Next Article in Journal
Mechanotransduction Circuits in Human Pathobiology
Next Article in Special Issue
Bisphenol-A in Drinking Water Accelerates Mammary Cancerogenesis and Favors an Immunosuppressive Tumor Microenvironment in BALB–neuT Mice
Previous Article in Journal
Comparative Genomic Analysis of PEBP Genes in Cucurbits Explores the Interactors of Cucumber CsPEBPs Related to Flowering Time
Previous Article in Special Issue
Postnatal Exposure to the Endocrine Disruptor Dichlorodiphenyltrichloroethane Affects Adrenomedullary Chromaffin Cell Physiology and Alters the Balance of Mechanisms Underlying Cell Renewal
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Endocrine Disruption Chemical-Regulated Aryl Hydrocarbon Receptor Activity in the Pathogenesis of Pancreatic Diseases and Cancer

by
Kyounghyun Kim
Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas Medical Sciences, Little Rock, AR 72225, USA
Int. J. Mol. Sci. 2024, 25(7), 3818; https://doi.org/10.3390/ijms25073818
Submission received: 20 February 2024 / Revised: 26 March 2024 / Accepted: 27 March 2024 / Published: 29 March 2024
(This article belongs to the Special Issue Endocrine Disruption and Human Diseases 2.0)

Abstract

:
The aryl hydrocarbon receptor (AHR) serves as a ligand-activated transcription factor crucial for regulating fundamental cellular and molecular processes, such as xenobiotic metabolism, immune responses, and cancer development. Notably, a spectrum of endocrine-disrupting chemicals (EDCs) act as agonists or antagonists of AHR, leading to the dysregulation of pivotal cellular and molecular processes and endocrine system disruption. Accumulating evidence suggests a correlation between EDC exposure and the onset of diverse pancreatic diseases, including diabetes, pancreatitis, and pancreatic cancer. Despite this association, the mechanistic role of AHR as a linchpin molecule in EDC exposure-related pathogenesis of pancreatic diseases and cancer remains unexplored. This review comprehensively examines the involvement of AHR in EDC exposure-mediated regulation of pancreatic pathogenesis, emphasizing AHR as a potential therapeutic target for the pathogenesis of pancreatic diseases and cancer.

1. Introduction

EDCs represent exogenous substances or mixtures that disrupt endocrine system function, leading to adverse effects in organisms, their progeny, or specific populations. These substances dysregulate the endocrine system, influencing hormone production, storage, and secretion, contributing to various detrimental effects on human health. EDCs can mimic, block, or interfere with the body’s hormone functions. Consequently, exposure to EDCs is associated with disruptions in sperm count, fertility, reproductive organs, endometriosis, puberty, cardiovascular function, immune response, nervous system activity, respiratory function, and metabolism. Ultimately, these disruptions may contribute to the development of various human diseases and cancers [1,2,3].
EDCs comprise a broad range of exogenous substances, including bisphenols [4], phthalates [5], organotin [6], pesticides [7], polychlorinated dibenzo-p-dioxins (PCBs) [8], dioxin-like compounds [9], polyaromatic hydrocarbons (PAHs) [10], flame retardants [11], and alkylphenols [12]. Exposure to EDCs is widespread in daily life through various products such as cosmetics, food and beverage packaging, toys, and carpets [13,14,15]. Naturally occurring EDCs, like phytoestrogens such as genistein and daidzein, contribute to this diverse array [16]. Certain heavy metals, including arsenic, chromium, or cadmium, can also function as endocrine disruptors [17,18,19]. Exposure routes to EDCs are diverse, involving ingestion, water consumption, inhalation, and dermal contact. Notably, many EDCs can act as agonists or antagonists of the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor crucial for environmental sensing and xenobiotic metabolism [20,21,22,23,24].

2. Aryl Hydrocarbon Receptor (AHR)

The aryl hydrocarbon receptor (AHR) was initially identified as a cytoplasmic receptor with a high affinity for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) [25]. The harmful health effects resulting from accidental TCDD exposure were first reported in Nitro, West Virginia, in 1949 [26]. TCDD gained notoriety as a contaminant in Agent Orange, an herbicide and defoliant mixture widely used during the Vietnam War, composed of N-butyl esters of 2,4-dichlorophenoxyacetic (2,4-D) and 2,4,5-trichlorophenoxyacetic (2,4,5-T) acids. Veterans with high exposure to Agent Orange have exhibited an increased incidence of cancer and congenital disabilities in their children [27]. TCDD, the most potent and extensively studied dioxin, serves as a prototype for dioxins that function as AHR agonists [28].
Basically, AHR functions as a ligand-activated transcription factor integral to cellular homeostasis, governing various physiological and pathological processes, including xenobiotic detoxification, metabolism, cardiovascular regulation, immunomodulation, and cancer development [29,30,31]. In its unliganded state, AHR forms an inactive complex, engaging with two heat shock protein 90 (HSP90) [32,33], AHR interacting protein (AIP) [34,35], and prostaglandin E synthase 3 (PTGES3 or p23) [36]. This interaction serves to maintain AHR stability, conformation, and cytoplasmic localization. AIP, in particular, safeguards AHR from ubiquitylation-induced degradation while contributing to AHR folding and stability through direct interactions with HSP90 and AHR [37].
Upon ligand binding, the aryl hydrocarbon receptor (AHR) undergoes nuclear translocation, exposing its nuclear localization signal (NLS). Within the nucleus, AHR forms a complex with the AHR nuclear translocator (ARNT), also known as Hypoxia Inducible Factor 1 Beta (HIF1β). This AHR/ARNT heterodimer binds specifically to DNA sequences termed Xenobiotic Response Elements (XREs), with a consensus sequence of 5′-TNGCGTG-3′. These XREs are situated in the promoter regions of downstream target genes, initiating the activation of gene expression (Figure 1). The interactions between AHR-ARNT-XRE lead to the induction of downstream target genes, including phase I detoxification enzymes such as CYP1A1 and CYP1B1, as well as phase II detoxification enzymes like UGT1A1 and UGT1A6 [36,37,38,39]. Following the activation of AHR signaling, the aryl hydrocarbon receptor repressor (AHRR) protein is induced, exerting inhibitory effects on AHR signaling activation. AHRR competes with ARNT to bind to AHR, constituting a negative feedback mechanism [40]. The resultant gene expressions have diverse physiological, pathological, and toxicological implications in various human diseases and cancers. This canonical AHR pathway activation, mediated through binding the AHR/ARNT complex to XRE, is recognized as the primary mechanism of AHR signaling.
In contrast to the canonical AHR pathway, which relies on AHR/ARNT/XRE interactions, activating the non-canonical AHR pathway occurs through interactions with other transcription factors. These factors include Kruppel-like factor 6 (KLF6), Estrogen Receptor α (ERα), and a member of the NF-κB family such as RelB. Independent of AHR/ARNT complex formation, AHR forms a heterodimer with KLF6 and binds to non-consensus XRE, inducing gene expression [41]. Ligand-activated AHR inhibits gene expression responses to the estrogen/ERα complex [42]. The interaction between ligand-bound AHR and RelB regulates IL8 expression, which is crucial in developing chronic inflammatory diseases [43] (Figure 1). Additionally, the non-canonical AHR pathway involves PKA (cAMP-dependent protein kinase)-mediated AHR activation in a ligand-independent manner [44]. The role of the non-canonical AHR pathway in various human diseases, including pancreatic diseases and cancer, remains largely unexplored.

3. AHR Structure and Its Interactions with Various Ligands

AHR belongs to the basic helix-loop-helix/per-ARNT-sim (bHLH/PAS) superfamily, characterized by three functional structural domains: a bHLH domain responsible for DNA binding, two PAS structural domains (A and B) facilitating dimerization with ARNT and ligand binding, and a transactivating domain for gene expression (Figure 2A). PAS domains function as ubiquitous and versatile sensor and interaction modules within signal transduction proteins. These PAS sensors can detect a diverse range of chemical and physical stimuli, consequently regulating the activity of various functionally diverse effector domains. Despite the extensive chemical, physical, and functional diversity associated with PAS sensors, the core structures of PAS domains remain broadly conserved [45]. AHR belongs to the distinctive bHLH/PAS protein family and is uniquely activated by small molecules, including various EDCs.
Protein–protein interactions occur within the PAS-A domain, which lacks a ligand-binding cavity. In contrast, the PAS-B domain forms a ligand-binding pocket (LBP) capable of accommodating diverse ligands. A distinctive feature of AHR is its versatile binding capacity to various ligands, including EDCs, phytochemicals, and endogenous metabolites [46,47,48]. A recent cryo-EM structural analysis of the indirubin-bound AHR complex unveiled the structural determinants of the PAS-B domain in promiscuous ligand binding. Notably, all secondary structures of the PAS-B domain, including a five-stranded antiparallel β-sheet (Aβ, Bβ, Gβ, Hβ, and Iβ) flanked by four consecutive α-helices (Cα, Dα, Eα, and Fα), contribute to the ligand binding pocket (LBP). This elongated channel—perpendicular to two helical structures—partially occupies the LBP, leaving a significant portion void, suggesting its capability to accommodate various small molecules of different sizes [49] (Figure 2B). A comparative study investigating the ligand-binding pockets (LBPs) of drosophila AHR (dAHR) and mouse AHR (mAHR) revealed that the larger size and structural variation of the mAHR PAS-B domain, which forms the LBP, contribute to its extensive ligand adaptability [50,51,52]. In contrast, dAHR possesses a smaller LBP and demonstrates constitutive activation in the absence of ligand binding, suggesting a species-dependent disparity in AHR actions [53].
Another notable aspect of AHR signaling involves the planar structure of AHR ligands. For instance, indirubin, characterized by its planar molecular structure and asymmetric double indole structure, intercalates between two layers of amino acid residues in the ligand-binding pocket (LBP). This characteristic underscores the crucial role of planarity in AHR ligands for selective ligand–AHR interactions [54,55,56,57]. Moreover, an interspecies difference exists in the binding affinity of AHR for the same ligand, despite a high level of sequence homology between mouse Ahr and human AHR (approximately 85%). For example, mouse AHR exhibits a ten-fold higher affinity for TCDD than human AHR [54,55,56]. In comparison, human AHR shows a much higher affinity for indirubin than mouse AHR, highlighting that variations in the ligand-binding pocket finely tune the specificity of ligand–AHR interactions despite its inherent promiscuity [57]. However, the ligand-dependent interactions between the PAS-A and PAS-B domains, which are likely responsible for recruiting different transcription factors and coregulators for activating a specific battery of genes, depending on ligand type (agonist vs. antagonist), remain to be further investigated.

4. EDCs from Environmental Pollutants and AHR

4.1. Dioxins and Dioxin-like Compounds

These compounds, identified and defined by the Stockholm Convention in 2001 as persistent organic pollutants (POPs), encompass polychlorinated dibenzofurans (PCDFs), polychlorinated dibenzo-p-dioxins (PCDDs), dibenzofurans (PCDFs), and polychlorinated biphenyls (PCBs). Recognizing their adverse effects on human health and the environment, the convention aimed to restrict their production. POPs, resistant to degradation through chemical, biological, and photolytic processes, are carbon-based substances. Their high stability and lipophilic nature lead to accumulation in the fatty tissues of humans and animals, causing adverse health effects. Beyond dioxin-like compounds, POPs encompass organochlorine pesticides (DDT, chlordane, dieldrin, heptachlor, hexachlorobenzene, mirex, and toxaphene) [58,59]. Accumulating evidence suggests that many POPs, including dioxins and dioxin-like compounds, exert biologically harmful effects by activating AHR function [60,61,62]. Dioxins and dioxin-like compounds are inadvertent byproducts resulting from high-temperature processes, including incomplete combustion of waste, coal, and wood, as well as automobile emissions. Industrial activities such as manufacturing chemicals, smelting, chlorine bleaching of paper pulp, and herbicide or pesticide production also contribute to their formation. The carbon skeleton of dioxins is represented by dibenzodioxin or dibenzo-p-dioxin. These compounds encompass polychlorinated dibenzo-p-dioxin (PCDD), polychlorinated dibenzofuran (PCDF) congeners, coplanar polychlorinated biphenyls (PCBs), and polybrominated biphenyl (PBB), which are bromine analogs of PCBs [63,64]. Natural disasters, including volcanic eruptions or forest fires, can also generate these toxic compounds [65]. Cigarette smoke contains elevated levels of dioxins or dioxin-like compounds.
TCDD, a prototypical dioxin and a most potent AHR agonist, exhibits a long half-life of 8 years in humans [66,67]. Short-term high exposure to dioxin can result in skin lesions, such as chloracne, and abnormal liver function [68,69,70]. Epidemiological studies have demonstrated that chronic dioxin exposure leads to impairments in the immune, nervous, cardiovascular, and reproductive systems and is associated with various types of cancers [61,71]. The International Agency for Research on Cancer (IARC) classifies dioxin as a Group 1 carcinogen [72]. The next section will describe the effects of the dioxin and dioxin-like compound-regulated AHR signaling axis on the pathogenesis of pancreatic diseases and cancer.

