Fact-checked by Grok 2 weeks ago

Steroid hormone receptor

Steroid hormone receptors (SHRs) are a subclass of s that function as ligand-activated transcription factors, binding steroid hormones such as estrogens, androgens, progestogens, glucocorticoids, and mineralocorticoids to regulate and mediate diverse physiological processes including , , , and stress responses. The discovery of steroid hormone receptors dates back to the mid-20th century. In 1958, Elwood V. Jensen identified the using radiolabeled , revealing specific high-affinity binding in target tissues such as the . This work laid the for understanding hormone-receptor interactions. In 1963, Peter Karlson proposed the "hormone-gene" hypothesis, positing that steroid hormones exert their effects by binding intracellular receptors that modulate gene transcription. These foundational insights spurred further research into the nuclear receptor superfamily. These receptors, comprising six classical members in humans (e.g., estrogen receptors α and β, , , , ), translate endocrine signals into transcriptional outcomes by interacting with and coregulatory proteins. Structurally, SHRs exhibit a modular organization with four main domains: an N-terminal domain (NTD) containing the ligand-independent 1 (AF-1), which is often intrinsically disordered to enable dynamic protein interactions; a central (DBD) featuring two motifs for sequence-specific recognition; a flexible region that undergoes post-translational modifications; and a C-terminal ligand-binding domain (LBD) housing the ligand-dependent 2 (AF-2) and a hydrophobic pocket for hormone binding. Upon ligand binding, SHRs dissociate from inhibitory heat shock proteins, dimerize, translocate to the , and bind to hormone response elements (HREs) such as estrogen response elements (EREs) or response elements (GREs), thereby recruiting coactivators to initiate and II-mediated transcription. In addition to this classical genomic pathway, SHRs can elicit rapid non-genomic effects through membrane localization, activating signaling cascades like MAPK/ERK or PI3K/Akt to influence and survival. Dysregulation of SHR signaling is implicated in numerous pathologies, including hormone-dependent cancers (e.g., , ), metabolic disorders, and autoimmune diseases, making these receptors key therapeutic targets for selective modulators like or dexamethasone. The synergistic action of AF-1 and AF-2 domains, modulated by and coregulator exchange, underscores the context-dependent nature of SHR activity across cell types and physiological states.

Introduction

Definition

Steroid hormones are a class of lipophilic signaling molecules derived from that readily diffuse across cell membranes due to their nonpolar structure. These hormones encompass estrogens, androgens, glucocorticoids, mineralocorticoids, and progestogens, each regulating diverse physiological processes such as , , and responses. Steroid hormone receptors are intracellular or membrane-associated proteins that specifically bind these with high affinity, typically in the nanomolar range, to initiate cellular signaling. Upon binding, these receptors undergo conformational changes that enable them to modulate target or activate rapid signaling pathways. The receptors are primarily classified into classical s, which function as intracellular ligand-activated transcription factors, and non-classical receptors, which are often membrane-associated and mediate rapid, non-genomic actions. receptors belong to the nuclear receptor superfamily, specifically the NR3C subfamily (with receptors in the related NR3A group), which comprises 48 members in humans, though only select ones—such as ESR1/2 (), AR (), GR (), PR (progesterone), and MR ()—bind s.

Historical background

The discovery of steroid hormones and their physiological effects began in the early , with key isolations marking initial observations of their roles in and development. Estrogen was first isolated in 1929 from the urine of pregnant women, enabling studies on its influence on female reproductive tissues. followed in 1934, extracted from corpora lutea by Willard M. Allen and George W. Corner, revealing its essential function in maintaining pregnancy. These milestones, along with characterizations by , Tadeus Reichstein, and Edward Doisy in the 1920s and 1930s, laid the groundwork for understanding steroid hormone actions. Butenandt shared the 1939 with Leopold Ruzicka for their work on sex hormones. In the mid-20th century, the concept of specific receptor proteins mediating steroid effects emerged through pioneering biochemical experiments. In 1958, Elwood V. Jensen used tritium-labeled to demonstrate high-affinity binding in target tissues like the , identifying the first steroid hormone receptor—the —in mammals. This radiolabeling approach revolutionized receptor detection, confirming that steroids concentrate in responsive cells via saturable, specific proteins. Building on this, Peter Karlson proposed the "hormone-gene" hypothesis in 1963, positing that steroid hormones directly regulate gene transcription through intracellular receptors, shifting paradigms from metabolic to genomic actions. Throughout the and , similar receptors for progesterone, glucocorticoids, and androgens were identified using radioligands, establishing the receptor model as central to steroid signaling. The 1980s brought molecular insights via gene cloning, unveiling the superfamily. The human cDNA was cloned in 1985 from cells, revealing a homologous to oncogenes like v-erbA, suggesting a broader family of ligand-activated transcription factors. Subsequent clonings, including the in 1986, confirmed structural conservation across receptors, defining the superfamily that includes non-steroid members like receptors. These advances enabled functional expression studies, solidifying the mechanism of action at the genetic level. The 1990s expanded the receptor landscape to non-classical forms, highlighting diverse signaling pathways. In 1997, a novel , GPR30 (later renamed ), was cloned from cells, initially as an but soon linked to rapid responses independent of classical receptors. This discovery underscored membrane-associated signaling, broadening the field beyond genomic actions. advanced in the early , providing atomic-level insights into receptor-ligand interactions. The first X-ray of a receptor ligand-binding domain—the in complex with dexamethasone—was solved in 2002, revealing a helical fold that accommodates ligands and recruits coactivators, facilitating rational for hormone-related disorders. This milestone, followed by structures of other receptors like the in 2004, transformed understanding of conformational dynamics and therapeutic targeting.

