perspectives on forms, functions, and regulation of cytochrome p450 in aquatic species. In Aquatic Toxicology: Molecular, Biochemical and Cellular Perspectives, Boca Raton, FL, pp 87–206.
  202. Zhulin I. B.; Taylor B. L. (1997) PAS domain s-boxes in archaea, bacteria and sensors for oxygen and redox. Trends Biochem. Sci. 22 (9), 331–333. 10.1016/S0968-0004(97)01110-9. [DOI] [PubMed] [Google Scholar]
  203. Duncan D. M.; Burgess E.A.; Duncan I. (1998) Control of distal antennal identity and tarsal development in drosophila by spineless-aristapedia, a homolog of the mammalian dioxin receptor. Genes Dev. 12 (9), 1290–1303. 10.1101/gad.12.9.1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Wernet M. F. (2006) Stochastic spineless expression creates the retinal mosaic for colour vision. Nature 440 (7081), 174–180. 10.1038/nature04615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Qin H.; Powell-Coffman J. A. (2004) The caenorhabditis elegans aryl hydrocarbon receptor, ahr-1, regulates neuronal development. Dev. Biol. 270 (1), 64–75. 10.1016/j.ydbio.2004.02.004. [DOI] [PubMed] [Google Scholar]
  206. Smith C. J.; et al. (2013) Sensory neuron fates are distinguished by a transcriptional switch that regulates dendrite branch stabilization. Neuron 79 (2), 266–280. 10.1016/j.neuron.2013.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Gillner M.; et al. (1985) Interactions of indoles with specific binding-sites for 2,3,7,8-tetrachlorodibenzo-para-dioxin in rat-liver. Mol. Pharmacol. 28 (4), 357–363. [PubMed] [Google Scholar]
  208. Nguyen L. P.; Bradfield C. A. (2008) The search for endogenous activators of the aryl hydrocarbon receptor. Chem. Res. Toxicol. 21 (1), 102–116. 10.1021/tx7001965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Long G.; McKinney J.; Pedersen L. (1987) Polychlorinated dibenzofuran (PCDF) binding to the ah receptor(s) and associated enzyme-induction - theoretical-model based on molecular-parameters. Quant. Struct.-Act. Relat. 6 (1), 1–7. 10.1002/qsar.19870060102. [DOI] [Google Scholar]
  210. Miller G.; Sontum S.; Crosby D. G. (1977) Electron-acceptor properties of chlorinated dibenzo-p-dioxins. Bull. Environ. Contam. Toxicol. 18 (5), 611–616. 10.1007/BF01684008. [DOI] [PubMed] [Google Scholar]
  211. Safe S. H. (1986) Comparative toxicology and mechanism of action of polychlorinated dibenzo-p-dioxins and dibenzofurans. Annu. Rev. Pharmacol. Toxicol. 26, 371–399. 10.1146/annurev.pa.26.040186.002103. [DOI] [PubMed] [Google Scholar]
  212. Safe S.; et al. (1985) PCBs - structure-function-relationships and mechanism of action. Environ. Health Perspect. 60, 47–56. 10.1289/ehp.856047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Murray I. A.; Patterson A. D.; Perdew G. H. (2014) Aryl hydrocarbon receptor ligands in cancer: Friend and foe. Nat. Rev. Cancer 14 (12), 801–14. 10.1038/nrc3846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Wincent E.; et al. (2012) 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. U. S. A. 109 (12), 4479–4484. 10.1073/pnas.1118467109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Gao Q.; et al. (2019) Age-dependent human elimination half-lives of dioxin-like polychlorinated biphenyls derived from biomonitoring data in the general population. Chemosphere 222, 541–548. 10.1016/j.chemosphere.2019.01.168. [DOI] [PubMed] [Google Scholar]