4.2. Polycyclic Aromatic Hydrocarbons (PAHs)

PAHs are organic compounds characterized by multiple aromatic rings. Exposure to PAHs can occur through various routes, including smoking, consumption of food and beverages, and inhalation of air. Major PAHs include benz[a]anthracene, chrysene, benzo[b]fluoranthene, and benzo[a]pyrene (BaP), with BaP being extensively studied for its carcinogenic and genotoxic properties [73,74]. Unlike persistent organic pollutants (POPs), PAHs have relatively short half-lives, ranging from 2.5 to 6.1 h [75,76]. The liver predominantly metabolizes PAHs through CYP enzymes, and the resulting metabolites are excreted in feces and urine. PAHs constitute a significant class of organic chemicals in particulate matter (PM) [77,78]. Multiple studies have robustly established an association between exposure to particulate matter (PM) and adverse human health effects, primarily attributed to the carcinogenic and mutagenic properties of polycyclic aromatic hydrocarbons (PAHs) [79,80]. Furthermore, many reports have underscored the correlation of PAH exposure with pancreatic diseases and cancer [81,82,83]. Nevertheless, the underlying mechanisms by which PAH exposure mediates the development of pancreatic diseases and cancer through the aryl hydrocarbon receptor (AHR) signaling pathway remain unexplored.

4.3. Hexachlorobenzene (HCB)

HCB is a chlorinated hydrocarbon that was historically employed as a fungicide or pesticide. Owing to its highly lipophilic nature, HCB is a pervasive pollutant that accumulates in biological systems and the environment [84]. Oral absorption represents a major route of HCB exposure. The accidental poisoning through HCB in Turkey from 1955–1959 highlighted the severe health consequences, with 4000 individuals exhibiting porphyria and skin lesions, and later developing arthritis [85]. In cell culture, HCB exposure has been shown to enhance cancer cell proliferation, migration, and invasion by activating AHR as a weak agonist [86]. Unlike polycyclic aromatic hydrocarbons (PAHs), HCB is classified as a weak AHR agonist and nongenotoxic carcinogen. Despite the fact that HCB exposure is a risk factor for the development of pancreatic diseases and cancer [87,88,89], the role of the HCB-AHR signaling axis remains uninvestigated.

4.4. Bisphenol A (BPA)

BPA is a chemical that produces various polycarbonate plastics, including food containers, baby bottles, water bottles, medical devices, and hygiene products. It has been a widely used endocrine-disrupting chemical (EDC) since the early 1950s. It is a major constituent of polycarbonate plastics used in manufacturing epoxy resins, dental sealants, and recycled paper, and is used in the lining of food cans [90,91]. BPA is detectable in urine, blood, breast milk, and other tissues, with the primary human exposure route being ingestion. Upon ingestion, BPA is rapidly absorbed and metabolized in the liver, becoming hydrophilic and subsequently excreted primarily in urine, with a known half-life of less than six hours [92,93]. Despite being a non-persistent EDC with a short half-life, over 90% of individuals exhibit detectable urine BPA levels. Many studies have reported a positive correlation between urine BPA levels and diabetes in adults and children [94,95]. Many studies have also reported that BPA exposure disrupts pancreatic β-cell function by targeting estrogen receptors α or β [96,97,98,99,100]. Additionally, BPA activates aryl hydrocarbon receptor (AHR) signaling and inhibits mouse ovarian follicle growth [101]. Exposure to BPA during mouse embryo development increases the expression of AHR and its downstream target genes [102]. Additionally, low-dose BPA exposure activates AHR in breast cancer cells, increasing their aggressive cancer cell phenotype [103]. However, the effects of BPA on AHR signaling involving development of pancreatic diseases and cancer remain to be further investigated. A recent report demonstrated that exposure to bisphenol A (BPA) in a mouse model activated the aryl hydrocarbon receptor (AHR) in pancreatic islets, indicating a potentially harmful role of BPA-AHR signaling activation in the pancreas [104]. Nevertheless, further mechanistic studies investigating the role of BPA-regulated AHR pathways in the pathogenesis of pancreatic diseases and cancer are warranted.

4.5. Heavy Metals

Exposure to heavy metals, such as arsenic, cadmium, or chromium, can result in cellular injury, genetic alterations, or a combination of both [105,106]. Arsenic, a prevalent toxic metal in the environment, induces the generation of reactive oxygen species (ROS) upon exposure, disrupting antioxidant defense mechanisms and impacting mitochondrial morphology and integrity. Chromium, particularly in its hexavalent form [Cr(VI)], is highly toxic, mutagenic, and carcinogenic, producing hydroxy radicals and superoxide, contributing to adverse effects. Cadmium, identified as a human carcinogen, also induces oxidative stress. The toxicity and carcinogenicity associated with heavy metals often involve the production of ROS [107,108]. ROS generated by heavy metal exposure likely indirectly activate AHR signaling. Exposure to arsenic or cadmium alone increases AHR activity and the expression of downstream target genes, such as CYP1A1 [109,110], and is possibly linked to the generation of oxidative stress that increases the production of biliverdin or bilirubin, an AHR ligand [111]. Similarly, oxidative stress induced by chromium (VI) exposure activates AHR signaling by increasing the production of oxindole, an AHR ligand. Cadmium exposure also elevates AHR signaling and downstream gene expression [112]. Moreover, heavy metal exposure and AHR ligand treatment further enhance AHR signaling activation [113,114]. These reports emphasize the pivotal role of oxidative stress in heavy metal exposure-regulated aryl hydrocarbon receptor (AHR) actions. Many studies have identified heavy metal exposure as a significant risk factor for pancreatic cancer [115,116,117]. However, the underlying mechanisms connecting heavy metal exposure-mediated AHR signaling regulation to the development of pancreatic diseases and cancer remain unknown.
Collectively, we summarize the interplay between endocrine-disrupting chemicals (EDCs) and aryl hydrocarbon receptor (AHR) signaling, as well as epidemiological studies and relevant findings on the roles of these EDC-mediated AHR signaling regulations (Table 1).

5. Roles of EDC–AHR Interactions in the Pathogenesis of Pancreatic Diseases and Cancer

In its multifunctional role, the pancreas plays a crucial part in the endocrine and exocrine systems. Regarding endocrine function, the islets of Langerhans within the pancreas consist of five distinct cell types—alpha, beta, delta, epsilon, and upsilon—each responsible for secreting specific hormones. These hormones include glucagon, insulin, somatostatin, ghrelin, and pancreatic polypeptide. On the other hand, exocrine function involves acinar cells releasing digestive enzymes such as amylase, lipase, and proteases. These enzymes are channeled into the pancreatic duct, which merges with the common bile duct, and their combined secretions enter the duodenum, aiding in the breakdown of carbohydrates, fats, and proteins from ingested food.
Exposure to EDC can have adverse effects on the pancreas, contributing to conditions such as obesity, diabetes, insulin resistance, hyperinsulinemia, and pancreatitis. Importantly, these conditions also serve as known risk factors for the development of pancreatic cancer [118,119]. Understanding the impact of EDC exposure on the intricate functions of the pancreas is essential for comprehending the potential health risks associated with these environmental contaminants.

5.1. Role of EDC-Regulated AHR in Diabetes Mellitus

Diabetes mellitus (DM) is a chronic metabolic disorder marked by elevated blood glucose levels due to compromised insulin secretion and disrupted glucose homeostasis. According to the IDF Diabetes Atlas, 537 million adults aged 20–79 had diabetes globally in 2021. Projections indicate an increase to 643 million by 2030 and 783 million by 2045 [120]. Two primary types of DM exist: type 1 DM (T1DM), characterized by total insulin absence, frequently found in children; and type 2 DM (T2DM), primarily found in adults, resulting from diminished insulin secretion and functionality.

5.1.1. Type 1 Diabetes Mellitus (T1DM)

T1DM is an autoimmune disorder characterized by the destruction of pancreatic β cells by self-reactive T cells, resulting in insulin deficiency. The incidence of T1DM has markedly increased over past decades, and this surge cannot be exclusively attributed to genetic factors. A potential contributing factor is the heightened exposure to endocrine-disrupting chemicals (EDCs) during prenatal and early developmental stages. This exposure may disrupt immune homeostasis, which regulates the maintenance and survival of pancreatic β cells. Numerous epidemiological studies suggest that exposure to environmental contaminants, such as dioxins, PCBs, bisphenol A, and air pollutants containing PAHs, increases the risk of type 1 diabetes mellitus (T1DM) [121,122].
AHR is expressed in various immune cell types, including dendritic and T cells, with T cells playing a pivotal role in destroying pancreatic β cells [123]. In the non-obese diabetic (NOD) mouse model of type 1 diabetes mellitus (T1DM) development, TCDD activated AHR, increasing Foxp3+ T cells that exert anti-inflammatory effects against effector T cells, preventing T1DM development [124]. In the same mouse model, AHR activation by the exogenous ligand 10-CI-BBQ inhibited T1DM development [125]. These findings suggest that AHR signaling activation, depending on the immune cell type, is immunosuppressive and can modulate immune responses during T1DM development, highlighting the potential of AHR targeting in T1DM. In contrast to the role of AHR in T1DM, a recent study reported that high levels of urinary metabolites of polycyclic aromatic hydrocarbons (PAHs) are associated with an increased risk of T1DM in children and adolescents [126]. In animal studies, prenatal exposure to a mixture of PAHs resulted in dysfunctional pancreatic islets associated with T1DM [127]. PAHs may increase the risk of T1DM development through other unknown mechanisms.
High urinary levels of bisphenol A (BPA) in children and adolescents are associated with increased T1DM development [128]. In animal studies, transmaternal exposure to BPA significantly increases insulitis severity and diabetes incidence in female offspring in a dose-dependent manner [129]. The progeny of BPA-exposed mothers exhibit heightened apoptosis of both pancreatic α and β cells, promoting the development of T1DM in NOD mice [130]. In another streptozotocin (STZ)-induced T1DM murine model, low-dose multiple oral BPA exposures facilitated diabetes induction, potentially through BPA-mediated immunomodulation of T cells or reduced cytokine levels, suggesting that BPA acts as a risk factor for diabetes by altering immune modulatory activity [131]. A recent report showed that BPA-activated AHR in pancreatic islets disrupted glucose homeostasis and altered insulin sensitivity, implying that BPA-mediated AHR signaling activation plays a role in T1D development [104]. Epidemiological evidence and molecular studies linking BPA exposure, AHR, and T1DM development need further investigation.
Elevated levels of arsenic and fluoride in drinking water have been associated with an increased incidence of type 1 diabetes mellitus (T1DM) [132]. The high plasma level of arsenic is associated with an increased risk of T1DM development [133]. Low-level sub-chronic arsenic exposure from the prenatal stage has been shown to impair glucose metabolism in adult life [134]. It has been previously reported that arsenic and other heavy metals regulate AHR signaling [109,110,111,112,113,114]. However, the effects of arsenic exposure on AHR activity in the pancreas associated with T1DM development remain unclear. Further human studies and longitudinal evidence are needed to explore this relationship.