Classical Nuclear Receptors

Structure and domains

Classical steroid hormone receptors (SHRs), such as the estrogen receptor (ER), glucocorticoid receptor (GR), androgen receptor (AR), progesterone receptor (PR), and mineralocorticoid receptor (MR), are intracellular transcription factors belonging to the nuclear receptor superfamily. These proteins exhibit a modular architecture, typically comprising 500–900 amino acids, and exist as monomers in their unliganded state but form homodimers upon ligand binding to enhance DNA interaction and transcriptional activity. The conserved domain organization includes an N-terminal A/B domain, a central DNA-binding domain (DBD), a flexible hinge region, and a C-terminal ligand-binding domain (LBD), which collectively enable ligand recognition, DNA binding, and co-regulator recruitment. The N-terminal A/B domain, also known as the transactivation domain, is the most variable region among SHRs, ranging from 30–500 in length, and is often intrinsically disordered with a high propensity for conformation. It harbors activation function 1 (AF1), which mediates ligand-independent by recruiting transcriptional co-activators and is regulated by post-translational modifications such as at specific serine residues, which can enhance or modulate receptor activity. For instance, in the , within AF1 influences its interaction with the basal transcriptional machinery. The A/B domain's flexibility allows it to adopt ordered conformations upon binding partners, contributing to context-specific . The central DBD, or C domain, is highly conserved across SHRs, spanning approximately 65–70 amino acids, and features two zinc finger motifs formed by eight cysteine residues coordinating four zinc ions. The first zinc finger contains a proximal box (P-box) with residues like GR's Gly-Ser-Ala-Cys that recognize specific hormone response elements (HREs) in DNA, such as the inverted palindromic GRE consisting of two AGAACA half-sites separated by a three-nucleotide spacer (5'-AGAACAnnnTGTTCT-3'). The second zinc finger includes a dimerization box (D-box) that facilitates receptor dimerization on DNA. Early structures, including the 1990 NMR solution structure of the free GR DBD and the 1991 crystal structure of the rat GR DBD bound to DNA, revealed how these zinc fingers insert into the major groove of DNA for sequence-specific interactions. The hinge region, or D domain, is a flexible linker of 40–65 connecting the DBD and LBD, exhibiting the least sequence conservation among SHRs. It contains nuclear localization signals (NLS) essential for receptor translocation to the and serves as a site for post-translational modifications like sumoylation, which can fine-tune receptor function. This domain's plasticity allows conformational adjustments during dimerization and co-regulator binding. The C-terminal LBD, encompassing the E and F domains (approximately 250 ), forms a globular structure with 11–12 α-helices arranged in an antiparallel sandwich, creating a hydrophobic pocket for high-affinity . 12 within the LBD constitutes 2 (AF2), which repositions upon to form a co-activator groove, recruiting proteins with LXXLL motifs. specificity arises from pocket residues; for example, the AR's LBD prefers androgens like testosterone due to bulky residues like Met895 that exclude estrogens, as shown in crystal structures. In the unliganded state, the LBD associates with chaperone complexes, including , to maintain an open conformation and prevent premature activation. Seminal crystal structures from the late 1990s and early 2000s, such as PDB entry 1ERE for the ERα LBD bound to , illustrated these ligand-induced conformational shifts, while PDB 2AM9 for the AR LBD with testosterone highlighted androgen-specific features. The F domain, present in some SHRs like ER, extends the LBD and modulates ligand-dependent activity without directly the .

Mechanism of genomic action

In the unliganded state, steroid hormone receptors are stabilized by binding to heat shock protein 90 (HSP90) chaperones, which maintain their inactivity and proper folding. For receptors such as the (GR), this chaperone-bound complex is predominantly localized in the , whereas for the (ER), it is primarily nuclear. Ligand binding to the ligand-binding domain (LBD) triggers a conformational change that dissociates the HSP90 complex, exposing the nuclear localization signal (NLS) in the hinge region and enabling nuclear translocation for cytoplasmic receptors like GR. This process also promotes receptor dimerization, essential for subsequent DNA interactions. The binding exhibits high affinity, with dissociation constants (Kd) typically in the range of 1-10 nM, reflecting the physiological concentrations of steroid hormones. The simple equilibrium for this binding is: \text{Receptor} + \text{Hormone} \rightleftharpoons \text{RH complex} where the association constant is given by K_a = \frac{[\text{RH}]}{[\text{R}][\text{H}]}. The dimerized, ligand-bound receptors then bind to specific hormone response elements (HREs) in the DNA, such as the inverted palindromic sequence AGGTCA half-sites for ER. In the absence of ligand, corepressors like nuclear receptor corepressor (NCoR) associate with the receptor to inhibit transcription, but ligand binding shifts this to recruitment of coactivators, including steroid receptor coactivator-1 (SRC-1), which interacts via LXXLL motifs with the activation function-2 (AF2) surface in the LBD. This coactivator complex enhances chromatin accessibility at enhancers and promoters, facilitating the recruitment of RNA polymerase II and the basal transcription machinery to initiate target gene transcription. For instance, activated GR upregulates anti-inflammatory genes such as FKBP5, contributing to resolved inflammatory responses. The residence time of the activated receptor on DNA is relatively short (on the order of seconds to minutes), enabling dynamic cycling and regulated gene expression changes.

Non-Classical Receptors

Membrane-bound nuclear receptors

Membrane-bound nuclear receptors represent a subset of classical receptors, such as (ERα) and beta (ERβ) as well as the (AR), that localize to the plasma membrane to facilitate rapid, non-transcriptional signaling. These receptors maintain the core modular structure of their nuclear counterparts, including DNA-binding and ligand-binding domains, but a small fraction—approximately 5-10% of total cellular ERα and ERβ—is targeted to the membrane through post-translational modifications. Palmitoylation, mediated by DHHC-7 and DHHC-21 acyltransferases, attaches to residues in the ligand-binding domain (e.g., Cys447 in human ERα), anchoring the receptors to cholesterol-rich domains like caveolae and lipid rafts via interactions with caveolin-1. Myristoylation plays a lesser role but can contribute to membrane association in certain contexts for these receptors. Upon ligand binding, such as for ERα or testosterone for , these membrane-bound receptors do not translocate to the but instead rapidly associate with transmembrane proteins to initiate signaling cascades. For instance, membrane ERα interacts with (IGF1R), promoting downstream activation of pathways like PI3K/Akt, while membrane can engage (EGFR) to modulate cell proliferation signals. A prominent example is membrane ER (mER), which, in endothelial cells, binds to activate endothelial nitric oxide synthase (eNOS) via kinase and PI3K, leading to production and acute . These variants differ from classical receptors in their dynamics and discovery. They exhibit shorter half-lives (typically 1-4 hours upon binding) and higher rates compared to forms, enabling quick responses to fluctuating levels. localization was first identified in the through subcellular techniques that separated components from cytosolic and fractions in steroid-responsive cells.

