Abstract
Per-Arnt-Sim (PAS) domain-containing proteins are critical to homeostatic regulatory networks that mediate responsiveness to environmental change. PAS domains are multifunctional structural motifs that allow protein-protein interactions among family members, typically forming heterodimeric transcription factors to affect the transcription of target genes. Prototypical PAS domain-dependent pathways include the circadian clock network and metabolic regulation of the xenobiotic response through the aryl hydrocarbon receptor (AhR). Both pathways are increasingly linked to health, and alteration in their function contributes to development of disease. The aryl hydrocarbon receptor (AhR) demonstrates promiscuity in ligand binding and selectivity during heterodimer formation, which allows varied combinations of protein/protein interactions with other Per-Arnt-Sim (PAS) domain containing proteins and crosstalk among signaling pathways, including the molecular clockworks. AhR and the circadian signaling pathways are highly integrated and reciprocally regulated. AhR exhibits a rhythmic expression and time-dependent sensitivity to activation by AhR agonists. Conversely, AhR influences amplitude and phase of rhythms in circadian clock genes, hormones, and behavior. Understanding the molecular interactions between AhR and the clock provides insight into physiological regulation of rhythmic processes and provides an innovative approach to development of therapeutics.
Keywords: Circadian rhythm, Aryl hydrocarbon Receptor, Per1, xenobiotics, amplitude regulation
Introduction
The revolution of the Earth about its own axis, which divides the solar day into a pattern of light and darkness, may be the most persistent environmental factor subserving the evolution of modern species. The sense of time endowed by this environment is accentuated by the presence of an internal mechanism for keeping time on a 24 h (circadian) scale, inherent in the genetic framework of living organisms ranging from cyanobacteria to humans. The internal, molecular program drives circadian oscillations within the organism that manifest at the molecular, biochemical, physiological and behavioral levels. The internal timing mechanism allows persistence of timekeeping in the absence of environmental cues, and anticipatory responses to changes in the environment to promote survival (Takahashi et al., 2008; Moore, 2013). By programming internal events to occur consistently at specific times across the 24 h cycle, physiological activity coordinates with the light/dark environment (Gillette & Tischkau, 1999; Gillette & Mitchell, 2002). For example, animals typically have bouts of rest and activity that correspond to specific times of day, allowing them to fill a particular ecological niche, to find food and avoid predators. Other examples of processes whose timing is controlled by a circadian clock include Neurospora crassa asexual spore production, leaf movements in plants, photosynthesis and nitrogen fixation in cyanobacterium (Bhadra et al., 2017), the firing rate of the suprachiasmatic nucleus (SCN) in mammals (Kuhlman & McMahon, 2006), clock gene expression levels and numerous aspects of metabolism (Takahashi, 2015, 2017).
Coherent circadian rhythms in mammals may be viewed as an organismic network, with a central hub located in the SCN of the ventral hypothalamus, the so-called “master clock”. Although individual neurons within the SCN are independent oscillators, coordinated function of specific SCN cell types is necessary for managing properly phased rhythms throughout the body (Yamaguchi et al., 2003; Liu et al., 2007; Ko et al., 2010). The SCN detects environmental signals and receives homeostatic feedback, which it uses to convey timing information to the rest of the organism through a variety of neural and endocrine mechanisms. Disruption of coordinated timing between the SCN and peripheral oscillators located in every organ system may upset the intricate homeostatic balance necessary for health and contribute to numerous pathologies, including cardiovascular disease, metabolic disease, cancer and others (Arble et al., 2015; Bass, 2017).
An important feature of the circadian timing system is its ability to adjust to resetting stimuli. Although the mammalian circadian timing system is sensitive to a variety of external and internal stimuli, the signals most relevant to this review are light, food and environmental chemicals. Nestled above the optic chiasm, the SCN receives monosynaptic neuronal input from the retina via specialized intrinsically photoreceptive retinal ganglion cells to function as the light-entrainable oscillator for the systemic circadian timing system (Hattar et al., 2003, 2006; Do & Yau, 2010; Hughes et al., 2015; Fernandez et al., 2016). N methyl d-aspartate (NMDA) receptor-mediated, glutamatergic neurotransmission activates signal transduction processes within the SCN to align the nucleus, and subsequently, the organism to light/dark cycles (Ebling, 1996; Gillette & Tischkau, 1999; Golombek & Rosenstein, 2010). Food can act independent of the SCN to entrain the circadian system. Although an anatomical site for the food-entrainable oscillator is yet to be determined, rhythms can be entrained by presentation of food at specific times of day. In response to food, animals demonstrate food anticipatory behavior, which is accompanied by an anticipatory rise in body temperature, increased corticosterone, gut motility, digestive enzyme activity and insulin secretion. Furthermore, rhythms can be shifted by altering the time when food is present. Importantly, this oscillator is independent of the SCN. The timing of food presentation alters clock gene transcripts in peripheral tissues, but not in the SCN; SCN ablation does not alter the timing of food anticipatory behavior. Thus, the system circadian timing system can be differentially entrained by light and food (Olivo et al., 2014; Hatori & Panda, 2015; Refinetti, 2015). Normally, these two signals harmonize with each other and the SCN coordinates timing of peripheral clocks. However, when the timing of light and food are presented out of their usual balance, rest/activity cycles remain controlled by the SCN, whereas peripheral systems may respond to food. Systemic disruption caused by discordance between the SCN and peripheral clocks may contribute to subsequent pathologies.
Rapid industrialization over the past 100 years has transformed the environment to trigger sweeping change in modern life-styles. Artificial lighting, computer and television screens, social demands incurred by the internet, work obligations, and weekend schedule changes may be considered “social zeitgebers” that act as environmental signals to affect circadian fidelity, and lead to chronic circadian disruption (Vetter, 2018). In addition, the industrial revolution and urbanization has increased exposure to chemical pollutants, whose impact on the circadian system in only beginning to be appreciated. The biological effects of many environmental xenobiotics are mediated through a common receptor mechanism, the aryl hydrocarbon receptor (AhR) signaling pathway. The AhR, similar to circadian clock components, is an ancient, evolutionarily conserved protein and member of the PAS domain family of proteins. AhR may function as a physiological bridging molecule between external and internal chemical signals; the exact physiological roles of AhR remain unclear, but seem to be multifactorial and certainly not limited to mediation of xenobiotic toxicity (Tian et al., 2015). Genomic organization, intron/exon splice patterns and conservation of the PAS domain suggests that brain-muscle ARNT-like 1 (BMAL1) and AhR are common derivatives of the same ancestral gene (Yu et al., 1999). AhR and components of its signaling pathways are regulated by the circadian clock. Conversely, anthropogenic and natural AhR ligands may impact clock function though AhR. The physiological interactions of AhR and the clock are further explored in this review.
SCN Communication with Peripheral Clocks
The SCN communicates timing information to the body through regulation of autonomic and neuroendocrine systems (reviewed in Dibner et al., 2010). Proximal SCN signals impact endocrine, pre-autonomic and intermediate neurons primarily in the dorsomedial hypothalamus, the medial preoptic area, and the subparaventricular zone of the hypothalamus (Kalsbeek et al., 2006). SCN neurons also project outside the hypothalamus to the paraventricular nucleus of the thalamus (PVT), the intergeniculate leaflet and the ventrolateral preoptic area (VLPO) (Hermes et al., 1996; Sun et al., 2001). Signals are relayed to pre-autonomic neurons in the brainstem and spinal cord, as well as endocrine neurons associated with secretion of corticotropin-releasing hormone, thyrotropin-releasing hormone and gonadotropin-releasing hormone (Kalsbeek & Buijs, 2002). Glutamate and gamma amino-butyric acid (GABA) (Buijs et al., 1995; Hermes et al., 1996) are primary neurotransmitters associated with SCN output, although a number of other molecules, including arginine vasopressin (Kalsbeek et al., 2006), cardiotrophin-like cytokine (Kraves and Weitz, 2006), prokineticin 2 (Cheng et al., 2002), vasoactive intestinal peptide (Kalsbeek & Buijs, 2002) and transforming growth factor α (Kramer et al., 2001) have also been identified.
The SCN has multiple outputs that influence hormone release. Although SCN-derived paracrine factors are sufficient to drive locomotor activity rhythms, direct neuronal connections are required for certain neuroendocrine rhythms (Lehman et al., 1987; Silver et al., 1996; de la Iglesia et al., 2003; Kalsbeek et al., 2008). A well-studied example of circadian control of peripheral endocrine rhythms is SCN regulation of the hypothalamic-pituitary-adrenal axis (HPA). The SCN provides stimulatory and inhibitory signals to influence the HPA axis (Tsang et al., 2014; Oster et al., 2017). SCN signaling to the PVN of the hypothalamus influences activity of corticotrophin-releasing neurons that regulate the secretion of adrenocorticotropic hormone (ACTH) from the pituitary gland (Vujovic et al., 2015). Furthermore, autonomic projections from the SCN through the PVN and intermediolateral column (IML) regulate diurnal changes in adrenal gland sensitivity to ACTH (Buijs et al., 1999). Retrograde labeling studies indicate that peripheral tissues receive both sympathetic and parasympathetic input from the SCN, suggesting that the clock regulates “rest” and “activity” phases (Buijs et al., 2003).
SCN-driven autonomic modulation of peripheral tissues is not limited to the adrenal gland; the pineal gland, heart, pancreas, intestines, ovaries and liver may also receive timing information from an SCN-derived autonomic signals (Buijs et al., 1999). Autonomic input from the SCN to the liver influences expression of hepatic clock and metabolic genes to regulate the daily plasma glucose rhythm (Kalsbeek et al., 2006, 2014). Inappropriately timed light exposure immediately resets liver rhythms, unless autonomic innervation to the liver is disrupted (Cailotto et al., 2005). Although SCN lesion does not always eliminate peripheral clock function, phase desynchrony among tissues suggests that the SCN coordinates rhythmicity among organ systems (Yoo et al., 2004; Guo et al., 2006). Over 100 transcripts lose circadian rhythmicity in the liver after adrenalectomy, which highlights the interaction of the SCN and the HPA axis in physiological regulation of clock function (Oishi et al., 2005).