5.1.2. Type 2 Diabetes Mellitus (T2DM)

Many epidemiological studies have consistently indicated a connection between dioxin exposure and the development of type 2 diabetes mellitus (T2DM). American war veterans who engaged in defoliant spraying containing TCDD during the Vietnam War showed a correlation between their TCDD exposure levels and the incidence of T2DM [135]. A significant dose–response relationship was observed between the serum concentration of six selected persistent organic pollutants (POPs)—including dioxin and dioxin-like compounds—and the prevalence of diabetes [136]. Furthermore, prolonged exposure to persistent organic pollutants (POPs), including polychlorinated biphenyls (PCBs) and organochlorine pesticides, was associated with lower serum insulin levels. Correspondingly, in vitro studies consistently demonstrated impaired insulin secretion in pancreatic β-cells following low-level POP exposure [137].
Mounting evidence suggests that exposure to TCDD targets pancreatic β-cells, disrupting insulin secretion regulation. Isolated pancreatic islet cells from rats exposed to low chronic TCDD exhibited impaired glucose-stimulated insulin secretion [138]. Similarly, low TCDD exposure to the insulin-secreting pancreatic β-cell line INS-1E significantly impaired insulin secretion, accompanied by increased pancreatic β-cell death, implicating the sensitivity of pancreatic β-cells to dioxin exposure [139]. TCDD exposure to human embryonic stem cells compromised pancreatic lineage differentiation and altered DNA methylation and gene expression, highlighting that early embryonic TCDD exposure dysregulates pancreatic function and increases the risk of type 2 diabetes mellitus (T2DM) [140]. Furthermore, several animal studies revealed that TCDD-activated AHR reduced insulin secretion. Exposure to TCDD suppressed insulin secretion and increased pancreatic β cell death [141]. Intriguingly, in CYP1A1 and CYP1A2 knockout islets, the toxic effects of TCDD or 3-MC were decreased, emphasizing the crucial role of CYP1 enzyme activities in pancreatic beta cell survival and death [142]. In an experimental model of T2DM induced by streptozotocin (STZ), significantly elevated CYP1A1 activity levels were observed in diabetic rats, supporting the role of the AHR-CYP1 axis in pancreatic β-cell pathophysiology [143]. Moreover, the crucial role of AHR in TCDD-mediated toxic effects on insulin secretion and glucose homeostasis was emphasized through the use of AHR knockout mice, which demonstrated imbalanced glucose homeostasis and reduced insulin levels [144]. A single instance of acute dioxin exposure to mice suppressed insulin secretion for up to 6 weeks, suggesting that the toxic effects of acute dioxin exposure on pancreatic β-cells can have long-term consequences, even if the exposure is transient [145]. Low levels of POP exposure, including organochlorine pesticides and PCBs, decreased insulin secretion by disrupting pancreatic β cell function [146].
Exposure to BPA, PAHs, hexachlorobenzene, or heavy metals has been acknowledged as a risk factor for the development of T2DM [147,148,149,150,151]. As detailed in the preceding sections, these EDCs act as positive or negative AHR signaling regulators. Nevertheless, the underlying mechanisms through which EDC exposure modulates AHR signaling in the context of T2DM development remain to be further investigated.

5.1.3. Role of EDC-Regulated AHR in Pancreatitis

Pancreatitis is an inflammatory disease of the pancreas, accompanied by the gradual replacement of the pancreas by fibrotic tissue compartments. Heavy alcohol consumption and cigarette smoke are major risk factors of pancreatitis [152]. Notably, cigarette smoke contains high levels of dioxins—dioxin-like compounds that activate the AHR signaling pathway [153,154]. Pancreatitis is a key risk factor for pancreatic cancer development [155,156].
Acute pancreatitis, characterized by inflammatory damage to the pancreatic acini, leads to extensive necrosis and multi-organ failure, contributing to severe cases’ mortality. The aryl hydrocarbon receptor (AHR) emerges as a critical transcription factor pivotal for the production of IL22, a protective cytokine in acute pancreatitis. In a murine model induced by caerulein, AHR inactivation using the antagonist CH223191 decreased IL22 production. At the same time, AHR activation by biliverdin, an AHR agonist, increased pancreatic IL22 levels and provided protection against acute pancreatitis [157]. Conversely, acute exposure to benzo(a)pyrene or TCDD induces pancreatitis accompanied by oxidative stress-related mitochondrial respiratory dysfunction. Resveratrol, an AHR antagonist, prevents the harmful effects of pancreatitis and mitigates mitochondrial damage [158]. TCDD exposure also upregulates long noncoding RNA MALAT1 expression in pancreatic cancer cells and tissue. MALAT1 interacts with the histone methyltransferase EZH2, enhancing its enzymatic activity. Consequently, AHR-mediated MALAT1 induction amplifies EZH2’s histone methyltransferase activity, revealing a novel pathway through which TCDD exposure alters epigenetic status via activation of the AHR-MALAT1-EZH2 signaling axis [159]. In experimental autoimmune pancreatitis, AHR activation augments IL22 production in pancreatic α cells, suppressing chronic fibrotic and inflammatory processes via IL22 production [160].
Chronic pancreatitis is a progressive inflammatory condition characterized by increased fibrosis, serving as a predisposing factor for pancreatic cancer. Cigarette smoke—a significant risk factor for chronic pancreatitis—contains elevated levels of dioxin-like compounds that activate aryl hydrocarbon receptor (AHR) signaling. In a murine model of chronic pancreatitis, exposure to cigarette smoke resulted in increased IL22 production in T cells, promoting pancreatic fibrosis and contributing to the development of chronic pancreatitis. Consistently, cigarette smokers in this model exhibited higher serum levels of IL22 than non-smokers [161]. These reports suggest that the context-dependent effects of AHR activation, influenced by the disease model or AHR ligand type, add complexity to the nature of AHR signaling.

5.1.4. Role of EDC-Regulated AHR in Pancreatic Cancer

Pancreatic cancer stands as the seventh leading cause of global cancer-related mortality, characterized by its formidable nature, with symptoms often emerging only at an advanced stage, resulting in elevated mortality rates. In 2018, approximately 450,000 new cases of pancreatic cancer were reported worldwide, leading to 432,242 deaths [162]. In the United States, pancreatic cancer accounts for 3% of overall cancer incidence and contributes to 7% of cancer-related deaths. Projections for 2023 estimate 64,050 new cases with 50,550 fatalities. The five-year survival rate for pancreatic cancer is a mere 12%, emphasizing the challenge posed by late-stage diagnoses, with only 12% identified at an early, surgically removable stage. Over 50% of individuals receive diagnoses at later stages, characterized by distal metastasis. Current therapeutic modalities lack efficacy for advanced stages, and options for efficacious early diagnosis remain limited [163,164]. Pancreatic ductal epithelial cells, among the different pancreatic cell types, are the origin of pancreatic adenocarcinomas, a major type of pancreatic cancer.
Epidemiological studies have indicated a moderate increase in the risk of pancreatic cancer associated with high serum concentrations of persistent organic pollutants (POPs) [165,166]. However, further research is needed to substantiate these findings. Notably, cigarette smoking stands out as a significant risk factor for pancreatic cancer, with high levels of dioxin-like compounds present in cigarette smoke [153]. Cigarette smoke has been shown to induce aryl hydrocarbon receptor (AHR) activation [154]. These observations highlight a potential molecular link between dioxin exposure, AHR activation, and the development of pancreatic cancer, prompting further investigation.
Using rats as a model, it was found that chronic exposure of dioxin or dioxin-like compounds in the pancreas increased cytoplasmic vacuolation, inflammation, and atrophy in the exocrine pancreas, accompanied with low incidence of pancreatic acini adenoma and carcinoma, indicating that pancreatic acini is a target tissue of dioxin and dioxin-like compounds [167]. AHR displays heightened expression in the cytoplasm of pancreatic cancer tissues, and its activation by AHR agonists such as DIM (diindolymethane) inhibits the growth of pancreatic cancer cells [168]. Similarly, omeprazole, functioning as an AHR agonist, hampers the migration and invasion of pancreatic cancer cells [169]. Carbidopa, an FDA-approved drug for Parkinson’s disease, acts as an AHR agonist, impeding the growth of pancreatic cancer cells by inhibiting IDO1 (indoleamine 2,3-dioxygenase-1) [170]. Depletion of AHR using small interfering RNAs targeting AHR in pancreatic cancer cells heightens sensitivity to gemcitabine, a chemotherapeutic agent, and diminishes cells’ invasive and migratory potential [171]. However, the effects of ligand-dependent AHR activation or inhibition on pancreatic cancer cell proliferation, invasion, and migration have not been investigated. Oct4, a master transcription factor of pluripotency that mediates cancer stem cell features, is suppressed by the tryptophan-derived AHR ligand ITE. ITE interacts with AHR and suppresses Oct4 expression, directing cancer stemness and growth [172]. These reports introduced additional layers of complexity in AHR activities; the type of ligand appears to dictate the regulation of distinct gene subsets, potentially encompassing both pro-oncogenic and tumor-suppressive genes.
AHR also functions as a sensor of microbiome-derived metabolites, modulating immune function within the tumor microenvironment. Notably, tumor-associated macrophages (TAMs) express high levels of AHR. The depletion of AHR in myeloid cells or the inhibition of AHR by antagonistic treatment reduces the progression of pancreatic ductal adenocarcinoma (PDAC) by diminishing the immunosuppressive function of TAMs and enhancing immune surveillance by CD8+ T cells. Correspondingly, high AHR expression in PDAC patients correlates with poor clinical outcomes and features of immunosuppressive TAMs, supporting the tumor-promoting role of AHR in PDAC [173]. These findings underscore the immunomodulatory functions of AHR in TAMs while emphasizing the necessity for further investigations into the interactions between immune cell types and ductal epithelial cells during the development of pancreatic cancer. The role of AHR as a molecular interface between EDC exposure and the development of various pancreatic diseases, including cancer, is summarized below (Figure 3).
In summary, numerous epidemiological studies substantiate a positive correlation between exposure to various EDCs and the incidence of diverse pancreatic diseases, including cancer. Mounting evidence suggests that many EDCs activate or interfere with aryl hydrocarbon receptor (AHR) signaling. However, there is a gap in our understanding regarding AHR’s molecular and mechanistic roles in the EDC-mediated development of various pancreatic diseases and cancer. Moreover, the intricate nature of AHR signaling, manifesting as activation or inhibition depending on ligand type, tissue/cell context, or disease model, poses a major obstacle in investigating the Janus-like role of AHR in promoting or inhibiting pancreatic pathogenesis. Despite these challenges, the ligand-dependent activation or inhibition of AHR’s actions by selective AHR modulators (SAhRMS) underscore its potential as a promising molecular target [174].

Funding

This work was funded primarily by the Winthrop P Rockefeller Cancer Institute, the Office of the Vice Chancellor for Research, and the Arkansas Bioscience Institute (ABI) at the University of Arkansas for Medical Sciences.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AHRaryl hydrocarbon receptor
AHRR aryl hydrocarbon receptor repressor
bHLH helix-loop-helix
BPAbisphenol A
DIMdiindolymethane
EDCendocrine-disrupting chemical
HCBhexachlorobenzene
LBPligand-binding pocket
PAHspolycyclic aromatic hydrocarbons
PCBs polychlorinated biphenyls
PCDF polychlorinated dibenzofurans
PM particulate matter
PDACpancreatic ductal adenocarcinoma
POPs persistent organic pollutants
T1DMtype I diabetes mellitus
T2DMtype II diabetes mellitus
TAMtumor-associated macrophages
TCDD2,3,7,8-tetrachlorodibenzo-p-dioxin
2,4-D2,4-Dichlorophenoxyacetic acid