G protein-coupled receptors

G protein-coupled estrogen receptor (GPER), also known as GPR30, is a seven-transmembrane (GPCR) that mediates rapid non-genomic effects of estrogens. It was cloned in 1997 from breast cancer tissue as an and later identified as an estrogen-binding protein. GPER exhibits high-affinity binding to 17β-estradiol with a (Kd) of approximately 3 nM. Upon ligand binding, GPER couples to both Gαs and Gαi proteins, leading to bidirectional modulation of activity and intracellular cAMP levels. Activation of GPER triggers multiple downstream signaling cascades, including phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) via the (MAPK) pathway and activation of the (PI3K)/Akt pathway, which promote cell survival and proliferation. Additionally, GPER stimulates (PLC) to generate (IP3), resulting in intracellular calcium mobilization from stores. These rapid signaling events contribute to non-genomic estrogen actions, distinct from classical pathways. GPER is ubiquitously expressed across tissues, with particularly high levels in the , , and . In these tissues, GPER activation influences cellular processes such as and ; for instance, in and cells, stimulation of GPER enhances tumor cell motility and invasiveness through ERK1/2 and PI3K/Akt signaling. Beyond GPER, membrane progestin receptors (mPRs) represent another class of steroid-binding receptors with G protein-coupling properties, belonging to the progestin and AdipoQ receptor (PAQR) family. Unlike classical GPCRs, mPRs feature a distinct seven-transmembrane but associate with inhibitory G proteins (Gαi) to suppress activity upon progestin binding. This signaling is well-documented in and models, where mPRs mediate rapid progesterone effects on maturation and sperm , though their roles and expression in remain less clearly defined. Recent studies in the 2020s have highlighted the neuroprotective potential of activation using selective agonists like G-1. For example, G-1 administration in animal models of reduces and dopaminergic loss by modulating microglial activation and ERK1/2 signaling. Similarly, G-1 confers protection against cerebral ischemia in ovariectomized rats by attenuating inflammatory responses and via GPER-mediated pathways.

Ion channels and other effectors

Steroid hormones can directly modulate ion channels through non-receptor mechanisms, enabling rapid cellular responses independent of genomic transcription. For instance, estrogens enhance the activity of big potassium (BK) channels in various cell types, including vascular smooth muscle and neurons, by increasing channel open probability and burst duration without involving classical nuclear receptors. This activation occurs via nongenomic pathways, such as nitric oxide-mediated phosphorylation of the channel's β subunit, leading to membrane hyperpolarization and reduced excitability within seconds to minutes. Similarly, progesterone metabolites like allopregnanolone directly bind to and potentiate GABA_A receptors, which function as chloride channels, enhancing inhibitory chloride influx and promoting neuronal silencing. At low concentrations, allopregnanolone increases the frequency and duration of channel openings in response to GABA, while higher doses can directly gate the channel. Receptor-mediated modulation also plays a key role, where membrane-associated pools of steroid receptors influence voltage-gated calcium channels (VGCCs) to alter cellular excitability. Membrane androgen receptors (AR) and glucocorticoid receptors (GR) can rapidly regulate L-type VGCCs, such as CaV1.2, through protein-protein interactions or signaling cascades that adjust channel gating and calcium influx. For example, testosterone, acting via nonclassical AR signaling at the plasma membrane, opens ATP-sensitive potassium (KATP) channels in vascular smooth muscle cells, promoting potassium efflux, hyperpolarization, and vasodilation independent of endothelial factors. This effect is direct and occurs within minutes, as evidenced by increased single-channel activity in patch-clamp studies. Recent research highlights synergies between steroid hormones and vitamin D in regulating ion channels; for instance, sex steroids and vitamin D together modulate expression and function of channels like TRPV5 and L-type VGCCs in hormone-sensitive tissues, influencing calcium homeostasis through both transcriptional and rapid nongenomic actions. These modulations often involve post-translational mechanisms, such as phosphorylation of ion channels by kinases activated downstream of steroid receptor signaling, without requiring DNA binding. GPER-mediated estrogen signaling, for example, activates protein kinase A (PKA), which phosphorylates targets including potassium and calcium channels, thereby fine-tuning their conductance and voltage sensitivity. Such rapid effects are exemplified by estradiol's potentiation of L-type VGCCs in hippocampal neurons, increasing calcium influx to support synaptic plasticity within seconds. Overall, these interactions enable steroids to elicit fast, localized changes in membrane potential and ion flux, distinct from slower genomic pathways.