  216. Matsumoto S. (2015) Unexpectedly long half-lives of blood 2,3,4,7,8-pentachlorodibenzofuran (pecdf) levels in yusho patients. Environ. Health 14 (1), 76. 10.1186/s12940-015-0059-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Bradfield C. A.; Bjeldanes L. F. (1987) Structure-activity-relationships of dietary indoles - a proposed mechanism of action as modifiers of xenobiotic metabolism. J. Toxicol. Environ. Health 21 (3), 311–323. 10.1080/15287398709531021. [DOI] [PubMed] [Google Scholar]
  218. Bjeldanes L. F.; et al. (1991) Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol invitro and invivo - comparisons with 2,3,7,8-tetrachlorodibenzo-para-dioxin. Proc. Natl. Acad. Sci. U. S. A. 88 (21), 9543–9547. 10.1073/pnas.88.21.9543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Shertzer H. (2000) The micronutrient indole-3-carbinol: Implications for disease and chemoprevention. Drug Metab. Drug Interact. 17, 29. 10.1515/DMDI.2000.17.1-4.159. [DOI] [PubMed] [Google Scholar]
  220. Rogan E. G. (2006) The natural chemopreventive compound indole-3-carbinol: State of the science. In Vivo 20 (2), 221–228. [PubMed] [Google Scholar]
  221. Loub W. D.; Wattenberg L. W.; Davis D. W. (1975) Aryl-hydrocarbon hydroxylase induction in rat tissues by naturally occurring indoles of cruciferous plants. Journal of the National Cancer Institute 54 (4), 985–988. [PubMed] [Google Scholar]
  222. Shertzer H. G. (1982) Invivo protection by indole-3-carbinol against carcinogen covalent binding to mouse hepatic macromolecules. Federation Proceedings 41 (5), 1559–1559. [Google Scholar]
  223. Wattenberg L. W. (1996) Chemoprevention of cancer. Prev. Med. 25 (1), 44–45. 10.1006/pmed.1996.0015. [DOI] [PubMed] [Google Scholar]
  224. Bradfield C. A.; Bjeldanes L. F. (1987) Structure—activity relationships of dietary indoles: A proposed mechanism of action as modifiers of xenobiotic metabolism. J. Toxicol. Environ. Health 21 (3), 311–323. 10.1080/15287398709531021. [DOI] [PubMed] [Google Scholar]
  225. Chen Y. H.; et al. (1995) Regulation of cyp1a1 by indolo 3,2-b carbazole in murine hepatoma-cells. J. Biol. Chem. 270 (38), 22548–22555. 10.1074/jbc.270.38.22548. [DOI] [PubMed] [Google Scholar]
  226. Bittinger M. A.; Nguyen L. P.; Bradfield C. A. (2003) Aspartate aminotransferase generates proagonists of the aryl hydrocarbon receptor. Mol. Pharmacol. 64 (3), 550–556. 10.1124/mol.64.3.550. [DOI] [PubMed] [Google Scholar]
  227. Nguyen L. P.; et al. (2009) D-amino acid oxidase generates agonists of the aryl hydrocarbon receptor fromd-tryptophan. Chem. Res. Toxicol. 22 (12), 1897–1904. 10.1021/tx900043s. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Chowdhury G.; et al. (2009) Structural identification of diindole agonists of the aryl hydrocarbon receptor derived from degradation of indole-3-pyruvic acid. Chem. Res. Toxicol. 22 (12), 1905–1912. 10.1021/tx9000418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Mezrich J. D.; et al. (2010) An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory t cells. J. Immunol. 185 (6), 3190–3198. 10.4049/jimmunol.0903670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Seok S.-H. (2018) Trace derivatives of kynurenine potently activate the aryl hydrocarbon receptor (ahr). J. Biol. Chem. 293, 1994. 10.1074/jbc.RA117.000631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Lowe M. M.; et al. (2014) Identification of cinnabarinic acid as a novel endogenous aryl hydrocarbon receptor ligand that drives il-22 production. PLoS One 9 (2), e87877 10.1371/journal.pone.0087877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Soucek P. (2011) Xenobiotics. Encyclopedia of Cancer 3964–3967. 10.1007/978-3-642-16483-5_6276. [DOI] [Google Scholar]