References

  1. Kahn, L.G.; Philippat, C.; Nakayama, S.F.; Slama, R.; Trasande, L. Endocrine-disrupting chemicals: Implications for human health. Lancet Diabetes Endocrinol. 2020, 8, 703–718. [Google Scholar] [CrossRef] [PubMed]
  2. Schug, T.T.; Janesick, A.; Blumberg, B.; Heindel, J.J. Endocrine disrupting chemicals and disease susceptibility. J. Steroid Biochem. Mol. Biol. 2011, 127, 204–215. [Google Scholar] [CrossRef] [PubMed]
  3. Diamanti-Kandarakis, E.; Bourguignon, J.P.; Giudice, L.C.; Hauser, R.; Prins, G.S.; Soto, A.M.; Zoeller, R.T.; Gore, A.C. Endocrine-disrupting chemicals: An Endocrine Society scientific statement. Endocr. Rev. 2009, 30, 293–342. [Google Scholar] [CrossRef] [PubMed]
  4. Kim, J.J.; Kumar, S.; Kumar, V.; Lee, Y.M.; Kim, Y.S.; Kumar, V. Bisphenols as a Legacy Pollutant, and Their Effects on Organ Vulnerability. Int. J. Environ. Res. Public Health 2019, 17, 112. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, Y.; Qian, H. Phthalates and Their Impacts on Human Health. Healthcare 2021, 9, 603. [Google Scholar] [CrossRef]
  6. Kimbrough, R.D. Toxicity and health effects of selected organotin compounds: A review. Environ. Health Perspect. 1976, 14, 51–56. [Google Scholar] [CrossRef]
  7. Mnif, W.; Hassine, A.I.; Bouaziz, A.; Bartegi, A.; Thomas, O.; Roig, B. Effect of endocrine disruptor pesticides: A review. Int. J. Environ. Res. Public Health 2011, 8, 2265–2303. [Google Scholar] [CrossRef]
  8. Mukerjee, D. Health impact of polychlorinated dibenzo-p-dioxins: A critical review. J. Air Waste Manag. Assoc. 1998, 48, 157–165. [Google Scholar] [CrossRef]
  9. White, S.S.; Birnbaum, L.S. An overview of the effects of dioxins and dioxin-like compounds on vertebrates, as documented in human and ecological epidemiology. J. Environ. Sci. Health C Environ. Carcinog. Ecotoxicol. Rev. 2009, 27, 197–211. [Google Scholar] [CrossRef]
  10. Patel, A.B.; Shaikh, S.; Jain, K.R.; Desai, C.; Madamwar, D. Polycyclic Aromatic Hydrocarbons: Sources, Toxicity, and Remediation Approaches. Front. Microbiol. 2020, 11, 562813. [Google Scholar] [CrossRef]
  11. Dishaw, L.V.; Macaulay, L.J.; Roberts, S.C.; Stapleton, H.M. Exposures, mechanisms, and impacts of endocrine-active flame retardants. Curr. Opin. Pharmacol. 2014, 19, 125–133. [Google Scholar] [CrossRef] [PubMed]
  12. Acir, I.H.; Guenther, K. Endocrine-disrupting metabolites of alkylphenol ethoxylates—A critical review of analytical methods, environmental occurrences, toxicity, and regulation. Sci. Total Environ. 2018, 635, 1530–1546. [Google Scholar] [CrossRef] [PubMed]
  13. Gore, A.C.; Chappell, V.A.; Fenton, S.E.; Flaws, J.A.; Nadal, A.; Prins, G.S.; Toppari, J.; Zoeller, R.T. EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr. Rev. 2015, 36, E1–E150. [Google Scholar] [CrossRef] [PubMed]
  14. Yilmaz, B.; Terekeci, H.; Sandal, S.; Kelestimur, F. Endocrine disrupting chemicals: Exposure, effects on human health, mechanism of action, models for testing and strategies for prevention. Rev. Endocr. Metab. Disord. 2020, 21, 127–147. [Google Scholar] [CrossRef] [PubMed]
  15. He, J.; Xu, J.; Zheng, M.; Pan, K.; Yang, L.; Ma, L.; Wang, C.; Yu, J. Thyroid dysfunction caused by exposure to environmental endocrine disruptors and the underlying mechanism: A review. Chem. Biol. Interact. 2024, 391, 110909. [Google Scholar] [CrossRef] [PubMed]
  16. La Merrill, M.A.; Vandenberg, L.N.; Smith, M.T.; Goodson, W.; Browne, P.; Patisaul, H.B.; Guyton, K.Z.; Kortenkamp, A.; Cogliano, V.J.; Woodruff, T.J.; et al. Consensus on the key characteristics of endocrine-disrupting chemicals as a basis for hazard identification. Nat. Rev. Endocrinol. 2020, 16, 45–57. [Google Scholar] [CrossRef] [PubMed]
  17. Tchounwou, P.B.; Yedjou, C.G.; Patlolla, A.K.; Sutton, D.J. Heavy metal toxicity and the environment. Exp. Suppl. 2012, 101, 133–164. [Google Scholar] [CrossRef] [PubMed]
  18. Balali-Mood, M.; Naseri, K.; Tahergorabi, Z.; Khazdair, M.R.; Sadeghi, M. Toxic Mechanisms of Five Heavy Metals: Mercury, Lead, Chromium, Cadmium, and Arsenic. Front. Pharmacol. 2021, 12, 643972. [Google Scholar] [CrossRef]
  19. Sall, M.L.; Diaw, A.K.D.; Gningue-Sall, D.; Efremova Aaron, S.; Aaron, J.J. Toxic heavy metals: Impact on the environment and human health, and treatment with conducting organic polymers, a review. Environ. Sci. Pollut. Res. Int. 2020, 27, 29927–29942. [Google Scholar] [CrossRef]
  20. Vogel, C.F.; Van Winkle, L.S.; Esser, C.; Haarmann-Stemmann, T. The aryl hydrocarbon receptor as a target of environmental stressors—Implications for pollution mediated stress and inflammatory responses. Redox. Biol. 2020, 34, 101530. [Google Scholar] [CrossRef]
  21. Zhou, H.; Wu, H.; Liao, C.; Diao, X.; Zhen, J.; Chen, L.; Xue, Q. Toxicology mechanism of the persistent organic pollutants (POPs) in fish through AhR pathway. Toxicol. Mech. Methods 2010, 20, 279–286. [Google Scholar] [CrossRef] [PubMed]
  22. Zhou, H.; Qu, Y.; Wu, H.; Liao, C.; Zheng, J.; Diao, X.; Xue, Q. Molecular phylogenies and evolutionary behavior of AhR (aryl hydrocarbon receptor) pathway genes in aquatic animals: Implications for the toxicology mechanism of some persistent organic pollutants (POPs). Chemosphere 2010, 78, 193–205. [Google Scholar] [CrossRef] [PubMed]
  23. Doan, T.Q.; Connolly, L.; Igout, A.; Nott, K.; Muller, M.; Scippo, M.L. In vitro profiling of the potential endocrine disrupting activities affecting steroid and aryl hydrocarbon receptors of compounds and mixtures prevalent in human drinking water resources. Chemosphere 2020, 258, 127332. [Google Scholar] [CrossRef]
  24. Poland, A.; Glover, E. 2,3,7,8-Tetrachlorodibenzo-p-dioxin: A potent inducer of -aminolevulinic acid synthetase. Science 1973, 179, 476–477. [Google Scholar] [CrossRef] [PubMed]
  25. Sweeney, M.H.; Mocarelli, P. Human health effects after exposure to 2,3,7,8-TCDD. Food Addit. Contam. 2000, 17, 303–316. [Google Scholar] [CrossRef] [PubMed]
  26. National Academies of Sciences, Engineering, and Medicine; Health and Medicine Division; Board on Population Health and Public Health Practice; Committee to Review the Health Effects in Vietnam Veterans of Exposure to Herbicides (Eleventh Biennial Update). Veterans and Agent Orange: Update 11 (2018); National Academies Press: Washington, DC, USA, 2018. [Google Scholar]
  27. Poland, A.; Kende, A. 2,3,7,8-Tetrachlorodibenzo-p-dioxin: Environmental contaminant and molecular probe. Fed. Proc. 1976, 35, 2404–2411. [Google Scholar]
  28. Mandal, P.K. Dioxin: A review of its environmental effects and its aryl hydrocarbon receptor biology. J. Comp. Physiol. B 2005, 175, 221–230. [Google Scholar] [CrossRef] [PubMed]
  29. Opitz, C.A.; Holfelder, P.; Prentzell, M.T.; Trump, S. The complex biology of aryl hydrocarbon receptor activation in cancer and beyond. Biochem. Pharmacol. 2023, 216, 115798. [Google Scholar] [CrossRef]
  30. Puga, A.; Ma, C.; Marlowe, J.L. The aryl hydrocarbon receptor cross-talks with multiple signal transduction pathways. Biochem. Pharmacol. 2009, 77, 713–722. [Google Scholar] [CrossRef]
  31. Murray, I.A.; Patterson, A.D.; Perdew, G.H. Aryl hydrocarbon receptor ligands in cancer: Friend and foe. Nat. Rev. Cancer 2014, 14, 801–814. [Google Scholar] [CrossRef]
  32. McGuire, J.; Whitelaw, M.L.; Pongratz, I.; Gustafsson, J.A.; Poellinger, L. A cellular factor stimulates ligand-dependent release of hsp90 from the basic helix-loop-helix dioxin receptor. Mol. Cell. Biol. 1994, 14, 2438–2446. [Google Scholar] [CrossRef] [PubMed]
  33. Bell, D.R.; Poland, A. Binding of aryl hydrocarbon receptor (AhR) to AhR-interacting protein. The role of hsp90. J. Biol. Chem. 2000, 275, 36407–36414. [Google Scholar] [CrossRef]
  34. Nukaya, M.; Lin, B.C.; Glover, E.; Moran, S.M.; Kennedy, G.D.; Bradfield, C.A. 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. 2010, 285, 35599–35605. [Google Scholar] [CrossRef] [PubMed]
  35. Kazlauskas, A.; Poellinger, L.; Pongratz, I. Evidence that the co-chaperone p23 regulates ligand responsiveness of the dioxin (Aryl hydrocarbon) receptor. J. Biol. Chem. 1999, 274, 13519–13524. [Google Scholar] [CrossRef] [PubMed]
  36. Meyer, B.K.; Perdew, G.H. Characterization of the AhR-hsp90-XAP2 core complex and the role of the immunophilin-related protein XAP2 in AhR stabilization. Biochemistry 1999, 38, 8907–8917. [Google Scholar] [CrossRef] [PubMed]
  37. Sugatani, J.; Yamakawa, K.; Tonda, E.; Nishitani, S.; Yoshinari, K.; Degawa, M.; Abe, I.; Noguchi, H.; Miwa, M. The induction of human UDP-glucuronosyltransferase 1A1 mediated through a distal enhancer module by flavonoids and xenobiotics. Biochem. Pharmacol. 2004, 67, 989–1000. [Google Scholar] [CrossRef]
  38. Auyeung, D.J.; Kessler, F.K.; Ritter, J.K. Mechanism of rat UDP-glucuronosyltransferase 1A6 induction by oltipraz: Evidence for a contribution of the Aryl hydrocarbon receptor pathway. Mol. Pharmacol. 2003, 63, 119–127. [Google Scholar] [CrossRef] [PubMed]
  39. Münzel, P.A.; Schmohl, S.; Buckler, F.; Jaehrling, J.; Raschko, F.T.; Köhle, C.; Bock, K.W. Contribution of the Ah receptor to the phenolic antioxidant-mediated expression of human and rat UDP-glucuronosyltransferase UGT1A6 in Caco-2 and rat hepatoma 5L cells. Biochem. Pharmacol. 2003, 66, 841–847. [Google Scholar] [CrossRef]
  40. Mimura, J.; Ema, M.; Sogawa, K.; Fujii-Kuriyama, Y. Identification of a novel mechanism of regulation of Ah (dioxin) receptor function. Genes Dev. 1999, 13, 20–25. [Google Scholar] [CrossRef]
  41. Wilson, S.R.; Joshi, A.D.; Elferink, C.J. The tumor suppressor Kruppel-like factor 6 is a novel aryl hydrocarbon receptor DNA binding partner. J. Pharmacol. Exp. Ther. 2013, 345, 419–429. [Google Scholar] [CrossRef]
