- Review
- Open access
- Published:
The aryl hydrocarbon receptor: a new frontier in male reproductive system
Reproductive Biology and Endocrinology volume 23, Article number: 70 (2025)
Abstract
Background
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor historically recognized for its role in the regulation of toxicity mediated by environmental chemicals. Recent research points to AhR’s critical participation in male reproductive physiology, particularly in spermatogenesis, hormone signaling, and the maintenance of sperm quality. Both endogenous ligands (e.g., dietary and gut microbiota-derived metabolites) and exogenous pollutants (e.g., dioxins and benzo-α-pyrene) influence AhR-mediated pathways, making it a key link between environmental exposures and male fertility.
Results
This review highlights AhR’s influence on the male reproductive system, emphasizing the role of endogenous AhR ligands and AhR expression in the maturation and function of male reproductive organs. Environmental AhR agonists have been shown to induce oxidative stress, hormonal imbalance, and sperm DNA damage, which impact harmfully on the spermatogenesis process, which leads to reproductive abnormalities. Conversely, certain natural compounds such as resveratrol, curcumin, and lycopene appear to antagonize AhR activation and reduce its negative effects, thus offering potential protective benefits against male reproductive toxicity. Nevertheless, discrepancies persist regarding the exact interplay between AhR signaling and critical reproductive hormones such as testosterone and LH, and it remains unclear how transgenerational epigenetic changes triggered by AhR activation might affect long-term male fertility.
Conclusion
AhR is pivotal in male reproductive physiology, influencing spermatogenesis, sperm quality, and hormone regulation through its interactions with both endogenous and environmental ligands. Persistent pollutants such as dioxins and polycyclic aromatic hydrocarbons cause oxidative damage and hormonal disturbances via AhR, contributing to reduced sperm quality and fertility.
Graphical Abstract
The Impact of ligands of Aryl Hydrocarbon Receptor (AhR) on Male Reproductive Health. →, activation; \(\dashv\), inhibition; DEHP, Di(2-ethylhexyl) phthalate; I3C, indole-3-carbinol; PAH, polycyclic aromatic hydrocarbon; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin (Created by Biorender.com).

Introduction
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that connects various external stimuli, including environmental, dietary, microbial, and metabolic. The AhR regulates transcriptional programs in a ligand-specific, cell-type-specific, and context-specific manner [1]. Scientific research has elucidated the complexities of AhR’s structure and function since the first report on identifying the AhR was published in 1976 [2]. Despite decades of research on the AhR, its precise functions in male reproductive biology remain incompletely understood and sometimes contradictory. Since then, the biological effects of AhR have been extensively studied across various physiological functions such as the nervous system [3, 4], immune system [5, 6], digestive system [7, 8], hepatic function [9,10,11], respiratory system [5, 12, 13], renal system [14, 15], skin health [16], and others [17]. Moreover, the complex network of AhR extends its influence on the realm of reproduction, adding another layer of complexity to its biological significance. However, findings often diverge regarding whether AhR activation promotes or inhibits male germ cell development. It impacts the intricate cellular processes involved in reproduction, playing a pivotal role in fertility, embryonic development, and hormone signaling. While different studies report detrimental effects (e.g., reduced sperm counts, disrupted hormone levels, or testicular pathology following AhR ligand exposure), others highlight AhR’s protective or homeostatic roles through its endogenous ligands and crosstalk with various signaling pathways [18,19,20]. The interaction of AhR with endogenous and exogenous ligands modulates gene expression and contributes to the delicate balance of reproductive functions [18,19,20]. Investigating the relationship between AhR and reproductive processes not only enhances our understanding of fertility, reproductive disorders, and the impact of environmental factors on reproductive health but also unveils the intricate molecular mechanisms underlying critical aspects of life.
That critical gap relates to the interplay between AhR and other hormone receptors, such as androgen and estrogen receptors. AhR can antagonize or augment these signals through direct protein–protein interactions or co-occupancy at shared response elements on DNA, leading to uncertain net effects on testosterone production, luteinizing hormone levels, and Sertoli cell function. There has been a limited number of reviews on the role of AhR in the regulation of the male reproductive system. As the new role of AhR is becoming recognized, we believe that a timely review that explores its role in male reproductive systems is warranted. To this end, this review provides an overview of AhR’s structure, function, and signaling pathways. Additionally, we consider its impact on male reproductive health and disorders, focusing on the influence of environmental factors, and identifying potential therapeutic applications in these areas.
Aryl hydrocarbon receptor
The AhR is a diverse and dynamic cytosolic receptor with expression in an extensive range of tissues throughout both developmental stages and adulthood [21]. Its widespread presence underscores its crucial nature in myriad biological processes, highlighting its role as a key regulator in maintaining homeostasis and responding to environmental cues [22]. The varied expression of AhR across tissues and life stages provides solid evidence of its involvement in vital physiological functions, suggesting its role in orchestrating complex biological systems [23]. Renowned as a transcription factor, AhR is activated by ligands and holds significant importance in an array of physiological functions such as xenobiotic metabolism, cellular proliferation, and developmental pathways. Notably, its importance has been reaffirmed in the field of reproductive studies thanks to its crucial regulatory functions [24].
History of aryl hydrocarbon receptor discovery
The aryl hydrocarbon receptor (AhR) was initially characterized in the 1970 s when researchers investigated the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) [25, 26]. The human AhR cDNA was identified in 1993, as being expressed at the highest levels in the placenta, lungs, and heart [27]. Although first recognized as a key mediator of xenobiotic metabolism (e.g., driving cytochrome P450 induction), more recent work has highlighted AhR’s influence on reproductive processes and immune regulation [28,29,30,31,32].
Human epidemiological evidence linking AhR to male reproductive outcomes
The most notable human data that discusses the relationship between AhR and reproductive toxicity began in 1976 following Seveso chemical explosion in Italy, which exposed residents to extremely high levels of TCDD [33] Subsequent epidemiological follow-ups revealed several long-term health impacts, including changes in sex ratios (fewer male births) and reduced sperm quality in exposed men [34]. Additionally, the study noted decreases in sperm count, motility, and normal morphology, suggesting that acid-induced AhR activation contributed to these adverse effects on male fertility. Moreover, evidence arises from occupational exposure studies; the men working in industries with high dioxin or polycyclic aromatic hydrocarbon (PAH) exposure (e.g., aluminum smelting, steel production, or pesticide manufacturing) have shown altered semen parameters and, in some cohorts, lower testosterone levels or gonadotropin imbalances [35].
Outstanding questions in AhR-mediated male reproductive regulation
Despite the progress made so far, there remain several key uncertainties regarding how the AhR influences male reproductive biology. Also, a key question that remains to be addressed is whether germline epigenetic changes arising from AhR activation could be passed down through multiple generations. Yet another concern involves the degree to which chronic, low-level exposure to AhR ligands subtly affects hormone production and semen characteristics, and whether specific developmental windows—such as puberty or adulthood—are more vulnerable [28, 35]. Further complexities include the ways in which AhR may interact with other endocrine pathways and how dietary or microbial metabolites acting as AhR ligands might influence reproductive outcomes [29].
Structure of aryl hydrocarbon receptor
The AhR is a transcription factor that belongs to the basic helix-loop-helix Per-Arnt-Sim (bHLH-PAS) family. Its structure consists of three domains (Fig. 1) [36]: the core DNA-binding domain, the N-terminal PAS domain, and the C-terminal ligand-binding domain [37, 38]. Combined, these domains give the AhR the capacity to bind ligands, regulate gene expression, and recognize environmental stimuli [39].
The functional domains of the AhR, ARNT and AhRR proteins: AhR domain structure; PAS: Per-Arnt-Sim (A and B); Q-rich: glutamine; S/T/P: serine, threonine & proline [36]
To sense a wide array of ligands and initiate conformational changes that lead to AhR activation, the N-terminal bHLH/PAS domains are vital [37]. Furthermore, these domains facilitate the translocation of the ligand-bound AhR-ARNT complex into the nucleus, where it interacts with target gene promoters and binds to DNA [40]. Because of its complex ligand interactions, the central domain delicately manages gene expression by refining AhR’s response to external stimuli [41].
Moreover, AhR possesses nuclear export signals (NES) and nuclear localization signals that dictate its movement between the cytoplasm and nucleus [42, 43]. Guided to the nucleus by the NLS, the AhR regulates gene expression. The NES consequently facilitates AhR’s export from the nucleus, ensuring that transcriptional responses conclude promptly [44].
Trans-activation of transcription is achieved through the interaction of AhR’s trans-activation domain (TAD) with co-activators, such as ARNT, CBP/p300, and SRC-1. Through the dynamic modulation of the TAD’s activity by post-translational modifications like phosphorylation and acetylation of histones, AhR can tailor gene expression responses to different cellular contexts and enhance gene transcription.
Understanding of AhR’s three-dimensional structure and ligand-binding pockets has been advanced by structural studies, including homology modeling and ligand docking experiments. These studies have also illuminated the molecular mechanisms that allow AhR’s flexible ligand-binding capacity [45, 46]. In-depth investigations reveal the complex nuances of these conformational shifts, which are critical for AhR activation and subsequent gene transcription. These conformational alterations are prompted by ligand binding.
When AhR is idle, it is usually positioned in the cytosol where it is associated with a cluster of chaperone proteins that maintains its stability and prevent its degradation [47, 48]. This stable form of AhR in the cytosol, identified as the AhR-low activity variant, displays a diminished responsiveness to ligand activation [41].
Cytosolic aryl hydrocarbon receptor
Under normal physiological conditions, the AhR is naturally found in an idle mode in the cytosol of various cells. This status is also known as the AhR-low activity variant, which is less responsive to activation by ligands [49]. In its stable cytosolic form, the AhR is associated with a complex of chaperone proteins, including AhR-interacting protein (AIP), Heat shock protein 90 (HSP90), Prohibitin 23 (p23), and X-associated protein 2 (XAP2). These proteins help maintain its structural integrity and prevent its degradation in the absence of ligand binding. Furthermore, the stabilization of AhR within the cytoplasm, which prevents unliganded nucleocytoplasmic shuttling, protects AhR from degradation by the ubiquitin–proteasome pathway, as illustrated in Fig. 2 [50].
The AIP/AhR/Hsp90/p23 complex. AhR: Aryl hydrocarbon receptor; AIP: Aryl hydrocarbon receptor-interacting protein; Hsp: Heat shock protein Hsp90-AhR-p23 complex [50]
Aryl hydrocarbon receptor repressor
The aryl hydrocarbon receptor repressor (AhRR) has been identified as a target gene of AhR, providing a unique mechanism of feedback inhibition of AhR function where the transcription factor directly induces the expression of its repressor through binding to its cognate regulatory sequence located in the promoter of the target gene. The N-terminal portion of the AhRR protein shows significant structural resemblance to AhR, particularly housing the DNA-binding bHLH domain and the PAS-A domain (Fig. 1). However, the C-terminal segment lacks the PAS-B and Q-rich TADs, suggesting that AhRR does not possess the well-defined AhR ligand-binding domain and remains transcriptionally inactive. AhRR expression is regulated by one or more xenobiotic response elements (XREs) found in the enhancer/promoter sequence of the murine and human Ahrr gene. AhRR can dimerize with ARNT and function as a specific inhibitor of AhR activity by competing with AhR to form heterodimers with ARNT, thus preventing the binding and trans-activation of AhR/ARNT complexes via XREs (Fig. 3) [51]. The ability of AhRR to regulate AhR’s signaling in major cellular processes like cell cycling, inflammation, and apoptosis is complex and varies depending on the cellular and environmental context. Moreover, in vitro experiments with multiple distinct types of cancer cell lines indicated that AhRR acts as a tumor suppressor gene [52]. Notably, a recent investigation uncovered numerous unique DNA binding sites within the promoter regions of tumor suppressor genes and genes associated with cancer development, specifically targeted by AhRR [53]. Additionally, human studies found epigenetic modifications of AhRR’s regulatory region associated with exposure to cigarette smoke and the development of various cancer types [54]. Conversely, a study found that overexpressing AhRR in a transgenic mouse model led to the induction of Cytochrome P450 1 A1 (CYP1 A1) by TCDD in a tissue-specific manner and that the overexpression of AhRR protected against hepatic injury and acute TCDD toxicity. CYP1 A1 is known for its involvement in the bioactivation of certain procarcinogens, converting them into more reactive forms capable of binding to DNA and potentially causing mutations. Thus, in the mouse model, overexpression of AhRR protected the mice from cancer development by preventing TCDD from activating CYP1 A1 [55, 56].
Schematic illustration of the repression of the canonical AhR signaling pathway by AhRR [51]
Aryl hydrocarbon receptor nuclear translocator
The AhR nuclear translocator (ARNT) is a protein that is essential for the function and comprehensive regulatory network of the AhR signaling pathway [57]. When AhR is activated in the cytoplasm, and the appropriate compounds bind within the AhR’s PAS domain, there are conformational changes and nuclear localization signals are unmasked [58]. Subsequently, AhR is translocated into the nucleus, where it is heterodimerized with ARNT. The transcriptionally active AhR/ARNT complex binds to XREs within the regulatory region of target genes to initiate transcription [39].
The ARNT is made up of a bHLH domain necessary for DNA binding, two essential PAS domains (PAS-A and PAS-B) for dimerization, and one trans-activation domain that enables its dimerization with AhR and binding to XREs [59]. The nuclear localization signal plays a role in oxygen sensing and cellular adaptation to hypoxic conditions, earning it the alternative name of HIF-1β [60]. ARNT operates at the intersection of multiple signaling pathways, contributing to the fine-tuning of AhR-mediated responses. ARNT features characteristic PAS domains and a bHLH motif, which facilitates its dimerization with AhR and binding to XREs [39].
AhR activation mechanisms (ligand activation to gene expression)
The AhR was frequently referred to as the “dioxin receptor” due to early detection of its role in regulating toxic responses to environmental pollutants including dioxins. However, recent advancements have unveiled its multifaceted role in regulating immunity, cell proliferation, and differentiation [61]. Moreover, the activation of AhR is a complex process comprising several steps:
Endogenous role of AhR
AhR-null mouse models suggest that AhR has a role in liver development: the livers of AhR-null mice tend to be smaller, with portal fibrosis, premature lipid accumulation, and further changes leading to the differential expression of hundreds of genes [62, 63]. Moreover, another study revealed that AhR deficiency exhibits sex-dependent defects in rats [64], while AhR knockout mice show unregulated matrix remodeling [65] and various cardiac dysfunctions [66]. Hence, it appears that AhR participates in the development of the liver, ovaries, cardiovascular system, immune system, and kidney formation in mammals [67]. The AhR has also been identified in early metazoans and multiple invertebrate species like Drosophila melanogaster and Caenorhabditis elegans, where it contributes to neuronal development [68]. Interestingly, invertebrate AhR orthologues do not seem to bind toxic AhR ligands, such as TCDD [69]. There are multiple candidate endogenous AhR ligands, including indoles, which are produced from dietary tryptophan by gut bacteria, and amino acid metabolites like prostaglandins and lipoxins; or tetrapyrroles, like bilirubin and biliverdin, 6-formyl (3,2-b) carbazole (FICZ), and 2-(1’H-indole-3-carbonyl)thiazole-4-carboxylic acid methyl ester (ITE), etc. [70]. Furthermore, indole-3-carbinol (I3 C) is another indole found in high concentration in cruciferous vegetables such as broccoli, cabbage and cauliflower and serves as an agonist of AhR which leads to downregulation of estrogen metabolism in estrogen-related cancers [71]; and ameliorates the colitis outcomes through modulation of mucin production [72]. However, although these compounds have been shown to bind AhR and trigger the expression of AhR target genes in most cases, their actual in vivo significance necessitates further investigation.