Genome-wide transcriptional profiling indicates that between 4 (muscle, adipose tissue) and 16 (liver) percent of transcripts show circadian oscillations; moreover, over 50% of protein-coding transcripts are predicted to oscillate somewhere in the body (Hughes et al., 2009; Zhang et al., 2014). These oscillations subserve key organ-specific physiological functions. Most relevant to this review, xenobiotic detoxification in the liver, kidney and small intestine (Gachon et al., 2004), as well as carbohydrate (Lamia et al., 2008) and lipid (Le Martelot et al., 2009) metabolism in the liver and adipose tissue, are all clock-regulated events. The SCN likely coordinates inter-organ timing through autonomic and endocrine signals, to allow for systemic regulation of metabolic events.
The Molecular Clockworks
The molecular circadian clock, in mammals, consists of transcriptional activators and repressors that coordinate to function as interconnected positive and negative feedback loops (Reviewed in Takahashi, 2017). In the primary loop, transcriptional activators, CLOCK (Circadian locomotor output cycles kaput) and BMAL1, heterodimerize through their PAS domains bind at enhancer-box (Ebox) regions to drive transcription of PAS domain-containing clock genes, Period 1 (Per1), Per2 and Per3 and two Cryptochrome (Cry) genes Cry1 and Cry2 (Gekakis et al., 1998; Kume et al., 1999; Shearman et al., 2000). PER and CRY accumulate in the cytoplasm and form heterodimers that translocate to the nucleus, where they repress activity of CLOCK:BMAL1, effectively inhibiting their own expression (Lee et al., 2001; Lowrey & Takahashi, 2011). Posttranslational phosphorylation of PER by the casein kinase I family of enzymes (CKIδ/ε) regulate formation of dimers with CRY, nuclear import of the PER:CRY complex and degradation of PER (St John et al., 2014). CRY degradation is regulated by the F-box and leucine rich repeat proteins (FBLX3) and FBLX21 ubiquitin ligase complexes (Xing et al., 2013; Yoo et al., 2013). As PER and CRY are degraded, transcriptional repression is released and CLOCK:BMAL1 can begin a new cycle of transcription. Thus, PER and CRY stability are important for determining circadian period length.
The nuclear hormone receptors, Rev-erbα and Rev-erbβ are also rhythmically transcribed by CLOCK:BMAL1. REV-ERBα and REV-ERBβ compete with retinoic acid-related orphan receptors (RORα, β, γ) for binding at ROR elements (ROREs) in the promoter of the Bmal1 gene. REV-ERBs repress, whereas RORs activate, Bmal1 transcription (Preitner et al., 2002; Sato et al., 2004; Cho et al., 2012; Zhang et al., 2015), producing a rhythm that is anti-phase to that of Per and Cry. Finally, CLOCK:BMAL1 drive transcription of DBP (D-box binding protein) that can act together with the nuclear repressor NFIL3 (Nuclear Factor, Interleukin 3 regulated), a product of the REV-ERB/ROR loop, on D-boxes to affect rhythm transcriptional processes within the molecular clock (Mitsui et al., 2001; Gachon et al., 2004).
Dynamic epigenetic regulation of chromatin provides the appropriate template for activation and repression of transcription across the circadian cycle (reviewed in (Takahashi, 2017). CLOCK and BMAL1 associate with histone acetyltransferases to shape chromatin in preparation for transcription; CLOCK-derived histone acetyl transferase (HAT) activity acetylates BMAL1 (Etchegaray et al., 2003; Curtis et al., 2004; Doi et al., 2006; Hirayama et al., 2007). Interactions with the methyltransferase, MLL1 (Mixed lineage leukemia 1; Katada & Sassone-Corsi, 2010), and histone lysine demethylases, promote recruitment of CLOCK:BMAL1 to the target gene promoters, where co-activators are also recruited to enhance transcription (DiTacchio et al., 2011; Lande-Diner et al., 2013). As PER and CRY accumulate, they form a large repressor complex that interacts with CLOCK:BMAL1. The dbp-cullin-4 ubitquitin ligase complex covalently marks Ebox sites to recruit the PER complex. Subsequently, other repressor complexes get involved to deacetylate and methylate histones. RNA helicases are recruited to terminate transcription. Thus, chromatin remodeling facilitates transcriptional activation and repression across the circadian cycle to regulate rhythmic gene expression. Collectively, epigenetic regulation of chromatin, transcriptional activation and repression, and posttranslational modifications of repressor proteins establish period and amplitude of the self-sustained core clock oscillation. In addition, the circadian transcriptional drivers CLOCK:BMAL1 induce expression of other clock controlled genes, including numerous transcription factors and tissue specific output genes. This regulatory network allows for nycthemeral regulation of tissue function (Bozek et al., 2009; Sahar & Sassone-Corsi, 2013).
AhR Signaling
AhR is ligand-activated transcription factor of the basic helix-loop-helix-PAS (bHLH-PAS) family (Burbach et al., 1992). A large number of environmental pollutants, including polychlorinated dibenzo-p-dioxins, polychlorinated dibenzofurans, polychlorinated biphenyls and polycyclic aromatic hydrocarbons act through AhR to elicit their toxic effects (Denison & Nagy, 2003). Among those anthropogenic chemicals, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and β-Naphthoflavone (BNF) are prototypic AhR agonists with the highest affinity (Lindén et al., 2010; Tian et al., 2015). Dietary AhR ligands include, but may not be limited to, indole metabolites from cruciferous plants and flavonoids found in fruits and vegetables (Denison & Nagy, 2003). Arachidonic acid metabolites, equilenin, heme metabolites, indigoids and photoproducts of tryptophan metabolism are also endogenous ligands (Heath-Pagliuso et al., 1998; Nguyen & Bradfield, 2008). Finally, AhR may be activated in a ligand-independent manner by cyclic 3’5’ adenosine monophosphate (cAMP)-dependent activation of protein kinase A. AhR translocation into the nucleus in response to cAMP-signaling directs transcriptional activation that deviates significantly from xenobiotic-dependent events; cAMP-dependent activation may inhibit heterodimerization with ARNT (Oesch-Bartlomowicz et al., 2005). AhR responsiveness to a variety of man-made, natural and endogenous compounds, as well as its evolutionary conservation, raises important questions regarding its function in health and disease.
Beyond its role in xenobiotic metabolism, the AhR has physiologically relevant functions. Through multiple mechanisms, the versatile nuclear receptor AhR affects liver and vascular development, cell cycle progression, immune function and circadian rhythm (Jaeger, Khazaal, et al., 2017; Larigot et al., 2018). Genomic AhR signaling was elucidated experimentally using xenobiotic ligands (Figure 1A). In the absence of ligand, AhR partitions to the cytoplasm, complexed with two heat shock chaperones (hsp90), immunophilin-like protein X-associated protein 2 (XAP2) and the co-chaperone, p23 (Petrulis & Perdew, 2002). Hsp90 retains the structure of AhR needed for ligand binding and blocks a nuclear localization signal. XAP2 and p23 maintain the stability of the hsp90 complex (Guyot et al., 2013). Conformational change following ligand binding exposes a nuclear localization signal, which allows translocation of the AhR complex into the nucleus (Larigot et al., 2018). After the ligand is released, AhR heterodimerizes with aryl hydrocarbon receptor nuclear translocator (ARNT) in the nucleus, which subsequently targets AhR:ARNT to specific xenobiotic response elements (XRE) in target gene promoters. AhR:ARNT dimerization occurs through PAS domains, a feature shared with several circadian clock proteins. PAS-A and PAS-B regions act as an interactive surface for heterodimer and homodimer formation, interactions with hsp90, and ligand binding (Gu et al., 2000; McGuire et al., 2001). XRE binding and transcriptional initiation is facilitated by AhR:ARNT interactions with histone acetyltransferases and chromatin remodeling factors (Ko et al., 1996). A canonical AhR target gene is the Cytochrome P4501a1 (Cyp1a1), a phase I metabolizing enzyme that metabolizes polycyclic aromatic compounds and other P450 enzymes are canonical AhR:ARNT targets. In addition, AhR controls non-P450 transcripts that are involved in regulation of gene expression, drug metabolism, lipid metabolism, nucleic acid metabolism and small molecule biochemistry (Lo & Matthews, 2012). After DNA binding, a nuclear export signal signals AhR to exit the nucleus, where it is degraded by the 26S proteasome (Davarinos & Pollenz, 1999). AhR expression is attenuated by a negative feedback loop with the AhR repressor gene (AhRR), which is a target gene of AhR:ARNT (Baba et al., 2001; Haarmann-Stemmann & Abel, 2006). Although unable to bind ligand, AhRR inhibits AhR signal transduction by competing with AhR for ARNT dimerization and XRE binding, thereby acting as a transcriptional repressor (Figure 1A) (Gu et al., 2000; Haarmann-Stemmann & Abel, 2006).
Figure 1. AhR signaling pathways.

A. In genomic signaling, unbound aryl hydrocarbon receptor (AhR) typically partitions to the cytoplasm in a complex with heat shock protein 90 (hsp90), p23 and XAP. Ligand binding releases XAP and uncovers a nuclear localization signal. After translocation into the nucleus, AhR binds aryl hydrocarbon nuclear transporter (ARNT) and releases the hsp90/p23 complex for recycling into the cytoplasm. AhR:ARNT binds xenobiotic response elements (XREs) to drive transcription of target genes. Cytochrome P450 enzymes, particularly Cyp1A1 and Cyp1B1 are prototypical target genes that feed back to degrade xenobiotic ligands. AhR also induces expression of the aryl hydrocarbon receptor repressor (AHRR), which feeds back to inhibit AhR:ARNT-induced transcription. In addition AhR:ARNT activates other target genes involved in cell division and differentiation, and metabolism. B. As physiological roles for AhR are revealed, the importance of non-genomic signaling through AhR are becoming apparent. AhR can inhibit activity of other nuclear receptors, including estrogen receptor (ER), androgen receptor (AR), thyroid hormone receptor (TR), Wnt/β-catenin and nuclear factor kappa B (NFκB) by either binding directly to receptor proteins or by binding and diverting the action of coactivators. AhR promotes increases in intracellular Ca2+ by actions on membrane Ca2+ channels or through release from the endoplasmic reticulum, leading to increased arachidonic acid and inflammation. AhR can also increase MAPK activity, which can synergize with Ca2+ to promote inflammation through production of arachidonic acid. MAPK activity also mediates AhR’s regulation of cellular senescence, apoptosis and proliferation.