  42. Wormke, M.; Stoner, M.; Saville, B.; Walker, K.; Abdelrahim, M.; Burghardt, R.; Safe, S. The aryl hydrocarbon receptor mediates degradation of estrogen receptor alpha through activation of proteasomes. Mol. Cell. Biol. 2003, 23, 1843–1855. [Google Scholar] [CrossRef] [PubMed]
  43. Vogel, C.F.; Sciullo, E.; Li, W.; Wong, P.; Lazennec, G.; Matsumura, F. RelB, a new partner of aryl hydrocarbon receptor-mediated transcription. Mol. Endocrinol. 2007, 21, 2941–2955. [Google Scholar] [CrossRef] [PubMed]
  44. Oesch-Bartlomowicz, B.; Huelster, A.; Wiss, O.; Antoniou-Lipfert, P.; Dietrich, C.; Arand, M.; Weiss, C.; Bockamp, E.; Oesch, F. Aryl hydrocarbon receptor activation by cAMP vs. dioxin: Divergent signaling pathways. Proc. Natl. Acad. Sci. USA 2005, 102, 9218–9223. [Google Scholar] [CrossRef] [PubMed]
  45. Möglich, A.; Ayers, R.A.; Moffat, K. Structure and signaling mechanism of Per-ARNT-Sim domains. Structure 2009, 17, 1282–1294. [Google Scholar] [CrossRef] [PubMed]
  46. Denison, M.S.; Soshilov, A.A.; He, G.; DeGroot, D.E.; Zhao, B. Exactly the same but different: Promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol. Sci. 2011, 124, 1–22. [Google Scholar] [CrossRef] [PubMed]
  47. Soshilov, A.A.; Denison, M.S. Ligand promiscuity of aryl hydrocarbon receptor agonists and antagonists revealed by site-directed mutagenesis. Mol. Cell. Biol. 2014, 34, 1707–1719. [Google Scholar] [CrossRef] [PubMed]
  48. Denison, M.S.; Nagy, S.R. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu. Rev. Pharmacol. Toxicol. 2003, 43, 309–334. [Google Scholar] [CrossRef]
  49. Gruszczyk, J.; Grandvuillemin, L.; Lai-Kee-Him, J.; Paloni, M.; Savva, C.G.; Germain, P.; Grimaldi, M.; Boulahtouf, A.; Kwong, H.S.; Bous, J.; et al. Cryo-EM structure of the agonist-bound Hsp90-XAP2-AHR cytosolic complex. Nat. Commun. 2022, 13, 7010. [Google Scholar] [CrossRef] [PubMed]
  50. Ema, M.; Ohe, N.; Suzuki, M.; Mimura, J.; Sogawa, K.; Ikawa, S.; Fujii-Kuriyama, Y. Dioxin binding activities of polymorphic forms of mouse and human arylhydrocarbon receptors. J. Biol. Chem. 1994, 269, 27337–27343. [Google Scholar] [CrossRef]
  51. Poland, A.; Glover, E. Characterization and strain distribution pattern of the murine Ah receptor specified by the Ahd and Ahb-3 alleles. Mol. Pharmacol. 1990, 38, 306–312. [Google Scholar]
  52. Moriguchi, T.; Motohashi, H.; Hosoya, T.; Nakajima, O.; Takahashi, S.; Ohsako, S.; Aoki, Y.; Nishimura, N.; Tohyama, C.; Fujii-Kuriyama, Y.; et al. Distinct response to dioxin in an arylhydrocarbon receptor (AHR)-humanized mouse. Proc. Natl. Acad. Sci. USA 2003, 100, 5652–5657. [Google Scholar] [CrossRef] [PubMed]
  53. Dai, S.; Qu, L.; Li, J.; Zhang, Y.; Jiang, L.; Wei, H.; Guo, M.; Chen, X.; Chen, Y. Structural insight into the ligand binding mechanism of aryl hydrocarbon receptor. Nat. Commun. 2022, 13, 6234. [Google Scholar] [CrossRef] [PubMed]
  54. Denison, M.S.; Pandini, A.; Nagy, S.R.; Baldwin, E.P.; Bonati, L. Ligand binding and activation of the Ah receptor. Chem. Biol. Interact. 2002, 141, 3–24. [Google Scholar] [CrossRef] [PubMed]
  55. Xing, Y.; Nukaya, M.; Satyshur, K.A.; Jiang, L.; Stanevich, V.; Korkmaz, E.N.; Burdette, L.; Kennedy, G.D.; Cui, Q.; Bradfield, C.A. Identification of the Ah-receptor structural determinants for ligand preferences. Toxicol. Sci. 2012, 129, 86–97. [Google Scholar] [CrossRef] [PubMed]
  56. Safe, S.; Lee, S.O.; Jin, U.H. Role of the aryl hydrocarbon receptor in carcinogenesis and potential as a drug target. Toxicol. Sci. 2013, 135, 1–16. [Google Scholar] [CrossRef] [PubMed]
  57. Flaveny, C.A.; Murray, I.A.; Chiaro, C.R.; Perdew, G.H. Ligand selectivity and gene regulation by the human aryl hydrocarbon receptor in transgenic mice. Mol. Pharmacol. 2009, 75, 1412–1420. [Google Scholar] [CrossRef] [PubMed]
  58. Lohmann, R.; Breivik, K.; Dachs, J.; Muir, D. Global fate of POPs: Current and future research directions. Environ. Pollut. 2007, 150, 150–165. [Google Scholar] [CrossRef] [PubMed]
  59. Ashraf, M.A. Persistent organic pollutants (POPs): A global issue, a global challenge. Environ. Sci. Pollut. Res. Int. 2017, 24, 4223–4227. [Google Scholar] [CrossRef] [PubMed]
  60. Bock, K.W. Aryl hydrocarbon receptor (AHR)-mediated inflammation and resolution: Non-genomic and genomic signaling. Biochem. Pharmacol. 2020, 182, 114220. [Google Scholar] [CrossRef]
  61. Piwarski, S.A.; Salisbury, T.B. The effects of environmental aryl hydrocarbon receptor ligands on signaling and cell metabolism in cancer. Biochem. Pharmacol. 2023, 216, 115771. [Google Scholar] [CrossRef]
  62. Zhang, W.; Xie, H.Q.; Li, Y.; Zhou, M.; Zhou, Z.; Wang, R.; Hahn, M.E.; Zhao, B. The aryl hydrocarbon receptor: A predominant mediator for the toxicity of emerging dioxin-like compounds. J. Hazard. Mater. 2022, 426, 128084. [Google Scholar] [CrossRef] [PubMed]
  63. Hites, R.A. Dioxins: An overview and history. Environ. Sci. Technol. 2011, 45, 16–20. [Google Scholar] [CrossRef]
  64. Behnisch, P.A.; Hosoe, K.; Sakai, S. Bioanalytical screening methods for dioxins and dioxin-like compounds a review of bioassay/biomarker technology. Environ. Int. 2001, 27, 413–439. [Google Scholar] [CrossRef] [PubMed]
  65. Manisalidis, I.; Stavropoulou, E.; Stavropoulos, A.; Bezirtzoglou, E. Environmental and Health Impacts of Air Pollution: A Review. Front. Public Health 2020, 8, 14. [Google Scholar] [CrossRef] [PubMed]
  66. Pirkle, J.L.; Wolfe, W.H.; Patterson, D.G.; Needham, L.L.; Michalek, J.E.; Miner, J.C.; Peterson, M.R.; Phillips, D.L. Estimates of the half-life of 2,3,7,8-tetrachlorodibenzo-p-dioxin in Vietnam Veterans of Operation Ranch Hand. J. Toxicol. Environ. Health 1989, 27, 165–171. [Google Scholar] [CrossRef]
  67. Kerger, B.D.; Leung, H.W.; Scott, P.; Paustenbach, D.J.; Needham, L.L.; Patterson, D.G., Jr.; Gerthoux, P.M.; Mocarelli, P. Age- and concentration-dependent elimination half-life of 2,3,7,8-tetrachlorodibenzo-p-dioxin in Seveso children. Environ. Health Perspect. 2006, 114, 1596–1602. [Google Scholar] [CrossRef] [PubMed]
  68. Saurat, J.H.; Kaya, G.; Saxer-Sekulic, N.; Pardo, B.; Becker, M.; Fontao, L.; Mottu, F.; Carraux, P.; Pham, X.C.; Barde, C.; et al. The cutaneous lesions of dioxin exposure: Lessons from the poisoning of Victor Yushchenko. Toxicol. Sci. 2012, 125, 310–317. [Google Scholar] [CrossRef] [PubMed]
  69. Pelclová, D.; Urban, P.; Preiss, J.; Lukás, E.; Fenclová, Z.; Navrátil, T.; Dubská, Z.; Senholdová, Z. Adverse health effects in humans exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Rev. Environ. Health 2006, 21, 119–138. [Google Scholar] [CrossRef] [PubMed]
  70. Furue, M.; Tsuji, G. Chloracne and Hyperpigmentation Caused by Exposure to Hazardous Aryl Hydrocarbon Receptor Ligands. Int. J. Environ. Res. Public Health 2019, 16, 4864. [Google Scholar] [CrossRef]
  71. Cole, P.; Trichopoulos, D.; Pastides, H.; Starr, T.; Mandel, J.S. Dioxin and cancer: A critical review. Regul. Toxicol. Pharmacol. 2003, 38, 378–388. [Google Scholar] [CrossRef]
  72. Steenland, K.; Bertazzi, P.; Baccarelli, A.; Kogevinas, M. Dioxin revisited: Developments since the 1997 IARC classification of dioxin as a human carcinogen. Environ. Health Perspect. 2004, 112, 1265–1268. [Google Scholar] [CrossRef] [PubMed]
  73. Phillips, D.H. Polycyclic aromatic hydrocarbons in the diet. Mutat. Res. 1999, 443, 139–147. [Google Scholar] [CrossRef] [PubMed]
  74. Mastrangelo, G.; Fadda, E.; Marzia, V. Polycyclic aromatic hydrocarbons and cancer in man. Environ. Health Perspect. 1996, 104, 1166–1170. [Google Scholar] [CrossRef]
  75. Li, Z.; Romanoff, L.; Bartell, S.; Pittman, E.N.; Trinidad, D.A.; McClean, M.; Webster, T.F.; Sjödin, A. Excretion profiles and half-lives of ten urinary polycyclic aromatic hydrocarbon metabolites after dietary exposure. Chem. Res. Toxicol. 2012, 25, 1452–1461. [Google Scholar] [CrossRef]
  76. Motorykin, O.; Santiago-Delgado, L.; Rohlman, D.; Schrlau, J.E.; Harper, B.; Harris, S.; Harding, A.; Kile, M.L.; Massey Simonich, S.L. Metabolism and excretion rates of parent and hydroxy-PAHs in urine collected after consumption of traditionally smoked salmon for Native American volunteers. Sci. Total Environ. 2015, 514, 170–177. [Google Scholar] [CrossRef] [PubMed]
  77. Cecinato, A.; Bacaloni, A.; Romagnoli, P.; Perilli, M.; Balducci, C. Molecular signatures of organic particulates as tracers of emission sources. Environ. Sci. Pollut. Res. Int. 2022, 29, 65904–65923. [Google Scholar] [CrossRef]
  78. Yang, L.; Zhang, H.; Zhang, X.; Xing, W.; Wang, Y.; Bai, P.; Zhang, L.; Hayakawa, K.; Toriba, A.; Tang, N. Exposure to Atmospheric Particulate Matter-Bound Polycyclic Aromatic Hydrocarbons and Their Health Effects: A Review. Int. J. Environ. Res. Public Health 2021, 18, 2177. [Google Scholar] [CrossRef]
  79. Møller, M.; Alfheim, I. Mutagenicity and PAH-analysis of airborne particulate matter. Atmos. Environ. 1980, 14, 83–88. [Google Scholar] [CrossRef]
  80. Sjaastad, A.K.; Jørgensen, R.B.; Svendsen, K. Exposure to polycyclic aromatic hydrocarbons (PAHs), mutagenic aldehydes and particulate matter during pan frying of beefsteak. Occup. Environ. Med. 2010, 67, 228–232. [Google Scholar] [CrossRef]
  81. Anderson, K.E.; Kadlubar, F.F.; Kulldorff, M.; Harnack, L.; Gross, M.; Lang, N.P.; Barber, C.; Rothman, N.; Sinha, R. Dietary intake of heterocyclic amines and benzo(a)pyrene: Associations with pancreatic cancer. Cancer Epidemiol. Biomark. Prev. 2005, 14, 2261–2265. [Google Scholar] [CrossRef]
  82. Andreotti, G.; Silverman, D.T. Occupational risk factors and pancreatic cancer: A review of recent findings. Mol. Carcinog. 2012, 51, 98–108. [Google Scholar] [CrossRef] [PubMed]