One of the most intriguing internal functions of AhR appears to be the modulation of cell proliferation in situations devoid of xenobiotic binding. Studies involving AhR-null mouse embryonic fibroblasts have demonstrated that AhR promotes progressive cell cycle activity even in the absence of an exogenous ligand [25, 73]. Conversely, low levels of TCDD can inhibit DNA synthesis in mouse epithelial cell cultures, rat primary hepatocytes [58, 69], or in rat liver following partial hepatectomy [74]. Consider, for instance, rat hepatoma 5L (AhR-positive) and BP8 (AhR-negative) cells. It has been observed that 5L cells proliferate more rapidly than BP8 cells [25, 58, 69]; furthermore, TCDD exposure induces G1 arrest in 5L cells but does not affect the proliferation of BP8 cells. This cell cycle arrest is linked to an increased expression of the cyclin-dependent kinase 2 (CDK2) inhibitor p27kip1, which in turn prevents the phosphorylation of pRb and subsequent activation of the E2 F transcription factor, a component that governs genes crucial for entry into the S-phase and DNA replication. Additional findings indicate that AhR directly interacts with pRb and represses the E2 F-dependent transcription in hepatoma cells [75,76,77].
Conversely, in contact-inhibited rat liver epithelial cells, AhR ligands cause an opposing effect—the loss of contact inhibition, leading to elevated cell proliferation. This loss of contact inhibition is also an AhR-dependent event [78,79,80,81]. Up-regulation of cell proliferation in confluent WB-F344 rat liver progenitor cells exposed to AhR ligand correlates with increased expression of cyclin A and heightened cyclin A/cdk2 activity [79]. In cells that are not exposed during contact inhibition, cyclin A gene expression is suppressed along with cdk2 activity and cell proliferation [77]. Thus, AhR may have a dual role in cell cycle regulation: its activation might either promote proliferation or act against it, depending on circumstances [82].
Ligands of aryl hydrocarbon receptor
The interaction between a ligand and a receptor is characterized by multiple variables, and the final cellular response depends on the combination of these variables. In other words, the activation of the receptor by two different compounds may result in not just a quantitatively different, but also qualitatively unique cellular response [21, 83,84,85].
The majority of AhR ligands are partial agonists. Partial agonists such as 2,3,3′,4,4′-pentachlorobiphenyl, and galangin possess a similar affinity as that of full agonists, though the inherent activity of a partial agonist is lower than a full agonist. Consequently, partial agonists can never elicit a maximal response, irrespective of whether a partial agonist occupies all receptors. Crucially, partial agonists perform as functional antagonists; when combined with a full agonist, a partial agonist lessens the full agonist’s effect, thereby exhibiting antagonistic behavior [86, 87].
Aryl hydrocarbon receptor and the reproductive system
The 1976 Seveso chemical explosion in Italy exposed residents to elevated levels of TCDD, a toxic chemical compound known for its severe and long-lasting toxic effects. Moreover, TCDD is lipophilic, accumulates in fatty tissues, and has a long half-life in humans, which further exacerbates its impact on those exposed to it. Additionally, Kerger (2011) reported that the explosion caused chloracne and had potential long-term health consequences, including effects on cancer rates. Kerger further noted that the explosion led to altered dental development, changes in the sex ratio, and decreased sperm quality. Moreover, TCDD triggers a cascade of events that can disrupt sperm cell development [88]. TCDD exposure has been shown to interfere with several key steps, leading to reduced sperm production, impaired sperm motility, and other reproductive issues [89]. AhR modulation alters the expression of genes involved in spermatogenesis and sperm function [90]. The rodent studies confirmed that TCDD and similar AhR ligands disrupt multiple steps of spermatogenesis that range from spermatogonia proliferation and meiosis to final sperm maturation [24, 91]. Furthermore, the AhR‐knockout mice showed abnormalities in seminiferous tubules, and reduced expression of protamine (Prm1/Prm2) and other spermatid‐specific genes were consistently observed, which proved the AhR’s regulatory role even in the absence of exogenous ligands [92].
Role of AhR in cell cycle stage
The seminiferous tubule in the testis houses a multitude of germ cells at distinct stages of development and maturation within the spermatogenic cycle. These spermatogenic cells produce mature spermatozoa, which are released into the tubular lumen as functional sperm through the process of spermatogenesis. Spermatogenesis is a complex, dynamic process during which spermatogonia proliferates, differentiates, and transforms into mature spermatozoa. This process occurs in three major stages: the mitotic stage, the meiotic stage, and the maturation stage [92].
Role of AhR in spermatocytogenesis (Interphase and Mitosis)
Interphase
Gap 1 phase
The Gap 1 phase (pre-meiotic) is a critical stage in the development of male germ cells. This phase is characterized by the expression of specific genes that play a role in round spermatids. The Gap 1 phase is also associated with the formation of gap junctions, particularly those involving Cx43, which play a role in the synchronization of germ cell proliferation and differentiation [93, 94]. The role of AhR in the G1 phase of the cell cycle is intricate and context-dependent. The specifics of how AhR action, particularly without an exogenous ligand, promotes cell progression through the G1 phase remain unclear. Additionally, studies using AhR-defective cell line variants and AhR-null mouse embryonic fibroblasts suggest that cells lacking a functional AhR possess a prolonged doubling time, which can mainly be attributed to a delayed progression through the G1 phase [95]. Conversely, exposure to potent AhR ligands like TCDD inhibits G1 phase progression in various cell lines [95, 96]. TCDD-AhR-mediated inhibition of G1 phase progression involves increases in the levels of CDK2 and the inhibitor of p27 KIP1. Furthermore, the inhibition of CDK2 activity by p27 KIP1 leads to a stall in the cell cycle within the G1 phase [75, 97]. The impact of AhR on the cell cycle may differ based on the presence or absence of exogenous ligands and the specific cellular context [98]. Studies demonstrate that disruptions induced by AhR ligand lead to Cx43 degradation, which subsequently causes germ cell proliferation. This alters the differentiation and forms of round spermatids [79].
Synthesis phase (S-phase)
At the synthesis phase, cells undergo DNA replication in preparation for cell division, and it plays a pivotal role in maintaining the spermatogonia stem cell population, which is crucial for continuous sperm production [99]. Recent studies have indicated that AhR can contribute to the p300-mediated induction of DNA synthesis. Furthermore, AhR action in the absence of an exogenous ligand may expedite S-phase progression; however, exposure to the AhR ligand, such as TCDD, leads to the inhibition of DNA replication in various cell lines [69, 100, 101]. The presence of potent AhR ligands can tip the balance from a pro-proliferative role to an anti-proliferative outcome by altering expression of cell cycle regulatory genes and increasing metabolic enzyme expression (e.g., CYP1 A1), which influences the cell’s redox state [69, 100, 101]. Additionally, research shows that TCDD treatment in animals reduces the number of cells in the S-phase and results in the accumulation of cells in the G1 phase [96]. From past studies, it appears that AhR plays a significant role in regulating the S-phase of the cell cycle [91]. Also, AhR activity, influenced by the presence or absence of its ligands and the metabolic activity of P4501 A1, is important in regulating cell cycle progression, particularly during the G1-to-S-phase transition. The alteration of cell cycle progression benefits from the manipulation of AhR activity and its subsequent effects on cell cycle regulators.
Gap 2 phase
The specific role of AhR during the Gap 2 (G2) phase of the cell cycle is not yet well-defined. During the G2 phase, the cell checks itself for any DNA damage following replication, using a control mechanism known as the G2-M DNA damage checkpoint. This provides the cell with a period to correct potential errors in its genome before cell division [102]. Aurora kinase A, a serine/threonine kinase, plays a critical role in facilitating the transition from the G2 phase to mitosis. Recent studies have demonstrated that Aurora kinase A, which is integral to the regulation of the G2/M checkpoint, exhibits dysregulated expression in Aryl Hydrocarbon Receptor (AhR) knockout models (Jones et al., 2021). These findings suggest that AhR may indirectly safeguard genomic integrity during the G2 phase; however, further investigations are required to fully elucidate the mechanisms underlying this process, especially in germ cells [103, 104].
Mitosis
Aurora kinase A promotes entry into mitosis by activating cyclin-dependent kinase 1, essential for the G2/M phase transition of the cell cycle. The specific role of AhR in the various phases of mitosis is not thoroughly studied, with limited information available. A prior study suggests that AhR influences the expression of Aurora kinase A, and its absence or antagonism leads to increased intracellular levels of Aurora kinase A, critical in mitosis, particularly in spindle formation and chromosome segregation [105]. AhR directly binds to DNA at AhR-responsive elements upstream of the Aurora kinase A gene’s transcriptional start site. This binding potentially acts as a transcriptional regulator or enhancer controlling Aurora kinase A expression. Additionally, studies suggest that AhR plays a role in regulating the G2/M phase transition in hematopoietic stem and progenitor cells by influencing the expression of the key mitotic regulator, Aurora kinase A. Dysregulation of Aurora kinase A, as seen with altered AhR signaling, can impact the proliferative capacity of these cells and potentially contribute to diseases.
In prophase, chromosomes condense and become visible, the nuclear envelope disintegrates, and the mitotic spindle begins to form. AhR plays a crucial role in chromatin condensation regulation during prophase, the first stage of mitosis. Chromatin condensation comprises the compaction of DNA into a more organized structure, simplifying proper chromosome alignment and segregation in later stages [106]. Studies have shown that TCDD‐AhR signaling upregulates histone deacetylases by enhancing chromatin compaction. Moreover, excessive compaction or mis‐timed deacetylation, however, can impair accurate chromosome segregation [107]. This allows histones to wrap the DNA more tightly [108,109,110]. Lack of AhR correlates with increased Aurora Kinase A expression and faster progression through mitosis, whereas TCDD‐bound AhR can paradoxically cause mitotic arrest in some contexts [24, 92]. Throughout metaphase, AhR contributes to chromosome alignment at the metaphase plate by enhancing the expression of microtubule-associated and kinetochore proteins, ensuring accurate chromosome segregation [111,112,113]. In anaphase, AhR facilitates sister chromatid segregation by triggering the expression of the anaphase-promoting complex and microtubule motor proteins. These facilitate chromatids’ movement towards opposite poles [114,115,116]. During telophase, AhR is implicated in the reformation of the nuclear envelope and cytokinesis, bolstering the expression of nuclear envelope proteins, actin, and myosin filaments, generating two distinct daughter cells with intact nuclei and completing the mitotic process [117]. Despite AhR’s importance in these mitotic stages, further research is required to fully grasp its regulatory mechanisms.
Meiosis (spermatogenesis)
Meiotic division I and II
AhR is essential for managing meiosis during spermatogenesis [24]. During prophase I of meiotic division I, AhR activation has been linked to the regulation of gene expression impacting the progression of this phase. It manages momentous events such as homologous chromosome pairing and recombination [89]. The effect of AhR on spindle formation and chromosome alignment becomes evident as cells transition into metaphase I, as AhR signaling may assist in the accurate separation of homologous chromosomes during anaphase I [24]. In the context of telophase I, AhR plays a role in the establishment of two distinct haploid cell gametes. Moreover, AhR continues to exert its influence in meiotic division II. In prophase II, AhR contributes to the maintenance of proper chromosomal arrangement and integrity [118].
During metaphase II, AhR contributes to the alignment of chromosomes along the equatorial plane. In anaphase II, it participates in preserving chromatid integrity during their orderly segregation. Furthermore, during telophase II, AhR likely aids in the completion of meiosis, leading to the formation of four haploid cells. In conclusion, AhR appears to be a significant molecular factor in orchestrating the complex processes of meiosis during spermatogenesis, securing the precise transmission of genetic information to the forthcoming generation [21, 89].
Most mechanistic insights come from rodent studies exposing males to TCDD or using AhR‐null mice [21, 80, 95]. Human epidemiological data are more limited and generally rely on retrospective analysis of TCDD‐exposed populations (e.g., Seveso) showing elevated rates of sperm abnormalities [33, 88].
Spermiogenesis
In spermiogenesis, which involves the transformation of spermatids into motile spermatozoa, the AhR plays a pivotal role spanning multiple distinct phases: the Golgi Phase, Cap Phase, Tail Phase, and Maturation Phase. During the Golgi Phase, AhR orchestrates the formation of the acrosome, a specialized organelle that contains enzymes essential for fertilization [119]. As the process transitions to the Cap Phase, AhR may help influence the reshaping of the nucleus and the positioning of the centriole, which are crucial for the subsequent formation of the sperm head and tail. In the Tail Phase, AhR potentially contributes to the development of the flagellum, ensuring proper motility of the mature sperm [120]. In the final Maturation Phase, AhR is likely involved in finalizing the structural and molecular modifications necessary for the sperm cell to achieve full functionality [21, 120,121,122].
Previous studies have displayed abnormalities in the seminiferous tubules of AhR knockout subjects. The expression of genes that are active during spermiogenesis, including Prm1 and Prm2, which encode protamine, was affected [119]. These protamine replace histones late in spermatogenesis and contribute significantly to sperm head condensation [123]. Hspa2 encodes a heat shock protein specifically expressed in spermatogenic cells, acting as a chaperone for the transition proteins that precede protamination. Disruption of HSPA2 is associated with poorly remodeled germ cells with residual mitochondria that generate increased reactive oxygen species. All three of these genes showed lower levels of expression in AhR knockout testes compared to wild-type testes [124].
Spermiation
Spermiation, the process by which mature sperms are released into the lumen of the seminiferous tubules, signifies the conclusion of spermatogenesis. AhR modulates the molecular and cellular events needed for the separation and liberation of mature sperm from Sertoli cells, facilitating the completion of spermatogenesis, including the disassembly of Sertoli–spermatid junctions, cytoskeletal remodeling, and the local regulation of growth factors and cytokines [125]. When AhR is activated by potent ligands, AhR induces enzymes as CYP1 A1 and alters junctional proteins as connexins in a way that disrupts normal spermiation, prematurely detaching immature spermatozoa and ultimately compromising sperm quality as showed in exposure to TCDD [91, 126]. In other rodent models, AhR knockout or ligand‐induced activation correlates with abnormal protamine expression (Prm1, Prm2), dysregulated Aurora kinase A, and premature breakdown of actin‐based cytoskeletal structures, leading to defective spermiation [24, 127]. In summary, the AhR appears to be a critical factor in steering spermiogenesis, taking an active role in the ordered evolution of sperm cells and their final discharge during spermiation [128].
Overall, AhR signaling—whether through potent exogenous ligands such as TCDD or via endogenous modulators—can alter the expression of cell‐cycle regulators, structural proteins (e.g., Aurora Kinase A), and spermatid‐specific genes (Prm1, Prm2, Hspa2). In vivo rodent evidence strongly supports the notion that overactivation of AhR by TCDD disrupts each major phase of spermatogenesis, from spermatogonia proliferation to the release of mature sperm. Although human data are less experimentally controlled, the Seveso cohort studies point to similar effects on sperm quality and overall reproductive health [33, 88]. A consistent theme is that ligand‐independent AhR plays a permissive or positive role in normal germ cell development, whereas exogenous ligand‐activated AhR (particularly with high‐affinity toxicants like TCDD) often inhibits cell‐cycle progression, reduces sperm output, and alters sperm morphology. The precise outcomes can depend on factors such as ligand potency, timing of exposure, and species‐specific differences in AhR signaling [21, 74, 95, 96, 100, 129, 130].