SHBG receptor complex

Sex hormone-binding globulin (SHBG) is a homodimeric , approximately 90–100 kDa in size, primarily synthesized and secreted by hepatocytes in the liver. It exhibits high-affinity binding to androgens such as testosterone and (DHT), as well as estrogens like , with dissociation constants (Kd) around 1 nM, thereby serving as the principal carrier for these steroids in . In humans, SHBG binds 40–65% of circulating testosterone and 20–40% of , significantly influencing their by limiting free hormone diffusion while potentially facilitating targeted delivery to tissues. The SHBG receptor (SHBG-R) is hypothesized to be a multiprotein complex on the that binds SHBG to facilitate non-classical steroid signaling and potentially cellular uptake, though its exact molecular identity and mechanisms remain under . sites were first detected in the late with Kd ~10 on membranes of target cells like those in the and , and activity was further characterized in the as facilitating rapid actions independent of intracellular receptors. In certain tissues, such as leiomyomas, SHBG-R co-localizes with caveolin-1 in caveolae structures, suggesting involvement in microdomain . Proposed mechanisms include promotion of of SHBG-bound s, primarily via the endocytic receptor megalin (a receptor family member) in steroid-responsive tissues like the reproductive tract, allowing internalized steroids to dissociate and access intracellular receptors; however, this process remains controversial. This mechanism challenges the traditional free hormone hypothesis by demonstrating rather than passive alone. Upon binding an appropriate , the SHBG-steroid-SHBG-R complex has been reported to trigger G protein-mediated , rapidly activating to elevate intracellular cyclic AMP () levels within minutes, though these signaling pathways are still debated. This non-genomic pathway modulates downstream effectors, including , influencing cellular processes like gene transcription indirectly. For instance, the DHT-SHBG complex enhances in semen samples, correlating with higher local SHBG isoform abundance and supporting through rapid signaling in reproductive cells. Unlike classical nuclear receptors that directly bind free steroids for genomic effects, the SHBG-R complex primarily modulates steroid access to intracellular receptors while initiating these membrane-based signals. SHBG levels and function are tightly regulated, with plasma concentrations decreasing in conditions like obesity due to hyperinsulinemia and hepatic lipogenesis, which suppress hepatic synthesis and reduce overall hormone bioavailability. Genetic variants in the SHBG gene, such as single nucleotide polymorphisms in the promoter region (e.g., rs6259), are associated with altered SHBG expression and circulating levels, impacting sex steroid availability and contributing to metabolic and reproductive phenotypes. These regulatory aspects underscore SHBG's role as a modulator rather than a direct receptor, fine-tuning steroid hormone action across physiological contexts.

Signaling Integration

Non-genomic actions

Non-genomic actions of steroid hormone receptors refer to rapid signaling events that occur within seconds to minutes, mediated primarily through membrane-associated receptors or effectors, and independent of transcription. These effects contrast with the slower genomic pathways by enabling immediate physiological responses, such as alterations in fluxes, second messenger systems, and kinase activation, often without requiring protein synthesis. Key signaling pathways activated by non-genomic steroid actions include the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) cascade, phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) pathway, and protein kinase C (PKC) signaling. For instance, estrogen binding to the G protein-coupled estrogen receptor (GPER) rapidly phosphorylates endothelial nitric oxide synthase (eNOS) via Akt and ERK, leading to increased nitric oxide (NO) production and subsequent vasodilation in vascular endothelium. Similarly, these pathways facilitate quick modulation of intracellular calcium levels and cyclic AMP, contributing to diverse cellular responses across tissues. Representative examples illustrate the breadth of these actions. Progesterone, acting through membrane progestin receptors (mPRs), induces rapid oocyte maturation in species like Xenopus laevis by elevating intracellular calcium and activating , promoting meiotic resumption without transcriptional involvement. Glucocorticoids exert fast anti-inflammatory effects via membrane glucocorticoid receptors (mGR), rapidly inhibiting pro-inflammatory cytokine release and modulating activity in immune cells, which supports immediate suppression of inflammation in conditions like . These non-genomic actions provide advantages for fine-tuned, localized physiological regulation, such as enhancing neuronal excitability in response to steroids or enabling rapid behavioral adaptations in reproductive contexts. Evidence for their transcription-independent nature comes from experiments showing that inhibitors like actinomycin D, which block synthesis, do not abolish these rapid effects, confirming reliance on pre-existing cellular machinery. Recent research as of 2024 has highlighted roles of macrophages in regulating production through non-genomic interactions during stress, contributing to dynamic hormone modulation.

Cross-talk with other pathways

Steroid hormone receptors (SRs) engage in extensive cross-talk with other cellular signaling pathways, enabling integrated responses to diverse stimuli and fine-tuning physiological outcomes. This interplay occurs through both genomic and non-genomic mechanisms, where SRs modulate or are modulated by pathways such as those involving receptor tyrosine kinases, transcription factors, and metabolic regulators. Such interactions are crucial for processes like control, , and metabolic , often involving shared co-regulators or indirect DNA binding modes. A prominent example of SR-pathway cross-talk is the ligand-independent transactivation of (EGFR) and human epidermal growth factor receptor 2 (HER2) by (ER). In breast cancer cells, ER can phosphorylate and activate EGFR/HER2 without estrogen binding, leading to downstream MAPK/ERK signaling that promotes cell survival and . This non-canonical activation occurs via matrix metalloproteinase-dependent release of EGFR ligands like heparin-binding EGF-like growth factor. Similarly, the (GR) exerts effects by inhibiting nuclear factor kappa B (NFκB) and c-Jun (a component of AP-1), preventing their transcriptional activity on pro-inflammatory genes such as interleukin-6 and tumor necrosis factor-alpha. GR achieves this through direct protein-protein interactions that sequester NFκB or compete for co-activators, thereby suppressing cytokine production in immune cells. Non-genomic cross-talk further amplifies SR signaling integration. The links to MAPK pathways by transactivating growth factor receptors, including , via rapid calcium mobilization and kinase activation, which supports estrogen-dependent endothelial cell migration and vascular function. At the genomic level, SRs integrate with other pathways through shared co-regulators and feedback mechanisms. For instance, and AP-1 (comprising c-Fos/c-Jun) co-occupy promoters via the histone acetyltransferases p300/CBP, which bridge their activities to regulate genes involved in ; this tethering allows ER to enhance AP-1-driven transcription in a ligand-dependent manner. autoregulation exemplifies feedback loops, where ligand-bound GR downregulates its own expression by binding negative response elements, limiting excessive glucocorticoid signaling and maintaining in stress responses. In cancer contexts, ER cross-talk with the PI3K/AKT/mTOR pathway drives resistance to tamoxifen by sustaining cell survival signals; PI3K activation phosphorylates ER at serine 167, enhancing its transcriptional activity independently of estrogen, thus bypassing endocrine blockade. Mechanistically, these integrations operate via distinct DNA binding models: direct binding to composite elements, where SRs and partner factors (e.g., NFκB) simultaneously contact adjacent response elements for synergistic activation, or tethering, where SRs indirectly associate with DNA through protein-protein interactions with DNA-bound partners like AP-1, facilitating transrepression without canonical SR DNA motifs. Tethering predominates in anti-inflammatory contexts, such as GR-NFκB inhibition, while composite elements support cooperative gene induction.