Although genomic AhR signaling is essential to the toxicological effects of dioxins, more recent studies highlight the physiological relevance of AhR, which may be arbitrated through a combination of genomic and non-genomic mechanisms. Pathways downstream of AhR activation may be influenced by ligand affinity, cell-type and other environmental factors. Microarray and genome-wide analysis of XRE sequences have identified numerous genes regulated by AhR (Tijet et al., 2006; Boutros et al., 2009; Dere, Lee, et al., 2011; Dere, Lo, et al., 2011; Lo & Matthews, 2012). Formation of heterodimers with alternative partners allows AhR to target diverse signaling pathways. Alternative signaling through AhR includes genomic cross-talk with other nuclear receptors, and non-genomic regulation of cell cycle and mitogen-activated protein kinase (MAPK) cascades, modulation of the immune system and activation of immediate early genes (Matsumura, 2009; Patel et al., 2009; Tanos et al., 2012).
AhR cross-talk with other nuclear receptors, including those for steroid hormones, thyroid hormones and retinoids involves multiple mechanisms (Figure 1B) (Widerak et al., 2006; Swedenborg & Pongratz, 2010). Perhaps the best explored is the interaction with the estrogen receptor (ER), where both classical and non-classical mechanisms contribute to inhibition of the estrogen response by AhR activation (Beischlag & Perdew, 2005; Khan et al., 2006; Swedenborg & Pongratz, 2010). AhR:ARNT act as transcriptional repressors at inhibitory dioxin response elements (iDRE) to block activation of certain genes by ER, at least in part by blocking binding of ER to its DNA recognition site (Safe et al., 1998; Wang et al., 2001). In addition, AhR may directly bind co-activator proteins, including p300 and cAMP response element binding protein (CBP), rendering them unavailable as co-activators in ER-mediated transcription (Beischlag & Perdew, 2005; Ma et al., 2009). Formation of AhR:ARNT complexes decreases availability for ARNT to complex with ER, which inhibits ER beta transcriptional activation (Rüegg et al., 2008). Direct binding of AhR with ER diverts the complex away from estrogen-responsive elements (EREs) and towards AhR-responsive genes (Ahmed et al., 2009). Thus, AhR interacts at multiple levels to disrupt ER signaling.
AhR also interacts with various intracellular signaling pathways, including protein kinases, calcium signaling, and others, in a non-genomic manner (Figure 1B) (Haarmann-Stemmann & Abel, 2006; Matsumura, 2009; Larigot et al., 2018). Activation of AhR promotes increased intracellular calcium and subsequent activation of arachidonic acid production and MAPK signaling (Tsai et al., 2014). In breast cancer cells, AhR activation promotes phosphorylation of MAPK and inhibits phosphorylation of the phosphotidyl inositol 3 kinase/protein kinase B (PI3K/AKT) pathway leading to cell cycle arrest and cell senescence (Wang, Xu, et al., 2014). Similarly, AhR inhibits proliferation and differentiation of osteoblasts through MAPK phosphorylation (Yu et al., 2014). Finally, AhR promotes germ cell apoptosis through activation of extracellular signal-regulated kinase (ERK1/2), p38 and c-jun N-terminal kinase (JNK) (Banerjee et al., 2016). In addition, activated AhR modifies cellular adhesion by disrupting focal adhesion points (Tomkiewicz et al., 2013). Thus, AhR acts as a multifaceted hub to regulate intracellular signaling and numerous physiological processes, including inflammation, cell adhesion, proliferation, cell senescence and death, as well as modifying the transcriptional regulation of other nuclear receptors. Physiological functions of AhR may include interactions with the circadian clock.
Regulation of AhR Signaling by the Circadian Clock
AhR, Arnt and Cyp1a1 display variation with 24-h periodicity (Shimba & Watabe, 2009; Wang, Zhang, et al., 2014). AhR transcripts peak at the time of lights-off in the SCN and 4 h earlier in liver. Oscillations persist under constant darkness conditions, albeit with slightly reduced amplitude in the SCN; the similarity to changes in the pattern of Bmal1 suggest the potential for co-regulation of these transcripts (Mukai et al., 2008). The AhR target gene, Cyp1a1, also shows circadian variation, albeit out of phase with AhR (4–8 h delay) (Mukai et al., 2008). Nycethemeral expression of AhR protein demonstrates diurnal changes in the availability of AhR for activation (Richardson et al., 1998; Shimba & Watabe, 2009), which implies that sequelae associated with AhR activation may also be regulated by the circadian clock. Transcripts of AhR target genes, Cyp1a1, Cyp1b1, and Arnt, although expressed at low levels without stimulation, are diurnally rhythmic with increased expression during the day (Richardson et al., 1998; Huang et al., 2002; Mukai et al., 2008). Induction of the AhR target genes Cyp1a1 and Cyp1b1 in response to a prototypical AhR ligand provides evidence for differences in AhR responsiveness that are driven by the clock (Qu et al., 2007, 2009, 2010). Cyp1a1 and Cyb1b1 are induced by AhR activation regardless of the time of day, however, responsiveness is significantly enhanced when exposure occurs at night (Qu et al., 2007, 2010). Variation in induction of Cyp1a1 is abolished in Per1 and/or Per2 mutant mice (Qu et al., 2009, 2010). Furthermore, oscillations of AhR mRNA and time-dependent sensitivity to agonists are absent in CLOCK mutant mice (Tanimura et al., 2011). Although these data suggest that the clock is an important regulator of AhR, molecular mechanisms underlying these effects remain largely unknown.
Molecular Interactions between AhR and Clock Components
PAS domains are structural motifs that govern protein-protein interactions that ultimately target gene transcription in response to environmental change (McIntosh et al., 2010). PAS domains in AhR bind ligands and regulate interaction with partners that are required to direct it to specific sequences on the promoters of target genes (Reisz-Porszasz et al., 1994; McGuire et al., 2001). PAS domains in circadian clock proteins, including CLOCK, BMAL1 and PER, likewise facilitate protein-protein interactions that determine transcriptional activity to regulate molecular rhythmicity. PAS domain-containing proteins are notably promiscuous in both their ligand binding and selectivity of dimerization partners, which allows for multivariate protein-protein interactions and significant crosstalk among intracellular signaling pathways. The classic AhR partner, ARNT shares significant sequence similarity to BMAL1, including similar intron/exon splice patterns and conservation of five exons that compose the PAS domain (Yu et al., 1999). Known promiscuity of PAS interactions, coupled with conserved similarities to clock-relevant proteins, suggests that AhR may interact with clock proteins to influence clock function. The aggregate data suggest that interactions between the clock and AhR signaling are reciprocal and significant.
Activation of AhR alters circadian clock gene transcripts and suppresses molecular rhythms (Garrett & Gasiewicz, 2006; Mukai et al., 2008; Xu et al., 2013). Reciprocally, genetic alteration of the circadian clock influences AhR signaling and sensitivity to agonist-induced activation of AhR and AhR target genes (Qu et al., 2007, 2009, 2010). Nycthemeral expression of AhR mRNA and protein levels is regulated by CLOCK:BMAL1 interactions at Ebox elements within the AhR promoter (Garrison & Denison, 2000; Huang et al., 2002; Zhang et al., 2009). Molecular mechanisms by which AhR is regulated by the PER complex remain unknown.
AhR may directly interact with core clock proteins to affect the molecular clockworks. Subsequent to ligand activation, AhR can form multiple heterodimers. The dimerization partner determines the target site for DNA binding and transcriptional activity. AhR:ARNT heterodimers bind XRE elements to drive transcription of target genes. In contrast, AhR may also interact with BMAL1, which has high homology with ARNT (Tischkau et al., 2011; Xu et al., 2013). The AhR:BMAL1 heterodimer disrupts CLOCK:BMAL1 activation at Eboxes on the Per1 promoter. The AhR interaction with BMAL1 displaces CLOCK from the complex and acts as a repressor of Ebox-dependent Per1 transcription, thereby dampening and delaying the Per1 rhythm (Xu et al., 2013). Inhibition of Ebox function provides a mechanism for dampening the amplitude and perhaps delaying the phase of the endogenous Per1 rhythm (Figure 2A).
Figure 2. Mechanisms of crosstalk between AhR and the molecular clock.

A. Activated AhR can physically interact with BMAL1, thereby reducing CLK:BMAL1 interactions. AHR:BMAL1 heterodimers bind to Ebox elements in the Per1 promoter, where they act to repress Per1 transcription. In addition, AhR inhibits light-induced Per1 expression through activation of JNK, which blocks CREB:CBP activity at Cre elements in the Per1 promoter. B. Rhythmic transcription of AhR is driven by CLK:BMAL1 at EBox promoter elements. PER1 and PER2 inhibit AhR transcript levels. Although the molecular mechanism has not been determined, PERs can inhibit CLK:BMAL1 function at the EBox.
AhR activation may also affect Per levels through an Ebox-independent mechanism. CRE elements mediate acute induction of Per1 transcription in the SCN in response to light (Travnickova-Bendova et al., 2002; Tischkau et al., 2003). Light-induced glutamatergic neurotransmission increases intracellular calcium and activation of MAPK, phosphorylation of cAMP regulatory element binding protein (CREB) and CRE-mediated transcription of Per1 (Ginty et al., 1993; Obrietan et al., 1998). Similarly, AhR activation has extensive effects on MAPK signaling (Tan et al., 2002; Ma et al., 2009). AhR agonists can activate JNK through AhR (Weiss et al., 2005, 2008; Diry et al., 2006). AhR activation blocks CRE-dependent Per1 transcription without affecting phosphorylation of CREB. Rather, AhR phosphorylates and activates JNK, which represses CRE-mediated transcriptional activity (Zhang et al., 2004; Xu et al., 2013). Suppression of CRE-mediated Per1 transcription provides a non-genomic mechanism for AhR to influence responsiveness of the molecular clockworks to phase resetting signals (Figure 2A).
AhR Regulation of Circadian Rhythmicity
Mice bearing a germ-line deletion of the AhR (AhRKO) provide a model for investigation of interactions between AhR and the circadian clock. AhRKO mice readily entrain to a light/dark cycle, re-entrain their clock in response to changes in the timing of light/dark cycles, and maintain a free-running period similar to wild-type littermates (Mukai et al., 2008). However, increased variability in circadian parameters, which is commonly observed after deletion of genes associated with clock stability, has been observed. AhRKO mice display increased daytime activity, variability in activity onset and altered patterns of nocturnal activity compared to wild-type mice (S. Tischkau, unpublished observations). Common to the C57BL/6 strain, AhRKO mice have a period of decreased activity the night, which is often referred to as a “nap”. The timing of nocturnal inactivity occurs 1–2 h earlier in the AhRKO mice, similar to B6.D2NAahrd/j mice, which are genetically altered in the region that includes the AhR locus (Hofstetter et al., 2007). The aggregate behavioral data suggest a modulatory effect of AhR on clock function, which is similar to the type of effect of other nuclear receptors, such as REV-ERBα or RORa (Sato et al., 2004; Akashi & Takumi, 2005).