  83. Alguacil, J.; Porta, M.; Kauppinen, T.; Malats, N.; Kogevinas, M.; Carrato, A. PANKRAS II Study Group. Occupational exposure to dyes, metals, polycyclic aromatic hydrocarbons and other agents and K-ras activation in human exocrine pancreatic cancer. Int. J. Cancer 2003, 107, 635–641. [Google Scholar] [CrossRef] [PubMed]
  84. Starek-Świechowicz, B.; Budziszewska, B.; Starek, A. Hexachlorobenzene as a persistent organic pollutant: Toxicity and molecular mechanism of action. Pharmacol. Rep. 2017, 69, 1232–1239. [Google Scholar] [CrossRef] [PubMed]
  85. Gocmen, A.; Peters, H.A.; Cripps, D.J.; Bryan, G.T.; Morris, C.R. Hexachlorobenzene episode in Turkey. Biomed. Environ. Sci. 1989, 2, 36–43. [Google Scholar] [PubMed]
  86. Miret, N.V.; Pontillo, C.A.; Zárate, L.V.; Kleiman de Pisarev, D.; Cocca, C.; Randi, A.S. Impact of endocrine disruptor hexachlorobenzene on the mammary gland and breast cancer: The story thus far. Environ. Res. 2019, 173, 330–341. [Google Scholar] [CrossRef] [PubMed]
  87. Hoppin, J.A.; Tolbert, P.E.; Holly, E.A.; Brock, J.W.; Korrick, S.A.; Altshul, L.M.; Zhang, R.H.; Bracci, P.M.; Burse, V.W.; Needham, L.L. Pancreatic cancer and serum organochlorine levels. Cancer Epidemiol. Biomark. Prev. 2000, 9, 199–205. [Google Scholar]
  88. Bosch de Basea, M.; Porta, M.; Alguacil, J.; Puigdomènech, E.; Gasull, M.; Garrido, J.A.; López, T.; PANKRAS II Study Group. Relationships between occupational history and serum concentrations of organochlorine compounds in exocrine pancreatic cancer. Occup. Environ. Med. 2011, 68, 332–338. [Google Scholar] [CrossRef]
  89. Porta, M.; Bosch de Basea, M.; Benavides, F.G.; López, T.; Fernandez, E.; Marco, E.; Alguacil, J.; Grimalt, J.O.; Puigdomènech, E.; PANKRAS II Study Group. Differences in serum concentrations of organochlorine compounds by occupational social class in pancreatic cancer. Environ. Res. 2008, 108, 370–379. [Google Scholar] [CrossRef] [PubMed]
  90. Rochester, J.R. Bisphenol A and human health: A review of the literature. Reprod. Toxicol. 2013, 42, 132–155. [Google Scholar] [CrossRef]
  91. Abraham, A.; Chakraborty, P. A review on sources and health impacts of bisphenol, A. Rev. Environ. Health 2020, 35, 201–210. [Google Scholar] [CrossRef]
  92. Teeguarden, J.G.; Waechter, J.M., Jr.; Clewell, H.J., 3rd; Covington, T.R.; Barton, H.A. Evaluation of oral and intravenous route pharmacokinetics, plasma protein binding, and uterine tissue dose metrics of bisphenol A: A physiologically based pharmacokinetic approach. Toxicol. Sci. 2005, 85, 823–838. [Google Scholar] [CrossRef] [PubMed]
  93. Völkel, W.; Bittner, N.; Dekant, W. Quantitation of bisphenol A and bisphenol A glucuronide in biological samples by high performance liquid chromatography-tandem mass spectrometry. Drug Metab. Dispos. 2005, 33, 1748–1757. [Google Scholar] [CrossRef] [PubMed]
  94. Farrugia, F.; Aquilina, A.; Vassallo, J.; Pace, N.P. Bisphenol A and Type 2 Diabetes Mellitus: A Review of Epidemiologic, Functional, and Early Life Factors. Int. J. Environ. Res. Public Health 2021, 18, 716. [Google Scholar] [CrossRef] [PubMed]
  95. Hwang, S.; Lim, J.E.; Choi, Y.; Jee, S.H. Bisphenol A exposure and type 2 diabetes mellitus risk: A meta-analysis. BMC Endocr. Disord. 2018, 18, 81. [Google Scholar] [CrossRef] [PubMed]
  96. Alonso-Magdalena, P.; Morimoto, S.; Ripoll, C.; Fuentes, E.; Nadal, A. The estrogenic effect of bisphenol A disrupts pancreatic beta-cell function in vivo and induces insulin resistance. Environ. Health Perspect. 2006, 114, 106–112. [Google Scholar] [CrossRef] [PubMed]
  97. Martinez-Pinna, J.; Marroqui, L.; Hmadcha, A.; Lopez-Beas, J.; Soriano, S.; Villar-Pazos, S.; Alonso-Magdalena, P.; Dos Santos, R.S.; Quesada, I.; Martin, F.; et al. Oestrogen receptor β mediates the actions of bisphenol-A on ion channel expression in mouse pancreatic beta cells. Diabetologia 2019, 62, 1667–1680. [Google Scholar] [CrossRef] [PubMed]
  98. Boronat-Belda, T.; Ferrero, H.; Al-Abdulla, R.; Quesada, I.; Gustafsson, J.A.; Nadal, Á.; Alonso-Magdalena, P. Bisphenol-A exposure during pregnancy alters pancreatic β-cell division and mass in male mice offspring: A role for ERβ. Food Chem. Toxicol. 2020, 145, 111681. [Google Scholar] [CrossRef] [PubMed]
  99. Alonso-Magdalena, P.; Ropero, A.B.; Carrera, M.P.; Cederroth, C.R.; Baquié, M.; Gauthier, B.R.; Nef, S.; Stefani, E.; Nadal, A. Pancreatic insulin content regulation by the estrogen receptor ER alpha. PLoS ONE 2008, 3, e2069. [Google Scholar] [CrossRef]
  100. Soriano, S.; Alonso-Magdalena, P.; García-Arévalo, M.; Novials, A.; Muhammed, S.J.; Salehi, A.; Gustafsson, J.A.; Quesada, I.; Nadal, A. Rapid insulinotropic action of low doses of bisphenol-A on mouse and human islets of Langerhans: Role of estrogen receptor β. PLoS ONE 2012, 7, e31109. [Google Scholar] [CrossRef]
  101. Ziv-Gal, A.; Craig, Z.R.; Wang, W.; Flaws, J.A. Bisphenol A inhibits cultured mouse ovarian follicle growth partially via the aryl hydrocarbon receptor signaling pathway. Reprod. Toxicol. 2013, 42, 58–67. [Google Scholar] [CrossRef]
  102. Nishizawa, H.; Imanishi, S.; Manabe, N. Effects of exposure in utero to bisphenol a on the expression of aryl hydrocarbon receptor, related factors, and xenobiotic metabolizing enzymes in murine embryos. J. Reprod. Dev. 2005, 51, 593–605. [Google Scholar] [CrossRef] [PubMed]
  103. Donini, C.F.; El Helou, M.; Wierinckx, A.; Győrffy, B.; Aires, S.; Escande, A.; Croze, S.; Clezardin, P.; Lachuer, J.; Diab-Assaf, M.; et al. Long-Term Exposure of Early-Transformed Human Mammary Cells to Low Doses of Benzo[a]pyrene and/or Bisphenol A Enhances Their Cancerous Phenotype via an AhR/GPR30 Interplay. Front. Oncol. 2020, 10, 712. [Google Scholar] [CrossRef]
  104. Banerjee, O.; Singh, S.; Prasad, S.K.; Bhattacharjee, A.; Seal, T.; Mandal, J.; Sinha, S.; Banerjee, A.; Maji, B.K.; Mukherjee, S. Exploring aryl hydrocarbon receptor (AhR) as a target for Bisphenol-A (BPA)-induced pancreatic islet toxicity and impaired glucose homeostasis: Protective efficacy of ethanol extract of Centella asiatica. Toxicology 2023, 500, 153693. [Google Scholar] [CrossRef] [PubMed]
  105. Jaishankar, M.; Tseten, T.; Anbalagan, N.; Mathew, B.B.; Beeregowda, K.N. Toxicity, mechanism and health effects of some heavy metals. Interdiscip. Toxicol. 2014, 7, 60–72. [Google Scholar] [CrossRef] [PubMed]
  106. Witkowska, D.; Słowik, J.; Chilicka, K. Heavy Metals and Human Health: Possible Exposure Pathways and the Competition for Protein Binding Sites. Molecules 2021, 26, 6060. [Google Scholar] [CrossRef]
  107. Fu, Z.; Xi, S. The effects of heavy metals on human metabolism. Toxicol. Mech. Methods 2020, 30, 167–176. [Google Scholar] [CrossRef]
  108. Jan, A.T.; Azam, M.; Siddiqui, K.; Ali, A.; Choi, I.; Haq, Q.M. Heavy Metals and Human Health: Mechanistic Insight into Toxicity and Counter Defense System of Antioxidants. Int. J. Mol. Sci. 2015, 16, 29592–29630. [Google Scholar] [CrossRef] [PubMed]
  109. Elbekai, R.H.; El-Kadi, A.O. Modulation of aryl hydrocarbon receptor-regulated gene expression by arsenite, cadmium, and chromium. Toxicology 2004, 202, 249–269. [Google Scholar] [CrossRef]
  110. Kann, S.; Huang, M.Y.; Estes, C.; Reichard, J.F.; Sartor, M.A.; Xia, Y.; Puga, A. Arsenite-induced aryl hydrocarbon receptor nuclear translocation results in additive induction of phase I genes and synergistic induction of phase II genes. Mol. Pharmacol. 2005, 68, 336–346. [Google Scholar] [CrossRef]
  111. Albores, A.; Cebrián, M.E.; Bach, P.H.; Connelly, J.C.; Hinton, R.H.; Bridges, J.W. Sodium arsenite induced alterations in bilirubin excretion and heme metabolism. J. Biochem. Toxicol. 1989, 4, 73–78. [Google Scholar] [CrossRef]
  112. Kou, Z.; Yang, R.; Lee, E.; Cuddapah, S.; Choi, B.H.; Dai, W. Oxidative stress modulates expression of immune checkpoint genes via activation of AhR signaling. Toxicol. Appl. Pharmacol. 2022, 457, 116314. [Google Scholar] [CrossRef] [PubMed]
  113. Anwar-Mohamed, A.; Elbekai, R.H.; El-Kadi, A.O. Regulation of CYP1A1 by heavy metals and consequences for drug metabolism. Expert Opin. Drug Metab. Toxicol. 2009, 5, 501–521. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, Z.; Yang, P.; Xie, J.; Lin, H.P.; Kumagai, K.; Harkema, J.; Yang, C. Arsenic and benzo[a]pyrene co-exposure acts synergistically in inducing cancer stem cell-like property and tumorigenesis by epigenetically down-regulating SOCS3 expression. Environ Int. 2020, 137, 105560. [Google Scholar] [CrossRef] [PubMed]
  115. Antwi, S.O.; Eckert, E.C.; Sabaque, C.V.; Leof, E.R.; Hawthorne, K.M.; Bamlet, W.R.; Chaffee, K.G.; Oberg, A.L.; Petersen, G.M. Exposure to environmental chemicals and heavy metals, and risk of pancreatic cancer. Cancer Causes Control 2015, 26, 1583–1591. [Google Scholar] [CrossRef]
  116. Djordjevic, V.R.; Wallace, D.R.; Schweitzer, A.; Boricic, N.; Knezevic, D.; Matic, S.; Grubor, N.; Kerkez, M.; Radenkovic, D.; Bulat, Z.; et al. Environmental cadmium exposure and pancreatic cancer: Evidence from case control, animal and in vitro studies. Environ. Int. 2019, 128, 353–361. [Google Scholar] [CrossRef] [PubMed]
  117. Carrigan, P.E.; Hentz, J.G.; Gordon, G.; Morgan, J.L.; Raimondo, M.; Anbar, A.D.; Miller, L.J. Distinctive heavy metal composition of pancreatic juice in patients with pancreatic carcinoma. Cancer Epidemiol. Biomark. Prev. 2007, 16, 2656–2663. [Google Scholar] [CrossRef] [PubMed]
  118. Pothuraju, R.; Rachagani, S.; Junker, W.M.; Chaudhary, S.; Saraswathi, V.; Kaur, S.; Batra, S.K. Pancreatic cancer associated with obesity and diabetes: An alternative approach for its targeting. J. Exp. Clin. Cancer Res. 2018, 37, 319. [Google Scholar] [CrossRef] [PubMed]