Clinical Relevance

Role in diseases

Dysregulation of steroid hormone receptors plays a central role in numerous pathologies, often through mutations, overexpression, or altered signaling that disrupts normal hormonal control of cellular processes. In cancers, aberrant receptor activity promotes uncontrolled proliferation and therapeutic resistance. For instance, overexpression of the (ER) alpha occurs in approximately 70-80% of cancers, driving tumor growth via enhanced estrogen-dependent transcription. Similarly, mutations in the (AR) are implicated in castration-resistant , where they enable ligand-independent activation and confer resistance to therapies like . In hematological malignancies, the glucocorticoid receptor (GR) is essential for inducing in leukemic cells, but polymorphisms or downregulation lead to glucocorticoid resistance in up to 20-30% of cases, worsening prognosis. Endocrine disorders frequently arise from steroid receptor defects that impair hormone responsiveness. Complete androgen insensitivity syndrome, a form of 46,XY disorder of sex development, results from over 1,000 identified mutations in the AR gene, leading to resistance to androgens and female external genitalia despite male gonadal development. In polycystic ovary syndrome (PCOS), altered progesterone receptor (PR) expression in the endometrium contributes to progesterone resistance, disrupting normal menstrual cycling and increasing risks of endometrial hyperplasia; studies show reduced PR levels and impaired downstream signaling in PCOS patients compared to controls. Metabolic and inflammatory conditions are also linked to steroid receptor variants. GR polymorphisms, such as the A3669G variant, are associated with relative resistance that attenuates metabolic complications like and altered lipid profiles in patients with . Recent research highlights the role of nuclear receptors (NRs) in non-alcoholic (NAFLD) through immunometabolic pathways; for example, NRs like PPARs and FXR regulate lipid homeostasis and inflammation, and their dysregulation exacerbates and , with 2025 studies emphasizing FXR agonists for modulating immune responses in metabolic dysfunction-associated steatotic liver disease. Neurological diseases involve steroid receptors in neuroprotection and rapid signaling. Loss of ER and G protein-coupled estrogen receptor (GPER) function contributes to Alzheimer's disease progression, where GPER activation normally mitigates amyloid-β toxicity and tau pathology, but its downregulation in aging brains heightens vulnerability to neurodegeneration. In stroke, rapid non-genomic actions of steroid receptors, such as progesterone receptor-mediated signaling, provide by reducing ischemic injury through anti-inflammatory and anti-apoptotic effects, as demonstrated in preclinical models of cerebral ischemia. Clinically, these receptor dysregulations have significant implications; for example, about 70% of ER-positive s initially respond to endocrine due to receptor dependency, though resistance develops in many cases via pathway .

Therapeutic applications

receptors are key targets for therapeutic interventions in hormone-dependent diseases, primarily through the use of selective agonists, antagonists, and modulators that exploit their ligand-binding properties to alter and cellular responses. Selective modulators (SERMs) such as act as antagonists in tissue to inhibit -driven proliferation in -positive (ER+) , serving as a first-line endocrine that reduces recurrence risk by approximately 50% in early-stage disease. Similarly, (AR) antagonists like bind to the AR ligand-binding domain, preventing nuclear translocation and DNA binding, thereby suppressing tumor growth in metastatic castration-resistant (mCRPC) and extending overall survival by 4-5 months in post-chemotherapy patients. (GR) agonists, exemplified by dexamethasone, mimic endogenous to induce anti-inflammatory effects by promoting transrepression of pro-inflammatory genes, widely used in conditions like and to rapidly suppress storms. Emerging therapies target non-classical steroid hormone receptors to harness rapid, non-genomic signaling for cardioprotective and metabolic benefits. G protein-coupled estrogen receptor (GPER) agonists, such as G1, activate to elicit vasodilatory and anti-fibrotic effects in preclinical models of cardiac ischemia/, with ongoing research exploring their potential in cardiovascular protection through endothelial activation. These agents show promise in clinical development for metabolic , where GPER modulation improves insulin sensitivity and reduces adiposity in animal models of and . Advanced strategies focus on coregulator modulation to overcome limitations of traditional ligands, including proteolysis-targeting chimeras (PROTACs) that induce ubiquitin-mediated of receptors. AR-targeted PROTACs like ARV-110 and ARV-766 are in phase 1/2 clinical trials for mCRPC, demonstrating partial responses in 20-30% of heavily pretreated patients by selectively depleting AR protein levels without affecting wild-type steroid receptors. Similarly, ER degrader PROTACs such as ARV-471 (vepdegestrant) have advanced to phase 3 trials for ER+ , with the VERITAC-2 study (completed in 2025) demonstrating a statistically significant improvement in (median 2.9 months longer than ) in patients with ESR1 mutations and prior endocrine therapy. Early phase 1 data showed clinical benefit rates up to 40% in pretreated patients. Recent advances in 2025 emphasize (NR)-targeted therapies for metabolic diseases, including selective GR agonists (SEGRAs) designed to preferentially induce transrepression for effects while minimizing transactivation-linked metabolic side effects like . These SEGRAs, such as those in preclinical optimization, aim to treat conditions like by dissociating therapeutic efficacy from adverse glucose dysregulation observed in classical glucocorticoids. Therapeutic challenges include resistance mechanisms driven by receptor and pathway cross-talk, complicating long-term efficacy. For instance, ESR1 occur in approximately 30% of ER+ breast cancers resistant to endocrine , leading to constitutive ER activation and relapse rates of up to 30% despite initial SERM responsiveness. In prostate cancer, AR splice variants confer enzalutamide resistance in 20-40% of cases, underscoring the need for combination strategies targeting multiple signaling nodes.