Activation of AhR also impacts circadian timing. TCDD exposure alters circadian rhythms in feeding behavior, locomotor activity, hormone levels and gene expression (Pohjanvirta et al., 1996; Miller et al., 1999; Garrett & Gasiewicz, 2006; Xu et al., 2010). Locomotor activity is phase-advanced by low dose TCDD treatment, whereas high doses can cause rhythm splitting and arrythmicity (Miller et al., 1999). Rhythms of clock gene transcripts, specifically Per1 and Bmal1, and the PER1 proteins are also perturbed in the SCN of TCDD-exposed mice (Mukai et al., 2008). Similarly, large doses of TCDD alter the timing of food intake, such that 50% of daily food consumption occurs during the early morning in nocturnal rats (Pohjanvirta & Tuomisto, 1990). Unfortunately, that study did not explore locomotor activity, which may account for the changes in feeding. However, these results definitively demonstrate that AhR activation alters the timing of food intake; treated animals seemed to be unable to identify the appropriate time to eat. Effects of AhR activation on plasma hormones indicate a similar effect on clock-regulated function. Corticosterone, prolactin, thyroid hormone and melatonin rhythms are attenuated after AhR activation (Jones et al., 1987; Pohjanvirta et al., 1989). The common denominator is that rhythmicity of each of these factors is controlled by the circadian clock in the SCN. These studies indicate that acute and chronic activation of AhR can affect physiological rhythms.
Perhaps more important than the consequences of AhR activation on central clock function of the SCN are the effects of AhR on circadian rhythmicity in peripheral tissues, where AhR is more highly expressed. Genomic depletion of AhR has a greater effect on rhythmicity in peripheral tissues compared to the SCN. AhRKO mice have increased amplitude of clock gene rhythms, which may protect from environmentally induced circadian disruptions incurred by jet lag or high fat diet (Jaeger, Xu, et al., 2017). Conversely, AhR activation suppresses amplitude and delays phase of Per1 and Bmal1 rhythms in the liver (Mukai et al., 2008; Xu et al., 2010, 2013). Similarly, AhR activation subsequent to TCDD administration significantly altered the phase, period and amplitude of circadian rhythms of Per1, Per2, as well as the numbers of antigenically and functionally defined hematopoietic progenitor cell classes in bone marrow (Garrett & Gasiewicz, 2006). Because cyclic expression of Per is required for rhythmicity, and constitutive or persistently amplified expression of core clock genes can alter period or completely disrupt rhythms, the effects of AhR on cycling of core clock gene transcripts may be important (Zeng et al., 1994; Wang & Tobin, 1998; Kadener et al., 2008).
Physiologically, AhR interactions with the clock may provide a fundamental integration of cellular metabolism. AhR activation alters the expression of genes associated with cholesterol and fatty acid synthesis, glucose metabolism and circadian rhythm in the liver (Sato et al., 2008). Many of the same genes affected by AhR activation are controlled physiologically by the circadian clock. For example, 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR), the rate limiting enzyme in the synthesis of cholesterol, as well as the sterol element binding protein (SREBP), which is important in the regulation of sterol synthesis, are expressed with circadian periodicity under physiological conditions and downregulated by AhR activation (Brewer et al., 2005; Acimovic et al., 2008; Sato et al., 2008; Le Martelot et al., 2009; Matsumoto et al., 2010). These data make a compelling case that AhR interacts with the circadian system to regulate metabolic outputs. AhR activation weakens rhythm amplitude, whereas its inhibition bolsters rhythm amplitude (Figure 3). Although the physiological consequences of amplitude regulation remain to be determined, dampened amplitudes are associated with poor metabolic outcomes. Depletion of AhR increases healthy metabolic function and protects against the detrimental effects of poor diet (Xu et al., 2015; Jaeger, Xu, et al., 2017). It seems entirely plausible that these events are intricately connected.
Figure 3. Physiological relevance of AhR interactions with the Per1 promoter.

CLK:BMAL1 drive the endogenous rhythm of many transcripts in WT mice, including the Pers, AhR and Cytochrome P450’s (Cyps) and metabolically important genes such as 3-Hydroxy-3-Methylglutaryl-CoA Reductase (HMGCR) and sterol-regulatory element binding protein (SREBP). When AhR is deleted, the amplitude of circadian oscillations is increased. AhR activation inhibits CLK:BMAL1-mediated transcription, thereby suppressing amplitude and delays phase of target gene transcripts. Suppression of and changes of phase are associated with disease process including sleep disorders, metabolic syndrome, cancers and cardiovascular disease.
Cross-talk between AhR Signaling and Light Signaling in the Circadian Clock
Perception of the environmental presence of light and darkness is the perhaps most important function of the SCN. The SCN recognizes light at night as an error in timing and sets in motion a series of events to realign the clock with the environment. Light in the early night is perceived the same as an extension of daytime; thus, the SCN clock will respond by delaying the rhythm the next day. Alternatively, light in the late night is interpreted by the SCN as an early beginning to the forthcoming day. The clock responds to late night light by moving ahead, or phase advancing, on the ensuing day. Mechanisms that underlie clock resetting in response to changes in environmental light have been intensely investigated (reviewed in Antle et al., 2009; Golombek & Rosenstein, 2010). Essentially, phase resetting of the SCN clock in response to light proceeds through activation of NMDA-type glutamate receptors on retinorecipient SCN neurons, leading to an influx of calcium, activation of MAPKs and CREB, and ultimately to induction of the clock gene Per1. Although glutamate is the primary neurotransmitter that mediates the effect of light on the SCN, numerous other signals can interact with the glutamatergic input to modulate the light signal. For example, pituitary adenylate cyclase activating peptide (PACAP) interacts with glutamate to increase the magnitude of phase delays and to attenuate phase advances (Chen et al., 1999). Serotonin, or its precursor, tryptophan also acts to temper the magnitude of light and glutamate-induced responses in the SCN (Glass et al., 1995; Quintero & McMahon, 1999; Sterniczuk et al., 2008).
6-formylindolo[3,2-b]carbazole (FICZ), a high affinity AhR agonist physiologically derived from tryptophan metabolism (Helferich & Denison, 1991; Heath-Pagliuso et al., 1998; Bergander et al., 2004) blocks the glutamate-induced phase resetting of the SCN clock, in an AhR-dependent manner, similar to the effects of serotonin (Mukai & Tischkau, 2007). Activation of AhR with other ligands also attenuates light/glutamate-induced clock resetting (Xu et al., 2013). Furthermore, AhRKO mice have an increased responsiveness to light-induced phase resetting. These data are consistent with AhR acting as a modulatory brake on the circadian clockworks. Activation attenuates circadian clock responses, whereas removal of AhR activity from the system accentuates circadian responses to external signals.
Collectively, this provides compelling evidence for crosstalk between AhR signaling and the circadian clock.
Conclusions
Adaptation to environmental change is critical to survival. On a planet that rotates about its own axis creating a day that is consistently 24 hours in length, the value of measuring time on a daily scale is reflected in the incorporation of an internal clock into the genetic architecture of nearly all organisms. A robust defense in the face of exposure to environmental toxins, especially those present in food, is similarly adaptive and critical to survival. The bHLH-PAS domain family of proteins is central to regulation of each of these critical biological processes, which intersect in a complex and reciprocal manner. Disruption of clock function and alteration of AhR signaling contribute to a vast array of disease processes that include metabolic dysfunction and cancers. Components of the AhR signaling pathway are expressed in a diurnal pattern. Moreover, activation of AhR pathway is influenced by the circadian clock; sensitivity of the pathway to activation by high affinity agonists varies across the day. Conversely, both activation and downregulation of the AhR pathway alters the normal rhythmicity (Figure 3). How AhR activation impacts physiological rhythmicity remains to be determined. Interestingly, AhR activation has greater effects on clock processes in peripheral tissues, which may create discordance between the SCN master clock and clocks in other organs. The clock gene, Per1, appears to be central to the intersection of AhR and circadian signaling, although further studies are required to dissect molecular interactions. A better understanding of the crosstalk will provide insight into the physiological role of the AhR, as well as provide novel insight into the pathological events associated with clock disruption.
Acknowledgments
This work funded in part by NIH grant ES017774.
List of Abbreviations
- ACTH
Adrenocorticotropic hormone
- AhR
Aryl hydrocarbon receptor
- AhRKO
aryl hydrocarbon receptor knockout
- AhRR
Aryl hydrocarbon receptor repressor
- AKT
protein kinase B
- ARNT
aryl hydrocarbon receptor nuclear transporter
- bHLH
basic helix-loop-helix
- BMAL1
Brain muscle ARNT-like 1
- BNF
beta-napthoflavone
- Ca2+
calcium
- cAMP
cyclic 3’5’ -adenosine monophosphate
- CBP
CREB binding proten
- CLOCK/CLK
Circadian locomotor output cycles kaput
- CRE
cAMP response element
- CREB
cAMP response element binding protein
- Cry
Cryptochrome genes
- Cyp1a1/1b1
cytochrome P450 enzyme 1a1, 1b1
- DBP
D site of albumin promoter-binding protein
- ER
estrogen receptor
- ERK1/2
extracellular signal-regulated kinase 1/2
- FBLX3 or 21
F-Box and Leucine Rich Repeat Protein 3 or 21
- FICZ
6-Formylindolo[3,2-b]carbazole
- GABA
Gamma amino-butyric acid
- h
Hour
- HAT
histone acetyl transferase
- HMGCR
3-Hydroxy-3-Methylglutaryl-CoA Reductase
- Hsp90
heat shock protein 90
- JNK
c-jun N-terminal kinase
- MAPK
mitogen activated protein kinase
- MLL1
mixed lineage leukemia 1
- NFIL
Nuclear factor interleukin 3 regulated
- NFκB
Nuclear factor kappa B
- NMDA
N methyl d-aspartate
- PACAP
pituitary adenylate cyclase activating peptide
- PAS
Period-Aryl hydrocarbon receptor nuclear transporter-simpleminded
- Per
Period genes
- PI3K
Phosphatidylinositide 3-kinase
- PVN
Hypothalamic paraventricular nucleus
- PVT
Paraventricular nucleus of the thalamus
- RNA
Ribonucleic acid
- ROR
Retinoid-related orphan receptor
- SCN
Suprachiasmatic nucleus
- SREBP
sterol regulatory element-binding protein
- TCDD
2,3,7,8 Tetrachlorodibenzo-p-dioxin
- VLPO
Ventrolateral preoptic area
- XAP2
X associated protein 2
- XRE
xenobiotic response element
Footnotes
Conflict of Interest
The author has no conflicts to disclose.