  233. Quattrochi L. C.; Tukey R. H. (1993) Nuclear uptake of the ah (dioxin) receptor in response to omeprazole - transcriptional activation of the human Cyp1a1 gene. Mol. Pharmacol. 43 (4), 504–508. [PubMed] [Google Scholar]
  234. Zhang S.; Qin C. H.; Safe S. H. (2003) Flavonoids as aryl hydrocarbon receptor agonists/antagonists: Effects of structure and cell context. Environ. Health Perspect. 111 (16), 1877–1882. 10.1289/ehp.6322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Ciolino H. P.; Daschner P. J.; Yeh G. C. (1999) Dietary flavonols quercetin and kaempferol are ligands of the aryl hydrocarbon receptor that affect cyp1a1 transcription differentially. Biochem. J. 340, 715–722. 10.1042/bj3400715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Song J.; et al. (2002) A ligand for the aryl hydrocarbon receptor isolated from lung. Proc. Natl. Acad. Sci. U. S. A. 99 (23), 14694–14699. 10.1073/pnas.232562899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Phelan D.; et al. (1998) Activation of the Ah receptor signal transduction pathway by bilirubin and biliverdin. Arch. Biochem. Biophys. 357 (1), 155–163. 10.1006/abbi.1998.0814. [DOI] [PubMed] [Google Scholar]
  238. Wincent E.; et al. (2009) The suggested physiologic aryl hydrocarbon receptor activator and cytochrome p4501 substrate 6-formylindolo[3,2-b]carbazole is present in humans. J. Biol. Chem. 284 (5), 2690–2696. 10.1074/jbc.M808321200. [DOI] [PubMed] [Google Scholar]
  239. Sugihara K.; et al. (2004) Aryl hydrocarbon receptor-mediated induction of microsomal drug-metabolizing enzyme activity by indirubin and indigo. Biochem. Biophys. Res. Commun. 318 (2), 571–578. 10.1016/j.bbrc.2004.04.066. [DOI] [PubMed] [Google Scholar]
  240. Birnbaum L. S. (2017) Dioxin and the ah receptor: Synergy of discovery. Current Opinion in Toxicology 2, 120–123. 10.1016/j.cotox.2017.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Lahvis G. P.; et al. (2005) The aryl hydrocarbon receptor is required for developmental closure of the ductus venosus in the neonatal mouse. Mol. Pharmacol. 67 (3), 714–720. 10.1124/mol.104.008888. [DOI] [PubMed] [Google Scholar]
  242. Jiang W. (2004) Disruption of the Ah receptor gene alters the susceptibility of mice to oxygen-mediated regulation of pulmonary and hepatic cytochromes p4501a expression and exacerbates hyperoxic lung injury. J. Pharmacol. Exp. Ther. 310 (2), 512–519. 10.1124/jpet.103.059766. [DOI] [PubMed] [Google Scholar]
  243. Veldhoen M.; Duarte J.H. (2010) The aryl hydrocarbon receptor: Fine-tuning the immune-response. Curr. Opin. Immunol. 22 (6), 747–752. 10.1016/j.coi.2010.09.001. [DOI] [PubMed] [Google Scholar]
  244. Quintana F. J.; Weiner H. L. (2009) Environmental control of th17 differentiation. Eur. J. Immunol. 39 (3), 655–657. 10.1002/eji.200839198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Stockinger B.; et al. (2014) The aryl hydrocarbon receptor: Multitasking in the immune system. Annu. Rev. Immunol. 32 (1), 403–432. 10.1146/annurev-immunol-032713-120245. [DOI] [PubMed] [Google Scholar]