References

  1. [1]
    Chemogenomics for steroid hormone receptors (NR3) - Nature
    Feb 3, 2025 · The nine human NR3 nuclear receptors translate steroid hormone signals in transcriptomic responses and operate multiple highly important ...
  2. [2]
    Steroid Hormone Receptors: Links With Cell Cycle Machinery and ...
    With a brief section covering the structure and general functions of SHR, this review focuses on finding the SHR-driven cross-talk between cell cycle and breast ...Abstract · Introduction · Biology of Steroid Hormone... · SHR-Driven Cross-Link...
  3. [3]
    Structural and functional relationships of the steroid hormone ...
    In this review, we summarize and discuss current knowledge regarding the molecular mechanisms of AF1-mediated gene activation, focusing on AF1 conformation and ...
  4. [4]
    Free Diffusion of Steroid Hormones Across Biomembranes
    Since steroid hormones are derived from cholesterol and share an identical backbone, they might be expected to associate with the membrane in a similar way.
  5. [5]
    Cellular Cholesterol Delivery, Processing & Use for Steroids
    Cholesterol is a starting material for the biosynthesis of steroid hormones; these fat soluble, low molecular weight substances play diverse and important ...
  6. [6]
    Function and Evolution of Nuclear Receptors in Environmental ...
    However, all these effects are regulated by NRs, which bind a highly specific (nanomolar affinity) steroidal ligand comprising two ER (ERα and ERβ), an androgen ...
  7. [7]
    The Interface of Nuclear and Membrane Steroid Signaling - PMC
    The most widely studied class of steroid hormone receptors are the nuclear receptors, named for their function as ligand-dependent transcription factors in the ...
  8. [8]
    Nuclear receptors outside the nucleus: extranuclear signalling ... - NIH
    Oct 12, 2016 · In contrast to nuclear steroid receptors, membrane-associated steroid receptors do not directly engage DNA to modulate transcription.
  9. [9]
    [PDF] Nuclear Receptor Superfamily-Update 2023 - DigitalCommons@TMC
    Nov 1, 2023 · This document is the International Union of Basic and Clinical Pharmacology CXIII update on the Nuclear Receptor Superfamily from 2023.
  10. [10]
    The History of Estrogen - February 2016 - menoPAUSE Blog
    Feb 17, 2016 · Yet, it was not until 1929 that estrogen as a hormone was isolated. The first commercial preparation of estrogen began as an estrogen ...
  11. [11]
    Ninety years of progesterone: the 'other' ovarian hormone - PMC - NIH
    It is wholly 90 years since the first report of the isolation and characterisation of progesterone by Willard M. Allen and George W. Corner.
  12. [12]
    One hundred years of hormones - PMC - PubMed Central - NIH
    In the 1920s and 1930s, Adolf Butenandt, Tadeus Reichstein and Edward Adelbert Doisy discovered and characterized various steroid hormones, including oestrogen ...
  13. [13]
    Elwood V. Jensen (1920–2012): Father of the nuclear receptors - NIH
    Mar 4, 2013 · Jensen is popularly credited with the discovery of the mammalian estrogen receptor, and the promulgation of this important molecule as a key player in female ...
  14. [14]
    Review Fifty years ago: The quest for steroid hormone receptors
    Aug 15, 2013 · In 1963 Peter Karlson put forward the revolutionary “hormone-gene” hypothesis, which would change drastically the way in which steroid hormones ...
  15. [15]
    An abridged history of sex steroid hormone receptor action - PubMed
    The understanding that steroid hormone receptors interact with the general transcription machinery and alter chromatin structure came in the 1970s and 1980s, ...
  16. [16]
    Cloning of the human glucocorticoid receptor cDNA - PubMed
    Cloning of the human glucocorticoid receptor cDNA. Nucleic Acids Res. 1985 Dec 9;13(23):8293-304. doi: 10.1093/nar/13.23.8293.
  17. [17]
    Elwood V. Jensen (1920–2012): Father of the nuclear receptors
    Mar 4, 2013 · ... cloning of the estrogen receptor in the Chambon laboratory in 1986. The work of Evans on the nuclear receptor superfamily awakened ...
  18. [18]
    Nuclear Receptor Superfamily: A Rosetta Stone for Physiology
    In the December 1985 issue of Nature, we described the cloning of the first nuclear receptor cDNA encoding the human glucocorticoid receptor (GR) (1). In the 20 ...
  19. [19]
    Crystal Structure of the Glucocorticoid Receptor Ligand Binding ...
    Crystal structure of the glucocorticoid receptor ligand binding domain reveals a novel mode of receptor dimerization and coactivator recognition.
  20. [20]
    Structure-Based Design of Estrogen Receptor-β Selective Ligands
    In this paper we describe X-ray crystallographic studies and structure-based optimization of a series of ERβ selective ligands. Using ab initio quantum ...
  21. [21]
  22. [22]
  23. [23]
    Visualizing the action of steroid hormone receptors in living cells
    Mar 9, 2007 · B. All steroid receptors are composed of a variable N-terminal domain (A/B) containing the AF-1 transactivation region, a highly conserved DNA ...
  24. [24]
  25. [25]
  26. [26]
  27. [27]
  28. [28]
    Communication between genomic and non-genomic signaling ... - NIH
    Here, we describe the different types of genomic and non-genomic steroid hormone signaling mechanisms and how they can influence one another to ultimately ...
  29. [29]
  30. [30]
  31. [31]
  32. [32]
    The role of FKBP5, a co-chaperone of the glucocorticoid receptor in ...
    FK506 binding protein 51 or FKBP5 is a co-chaperone of hsp90 which regulates glucocorticoid receptor (GR) sensitivity.
  33. [33]
    The Steroid Hormone Receptors | GLOWM
    The steroid receptors are considered class I members of the nuclear receptor superfamily while the other receptors are class II receptors. A variety of ...
  34. [34]
    Emerging Evidence on Membrane Estrogen Receptors as Novel ...
    Feb 17, 2023 · A pool of 5–10% of the classical estrogen receptors ERα and ERβ is located in the cell membrane, where they are embedded in lipid rafts [15,16].