Exploring AhR signaling in male reproductive health
Reproduction is a crucial biological event, and any signs of threatened reproductive function provoke significant responses in the scientific community and public media [131]. Despite receiving comparatively less attention, male reproductive function is a concern in the Western world, with infertility affecting approximately 15% of all couples [132]. Studies using AhR knockout mice have demonstrated that AhR deficiency is associated with defects in the seminiferous epithelium, the presence of multinucleated giant cells, hypocellularity, apical sloughing, and an increased count of retained elongated spermatids [119]. Furthermore, the modification of the AhR pathway, through the use of agonists or antagonists has led to changes in spermatozoa morphology and acrosome integrity via the regulation of the Dnah1 gene [90]. Table 1 summarizes some of AhR’s agonists and antagonists that interfere with reproductive system function of males.
Hormonal control
Spermatogenesis is dependent on the pituitary hormone, follicle-stimulating hormone (FSH), and locally produced androgens in response to the luteinizing hormone. These hormones not only nurture oocytes but also produce steroid hormones that ensure optimal functioning conditions in the female reproductive system [175, 176]. Most AhR/steroid receptor interactions have been studied in human breast cancer and endometrial carcinoma cell lines [176]. In one study, the activation of AhR by xenobiotics was shown to prompt the degradation of the androgen receptor [177]. Another study demonstrated low fertility in AhR KO mice along with degenerative alterations in the testes, germ cell apoptosis, and a decreased number of early spermatids [178]. Most recently, work has been published demonstrating that the abolishment of AhR signaling led to a decline in LH levels in rat serum [90].
AhR interaction with steroid hormone receptors
Steroid hormones play critical roles in the regulation of both human and animal fertility. In particular, male androgens, such as testosterone, function as primary inducers of the development of primary and secondary genital organs, in addition to libido potency [179]. The interplay between AhR and steroid hormone receptors is pivotal in regulating diverse physiological processes. Disruptions in steroid hormone synthesis, activity, or metabolism have been associated with various male reproductive issues. These include varicocele, erectile dysfunction, and infertility [180]. Such disturbances also impact female reproductive processes, affecting follicular dysfunction and atresia [180]. Previous in vitro experiments showed the competitive binding of AhR-ARNT complexes in HEC-1 A human endometrial carcinoma cells. This inhibits the binding of estrogen receptor alpha (ER-alpha) to imperfect estrogen response elements (EREs) [181, 182]. The activation of AhR can modulate the activity of steroid hormone receptors, especially the estrogen receptor, with potential substantial effects on endocrine function and overall health. This implies a crosstalk between AhR and estrogen receptor signaling pathways. Furthermore, certain environmental chemicals can bind directly to steroid hormone receptors, mimicking their function, and potentially causing adverse effects on both wildlife and humans [58, 183].
In vivo, recent studies have demonstrated that AhR-dependent mechanisms inhibit the growth of estrogen receptor-positive breast cancer cells in mouse xenografts [184]. The activation of AhR by Carbidopa, decarboxylase inhibitor, induces nuclear localization, leading to an increase in AhR transcriptional activity—effects that are nullified by an AhR blocker [185]. Studies on porcine follicular cells indicated that TCDD exposure led to a reduction in estrogen and progesterone synthesis through the use of AhR or ER blockers [186]. Dioxin has been observed to cause reproductive abnormalities including endometriosis, teratogenesis, abortion, decreased fertility, and endocrine disruption, particularly in luteal and follicular steroidogenesis in an AhR-dependent manner [187]. Additionally, prior studies suggested that AhR activity is crucial for cell proliferation. The AhR signaling pathway acts as a critical transcription factor in trophectoderm cells, influencing the cell cycle by modulating genes targeted by AhR and potential genes involved in trophectoderm cell proliferation [188]. The interaction mediated by AhR with steroid hormone receptors significantly affects the regulation of endocrine function. Research suggests that AhR can inhibit estrogen signaling by attaching to estrogen-responsive elements and accelerating estrogen receptor degradation [189]. This interaction can result in modified hormone synthesis, increased ligand metabolism, reduced receptor levels, and interaction between AhR and steroid hormone signaling pathways at the transcriptional level [190]. A recent study revealed that administering resveratrol, which is considered to be an AhR agonist, to male rats enhances the steroidogenesis of the testosterone hormone [90].
AhR and the regulation of the hypothalamic-pituitary–gonadal (HPG) axis
Studies have demonstrated a significant reduction in FSH and LH during the preovulatory period in female rats exposed to TCDD or related AhR ligands. This strongly suggests that the AhR may be partly responsible for the dysregulation of the hypothalamic-hypophyseal axis, which regulates spermatogenesis and androgen biosynthesis [191, 192]. Furthermore, the activation of AhR-induced suppression of gonadotropin surges has been attributed to a decreased responsiveness of the hypothalamus to the positive feedback from estrogens, without influencing preovulatory serum estrogen levels [191, 193]. This suggests reproductive toxicity linked to AhR activation [58, 90, 194].
Experiments with exogenous Gonadotropin-Releasing Hormone (GnRH) indicate that TCDD-induced inhibition of gonadotropin surges may be due to insufficient production and/or release of GnRH, implying an impact of TCDD on the central nervous system [195]. The observed inhibition of gonadotropin surges is linked to a decrease in hypothalamus responsiveness to estrogen feedback. This hypothesis is backed by the reversal of TCDD effects with higher estrogen concentrations [189]. The intricate relationship between the two is further demonstrated by the interaction between aryl hydrocarbon and estrogen-mediated signaling pathways, as evidenced by the partial estrogen antagonist, tamoxifen [196]. The expression of AhR signaling pathway members, especially in areas of the brain that control reproductive functions, emphasizes the complexity of AhR involvement. The coincidence of AhR gene expression with that of Glutamic Acid Decarboxylase 67, vital for Gamma-Aminobutyric Acid synthesis, suggests its potential role in regulating GABAergic neurons influencing the onset of puberty and gonadotropin surges [58, 69, 100].
Additionally, research shows that low doses of TCDD expedite puberty and HPG axis maturation in female rats—a compelling indication of complex interaction [197, 198]. The antagonism between AhR and estrogen receptor (ER) signaling pathways plays a substantial role in the estrogen-sensitive pituitary gland. This is evident in the interaction between AhR and ERα in both prolactin-secreting and gonadotropin-secreting cells [199]. TCDD displays a variety of effects, such as preventing E2-induced prolactin expression, encouraging LH-ß and ERα mRNA expression, and restraining FSH mRNA in the pituitary [196]. Intriguingly, TCDD reduces the release of prolactin, potentially due to increased dopamine secretion [200]. Developmental stage sensitivity to TCDD is clear, as it lessens fetal gonadotropin production without affecting adults. The influence of TCDD on GnRH release and the calcium’s role in mediating these effects amplify the intricate mechanisms involved [201]. Furthermore, TCDD changes the metabolism of the hypothalamus and pituitary gland, impacting lipoic acid content, ATP levels, and gonadotropin secretion [202]. Thyroid hormones, which the HPG axis regulates, are also influenced by TCDD, with conflicting findings observed on T4 and T3 levels. The TCDD-mediated induction of UDP-glucuronosyltransferase might contribute to decreased thyroid hormone levels [203].
Biological effects of AhR in male reproduction
The specific role of AhR in spermatogenesis has not been extensively studied, despite the well-established effects of AhR activation on cellular processes like the cell cycle, stem cell proliferation, and tissue differentiation [119]. It appears that AhR activity is crucial for cell proliferation and progression through the cell cycle [204, 205]. Conversely, some studies suggest that AhR signaling has anti-proliferative effects as its activation can induce cell cycle arrest at the G1/G0 phase [204]. AhR has been observed in the cells of the seminiferous epithelium, including both Sertoli cells and germ cells, as well as in Leydig cells of the interstitial tissue [206]. In vitro studies of cell cycle dynamics with ongoing TCDD exposure showed inhibited proliferation and G1-phase cell cycle arrest in various cell types, including hepatocytes [188], neuronal cells [207], thymocytes [188], and many kinds of cancer cells [105, 208] through activating AhR [209]. Broadly speaking, it should be noted that the effect of AhR on cell proliferation may vary based on the cell type and the specific phase of the cell cycle [36].
A substantial body of research suggests that the absence of AhR ligand binding or AhR activation may contribute to inflammation [210], apoptosis [119], and oxidative stress in sperm leading to DNA damage [211]. A study revealed that AhR expression in the rat seminiferous tubule is restricted to primary pachytene spermatocytes during stages VII–XI and round spermatids during stages II–XIV of the spermatogenic cycle [212, 213]. In contrast, both AhR and ARNT were found to be expressed in all stages of the seminiferous tubules in human testes [212]. The presence of AhR in sperm has suggested a mechanism by which environmental dioxins, polycyclic aromatic hydrocarbons, and polyhalogenated biphenyls could directly affect sperm function [214]. Mice deficient in AhR show decreased male fertility, sperm count, and weights of seminal vesicles and dorsolateral prostate [119]. Recent reports have also identified AhR immunoreactivity in the Sertoli cells of both rat and human testes [101]. Additionally, AhR signaling is needed to induce the expression of indoleamine 2,3-dioxygenase. Several studies have shown that this enzyme initiates the pathway of tryptophan catabolism [215, 216]. Furthermore, a deficiency in indoleamine 2,3-dioxygenase has been associated with significant increases in pro-inflammatory markers and the number and percentage of morphologically abnormal sperm [217, 218].
The role of AhR agonists in spermatogenesis
Effects of cigarette smoking and benzo-α-pyrene exposure
The decline in sperm counts and motility, along with abnormal sperm morphology, are among the negative consequences linked to cigarette smoking, which contains Benzo-α-pyrene [158]. This compound serves as an AhR agonist and can harm human health by causing DNA adduct formation and apoptosis in seminiferous tubules [219]. Furthermore, co-culture experiments involving human sperm and benzo-α-pyrene have demonstrated premature acrosome reactions and accelerated hyperactivation, both of which result in dysmorphic sperm and decreased sperm counts in exposed men [220]. Numerous studies have illustrated the destructive impact of Benzo-α-pyrene on sperm motility, suggesting alterations in mitochondrial function, the up-regulation of pro-apoptotic genes at the mitochondrial level, and DNA damage [221]. Similarly, wild-type mice exposed daily to Benzo-α-pyrene showed lower sperm counts and subfertility, and the repercussions are seen in subsequent generations [222]. Metabolically activated Benzo-α-pyrene leads to an increase in ROS generation, inducing oxidative stress, amplified lipid peroxidation, and the activation of caspases and endonucleases [159]. Moreover, the toxic effects of Benzo-α-pyrene are highlighted in in vitro and in vivo studies, depicting p53-mediated male germ cell apoptosis [223]. This process involves the activation of caspases 3, 6, 8, and 9, modification of Bcl-2, modulation of the Fas/FasL system, and activation of MAPKs (ERK 1/2, JNK 1/2, P38 MAPK) which contribute to p53 phosphorylation [158]. Interestingly, studies suggest a negative connection between p53 and AhR activity, implying a potential counteracting role of AhR against p53 activity [224]. Exposure to Benzo-α-pyrene involves AhR activation, nuclear translocation, DNA binding, and the subsequent decrease in the transcriptional activation of CYP1 A1 in various tissue systems [225]. Experimental studies have shown that curcumin and resveratrol, which function as AhR antagonists, can effectively shield against Benzo-α-pyrene-induced testicular germ cell apoptosis [226]. This protective effect corresponds to reduced protein and mRNA expression of CYP1 A1, diminished total AhR levels, and inhibited nuclear translocation of AhR [11, 158, 159, 227].
Activation of MAPK pathways and glucose uptake
Recent empirical evidence robustly supports the involvement of the JNK signaling pathway in maintaining blood-testes barrier function and facilitating germ cell migration. Studies have shown that the activation of the JNK signaling pathway mitigates the disruptive effects of CdCl2 on the blood-testes barrier in adult rats [228, 229]. The ERK signaling pathway plays a direct regulatory role in apoptosis, mitosis, and the progression of germ cell meiosis [230]. Furthermore, the meiosis of spermatocytes depends on the activation of the ERK signaling pathway, as demonstrated in co-culture experiments involving stem cells and pachytene spermatocytes [122].
Curcumin and resveratrol, which act through AhR, initiate the activation of ERK, p38 MAPK, and JNK pathways via the AhR pathway [158, 231,232,233,234]. The activation of the p38 MAPK signaling pathway correlates with an elevation in GLUT1 mRNA levels, promoting glucose uptake. Importantly, most genes associated with the MAPK pathway are present in immature rat stem cells. The p38 MAPK signaling pathway is pivotal in promoting JAM-B transcription in response to interleukin-1α stimulation in these cells. Additionally, both the p38 MAPK and ERK signaling pathways play a role in regulating cell junctions [235]. Resveratrol administration to male rats increases the percentages of normal sperm morphology and enhances acrosomal integrity as well as testicular parenchyma features through Dnah1 mRNA regulation [90].
Environmental impacts of AhR on the male reproductive system
Dioxin or TCDD, a well-established environmental contaminant, is associated with reproductive defects (such as endometriosis, teratogenesis, abortion, and diminished fertility) and endocrine disruption, affecting luteal and follicular steroidogenesis [236]. It is believed that the toxic effects of TCDD are primarily mediated through its ability to activate AhR [237]. Additionally, studies have proven that mice lacking a functioning AhR are resistant to dioxin toxicity [69]. Recent studies suggest a role for oxidative stress, induced by AhR-mediated production of reactive oxygen, in dioxin toxicity [157, 238]. Dioxin-induced production of mitochondrial reactive oxygen species in the testis and other tissues and organs has also been reported [239]. Moreover, dioxin can interfere with endocrine functions during development and adulthood, due to the AhR pathway’s crosstalk with several other signaling pathways, including the ER, retinoblastoma protein, hypoxia, NFκB, and TGF-β [240].
Diesel exhaust particles are known to increase serum concentrations of testosterone and the weight of the accessory glands in rats [241, 242]. These particles contain polycyclic aromatic hydrocarbons that activate AhR and decrease sperm production. Additionally, studies have demonstrated a decrease in the number of sperm and Sertoli cells in mature rats exposed to diesel exhaust particles [150]. The effects of diesel exhaust on spermatids in the testis and epididymis are dependent on AhR [121, 243].
A recent study found that long-term exposure of Zebra fish to an AhR agonist, known as nuburon, led to reproductive toxicity, which was apparent through a decrease in the number of sperm and an increase in oxidative stress levels due to the hyperactivity of the AhR pathway [3]. Moreover, exposing mice to chloro-choline chloride, a plant growth enhancer, could cause environmental concerns through an increased incidence of reproductive toxicity. This toxicity arises from the activation of the AhR/PERK axis, which subsequently leads to poor semen quality and degenerated testicular tissue [4]. However, some environmental pollutants, such as tris(2,3-dibromopropyl) iso-cyanurate, can trigger CYP19a1 toxicity directly via the estrogenic receptor in an AhR-independent mechanism, as revealed in an investigation run on a mouse-spermatogenic cell line [5].