References
- Acimovic J, Fink M, Pompon D, Bjorkhem I, Hirayama J, Sassone-Corsi P, Golicnik M, & Rozman D (2008) CREM modulates the circadian expression of CYP51, HMGCR and cholesterogenesis in the liver. Biochem Biophys Res Commun, 376, 206–210. [DOI] [PubMed] [Google Scholar]
- Ahmed S, Valen E, Sandelin A, & Matthews J (2009) Dioxin increases the interaction between aryl hydrocarbon receptor and estrogen receptor alpha at human promoters. Toxicol Sci, 111, 254–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akashi M & Takumi T (2005) The orphan nuclear receptor RORalpha regulates circadian transcription of the mammalian core-clock Bmal1. Nat Struct Mol Biol, 12, 441–448. [DOI] [PubMed] [Google Scholar]
- Antle MC, Smith VM, Sterniczuk R, Yamakawa GR, & Rakai BD (2009) Physiological responses of the circadian clock to acute light exposure at night. Rev Endocr Metab Disord, 10, 279–291. [DOI] [PubMed] [Google Scholar]
- Arble DM, Bass J, Behn CD, Butler MP, Challet E, Czeisler C, Depner CM, Elmquist J, Franken P, Grandner MA, Hanlon EC, Keene AC, Joyner MJ, Karatsoreos I, Kern PA, Klein S, Morris CJ, Pack AI, Panda S, Ptacek LJ, Punjabi NM, Sassone-Corsi P, Scheer FA, Saxena R, Seaquest ER, Thimgan MS, Van Cauter E, & Wright KP (2015) Impact of Sleep and Circadian Disruption on Energy Balance and Diabetes: A Summary of Workshop Discussions. Sleep, 38, 1849–1860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baba T, Mimura J, Gradin K, Kuroiwa A, Watanabe T, Matsuda Y, Inazawa J, Sogawa K, & Fujii-Kuriyama Y (2001) Structure and expression of the Ah receptor repressor gene. J Biol Chem, 276, 33101–33110. [DOI] [PubMed] [Google Scholar]
- Banerjee B, Nandi P, Chakraborty S, Raha S, Sen PC, & Jana K (2016) Resveratrol ameliorates benzo(a)pyrene-induced testicular dysfunction and apoptosis: involvement of p38 MAPK/ATF2/iNOS signaling. J Nutr Biochem, 34, 17–29. [DOI] [PubMed] [Google Scholar]
- Bass JT (2017) The circadian clock system’s influence in health and disease. Genome Med, 9, 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beischlag TV & Perdew GH (2005) ER alpha-AHR-ARNT protein-protein interactions mediate estradiol-dependent transrepression of dioxin-inducible gene transcription. J Biol Chem, 280, 21607–21611. [DOI] [PubMed] [Google Scholar]
- Bergander L, Wincent E, Rannug A, Foroozesh M, Alworth W, & Rannug U (2004) Metabolic fate of the Ah receptor ligand 6-formylindolo[3,2-b]carbazole. Chem Biol Interact, 149, 151–164. [DOI] [PubMed] [Google Scholar]
- Bhadra U, Thakkar N, Das P, & Pal Bhadra M (2017) Evolution of circadian rhythms: from bacteria to human. Sleep Med, 35, 49–61. [DOI] [PubMed] [Google Scholar]
- Boutros PC, Bielefeld KA, Pohjanvirta R, & Harper PA (2009) Dioxin-dependent and dioxin-independent gene batteries: comparison of liver and kidney in AHR-null mice. Toxicol Sci, 112, 245–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bozek K, Relógio A, Kielbasa SM, Heine M, Dame C, Kramer A, & Herzel H (2009) Regulation of clock-controlled genes in mammals. PLoS One, 4, e4882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brewer M, Lange D, Baler R, & Anzulovich A (2005) SREBP-1 as a transcriptional integrator of circadian and nutritional cues in the liver. J Biol Rhythms, 20, 195–205. [DOI] [PubMed] [Google Scholar]
- Buijs RM, la Fleur SE, Wortel J, Van Heyningen C, Zuiddam L, Mettenleiter TC, Kalsbeek A, Nagai K, & Niijima A (2003) The suprachiasmatic nucleus balances sympathetic and parasympathetic output to peripheral organs through separate preautonomic neurons. J Comp Neurol, 464, 36–48. [DOI] [PubMed] [Google Scholar]
- Buijs RM, Wortel J, & Hou YX (1995) Colocalization of gamma-aminobutyric acid with vasopressin, vasoactive intestinal peptide, and somatostatin in the rat suprachiasmatic nucleus. J Comp Neurol, 358, 343–352. [DOI] [PubMed] [Google Scholar]
- Buijs RM, Wortel J, Van Heerikhuize JJ, Feenstra MG, Ter Horst GJ, Romijn HJ, & Kalsbeek A (1999) Anatomical and functional demonstration of a multisynaptic suprachiasmatic nucleus adrenal (cortex) pathway. Eur J Neurosci, 11, 1535–1544. [DOI] [PubMed] [Google Scholar]
- Burbach KM, Poland A, & Bradfield CA (1992) Cloning of the Ah-receptor cDNA reveals a distinctive ligand-activated transcription factor. Proc Natl Acad Sci U A, 89, 8185–8189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cailotto C, La Fleur SE, Van Heijningen C, Wortel J, Kalsbeek A, Feenstra M, Pévet P, & Buijs RM (2005) The suprachiasmatic nucleus controls the daily variation of plasma glucose via the autonomic output to the liver: are the clock genes involved? Eur J Neurosci, 22, 2531–2540. [DOI] [PubMed] [Google Scholar]
- Chen D, Buchanan GF, Ding JM, Hannibal J, & Gillette MU (1999) Pituitary adenylyl cyclase-activating peptide: a pivotal modulator of glutamatergic regulation of the suprachiasmatic circadian clock. Proc Natl Acad Sci U A, 96, 13468–13473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng MY, Bullock CM, Li C, Lee AG, Bermak JC, Belluzzi J, Weaver DR, Leslie FM, & Zhou QY (2002) Prokineticin 2 transmits the behavioural circadian rhythm of the suprachiasmatic nucleus. Nature, 417, 405–410. [DOI] [PubMed] [Google Scholar]
- Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong L-W, DiTacchio L, Atkins AR, Glass CK, Liddle C, Auwerx J, Downes M, Panda S, & Evans RM (2012) Regulation of circadian behaviour and metabolism by REV-ERB-α and REV-ERB-β. Nature, 485, 123–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Curtis AM, Seo SB, Westgate EJ, Rudic RD, Smyth EM, Chakravarti D, FitzGerald GA, & McNamara P (2004) Histone acetyltransferase-dependent chromatin remodeling and the vascular clock. J Biol Chem, 279, 7091–7097. [DOI] [PubMed] [Google Scholar]
- Davarinos NA & Pollenz RS (1999) Aryl hydrocarbon receptor imported into the nucleus following ligand binding is rapidly degraded via the cytosplasmic proteasome following nuclear export. J Biol Chem, 274, 28708–28715. [DOI] [PubMed] [Google Scholar]
- de la Iglesia HO, Meyer J, & Schwartz WJ (2003) Lateralization of circadian pacemaker output: Activation of left- and right-sided luteinizing hormone-releasing hormone neurons involves a neural rather than a humoral pathway. J. Neurosci. Off. J. Soc. Neurosci, 23, 7412–7414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denison MS & Nagy SR (2003) Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol, 43, 309–334. [DOI] [PubMed] [Google Scholar]
- Dere E, Lee AW, Burgoon LD, & Zacharewski TR (2011) Differences in TCDD-elicited gene expression profiles in human HepG2, mouse Hepa1c1c7 and rat H4IIE hepatoma cells. BMC Genomics, 12, 193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dere E, Lo R, Celius T, Matthews J, & Zacharewski TR (2011) Integration of genome-wide computation DRE search, AhR ChIP-chip and gene expression analyses of TCDD-elicited responses in the mouse liver. BMC Genomics, 12, 365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dibner C, Schibler U, & Albrecht U (2010) The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu Rev Physiol, 72, 517–549. [DOI] [PubMed] [Google Scholar]
- Diry M, Tomkiewicz C, Koehle C, Coumoul X, Bock KW, Barouki R, & Transy C (2006) Activation of the dioxin/aryl hydrocarbon receptor (AhR) modulates cell plasticity through a JNK-dependent mechanism. Oncogene, 25, 5570–5574. [DOI] [PubMed] [Google Scholar]
- DiTacchio L, Le HD, Vollmers C, Hatori M, Witcher M, Secombe J, & Panda S (2011) Histone lysine demethylase JARID1a activates CLOCK-BMAL1 and influences the circadian clock. Science, 333, 1881–1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Do MT & Yau KW (2010) Intrinsically photosensitive retinal ganglion cells. Physiol Rev, 90, 1547–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doi M, Hirayama J, & Sassone-Corsi P (2006) Circadian regulator CLOCK is a histone acetyltransferase. Cell, 125, 497–508. [DOI] [PubMed] [Google Scholar]
- Ebling FJ (1996) The role of glutamate in the photic regulation of the suprachiasmatic nucleus. Prog Neurobiol, 50, 109–132. [DOI] [PubMed] [Google Scholar]
- Etchegaray JP, Lee C, Wade PA, & Reppert SM (2003) Rhythmic histone acetylation underlies transcription in the mammalian circadian clock. Nature, 421, 177–182. [DOI] [PubMed] [Google Scholar]
- Fernandez DC, Chang YT, Hattar S, & Chen SK (2016) Architecture of retinal projections to the central circadian pacemaker. Proc Natl Acad Sci U A, 113, 6047–6052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gachon F, Fonjallaz P, Damiola F, Gos P, Kodama T, Zakany J, Duboule D, Petit B, Tafti M, & Schibler U (2004) The loss of circadian PAR bZip transcription factors results in epilepsy. Genes Dev, 18, 1397–1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garrett RW & Gasiewicz TA (2006) The aryl hydrocarbon receptor agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin alters the circadian rhythms, quiescence, and expression of clock genes in murine hematopoietic stem and progenitor cells. Mol Pharmacol, 69, 2076–2083. [DOI] [PubMed] [Google Scholar]
- Garrison PM & Denison MS (2000) Analysis of the murine AhR gene promoter. J Biochem Mol Toxicol, 14, 1–10. [DOI] [PubMed] [Google Scholar]