  35. [35]
    The role of membrane ERα signaling in bone and other major ...
    Jul 8, 2016 · This membrane-bound fraction is estimated to be approximately 5–10%, depending on cell-type12.
  36. [36]
    DHHC-7 and -21 are palmitoylacyltransferases for sex steroid ...
    Oct 26, 2011 · In this paper, we report that DHHC-7 and -21 proteins serve as PATs for ER, PR, and AR, driving membrane localization and rapid signaling by ...
  37. [37]
    Mutation of the palmitoylation site of estrogen receptor α in vivo ...
    We created a mouse with a point mutation of the palmitoylation site of ERα (C451A-ERα) to obtain membrane-specific loss of function.
  38. [38]
    Mutation of the palmitoylation site of estrogen receptor α in vivo ...
    In the plasma membrane, ERα has been localized to caveolae/lipid rafts by direct binding to caveolin-1 through Ser-522 or indirectly via the scaffold protein ...
  39. [39]
    Rapid Actions of the Nuclear Progesterone Receptor through cSrc in ...
    In the case of the PR, ER, and AR, their attachment to caveolae lipid rafts of the plasma membrane is mediated by their palmitoylation at the ligand-binding ...
  40. [40]
    The role of Shc and insulin-like growth factor 1 receptor in ... - PNAS
    Down-regulation of IGF-1R clearly blocked ERα membrane translocation in the cells (Fig. 4 Ah and Bc), confirming the role of IGF-1R on ERα membrane association.
  41. [41]
    Membrane-Initiated Estrogen, Androgen, and Progesterone ...
    We provide a review and update on the current state of knowledge of membrane-initiated estrogen (ER), androgen (AR) and progesterone (PR) receptor signaling.Membrane Estrogen Signaling · G Protein--Coupled Estrogen... · Membrane Androgen Signaling<|separator|>
  42. [42]
    Membrane Estrogen Receptor Engagement Activates Endothelial ...
    Here we demonstrate for the first time, to our knowledge, that E2 rapidly induces phosphorylation and activation of eNOS through the phosphatidylinositol 3 (PI3)- ...Missing: mER | Show results with:mER
  43. [43]
    Membrane estrogen receptor engagement activates endothelial ...
    Here we demonstrate for the first time, to our knowledge, that E(2) rapidly induces phosphorylation and activation of eNOS through the phosphatidylinositol 3 ( ...Missing: mER | Show results with:mER
  44. [44]
    Temporal variation in estrogen receptor-α protein turnover in the ...
    Upon initial hormone treatment, ERα half-life is shortened from 3 to 1 h. However, ERα half-life increases over time, achieving a half-life of ∼6 h in 72 h of ...<|control11|><|separator|>
  45. [45]
    Membrane and Nuclear Estrogen Receptor Alpha Actions
    This review mainly focuses on ERα mechanisms of action and its major role in reproduction (in particular in uterus and mammary gland) and in vascular ...
  46. [46]
    Steroid hormones: Interactions with membrane-bound receptors
    them were cloned in the 1980s and 1990s. These discoveries were the origins of the classical model for steroid hormone action (Figure 1a). In this model ...
  47. [47]
    G Protein-Coupled Estrogen Receptor: A Potential Therapeutic ...
    GPER, also known as GPER1 or GPR30, was first discovered in 1996 in breast cancer tissue (5). Its cDNA sequence was cloned by Takada et al. in 1997 via ...
  48. [48]
    GPER1 - G-protein coupled estrogen receptor 1 | UniProtKB - UniProt
    Binds 17-beta-estradiol (E2) in plasma membranes with high affinity (Kd is 3.3 nM) and displays rapid kinetics of association and dissociation. 3 ...
  49. [49]
    G Protein–Coupled Estrogen Receptor GPER - PubMed Central - NIH
    Competitive binding assays with radiolabeled or fluorescent probes revealed that E2 has the highest GPER binding affinity (3–6 nM) and greater than 1,000-fold ...Gper Ligands And... · Gper Pharmacology With... · Xenoestrogens As Gper...
  50. [50]
    Role of G Protein-Coupled Estrogen Receptor in Digestive System ...
    Apr 14, 2021 · ... MAPK/ERK and PI3K/AKT pathways. Additionally, the additional signals activated by GPER include PLC/IP3/calcium mobilization, PKC, and Hippo ...
  51. [51]
    NHERF1, a novel GPER associated protein, increases stability and ...
    Aug 23, 2016 · GPER is extensively expressed in numerous tissues such as breast, uterus, ovary and brain. It has been reported to play physiological roles ...
  52. [52]
    Role of G Protein-Coupled Estrogen Receptor in Cancer Progression
    Jul 15, 2019 · GPER is known to contribute to cancer cell proliferation, migration, and invasion in breast cancer (18).Gper-Activated Signaling... · Gper In Breast Cancer · Gper In Ovarian Cancer
  53. [53]
    Membrane Progesterone Receptors (mPRs, PAQRs) - NIH
    May 30, 2022 · However, mPRs are not members of the large GPCR superfamily and instead belong to the progestin and adipoQ receptor (PAQR) family, which has a ...
  54. [54]
    Anti-Inflammatory Actions of G-Protein-Coupled Estrogen Receptor ...
    Jan 9, 2023 · The study shows that brain-derived estrogen helps to mediate the anti-inflammatory effects of G-protein-coupled estrogen receptor 1 signaling.2.5. Aromatase Activity · 3. Results · 3.4. Gper Plays An Important...Missing: 2020s | Show results with:2020s
  55. [55]
    The modulation of potassium channels by estrogens facilitates ...
    Interestingly, administration of estrogens produces an inhibitory effect on the Kv and TASK-1 K2P channels but activates the BK, KATP, and TREK-1 K2P channels ...
  56. [56]
    Estrogenic Modulation of Ionic Channels, Pumps and Exchangers in ...
    Estrogen acutely increases the BKCa activity through the NO/GC/cGMP/PKG pathway phosphorylation of the β subunit, and when the SNP C818T is expressed, E2 ...
  57. [57]
    Allopregnanolone Activates GABA(A) receptor/Cl(-) Channels in a ...
    The results indicate that allopregnanolone interacts with GABA(A) receptor/Cl(-) channels expressed by embryonic hippocampal neurons in multiple ways, some of ...
  58. [58]
    Role of allopregnanolone-mediated γ-aminobutyric acid A receptor ...