Phthalate-induced male reproductive toxicity
Phthalates are used extensively to enhance the durability of plastics. Studies have shown that Di(2-ethylhexyl) phthalate (DEHP) can cause male reproductive toxicity [244]. This toxicity involves effects on spermatogenesis, disrupting self-renewal, meiosis, and spermatogonia activities [245]. Moreover, phthalates may inhibit testosterone synthesis by impacting the gonadal axis, thereby reducing the quality and quantity of sperm and leading to reproductive disorders [246]. A previous study suggested that phthalates caused DNA damage and apoptosis in sperm, affecting sperm density, vitality, and progressive motility. This decrease in sperm quality, including DNA damage and impaired motility, has been linked to testicular spermatogenesis disorders [247, 248]. Additionally, phthalates increase the expression of CYP1 A1, suggesting AhR pathway activation, which has a variety of effects on different cell functions [249, 250]. Remarkably, phthalate treatment seems to promote the nuclear accumulation of AhR and ARNT, both localizing to the spermatogenic cell nucleus. Interestingly, as the downstream gene targets of the AhR/ARNT signaling system, the levels of CYP1 A1, CYP1 A2, and CYP1B1 were significantly increased following phthalate exposure [165]. Phthalates also seem to induce impairment of blood-testis barrier integrity, which is crucial for normal spermatogenesis [251].
Resveratrol as an AhR antagonist
Resveratrol is a phytochemical present in peanuts, grapes, blueberries, rhubarb, and wine, possessing cytoprotective and antioxidant properties. It functions as an antagonist of the AhR, with one of its mechanisms involving the inhibition of AhR expression [160, 252]. Moreover, resveratrol obstructs the activation of CYP1 A1 and CYP1B1, and this action is associated with a decrease in ROS production [253]. The protective effect of resveratrol on ROS generation is highly significant because peroxidation of polyunsaturated fatty acids can result in lowered membrane fluidity and reduced activity of membrane enzymes and ion channels, potentially endangering sperm motility [254]. Researchers have detailed the mechanism behind resveratrol’s actions on AhR, emphasizing its capability to restore the reduction in ERK and p38 MAPK phosphorylation prompted by AhR’s ligand agonists [158, 231]. Resveratrol defends cells from DNA damage and apoptosis by moderating the anti- and pro-apoptotic mediators, thus enhancing the antioxidant status [222]. Resveratrol inhibits the enzymatic activity of various cytochrome P450 s and deters their transcription through the antagonism of AhR, implying that resveratrol might lessen cells’ cancer-causing exposure, including TCDD [159, 222].
Lycopene supplementation alleviates male infertility
Lycopene is a carotenoid antioxidant located in plants, such as tomatoes. Lycopene supplementation studies in humans and animals have demonstrated potential in alleviating male infertility where the sperm count, and viability increased with Lycopene treatment [255]. Furthermore, Lycopene can abate testicular toxicity, thus offsetting the harmful effects of pollution by managing the CYP450 s homeostasis and the AhR/ARNT signaling system [245]. Studies have shown that administering Lycopene to rats mitigated nearly all testicular structural damage, which included desquamative germinal cells and the slowing of spermatogenesis [251, 256, 257]. Current evidence suggests that lycopene functions as an antagonist (or at least an inhibitor) of AhR. Additionally, Lycopene precipitated a substantial decrease in the nuclear accumulation of AhR and ARNT, while their downstream target genes, inhibitors of CYP1 enzymes (CYP1 A1, CYP1 A2, and CYP1B1) were significantly reduced to normal levels. Concurrently, the values of sperm motility, number, and density exhibited an increase [56, 258].
Curcumin supplementation alleviates male infertility
Curcumin is a naturally occurring plant polyphenol found in the ancient Indian spice Turmeric, boasting various beneficial properties. It is known for its anti-oxidative and anti-inflammatory activity [168, 259]. It has been reported that curcumin acts as both an agonist and antagonist for the AhR depending on the context of exposure [81, 259]. Without strong ligands like dioxin, curcumin mildly stimulate AhR, effectively behaving as a weak agonist by upregulating certain AhR-responsive genes [81]. Conversely, when potent ligands are present or when the cellular environment includes heightened AhR sensitivity, curcumin frequently serves as an antagonist by blocking ligand binding, promoting AhR degradation, or altering co-factor interactions in a way that diminishes AhR-driven gene expression [169, 259]. A few studies indicate that curcumin can prevent testicular germ cell apoptosis under various stressful conditions [260]. Curcumin enhances resveratrol’s effectiveness, and together, they regulate p53 phosphorylation specifically at ser 15 involving MAPKs [159]. Curcumin can also mitigate oxidative stress [261] and inflammation, which are two major contributors to male infertility [260].
The affinity of selected AhR ligands
Table 2 provides an overview of the relative binding affinity of various AhR ligands, indicating whether each ligand exhibits high or low affinity. This summary, based on findings from several studies, highlights that potent agonists such as TCDD and FICZ bind with high affinity. In contrast, other compounds—like benzo[a]pyrene, DEHP, and several dietary modulators (resveratrol and curcumin)—generally display lower affinity.
Epigenetic modifications and transgenerational effects
Epigenetic landscape changes induced by AhR activation
Transgenerational inheritance refers to the transfer of traits or characteristics from one generation to the next, not through alterations in DNA sequences, but rather via modifications of the epigenome. Environmental factors can trigger these modifications and potentially influence the phenotypes of the progeny. Several studies suggest that environmental stimuli can modify parental traits, thus influencing the phenotypes of offspring via gametic epigenetic inheritance. As a result, the role of epigenetic factors and their heritability merit careful consideration in the context of disease risk assessment [266, 267]. The AhR plays a significant role in influencing the epigenetic landscape, particularly regarding transgenerational effects resulting from chemical exposures. Research indicates that AhR activation alters DNA methylation patterns and gene expression, which can affect phenotypic outcomes across generations [268]. In zebrafish exposed to AhR agonists such as benzo-α-pyrene and 7,12-dimethylbenz(a)anthracene, changes in DNA methylation and gene expression have been discovered, suggesting a potential role of AhR-mediated epigenetic modifications in transgenerational effects [269]. Hence, the observed phenotypic alterations in AhR lineage zebrafish raise questions about the contribution of epigenetic changes in the AhR-ARNT signaling regulation to reproductive and skeletal phenotypes. Understanding the persistence and reversibility of these effects is vital for a comprehensive assessment of the impact of AhR activation on transgenerational outcomes [270,271,272]. Furthermore, it has been demonstrated that AhR activation can modulate DNA methylation patterns in specific genomic regions, impacting the expression of genes involved in various cellular processes [273]. Additionally, histone modifications, such as acetylation and methylation, may be influenced by AhR signaling, further contributing to the regulation of gene expression [273].
Transgenerational inheritance of AhR-induced reproductive alterations
Some studies suggest that environmental cues can induce parental changes and affect the phenotypes of offspring through gametic epigenetic inheritance. As a result, epigenetic factors and their heritability should be considered during disease risk assessment. One intriguing aspect of AhR signaling is its potential to induce transgenerational effects, which refer to the transmission of traits or alterations in phenotype across generations, without direct exposure to the environmental stimuli. Research suggests that AhR activation could lead to reproductive changes that can be passed on to following generations [269, 270, 274, 275].
AhR-mediated epigenetic dysregulation and male fertility
Several factors seem to contribute to the outcome of gene transcriptional regulation by AhR, such as the nature of the ligand and its further metabolism by AhR-induced enzymes, the local tissue microenvironment, and the presence of co-regulators or specific transcription factors in cells. Studies have suggested that AhR activations may lead to epigenetic dysregulation in the male reproductive system, impacting the development and function of sperm [55, 267, 268, 276]. Epigenetic modifications driven by AhR signaling in the male germline can affect the expression of genes involved in spermatogenesis, sperm motility, and fertilization. Understanding these molecular shifts is crucial for discerning the connections between environmental exposures, AhR activation, and male fertility outcomes [268, 277, 278].
In summary, the AhR significantly impacts spermatogenesis, hormonal regulation, and reproductive function, asserting its importance as a regulator of male reproductive health. It underlines its role in male fertility by directing spermiogenesis and ensuring the completion of spermiation. AhR’s influence on both reproductive and endocrine processes is demonstrated through its interaction with steroid hormone receptors, particularly estrogen receptors. Male reproductive function can be compromised by environmental chemicals or factors such as phthalates, benzo-a-pyrene, dioxins, and cigarette smoke, through AhR activation. This may subsequently lead to sperm abnormalities and infertility. The complex relationship between AhR and the hypothalamic-pituitary–gonadal axis emphasizes its regulatory role in hormonal and reproductive processes. AhR activation, which can influence reproductive outcomes, may also affect estrogen signaling and modulate gonadotropin surges. Furthermore, AhR-mediated epigenetic modifications may affect gene expression in the male germline, potentially impacting sperm development and fertility.
Most studies examine the effects of xenobiotic AhR ligands such as TCDD are based on animal or in vitro studies with cell lines. There are few epidemiological studies involving people accidentally exposed to xenobiotics. Therefore, more human studies are crucial to verify the effects of environmental AhR ligands on the male reproductive system. Currently, there are no effective treatments for reversing the toxic effects of environmental contaminants like dioxins on the male reproductive system.
Interestingly, dietary AhR ligands or antagonists, including lycopene, resveratrol, and curcumin, may offer some protection against male reproductive toxicity caused by environmental pollutants. Their potential ability to reduce oxidative stress and AhR activation suggests a possible treatment path for enhancing male reproductive health (Graphical Abstract).
Data availability
No datasets were generated or analysed during the current study.
References
Rothhammer V, Quintana FJ. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol. 2019;19:184–97.
Sondermann NC, Faßbender S, Hartung F, Hätälä AM, Rolfes KM, Vogel CFA, et al. Functions of the aryl hydrocarbon receptor (AHR) beyond the canonical AHR/ARNT signaling pathway. Biochem Pharmacol. 2022;208:115371.
Qian C, Yang C, Lu M, Bao J, Shen H, Deng B, et al. Activating AhR alleviates cognitive deficits of Alzheimer’s disease model mice by upregulating endogenous Aβ catabolic enzyme Neprilysin. Theranostics. 2021;11:8797.
Rothhammer V, Mascanfroni ID, Bunse L, Takenaka MC, Kenison JE, Mayo L, et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med. 2016;22:586–97.
Alghetaa H, Mohammed A, Singh N, Wilson K, Cai G, Putluri N, et al. Resveratrol attenuates staphylococcal enterotoxin B-activated immune cell metabolism via upregulation of miR-100 and suppression of mTOR signaling pathway. Front Pharmacol. 2023;14:1106733.
Abdulla OA, Neamah W, Sultan M, Alghetaa HK, Singh N, Busbee PB, et al. The ability of AhR ligands to attenuate delayed type hypersensitivity reaction is associated with alterations in the gut microbiota. Front Immunol. 2021;12:684727.
Li Y, Wang X, Su Y, Wang Q, Huang S, Pan Z, et al. Baicalein ameliorates ulcerative colitis by improving intestinal epithelial barrier via AhR/IL-22 pathway in ILC3s. Acta Pharmacol Sin. 2022;43:1495–507.
Wiggins BG, Wang Y-F, Burke A, Grunberg N, Vlachaki Walker JM, Dore M, et al. Endothelial sensing of AHR ligands regulates intestinal homeostasis. Nature. 2023;621:821–9.
Dawood RA, Alghetaa H. Deleterious effects of mercuric chloride toxicity initiated partially from physiological disorder of kidney, immunocytes and redox system, can be reversed by resveratrol administration. Adv Anim Vet Sci. 2023;11(9):1471.
Cannon AS, Holloman BL, Wilson K, Miranda K, Dopkins N, Nagarkatti P, et al. Ahr activation leads to attenuation of murine autoimmune hepatitis: single-cell rna-seq analysis reveals unique immune cell phenotypes and gene expression changes in the liver. Front Immunol. 2022;13:899609.
Aghetaa HFK, Dawood RA, Aladhami AK. Resveratrol administration ameliorates hepatotoxicity in mercuric chloride- induced liver injury in rats. Iraqi J of Vet Med. 2023;47:1–8.
Alghetaa H, Mohammed A, Zhou J, Singh N, Nagarkatti M, Nagarkatti P. Resveratrol-mediated attenuation of superantigen-driven acute respiratory distress syndrome is mediated by microbiota in the lungs and gut. Pharmacol Res. 2021;167:105548.
Alharris E, Mohammed A, Alghetaa H, Zhou J, Nagarkatti M, Nagarkatti P. The ability of resveratrol to attenuate ovalbumin-mediated allergic asthma is associated with changes in microbiota involving the gut-lung axis, enhanced barrier function and decreased inflammation in the lungs. Front Immunol. 2022;13:805770.
Chen C, Sun S, Zhao J, Wu Q, He W, Sun W. Yishen-Qingli-Huoxue formula attenuates renal fibrosis by inhibiting indoxyl sulfate via AhR/snai1 signaling. Phytomedicine. 2023;108:154546.
Itkin B, Breen A, Turyanska L, Sandes EO, Bradshaw TD, Loaiza-Perez AI. New treatments in renal cancer: the AhR ligands. Int J Mol Sci. 2020;21:3551.
Carlucci CD, Hui Y, Chumanevich AP, Robida PA, Fuseler JW, Sajish M, et al. Resveratrol protects against skin inflammation through inhibition of mast cell, sphingosine Kinase-1, Stat3 and NF-κB p65 signaling activation in mice. Int J Mol Sci. 2023;24:6707.
Alabsawy S, Alghetaa HF. Resveratrol Administration Reverses The Osteoporotic Bone In Independent-Manner Of Ahr-Esr1 Axis In Rats. J Neonatal Surg. 2025;14:1143. https://doiorg.publicaciones.saludcastillayleon.es/10.63682/jns.v14i15S.3722.
Kojima H, Takeuchi S, Nagai T. Endocrine-disrupting potential of pesticides via nuclear receptors and aryl hydrocarbon receptor. J Health Sci. 2010;56:374–86.
Memari B, Nguyen-Yamamoto L, Salehi-Tabar R, Zago M, Fritz JH, Baglole CJ, et al. Endocrine aryl hydrocarbon receptor signaling is induced by moderate cutaneous exposure to ultraviolet light. Sci Rep. 2019;9:8486.
Yoshizawa K, Marsh T, Foley JF, Cai B, Peddada S, Walker NJ, et al. Mechanisms of exocrine pancreatic toxicity induced by oral treatment with 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin in female Harlan Sprague-Dawley Rats. Toxicol Sci. 2005;85:594–606.
Gonzalez FJ, Fernandez-Salguero P. The aryl hydrocarbon receptor: studies using the AHR-null mice. Drug Metab Dispos. 1998;26:1194–8.
Seok S-H, Lee W, Jiang L, Molugu K, Zheng A, Li Y, et al. Structural hierarchy controlling dimerization and target DNA recognition in the AHR transcriptional complex. Proc Natl Acad Sci. 2017;114:5431–6.
Otte JC, Schmidt AD, Hollert H, Braunbeck T. Spatio-temporal development of CYP1 activity in early life-stages of zebrafish (Danio rerio). Aquat Toxicol. 2010;100:38–50.
Karman BN, Hernández-Ochoa I, Ziv-Gal A, Flaws JA. Involvement of the AHR in development and functioning of the female and male reproductive systems. The AH Receptor in Biology and Toxicology Hoboken, NJ: Wiley. 2012;437–66.
Prasad Singh N, Nagarkatti M, Nagarkatti P. From suppressor T cells to regulatory T cells: how the journey that began with the discovery of the toxic effects of TCDD led to better understanding of the role of AhR in immunoregulation. Int J Mol Sci. 2020;21:7849.
Busbee PB, Rouse M, Nagarkatti M, Nagarkatti PS. Use of natural AhR ligands as potential therapeutic modalities against inflammatory disorders. Nutr Rev. 2013;71:353–69.
Dolwick KM, Schmidt JV, Carver LA, Swanson HI, Bradfield CA. Cloning and expression of a human Ah receptor cDNA. Mol Pharmacol. 1993;44:911–7.
Esser C, Rannug A, Stockinger B. The aryl hydrocarbon receptor in immunity. Trends Immunol. 2009;30:447–54.
Neamah WH, Busbee PB, Alghetaa H, Abdulla OA, Nagarkatti M, Nagarkatti P. Ahr activation leads to alterations in the gut microbiome with consequent effect on induction of myeloid derived suppressor cells in a CXCR2-dependent manner. Int J Mol Sci. 2020;21:9613.
Alghetaa H, Mohammed A, Zumbrun E, Elliot D, Nagarkatti P, Nagarkatti M. Epigenetic-immunological axis of resveratrol-mediated amelioration of acute lung injury. Eur J Immunol. 2016;46:391 WIley-Blackwell 111 River St, Hoboken 07030–5774, NJ USA.
Al-Kinany, Qammar Shaker Hmood Enayah SH. The effect of polycyclic aromatic hydrocarbons on AHR and Cyp450 enzymes in oil field workers in south of Iraq. Iran J Ichthyol. 2021;8:359–63.
Esakky P, Hansen DA, Drury AM, Moley KH. Cigarette smoke condensate induces aryl hydrocarbon receptor-dependent changes in gene expression in spermatocytes. Reprod Toxicol. 2012;34:665–76.
Ye M, Warner M, Mocarelli P, Brambilla P, Eskenazi B. Prenatal exposure to TCDD and atopic conditions in the Seveso second generation: a prospective cohort study. Environ Health. 2018;17:1–9.
Mocarelli P, Brambilla P, Gerthoux PM, Patterson DG Jr, Needham LL. Change in sex ratio with exposure to dioxin. The Lancet. 1996;348:409.
Mocarelli P, Gerthoux PM, Patterson DG Jr, Milani S, Limonta G, Bertona M, et al. Dioxin exposure, from infancy through puberty, produces endocrine disruption and affects human semen quality. Environ Health Perspect. 2008;116:70–7.
Larigot L, Juricek L, Dairou J, Coumoul X. AhR signaling pathways and regulatory functions. Biochim Open. 2018;7:1–9.
Shivanna B, Chu C, Moorthy B. The Aryl Hydrocarbon Receptor (AHR): a novel therapeutic target for pulmonary diseases? Int J Mol Sci. 2022;23(3):1516.
Neamah WH, Singh NP, Alghetaa H, Abdulla OA, Chatterjee S, Busbee PB, et al. AhR activation leads to massive mobilization of myeloid-derived suppressor cells with immunosuppressive activity through regulation of CXCR2 and microRNA miR-150-5p and miR-543-3p that target anti-inflammatory genes. J Immunol. 2019;203:1830–44.
Dai S, Qu L, Li J, Zhang Y, Jiang L, Wei H, et al. Structural insight into the ligand binding mechanism of aryl hydrocarbon receptor. Nat Commun. 2022;13(1):6234.
Haidar R, Henkler F, Kugler J, Rosin A, Genkinger D, Laux P, et al. The role of DNA-binding and ARNT dimerization on the nucleo-cytoplasmic translocation of the aryl hydrocarbon receptor. Sci Rep. 2021;11:18194.
Gargaro M, Scalisi G, Manni G, Mondanelli G, Grohmann U, Fallarino F. The landscape of AhR regulators and coregulators to fine-tune AhR functions. Int J Mol Sci. 2021;22(2):757.
Xu D, Farmer A, Collett G, Grishin NV, Chook YM. Sequence and structural analyses of nuclear export signals in the NESdb database. Mol Biol Cell. 2012;23:3677–93.
Huang TT, Kudo N, Yoshida M, Miyamoto S. A nuclear export signal in the N-terminal regulatory domain of IB controls cytoplasmic localization of inactive NF-BIB complexes. Proc Natl Acad Sci U S A. 2000;97(3):1014–9. Available from: https://www.pnas.org.
BorkГєti P, Bajusz I, Bajusz C, KristГi I, KovГЎcs Z, Vilmos P. Testing the biological significance of the nuclear localization of actin. Biopolym Cell. 2019;35:204.
Ramos KS, Nanez A. Introduction and overview of receptor systems. 2010.
Barton CE, Pietenpol JA. p53 Family. 2013.
Chang YM, Chen CKM, Hou MH. Conformational changes in DNA upon ligand binding monitored by circular dichroism. Int J Mol Sci. 2012;13:3394–413.
Kudo I, Hosaka M, Haga A, Tsuji N, Nagata Y, Okada H, et al. The regulation mechanisms of AhR by molecular chaperone complex. The journal of biochemistry. 2018;163:223–32.
Szelest M, Walczak K, Plech T. A new insight into the potential role of tryptophan-derived AhR ligands in skin physiological and pathological processes. Int J Mol Sci. 2021;22:1104.
Cain JW, Miljic D, Popovic V, Korbonits M. Role of the aryl hydrocarbon receptor-interacting protein in familial isolated pituitary adenoma. Expert Rev Endocrinol Metab. 2010;5(5):681–95.
Vogel CFA, Ishihara Y, Campbell CE, Kado SY, Nguyen-Chi A, Sweeney C, et al. A protective role of aryl hydrocarbon receptor repressor in inflammation and tumor growth. Cancers (Basel). 2019;11:589.
Vogel CFA, Haarmann-Stemmann T. The aryl hydrocarbon receptor repressor–more than a simple feedback inhibitor of AhR signaling: clues for its role in inflammation and cancer. Curr Opin Toxicol. 2017;2:109–19.
Yang SY, Ahmed S, Satheesh SV, Matthews J. Genome-wide mapping and analysis of aryl hydrocarbon receptor (AHR)-and aryl hydrocarbon receptor repressor (AHRR)-binding sites in human breast cancer cells. Arch Toxicol. 2018;92:225–40.
Lee KWK, Richmond R, Hu P, French L, Shin J, Bourdon C, et al. Prenatal exposure to maternal cigarette smoking and DNA methylation: epigenome-wide association in a discovery sample of adolescents and replication in an independent cohort at birth through 17 years of age. Environ Health Perspect. 2015;123:193–9.
Beedanagari SR, Taylor RT, Bui P, Wang F, Nickerson DW, Hankinson O. Role of epigenetic mechanisms in differential regulation of the dioxin-inducible human CYP1A1 and CYP1B1 genes. Mol Pharmacol. 2010;78:608–16.
Kim YJ, Lim Y, Lee E. CARM1 is involved in CYP1A1 gene expression as a transcriptional coactivator. Mol Cell Toxicol. 2017;13:263–70.
Wolff M, Jelkmann W, Dunst J, Depping R. The aryl hydrocarbon receptor nuclear translocator (ARNT/HIF-1β) is influenced by hypoxia and hypoxia-mimetics. Cell Physiol Biochem. 2013;32:849–58.
Motta S, Bonati L. TCDD-induced allosteric perturbation of the AhR: ARNT binding to DNA. Int J Mol Sci. 2023;24:9339.
Sun X, Jing L, Li F, Zhang M, Diao X, Zhuang J, et al. Structures of NPAS4-ARNT and NPAS4-ARNT2 heterodimers reveal new dimerization modalities in the bHLH-PAS transcription factor family. Proc Natl Acad Sci. 2022;119:e2208804119.
Mandl M, Depping R. Hypoxia-inducible aryl hydrocarbon receptor nuclear translocator (ARNT)(HIF-1β): is it a rare exception? Mol Med. 2014;20:215–20.
Mulero-Navarro S, Fernandez-Salguero PM. New trends in aryl hydrocarbon receptor biology. Front Cell Dev Biol. 2016;4:45.
Heo MJ, Suh JH, Lee SH, Poulsen KL, An YA, Moorthy B, et al. Aryl hydrocarbon receptor maintains hepatic mitochondrial homeostasis in mice. Mol Metab. 2023;72:101717.
Csanaky IL, Lickteig AJ, Zhang Y, Klaassen CD. Effects of patent ductus venosus on bile acid homeostasis in aryl hydrocarbon receptor (AhR)-null mice. Toxicol Appl Pharmacol. 2020;403:115136.
Eti NA, Flor S, Iqbal K, Scott RL, Klenov VE, Gibson-Corley KN, et al. PCB126 induced toxic actions on liver energy metabolism is mediated by AhR in rats. Toxicology. 2022;466:153054.
Shi Y, Zeng Z, Yu J, Tang B, Tang R, Xiao R. The aryl hydrocarbon receptor: An environmental effector in the pathogenesis of fibrosis. Pharmacol Res. 2020;160:105180.
da Silva JF, Bolsoni JA, da Costa RM, Alves JV, Bressan AFM, LE Silva V, et al. Aryl hydrocarbon receptor (AhR) activation contributes to high-fat diet-induced vascular dysfunction. Br J Pharmacol. 2022;179:2938–52.
Coelho NR, Pimpão AB, Correia MJ, Rodrigues TC, Monteiro EC, Morello J, et al. Pharmacological blockage of the AHR-CYP1A1 axis: A call for in vivo evidence. J Mol Med. 2022;100:215–43.
Brinkmann V, Schiavi A, Shaik A, Puchta DR, Ventura N. Dietary and environmental factors have opposite AhR-dependent effects on C. elegans healthspan. Aging (Albany NY). 2021;13:104.
Bock KW. From TCDD-mediated toxicity to searches of physiologic AHR functions. Biochem Pharmacol. 2018;155:419–24.
Miao H, Wang Y, Yu X, Zou L, Guo Y, Su W, et al. Lactobacillus species ameliorate membranous nephropathy through inhibiting the aryl hydrocarbon receptor pathway via tryptophan-produced indole metabolites. Br J Pharmacol. 2024;181:162–79.
Arellano-Gutiérrez CV, Quintas-Granados LI, Cortés H, González del Carmen M, Leyva-Gómez G, Bustamante-Montes LP, et al. Indole-3-Carbinol, a phytochemical aryl hydrocarbon receptor-ligand, induces the mRNA overexpression of UBE2L3 and cell proliferation arrest. Curr Issues Mol Biol. 2022;44:2054–68.
Saxena A, Mitchell C, Bogdon R, Roark K, Wilson K, Staley S, et al. Aryl hydrocarbon receptor regulates muc2 production independently of IL-22 during colitis. Int J Mol Sci. 2024;25:2404.
Kamenickova A, Anzenbacherova E, Pavek P, Soshilov AA, Denison MS, Zapletalova M, et al. Effects of anthocyanins on the AhR–CYP1A1 signaling pathway in human hepatocytes and human cancer cell lines. Toxicol Lett. 2013;221:1–8.
Bailey-Downs LC, Mitschelen M, Sosnowska D, Toth P, Pinto JT, Ballabh P, et al. Liver-specific knockdown of IGF-1 decreases vascular oxidative stress resistance by impairing the Nrf2-dependent antioxidant response: a novel model of vascular aging. J Gerontol A Biol Sci Med Sci. 2012;67:313–29.
Kolluri SK, Weiss C, Koff A, Göttlicher M. p27Kip1 induction and inhibition of proliferation by the intracellular Ah receptor in developing thymus and hepatoma cells. Genes Dev. 1999;13:1742–53.
Kahalehili HM, Newman NK, Pennington JM, Kolluri SK, Kerkvliet NI, Shulzhenko N, et al. Dietary indole-3-carbinol activates AhR in the gut, alters Th17-microbe interactions, and exacerbates insulitis in NOD mice. Front Immunol. 2021;11:606441.
Pevarello P, Brasca MG, Amici R, Orsini P, Traquandi G, Corti L, et al. 3-Aminopyrazole inhibitors of CDK2/cyclin A as antitumor agents. 1. Lead finding. J Med Chem. 2004;47:3367–80.
Vondráček J, Machala M, Bryja V, Chramostová K, Krčmář P, Dietrich C, et al. Aryl hydrocarbon receptor-activating polychlorinated biphenyls and their hydroxylated metabolites induce cell proliferation in contact-inhibited rat liver epithelial cells. Toxicol Sci. 2005;83:53–63.
Andrysík Z, Procházková J, Kabátková M, Umannová L, Šimečková P, Kohoutek J, et al. Aryl hydrocarbon receptor-mediated disruption of contact inhibition is associated with connexin43 downregulation and inhibition of gap junctional intercellular communication. Arch Toxicol. 2013;87:491–503.
Faust D, Vondráček J, Krčmář P, Šmerdová L, Procházková J, Hrubá E, et al. AhR-mediated changes in global gene expression in rat liver progenitor cells. Arch Toxicol. 2013;87:681–98.
Brinkmann V, Romeo M, Larigot L, Hemmers A, Tschage L, Kleinjohann J, et al. Aryl hydrocarbon receptor-dependent and-independent pathways mediate curcumin anti-aging effects. Antioxidants. 2022;11:613.
Al Zoubi MS, Al Khateeb W, El-Oqlah M, Migdady M, Abu Al-Arja MI, Bzour M, et al. Anti-proliferative, Anti-angiogenic and Anti-inflammatory Effects of Moringa peregrina Leaf Extracts on Testosterone-Induced Benign Prostatic Hyperplasia in Rats. Asian Pac J Cancer Prev. 2022;23:161–9.
Al-Khaqani B, Mohammed A. Ovalbumin-induced asthma in rats is alleviated by resveratrol treatment. J Anim Health Prod. 2024;12:121–7.
Ahmed RM, Mohammed AK. Amelioration of hepatotoxicity by sodium butyrate administration in rats. World’s Vet J. 2022;12(3):323–9.
Ahmed RM, Mohammed AK. Role of sodium butyrate supplement on reducing hepatotoxicity induced by lead acetate in rats. Iraqi J Vet Med. 2022;46:29–35.
Smith KJ, Murray IA, Tanos R, Tellew J, Boitano AE, Bisson WH, et al. Identification of a high-affinity ligand that exhibits complete aryl hydrocarbon receptor antagonism. J Pharmacol Exp Ther. 2011;338:318–27.
Howard GJ, Schlezinger JJ, Hahn ME, Webster TF. Generalized concentration addition predicts joint effects of aryl hydrocarbon receptor agonists with partial agonists and competitive antagonists. Environ Health Perspect. 2010;118:666–72.
Kerger BD, Gerthoux PM, Mocarelli P. 1976 Trichlorophenol Reactor Explosion at Seveso, Italy. 2019;
Bircsak KM, Copes LT, King S, Prantner AM, Hwang W-T, Gerton GL. The aryl hydrocarbon receptor mediates sex ratio distortion in the embryos sired by TCDD-exposed male mice. Reprod Toxicol. 2020;94:75–83.
Bustani G, Al-Humadi N, Alghetaa H. Aryl hydrocarbon receptor modulation and resveratrol influence on Dnah1 expression: implications for male reproductive health. Iraqi J Vet Med. 2024;48:15–24.
Bunger MK, Moran SM, Glover E, Thomae TL, Lahvis GP, Lin BC, et al. Resistance to 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin toxicity and abnormal liver development in mice carrying a mutation in the nuclear localization sequence of the aryl hydrocarbon receptor. J Biol Chem. 2003;278:17767–74.
Hernández-Ochoa I, Karman BN, Flaws JA. The role of the aryl hydrocarbon receptor in the female reproductive system. Biochem Pharmacol. 2009;77(4):547–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bcp.2008.09.037.
Hilbold E, Distl O, Hoedemaker M. Loss of Cx43 in murine sertoli cells leads to altered prepubertal sertoli cell maturation. Cells. 2020;9(3):676.
Rode K, Langeheine M, Seeger B, Brehm R. Connexin43 in germ cells seems to be dispensable for murine spermatogenesis. Int J Mol Sci. 2021;22:7924.
Elizondo G, Fernandez-Salguero P, Sheikh MS, Kim G-Y, Fornace AJ, Lee KS, et al. Altered cell cycle control at the G2/M phases in aryl hydrocarbon receptor-null embryo fibroblast. Mol Pharmacol. 2000;57:1056–63.
Marlowe JL, Knudsen ES, Schwemberger S, Puga A. The aryl hydrocarbon receptor displaces p300 from E2F-dependent promoters and represses S phase-specific gene expression. J Biol Chem. 2004;279:29013–22.
Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 1999;13:1501–12.
Sahebnasagh A, Hashemi J, Khoshi A, Saghafi F, Avan R, Faramarzi F, et al. Aromatic hydrocarbon receptors in mitochondrial biogenesis and function. Mitochondrion. 2021;61:85–101.
Limas JC, Cook JG. Preparation for DNA replication: the key to a successful S phase. FEBS Lett. 2019;593:2853–67.
Cho TE, Bott D, Ahmed S, Hutin D, Gomez A, Tamblyn L, et al. 3-methylcholanthrene induces chylous ascites in TCDD-inducible poly-ADP-ribose polymerase (TIPARP) knockout mice. Int J Mol Sci. 2019;20:2312.
Ribeiro MA, Estill MS, Fernandez GJ, Moraes LN, Krawetz SA, Scarano WR. Integrative transcriptome and microRNome analysis identifies dysregulated pathways in human Sertoli cells exposed to TCDD. Toxicology. 2018;409:112–8.
Shan Z, Liu L, Shen J, Hao H, Zhang H, Lei L, et al. Enhanced UV resistance role of death domain-associated protein in human MDA-MB-231 breast cancer cells by regulation of G2 DNA damage checkpoint. Cell Transplant. 2020;29:0963689720920277.
Franchini AM, Vaughan KL, Bhattacharya S, Singh KP, Gasiewicz TA, Lawrence BP. Impact of the aryl hydrocarbon receptor on Aurora A kinase and the G2/M phase pathway in hematopoietic stem and progenitor cells. Receptors. 2023;2:100–15.
Soncini C, Carpinelli P, Gianellini L, Fancelli D, Vianello P, Rusconi L, et al. PHA-680632, a novel Aurora kinase inhibitor with potent antitumoral activity. Clin Cancer Res. 2006;12:4080–9.
Barhoover MA, Hall JM, Greenlee WF, Thomas RS. Aryl hydrocarbon receptor regulates cell cycle progression in human breast cancer cells via a functional interaction with cyclin-dependent kinase 4. Mol Pharmacol. 2010;77:195–201.
Zhang J, Gao X, Yu L. Roles of histone deacetylases in acute myeloid leukemia with fusion proteins. Front Oncol. 2021;11:741746.
Bustani GS, Alghetaa HF. Aryl Hydrocarbon Receptor Modulation Impacts on the Blood-Testis Barrier Integrity via TJP1 function. Iran J Vet Med. 2025;19(3):3.
Wang L-T, Chiou S-S, Chai C-Y, Hsi E, Wang S-N, Huang S-K, et al. Aryl hydrocarbon receptor regulates histone deacetylase 8 expression to repress tumor suppressive activity in hepatocellular carcinoma. Oncotarget. 2017;8:7489.
Garrison PM, Rogers JM, Brackney WR, Denison MS. Effects of histone deacetylase inhibitors on the Ah receptor gene promoter. Arch Biochem Biophys. 2000;374:161–71.
Tsai C-H, Li C-H, Liao P-L, Chang Y-W, Cheng Y-W, Kang J-J. Aza-PBHA, a potent histone deacetylase inhibitor, inhibits human gastric-cancer cell migration via PKCα-mediated AHR-HDAC interactions. Biochim Biophys Acta Mol Cell Res. 2020;1867:118564.
Rody A, Karn T, Gätje R, Ahr A, Solbach C, Kourtis K, et al. Gene expression profiling of breast cancer patients treated with docetaxel, doxorubicin, and cyclophosphamide within the GEPARTRIO trial: HER-2, but not topoisomerase II alpha and microtubule-associated protein tau, is highly predictive of tumor response. The Breast. 2007;16:86–93.
Oikawa K, Ohbayashi T, Mimura J, Iwata R, Kameta A, Evine K, et al. Dioxin suppresses the checkpoint protein, MAD2, by an aryl hydrocarbon receptor-independent pathway. Cancer Res. 2001;61:5707–9.
Kim JE, Kim HR, Kang SY, Jung MJ, Heo NH, Lee HJ, et al. Aryl hydrocarbon Receptor and autophagy-related protein microtubule-associated protein light chain 3 expression in psoriasis. Ann Dermatol. 2021;33:138.
Zhang X, Xiao T, Cheng S, Tong T, Gao Y. Cigarette smoke suppresses the ubiquitin-dependent degradation of OLC1. Biochem Biophys Res Commun. 2011;407:753–7.
Yamashita N, Yoshizuka A, Kase A, Ozawa M, Taga C, Sanada N, et al. Activation of the aryl hydrocarbon receptor by 3-methylcholanthrene, but not by indirubin, suppresses mammosphere formation via downregulation of CDC20 expression in breast cancer cells. Biochem Biophys Res Commun. 2021;570:131–6.
Ko C-I, Fan Y, de Gannes M, Wang Q, Xia Y, Puga A. Repression of the aryl hydrocarbon receptor is required to maintain mitotic progression and prevent loss of pluripotency of embryonic stem cells. Stem Cells. 2016;34:2825–39.
Paolo DP. How to scaffold the contractile ring for a safe cytokinesis–lessons from Anillin-related proteins. J Cell Sci. 2009;122:1071–9.
Wurlina W, Mustofa I, Meles DK, Mulyati S, Cempaka Putri DKS, Suwasanti N. Administration of the α-tocopherol for repairing testicle histological damage in rats exposed to dioxin. Thai J Vet Med. 2021;51:293–301.
Hansen DA, Esakky P, Drury A, Lamb L, Moley KH. The aryl hydrocarbon receptor is important for proper seminiferous tubule architecture and sperm development in mice. Biol Reprod. 2014;90:1–8.
Hermann B, Sukhwani M, Winkler F, Pascarella J, Peters K, Sheng Y, et al. Spermatogonial stem cell transplantation into rhesus testes regenerates spermatogenesis producing functional sperm. Cell Stem Cell. 2012;11:715–26.
Mahendran D. Evaluation of spermatogenic, aphrodisiac and anti-oxidant activity of classical siddha drug Thathu Viruthi Chooranam in rodents. Chennai: Government Siddha Medical College; 2017.
Venanzi A, Di Sante M, Bruscoli S, Biagioli M, Sorcini D, Cimino M, et al. Recombinant long-glucocorticoid-induced leucine zipper (L-GILZ) protein restores the control of proliferation in gilz KO spermatogonia. Eur J Pharm Sci. 2014;63:22–8.
Eddy EM. Role of heat shock protein HSP70-2 in spermatogenesis. Rev Reprod. 1999;4:23–30.
Aitken RJ, De Iuliis GN. On the possible origins of DNA damage in human spermatozoa. Mol Hum Reprod. 2009;16:3–13.
Elsayed HYA, Borroto ET, Pliego AB, Dibarrat JA, Ramirez FR, Chagoyán JCV, et al. Sperm quality in mouse after exposure to low doses of TCDD. Curr Top Med Chem. 2019;19:931–43.
Hahn ME. Aryl hydrocarbon receptors: diversity and evolution. Chem Biol Interact. 2002;141:131–60.
Esteban J, Sánchez-Pérez I, Hamscher G, Miettinen HM, Korkalainen M, Viluksela M, et al. Role of aryl hydrocarbon receptor (AHR) in overall retinoid metabolism: Response comparisons to 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) exposure between wild-type and AHR knockout mice. Reprod Toxicol. 2021;101:33–49.
Faiad W, Soukkarieh C, Murphy DJ, Hanano A. Effects of dioxins on animal spermatogenesis: A state-of-the-art review. Front Reprod Health. 2022;4:1009090.
Ruszkowska M, Sadowska A, Nynca A, Orlowska K, Swigonska S, Molcan T, et al. The effects of 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) on the transcriptome of aryl hydrocarbon receptor (AhR) knock-down porcine granulosa cells. PeerJ. 2020;8:e8371.
Sundekilde UK, Kristensen CM, Olsen MA, Pilegaard H, Rasmussen MK. Time-dependent regulation of hepatic cytochrome P450 mRNA in male liver-specific PGC-1α knockout mice. Toxicology. 2022;469:153121.
Mussgnug F, Corso S, Sanchini V. Human reproduction and parental responsibility: new theories, narratives. Ethics Phenomenology and Mind. 2020;19:1–260.
Alabedi T, AL-Baghdady HF, Alahmer MA, Bustani GS, Al-Dhalimy AM. Effects of Ocimum tenuiflorum on Induced Testicular Degeneration by Filgrastim in Wistar Rats. Arch Razi Inst. 2021;76:1555–9.
Amare DE, Bovee TFH, Mulder PPJ, Hamers A, Hoogenboom RLAP. Acid condensation products of indole-3-carbinol and their in-vitro (anti) estrogenic,(anti) androgenic and aryl hydrocarbon receptor activities. Arab J Chem. 2020;13:7199–211.
Tüfek NH, Yahyazadeh A. Protective effect of indole-3-carbinol on sperm morphometric alteration in a high-fat diet-induced obese rat model. J Clin Pract Res. 2021;43:161.
Aggarwal BB, Ichikawa H. Molecular targets and anticancer potential of indole-3-carbinol and its derivatives. Cell Cycle. 2005;4:1201–15.
Rey-Ares V, Rossi SP, Dietrich K-G, Köhn F-M, Schwarzer JU, Welter H, et al. Prostaglandin E2 (PGE2) is a testicular peritubular cell-derived factor involved in human testicular homeostasis. Mol Cell Endocrinol. 2018;473:217–24.
Wang H, Dong B, Zheng Z, Wu Z, Li W, Ding J. Metastasis-associated protein 1 (MTA1) signaling in rheumatoid synovium: Regulation of inflammatory response and cytokine-mediated production of prostaglandin E2 (PGE2). Biochem Biophys Res Commun. 2016;473:442–8.
Seidel SD, Winters GM, Rogers WJ, Ziccardi MH, Li V, Keser B, et al. Activation of the Ah receptor signaling pathway by prostaglandins. J Biochem Mol Toxicol. 2001;15:187–96.
Nebert DW, Karp CL. Endogenous functions of the aryl hydrocarbon receptor (AHR): intersection of cytochrome P450 1 (CYP1)-metabolized eicosanoids and AHR biology. J Biol Chem. 2008;283:36061–5.
Chiang N, Serhan CN. Specialized pro-resolving mediator network: an update on production and actions. Essays Biochem. 2020;64:443–62.
Brokken LJS, Lundberg PJ, Spanò M, Manicardi GC, Pedersen HS, Struciński P, et al. Interactions between polymorphisms in the aryl hydrocarbon receptor signalling pathway and exposure to persistent organochlorine pollutants affect human semen quality. Reprod Toxicol. 2014;49:65–73.
Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature. 2014;510:92–101.
Bradshaw TD, Bell DR. Relevance of the aryl hydrocarbon receptor (AhR) for clinical toxicology. Clin Toxicol. 2009;47:632–42.
Sedlak TW, Snyder SH. Bilirubin benefits: cellular protection by a biliverdin reductase antioxidant cycle. Pediatrics. 2004;113:1776–82.
Togawa H, Shinkai S, Mizutani T. Induction of human UGT1A1 by bilirubin through AhR dependent pathway. Drug Metab Lett. 2008;2:231–7.
Farmahin R, Crump D, Kennedy SW. Sensitivity of avian species to the aryl hydrocarbon receptor ligand 6-formylindolo [3, 2-b] carbazole (FICZ). Chem Biol Interact. 2014;221:61–9.
Jin U-H, Park H, Li X, Davidson LA, Allred C, Patil B, et al. Structure-dependent modulation of aryl hydrocarbon receptor-mediated activities by flavonoids. Toxicol Sci. 2018;164:205–17.
Tan Y-Q, Wang Y-N, Feng H-Y, Guo Z-Y, Li X, Nie X-L, et al. Host/microbiota interactions-derived tryptophan metabolites modulate oxidative stress and inflammation via aryl hydrocarbon receptor signaling. Free Radic Biol Med. 2022;184:30–41.
Henry EC, Bemis JC, Henry O, Kende AS, Gasiewicz TA. A potential endogenous ligand for the aryl hydrocarbon receptor has potent agonist activity in vitro and in vivo. Arch Biochem Biophys. 2006;450:67–77.
Plunk EC, Richards SM. Endocrine-disrupting air pollutants and their effects on the hypothalamus-pituitary-gonadal axis. Int J Mol Sci. 2020;21:9191.
Xu B, Yang H, Sun M, Chen H, Jiang L, Zheng X, et al. 2, 3′, 4, 4′, 5-Pentachlorobiphenyl induces inflammatory responses in the thyroid through JNK and aryl hydrocarbon receptor-mediated pathway. Toxicol Sci. 2016;149:300–11.
Wang Q-Y, Zhang L, Han X-Y, Wang D-M, Ding M-L, Cheng D, et al. 2, 3′, 4, 4′, 5-Pentachlorobiphenyl induces mitochondria-dependent apoptosis mediated by AhR/Cyp1a1 in mouse germ cells. J Hazard Mater. 2023;445:130547.
Cooke PS, Sato T, Buchanan DL. Disruption of Steroid Hormone Signaling by PCBs. In: L. W. Robertson and L. G. Hansen, Eds., PCBs: Recent Advances in Environmental Toxicology and Health Effects. Lexington: The University Press of Kentucky; 2001. p. 257–63.
Wu Y, Luo J, Xu B. Insights into the anticancer effects of galangal and galangin: A comprehensive review. Phytomedicine. 2024;135:156085.
Liu J-R, Miao H, Deng D-Q, Vaziri ND, Li P, Zhao Y-Y. Gut microbiota-derived tryptophan metabolism mediates renal fibrosis by aryl hydrocarbon receptor signaling activation. Cell Mol Life Sci. 2021;78:909–22.
Puppala D, Gairola CG, Swanson HI. Identification of kaempferol as an inhibitor of cigarette smoke-induced activation of the aryl hydrocarbon receptor and cell transformation. Carcinogenesis. 2007;28:639–47.
Hsu C-N, Lin Y-J, Lu P-C, Tain Y-L. Maternal resveratrol therapy protects male rat offspring against programmed hypertension induced by TCDD and dexamethasone exposures: Is it relevant to aryl hydrocarbon receptor? Int J Mol Sci. 2018;19:2459.
Alamo A, Condorelli RA, Mongioì LM, Cannarella R, Giacone F, Calabrese V, et al. Environment and male fertility: Effects of benzo-α-pyrene and resveratrol on human sperm function in vitro. J Clin Med. 2019;8:561.
Banerjee B, Chakraborty S, Ghosh D, Raha S, Sen PC, Jana K. Benzo (a) pyrene induced p53 mediated male germ cell apoptosis: Synergistic protective effects of curcumin and resveratrol. Front Pharmacol. 2016;7:245.
Revel A, Raanani H, Younglai E, Xu J, Han R, Savouret J-F, et al. Resveratrol, a natural aryl hydrocarbon receptor antagonist, protects sperm from DNA damage and apoptosis caused by benzo (a) pyrene. Reprod Toxicol. 2001;15:479–86.
Wang R, Huang Y, Gan X, Fu C, Li Y, Chen N, et al. Switch of phosphorylation to O-GlcNAcylation of AhR contributes to vascular oxidative stress induced by benzo[a]pyrene. Food Sci Human Wellness. 2023;12:2263–75.
Xu A, Wang J, Wang H, Sun Y, Hao T. Protective effect of lycopene on testicular toxicity induced by Benzo [a] pyrene intake in rats. Toxicology. 2019;427:152301.
Ge C, Ye J, Wang Q, Zhang C, Yang J-M, Qian G. Polycyclic aromatic hydrocarbons suppress meiosis in primordial germ cells via the AHR signaling pathway. Toxicol Lett. 2012;210:285–92.
O’Driscoll CA, Gallo ME, Hoffmann EJ, Fechner JH, Schauer JJ, Bradfield CA, et al. Polycyclic aromatic hydrocarbons (PAHs) present in ambient urban dust drive proinflammatory T cell and dendritic cell responses via the aryl hydrocarbon receptor (AHR) in vitro. PLoS ONE. 2018;13:e0209690.
Ventrice P, Ventrice D, Russo E, De Sarro G. Phthalates: European regulation, chemistry, pharmacokinetic and related toxicity. Environ Toxicol Pharmacol. 2013;36:88–96.
Mohammadi H, Ashari S. Mechanistic insight into toxicity of phthalates, the involved receptors, and the role of Nrf2, NF-κB, and PI3K/AKT signaling pathways. Environ Sci Pollut Res. 2021;28:35488–527.
Radeef AM, Wadday AS, Obayes AM, Baqir Al-Dhalimy AM, Al-Hasan BA, Bustani GS. Piracetam and Citicoline Effect on the General Anesthesia Animals Model. J Pharm Negat Results. 2022;13(3):241.
Momeni HR, Eskandari N. Curcumin protects the testis against cadmium-induced histopathological damages and oxidative stress in mice. Hum Exp Toxicol. 2020;39:653–61.
Potter PE. Curcumin: a natural substance with potential efficacy in Alzheimer’s disease. J Exp Pharmacol. 2013;5:23.
Alibraheemi NAA, Bustani GS, Al-Dhalimy AMB. Effect of curcumin on LH and FSH hormones of polycystic syndrome induced by letrozole in female rats. Lat Am J Pharm. 2021;40:179–83.
Shahedi A, Talebi AR, Mirjalili A, Pourentezari M. Protective effects of curcumin on chromatin quality, sperm parameters, and apoptosis following testicular torsion-detorsion in mice. Clin Exp Reprod Med. 2021;48:27–33.
Uçar S, Pandir D. Furan induced ovarian damage in non-diabetic and diabetic rats and cellular protective role of lycopene. Arch Gynecol Obstet. 2017;296:1027–37.
Al-Mousaw M, Bustani GS, Barqaawee MJA, AL-Shamma YM. Evaluation of histology and sperm parameters of testes treated by lycopene against cyclophosphamide that induced testicular toxicity in Male rats. AIP Conf Proc. AIP Publishing LLC;2022. p. 20040.
Çeribaşi AO, Türk G, Sönmez M, Sakin F, Ateşşahin A. Toxic effect of cyclophosphamide on sperm morphology, testicular histology and blood oxidant-antioxidant balance, and protective roles of lycopene and ellagic acid. Basic Clin Pharmacol Toxicol. 2010;107:730–6.
Nandi S, Tripathi SK, Gupta PSP, Mondal S. Molecular Insights of Compromised Female in Under Metabolic and Nutritional Stress. Sustainable Agriculture Reviews 57. Springer; 2022. p. 229–51.
van den Brand AD. Female reproductive health: Species and ligand-specific endometrial differences to hormones and aryl hydrocarbon receptor activation. Doctoral Thesis. Utrecht: Universiteit Utrecht; 2020. p. 186.
Zgarbová E, Vrzal R. The impact of indoles activating the aryl hydrocarbon receptor on androgen receptor activity in the 22Rv1 prostate cancer cell line. Int J Mol Sci. 2022;24:502.
Huang B, Butler R, Miao Y, Dai Y, Wu W, Su W, et al. Dysregulation of Notch and ERα signaling in AhR−/− male mice. Proc Natl Acad Sci. 2016;113:11883–8.
Pencina KM, Travison TG, Cunningham GR, Lincoff AM, Nissen SE, Khera M, et al. Effect of testosterone replacement therapy on sexual function and hypogonadal symptoms in men with hypogonadism. J Clin Endocrinol Metab. 2024;109:569–80.
Ohtake F, Baba A, Fujii-Kuriyama Y, Kato S. Intrinsic AhR function underlies cross-talk of dioxins with sex hormone signalings. Biochem Biophys Res Commun. 2008;370:541–6.
Pocar P, Fischer B, Klonisch T, Hombach-Klonisch S. Molecular interactions of the aryl hydrocarbon receptor and its biological and toxicological relevance for reproduction. Reproduction. 2005;129:379–89.
Wang SY, Cheng YY, Liu SC, Xu YX, Gao Y, Wang CL, et al. A synonymous mutation in IGF-1 impacts the transcription and translation process of gene expression. Mol Ther-Nucleic Acids. 2021;26:1446–65.
Kodama S, Okada K, Akimoto K, Inui H, Ohkawa H. Recombinant aryl hydrocarbon receptors for bioassay of aryl hydrocarbon receptor ligands in transgenic tobacco plants. Plant Biotechnol J. 2009;7:119–28.
Miyazaki T, Chung S, Sakai H, Ohata H, Obata Y, Shiokawa D, et al. Stemness and immune evasion conferred by the TDO2-AHR pathway are associated with liver metastasis of colon cancer. Cancer Sci. 2022;113:170–81.
Chen Z, Xia X, Chen H, Huang H, An X, Sun M, et al. Carbidopa suppresses estrogen receptor-positive breast cancer via AhR-mediated proteasomal degradation of ERα. Invest New Drugs. 2022;40(6):1216–30.
Jablonska O, Piasecka-Srader J, Nynca A, Kołomycka A, Robak A, Wąsowska B, et al. 2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin alters steroid secretion but does not affect cell viability and the incidence of apoptosis in porcine luteinised granulosa cells. Acta Vet Hung. 2014;62:408–21.
Ren R, Zhang S. Non-classical symmetry and analytic self-similar solutions for a non-homogenous time-fractional vector NLS system. Adv Differ Equ. 2021;2021:1–21.
Xu X, Zhang X, Yuan Y, Zhao Y, Fares HM, Yang M, et al. Species-specific differences in aryl hydrocarbon receptor responses: how and why? Int J Mol Sci. 2021;22:13293.
Swedenborg E, Pongratz I. AhR and ARNT modulate ER signaling. Toxicology. 2010;268:132–8.
Ikuta K, Ejima A, Abe S, Shimba A. Control of immunity and allergy by steroid hormones. Allergol Int. 2022;71:432–6.
Matvere A, Teino I, Varik I, Kuuse S, Tiido T, Kristjuhan A, et al. FSH/LH-dependent upregulation of Ahr in murine granulosa cells is controlled by PKA signaling and involves epigenetic regulation. Int J Mol Sci. 2019;20:3068.
Wójtowicz AK, Szychowski KA, Wnuk A, Kajta M. Dibutyl phthalate (DBP)-induced apoptosis and neurotoxicity are mediated via the aryl hydrocarbon receptor (AhR) but not by estrogen receptor alpha (ERα), estrogen receptor beta (ERβ), or peroxisome proliferator-activated receptor gamma (PPARγ) in mouse c. Neurotox Res. 2017;31:77–89.
Ma F, Feng Y, Zhang Y, Wang R, Su D. The roles of stress-induced immune response in female reproduction. Environment and Female Reproductive Health: Springer; 2021. p. 161–79.
Słowikowska-Hilczer J. Xenobiotics with estrogen or antiandrogen action—disruptors of the male reproductive system. Open Medicine. 2006;1:205–27.
Denison MS, Nagy SR. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol. 2003;43:309–34.
Cano-Sánchez J, Murillo-González FE, de Jesús-Aguilar J, Cabañas-Cortés MA, Tirado-Garibay AC, Elizondo G. The Aryl hydrocarbon receptor ligand 6-Formylindolo (3, 2-b) carbazole promotes estrogen receptor Alpha and c-Fos protein degradation and inhibits MCF-7 cell proliferation and migration. Pharmacology. 2023;108:157–65.
Kakeyama M, Sone H, Tohyama C. Perinatal exposure of female rats to 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin induces central precocious puberty in the offspring. J Endocrinol. 2008;197:351–8.
Koga T, Ishida T, Takeda T, Ishii Y, Uchi H, Tsukimori K, et al. Restoration of dioxin-induced damage to fetal steroidogenesis and gonadotropin formation by maternal co-treatment with α-lipoic acid. PLoS ONE. 2012;7:e40322.
Kajta M, Domin H, Grynkiewicz G, Lason W. Genistein inhibits glutamate-induced apoptotic processes in primary neuronal cell cultures: An involvement of aryl hydrocarbon receptor and estrogen receptor/glycogen synthase kinase-3β intracellular signaling pathway. Neuroscience. 2007;145:592–604.
Di Nisio A, Corsini C, Foresta C. Environmental Impact on the Hypothalamus-Pituitary-Testis Axis. Environmental Endocrinology and Endocrine Disruptors: Endocrine and Endocrine-targeted Actions and Related Human Diseases. Springer; 2023. p. 207–38.
Aldeli N, Murphy D, Hanano A. Impact of dioxins on reproductive health in female mammals. Front Toxicol. 2024;6:1392257.
Juricek L, Coumoul X. The aryl hydrocarbon receptor and the nervous system. Int J Mol Sci. 2018;19:2504.
Long M, Ghisari M, Bonefeld-Jørgensen EC. Effects of perfluoroalkyl acids on the function of the thyroid hormone and the aryl hydrocarbon receptor. Environ Sci Pollut Res. 2013;20:8045–56.
Bock KW. Human and rodent aryl hydrocarbon receptor (AHR): from mediator of dioxin toxicity to physiologic AHR functions and therapeutic options. Biol Chem. 2017;398:455–64.
Ma Q, Whitlock JP Jr. The aromatic hydrocarbon receptor modulates the Hepa 1c1c7 cell cycle and differentiated state independently of dioxin. Mol Cell Biol. 1996;16:2144–50.
Zarzycka M, Gorowska-Wojtowicz E, Tworzydlo W, Klak A, Kozub K, Hejmej A, et al. Are aryl hydrocarbon receptor and G-protein–coupled receptor 30 involved in the regulation of seasonal testis activity in photosensitive rodent—the bank vole (Myodes glareolus)? Theriogenology. 2016;86:674–86.
Chen Y, Xie HQ, Sha R, Xu T, Zhang S, Fu H, et al. 2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin and up-regulation of neurofilament expression in neuronal cells: Evaluation of AhR and MAPK pathways. Environ Int. 2020;134:105193.
Cheng Y-H, Li L-A, Lin P, Cheng L-C, Hung C-H, Chang NW, et al. Baicalein induces G1 arrest in oral cancer cells by enhancing the degradation of cyclin D1 and activating AhR to decrease Rb phosphorylation. Toxicol Appl Pharmacol. 2012;263:360–7.
Wilson SR, Joshi AD, Elferink CJ. The tumor suppressor Kruppel-like factor 6 is a novel aryl hydrocarbon receptor DNA binding partner. J Pharmacol Exp Ther. 2013;345:419–29.
Meng Y, Lin R, Wu F, Sun Q, Jia L. Decreased capacity for sperm production induced by perinatal bisphenol a exposure is associated with an increased inflammatory response in the offspring of C57BL/6 male mice. Int J Environ Res Public Health. 2018;15:2158.
Mostafa T, Fouad H, Nabil N, Rashed L, Sabry D, Abougabal K, et al. Aryl hydrocarbon receptor (AhR) rs2066853 gene polymorphism association with infertile oligoasthenoteratozoospermic men and seminal oxidative stress. Environ Sci Pollut Res. 2017;24:8297–301.
Schultz R, Suominen J, Värre T, Hakovirta H, Parvinen M, Toppari J, et al. Expression of aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator messenger ribonucleic acids and proteins in rat and human testis. Endocrinology. 2003;144:767–76.
Choi J, Kim I, Hwang S, Shin B, Kim S. Effect of 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin on testicular spermatogenesis-related panels and serum sex hormone levels in rats. BJU Int. 2008;101:250–5.
Bidgoli SA, Karimi M, Asami Z, Baher H, Zavarhei MD. Association between testicular Aryl hydrocarbon Receptor levels and idiopathic male infertility: A case–control study in Iran. Sci Total Environ. 2011;409:3267–73.
Vogel CFA, Goth SR, Dong B, Pessah IN, Matsumura F. Aryl hydrocarbon receptor signaling mediates expression of indoleamine 2, 3-dioxygenase. Biochem Biophys Res Commun. 2008;375:331–5.
Grunewald ME, Shaban MG, Mackin SR, Fehr AR, Perlman S. Murine coronavirus infection activates the aryl hydrocarbon receptor in an indoleamine 2, 3-dioxygenase-independent manner, contributing to cytokine modulation and proviral TCDD-inducible-PARP expression. J Virol. 2020;94:e01743-e1819.
Dai X, Zhu BT. Indoleamine 2, 3-dioxygenase tissue distribution and cellular localization in mice: implications for its biological functions. J Histochem Cytochem. 2010;58:17–28.
Jrad-Lamine A, Henry-Berger J, Gourbeyre P, Damon-Soubeyrand C, Lenoir A, Combaret L, et al. Deficient tryptophan catabolism along the kynurenine pathway reveals that the epididymis is in a unique tolerogenic state. J Biol Chem. 2011;286:8030–42.