  119. Sun, H.; Saeedi, P.; Karuranga, S.; Pinkepank, M.; Ogurtsova, K.; Duncan, B.B.; Stein, C.; Basit, A.; Chan, J.C.N.; Claude Mbanya, J.; et al. Erratum to “IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045” [Diabetes Res. Clin. Pract. 183 (2022) 109119]. Diabetes Res. Clin. Pract. 2023, 204, 110945. [Google Scholar] [CrossRef] [PubMed]
  120. Ma, D.M.; Dong, X.W.; Han, X.; Ling, Z.; Lu, G.T.; Sun, Y.Y.; Yin, X.D. Pancreatitis and Pancreatic Cancer Risk. Technol. Cancer Res. Treat. 2023, 22, 15330338231164875. [Google Scholar] [CrossRef]
  121. Bodin, J.; Stene, L.C.; Nygaard, U.C. Can exposure to environmental chemicals increase the risk of diabetes type 1 development? Biomed. Res. Int. 2015, 2015, 208947. [Google Scholar] [CrossRef]
  122. Lim, C.C.; Thurston, G.D. Air Pollution, Oxidative Stress, and Diabetes: A Life Course Epidemiologic Perspective. Curr. Diabetes Rep. 2019, 19, 58. [Google Scholar] [CrossRef]
  123. Hao, N.; Whitelaw, M.L. The emerging roles of AhR in physiology and immunity. Biochem. Pharmacol. 2013, 86, 561–570. [Google Scholar] [CrossRef]
  124. Kerkvliet, N.I.; Steppan, L.B.; Vorachek, W.; Oda, S.; Farrer, D.; Wong, C.P.; Pham, D.; Mourich, D.V. Activation of aryl hydrocarbon receptor by TCDD prevents diabetes in NOD mice and increases Foxp3+ T cells in pancreatic lymph nodes. Immunotherapy 2009, 1, 539–547. [Google Scholar] [CrossRef]
  125. Ehrlich, A.K.; Pennington, J.M.; Wang, X.; Rohlman, D.; Punj, S.; Löhr, C.V.; Newman, M.T.; Kolluri, S.K.; Kerkvliet, N.I. Activation of the Aryl Hydrocarbon Receptor by 10-Cl-BBQ Prevents Insulitis and Effector T Cell Development Independently of Foxp3+ Regulatory T Cells in Nonobese Diabetic Mice. J. Immunol. 2016, 196, 264–273. [Google Scholar] [CrossRef]
  126. Kelishadi, R.; Hovsepian, S.; Amin, M.M.; Mozafarian, N.; Sedaghat, S.; Hashemipour, M. Association of Polycyclic Aromatic Hydrocarbons Urine Metabolites with Type 1 Diabetes. J. Diabetes Res. 2023, 2023, 6692810. [Google Scholar] [CrossRef]
  127. Ou, K.; Song, J.; Zhang, S.; Fang, L.; Lin, L.; Lan, M.; Chen, M.; Wang, C. Prenatal exposure to a mixture of PAHs causes the dysfunction of islet cells in adult male mice: Association with type 1 diabetes mellitus. Ecotoxicol. Environ. Saf. 2022, 239, 113695. [Google Scholar] [CrossRef]
  128. Tosirisuk, N.; Sakorn, N.; Jantarat, C.; Nosoongnoen, W.; Aroonpakmongkol, S.; Supornsilchai, V. Increased bisphenol A levels in Thai children and adolescents with type 1 diabetes mellitus. Pediatr. Int. 2022, 64, e14944. [Google Scholar] [CrossRef]
  129. Bodin, J.; Bølling, A.K.; Becher, R.; Kuper, F.; Løvik, M.; Nygaard, U.C. Transmaternal bisphenol A exposure accelerates diabetes type 1 development in NOD mice. Toxicol. Sci. 2014, 137, 311–323. [Google Scholar] [CrossRef]
  130. Cetkovic-Cvrlje, M.; Thinamany, S.; Bruner, K.A. Bisphenol A (BPA) aggravates multiple low-dose streptozotocin-induced Type 1 diabetes in C57BL/6 mice. J. Immunotoxicol. 2017, 14, 160–168. [Google Scholar] [CrossRef]
  131. Bodin, J.; Kocbach Bølling, A.; Wendt, A.; Eliasson, L.; Becher, R.; Kuper, F.; Løvik, M.; Nygaard, U.C. Exposure to bisphenol, A. but not phthalates, increases spontaneous diabetes type 1 development in NOD mice. Toxicol. Rep. 2015, 2, 99–110. [Google Scholar] [CrossRef]
  132. Chafe, R.; Aslanov, R.; Sarkar, A.; Gregory, P.; Comeau, A.; Newhook, L.A. Association of type 1 diabetes and concentrations of drinking water components in Newfoundland and Labrador, Canada. BMJ Open Diabetes Res. Care 2018, 6, e000466. [Google Scholar] [CrossRef]
  133. Grau-Pérez, M.; Kuo, C.C.; Spratlen, M.; Thayer, K.A.; Mendez, M.A.; Hamman, R.F.; Dabelea, D.; Adgate, J.L.; Knowler, W.C.; Bell, R.A.; et al. The Association of Arsenic Exposure and Metabolism With Type 1 and Type 2 Diabetes in Youth: The SEARCH Case-Control Study. Diabetes Care 2017, 40, 46–53. [Google Scholar] [CrossRef]
  134. Dávila-Esqueda, M.E.; Morales, J.M.; Jiménez-Capdeville, M.E.; De la Cruz, E.; Falcón-Escobedo, R.; Chi-Ahumada, E.; Martin-Pérez, S. Low-level subchronic arsenic exposure from prenatal developmental stages to adult life results in an impaired glucose homeostasis. Exp. Clin. Endocrinol. Diabetes 2011, 119, 613–617. [Google Scholar] [CrossRef]
  135. Henriksen, G.L.; Ketchum, N.S.; Michalek, J.E.; Swaby, J.A. Serum dioxin and diabetes mellitus in veterans of Operation Ranch Hand. Epidemiology 1997, 8, 252–258. [Google Scholar] [CrossRef]
  136. Remillard, R.B.; Bunce, N.J. Linking dioxins to diabetes: Epidemiology and biologic plausibility. Environ. Health Perspect. 2002, 110, 853–858. [Google Scholar] [CrossRef]
  137. Lee, D.H.; Lee, I.K.; Song, K.; Steffes, M.; Toscano, W.; Baker, B.A.; Jacobs, D.R., Jr. A strong dose-response relation between serum concentrations of persistent organic pollutants and diabetes: Results from the National Health and Examination Survey 1999-2002. Diabetes Care 2006, 29, 1638–1644. [Google Scholar] [CrossRef]
  138. Novelli, M.; Piaggi, S.; De Tata, V. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced impairment of glucose-stimulated insulin secretion in isolated rat pancreatic islets. Toxicol. Lett. 2005, 156, 307–314. [Google Scholar] [CrossRef]
  139. Piaggi, S.; Novelli, M.; Martino, L.; Masini, M.; Raggi, C.; Orciuolo, E.; Masiello, P.; Casini, A.; De Tata, V. Cell death and impairment of glucose-stimulated insulin secretion induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the beta-cell line INS-1E. Toxicol. Appl. Pharmacol. 2007, 220, 333–340. [Google Scholar] [CrossRef]
  140. Kubi, J.A.; Chen, A.C.H.; Fong, S.W.; Lai, K.P.; Wong, C.K.C.; Yeung, W.S.B.; Lee, K.F.; Lee, Y.L. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the differentiation of embryonic stem cells towards pancreatic lineage and pancreatic beta cell function. Environ. Int. 2019, 130, 104885. [Google Scholar] [CrossRef]
  141. Novelli, M.; Beffy, P.; Masini, M.; Vantaggiato, C.; Martino, L.; Marselli, L.; Marchetti, P.; De Tata, V. Selective beta-cell toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin on isolated pancreatic islets. Chemosphere 2021, 265, 129103. [Google Scholar] [CrossRef]
  142. Ibrahim, M.; MacFarlane, E.M.; Matteo, G.; Hayek, M.P.; Rick, K.R.C.; Farokhi, S.; Copley, C.M.; O’Dwyer, S.; Bruin, J.E. Functional cytochrome P450 1A enzymes are induced in mouse and human islets following pollutant exposure. Diabetologia 2020, 63, 162–178. [Google Scholar] [CrossRef]
  143. Kuzgun, G.; Başaran, R.; Arıoğlu İnan, E.; Can Eke, B. Effects of insulin treatment on hepatic CYP1A1 and CYP2E1 activities and lipid peroxidation levels in streptozotocin-induced diabetic rats. J. Diabetes Metab. Disord. 2020, 19, 1157–1164. [Google Scholar] [CrossRef]
  144. Wang, C.; Xu, C.X.; Krager, S.L.; Bottum, K.M.; Liao, D.F.; Tischkau, S.A. Aryl hydrocarbon receptor deficiency enhances insulin sensitivity and reduces PPAR-α pathway activity in mice. Environ. Health Perspect. 2011, 119, 1739–1744. [Google Scholar] [CrossRef]
  145. Hoyeck, M.P.; Blair, H.; Ibrahim, M.; Solanki, S.; Elsawy, M.; Prakash, A.; Rick, K.R.C.; Matteo, G.; O’Dwyer, S.; Bruin, J.E. Long-term metabolic consequences of acute dioxin exposure differ between male and female mice. Sci. Rep. 2020, 10, 1448. [Google Scholar] [CrossRef]
  146. Lee, Y.M.; Ha, C.M.; Kim, S.A.; Thoudam, T.; Yoon, Y.R.; Kim, D.J.; Kim, H.C.; Moon, H.B.; Park, S.; Lee, I.K.; et al. Low-Dose Persistent Organic Pollutants Impair Insulin Secretory Function of Pancreatic β-Cells: Human and In Vitro Evidence. Diabetes 2017, 66, 2669–2680. [Google Scholar] [CrossRef]
  147. Pérez-Bermejo, M.; Mas-Pérez, I.; Murillo-Llorente, M.T. The Role of the Bisphenol A in Diabetes and Obesity. Biomedicines 2021, 9, 666. [Google Scholar] [CrossRef]
  148. Stallings-Smith, S.; Mease, A.; Johnson, T.M.; Arikawa, A.Y. Exploring the association between polycyclic aromatic hydrocarbons and diabetes among adults in the United States. Environ. Res. 2018, 166, 588–594. [Google Scholar] [CrossRef]
  149. Wu, H.; Bertrand, K.A.; Choi, A.L.; Hu, F.B.; Laden, F.; Grandjean, P.; Sun, Q. Persistent organic pollutants and type 2 diabetes: A prospective analysis in the nurses’ health study and meta-analysis. Environ. Health Perspect. 2013, 121, 153–161. [Google Scholar] [CrossRef]
  150. Ji, J.H.; Jin, M.H.; Kang, J.H.; Lee, S.I.; Lee, S.; Kim, S.H.; Oh, S.Y. Relationship between heavy metal exposure and type 2 diabetes: A large-scale retrospective cohort study using occupational health examinations. BMJ Open 2021, 11, e039541. [Google Scholar] [CrossRef]
  151. Khan, A.R.; Awan, F.R. Metals in the pathogenesis of type 2 diabetes. J. Diabetes Metab. Disord. 2014, 13, 16. [Google Scholar] [CrossRef]
  152. Weiss, F.U.; Laemmerhirt, F.; Lerch, M.M. Etiology and Risk Factors of Acute and Chronic Pancreatitis. Visc. Med. 2019, 35, 73–81. [Google Scholar] [CrossRef]
  153. Kasai, A.; Hiramatsu, N.; Hayakawa, K.; Yao, J.; Maeda, S.; Kitamura, M. High levels of dioxin-like potential in cigarette smoke evidenced by in vitro and in vivo biosensing. Cancer Res. 2006, 66, 7143–7150. [Google Scholar] [CrossRef]
  154. Kitamura, M.; Kasai, A. Cigarette smoke as a trigger for the dioxin receptor-mediated signaling pathway. Cancer Lett. 2007, 252, 184–194. [Google Scholar] [CrossRef]
  155. Park, S.M.; Kim, K.B.; Han, J.H.; Kim, N.; Kang, T.U.; Swan, H.; Kim, H.J. Incidence and risk of pancreatic cancer in patients with acute or chronic pancreatitis: A population-based cohort study. Sci. Rep. 2023, 13, 18930. [Google Scholar] [CrossRef]
  156. Umans, D.S.; Hoogenboom, S.A.; Sissingh, N.J.; Lekkerkerker, S.J.; Verdonk, R.C.; van Hooft, J.E. Pancreatitis and pancreatic cancer: A case of the chicken or the egg. World J. Gastroenterol. 2021, 27, 3148–3157. [Google Scholar] [CrossRef]