- Gekakis N, Staknis D, Nguyen HB, Davis FC, Wilsbacher LD, King DP, Takahashi JS, & Weitz CJ (1998) Role of the CLOCK protein in the mammalian circadian mechanism. Science, 280, 1564–1569. [DOI] [PubMed] [Google Scholar]
- Gillette MU & Mitchell JW (2002) Signaling in the suprachiasmatic nucleus: selectively responsive and integrative. Cell Tissue Res, 309, 99–107. [DOI] [PubMed] [Google Scholar]
- Gillette MU & Tischkau SA (1999) Suprachiasmatic nucleus: the brain’s circadian clock. Recent Prog Horm Res, 54, 33–58; discussion 58–9. [PubMed] [Google Scholar]
- Ginty DD, Kornhauser JM, Thompson MA, Bading H, Mayo KE, Takahashi JS, & Greenberg ME (1993) Regulation of CREB phosphorylation in the suprachiasmatic nucleus by light and a circadian clock. Science, 260, 238–241. [DOI] [PubMed] [Google Scholar]
- Glass JD, Selim M, Srkalovic G, & Rea MA (1995) Tryptophan loading modulates light-induced responses in the mammalian circadian system. J Biol Rhythms, 10, 80–90. [DOI] [PubMed] [Google Scholar]
- Golombek DA & Rosenstein RE (2010) Physiology of circadian entrainment. Physiol Rev, 90, 1063–1102. [DOI] [PubMed] [Google Scholar]
- Gu YZ, Hogenesch JB, & Bradfield CA (2000) The PAS superfamily: sensors of environmental and developmental signals. Annu Rev Pharmacol Toxicol, 40, 519–561. [DOI] [PubMed] [Google Scholar]
- Guo H, Brewer JM, Lehman MN, & Bittman EL (2006) Suprachiasmatic regulation of circadian rhythms of gene expression in hamster peripheral organs: effects of transplanting the pacemaker. J Neurosci, 26, 6406–6412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guyot E, Chevallier A, Barouki R, & Coumoul X (2013) The AhR twist: ligand-dependent AhR signaling and pharmaco-toxicological implications. Drug Discov Today, 18, 479–486. [DOI] [PubMed] [Google Scholar]
- Haarmann-Stemmann T & Abel J (2006) The arylhydrocarbon receptor repressor (AhRR): structure, expression, and function. Biol Chem, 387, 1195–1199. [DOI] [PubMed] [Google Scholar]
- Hatori M & Panda S (2015) Response of peripheral rhythms to the timing of food intake. Methods Enzym, 552, 145–161. [DOI] [PubMed] [Google Scholar]
- Hattar S, Kumar M, Park A, Tong P, Tung J, Yau KW, & Berson DM (2006) Central projections of melanopsin-expressing retinal ganglion cells in the mouse. J Comp Neurol, 497, 326–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hattar S, Lucas RJ, Mrosovsky N, Thompson S, Douglas RH, Hankins MW, Lem J, Biel M, Hofmann F, Foster RG, & Yau KW (2003) Melanopsin and rod-cone photoreceptive systems account for all major accessory visual functions in mice. Nature, 424, 76–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heath-Pagliuso S, Rogers WJ, Tullis K, Seidel SD, Cenijn PH, Brouwer A, & Denison MS (1998) Activation of the Ah receptor by tryptophan and tryptophan metabolites. Biochemistry, 37, 11508–11515. [DOI] [PubMed] [Google Scholar]
- Helferich WG & Denison MS (1991) Ultraviolet photoproducts of tryptophan can act as dioxin agonists. Mol Pharmacol, 40, 674–678. [PubMed] [Google Scholar]
- Hermes ML, Coderre EM, Buijs RM, & Renaud LP (1996) GABA and glutamate mediate rapid neurotransmission from suprachiasmatic nucleus to hypothalamic paraventricular nucleus in rat. J Physiol, 496 ( Pt 3), 749–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirayama J, Sahar S, Grimaldi B, Tamaru T, Takamatsu K, Nakahata Y, & Sassone-Corsi P (2007) CLOCK-mediated acetylation of BMAL1 controls circadian function. Nature, 450, 1086–1090. [DOI] [PubMed] [Google Scholar]
- Hofstetter JR, Svihla-Jones DA, & Mayeda AR (2007) A QTL on mouse chromosome 12 for the genetic variance in free-running circadian period between inbred strains of mice. J Circadian Rhythms, 5, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang P, Ceccatelli S, & Rannug A (2002) A study on diurnal mRNA expression of CYP1A1, AHR, ARNT, and PER2 in rat pituitary and liver. Env. Toxicol Pharmacol, 11, 119–126. [DOI] [PubMed] [Google Scholar]
- Hughes ME, DiTacchio L, Hayes KR, Vollmers C, Pulivarthy S, Baggs JE, Panda S, & Hogenesch JB (2009) Harmonics of circadian gene transcription in mammals. PLoS Genet, 5, e1000442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hughes S, Jagannath A, Hankins MW, Foster RG, & Peirson SN (2015) Photic regulation of clock systems. Methods Enzym, 552, 125–143. [DOI] [PubMed] [Google Scholar]
- Jaeger C, Khazaal AQ, Xu C, Sun M, Krager SL, & Tischkau SA (2017) Aryl Hydrocarbon Receptor Deficiency Alters Circadian and Metabolic Rhythmicity. J Biol Rhythms, 32, 109–120. [DOI] [PubMed] [Google Scholar]
- Jaeger C, Xu C, Sun M, Krager S, & Tischkau SA (2017) Aryl hydrocarbon receptor-deficient mice are protected from high fat diet-induced changes in metabolic rhythms. Chronobiol Int, 34, 318–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones MK, Weisenburger WP, Sipes IG, & Russell DH (1987) Circadian alterations in prolactin, corticosterone, and thyroid hormone levels and down-regulation of prolactin receptor activity by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol, 87, 337–350. [DOI] [PubMed] [Google Scholar]
- Kadener S, Menet JS, Schoer R, & Rosbash M (2008) Circadian transcription contributes to core period determination in Drosophila. PLoS Biol, 6, e119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalsbeek A & Buijs RM (2002) Output pathways of the mammalian suprachiasmatic nucleus: coding circadian time by transmitter selection and specific targeting. Cell Tissue Res, 309, 109–118. [DOI] [PubMed] [Google Scholar]
- Kalsbeek A, la Fleur S, & Fliers E (2014) Circadian control of glucose metabolism. Mol Metab, 3, 372–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalsbeek A, Perreau-Lenz S, & Buijs RM (2006) A network of (autonomic) clock outputs. Chronobiol Int, 23, 521–535. [DOI] [PubMed] [Google Scholar]
- Kalsbeek A, Verhagen LAW, Schalij I, Foppen E, Saboureau M, Bothorel B, Buijs RM, & Pévet P (2008) Opposite actions of hypothalamic vasopressin on circadian corticosterone rhythm in nocturnal versus diurnal species. Eur. J. Neurosci, 27, 818–827. [DOI] [PubMed] [Google Scholar]
- Katada S & Sassone-Corsi P (2010) The histone methyltransferase MLL1 permits the oscillation of circadian gene expression. Nat Struct Mol Biol, 17, 1414–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khan S, Barhoumi R, Burghardt R, Liu S, Kim K, & Safe S (2006) Molecular mechanism of inhibitory aryl hydrocarbon receptor-estrogen receptor/Sp1 cross talk in breast cancer cells. Mol Endocrinol, 20, 2199–2214. [DOI] [PubMed] [Google Scholar]
- Ko CH, Yamada YR, Welsh DK, Buhr ED, Liu AC, Zhang EE, Ralph MR, Kay SA, Forger DB, & Takahashi JS (2010) Emergence of noise-induced oscillations in the central circadian pacemaker. PLoS Biol, 8, e1000513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ko HP, Okino ST, Ma Q, & Whitlock JP (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, 430–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kramer A, Yang FC, Snodgrass P, Li X, Scammell TE, Davis FC, & Weitz CJ (2001) Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling. Science, 294, 2511–2515. [DOI] [PubMed] [Google Scholar]
- Kuhlman SJ & McMahon DG (2006) Encoding the ins and outs of circadian pacemaking. J Biol Rhythms, 21, 470–481. [DOI] [PubMed] [Google Scholar]
- Kume K, Zylka MJ, Sriram S, Shearman LP, Weaver DR, Jin X, Maywood ES, Hastings MH, & Reppert SM (1999) mCRY1 and mCRY2 are essential components of the negative limb of the circadian clock feedback loop. Cell, 98, 193–205. [DOI] [PubMed] [Google Scholar]
- Lamia KA, Storch KF, & Weitz CJ (2008) Physiological significance of a peripheral tissue circadian clock. Proc Natl Acad Sci U A, 105, 15172–15177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lande-Diner L, Boyault C, Kim JY, & Weitz CJ (2013) A positive feedback loop links circadian clock factor CLOCK-BMAL1 to the basic transcriptional machinery. Proc Natl Acad Sci U A, 110, 16021–16026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larigot L, Juricek L, Dairou J, & Coumoul X (2018) AhR signaling pathways and regulatory functions. Biochim Open, 7, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Martelot G, Claudel T, Gatfield D, Schaad O, Kornmann B, Lo Sasso G, Moschetta A, & Schibler U (2009) REV-ERBalpha participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biol, 7, e1000181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee C, Etchegaray JP, Cagampang FR, Loudon AS, & Reppert SM (2001) Posttranslational mechanisms regulate the mammalian circadian clock. Cell, 107, 855–867. [DOI] [PubMed] [Google Scholar]
- Lehman MN, Silver R, Gladstone WR, Kahn RM, Gibson M, & Bittman EL (1987) Circadian rhythmicity restored by neural transplant. Immunocytochemical characterization of the graft and its integration with the host brain. J Neurosci, 7, 1626–1638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindén J, Lensu S, Tuomisto J, & Pohjanvirta R (2010) Dioxins, the aryl hydrocarbon receptor and the central regulation of energy balance. Front Neuroendocr, 31, 452–478. [DOI] [PubMed] [Google Scholar]
- Liu AC, Welsh DK, Ko CH, Tran HG, Zhang EE, Priest AA, Buhr ED, Singer O, Meeker K, Verma IM, Doyle FJ, Takahashi JS, & Kay SA (2007) Intercellular coupling confers robustness against mutations in the SCN circadian clock network. Cell, 129, 605–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lo R & Matthews J (2012) High-resolution genome-wide mapping of AHR and ARNT binding sites by ChIP-Seq. Toxicol Sci, 130, 349–361. [DOI] [PubMed] [Google Scholar]
- Lowrey PL & Takahashi JS (2011) Genetics of circadian rhythms in Mammalian model organisms. Adv Genet, 74, 175–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma C, Marlowe JL, & Puga A (2009) The aryl hydrocarbon receptor at the crossroads of multiple signaling pathways. EXS, 99, 231–257. [DOI] [PubMed] [Google Scholar]
- Matsumoto E, Ishihara A, Tamai S, Nemoto A, Iwase K, Hiwasa T, Shibata S, & Takiguchi M (2010) Time of day and nutrients in feeding govern daily expression rhythms of the gene for sterol regulatory element-binding protein (SREBP)-1 in the mouse liver. J Biol Chem, 285, 33028–33036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsumura F (2009) The significance of the nongenomic pathway in mediating inflammatory signaling of the dioxin-activated Ah receptor to cause toxic effects. Biochem Pharmacol, 77, 608–626. [DOI] [PubMed] [Google Scholar]
- McGuire J, Okamoto K, Whitelaw ML, Tanaka H, & Poellinger L (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, 41841–41849. [DOI] [PubMed] [Google Scholar]
- McIntosh BE, Hogenesch JB, & Bradfield CA (2010) Mammalian Per-Arnt-Sim proteins in environmental adaptation. Annu Rev Physiol, 72, 625–645. [DOI] [PubMed] [Google Scholar]
- Miller J, Settachan D, Frame L, & Dickerson R (1999) 2,3,7,8-tetrachlorodibenzo-p-dioxin phase advance the deer mouse (Peromyscus maniculatis) circadian rhythm by altering expression of clock proteins. Organohalogen Compd, 42, 23–28. [Google Scholar]
- Mitsui S, Yamaguchi S, Matsuo T, Ishida Y, & Okamura H (2001) Antagonistic role of E4BP4 and PAR proteins in the circadian oscillatory mechanism. Genes Dev, 15, 995–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore RY (2013) The suprachiasmatic nucleus and the circadian timing system. Prog Mol Biol Transl Sci, 119, 1–28. [DOI] [PubMed] [Google Scholar]
- Mukai M, Lin TM, Peterson RE, Cooke PS, & Tischkau SA (2008) Behavioral rhythmicity of mice lacking AhR and attenuation of light-induced phase shift by 2,3,7,8-tetrachlorodibenzo-p-dioxin. J Biol Rhythms, 23, 200–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukai M & Tischkau SA (2007) Effects of tryptophan photoproducts in the circadian timing system: searching for a physiological role for aryl hydrocarbon receptor. Toxicol Sci, 95, 172–181. [DOI] [PubMed] [Google Scholar]
- Nguyen LP & Bradfield CA (2008) The search for endogenous activators of the aryl hydrocarbon receptor. Chem Res Toxicol, 21, 102–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obrietan K, Impey S, & Storm DR (1998) Light and circadian rhythmicity regulate MAP kinase activation in the suprachiasmatic nuclei. Nat Neurosci, 1, 693–700. [DOI] [PubMed] [Google Scholar]
- Oesch-Bartlomowicz B, Huelster A, Wiss O, Antoniou-Lipfert P, Dietrich C, Arand M, Weiss C, Bockamp E, & Oesch F (2005) Aryl hydrocarbon receptor activation by cAMP vs. dioxin: divergent signaling pathways. Proc Natl Acad Sci U A, 102, 9218–9223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oishi K, Amagai N, Shirai H, Kadota K, Ohkura N, & Ishida N (2005) Genome-wide expression analysis reveals 100 adrenal gland-dependent circadian genes in the mouse liver. DNA Res, 12, 191–202. [DOI] [PubMed] [Google Scholar]
- Olivo D, Caba M, Gonzalez-Lima F, Vázquez A, & Corona-Morales A (2014) Circadian feeding entrains anticipatory metabolic activity in piriform cortex and olfactory tubercle, but not in suprachiasmatic nucleus. Brain Res, 1592, 11–21. [DOI] [PubMed] [Google Scholar]
- Oster H, Challet E, Ott V, Arvat E, de Kloet ER, Dijk DJ, Lightman S, Vgontzas A, & Van Cauter E (2017) The Functional and Clinical Significance of the 24-Hour Rhythm of Circulating Glucocorticoids. Endocr Rev, 38, 3–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patel RD, Murray IA, Flaveny CA, Kusnadi A, & Perdew GH (2009) Ah receptor represses acute-phase response gene expression without binding to its cognate response element. Lab Invest, 89, 695–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petrulis JR & Perdew GH (2002) The role of chaperone proteins in the aryl hydrocarbon receptor core complex. Chem Biol Interact, 141, 25–40. [DOI] [PubMed] [Google Scholar]
- Pohjanvirta R, Laitinen J, Vakkuri O, Lindén J, Kokkola T, Unkila M, & Tuomisto J (1996) Mechanism by which 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) reduces circulating melatonin levels in the rat. Toxicology, 107, 85–97. [DOI] [PubMed] [Google Scholar]
- Pohjanvirta R & Tuomisto J (1990) 2,3,7,8-Tetrachlorodibenzo-p-dioxin enhances responsiveness to post-ingestive satiety signals. Toxicology, 63, 285–299. [DOI] [PubMed] [Google Scholar]
- Pohjanvirta R, Tuomisto J, Linden J, & Laitinen J (1989) TCDD reduces serum melatonin levels in Long-Evans rats. Pharmacol Toxicol, 65, 239–240. [DOI] [PubMed] [Google Scholar]
- Preitner N, Damiola F, Lopez-Molina L, Zakany J, Duboule D, Albrecht U, & Schibler U (2002) The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell, 110, 251–260. [DOI] [PubMed] [Google Scholar]
- Qu X, Metz RP, Porter WW, Cassone VM, & Earnest DJ (2007) Disruption of clock gene expression alters responses of the aryl hydrocarbon receptor signaling pathway in the mouse mammary gland. Mol Pharmacol, 72, 1349–1358. [DOI] [PubMed] [Google Scholar]
- Qu X, Metz RP, Porter WW, Cassone VM, & Earnest DJ (2009) Disruption of period gene expression alters the inductive effects of dioxin on the AhR signaling pathway in the mouse liver. Toxicol Appl Pharmacol, 234, 370–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qu X, Metz RP, Porter WW, Neuendorff N, Earnest BJ, & Earnest DJ (2010) The clock genes period 1 and period 2 mediate diurnal rhythms in dioxin-induced Cyp1A1 expression in the mouse mammary gland and liver. Toxicol Lett, 196, 28–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quintero JE & McMahon DG (1999) Serotonin modulates glutamate responses in isolated suprachiasmatic nucleus neurons. J Neurophysiol, 82, 533–539. [DOI] [PubMed] [Google Scholar]
- Refinetti R (2015) Comparison of light, food, and temperature as environmental synchronizers of the circadian rhythm of activity in mice. J Physiol Sci, 65, 359–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reisz-Porszasz S, Probst MR, Fukunaga BN, & Hankinson O (1994) Identification of functional domains of the aryl hydrocarbon receptor nuclear translocator protein (ARNT). Mol Cell Biol, 14, 6075–6086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richardson VM, Santostefano MJ, & Birnbaum LS (1998) Daily cycle of bHLH-PAS proteins, Ah receptor and Arnt, in multiple tissues of female Sprague-Dawley rats. Biochem Biophys Res Commun, 252, 225–231. [DOI] [PubMed] [Google Scholar]
- Rüegg J, Swedenborg E, Wahlström D, Escande A, Balaguer P, Pettersson K, & Pongratz I (2008) The transcription factor aryl hydrocarbon receptor nuclear translocator functions as an estrogen receptor beta-selective coactivator, and its recruitment to alternative pathways mediates antiestrogenic effects of dioxin. Mol Endocrinol, 22, 304–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Safe S, Wang F, Porter W, Duan R, & McDougal A (1998) Ah receptor agonists as endocrine disruptors: antiestrogenic activity and mechanisms. Toxicol Lett, 102–103, 343–347. [DOI] [PubMed] [Google Scholar]
- Sahar S & Sassone-Corsi P (2013) The epigenetic language of circadian clocks. Handb Exp Pharmacol, 29–44. [DOI] [PubMed] [Google Scholar]
- Sato S, Shirakawa H, Tomita S, Ohsaki Y, Haketa K, Tooi O, Santo N, Tohkin M, Furukawa Y, Gonzalez FJ, & Komai M (2008) Low-dose dioxins alter gene expression related to cholesterol biosynthesis, lipogenesis, and glucose metabolism through the aryl hydrocarbon receptor-mediated pathway in mouse liver. Toxicol Appl Pharmacol, 229, 10–19. [DOI] [PubMed] [Google Scholar]
- Sato TK, Panda S, Miraglia LJ, Reyes TM, Rudic RD, McNamara P, Naik KA, FitzGerald GA, Kay SA, & Hogenesch JB (2004) A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron, 43, 527–537. [DOI] [PubMed] [Google Scholar]
- Shearman LP, Sriram S, Weaver DR, Maywood ES, Chaves I, Zheng B, Kume K, Lee CC, van der Horst GT, Hastings MH, & Reppert SM (2000) Interacting molecular loops in the mammalian circadian clock. Science, 288, 1013–1019. [DOI] [PubMed] [Google Scholar]
- Shimba S & Watabe Y (2009) Crosstalk between the AHR signaling pathway and circadian rhythm. Biochem Pharmacol, 77, 560–565. [DOI] [PubMed] [Google Scholar]
- Silver R, LeSauter J, Tresco PA, & Lehman MN (1996) A diffusible coupling signal from the transplanted suprachiasmatic nucleus controlling circadian locomotor rhythms. Nature, 382, 810–813. [DOI] [PubMed] [Google Scholar]
- St John PC, Hirota T, Kay SA, & Doyle FJ (2014) Spatiotemporal separation of PER and CRY posttranslational regulation in the mammalian circadian clock. Proc Natl Acad Sci U A, 111, 2040–2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sterniczuk R, Stepkowski A, Jones M, & Antle MC (2008) Enhancement of photic shifts with the 5-HT1A mixed agonist/antagonist NAN-190: intra-suprachiasmatic nucleus pathway. Neuroscience, 153, 571–580. [DOI] [PubMed] [Google Scholar]
- Sun X, Whitefield S, Rusak B, & Semba K (2001) Electrophysiological analysis of suprachiasmatic nucleus projections to the ventrolateral preoptic area in the rat. Eur J Neurosci, 14, 1257–1274. [DOI] [PubMed] [Google Scholar]
- Swedenborg E & Pongratz I (2010) AhR and ARNT modulate ER signaling. Toxicology, 268, 132–138. [DOI] [PubMed] [Google Scholar]
- Takahashi JS (2015) Molecular components of the circadian clock in mammals. Diabetes Obes Metab, 17 Suppl 1, 6–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takahashi JS (2017) Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet, 18, 164–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takahashi JS, Hong HK, Ko CH, & McDearmon EL (2008) The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat Rev Genet, 9, 764–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan Z, Chang X, Puga A, & Xia Y (2002) Activation of mitogen-activated protein kinases (MAPKs) by aromatic hydrocarbons: role in the regulation of aryl hydrocarbon receptor (AHR) function. Biochem Pharmacol, 64, 771–780. [DOI] [PubMed] [Google Scholar]
- Tanimura N, Kusunose N, Matsunaga N, Koyanagi S, & Ohdo S (2011) Aryl hydrocarbon receptor-mediated Cyp1a1 expression is modulated in a CLOCK-dependent circadian manner. Toxicology, 290, 203–207. [DOI] [PubMed] [Google Scholar]
- Tanos R, Patel RD, Murray IA, Smith PB, Patterson AD, & Perdew GH (2012) Aryl hydrocarbon receptor regulates the cholesterol biosynthetic pathway in a dioxin response element-independent manner. Hepatology, 55, 1994–2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian J, Feng Y, Fu H, Xie HQ, Jiang JX, & Zhao B (2015) The Aryl Hydrocarbon Receptor: A Key Bridging Molecule of External and Internal Chemical Signals. Env. Sci Technol, 49, 9518–9531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tijet N, Boutros PC, Moffat ID, Okey AB, Tuomisto J, & Pohjanvirta R (2006) Aryl hydrocarbon receptor regulates distinct dioxin-dependent and dioxin-independent gene batteries. Mol Pharmacol, 69, 140–153. [DOI] [PubMed] [Google Scholar]
- Tischkau SA, Jaeger CD, & Krager SL (2011) Circadian clock disruption in the mouse ovary in response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Lett, 201, 116–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tischkau SA, Mitchell JW, Tyan S-H, Buchanan GF, & Gillette MU (2003) Ca2+/cAMP response element-binding protein (CREB)-dependent activation of Per1 is required for light-induced signaling in the suprachiasmatic nucleus circadian clock. J. Biol. Chem, 278, 718–723. [DOI] [PubMed] [Google Scholar]
- Tomkiewicz C, Herry L, Bui LC, Métayer C, Bourdeloux M, Barouki R, & Coumoul X (2013) The aryl hydrocarbon receptor regulates focal adhesion sites through a non-genomic FAK/Src pathway. Oncogene, 32, 1811–1820. [DOI] [PubMed] [Google Scholar]
- Travnickova-Bendova Z, Cermakian N, Reppert SM, & Sassone-Corsi P (2002) Bimodal regulation of mPeriod promoters by CREB-dependent signaling and CLOCK/BMAL1 activity. Proc Natl Acad Sci U A, 99, 7728–7733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai MJ, Hsu YL, Wang TN, Wu LY, Lien CT, Hung CH, Kuo PL, & Huang MS (2014) Aryl hydrocarbon receptor (AhR) agonists increase airway epithelial matrix metalloproteinase activity. J Mol Med Berl, 92, 615–628. [DOI] [PubMed] [Google Scholar]
- Tsang AH, Barclay JL, & Oster H (2014) Interactions between endocrine and circadian systems. J Mol Endocrinol, 52, R1–16. [DOI] [PubMed] [Google Scholar]
- Vetter C (2018) Circadian disruption: What do we actually mean? Eur. J. Neurosci,. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vujovic N, Gooley JJ, Jhou TC, & Saper CB (2015) Projections from the subparaventricular zone define four channels of output from the circadian timing system. J Comp Neurol, 523, 2714–2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, Xu CX, Bu Y, Bottum KM, & Tischkau SA (2014) Beta-naphthoflavone (DB06732) mediates estrogen receptor-positive breast cancer cell cycle arrest through AhR-dependent regulation of PI3K/AKT and MAPK/ERK signaling. Carcinogenesis, 35, 703–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, Zhang ZM, Xu CX, & Tischkau SA (2014) Interplay between Dioxin-mediated signaling and circadian clock: a possible determinant in metabolic homeostasis. Int J Mol Sci, 15, 11700–11712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang F, Samudio I, & Safe S (2001) Transcriptional activation of cathepsin D gene expression by 17beta-estradiol: mechanism of aryl hydrocarbon receptor-mediated inhibition. Mol Cell Endocrinol, 172, 91–103. [DOI] [PubMed] [Google Scholar]
- Wang ZY & Tobin EM (1998) Constitutive expression of the CIRCADIAN CLOCK ASSOCIATED 1 (CCA1) gene disrupts circadian rhythms and suppresses its own expression. Cell, 93, 1207–1217. [DOI] [PubMed] [Google Scholar]
- Weiss C, Faust D, Dürk H, Kolluri SK, Pelzer A, Schneider S, Dietrich C, Oesch F, & Göttlicher M (2005) TCDD induces c-jun expression via a novel Ah (dioxin) receptor-mediated p38-MAPK-dependent pathway. Oncogene, 24, 4975–4983. [DOI] [PubMed] [Google Scholar]
- Weiss C, Faust D, Schreck I, Ruff A, Farwerck T, Melenberg A, Schneider S, Oesch-Bartlomowicz B, Zatloukalová J, Vondrácek J, Oesch F, & Dietrich C (2008) TCDD deregulates contact inhibition in rat liver oval cells via Ah receptor, JunD and cyclin A. Oncogene, 27, 2198–2207. [DOI] [PubMed] [Google Scholar]
- Widerak M, Ghoneim C, Dumontier MF, Quesne M, Corvol MT, & Savouret JF (2006) The aryl hydrocarbon receptor activates the retinoic acid receptoralpha through SMRT antagonism. Biochimie, 88, 387–397. [DOI] [PubMed] [Google Scholar]
- Xing W, Busino L, Hinds TR, Marionni ST, Saifee NH, Bush MF, Pagano M, & Zheng N (2013) SCF(FBXL3) ubiquitin ligase targets cryptochromes at their cofactor pocket. Nature, 496, 64–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu CX, Krager SL, Liao DF, & Tischkau SA (2010) Disruption of CLOCK-BMAL1 transcriptional activity is responsible for aryl hydrocarbon receptor-mediated regulation of Period1 gene. Toxicol Sci, 115, 98–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu CX, Wang C, Krager SL, Bottum KM, & Tischkau SA (2013) Aryl hydrocarbon receptor activation attenuates Per1 gene induction and influences circadian clock resetting. Toxicol Sci, 132, 368–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu CX, Wang C, Zhang ZM, Jaeger CD, Krager SL, Bottum KM, Liu J, Liao DF, & Tischkau SA (2015) Aryl hydrocarbon receptor deficiency protects mice from diet-induced adiposity and metabolic disorders through increased energy expenditure. Int. J. Obes, 39, 1300–1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamaguchi S, Isejima H, Matsuo T, Okura R, Yagita K, Kobayashi M, & Okamura H (2003) Synchronization of cellular clocks in the suprachiasmatic nucleus. Science, 302, 1408–1412. [DOI] [PubMed] [Google Scholar]
- Yoo SH, Mohawk JA, Siepka SM, Shan Y, Huh SK, Hong HK, Kornblum I, Kumar V, Koike N, Xu M, Nussbaum J, Liu X, Chen Z, Chen ZJ, Green CB, & Takahashi JS (2013) Competing E3 ubiquitin ligases govern circadian periodicity by degradation of CRY in nucleus and cytoplasm. Cell, 152, 1091–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoo SH, Yamazaki S, Lowrey PL, Shimomura K, Ko CH, Buhr ED, Siepka SM, Hong HK, Oh WJ, Yoo OJ, Menaker M, & Takahashi JS (2004) PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals persistent circadian oscillations in mouse peripheral tissues. Proc Natl Acad Sci U A, 101, 5339–5346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu H, Du Y, Zhang X, Sun Y, Li S, Dou Y, Li Z, Yuan H, & Zhao W (2014) The aryl hydrocarbon receptor suppresses osteoblast proliferation and differentiation through the activation of the ERK signaling pathway. Toxicol Appl Pharmacol, 280, 502–510. [DOI] [PubMed] [Google Scholar]
- Yu W, Ikeda M, Abe H, Honma S, Ebisawa T, Yamauchi T, Honma K, & Nomura M (1999) Characterization of three splice variants and genomic organization of the mouse BMAL1 gene. Biochem Biophys Res Commun, 260, 760–767. [DOI] [PubMed] [Google Scholar]
- Zeng H, Hardin PE, & Rosbash M (1994) Constitutive overexpression of the Drosophila period protein inhibits period mRNA cycling. EMBO J, 13, 3590–3598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang R, Lahens NF, Ballance HI, Hughes ME, & Hogenesch JB (2014) A circadian gene expression atlas in mammals: implications for biology and medicine. Proc. Natl. Acad. Sci. U. S. A, 111, 16219–16224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W, Moorthy B, Chen M, Muthiah K, Coffee R, Purchio AF, & West DB (2004) A Cyp1a2-luciferase transgenic CD-1 mouse model: responses to aryl hydrocarbons similar to the humanized AhR mice. Toxicol Sci, 82, 297–307. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Fang B, Emmett MJ, Damle M, Sun Z, Feng D, Armour SM, Remsberg JR, Jager J, Soccio RE, Steger DJ, & Lazar MA (2015) GENE REGULATION. Discrete functions of nuclear receptor Rev-erbα couple metabolism to the clock. Science, 348, 1488–1492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang YK, Yeager RL, & Klaassen CD (2009) Circadian expression profiles of drug-processing genes and transcription factors in mouse liver. Drug Metab Dispos, 37, 106–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