  246. Rannug A.; Rannug U. (2018) The tryptophan derivative 6-formylindolo[3,2-b]carbazole, ficz, a dynamic mediator of endogenous aryl hydrocarbon receptor signaling, balances cell growth and differentiation. Crit. Rev. Toxicol. 48 (7), 555–574. 10.1080/10408444.2018.1493086. [DOI] [PubMed] [Google Scholar]
  247. Murray I. A.; Perdew G. H. (2017) Ligand activation of the ah receptor contributes to gastrointestinal homeostasis. Current Opinion in Toxicology 2, 15–23. 10.1016/j.cotox.2017.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Jiang W.; et al. (2004) Disruption of the Ah receptor gene alters the susceptibility of mice to oxygen-mediated regulation of pulmonary and hepatic cytochromes p4501a expression and exacerbates hyperoxic lung injury. J. Pharmacol. Exp. Ther. 310 (2), 512–519. 10.1124/jpet.103.059766. [DOI] [PubMed] [Google Scholar]
  249. Hankinson O. (2016) The role of AHR-inducible cytochrome p450s in metabolism of polyunsaturated fatty acids. Drug Metab. Rev. 48 (3), 342–350. 10.1080/03602532.2016.1197240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Schaldach C. M.; Riby J.; Bjeldanes L. F. (1999) Lipoxin a4: A new class of ligand for the Ah receptor. Biochemistry 38 (23), 7594–7600. 10.1021/bi982861e. [DOI] [PubMed] [Google Scholar]
  251. Smirnova A.; et al. (2016) Evidence for new light-independent pathways for generation of the endogenous aryl hydrocarbon receptor agonist FICZ. Chem. Res. Toxicol. 29 (1), 75–86. 10.1021/acs.chemrestox.5b00416. [DOI] [PubMed] [Google Scholar]
  252. Hubbard T. D.; Murray I. A.; Perdew G. H. (2015) Indole and tryptophan metabolism: Endogenous and dietary routes to ah receptor activation. Drug Metab. Dispos. 43 (10), 1522–35. 10.1124/dmd.115.064246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Noakes R. (2015) The aryl hydrocarbon receptor: A review of its role in the physiology and pathology of the integument and its relationship to the tryptophan metabolism. Int. J. Tryptophan Res. 8, IJTR.S19985. 10.4137/IJTR.S19985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Rannug A.; et al. (1987) 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 (32), 15422–15427. [PubMed] [Google Scholar]
  255. Li Y.; et al. (2011) Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell 147 (3), 629–640. 10.1016/j.cell.2011.09.025. [DOI] [PubMed] [Google Scholar]
  256. Hubbard T. D. (2015) Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Sci. Rep. 5, 12689. 10.1038/srep12689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Metidji A.; et al. (2018) The environmental sensor ahr protects from inflammatory damage by maintaining intestinal stem cell homeostasis and barrier integrity. Immunity 49 (2), 353–362. e5. 10.1016/j.immuni.2018.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Villa M.; et al. (2016) The aryl hydrocarbon receptor controls cyclin o to promote epithelial multiciliogenesis. Nat. Commun. 7 (1), 12652. 10.1038/ncomms12652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. McMillan B. J.; Bradfield C. A. (2007) The aryl hydrocarbon receptor is activated by modified low-density lipoprotein. Proc. Natl. Acad. Sci. U. S. A. 104 (4), 1412–1417. 10.1073/pnas.0607296104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Lahvis G. P.; et al. (2000) Portosystemic shunting and persistent fetal vascular structures in aryl hydrocarbon receptor-deficient mice. Proc. Natl. Acad. Sci. U. S. A. 97 (19), 10442–10447. 10.1073/pnas.190256997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Schiering C.; et al. (2017) Feedback control of AHR signalling regulates intestinal immunity. Nature 542 (7640), 242–245. 10.1038/nature21080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Dalton T. P.; Puga A.; Shertzer H. G. (2002) Induction of cellular oxidative stress by aryl hydrocarbon receptor activation. Chem.-Biol. Interact. 141 (1–2), 77–95. 10.1016/S0009-2797(02)00067-4. [DOI] [PubMed] [Google Scholar]