  157. Xue, J.; Nguyen, D.T.; Habtezion, A. Aryl hydrocarbon receptor regulates pancreatic IL-22 production and protects mice from acute pancreatitis. Gastroenterology 2012, 143, 1670–1680. [Google Scholar] [CrossRef]
  158. Ghosh, J.; Chowdhury, A.R.; Srinivasan, S.; Chattopadhyay, M.; Bose, M.; Bhattacharya, S.; Raza, H.; Fuchs, S.Y.; Rustgi, A.K.; Gonzalez, F.J.; et al. Cigarette Smoke Toxins-Induced Mitochondrial Dysfunction and Pancreatitis Involves Aryl Hydrocarbon Receptor Mediated Cyp1 Gene Expression: Protective Effects of Resveratrol. Toxicol. Sci. 2018, 166, 428–440. [Google Scholar] [CrossRef]
  159. Lee, J.E.; Cho, S.G.; Ko, S.G.; Ahrmad, S.A.; Puga, A.; Kim, K. Regulation of a long noncoding RNA MALAT1 by aryl hydrocarbon receptor in pancreatic cancer cells and tissues. Biochem. Biophys. Res. Commun. 2020, 532, 563–569. [Google Scholar] [CrossRef]
  160. Kamata, K.; Hara, A.; Minaga, K.; Yoshikawa, T.; Kurimoto, M.; Sekai, I.; Okai, N.; Omaru, N.; Masuta, Y.; Otsuka, Y.; et al. Activation of the aryl hydrocarbon receptor inhibits the development of experimental autoimmune pancreatitis through IL-22-mediated signaling pathways. Clin. Exp. Immunol. 2023, 212, 171–183. [Google Scholar] [CrossRef]
  161. Xue, J.; Zhao, Q.; Sharma, V.; Nguyen, L.P.; Lee, Y.N.; Pham, K.L.; Edderkaoui, M.; Pandol, S.J.; Park, W.; Habtezion, A. Aryl Hydrocarbon Receptor Ligands in Cigarette Smoke Induce Production of Interleukin-22 to Promote Pancreatic Fibrosis in Models of Chronic Pancreatitis. Gastroenterology 2016, 151, 1206–1217. [Google Scholar] [CrossRef] [PubMed]
  162. Rawla, P.; Sunkara, T.; Gaduputi, V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J. Oncol. 2019, 10, 10–27. [Google Scholar] [CrossRef] [PubMed]
  163. Hu, J.X.; Zhao, C.F.; Chen, W.B.; Liu, Q.C.; Li, Q.W.; Lin, Y.Y.; Gao, F. Pancreatic cancer: A review of epidemiology, trend, and risk factors. World J. Gastroenterol. 2021, 27, 4298–4321. [Google Scholar] [CrossRef] [PubMed]
  164. Zhang, Q.; Zeng, L.; Chen, Y.; Lian, G.; Qian, C.; Chen, S.; Li, J.; Huang, K. Pancreatic Cancer Epidemiology, Detection, and Management. Gastroenterol. Res. Pract. 2016, 2016, 8962321. [Google Scholar] [CrossRef] [PubMed]
  165. Porta, M.; Gasull, M.; Pumarega, J.; Kiviranta, H.; Rantakokko, P.; Raaschou-Nielsen, O.; Bergdahl, I.A.; Sandanger, T.M.; Agudo, A.; Rylander, C.; et al. Plasma concentrations of persistent organic pollutants and pancreatic cancer risk. Int. J. Epidemiol. 2022, 51, 479–490. [Google Scholar] [CrossRef] [PubMed]
  166. Helou, K.; Harmouche-Karaki, M.; Karake, S.; Narbonne, J.F. A review of organochlorine pesticides and polychlorinated biphenyls in Lebanon: Environmental and human contaminants. Chemosphere 2019, 231, 357–368. [Google Scholar] [CrossRef] [PubMed]
  167. Nyska, A.; Jokinen, M.P.; Brix, A.E.; Sells, D.M.; Wyde, M.E.; Orzech, D.; Haseman, J.K.; Flake, G.; Walker, N.J. Exocrine pancreatic pathology in female Harlan Sprague-Dawley rats after chronic treatment with 2,3,7,8-tetrachlorodibenzo-p-dioxin and dioxin-like compounds. Environ. Health Perspect. 2004, 112, 903–909. [Google Scholar] [CrossRef] [PubMed]
  168. Koliopanos, A.; Kleeff, J.; Xiao, Y.; Safe, S.; Zimmermann, A.; Büchler, M.W.; Friess, H. Increased arylhydrocarbon receptor expression offers a potential therapeutic target for pancreatic cancer. Oncogene 2002, 21, 6059–6070. [Google Scholar] [CrossRef] [PubMed]
  169. Jin, U.H.; Kim, S.B.; Safe, S. Omeprazole Inhibits Pancreatic Cancer Cell Invasion through a Nongenomic Aryl Hydrocarbon Receptor Pathway. Chem. Res. Toxicol. 2015, 28, 907–918. [Google Scholar] [CrossRef] [PubMed]
  170. Korac, K.; Rajasekaran, D.; Sniegowski, T.; Schniers, B.K.; Ibrahim, A.F.; Bhutia, Y.D. Carbidopa, an activator of aryl hydrocarbon receptor, suppresses IDO1 expression in pancreatic cancer and decreases tumor growth. Biochem. J. 2022, 479, 1807–1824. [Google Scholar] [CrossRef]
  171. Stukas, D.; Jasukaitiene, A.; Bartkeviciene, A.; Matthews, J.; Maimets, T.; Teino, I.; Jaudzems, K.; Gulbinas, A.; Dambrauskas, Z. Targeting AHR Increases Pancreatic Cancer Cell Sensitivity to Gemcitabine through the ELAVL1-DCK Pathway. Int. J. Mol. Sci. 2023, 24, 13155. [Google Scholar] [CrossRef]
  172. Cheng, J.; Li, W.; Kang, B.; Zhou, Y.; Song, J.; Dan, S.; Yang, Y.; Zhang, X.; Li, J.; Yin, S.; et al. Tryptophan derivatives regulate the transcription of Oct4 in stem-like cancer cells. Nat. Commun. 2015, 6, 7209. [Google Scholar] [CrossRef] [PubMed]
  173. Hezaveh, K.; Shinde, R.S.; Klötgen, A.; Halaby, M.J.; Lamorte, S.; Ciudad, M.T.; Quevedo, R.; Neufeld, L.; Liu, Z.Q.; Jin, R.; et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 2022, 55, 324–340.e8. [Google Scholar] [CrossRef] [PubMed]
  174. Safe, S.; Jin, U.H.; Park, H.; Chapkin, R.S.; Jayaraman, A. Aryl Hydrocarbon Receptor (AHR) Ligands as Selective AHR Modulators (SAhRMs). Int. J. Mol. Sci. 2020, 21, 6654. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Activation of AHR signaling in canonical and non-canonical manners. Upon ligand binding, AHR translocates into the nucleus in the canonical pathway, where it forms a heterodimer with ARNT. This complex then interacts with Xenobiotic Response Elements (XRE) on target gene promoters, initiating downstream gene transcription. Conversely, in the non-canonical pathway, ligand-bound AHR interacts with alternative transcription factors such as KLF6, ERα, and NFκB, leading to the activation of gene transcription or degradation of transcription factors independent of XRE interaction.
Figure 1. Activation of AHR signaling in canonical and non-canonical manners. Upon ligand binding, AHR translocates into the nucleus in the canonical pathway, where it forms a heterodimer with ARNT. This complex then interacts with Xenobiotic Response Elements (XRE) on target gene promoters, initiating downstream gene transcription. Conversely, in the non-canonical pathway, ligand-bound AHR interacts with alternative transcription factors such as KLF6, ERα, and NFκB, leading to the activation of gene transcription or degradation of transcription factors independent of XRE interaction.
Ijms 25 03818 g001
Figure 2. AHR structure and ligand binding pocket (LBP). The domain structure of human AHR contains bHLH, PAS A, PASB, and TAD, totaling 850 amino acids. (A) A close-up view of the ligand binding pocket (LBP in magenta color) shows a transparent surface with the ligand indirubin (middle). The AHR LBP interacts with the AHR agonist indirubin (B).
Figure 2. AHR structure and ligand binding pocket (LBP). The domain structure of human AHR contains bHLH, PAS A, PASB, and TAD, totaling 850 amino acids. (A) A close-up view of the ligand binding pocket (LBP in magenta color) shows a transparent surface with the ligand indirubin (middle). The AHR LBP interacts with the AHR agonist indirubin (B).
Ijms 25 03818 g002
Figure 3. Role of AHR in EDC-mediated pancreatic pathogenesis. The references related to T1DM are [124,125,126,127], T2DM [138,139,140,141,142,143,144,145,146], acute pancreatitis [157,158,159,160], chronic pancreatitis [161], and pancreatic cancer [167,168,169,170,171,172,173].
Figure 3. Role of AHR in EDC-mediated pancreatic pathogenesis. The references related to T1DM are [124,125,126,127], T2DM [138,139,140,141,142,143,144,145,146], acute pancreatitis [157,158,159,160], chronic pancreatitis [161], and pancreatic cancer [167,168,169,170,171,172,173].
Ijms 25 03818 g003
Table 1. A summary of the interplays between endocrine-disrupting chemicals (EDCs), AHR signaling, epidemiological reports, and relevant findings on the roles of these EDC-mediated AHR signaling regulations in the development of pancreatic diseases and cancer. References are indicated as [ ].
Table 1. A summary of the interplays between endocrine-disrupting chemicals (EDCs), AHR signaling, epidemiological reports, and relevant findings on the roles of these EDC-mediated AHR signaling regulations in the development of pancreatic diseases and cancer. References are indicated as [ ].
EDCRegulation of AHR SignalingEpidemiological Studies Relevant to Pancreatic Diseases or CancerMechanistic Role of AHR in Pancreatic Diseases and Cancer
Dioxin and dioxin-like compoundsAHR agonists [60,61,62]See 5. Roles of EDC–AHR Interactions in the Pathogenesis of Pancreatic Diseases and CancerSee 5. Roles of EDC–AHR Interactions in the Pathogenesis of Pancreatic Diseases and Cancer
Polycyclic aromatic hydrocarbonsAHR agonists and oxidative stress inducers [73,74,75,76,77,78,79,80][81,82,83]Unknown
HexachlorobenzeneWeak AHR agonist [86][87,88,89]Unknown
Bisphenol AWeak AHR agonist
[101,102,103,104]
[94,95,96,97,98,99,100]Unknown
Heavy metalsAHR agonists and oxidative stress inducers [109,110,111,112,113,114][115,116,117]Unknown
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, K. The Role of Endocrine Disruption Chemical-Regulated Aryl Hydrocarbon Receptor Activity in the Pathogenesis of Pancreatic Diseases and Cancer. Int. J. Mol. Sci. 2024, 25, 3818. https://doi.org/10.3390/ijms25073818

AMA Style

Kim K. The Role of Endocrine Disruption Chemical-Regulated Aryl Hydrocarbon Receptor Activity in the Pathogenesis of Pancreatic Diseases and Cancer. International Journal of Molecular Sciences. 2024; 25(7):3818. https://doi.org/10.3390/ijms25073818

Chicago/Turabian Style

Kim, Kyounghyun. 2024. "The Role of Endocrine Disruption Chemical-Regulated Aryl Hydrocarbon Receptor Activity in the Pathogenesis of Pancreatic Diseases and Cancer" International Journal of Molecular Sciences 25, no. 7: 3818. https://doi.org/10.3390/ijms25073818

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop