Abstract
The aryl hydrocarbon receptor (AhR) has been implicated in regulating skeletal progenitor cells and the activity of bone forming osteoblasts and bone resorbing osteoclasts, thereby impacting bone mass and the risk of skeletal fractures. The AhR also plays an important role in the immune system within the skeletal niche and the differentiation of mesenchymal stem cells into other cell lineages including chondrocytes and adipocytes. This transcription factor responds to environmental pollutants which can act as AhR ligands, initiating or interfering with various signaling cascades to mediate downstream effects, and also responds to endogenous ligands including tryptophan metabolites. This review comprehensively describes the reported roles of the AhR in skeletal cell biology, focusing on mesenchymal stem cells, osteoblasts, and osteoclasts, and discusses how AhR exhibits sexually dimorphic effects in bone. The molecular mechanisms mediating AhR’s downstream effects are highlighted to emphasize the potential importance of targeting this signaling cascade in skeletal disorders.
Keywords: bone, nuclear receptors, osteoblast, osteoclast, skeletal
1. Introduction
Bone is a critical and specialized organ in vertebrates (Su, Yang et al. 2019) that plays key roles in movement, internal organ protection, hematopoiesis, and storage of minerals. The function of bone is maintained through highly controlled mechanisms that regulate the activity of bone forming cells (osteoblasts) and bone resorbing cells (osteoclasts). Bone modeling describes the process of bone growth achieved through the independent action of osteoblasts and osteoclasts (Langdahl, Ferrari et al. 2016). This process begins during fetal development and is a major contributor to both peak bone mass and overall bone shape (Siddiqui and Partridge 2016). Bone also undergoes constant remodeling activity where osteoblasts and osteoclasts work together to renew, repair, and adapt the tissue as needed in response to various stimuli and signals such as hormones, cytokines, growth factors, and biomechanical forces (Siddiqui and Partridge 2016, Eisa, Reddy et al. 2020). Skeletal diseases can reflect the effects of inadequate balance between bone formation and resorption activity. In osteoporosis, osteoclast-mediated bone resorption becomes unbalanced with osteoblast-mediated bone formation, leading to a net decrease in bone mass and a corresponding increase in risk of bone fracture.
Aging is often associated with bone loss and increased skeletal fragility that adversely impact patients’ quality of life (Kim, Hamrick et al. 2019). While many causative factors for osteoporosis exist, one proposed contributor to bone loss with age is activation of a specialized nuclear hormone receptor called the aryl hydrocarbon receptor (AhR) (Refaey, McGee-Lawrence et al. 2017, Eisa, Reddy et al. 2020). This receptor acts as a transcription factor that responds to environmental pollutants and has been implicated in regulatory mechanisms influencing the immune system, liver homeostasis, and metabolic diseases as well as bone (Wright, De Castro et al. 2017, Eisa, Reddy et al. 2020, Kondrikov, Elmansi et al. 2020). AhR functions through canonical and non-canonical pathways to regulate these biological processes. Canonical signaling involves the binding of AhR to a nucleotide sequence (5’-GCGTG-3’) called the xenobiotic response element (XRE), also referred to as dioxin response elements (DRE), via molecular mechanisms that have been studied and reviewed extensively in several previous papers (McMillan and Bradfield 2007, Beischlag, Luis Morales et al. 2008). However, there are relatively fewer studies describing the process of non-canonical AhR signaling (Jackson, Joshi et al. 2015, Wright, De Castro et al. 2017). The latter pathway has been described after failing to observe a readily identifiable XRE sequence in genes responsive to AhR activation (Huang and Elferink 2012, Wright, De Castro et al. 2017). In fact, AhR has been shown to form complexes with other proteins and interact with a variety of response elements, exhibiting a plethora of effects (Huang and Elferink 2012, Wilson, Joshi et al. 2013, Jackson, Li et al. 2014, Jackson, Joshi et al. 2015, Wright, De Castro et al. 2017).
Tryptophan metabolites such as kynurenine are well-known endogenous ligands to the AhR (Lanis, Alexeev et al. 2017). Since these compounds are polar in nature, they gain access to the intracellular compartment through a transporter known as solute carrier transporter 7a5 (SLC7A5), also referred to as large amino acid transporter 1 (LAT1) (Napolitano, Scalise et al. 2015, Scalise, Galluccio et al. 2018, Sinclair, Neyens et al. 2018). Once inside the cell, these ligands can exhibit their biological effects either through canonical or non-canonical AhR signaling cascades. Because AhR mediates diverse endogenous functions, its effect on the skeletal system have not been fully described. Therefore, understanding how AhR could affect the balance between bone formation and bone resorption activity paves the way for new therapeutics in the field. The purpose of this review is to explore the effects of signaling through AhR in mechanisms of bone cell activity.
2. Bone cells
Mesenchymal stem cells (MSCs) are multipotent progenitor cells that can differentiate into bone-forming osteoblasts (Stein and Lian 1993). MSC undergo an initial proliferation phase where genes involved in cell cycle and cell growth (e.g., c-myc, c-jun), are highly expressed (Stein and Lian 1993). This phase is accompanied by the initial stages of extracellular matrix production through the expression of proteins including type I collagen, transforming growth factor beta (TGF-β), and fibronectin (Stein and Lian 1993). During the early stages of differentiation, these cells begin to express genes associated with an osteoblastic bone cell phenotype including runt-related transcription factor 2 (RUNX2) (Stein and Lian 1993, Komori, Yagi et al. 1997, Korkalainen, Kallio et al. 2009). The expression of type I collagen persists during differentiation, although extracellular matrix composition changes considerably as the cells mature (Assis-Ribas, Forni et al. 2018). Furthermore, during the differentiation phase, proteins related to osteoblast phenotype including alkaline phosphatase, osteocalcin, and osteopontin are synthesized and released in part to promote matrix mineralization (Stein and Lian 1993), providing bone with its unique biochemical properties (Murshed 2018).
Osteoclasts, which resorb bone, are multinucleated cells that arise from hematopoietic stem cells (Boyle, Simonet et al. 2003). These cells solubilize the organic component of bone matrix by secreting proteases such as cathepsin K, matrix metalloproteinases (MMPs), and tartrate resistant acid phosphatase (TRAP) (Boyle, Simonet et al. 2003). They also solubilize the mineral component of bone matrix through the release of acidic hydrogen ions in the form of hydrochloric acid (Roodman 1999). Osteoclast differentiation is initiated by two fundamental molecules; macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-B ligand (RANKL) (Boyle, Simonet et al. 2003). M-CSF, which is produced by a variety of cells including osteoblasts and stromal cells (Mun, Park et al. 2020) drives pre-osteoclast survival, proliferation, and surface expression of RANK (Korkalainen, Kallio et al. 2009). The binding of RANKL to RANK triggers a series of signaling cascades that results in the fusion of pre-osteoclasts to form polykaryons, which then mature to give rise to functional, bone-resorbing osteoclasts (Boyle, Simonet et al. 2003). Osteoblast-lineage cells can also secrete osteoprotegerin (OPG) which serves as a decoy receptor for RANKL, thus blocking RANKL-RANK signaling and suppresses osteoclastogenesis (Boyle, Simonet et al. 2003).
The coupled action of osteoblasts and osteoclasts is key to bone homeostasis. In addition to being regulated by endocrine factors, exogenous factors such as environmental chemicals and pollutants influence bone remodeling balance (Iqbal, Sun et al. 2013). For instance, cigarette smoke is known to impair bone formation and increase osteoclastic bone resorption (Iqbal, Sun et al. 2013). In fact, long term smoking has been associated with elevated risk of osteoporosis and bone fractures (Ward and Klesges 2001, Al-Bashaireh, Haddad et al. 2018). Smoke toxicants contain molecules like 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; dioxin) that adversely affect the bone (Iqbal, Sun et al. 2013). Interestingly, TCDD mediates its effect at least in part through activating the AhR which in turn drives the expression of genes involved in bone resorption (Iqbal, Sun et al. 2013). The role of AhR in bone health has been investigated by several research groups (Herlin, Finnila et al. 2013, Tong, Niu et al. 2017, Eisa, Reddy et al. 2020), but its exact action remains elusive due to the diversity of cell type specific effects.
3. AhR signaling and regulation
AhR is a ligand-activated transcription factor involved in the response to environmental pollutants and chemicals such as aromatic hydrocarbons (Swedenborg and Pongratz 2010). In addition, it plays a role in a variety of biological processes like drug metabolism (Ramadoss, Marcus et al. 2005), immune regulation (Stevens, Mezrich et al. 2009), and cardiovascular activity (Zhu, Meng et al. 2019). In the absence of a ligand, AhR is present in the cytoplasm in a chaperone complex consisting of heat shock protein 90 (Hsp90), P23, aryl hydrocarbon receptor associated 9 (ARA9), and c-SRC (Carambia and Schuran 2021). This complex ensures that AhR is properly folded and maintained in the correct three-dimensional confirmation that allows ligand interaction (Stevens, Mezrich et al. 2009). Upon agonist binding to AhR, a conformational change is induced which exposes the nuclear localization signal and allows the AhR-chaperone complex to translocate to the nucleus (Stevens, Mezrich et al. 2009). In the nucleus, the chaperone complex disassembles and AhR forms a heterodimer with aryl hydrocarbon receptor nuclear translocator (ARNT) to generate an activated transcription factor (Stevens, Mezrich et al. 2009). Several co-activators can be recruited to the new complex facilitating its binding to an XRE sequence (Stevens, Mezrich et al. 2009). This binding triggers the expression of AhR target genes such as cytochrome P450 family 1 subfamily A member 1 (CYP1A1), and subfamily B member 1 (CYP1B1) (Zhu, Meng et al. 2019). The process described above is referred to as canonical AhR signaling (Figure 1).
Figure 1: Canonical and non-canonical AhR signaling pathways.
Upon binding to an agonist, such as kynurenine (KYN), AhR leaves its chaperone complex and translocates to the nucleus. In the canonical pathway, AhR interacts with ARNT and binds the xenobiotic response element (XRE) nucleotide sequence to drive the expression of target genes including CYP1A1 and CYP1B1. The non-canonical pathway, in contrast, involves the interaction of AhR and KLF6 binding to a non-consensus XRE (NC-XRE) nucleotide sequence to drive expression of target genes including PAI-1 and p21cip1. Figure created with BioRender.com.
Another pathway for AhR-mediated effects is through non-canonical signaling, where AhR forms a complex with other molecules allowing it to bind to genes lacking XRE sequences (Wright, De Castro et al. 2017) (Figure 1). A non-consensus xenobiotic response element (NC-XRE) was first described by Huang and Elferink in 2012 (Huang and Elferink 2012), consisting of a 5'-GGGA-3’ tetranucleotide repeat. Subsequent studies demonstrated that this site utilizes Kruppel-like factor 6 (KLF6) as a binding partner rather than ARNT (Huang and Elferink 2012, Wilson, Joshi et al. 2013, Jackson, Joshi et al. 2015), and that target genes regulated through this mechanism include PAI-1 (Huang and Elferink 2012) and p21cip1 (Jackson, Li et al. 2014).