  263. Matsumura F. (2003) On the significance of the role of cellular stress response reactions in the toxic actions of dioxin. Biochem. Pharmacol. 66 (4), 527–540. 10.1016/S0006-2952(03)00157-6. [DOI] [PubMed] [Google Scholar]
  264. Dietrich C. (2016) Antioxidant functions of the aryl hydrocarbon receptor. Stem Cells Int. 2016, 1–10. 10.1155/2016/7943495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Robertson J. D.; Orrenius S. (2000) Molecular mechanisms of apoptosis induced by cytotoxic chemicals. Crit. Rev. Toxicol. 30 (5), 609–627. 10.1080/10408440008951122. [DOI] [PubMed] [Google Scholar]
  266. Veith A.; Moorthy B. (2018) Role of cytochrome p450s in the generation and metabolism of reactive oxygen species. Current Opinion in Toxicology 7, 44–51. 10.1016/j.cotox.2017.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Nebert D. W.; et al. (2000) Role of the aromatic hydrocarbon receptor and [Ah] gene battery in the oxidative stress response, cell cycle control, and apoptosis. Biochem. Pharmacol. 59 (1), 65–85. 10.1016/S0006-2952(99)00310-X. [DOI] [PubMed] [Google Scholar]
  268. Orrenius S. (2019) Role of cell death in toxicology: Does it matter how cells die?. Annu. Rev. Pharmacol. Toxicol. 59 (1), 1–14. 10.1146/annurev-pharmtox-010818-021725. [DOI] [PubMed] [Google Scholar]
  269. Nukaya M.; et al. (2010) The aryl hydrocarbon receptor-interacting protein (AIP) is required for dioxin-induced hepatotoxicity but not for the induction of the cyp1a1 and cyp1a2 genes. J. Biol. Chem. 285 (46), 35599–35605. 10.1074/jbc.M110.132043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Nukaya M.; Moran S.; Bradfield C. A. (2009) The role of the dioxin-responsive element cluster between the cyp1a1 and cyp1a2 loci in aryl hydrocarbon receptor biology. Proc. Natl. Acad. Sci. U. S. A. 106 (12), 4923–4928. 10.1073/pnas.0809613106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. van Larebeke N.; et al. (2001) The belgian pcb and dioxin incident of january-june 1999: Exposure data and potential impact on health. Environ. Health Perspect. 109 (3), 265–273. 10.1289/ehp.01109265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Heres L.; et al. (2010) Tracing and analytical results of the dioxin contamination incident in 2008 originating from the republic of ireland. Food Addit. Contam., Part A 27 (12), 1733–1744. 10.1080/19440049.2010.522598. [DOI] [PubMed] [Google Scholar]
  273. Marnane I. (2012) Comprehensive environmental review following the pork PCB/dioxin contamination incident in ireland. J. Environ. Monit. 14 (10), 2551–2556. 10.1039/c2em30374d. [DOI] [PubMed] [Google Scholar]
  274. Bertazzi P. A.; et al. (1998) The seveso studies on early and long-term effects of dioxin exposure: A review. Environ. Health Perspect. 106, 625–633. 10.2307/3433813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. Warner M.; et al. (2011) Dioxin exposure and cancer risk in the seveso women’s health study. Environ. Health Perspect. 119 (12), 1700–1705. 10.1289/ehp.1103720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. National Academies of Sciences, Engineering, and Medicine. (2018) Veterans and agent orange: Update 11 (2018), The National Academies Press, Washington, DC. [PubMed] [Google Scholar]