Yang W, Zhang G, Jiang F, Zeng Y, Zou P, An H, et al. BPDE and B [a] P induce mitochondrial compromise by ROS-mediated suppression of the SIRT1/TERT/PGC-1α pathway in spermatogenic cells both in vitro and in vivo. Toxicol Appl Pharmacol. 2019;376:17–37.
Daoud NM, Aly MS, Ezzo OH, Ali NA. Zinc oxide nanoparticles improve testicular steroidogenesis machinery dysfunction in benzo [α] pyrene-challenged rats. Sci Rep. 2021;11:1–14.
Sipinen V, Laubenthal J, Baumgartner A, Cemeli E, Linschooten JO, Godschalk RWL, et al. In vitro evaluation of baseline and induced DNA damage in human sperm exposed to benzo [a] pyrene or its metabolite benzo [a] pyrene-7, 8-diol-9, 10-epoxide, using the comet assay. Mutagenesis. 2010;25:417–25.
Mongioì LM, Perelli S, Condorelli RA, Barbagallo F, Crafa A, Cannarella R, et al. The role of resveratrol in human male fertility. Molecules. 2021;26:2495.
Tartarelli I. Toxicogenomic effects of direct Benzo [a] pyrene-exposure on adult testicular organotypic culture. 2023;
Li Z-D, Wang K, Yang X-W, Zhuang Z-G, Wang J-J, Tong X-W. Expression of aryl hydrocarbon receptor in relation to p53 status and clinicopathological parameters in breast cancer. Int J Clin Exp Pathol. 2014;7:7931.
Garg R, Gupta S, Maru GB. Dietary curcumin modulates transcriptional regulators of phase I and phase II enzymes in benzo [a] pyrene-treated mice: mechanism of its anti-initiating action. Carcinogenesis. 2008;29:1022–32.
Perepechaeva ML, Seredina TA, Sidorova YA, Pivovarova EN, Markel AL, Lyakhovich VV, et al. Quercetin attenuates benzo (α) pyrene-induced CYP1A expression. Biomed Environ Sci. 2017;30:308–13.
Abdulla JM, Al-Okaily BN. Histomorphometric and histopathological alterations of rat testis following exposure to hydrogen peroxide: Protective role of resveratrol supplement. Iraqi J Vet Med. 2022;46:17–23.
Mruk D, Zhu L, SilvestrinI B, Lee WM, Cheng CYAN. Interactions of proteases and protease inhibitors in Sertoli-germ cell cocultures preceding the formation of specialized Sertoli-germ cell junctions in vitro. J Androl. 1997;18:612–22.
Shati AA. Resveratrol improves sperm parameter and testicular apoptosis in cisplatin-treated rats: Effects on ERK1/2, JNK, and Akt pathways. Syst Biol Reprod Med. 2019;65:236–49.
Fung MKL, Cheung HW, Ling MT, Cheung ALM, Wong YC, Wang X. Role of MEK/ERK pathway in the MAD2-mediated cisplatin sensitivity in testicular germ cell tumour cells. Br J Cancer. 2006;95:475–84.
Liu W-C, Shyu J-F, Lin Y-F, Chiu H-W, Lim PS, Lu C-L, et al. Resveratrol rescue indoxyl sulfate-induced deterioration of osteoblastogenesis via the aryl hydrocarbon receptor/MAPK pathway. Int J Mol Sci. 2020;21:7483.
Khayoon HA, Al-Rekabi FMK. Cytotoxic effect of resveratrol on colorectal cancer cell line. Iraqi J Vet Med. 2020;44:68–74.
Jawad RAM, Sahib HB. Estimation the safety of parenteral resveratrol in mice (P-ISSN 1683-3597 E-ISSN 2521-3512). Iraqi J Pharmaceutical. 2022;31:167–75.
Alharris ES, Alghetaa HF, Busbee PB, Nagarkatti M, Nagarkatti P. Resveratrol improves a murine model of asthma through alterations in the gut microbiome. J Immunol. 2017;198:17–53.
Ni F-D, Hao S-L, Yang W-X. Multiple signaling pathways in Sertoli cells: recent findings in spermatogenesis. Cell Death Dis. 2019;10:1–15.
Meles DK, Rachmawati K, Hamid IS, Mustofa I, Wurlina W, Suwasanti N, et al. α-Tocopherol Prevents Sperm Apoptosis and Necrosis in Rats Exposed to 2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin. Vet Med Int. 2022;2022(1):3685686.
Al-Ghezi ZZ, Singh N, Mehrpouya-Bahrami P, Busbee PB, Nagarkatti M, Nagarkatti PS. AhR activation by TCDD (2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin) attenuates pertussis toxin-induced inflammatory responses by differential regulation of tregs and Th17 cells through specific targeting by microRNA. Front Microbiol. 2019;10:2349.
Abdulkareem SM, Nanakali NM. Quercetin reduces oxidative stress damage to reproductive profile induced by 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin in male albino rats (Rattus norvegicus L.). Appl Ecol Environ Res. 2019;17:13185–97.
Fisher MT, Nagarkatti M, Nagarkatti PS. Aryl hydrocarbon receptor-dependent induction of loss of mitochondrial membrane potential in epididydimal spermatozoa by 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol Lett. 2005;157:99–107.
Petersen SL, Krishnan S, Hudgens ED. The aryl hydrocarbon receptor pathway and sexual differentiation of neuroendocrine functions. Endocrinology. 2006;147:s33-42.
Sobolewski M, Anderson T, Conrad K, Marvin E, Klocke C, Morris-Schaffer K, et al. Developmental exposures to ultrafine particle air pollution reduces early testosterone levels and adult male social novelty preference: risk for children’s sex-biased neurobehavioral disorders. Neurotoxicology. 2018;68:203–11.
Skovmand A, Erdely A, Antonini JM, Nurkiewicz TR, Shoeb M, Eye T, et al. Inhalation of welding fumes reduced sperm counts and high fat diet reduced testosterone levels; differential effects in Sprague Dawley and Brown Norway rats. Part Fibre Toxicol. 2020;17:1–14.
Izawa H, Kohara M, Watanabe G, Taya K, Sagai M. Effects of diesel exhaust particles on the male reproductive system in strains of mice with different aryl hydrocarbon receptor responsiveness. J Reprod Dev. 2007;53(6):1191–7.
Alam MS, Hoque MN. Prophylactic effects of vitamin E and selenium on di (n-butyl) phthalate-induced testicular damage in prepubertal rats. J Adv Biotechnol Exp Ther. 2018;1:65–71.
Zhao Y, Lin J, Talukder M, Zhu S-Y, Li M-Z, Wang H-R, et al. Aryl hydrocarbon receptor as a target for lycopene preventing DEHP-induced spermatogenic disorders. J Agric Food Chem. 2020;68:4355–66.
Ji B, Wen Z, Ni C, Zhu Q, Wang Y, Li X, et al. The production of testosterone and gene expression in neonatal testes of rats exposed to diisoheptyl phthalate during pregnancy is inhibited. Front Pharmacol. 2021;12:568311.
Mondal S, Ghosh S, Bhattacharya S, Mukherjee S. Chronic dietary administration of lower levels of diethyl phthalate induces murine testicular germ cell inflammation and sperm pathologies: Involvement of oxidative stress. Chemosphere. 2019;229:443–51.
Mann U, Shiff B, Patel P. Reasons for worldwide decline in male fertility. Curr Opin Urol. 2020;30:296–301.
Wójtowicz AK, Sitarz-Głownia AM, Szczęsna M, Szychowski KA. The action of di-(2-ethylhexyl) phthalate (DEHP) in mouse cerebral cells involves an impairment in aryl hydrocarbon receptor (AhR) signaling. Neurotox Res. 2019;35:183–95.
Zhao Y, Li X-N, Zhang H, Cui J-G, Wang J-X, Chen M-S, et al. Phthalate-induced testosterone/androgen receptor pathway disorder on spermatogenesis and antagonism of lycopene. J Hazard Mater. 2022;439:129689.
Zhao Y, Chen M-S, Wang J-X, Cui J-G, Zhang H, Li X-N, et al. Connexin-43 is a promising target for lycopene preventing phthalate-induced spermatogenic disorders. J Adv Res. 2022;49:115–26.
Assefa EG, Yan Q, Gezahegn SB, Salissou MTM, He S, Wu N, et al. Role of resveratrol on indoxyl sulfate-induced endothelial hyperpermeability via aryl hydrocarbon receptor (AHR)/Src-dependent pathway. Oxid Med Cell Longev. 2019;2019:5847040.
Nsaif GS, Al-Mualm M. The effect of e-cigarettes smoking on expression and methylation of CYP1A1 and CYP1B1 genes and other biochemical parameters. Mater Today Proc. 2021;80(6):3863–6.
Bustani GS, Baiee FH. Semen extenders: An evaluative overview of preservative mechanisms of semen and semen extenders. Vet World. 2021;14:1220.
Bucak MN, Ataman MB, Başpınar N, Uysal O, Taşpınar M, Bilgili A, et al. Lycopene and resveratrol improve post-thaw bull sperm parameters: sperm motility, mitochondrial activity and DNA integrity. Andrologia. 2015;47:545–52.
Zhao Y, Bao R-K, Zhu S-Y, Talukder M, Cui J-G, Zhang H, et al. Lycopene prevents DEHP-induced hepatic oxidative stress damage by crosstalk between AHR–Nrf2 pathway. Environ Pollut. 2021;285:117080.
Zhao Y, Li M-Z, Talukder M, Luo Y, Shen Y, Wang H-R, et al. Effect of mitochondrial quality control on the lycopene antagonizing DEHP-induced mitophagy in spermatogenic cells. Food Funct. 2020;11:5815–26.
Vogel CFA, Chang WLW, Kado S, McCulloh K, Vogel H, Wu D, et al. Transgenic overexpression of aryl hydrocarbon receptor repressor (AhRR) and AhR-mediated induction of CYP1A1, cytokines, and acute toxicity. Environ Health Perspect. 2016;124:1071–83.
Lin C-H, Chou C-C, Lee Y-H, Hung C-C. Curcumin facilitates aryl hydrocarbon receptor activation to ameliorate inflammatory astrogliosis. Molecules. 2022;27:2507.
Bustani GS, Jabbar MK, AL-Baghdady HF, Al-Dhalimy AMB. Protective effects of curcumin on testicular and sperm parameters abnormalities induced by nicotine in male rats. AIP Conf Proc. AIP Publishing; 2022.
Alharris E, Alghetaa H, Seth R, Chatterjee S, Singh NP, Nagarkatti M, et al. Resveratrol attenuates allergic asthma and associated inflammation in the lungs through regulation of miRNA-34a that targets FoxP3 in mice. Front Immunol. 2018;9:2992.
Li M-Z, Zhao Y, Wang H-R, Talukder M, Li J-L. Lycopene preventing dehp-induced renal cell damage is targeted by aryl hydrocarbon receptor. J Agric Food Chem. 2021;69:12853–61.
Wang L, Xue J, Wei F, Zheng G, Cheng M, Liu S. Chemopreventive effect of galangin against benzo (a) pyrene-induced stomach tumorigenesis through modulating aryl hydrocarbon receptor in Swiss albino mice. Hum Exp Toxicol. 2021;40:1434–44.
Lněničková K, Skálová L, Stuchlíková Raisová L, Szotáková B, Matoušková P. Induction of xenobiotic-metabolizing enzymes in hepatocytes by beta-naphthoflavone: Time-dependent changes in activities, protein and mRNA levels. Acta Pharmaceutica. 2018;68:75–85.
Shi LZ, Czuprynski CJ. Beta-naphthoflavone causes an AhR-independent inhibition of invasion and intracellular multiplication of Listeria monocytogenes in murine hepatocytes. Microb Pathog. 2009;47:258–66.
Ben Maamar M, Nilsson EE, Skinner MK. Epigenetic transgenerational inheritance, gametogenesis and germline development. Biol Reprod. 2021;105:570–92.
Legoff L, D’Cruz SC, Tevosian S, Primig M, Smagulova F. Transgenerational inheritance of environmentally induced epigenetic alterations during mammalian development. Cells. 2019;8:1559.
Wajda A, Łapczuk-Romańska J, Paradowska-Gorycka A. Epigenetic regulations of AhR in the aspect of immunomodulation. Int J Mol Sci. 2020;21:6404.
Knecht AL, Truong L, Marvel SW, Reif DM, Garcia A, Lu C, et al. Transgenerational inheritance of neurobehavioral and physiological deficits from developmental exposure to benzo [a] pyrene in zebrafish. Toxicol Appl Pharmacol. 2017;329:148–57.
Baker TR, King-Heiden TC, Peterson RE, Heideman W. Dioxin induction of transgenerational inheritance of disease in zebrafish. Mol Cell Endocrinol. 2014;398:36–41.
Kuroki S, Matoba S, Akiyoshi M, Matsumura Y, Miyachi H, Mise N, et al. Epigenetic regulation of mouse sex determination by the histone demethylase Jmjd1a. Science. 1979;2013(341):1106–9.
Mirbahai L, Williams TD, Zhan H, Gong Z, Chipman JK. Comprehensive profiling of zebrafish hepatic proximal promoter CpG island methylation and its modification during chemical carcinogenesis. BMC Genomics. 2011;12:1–16.
Habano W, Miura T, Terashima J, Ozawa S. Aryl hydrocarbon receptor as a DNA methylation reader in the stress response pathway. Toxicology. 2022;470:153154.
Viluksela M, Pohjanvirta R. Multigenerational and transgenerational effects of dioxins. Int J Mol Sci. 2019;20:2947.
Bruner-Tran KL, Ding T, Yeoman KB, Archibong A, Arosh JA, Osteen KG. Developmental exposure of mice to dioxin promotes transgenerational testicular inflammation and an increased risk of preterm birth in unexposed mating partners. PLoS ONE. 2014;9:e105084.
Tomikawa J, Uenoyama Y, Ozawa M, Fukanuma T, Takase K, Goto T, et al. Epigenetic regulation of Kiss1 gene expression mediating estrogen-positive feedback action in the mouse brain. Proc Natl Acad Sci. 2012;109:E1294–301.
Romagnolo DF, Daniels KD, Grunwald JT, Ramos SA, Propper CR, Selmin OI. Epigenetics of breast cancer: Modifying role of environmental and bioactive food compounds. Mol Nutr Food Res. 2016;60:1310–29.
Kajta M, Wnuk A, Rzemieniec J, Litwa E, Lason W, Zelek-Molik A, et al. Depressive-like effect of prenatal exposure to DDT involves global DNA hypomethylation and impairment of GPER1/ESR1 protein levels but not ESR2 and AHR/ARNT signaling. J Steroid Biochem Mol Biol. 2017;171:94–109.
Acknowledgements
Not applicable.
Funding
No institutional funds are available.
Author information
Authors and Affiliations
Contributions
Conceptualization, GB, HA, AM, MN, and PN; Investigation, GB, HA, AM, MN, and PN; Writing—original draft preparation, GB and HA; Writing—review and editing, GB, HA, AM, MN and PN; Visualization, GB and AM; Supervision, HA, and PN. Financial support, MN, and PN. All authors have read and agreed to the current version of the manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
This review article does not have any individual-related identifiable data or images.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Bustani, G., Alghetaa, H., Mohammed, A. et al. The aryl hydrocarbon receptor: a new frontier in male reproductive system. Reprod Biol Endocrinol 23, 70 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12958-025-01401-3
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12958-025-01401-3