The AhR-ARNT dimer can directly interact with estrogen receptor (ER) to create a transcriptionally active complex that binds the estrogen response element (ERE). (Ohtake, Takeyama et al. 2003, Jackson, Joshi et al. 2015). AhR can also regulate the cell cycle after complexing with S phase proteins including E2F, p300, pRb (Puga, Barnes et al. 2000, Marlowe, Knudsen et al. 2004). Accumulating evidence also demonstrates that AhR directly interacts with RelB to regulate NF-κB target genes (Vogel, Sciullo et al. 2007). In addition to the genomic effects induced by AhR, it can act through non-genomic manner to control biological processes (Carambia and Schuran 2021). For example, ligand binding triggers the dissociation of the chaperone complex and releases c-SRC, allowing c-SRC to act as a protein kinase that phosphorylates downstream proteins (Carambia and Schuran 2021). One effect of this c-SRC activity is increased focal adhesion kinase activation that can lead to increased cellular adhesion and decreased migration (Larigot, Juricek et al. 2018). Ligand binding to AhR can also increase intracellular calcium, which when coupled with its c-SRC activation ability, promotes cyclooxygenase 2 (COX2) and arachidonic acid production leading to inflammation (Larigot, Juricek et al. 2018). In addition, AhR-ligand-binding can promote the degradation of Indolamine-2,3-dioxygenase 1 (IDO1) by generating E3 ubiquitin ligase complex (Pallotta, Fallarino et al. 2014). IDO1 is responsible for the conversion of tryptophan to kynurenine and has been implicated in inflammatory processes (Pallotta, Fallarino et al. 2014).
AhR can be activated by a wide range of endogenous and exogenous ligands (Denison and Nagy 2003), and it is important to note that the downstream signaling mediated by AhR upon binding these molecules is both ligand- and cell-type dependent (Safe, Jin et al. 2020). Smoke toxicants such as TCDD and benzo [a] pyrene (BaP) are common AhR agonists that have been reported to influence the skeletal and immune systems (Table 1) (Iqbal, Sun et al. 2013). However, to our knowledge, less is known about the skeletal effects of non-classical AhR agonists such as thiabendazole (Seidel, Winters et al. 2001, Iqbal, Sun et al. 2013). Furthermore, proton pump inhibitors like omeprazole have been described as AhR agonists in breast cancer cells, but their influence on bone also occurs through non-AhR mediated effects such as blocking acid secretion and hindering calcium absorption (Hyun, Chun et al. 2010, Jin, Lee et al. 2014).
Table 1:
Known AhR ligands affecting the skeletal and immune systems
Compound | Reference |
---|---|
Agonists | |
TCDD | (Herlin, Finnila et al. 2013, Iqbal, Sun et al. 2013, Tong, Niu et al. 2017, Fader, Nault et al. 2018) |
BaP | (Iqbal, Sun et al. 2013, Zhou, Jiang et al. 2017, An, Shi et al. 2020) |
Kynurenine | (Kim, Hamrick et al. 2019, Eisa, Reddy et al. 2020) |
Kynurenic acid | (DiNatale, Murray et al. 2010) |
FICZ | (Oberg, Bergander et al. 2005) |
3MC | (Naruse, Otsuka et al. 2004, Yu, Kondo et al. 2014, Yu, Pang et al. 2015) |
Tetrandrine | (Jia, Tao et al. 2019) |
Norisoboldine | (Wei, Lv et al. 2015, Lv, Wang et al. 2018) |
PCB126 | (Lee and Yang 2012) |
Lipoxin A4 | (Schaldach, Riby et al. 1999, Liu, Guan et al. 2017) |
Leflunomide | (Liang, Li et al. 2020) |
Serotonin | (Nieto, Rayo et al. 2020) |
4-Hydroxytamoxifen | (DuSell, Nelson et al. 2010) |
Sinomenine | (Tong, Yuan et al. 2016) |
Antagonists | |
BAY 2416964 | (Lu, Liu et al. 2022) |
CH-223191 | (Cui, Feng et al. 2020) |
3’4’DMF | (Kido, Fujihara et al. 2014) |
GNF351 | (Watson, Nordberg et al. 2019) |
α-naphthoflavone | (Cedervall, Lind et al. 2015) |
AhR activity can be downregulated by negative feedback and proteasomal degradation (Davarinos and Pollenz 1999, Mimura, Ema et al. 1999). When the AhR/ARNT heterodimer binds an XRE, it induces the expression of AhR repressor (AhRR), which is a polypeptide structurally similar to AhR but carries a potent transcriptional repressor domain (Mimura, Ema et al. 1999). AhRR can dimerize with ARNT and reduce AhR transcriptional activity by competing for XRE binding (Mimura, Ema et al. 1999). Similarly, CYP1A1 and CYP1A2 are directly activated by AhR to promote the degradation of AhR ligands and limit the activity of this pathway (Carambia and Schuran 2021). Another mechanism to reduce AhR activity is through proteasomal degradation of the ligand-activated AhR after being exported from the nuclear compartment (Davarinos and Pollenz 1999). In addition to the processes described above, AhR-mediated signaling activity can also be epigenetically regulated via mechanisms including changes in DNA methylation, microRNAs, and histone deacetylases (Gomez-Duran, Ballestar et al. 2008, Amenya, Tohyama et al. 2016, Elmansi, Hussein et al. 2020, Tantoh, Wu et al. 2020, Disner, Lopes-Ferreira et al. 2021).
Growing evidence indicates that AhR signaling interacts with critical cellular pathways including vascular endothelial growth factor (VEGF) (Takeuchi, Takeuchi et al. 2009), WNT (Arinze, Yin et al. 2022), fibroblast growth factor (FGF) (Girer, Murray et al. 2016), TGF-β (Gramatzki, Pantazis et al. 2009), toll-like receptor (TLR) (Kado, Chang et al. 2017), the CXCL12/CXCR4 axis (Elmansi, Hussein et al. 2020), and ER (Vert and Chory 2011) signaling, among others. While a detailed summary of each of these mechanisms is beyond the scope of this review, clearly these pathways are all well-established regulators of myriad MSC, chondrocyte, osteoblast, and osteoclast functions as reviewed elsewhere (Plotkin and Bruzzaniti 2019, Hallett, Ono et al. 2021, Yahara, Nguyen et al. 2022), and accordingly AhR crosstalk with these pathways may drive downstream cellular responses. However, the relative impact of signaling directly mediated by AhR as compared to its modulation of other pathways remains to be elucidated in discrete skeletal cell populations.
4. Role of AhR in MSCs
MSCs constitute a heterogenous population of cells that can differentiate into several mesodermal cell lineages such as chondrocytes, adipocytes, and osteoblasts (Abney and Galipeau 2021). The AhR is strongly expressed in MSCs (Huang, Wang et al. 2022), and many independent reports indicate that AhR-mediated signaling inhibits MSC proliferation (Zhou, Jiang et al. 2017, Jia, Zhao et al. 2021, Huang, Wang et al. 2022). Human bone marrow MSC proliferation was inhibited by AhR ligand kynurenine and stimulated by AhR inhibitor SR1 in a concentration and time-dependent manner (Jia, Zhao et al. 2021); proliferation was also stimulated through siRNA-mediated knockdown of AhR expression, which protected against the inhibitory effects of kynurenine (Jia, Zhao et al. 2021). The cigarette smoke toxicant BaP decreased the proliferation and self-renewal of rat MSCs, comparable to the reduced proliferation rate seen in MSCs isolated from human smokers as compared to non-smokers (Zhou, Jiang et al. 2017). However, some disagreement exists, as the AhR ligand FICZ did not affect rat bone marrow MSC proliferation (Huang, Wang et al. 2022). Other cellular processes relating to cell survival are also affected by AhR signaling in MSC, for instance kynurenine disrupted autophagy and promoted senescence in murine BMSC in an AhR-dependent manner (Kondrikov, Elmansi et al. 2020).
MSCs play a role in a variety of biological processes including inflammation (Abney and Galipeau 2021), and the AhR is implicated in the anti-inflammatory effect of MSCs (de Almeida, Evangelista et al. 2017, Abney and Galipeau 2021). Upon tissue injury, MSCs migrate to the affected site to modulate inflammatory response (Ou-Yang, Huang et al. 2011). Xu et al. demonstrated that AhR knockout mice display heightened lung inflammation following allergen exposure due to impaired migratory potential of MSCs (Xu, Zhou et al. 2015). Mechanistically, after MSCs arrive to the site of injury, they induce a switch in macrophage phenotype from the proinflammatory M1 to the anti-inflammatory M2 (Cui, Feng et al. 2020) and pharmacological inhibition of AhR by CH223191 reduced the polarization of macrophages from M1 to M2 phenotype and exacerbated inflammation (Cui, Feng et al. 2020). Moreover, AhR signaling affected the ability of MSCs to produce soluble cytokines (Jensen, Leeman et al. 2003). This was revealed when an AhR-expressing bone marrow stromal cell line (BMS2) was treated with the AhR agonists 7,12-dimethylbenz [a]anthracene (DMBA) and TCDD, both of which reduced NF-κB levels (Jensen, Leeman et al. 2003). Since the interleukin-6 (IL-6) promoter features NF-κB binding sites, the reduction of NF-κB reduced the expression of IL-6, and lipopolysaccharide-induced increases in IL-6 levels were blunted in the presence of DMBA and TCDD (Jensen, Leeman et al. 2003). In addition, the AhR antagonists DMF and CH-223191 inhibited kynurenine-induced downregulation of gene and protein levels of the inflammatory and multifunctional cytokine CXCL12 in human and murine BMSCs (Elmansi, Hussein et al. 2020). Collectively, these effects indicate that AhR stimulation directly affects the inflammatory responses of MSCs.
The well-regulated expression of transcription factors is critical for MSCs differentiation into different cell lineage including osteoblasts (which will be described in the next section) and adipocytes, and disruptions in the expression of transcription factors can therefore compromise MSCs differentiation capacity. With regards to adipocyte commitment of MSCs, the AhR ligand BaP was administered to human bone marrow derived MSCs and suppressed adipogenic differentiation (Podechard, Fardel et al. 2009) by reducing mRNA expression of CCAAT/enhancer-binding protein beta (CEBPβ) and peroxisome proliferator-activated receptor γ (PPARγ), key transcription factors for adipogenesis (Podechard, Fardel et al. 2009). BaP also repressed expression of adipocyte markers like fatty acid binding protein-4 (Fabp4), whereas the AhR antagonist α-naphthoflavone successfully counteracted BaP’s inhibitory effects on adipogenesis (Podechard, Fardel et al. 2009). A similar study reported that treating canine MSCs with BaP inhibited adipocyte differentiation by activating AhR and downregulating PPARγ (Rathore and Cekanova 2015). Intriguingly, in vivo administration of the AhR ligand TCDD also decreased bone marrow adiposity (Fader, Nault et al. 2018).