  277. Dwernychuk L. W.; et al. (2002) Dioxin reservoirs in southern Vietnam—a legacy of agent orange. Chemosphere 47 (2), 117–137. 10.1016/S0045-6535(01)00300-9. [DOI] [PubMed] [Google Scholar]
  278. Evans R. G.; et al. (2000) Dioxin incinerator emissions exposure study times beach, missouri. Chemosphere 40 (9–11), 1063–1074. 10.1016/S0045-6535(99)00354-9. [DOI] [PubMed] [Google Scholar]
  279. Sun M. (1983) Missouri costly dioxin lesson. Science 219 (4583), 367–369. 10.1126/science.6849139. [DOI] [PubMed] [Google Scholar]
  280. Schulte K. W.; et al. (2017) Structural basis for aryl hydrocarbon receptor-mediated gene activation. Structure 25 (7), 1025–1033. 10.1016/j.str.2017.05.008. [DOI] [PubMed] [Google Scholar]
  281. Jones L. C.; Whitlock J. P. (2001) Dioxin-inducible transactivation in a chromosomal setting - analysis of the acidic domain of the Ah receptor. J. Biol. Chem. 276 (27), 25037–25042. 10.1074/jbc.M102910200. [DOI] [PubMed] [Google Scholar]
  282. Watt K.; et al. (2005) Induced α-helix structure in the aryl hydrocarbon receptor transactivation domain modulates protein-protein interactions. Biochemistry 44 (2), 734–743. 10.1021/bi0487701. [DOI] [PubMed] [Google Scholar]
  283. Kang H. J.; et al. (2008) Brca1 transcriptional activity is enhanced by interactions between its ad1 domain and AHR. Cancer Chemother. Pharmacol. 62 (6), 965–975. 10.1007/s00280-008-0686-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Kang H. J.; et al. (2006) Brca1 modulates xenobiotic stress-inducible gene expression by interacting with ARNT in human breast cancer cells. J. Biol. Chem. 281 (21), 14654–14662. 10.1074/jbc.M601613200. [DOI] [PubMed] [Google Scholar]
  285. Wang S.; Hankinson O. (2002) Functional involvement of the brahma/swi2-related gene 1 protein in cytochrome p4501a1 transcription mediated by the aryl hydrocarbon receptor complex. J. Biol. Chem. 277 (14), 11821–11827. 10.1074/jbc.M110122200. [DOI] [PubMed] [Google Scholar]
  286. Jin H. L.; Jeong K. W. (2016) Regulation of aryl hydrocarbon receptor-mediated transcription in human retinal pigmented epithelial cells. Biochem. Biophys. Res. Commun. 472 (2), 366–372. 10.1016/j.bbrc.2016.03.006. [DOI] [PubMed] [Google Scholar]
  287. Zhang S.; Rowlands C.; Safe S. (2008) Ligand-dependent interactions of the Ah receptor with coactivators in a mammalian two-hybrid assay. Toxicol. Appl. Pharmacol. 227 (2), 196–206. 10.1016/j.taap.2007.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Kim J. H.; Stallcup M. R. (2004) Role of the coiled-coil coactivator (COCOA) in aryl hydrocarbon receptor-mediated transcription. J. Biol. Chem. 279 (48), 49842–49848. 10.1074/jbc.M408535200. [DOI] [PubMed] [Google Scholar]
  289. Klinge C. M.; Kaur K.; Swanson H. I. (2000) The aryl hydrocarbon receptor interacts with estrogen receptor alpha and orphan receptors coup-tfi and errα1. Arch. Biochem. Biophys. 373 (1), 163–174. 10.1006/abbi.1999.1552. [DOI] [PubMed] [Google Scholar]
  290. Nguyen T. A.; et al. (1999) Interactions of nuclear receptor coactivator/corepressor proteins with the aryl hydrocarbon receptor complex. Arch. Biochem. Biophys. 367 (2), 250–257. 10.1006/abbi.1999.1282. [DOI] [PubMed] [Google Scholar]