5. Role of AhR in osteoblasts
Several studies suggest that activation of AhR by environmental toxicants like TCDD and BaP impedes the differentiation of MSCs into osteoblasts (Figure 2) (Korkalainen, Kallio et al. 2009, Tong, Niu et al. 2017, Zhou, Jiang et al. 2017, Watson, Nordberg et al. 2019). For instance, TCDD-treated MSCs showed a dose dependent decrease in mRNA levels of osteoblastic markers such as RUNX2, osteocalcin and alkaline phosphatase, mediated through inhibiting β-catenin expression (Tong, Niu et al. 2017). Zhou et al. verified that BaP inhibited alkaline phosphatase and matrix mineralization of MSCs and showed that in vitro treatment with BaP inhibited TGF-β1/SMAD4 and TGF-β1/ERK/AKT signaling pathways (Zhou, Jiang et al. 2017). Korkalainen et al. treated mice and rats with TCDD and isolated MSCs from bone marrow of tibia and femur, monitoring the progress of osteoblastic differentiation in these cells (Korkalainen, Kallio et al. 2009). Their quantitative real time-polymerase chain reaction (RT-PCR) results showed reduced mRNA levels of RUNX2, osteocalcin and alkaline phosphatase, suggesting impaired osteoblastic differentiation (Korkalainen, Kallio et al. 2009). To directly test the role of the AhR in these mechanisms, the authors repeated several experiments in AhR-knockout mice and found that the impaired osteoblastic differentiation attributed to TCDD was rescued in the knockout strain (Korkalainen, Kallio et al. 2009). In another study, skeletal changes including decreased cortical thickness and elevated cortical bone porosity were evident after 10 weeks of TCDD treatment (Herlin, Finnila et al. 2013). Bone matrix was harder after TCDD treatment, suggesting increased matrix brittleness in TCDD treated mice comparable to what is seen in older bone with impaired osteogenesis (Herlin, Finnila et al. 2013). A suppressive role of AhR activation on osteoblast differentiation was also evident in human derived MSCs (Watson, Nordberg et al. 2019), where TCDD treatment suppressed alkaline phosphatase activity, matrix mineralization, and expression of the transcription factor DLX5 and the osteogenic markers osteopontin and integrin-binding sialoprotein (Watson, Nordberg et al. 2019). Fibroblast growth factor 9 (FGF9) and FGF18 expression, known to inhibit MSC differentiation into osteoblasts, were upregulated by TCDD treatment (Watson, Nordberg et al. 2019). Blocking AhR with GNF351 attenuated TCDD-induced effects on matrix mineralization and rescued expression of genes related to extracellular matrix, osteogenic regulation and maintenance of multipotency (Watson, Nordberg et al. 2019).
Figure 2: Reported effects of AhR signaling in bone cells.
AhR has been reported to exhibit a diverse array of effects in cells of the MSC and HSC lineages, with many effects occurring in a ligand- and cell type-dependent fashion. Figure created with BioRender.com.
Surprisingly however, at least two studies have shown a net benefit of TCDD treatment on bone (Herlin, Finnila et al. 2013, Fader, Nault et al. 2018), resulting in part from increasing osteoblasts (Fader, Nault et al. 2018) (Figure 2). Likewise, treatment of MC3T3-E1 pre-osteoblasts with the endogenous AhR ligand 6-Formylindolo[3,2-b] carbazole (FICZ) accelerated osteoblastic differentiation and expression of osteoblastic genes including alkaline phosphatase, osteocalcin, and type 1 collagen (Yoshikawa, Izawa et al. 2021). Moreover, in vivo administration of FICZ increased bone formation rate and mineral apposition rate in wildtype mice (Yoshikawa, Izawa et al. 2021). Similarly, a recent report by Huang et al. demonstrated that FICZ treatment promoted osteogenic differentiation of rat bone marrow stromal cells (Huang, Wang et al. 2022). However, kynurenine (another endogenous AhR ligand) inhibited differentiation of BMSC-derived osteoblasts, reducing mineralized matrix production by these cells, and also decreased dynamic indices of bone mineralization activity (Refaey, McGee-Lawrence et al. 2017). These studies highlight the critical point that AhR-mediated signaling can be ligand-dependent, with different ligands capable of producing opposite phenotypic responses both in vivo and in vitro in a cell type-specific manner, as previously reviewed (Safe, Jin et al. 2020) (Figure 2). Many studies have also demonstrated that the effect of AhR can be species dependent (Unkila, Ruotsalainen et al. 1995, Karchner, Franks et al. 2006, Xu, Zhang et al. 2021). One such example is the distinctive sensitivity to TCDD among mammals, fish, toads and birds (Xu, Zhang et al. 2021). Among all vertebrates, the most sensitive species to TCDD are fish, while toads and frogs are considered the least sensitive (Peterson, Theobald et al. 1993, Xu, Zhang et al. 2021). Intraspecies variability is also observed, with 120-fold difference in the response to TCDD between two fish species; bull trout (Salvelinus confluentus) and zebrafish (Danio rerio) (King-Heiden, Mehta et al. 2012). Mammals exhibit differential responses to TCDD as well, for example, rats and hamsters demonstrate teratogenic effects upon exposure to this agent, but this impact is undetectable in guinea pigs (Kransler, McGarrigle et al. 2007).
Discrepancies in the sensitivity of different species to AhR-mediated signaling could be impacted by differing primary structures of the AhR ligand binding domain among species, where small amino acid variations in this domain greatly alter the extent of AhR activation (Xu, Zhang et al. 2021). Supporting this idea is a site directed mutagenesis study demonstrating that TCDD binding affinity was reduced upon replacing Ala375 with Val (Poland, Palen et al. 1994). Changes in the primary structure of AhR affect ligand and coregulator interactions, likely contributing to varied downstream responses (Pandini, Denison et al. 2007). Many different transcription factors and coregulators can bind to AhR depending on its microenvironment (Gargaro, Scalisi et al. 2021). For instance, AhR can either directly interact with signal transducers and activator of transcription (STAT) to regulate proinflammatory responses, or it can form a complex with STAT which then binds to NF-κB, suppressing the transcription of inflammatory mediators (Kimura, Naka et al. 2009). It has also been reported that a ligand-bound AhR/ARNT complex interacts with hypo-phosphorylated retinoblastoma protein which inhibits the cell cycle transition from G1 to S phase (Puga, Barnes et al. 2000). Through this interaction, cell cycle progression and cellular proliferation are slowed down by AhR in humans and rats (Gargaro, Scalisi et al. 2021). Taken together, these findings suggest that different coregulators of AhR present in different species could promote signaling through various AhR pathways to drive the expression of distinct gene sets.
Tissue-targeted genetic models used to directly investigate the role of AhR in osteoblasts are sparsely reported in the literature. AhR floxed mice crossed with α1(I)-Collagen-Cre were developed to investigate the effects of osteoblast-targeted AhR knockout, but surprisingly these mice presented with a bone phenotype that was comparable to their wildtype littermates when examined at young ages (Yu, Kondo et al. 2014). However, it is worth noting that the detrimental effects of the AhR ligand kynurenine on bone mass are attributed to an aging skeletal phenotype (Refaey, McGee-Lawrence et al. 2017), raising the possibility that the role of AhR in osteoblasts is age dependent. It is surprising that, to our knowledge, the role of AhR in osteocytes (which are terminally differentiated osteoblasts) has not yet been directly tested. Given the importance of osteocytes in the regulation of skeletal mechanobiology, mineral homeostasis, and the regulation of bone remodeling activity, such studies are likely warranted to better understand the overall role of AhR in skeletal biology
6. Role of AhR in osteoclasts
Osteoclasts are derived from hematopoietic stem cells, and several reports have found that genetic deletion of AhR promotes HSC proliferation (Singh, Bennett et al. 2014, Bennett, Singh et al. 2015, Unnisa, Singh et al. 2016) (Figure 2). The literature presents conflicting findings for the role for AhR in osteoclasts (Figure 2). Some studies show that AhR stimulation drives osteoclastogenesis and increases bone resorption (Iqbal, Sun et al. 2013, Eisa, Reddy et al. 2020), while others demonstrate that AhR inhibits osteoclastogenesis (Voronov, Heersche et al. 2005, Jia, Tao et al. 2019). Kynurenine is an endogenous metabolite that activates AhR signaling pathway (Opitz, Litzenburger et al. 2011), and in fact, AhR is the only known receptor for kynurenine thus far, responsible for mediating many of kynurenine’s effects (Eisa, Reddy et al. 2020). Therefore, kynurenine-centric studies may be informative regarding understanding AhR’s effects in osteoclasts. However, careful interpretation of these studies is critical as kynurenine has been shown to modulate cell bioenergetics (Pierce, Roberts et al. 2020), and it is not yet known whether this impact is AhR-dependent. A case-control study on elderly patients investigated relationships between bone marrow kynurenine levels, age, and osteoporosis-related phenotypes (Kim, Hamrick et al. 2019). Interestingly, the results revealed that kynurenine accumulates with age, and that patients with fragility hip fractures had 39.7% higher bone barrow kynurenine levels than patients without hip fractures (Kim, Hamrick et al. 2019). Relevant to osteoclasts, the researchers also analyzed bone marrow samples for markers of osteoclast activity like TRAP-5b and RANKL, finding elevated plasma level of these biochemical molecules and reduced bone mass density at the total femur with increasing age (Kim, Hamrick et al. 2019). These results suggest that elevated kynurenine levels (and by extension activating the AhR) could contribute to increased bone resorption and higher fragility observed within the elderly population (Kim, Hamrick et al. 2019). The negative influence of kynurenine on bone density and architecture was also noted in mice fed kynurenine or intraperitoneally injected with the agent (Refaey, McGee-Lawrence et al. 2017). Markers of osteoclastic activity such as RANKL and pyridinoline cross-links (PYD) were elevated in the serum of kynurenine treated mice, and histological markers of osteoclastic activity and bone marrow adiposity were also increased in the treated animals (Refaey, McGee-Lawrence et al. 2017). These results suggested that kynurenine signaling (likely mediated through the AhR) is involved in promoting osteoclastic resorption and skeletal aging.
On a mechanistic level, Eisa et al. investigated AhR’s role in the molecular pathway for kynurenine’s effect in osteoclasts in vitro, showing that kynurenine acted through the AhR pathway to enhance RANKL-dependent osteoclastogenesis and bone resorption (Eisa, Reddy et al. 2020). Cotreatment of Raw 264.7 cells with kynurenine and RANKL increased the mRNA and protein levels of c-fos and NFATc1, which are critical transcription factors that induce osteoclast differentiation (Eisa, Reddy et al. 2020). The number of TRAP positive cells was also elevated after kynurenine/RANKL treatment (Eisa, Reddy et al. 2020). Pharmacological and genetic blockade of AhR pathway attenuated kynurenine’s effect on osteoclasts, which confirmed the involvement of this cascade in osteoclast biology (Eisa, Reddy et al. 2020). The function of AhR in osteoclasts has been explored in vivo by using systemic and tissue specific AhR knockout mice (Yu, Kondo et al. 2014). When AhR was knocked out systemically (body-wide), mice exhibited greater bone mineral density, trabecular bone connectivity density, trabecular bone volume, and trabecular number compared to wild-type mice (Yu, Kondo et al. 2014). Both males and females with whole-body AhR knockout had fewer osteoclasts than wild-type mice with no changes in mineral apposition rate and bone formation rate, suggesting that skeletal effects of body-wide AhR deletion were primarily mediated through reduced bone resorption (Yu, Kondo et al. 2014). AhR floxed mice were then crossed with Cathepsin K-Cre mice to drive AhR conditional knockout in cells of the osteoclast lineage (Yu, Kondo et al. 2014), which showed that osteoclast-targeted knockout mice had higher bone mass with lower bone resorption (Yu, Kondo et al. 2014).
There is also contrasting evidence in the literature suggesting that AhR signaling inhibits osteoclast activity. For example, at least one report demonstrates that TCDD treatment of juvenile rodents reduced osteoclastic bone resorption (Fader, Nault et al. 2018). Through stimulating the AhR pathway, 3-methylcholanthrene (3MC) repressed the differentiation of osteoclasts via a reducing expression of the pro-osteoclastogenic molecule RANKL expression in osteoblast-lineage ST2 progenitor cells (Naruse, Otsuka et al. 2004). These studies suggested that the AhR pathway might reduce osteoclast differentiation indirectly by affecting osteoclast supporting cells rather than osteoclast precursors (Naruse, Otsuka et al. 2004). However, literature support also exists for the idea that AhR-mediated signaling can directly inhibit osteoclasts. For example, the endogenous AhR ligand FICZ dose-dependently inhibited actin ring formation and pit formation by bone marrow macrophage-derived osteoclasts (Yoshikawa, Izawa et al. 2021). Voronov et al. demonstrated that the AhR agonist BaP also inhibited osteoclast differentiation and bone resorption (Voronov, Heersche et al. 2005, Voronov, Li et al. 2008), attributed to crosstalk between AhR and NF-kB pathways (Voronov, Li et al. 2008). NF-kB is a transcription factor that resides in the cytoplasm in complex with IkBα, p65 and p50 (Oeckinghaus and Ghosh 2009). When NF-kB pathway is stimulated, IKK phosphorylates IkBα and targets it for degradation, allowing NF-kB to translocate to the nucleus and drive gene expression (Oeckinghaus and Ghosh 2009). NF-kB is a common transcription factor for AhR and RANKL signaling cascades, therefore, stimulating AhR creates a competition for NF-kB and limits RANKL induced osteoclastogenesis (Voronov, Li et al. 2008). Although the exact mechanism for the crosstalk between AhR and RANKL is not clearly understood, it was proposed that AhR can complex with p65 and IkBα in the cytoplasm (Voronov, Li et al. 2008). Upon AhR stimulation, AhR could act as a ligand-dependent E3 ubiquitin ligase and activate the IKK complex, driving the translocation of NF-kB to the nucleus and subsequent gene expression (Voronov, Li et al. 2008). All together, these events reduce the availability of NF-kB for RANKL mediated signaling, limiting osteoclastogenesis (Voronov, Li et al. 2008).
7. Role of AhR in cartilage and endochondral ossification
Endochondral bone development begins with a cartilaginous template which osteoblasts and osteoclasts then replace with bone tissue. This process is regulated by various endocrine factors including growth hormone and insulin-like growth factor-1 (IGF-1) (Cedervall, Lind et al. 2015). The AhR has been reported to play a role in cartilage development and subsequent bone growth. For example, AhR expression was suppressed in cartilage templates, but activation of AhR by the endogenous ligand FICZ positively promoted later processes of endochondral ossification (Huang, Wang et al. 2022). Cedervall et al. investigated how AhR acts in growth plate chondrocytes to regulate bone elongation, beginning with the finding that AhR was broadly expressed in the growth plate cartilage of human subjects and more highly expressed in hypertrophic as compared to resting chondrocytes (Cedervall, Lind et al. 2015). For mechanistic studies, fetal rat metatarsal bones were cultured and monitored for longitudinal growth after exposure to AhR modulators (Cedervall, Lind et al. 2015). TCDD did not affect bone growth at the concentrations tested (1 pM −10 nM), but higher dose of the AhR antagonist, α-naphthoflavone, increased chondrocyte apoptosis and suppressed bone growth (Cedervall, Lind et al. 2015). It is worth noting that at lower doses (10 pM and 10 nM), α-naphthoflavone did not affect bone growth and cytotoxic effects may have occurred through AhR independent mechanisms (Cedervall, Lind et al. 2015). The authors therefore concluded that AhR activation may not directly affect endochondral bone growth, and that AhR activation in the rat growth plate may require an unknown co-factor to affect cartilage formation (Cedervall, Lind et al. 2015). Yang et al. demonstrated that rabbit chondrocytes were exposed to TCDD at a concentration of 10 nM developed elevated levels of reactive oxygen species and nitric oxide (Yang and Lee 2010). This created cellular stress which eventually triggered chondrocyte apoptosis (Yang and Lee 2010). Inhibiting AhR using 10 μM of α-naphthoflavone attenuated these harmful effects of TCDD (Yang and Lee 2010), raising the possibility that apoptosis of chondrocytes driven by AhR activation could lead to cartilage damage and arthritis development (Yang and Lee 2010). In a parallel study, treating Japanese rice fish (medaka) with TCDD (ppt-ppb concentrations) impaired chondrogenesis and osteogenesis (Dong, Hinton et al. 2012). The recruitment of MSCs to hypural cartilage anlage was attenuated following TCDD treatment, and chondrocyte proliferation and differentiation were also impaired in a dose-dependent manner (Dong, Hinton et al. 2012). On a molecular level, the expression of type 2 collagen (a major cartilage extracellular matrix protein) was markedly blunted after TCDD treatment (Dong, Hinton et al. 2012). This effect was mediated through downregulating the key chondrogenic transcription factor, SRY-box transcription factor 9 (SOX9) (Dong, Hinton et al. 2012). With regards to subsequent bone formation, TCDD treatment attenuated Osterix expression and reduced perichondral ossification within the fish hypural anlage (Dong, Hinton et al. 2012). Mechanisms of endochondral ossification are initiated postnatally during fracture repair, and, when administered to rats with tibial fractures, the AhR ligand BaP delayed healing and resulted in a less mineralized callus (Zhou, Jiang et al. 2017). Together, these results imply that AhR activation may negatively influence cartilage and endochondral bone formation.
In addition to AhR’s role in cartilage during developmental and repair-mediated processes of endochondral ossification, AhR may also play a role in diseases affecting cartilage like osteoarthritis. Expression of AhR was up-regulated in human osteoarthritic cartilage as compared to intact controls (Klinger, Beyer et al. 2013), and was also highly expressed in meniscus and subchondral bone from osteoarthritic patients (Chang, Yao et al. 2021). The AhR signaling pathway was reported to be more active in osteoarthritic synovial tissues as compared to those from rheumatoid arthritis (Ogando, Tardaguila et al. 2016). As with other cell types described above, the effects of AhR-mediated signaling in chondrocytes with respect to osteoarthritis phenotypes are likely ligand dependent. Exposure to the exogenous AhR ligand BaP led to the development of osteoarthritis-like lesions, loss of proteoglycans, and reduced expression of aggrecan, type 2 collagen, and Sox-9 in the articular cartilage of wildtype (but not AhR-knockout) mice (Yoshikawa, Izawa et al. 2021). Interestingly, however, administration of the endogenous AhR ligand FICZ was protective against the development of mechanically induced osteoarthritis in mice (Yoshikawa, Izawa et al. 2021). Mechanistically, BaP promoted expression of the active forms of caspase-3 and caspase-9 in ATCD5 chondrocyte progenitor cells, and increased expression of cleaved caspase-3 in the mandibular chondrocytes of wildtype (but not AhR-knockout) mice, but administration of FICZ reduced metrics of apoptosis (Yoshikawa, Izawa et al. 2021). The beneficial effects of FICZ described above are consistent with a recent bioRxiv report, where intra-articular delivery of an indoleamine 2,3-dioxygenase and galectin-3 fusion protein (IDO-Gal3), intended to drive local expression of the endogenous AhR ligand kynurenine, improved functional outcomes in a rat surgical model of osteoarthritis (Partain, Bracho-Sanchez et al. 2021).
8. Sexually dimorphic effects of AhR in bone
Crosstalk between signaling cascades is a common phenomenon driving biological events (Vert and Chory 2011). Some evidence of sexually dimorphic effects of AhR have been reported in bone, which may be due in part to the interaction between AhR and estrogen receptors (ER) (Wejheden, Brunnberg et al. 2010, Tarnow, Bross et al. 2017). Wejhedem et al. designed a study to explore the long-term effect of exposing mice to AhR ligands (Wejheden, Brunnberg et al. 2010). As their experimental model, they studied three-month-old transgenic mice expressing constitutively active AhR. Results revealed that continuous AhR stimulation reduced bone formation in transgenic female mice but not in males (Wejheden, Brunnberg et al. 2010). Bone dimensions, density, and content were notably altered in female mice, while males were minimally affected (Wejheden, Brunnberg et al. 2010). Osteoclastic markers such as carboxy-terminal cross-linked telopeptide of type 1 collagen (CTX-1) and osteoclast volume density were increased by more than 60% and the resorption index of CTX-1/TRAP 5b was increased by 90% in females but not in males (Wejheden, Brunnberg et al. 2010). On an mRNA level, female but not male mice demonstrated altered expression of osteoclastic and osteoblastic markers in these studies (Wejheden, Brunnberg et al. 2010). Similarly, treatment with the endogenous AhR ligand kynurenine was reported to decrease osteoblasts in young female but not young male mice, and likewise impaired cellular energetics in BMSC-derived osteoblasts from females but not males (Pierce, Roberts et al. 2020). Another report demonstrated that TCDD treatment exhibited gender dependent effect in C57BL/6J mice (Herlin, Finnila et al. 2013). For instance, bone biomechanical properties were decreased in females following TCDD treatment, but males were not affected (Herlin, Finnila et al. 2013). Also, unlike males, TCDD treated females demonstrated unbalanced bone remodeling activity as indicated by lower PINP/CTX-1 ratio (Herlin, Finnila et al. 2013). In the same report, bone phenotype of both genders was compared between AhR knockout mice and wild-type mice, showing that AhR deletion resulted in higher trabecular number and trabecular bone volume fraction in both males and females (Herlin, Finnila et al. 2013). However, female knockout mice had lower cortical area, cortical thickness and bone mineral density compared to wild-type females (Herlin, Finnila et al. 2013), whereas male knockout mice displayed lower plasticity and harder cortical bone matrix than their wild-type controls (Herlin, Finnila et al. 2013). In another report, eight-week-old osteoclast-targeted AhR knockout mice were orchiectomized or ovariectomized then monitored for four weeks, showing that the osteoclast-targeted AhR knockout mice were resistant to bone loss induced by gonadectomy (Yu, Kondo et al. 2014). This implied that sex hormones might influence bone remodeling through AhR.
Multiple mechanisms have been proposed for the crosstalk between AhR and ER (Tarnow, Bross et al. 2017). Firstly, AhR could directly interact with ERα to form a complex which binds to AhR target genes (Matthews and Gustafsson 2006). Alternatively, the complex might bind to ERE and inhibit ER target gene expression (Jackson, Joshi et al. 2015) . The competition for shared co-factors between the two signaling cascades also influences the outcome of AhR activation (Swedenborg and Pongratz 2010). Moreover, AhR could increase proteasomal degradation of ER through upregulating CYP expression and estrogen metabolism (Wormke, Stoner et al. 2003). Estrogen has also been reported to alter kynurenine pathway by inhibiting kynurenine aminotransferase enzymes (Jayawickrama, Nematollahi et al. 2017). All together, these mechanisms suggest that AhR stimulation might exert antiestrogenic effects in bone (Wejheden, Brunnberg et al. 2010). Since estrogen loss usually increases bone resorption, the crosstalk between AhR and ER could contribute to the higher sensitivity of female mice to AhR activation (Wejheden, Brunnberg et al. 2010).
9. Conclusions
Taken together, the literature suggests that signaling through AhR likely plays an important role in skeletal homeostasis. Several reports have shown that activating AhR reduces osteoblast differentiation and activity (Korkalainen, Kallio et al. 2009, Tong, Niu et al. 2017, Zhou, Jiang et al. 2017, Watson, Nordberg et al. 2019), and that treating MSCs with AhR agonists suppressed osteoblastic markers (Siddiqui and Partridge 2016, Refaey, McGee-Lawrence et al. 2017, Tong, Niu et al. 2017). Additionally, AhR stimulation increased bone porosity and reduced matrix mineralization in vivo (Herlin, Finnila et al. 2013). However, as mentioned above, some AhR ligands like FICZ appear to have a beneficial effect on osteoblasts (Yoshikawa, Izawa et al. 2021). Similarly, the role of AhR in osteoclasts is still unclear, as some support exists for the idea that AhR activation drives osteoclastogenesis (Iqbal, Sun et al. 2013, Kim, Hamrick et al. 2019, Eisa, Reddy et al. 2020), whereas other research groups showed that AhR stimulation inhibits osteoclastogenesis (Naruse, Otsuka et al. 2004, Voronov, Li et al. 2008). The inconsistent findings suggest that the resultant effect of stimulating AhR likely depends on the specific ligand used, the ligand’s concentration, and duration of treatment (Park, Madhavaram et al. 2020, Safe, Jin et al. 2020). Indeed, these factors could act through different AhR signaling pathways (e.g., canonical, non-canonical, or non-genomic) to influence distinct downstream signaling proteins (Eisa, Reddy et al. 2020).
While the effect of AhR on muscle is beyond the scope of this review, the importance of bone-muscle crosstalk has recently come to light and is an area of increasing interest; in this context, it is important to note that AhR activation can also lead to muscle atrophy (Kaiser, Yu et al. 2019, He, He et al. 2020, Thome, Miguez et al. 2022). In fact, a recent publication from Thome et al demonstrated that muscle atrophy due to overexpression of AhR could be rescued via AhR antagonism (Thome, Miguez et al. 2022). However, the role of AhR in mechanisms of bone-muscle crosstalk has yet to be clearly defined, and since AhR modulation exhibits context-dependent effects, therapeutically targeting this pathway to combat musculoskeletal frailty in aging might require personalizing the treatment plan according to a variety of patient-specific and environmental factors such as gender, age, and lifestyle habits.
While much has been learned about the role of AhR in the skeleton, much remains to be explored. Many of the studies described above have utilized pharmacological inhibitors of AhR that are likely to have off-targeted effects, and many of the ligand-based studies have employed environmental pollutant AhR ligands like TCDD and BaP. To address this issue, the specific role of AhR in bone cells should be tested in future in vitro studies employing knockdown mediated by sequence-specific small interfering RNA (siRNA), small hairpin RNA (shRNA), or CRISPR/Cas9-mediated knockouts of AhR, along with creation of novel cell type-specific in vivo knockout mouse models of the AhR. Moreover, it will be critical to ascertain the contributions of endogenous AhR ligands (like kynurenine and other tryptophan metabolites) to skeletal biology, particularly in the context of aging. Although the literature demonstrates that kynurenine activates AhR, the precise mechanism is not yet understood. For example, it is not clear whether kynurenine and/or other tryptophan metabolites activate canonical, non-canonical or non-genomic modes of AhR signaling, and the specific signaling molecules and enzymes involved in the effect of kynurenine and other tryptophan metabolites on bone cells are not well defined. It has been recognized that the AhR cascade undergoes important crosstalk with other nuclear hormone receptors such as ER and the glucocorticoid receptor (Widerak, Ghoneim et al. 2006, Monostory, Pascussi et al. 2009, Denison, Soshilov et al. 2011). Among those, the crosstalk between AhR and ER is the best understood, while other interactions are less studied (Denison, Soshilov et al. 2011). Future studies focused on elucidating such molecular interactions would foster better understanding of the role of AhR in musculoskeletal disorders, ultimately promoting the discovery of new, effective therapeutic skeletal agents
Funding
The authors are supported by funding provided by the National Institute on Aging (NIA P01 AG036675 and R01 AG 067510). The contents of this publication do not represent the views of the Department of Veterans Affairs or the United States Government.
Abbreviations
- 3’4’DMF
3',4'-dimethoxyflavone
- 3MC
3-methylcholanthrene
- AhR
Aryl hydrocarbon receptor
- AHRR
Aryl hydrocarbon receptor repressor
- ARA9
Aryl hydrocarbon receptor associated 9
- ARNT
Aryl hydrocarbon receptor nuclear translocator
- BaP
Benzo(a)pyrene
- CEBPβ
CCAAT/enhancer-binding protein beta
- COX2
Cyclooxygenase 2
- CTX-1
Carboxy-terminal cross-linked telopeptide of type 1 collagen
- CYP1A1
Cytochrome P450 family 1 subfamily A member 1
- CYP1B1
Cytochrome P450 family 1 subfamily B member 1
- DMBA
7,12-dimethylbenz [a]anthracene
- ER
Estrogen receptor
- ERE
Estrogen response element
- FGF9
Fibroblast growth factor 9
- FICZ
6-Formylindolo[3,2-b] carbazole
- HSC
Hematopoietic stem cells
- Hsp90
Heat shock protein 90
- IDO
Indolamine-2,3-dioxygenase
- IGF-1
Insulin-like growth factor-1
- IL-6
Interleukin-6
- KLF6
Kruppel-like factor 6
- KYN
Kynurenine
- LAT1
Large amino acid transporter 1
- m-CSF
Macrophage colony stimulating factor
- MMP
Matrix metalloproteinases
- MSC
Mesenchymal stem cells
- NC-XRE
Non-consensus xenobiotic response element
- OPG
Osteoprotegerin
- PCB126
3,3′,4,4′,5-Pentachlorobiphenyl
- PPARγ
Peroxisome proliferator-activated receptor γ
- RANKL
Receptor activator of nuclear factor kappa-B ligand
- RUNX2
Runt-related transcription factor 2
- SLC7A5
Solute carrier transporter 7a5
- SOX9
SRY-Box Transcription Factor 9
- STAT
Signal transducers and activator of transcription
- TCDD
2,3,7,8-tetrachlorodibenzo-p-dioxin
- TGF-β
Transforming growth factor-β
- TLR
Toll-like receptor
- TRAP
Tartrate resistant acid phosphatase
- Trp
Tryptophan
- VEGF
Vascular endothelial growth factor
- XRE
Xenobiotic response element
Footnotes
Declaration of Interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported in this review.
References
- Abney KK and Galipeau J (2021). "Aryl hydrocarbon receptor in mesenchymal stromal cells: new frontiers in AhR biology." FEBS J 288(13): 3962–3972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al-Bashaireh AM, Haddad LG, Weaver M, Chengguo X, Kelly DL and Yoon S (2018). "The Effect of Tobacco Smoking on Bone Mass: An Overview of Pathophysiologic Mechanisms." J Osteoporos 2018: 1206235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amenya HZ, Tohyama C and Ohsako S (2016). "Dioxin induces Ahr-dependent robust DNA demethylation of the Cyp1a1 promoter via Tdg in the mouse liver." Sci Rep 6: 34989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- An L, Shi Q, Fan M, Huang G, Zhu M, Zhang M, Liu Y and Weng Y (2020). "Benzo[a]pyrene injures BMP2-induced osteogenic differentiation of mesenchymal stem cells through AhR reducing BMPRII." Ecotoxicol Environ Saf 203: 110930. [DOI] [PubMed] [Google Scholar]
- Arinze NV, Yin W, Lotfollahzadeh S, Napoleon MA, Richards S, Walker JA, Belghasem M, Ravid JD, Hassan Kamel M, Whelan SA, Lee N, Siracuse JJ, Anderson S, Farber A, Sherr D, Francis J, Hamburg NM, Rahimi N and Chitalia VC (2022). "Tryptophan metabolites suppress the Wnt pathway and promote adverse limb events in chronic kidney disease." J Clin Invest 132(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Assis-Ribas T, Forni MF, Winnischofer SMB, Sogayar MC and Trombetta-Lima M (2018). "Extracellular matrix dynamics during mesenchymal stem cells differentiation." Dev Biol 437(2): 63–74. [DOI] [PubMed] [Google Scholar]
- Beischlag TV, Luis Morales J, Hollingshead BD and Perdew GH (2008). "The aryl hydrocarbon receptor complex and the control of gene expression." Crit Rev Eukaryot Gene Expr 18(3): 207–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bennett JA, Singh KP, Unnisa Z, Welle SL and Gasiewicz TA (2015). "Deficiency in Aryl Hydrocarbon Receptor (AHR) Expression throughout Aging Alters Gene Expression Profiles in Murine Long-Term Hematopoietic Stem Cells." PLoS One 10(7): e0133791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boyle WJ, Simonet WS and Lacey DL (2003). "Osteoclast differentiation and activation." Nature 423(6937): 337–342. [DOI] [PubMed] [Google Scholar]
- Carambia A and Schuran FA (2021). "The aryl hydrocarbon receptor in liver inflammation." Semin Immunopathol 43(4): 563–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cedervall T, Lind PM and Savendahl L (2015). "Expression of the aryl hydrocarbon receptor in growth plate cartilage and the impact of its local modulation on longitudinal bone growth." Int J Mol Sci 16(4): 8059–8069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang L, Yao H, Yao Z, Ho KK, Ong MT, Dai B, Tong W, Xu J and Qin L (2021). "Comprehensive Analysis of Key Genes, Signaling Pathways and miRNAs in Human Knee Osteoarthritis: Based on Bioinformatics." Front Pharmacol 12: 730587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cui Z, Feng Y, Li D, Li T, Gao P and Xu T (2020). "Activation of aryl hydrocarbon receptor (AhR) in mesenchymal stem cells modulates macrophage polarization in asthma." J Immunotoxicol 17(1): 21–30. [DOI] [PubMed] [Google Scholar]
- Davarinos NA and Pollenz RS (1999). "Aryl hydrocarbon receptor imported into the nucleus following ligand binding is rapidly degraded via the cytosplasmic proteasome following nuclear export." J Biol Chem 274(40): 28708–28715. [DOI] [PubMed] [Google Scholar]
- de Almeida DC, Evangelista LSM and Câmara NOS (2017). "Role of aryl hydrocarbon receptor in mesenchymal stromal cell activation: A minireview." World J Stem Cells 9(9): 152–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denison MS and Nagy SR (2003). "Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals." Annu Rev Pharmacol Toxicol 43: 309–334. [DOI] [PubMed] [Google Scholar]
- Denison MS, Soshilov AA, He G, DeGroot DE and Zhao B (2011). "Exactly the same but different: promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor." Toxicol Sci 124(1): 1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DiNatale BC, Murray IA, Schroeder JC, Flaveny CA, Lahoti TS, Laurenzana EM, Omiecinski CJ and Perdew GH (2010). "Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling." Toxicol Sci 115(1): 89–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Disner GR, Lopes-Ferreira M and Lima C (2021). "Where the Aryl Hydrocarbon Receptor Meets the microRNAs: Literature Review of the Last 10 Years." Front Mol Biosci 8: 725044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong W, Hinton DE and Kullman SW (2012). "TCDD disrupts hypural skeletogenesis during medaka embryonic development." Toxicol Sci 125(1): 91–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DuSell CD, Nelson ER, Wittmann BM, Fretz JA, Kazmin D, Thomas RS, Pike JW and McDonnell DP (2010). "Regulation of aryl hydrocarbon receptor function by selective estrogen receptor modulators." Mol Endocrinol 24(1): 33–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eisa NH, Reddy SV, Elmansi AM, Kondrikova G, Kondrikov D, Shi XM, Novince CM, Hamrick MW, McGee-Lawrence ME, Isales CM, Fulzele S and Hill WD (2020). "Kynurenine Promotes RANKL-Induced Osteoclastogenesis In Vitro by Activating the Aryl Hydrocarbon Receptor Pathway." Int J Mol Sci 21(21). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elmansi AM, Hussein KA, Herrero SM, Periyasamy-Thandavan S, Aguilar-Perez A, Kondrikova G, Kondrikov D, Eisa NH, Pierce JL, Kaiser H, Ding KH, Walker AL, Jiang X, Bollag WB, Elsalanty M, Zhong Q, Shi XM, Su Y, Johnson M, Hunter M, Reitman C, Volkman BF, Hamrick MW, Isales CM, Fulzele S, McGee-Lawrence ME and Hill WD (2020). "Age-related increase of kynurenine enhances miR29b-1-5p to decrease both CXCL12 signaling and the epigenetic enzyme Hdac3 in bone marrow stromal cells." Bone Rep 12: 100270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fader KA, Nault R, Raehtz S, McCabe LR and Zacharewski TR (2018). "2,3,7,8-Tetrachlorodibenzo-p-dioxin dose-dependently increases bone mass and decreases marrow adiposity in juvenile mice." Toxicol Appl Pharmacol 348: 85–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gargaro M, Scalisi G, Manni G, Mondanelli G, Grohmann U and Fallarino F (2021). "The Landscape of AhR Regulators and Coregulators to Fine-Tune AhR Functions." Int J Mol Sci 22(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Girer NG, Murray IA, Omiecinski CJ and Perdew GH (2016). "Hepatic Aryl Hydrocarbon Receptor Attenuates Fibroblast Growth Factor 21 Expression." J Biol Chem 291(29): 15378–15387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomez-Duran A, Ballestar E, Carvajal-Gonzalez JM, Marlowe JL, Puga A, Esteller M and Fernandez-Salguero PM (2008). "Recruitment of CREB1 and histone deacetylase 2 (HDAC2) to the mouse Ltbp-1 promoter regulates its constitutive expression in a dioxin receptor-dependent manner." J Mol Biol 380(1): 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gramatzki D, Pantazis G, Schittenhelm J, Tabatabai G, Köhle C, Wick W, Schwarz M, Weller M and Tritschler I (2009). "Aryl hydrocarbon receptor inhibition downregulates the TGF-beta/Smad pathway in human glioblastoma cells." Oncogene 28(28): 2593–2605. [DOI] [PubMed] [Google Scholar]
- Hallett SA, Ono W and Ono N (2021). "The hypertrophic chondrocyte: To be or not to be." Histol Histopathol 36(10): 1021–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He C, He W, Hou J, Chen K, Huang M, Yang M, Luo X and Li C (2020). "Bone and Muscle Crosstalk in Aging." Front Cell Dev Biol 8: 585644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herlin M, Finnila MA, Zioupos P, Aula A, Risteli J, Miettinen HM, Jamsa T, Tuukkanen J, Korkalainen M, Hakansson H and Viluksela M (2013). "New insights to the role of aryl hydrocarbon receptor in bone phenotype and in dioxin-induced modulation of bone microarchitecture and material properties." Toxicol Appl Pharmacol 273(1): 219–226. [DOI] [PubMed] [Google Scholar]
- Huang G and Elferink CJ (2012). "A novel nonconsensus xenobiotic response element capable of mediating aryl hydrocarbon receptor-dependent gene expression." Mol Pharmacol 81(3): 338–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang J, Wang Y and Zhou Y (2022). "Beneficial roles of the AhR ligand FICZ on the regenerative potentials of BMSCs and primed cartilage templates." RSC Adv 12(18): 11505–11516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hyun JJ, Chun HJ, Keum B, Seo YS, Kim YS, Jeen YT, Lee HS, Um SH, Kim CD, Ryu HS, Kim SG and Jung WW (2010). "Effect of omeprazole on the expression of transcription factors in osteoclasts and osteoblasts." Int J Mol Med 26(6): 877–883. [DOI] [PubMed] [Google Scholar]
- Iqbal J, Sun L, Cao J, Yuen T, Lu P, Bab I, Leu NA, Srinivasan S, Wagage S, Hunter CA, Nebert DW, Zaidi M and Avadhani NG (2013). "Smoke carcinogens cause bone loss through the aryl hydrocarbon receptor and induction of Cyp1 enzymes." Proc Natl Acad Sci U S A 110(27): 11115–11120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jackson DP, Joshi AD and Elferink CJ (2015). "Ah Receptor Pathway Intricacies; Signaling Through Diverse Protein Partners and DNA-Motifs." Toxicol Res (Camb) 4(5): 1143–1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jackson DP, Li H, Mitchell KA, Joshi AD and Elferink CJ (2014). "Ah receptor-mediated suppression of liver regeneration through NC-XRE-driven p21Cip1 expression." Mol Pharmacol 85(4): 533–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jayawickrama GS, Nematollahi A, Sun G, Gorrell MD and Church WB (2017). "Inhibition of human kynurenine aminotransferase isozymes by estrogen and its derivatives." Sci Rep 7(1): 17559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jensen BA, Leeman RJ, Schlezinger JJ and Sherr DH (2003). "Aryl hydrocarbon receptor (AhR) agonists suppress interleukin-6 expression by bone marrow stromal cells: an immunotoxicology study." Environ Health 2(1): 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia Y, Tao Y, Lv C, Xia Y, Wei Z and Dai Y (2019). "Tetrandrine enhances the ubiquitination and degradation of Syk through an AhR-c-src-c-Cbl pathway and consequently inhibits osteoclastogenesis and bone destruction in arthritis." Cell Death Dis 10(2): 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia Y, Zhao Y, Zhang Z, Shi L, Fang Y and Chang C (2021). "Aryl hydrocarbon receptor signaling pathway plays important roles in the proliferative and metabolic properties of bone marrow mesenchymal stromal cells." Acta Biochim Biophys Sin (Shanghai) 53(11): 1428–1439. [DOI] [PubMed] [Google Scholar]
- Jin UH, Lee SO, Pfent C and Safe S (2014). "The aryl hydrocarbon receptor ligand omeprazole inhibits breast cancer cell invasion and metastasis." BMC Cancer 14: 498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kado S, Chang WLW, Chi AN, Wolny M, Shepherd DM and Vogel CFA (2017). "Aryl hydrocarbon receptor signaling modifies Toll-like receptor-regulated responses in human dendritic cells." Arch Toxicol 91(5): 2209–2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaiser H, Yu K, Pandya C, Mendhe B, Isales CM, McGee-Lawrence ME, Johnson M, Fulzele S and Hamrick MW (2019). "Kynurenine, a Tryptophan Metabolite That Increases with Age, Induces Muscle Atrophy and Lipid Peroxidation." Oxid Med Cell Longev 2019: 9894238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karchner SI, Franks DG, Kennedy SW and Hahn ME (2006). "The molecular basis for differential dioxin sensitivity in birds: role of the aryl hydrocarbon receptor." Proc Natl Acad Sci U S A 103(16): 6252–6257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kido S, Fujihara M, Nomura K, Sasaki S, Mukai R, Ohnishi R, Kaneko I, Segawa H, Tatsumi S, Izumi H, Kohno K and Miyamoto K (2014). "Molecular mechanisms of cadmium-induced fibroblast growth factor 23 upregulation in osteoblast-like cells." Toxicol Sci 139(2): 301–316. [DOI] [PubMed] [Google Scholar]
- Kim BJ, Hamrick MW, Yoo HJ, Lee SH, Kim SJ, Koh JM and Isales CM (2019). "The Detrimental Effects of Kynurenine, a Tryptophan Metabolite, on Human Bone Metabolism." J Clin Endocrinol Metab 104(6): 2334–2342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimura A, Naka T, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y and Kishimoto T (2009). "Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses." J Exp Med 206(9): 2027–2035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- King-Heiden TC, Mehta V, Xiong KM, Lanham KA, Antkiewicz DS, Ganser A, Heideman W and Peterson RE (2012). "Reproductive and developmental toxicity of dioxin in fish." Mol Cell Endocrinol 354(1-2): 121–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klinger P, Beyer C, Ekici AB, Carl HD, Schett G, Swoboda B, Hennig FF and Gelse K (2013). "The Transient Chondrocyte Phenotype in Human Osteophytic Cartilage: A Role of Pigment Epithelium-Derived Factor?" Cartilage 4(3): 249–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M, Sato M, Okamoto R, Kitamura Y, Yoshiki S and Kishimoto T (1997). "Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts." Cell 89(5): 755–764. [DOI] [PubMed] [Google Scholar]
- Kondrikov D, Elmansi A, Bragg RT, Mobley T, Barrett T, Eisa N, Kondrikova G, Schoeinlein P, Aguilar-Perez A, Shi XM, Fulzele S, Lawrence MM, Hamrick M, Isales C and Hill W (2020). "Kynurenine inhibits autophagy and promotes senescence in aged bone marrow mesenchymal stem cells through the aryl hydrocarbon receptor pathway." Exp Gerontol 130: 110805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korkalainen M, Kallio E, Olkku A, Nelo K, Ilvesaro J, Tuukkanen J, Mahonen A and Viluksela M (2009). "Dioxins interfere with differentiation of osteoblasts and osteoclasts." Bone 44(6): 1134–1142. [DOI] [PubMed] [Google Scholar]
- Kransler KM, McGarrigle BP and Olson JR (2007). "Comparative developmental toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the hamster, rat and guinea pig." Toxicology 229(3): 214–225. [DOI] [PubMed] [Google Scholar]
- Langdahl B, Ferrari S and Dempster DW (2016). "Bone modeling and remodeling: potential as therapeutic targets for the treatment of osteoporosis." Ther Adv Musculoskelet Dis 8(6): 225–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lanis JM, Alexeev EE, Curtis VF, Kitzenberg DA, Kao DJ, Battista KD, Gerich ME, Glover LE, Kominsky DJ and Colgan SP (2017). "Tryptophan metabolite activation of the aryl hydrocarbon receptor regulates IL-10 receptor expression on intestinal epithelia." Mucosal Immunol 10(5): 1133–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larigot L, Juricek L, Dairou J and Coumoul X (2018). "AhR signaling pathways and regulatory functions." Biochim Open 7: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee HG and Yang JH (2012). "PCB126 induces apoptosis of chondrocytes via ROS-dependent pathways." Osteoarthritis Cartilage 20(10): 1179–1185. [DOI] [PubMed] [Google Scholar]
- Liang C, Li J, Lu C, Xie D, Liu J, Zhong C, Wu X, Dai R, Zhang H, Guan D, Guo B, He B, Li F, He X, Zhang W, Zhang BT, Zhang G and Lu A (2020). "Author Correction: HIF1alpha inhibition facilitates Leflunomide-AHR-CRP signaling to attenuate bone erosion in CRP-aberrant rheumatoid arthritis." Nat Commun 11(1): 3098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu C, Guan H, Cai C, Li F and Xiao J (2017). "Lipoxin A4 suppresses osteoclastogenesis in RAW264.7 cells and prevents ovariectomy-induced bone loss." Exp Cell Res 352(2): 293–303. [DOI] [PubMed] [Google Scholar]
- Lu Z, Liu R, Wang Y, Jiao M, Li Z, Wang Z, Huang C, Shi G, Ke A, Wang L, Fu Y, Xia J, Wen H, Zhou J, Wang X, Ye D, Fan J, Chu Y and Cai J (2022). "TET2 Inactivation Restrains IL-10-producing Regulatory B cells to Enable Anti-tumor Immunity in Hepatocellular Carcinoma." Hepatology. [DOI] [PubMed] [Google Scholar]
- Lv Q, Wang K, Qiao S, Yang L, Xin Y, Dai Y and Wei Z (2018). "Norisoboldine, a natural AhR agonist, promotes Treg differentiation and attenuates colitis via targeting glycolysis and subsequent NAD(+)/SIRT1/SUV39H1/H3K9me3 signaling pathway." Cell Death Dis 9(3): 258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marlowe JL, Knudsen ES, Schwemberger S and Puga A (2004). "The aryl hydrocarbon receptor displaces p300 from E2F-dependent promoters and represses S phase-specific gene expression." J Biol Chem 279(28): 29013–29022. [DOI] [PubMed] [Google Scholar]
- Matthews J and Gustafsson JA (2006). "Estrogen receptor and aryl hydrocarbon receptor signaling pathways." Nucl Recept Signal 4: e016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McMillan BJ and Bradfield CA (2007). "The aryl hydrocarbon receptor sans xenobiotics: endogenous function in genetic model systems." Mol Pharmacol 72(3): 487–498. [DOI] [PubMed] [Google Scholar]
- Mimura J, Ema M, Sogawa K and Fujii-Kuriyama Y (1999). "Identification of a novel mechanism of regulation of Ah (dioxin) receptor function." Genes Dev 13(1): 20–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monostory K, Pascussi JM, Kobori L and Dvorak Z (2009). "Hormonal regulation of CYP1A expression." Drug Metab Rev 41(4): 547–572. [DOI] [PubMed] [Google Scholar]
- Mun SH, Park PSU and Park-Min KH (2020). "The M-CSF receptor in osteoclasts and beyond." Exp Mol Med 52(8): 1239–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murshed M (2018). "Mechanism of Bone Mineralization." Cold Spring Harb Perspect Med 8(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Napolitano L, Scalise M, Galluccio M, Pochini L, Albanese LM and Indiveri C (2015). "LAT1 is the transport competent unit of the LAT1/CD98 heterodimeric amino acid transporter." Int J Biochem Cell Biol 67: 25–33. [DOI] [PubMed] [Google Scholar]
- Naruse M, Otsuka E, Naruse M, Ishihara Y, Miyagawa-Tomita S and Hagiwara H (2004). "Inhibition of osteoclast formation by 3-methylcholanthrene, a ligand for arylhydrocarbon receptor: suppression of osteoclast differentiation factor in osteogenic cells." Biochem Pharmacol 67(1): 119–127. [DOI] [PubMed] [Google Scholar]
- Nieto C, Rayo I, de Las Casas-Engel M, Izquierdo E, Alonso B, Bechade C, Maroteaux L, Vega MA and Corbi AL (2020). "Serotonin (5-HT) Shapes the Macrophage Gene Profile through the 5-HT2B-Dependent Activation of the Aryl Hydrocarbon Receptor." J Immunol 204(10): 2808–2817. [DOI] [PubMed] [Google Scholar]
- Oberg M, Bergander L, Hakansson H, Rannug U and Rannug A (2005). "Identification of the tryptophan photoproduct 6-formylindolo[3,2-b]carbazole, in cell culture medium, as a factor that controls the background aryl hydrocarbon receptor activity." Toxicol Sci 85(2): 935–943. [DOI] [PubMed] [Google Scholar]
- Oeckinghaus A and Ghosh S (2009). "The NF-kappaB family of transcription factors and its regulation." Cold Spring Harb Perspect Biol 1(4): a000034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogando J, Tardaguila M, Diaz-Alderete A, Usategui A, Miranda-Ramos V, Martinez-Herrera DJ, de la Fuente L, Garcia-Leon MJ, Moreno MC, Escudero S, Canete JD, Toribio ML, Cases I, Pascual-Montano A, Pablos JL and Manes S (2016). "Notch-regulated miR-223 targets the aryl hydrocarbon receptor pathway and increases cytokine production in macrophages from rheumatoid arthritis patients." Sci Rep 6: 20223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohtake F, Takeyama K, Matsumoto T, Kitagawa H, Yamamoto Y, Nohara K, Tohyama C, Krust A, Mimura J, Chambon P, Yanagisawa J, Fujii-Kuriyama Y and Kato S (2003). "Modulation of oestrogen receptor signalling by association with the activated dioxin receptor." Nature 423(6939): 545–550. [DOI] [PubMed] [Google Scholar]
- Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, Schumacher T, Jestaedt L, Schrenk D, Weller M, Jugold M, Guillemin GJ, Miller CL, Lutz C, Radlwimmer B, Lehmann I, von Deimling A, Wick W and Platten M (2011). "An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor." Nature 478(7368): 197–203. [DOI] [PubMed] [Google Scholar]
- Ou-Yang HF, Huang Y, Hu XB and Wu CG (2011). "Suppression of allergic airway inflammation in a mouse model of asthma by exogenous mesenchymal stem cells." Exp Biol Med (Maywood) 236(12): 1461–1467. [DOI] [PubMed] [Google Scholar]
- Pallotta MT, Fallarino F, Matino D, Macchiarulo A and Orabona C (2014). "AhR-Mediated, Non-Genomic Modulation of IDO1 Function." Front Immunol 5: 497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pandini A, Denison MS, Song Y, Soshilov AA and Bonati L (2007). "Structural and functional characterization of the aryl hydrocarbon receptor ligand binding domain by homology modeling and mutational analysis." Biochemistry 46(3): 696–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park R, Madhavaram S and Ji JD (2020). "The Role of Aryl-Hydrocarbon Receptor (AhR) in Osteoclast Differentiation and Function." Cells 9(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Partain BD, Bracho-Sanchez E, Farhadi SA, Yarmola EG, Keselowsky BG, Hudalla GA and Allen KD (2021). "Intra-Articular Delivery of an Indoleamine 2,3-Dioxygenase Galectin-3 Fusion Protein for Osteoarthritis Treatmetn in Male Lewis Rats." bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peterson RE, Theobald HM and Kimmel GL (1993). "Developmental and reproductive toxicity of dioxins and related compounds: cross-species comparisons." Crit Rev Toxicol 23(3): 283–335. [DOI] [PubMed] [Google Scholar]
- Pierce JL, Roberts RL, Yu K, Kendall RK, Kaiser H, Davis C, Johnson MH, Hill WD, Isales CM, Bollag WB, Hamrick MW and McGee-Lawrence ME (2020). "Kynurenine suppresses osteoblastic cell energetics in vitro and osteoblast numbers in vivo." Exp Gerontol 130: 110818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plotkin LI and Bruzzaniti A (2019). "Molecular signaling in bone cells: Regulation of cell differentiation and survival." Adv Protein Chem Struct Biol 116: 237–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Podechard N, Fardel O, Corolleur M, Bernard M and Lecureur V (2009). "Inhibition of human mesenchymal stem cell-derived adipogenesis by the environmental contaminant benzo(a)pyrene." Toxicol In Vitro 23(6): 1139–1144. [DOI] [PubMed] [Google Scholar]
- Poland A, Palen D and Glover E (1994). "Analysis of the four alleles of the murine aryl hydrocarbon receptor." Mol Pharmacol 46(5): 915–921. [PubMed] [Google Scholar]
- Puga A, Barnes SJ, Dalton TP, Chang C, Knudsen ES and Maier MA (2000). "Aromatic hydrocarbon receptor interaction with the retinoblastoma protein potentiates repression of E2F-dependent transcription and cell cycle arrest." J Biol Chem 275(4): 2943–2950. [DOI] [PubMed] [Google Scholar]
- Ramadoss P, Marcus C and Perdew GH (2005). "Role of the aryl hydrocarbon receptor in drug metabolism." Expert Opin Drug Metab Toxicol 1(1): 9–21. [DOI] [PubMed] [Google Scholar]
- Rathore K and Cekanova M (2015). "Effects of environmental carcinogen benzo(a)pyrene on canine adipose-derived mesenchymal stem cells." Res Vet Sci 103: 34–43. [DOI] [PubMed] [Google Scholar]
- Refaey ME, McGee-Lawrence ME, Fulzele S, Kennedy EJ, Bollag WB, Elsalanty M, Zhong Q, Ding KH, Bendzunas NG, Shi XM, Xu J, Hill WD, Johnson MH, Hunter M, Pierce JL, Yu K, Hamrick MW and Isales CM (2017). "Kynurenine, a Tryptophan Metabolite That Accumulates With Age, Induces Bone Loss." J Bone Miner Res 32(11): 2182–2193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roodman GD (1999). "Cell biology of the osteoclast." Exp Hematol 27(8): 1229–1241. [DOI] [PubMed] [Google Scholar]
- Safe S, Jin UH, Park H, Chapkin RS and Jayaraman A (2020). "Aryl Hydrocarbon Receptor (AHR) Ligands as Selective AHR Modulators (SAhRMs)." Int J Mol Sci 21(18). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scalise M, Galluccio M, Console L, Pochini L and Indiveri C (2018). "The Human SLC7A5 (LAT1): The Intriguing Histidine/Large Neutral Amino Acid Transporter and Its Relevance to Human Health." Front Chem 6: 243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schaldach CM, Riby J and Bjeldanes LF (1999). "Lipoxin A4: a new class of ligand for the Ah receptor." Biochemistry 38(23): 7594–7600. [DOI] [PubMed] [Google Scholar]
- Seidel SD, Winters GM, Rogers WJ, Ziccardi MH, Li V, Keser B and Denison MS (2001). "Activation of the Ah receptor signaling pathway by prostaglandins." J Biochem Mol Toxicol 15(4): 187–196. [DOI] [PubMed] [Google Scholar]
- Siddiqui JA and Partridge NC (2016). "Physiological Bone Remodeling: Systemic Regulation and Growth Factor Involvement." Physiology (Bethesda) 31(3): 233–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sinclair LV, Neyens D, Ramsay G, Taylor PM and Cantrell DA (2018). "Single cell analysis of kynurenine and System L amino acid transport in T cells." Nat Commun 9(1): 1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh KP, Bennett JA, Casado FL, Walrath JL, Welle SL and Gasiewicz TA (2014). "Loss of aryl hydrocarbon receptor promotes gene changes associated with premature hematopoietic stem cell exhaustion and development of a myeloproliferative disorder in aging mice." Stem Cells Dev 23(2): 95–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stein GS and Lian JB (1993). "Molecular mechanisms mediating proliferation/differentiation interrelationships during progressive development of the osteoblast phenotype." Endocr Rev 14(4): 424–442. [DOI] [PubMed] [Google Scholar]
- Stevens EA, Mezrich JD and Bradfield CA (2009). "The aryl hydrocarbon receptor: a perspective on potential roles in the immune system." Immunology 127(3): 299–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su N, Yang J, Xie Y, Du X, Chen H, Zhou H and Chen L (2019). "Bone function, dysfunction and its role in diseases including critical illness." Int J Biol Sci 15(4): 776–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swedenborg E and Pongratz I (2010). "AhR and ARNT modulate ER signaling." Toxicology 268(3): 132–138. [DOI] [PubMed] [Google Scholar]
- Takeuchi A, Takeuchi M, Oikawa K, Sonoda K-H, Usui Y, Okunuki Y, Takeda A, Oshima Y, Yoshida K, Usui M, Goto H and Kuroda M (2009). "Effects of Dioxin on Vascular Endothelial Growth Factor (VEGF) Production in the Retina Associated with Choroidal Neovascularization." Investigative Ophthalmology & Visual Science 50(7): 3410–3416. [DOI] [PubMed] [Google Scholar]
- Tantoh DM, Wu MC, Chuang CC, Chen PH, Tyan YS, Nfor ON, Lu WY and Liaw YP (2020). "AHRR cg05575921 methylation in relation to smoking and PM2.5 exposure among Taiwanese men and women." Clin Epigenetics 12(1): 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarnow P, Bross S, Wollenberg L, Nakajima Y, Ohmiya Y, Tralau T and Luch A (2017). "A Novel Dual-Color Luciferase Reporter Assay for Simultaneous Detection of Estrogen and Aryl Hydrocarbon Receptor Activation." Chem Res Toxicol 30(7): 1436–1447. [DOI] [PubMed] [Google Scholar]
- Thome T, Miguez K, Willms AJ, Burke SK, Chandran V, de Souza AR, Fitzgerald LF, Baglole C, Anagnostou ME, Bourbeau J, Jagoe RT, Morais JA, Goddard Y, Taivassalo T, Ryan TE and Hepple RT (2022). "Chronic aryl hydrocarbon receptor activity phenocopies smoking-induced skeletal muscle impairment." J Cachexia Sarcopenia Muscle 13(1): 589–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tong B, Yuan X, Dou Y, Wu X, Wang Y, Xia Y and Dai Y (2016). "Sinomenine induces the generation of intestinal Treg cells and attenuates arthritis via activation of aryl hydrocarbon receptor." Lab Invest 96(10): 1076–1086. [DOI] [PubMed] [Google Scholar]
- Tong Y, Niu M, Du Y, Mei W, Cao W, Dou Y, Yu H, Du X, Yuan H and Zhao W (2017). "Aryl hydrocarbon receptor suppresses the osteogenesis of mesenchymal stem cells in collagen-induced arthritic mice through the inhibition of beta-catenin." Exp Cell Res 350(2): 349–357. [DOI] [PubMed] [Google Scholar]
- Unkila M, Ruotsalainen M, Pohjanvirta R, Viluksela M, MacDonald E, Tuomisto JT, Rozman K and Tuomisto J (1995). "Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on tryptophan and glucose homeostasis in the most TCDD-susceptible and the most TCDD-resistant species, guinea pigs and hamsters." Arch Toxicol 69(10): 677–683. [DOI] [PubMed] [Google Scholar]
- Unnisa Z, Singh KP, Henry EC, Donegan CL, Bennett JA and Gasiewicz TA (2016). "Aryl Hydrocarbon Receptor Deficiency in an Exon 3 Deletion Mouse Model Promotes Hematopoietic Stem Cell Proliferation and Impacts Endosteal Niche Cells." Stem Cells Int 2016: 4536187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vert G and Chory J (2011). "Crosstalk in cellular signaling: background noise or the real thing?" Dev Cell 21(6): 985–991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vogel CF, Sciullo E and Matsumura F (2007). "Involvement of RelB in aryl hydrocarbon receptor-mediated induction of chemokines." Biochem Biophys Res Commun 363(3): 722–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Voronov I, Heersche JN, Casper RF, Tenenbaum HC and Manolson MF (2005). "Inhibition of osteoclast differentiation by polycyclic aryl hydrocarbons is dependent on cell density and RANKL concentration." Biochem Pharmacol 70(2): 300–307. [DOI] [PubMed] [Google Scholar]
- Voronov I, Li K, Tenenbaum HC and Manolson MF (2008). "Benzo[a]pyrene inhibits osteoclastogenesis by affecting RANKL-induced activation of NF-kappaB." Biochem Pharmacol 75(10): 2034–2044. [DOI] [PubMed] [Google Scholar]
- Ward KD and Klesges RC (2001). "A meta-analysis of the effects of cigarette smoking on bone mineral density." Calcif Tissue Int 68(5): 259–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watson ATD, Nordberg RC, Loboa EG and Kullman SW (2019). "Evidence for Aryl hydrocarbon Receptor-Mediated Inhibition of Osteoblast Differentiation in Human Mesenchymal Stem Cells." Toxicol Sci 167(1): 145–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei ZF, Lv Q, Xia Y, Yue MF, Shi C, Xia YF, Chou GX, Wang ZT and Dai Y (2015). "Norisoboldine, an Anti-Arthritis Alkaloid Isolated from Radix Linderae, Attenuates Osteoclast Differentiation and Inflammatory Bone Erosion in an Aryl Hydrocarbon Receptor-Dependent Manner." Int J Biol Sci 11(9): 1113–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wejheden C, Brunnberg S, Larsson S, Lind PM, Andersson G and Hanberg A (2010). "Transgenic mice with a constitutively active aryl hydrocarbon receptor display a gender-specific bone phenotype." Toxicol Sci 114(1): 48–58. [DOI] [PubMed] [Google Scholar]
- Widerak M, Ghoneim C, Dumontier MF, Quesne M, Corvol MT and Savouret JF (2006). "The aryl hydrocarbon receptor activates the retinoic acid receptoralpha through SMRT antagonism." Biochimie 88(3-4): 387–397. [DOI] [PubMed] [Google Scholar]
- Wilson SR, Joshi AD and Elferink CJ (2013). "The tumor suppressor Kruppel-like factor 6 is a novel aryl hydrocarbon receptor DNA binding partner." J Pharmacol Exp Ther 345(3): 419–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wormke M, Stoner M, Saville B, Walker K, Abdelrahim M, Burghardt R and Safe S (2003). "The aryl hydrocarbon receptor mediates degradation of estrogen receptor alpha through activation of proteasomes." Mol Cell Biol 23(6): 1843–1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright EJ, De Castro KP, Joshi AD and Elferink CJ (2017). "Canonical and non-canonical aryl hydrocarbon receptor signaling pathways." Curr Opin Toxicol 2: 87–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu T, Zhou Y, Qiu L, Do DC, Zhao Y, Cui Z, Wang H, Liu X, Saradna A, Cao X, Wan M and Gao P (2015). "Aryl Hydrocarbon Receptor Protects Lungs from Cockroach Allergen-Induced Inflammation by Modulating Mesenchymal Stem Cells." J Immunol 195(12): 5539–5550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu X, Zhang X, Yuan Y, Zhao Y, Fares HM, Yang M, Wen Q, Taha R and Sun L (2021). "Species-Specific Differences in Aryl Hydrocarbon Receptor Responses: How and Why?" Int J Mol Sci 22(24). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yahara Y, Nguyen T, Ishikawa K, Kamei K and Alman BA (2022). "The origins and roles of osteoclasts in bone development, homeostasis and repair." Development 149(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang JH and Lee HG (2010). "2,3,7,8-Tetrachlorodibenzo-p-dioxin induces apoptosis of articular chondrocytes in culture." Chemosphere 79(3): 278–284. [DOI] [PubMed] [Google Scholar]
- Yoshikawa Y, Izawa T, Hamada Y, Takenaga H, Wang Z, Ishimaru N and Kamioka H (2021). "Roles for B[a]P and FICZ in subchondral bone metabolism and experimental temporomandibular joint osteoarthritis via the AhR/Cyp1a1 signaling axis." Sci Rep 11(1): 14927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu TY, Kondo T, Matsumoto T, Fujii-Kuriyama Y and Imai Y (2014). "Aryl hydrocarbon receptor catabolic activity in bone metabolism is osteoclast dependent in vivo." Biochem Biophys Res Commun 450(1): 416–422. [DOI] [PubMed] [Google Scholar]
- Yu TY, Pang WJ and Yang GS (2015). "Aryl hydrocarbon receptors in osteoclast lineage cells are a negative regulator of bone mass." PLoS One 10(1): e0117112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Y, Jiang R, An L, Wang H, Cheng S, Qiong S and Weng Y (2017). "Benzo[a]pyrene impedes self-renewal and differentiation of mesenchymal stem cells and influences fracture healing." Sci Total Environ 587-588: 305–315. [DOI] [PubMed] [Google Scholar]
- Zhu K, Meng Q, Zhang Z, Yi T, He Y, Zheng J and Lei W (2019). "Aryl hydrocarbon receptor pathway: Role, regulation and intervention in atherosclerosis therapy (Review)." Mol Med Rep 20(6): 4763–4773. [DOI] [PMC free article] [PubMed] [Google Scholar]