  291. Chen Y.-H.; et al. (2006) Role of gac63 in transcriptional activation mediated by the aryl hydrocarbon receptor. J. Biol. Chem. 281 (18), 12242–12247. 10.1074/jbc.M512537200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  292. Tohkin M.; et al. (2000) Aryl hydrocarbon receptor is required for p300-mediated induction of DNA synthesis by adenovirus e1a. Mol. Pharmacol. 58 (4), 845–851. 10.1124/mol.58.4.845. [DOI] [PubMed] [Google Scholar]
  293. Kobayashi A.; et al. (1997) Cbp/p300 functions as a possible transcriptional coactivator of Ah receptor nuclear translocator (ARNT). J. Biochem. 122 (4), 703–710. 10.1093/oxfordjournals.jbchem.a021812. [DOI] [PubMed] [Google Scholar]
  294. Elferink C. J.; Ge N.-L.; Levine A. (2001) Maximal aryl hydrocarbon receptor activity depends on an interaction with the retinoblastoma protein. Mol. Pharmacol. 59 (4), 664–673. 10.1124/mol.59.4.664. [DOI] [PubMed] [Google Scholar]
  295. Madak-Erdogan Z.; Katzenellenbogen B. S. (2012) Aryl hydrocarbon receptor modulation of estrogen receptor α-mediated gene regulation by a multimeric chromatin complex involving the two receptors and the coregulator rip140. Toxicol. Sci. 125 (2), 401–411. 10.1093/toxsci/kfr300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Kumar M. B.; Tarpey R. W.; Perdew G. H. (1999) Differential recruitment of coactivator rip140 byahand estrogen receptors. J. Biol. Chem. 274 (32), 22155–22164. 10.1074/jbc.274.32.22155. [DOI] [PubMed] [Google Scholar]
  297. Klinge C. M.; et al. (2001) Short heterodimer partner (shp) orphan nuclear receptor inhibits the transcriptional activity of aryl hydrocarbon receptor (ahr)/ahr nuclear translocator (ARNT). Arch. Biochem. Biophys. 390 (1), 64–70. 10.1006/abbi.2001.2366. [DOI] [PubMed] [Google Scholar]
  298. Fallone F.; et al. (2004) Retinoids repress Ah receptor Cyp1a1 induction pathway through the smrt corepressor. Biochem. Biophys. Res. Commun. 322 (2), 551–556. 10.1016/j.bbrc.2004.07.153. [DOI] [PubMed] [Google Scholar]
  299. Beischlag T. V.; et al. (2004) Recruitment of thyroid hormone receptor/retinoblastoma-interacting protein 230 by the aryl hydrocarbon receptor nuclear translocator is required for the transcriptional response to both dioxin and hypoxia. J. Biol. Chem. 279 (52), 54620–54628. 10.1074/jbc.M410456200. [DOI] [PubMed] [Google Scholar]
  300. Solaimani P.; Wang F.; Hankinson O. (2014) Sin3a, generally regarded as a transcriptional repressor, is required for induction of gene transcription by the aryl hydrocarbon receptor. J. Biol. Chem. 289 (48), 33655–33662. 10.1074/jbc.M114.611236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Harper P. A.; et al. (1991) Detection and characterization of the ah-receptor for 2,3,7,8-tetrachlorodibenzo-p-dioxin in the human colon adenocarcinoma cell-line ls180. Arch. Biochem. Biophys. 290 (1), 27–36. 10.1016/0003-9861(91)90587-9. [DOI] [PubMed] [Google Scholar]
  302. Barnes D. G. (1991) Toxicity equivalents and APS’s risk assessment of 2,3,7,8-TCDD. Sci. Total Environ. 104, 73–86. 10.1016/0048-9697(91)90008-3. [DOI] [PubMed] [Google Scholar]
  303. Safe H. S. (1998) Hazard and risk assessment of chemical mixtures using the toxic equivalency factor approach. Environmental Health Perspectives: Supplement 106, 4. 10.2307/3434151. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Chemical Research in Toxicology are provided here courtesy of American Chemical Society

RESOURCES