    When ALLO binds to GABAA receptor, it will enhance the GABA evoked chloride current through increasing the frequency and/or duration of openings of the GABA- ...
  59. [59]
    Activation of the glucocorticoid receptor rapidly triggers calcium ...
    In close proximity to 5-HT releasing sites, activated GR rapidly triggers L-type VGCC-dependent vesicular 5-HT release.Missing: pools excitability
  60. [60]
    The vasodilatory action of testosterone: a potassium-channel ... - NIH
    Taken together, these four studies provide evidence that testosterone-induced vasodilatation occurs via potassium channel opening, having a modulatory effect ...
  61. [61]
    Effect of Testosterone on Potassium Channel Opening in Human ...
    Jan 18, 2008 · Testosterone (200 nM) significantly increased the single-channel activity of calcium-activated potassium (BK Ca ) channels and whole-cell K + currents by 443.4 ...
  62. [62]
    Ion Channel Regulation by Sex Steroid Hormones and Vitamin D in ...
    Here, we reviewed the SSH and vitamin D regulation of ion channels involved in cancer, and analyzed the potential molecular pathways implicated. In addition, we ...
  63. [63]
    Kinases and protein phosphorylation as regulators of steroid ... - NIH
    Many of the phosphorylation sites contain Ser/Thr-Pro motifs implicating proline-directed kinases such as the cyclin-dependent kinases and the mitogen-activated ...
  64. [64]
    Estrogens directly potentiate neuronal L-type Ca2+ channels - NIH
    Sep 30, 2008 · Recent studies have demonstrated that the gonadal steroid estrogen rapidly induces Ca2+ influx in hippocampal neurons, which is required for ...
  65. [65]
    Sex Hormone-Binding Globulin and Metabolic Syndrome in ... - MDPI
    SHBG is a 90–100 KDa homodimeric glycoprotein that transports testosterone and other steroids in the circulation with high affinity (KD ~1 nmol/L), reduces ...
  66. [66]
    Recent Advances on Sex Hormone-Binding Globulin Regulation by ...
    Jun 27, 2024 · Sex hormone-binding globulin (SHBG) is a homodimeric glycoprotein produced by the human liver and secreted into the systemic circulation ...
  67. [67]
    Associations of Sex-Hormone-Binding Globulin (SHBG) with Non ...
    Oct 27, 2004 · In normal men and women, between 40 and 65% of circulating testosterone (T) and between 20 and 40% of circulating estradiol (E2) is bound to ...
  68. [68]
    Sex hormone-binding globulin is synthesized in target cells
    **Summary of SHBG Receptor Discovery and Related Pathways:**
  69. [69]
    Expression of sex hormone-binding globulin, oxytocin receptor ...
    Our observations indicate an interaction of SHBG and OTR, associated with caveolin-1, which may account in part for known non-genomic actions of ovarian ...
  70. [70]
  71. [71]
    Sex hormone-binding globulin mediates steroid ... - PubMed - NIH
    SHBG binds to a receptor on cell membranes, transduces signal via G protein, activates adenylyl cyclase, and generates cAMP. Only certain steroids can activate ...Missing: caveolin- uptake endocytosis discovery 1990s
  72. [72]
    Sex hormone-binding globulin mediates steroid ... - ScienceDirect.com
    The SHBG–RSHBG complex causes the activation of adenylyl cyclase and the generation of cAMP within a matter of minutes after exposure to an appropriate steroid.
  73. [73]
    Human Sperm Sex Hormone-Binding Globulin Isoform
    Sperm SHBG isoform levels were significantly lower in semen samples with poor motility compared with specimens with good motility (Fig. 7). A multiple ...
  74. [74]
    The association of obesity with sex hormone-binding globulin is ...
    We found that the association between obesity and lowered SHBG is greater than the association of ageing with increased SHBG.
  75. [75]
    Human sex hormone–binding globulin variants associated with ... - JCI
    The genetic screening methods reported here will help identify individuals in whom genetic variants contribute to low serum SHBG levels, as opposed to others ...
  76. [76]
    Membrane-initiated actions of sex steroids and reproductive behavior
    Dec 1, 2021 · In this review we present a selection of these early data that changed the conceptual landscape and we provide a summary the different types of ...
  77. [77]
    Non-genomic actions of steroid hormones on the contractility of non ...
    Feb 17, 2024 · Non-genomic pathways of steroid actions are also known, mediated by cell membrane-located seven transmembrane domain receptors. Sex steroids ...
  78. [78]
    The Role of G Protein-Coupled Estrogen Receptor (GPER) in ... - NIH
    GPER phosphorylates eNOS through the non-genomic Akt/ERK pathway, which in turn promotes NO production. NO produced in ECs stimulates sGC in VSMCs to produce ...
  79. [79]
    Non-genomic Effects of Estrogen on Cell Homeostasis ... - Frontiers
    Estrogen increases NO bioavailability in the vascular system through both the signaling pathways (genomic and non-genomic). Through the non-genomic signaling, ...Abstract · Introduction · Estrogen and Estrogen... · Estrogen Regulated miRNAs...
  80. [80]
  81. [81]
    Membrane progesterone receptor induces meiosis in Xenopus ...
    Nov 2, 2020 · Because APPL1 is essential for P4-dependent oocyte maturation, we tested whether the interaction between APPL1 and mPR is regulated by P4.<|separator|>
  82. [82]
    Non-genomic Effects of Glucocorticoids: An Updated View - PubMed
    Nov 26, 2018 · In this review article, we discuss (i) the non-genomic effects of GCs on pathways relevant to the pathogenesis of inflammatory diseases and (ii) the putative ...
  83. [83]
    Sex hormone signaling and regulation of immune function - PubMed
    Nov 14, 2023 · The effect of sex steroids on immunity involves both the concentration of the ligand and the density and distribution of genomic and nongenomic ...Missing: actions | Show results with:actions
  84. [84]
    Making sense of cross-talk between steroid hormone receptors and ...
    This review provides an overview of the primary ways in which steroid hormone receptor and growth factor cross-talk occurs, using the human progesterone ...
  85. [85]
    Genome-wide crosstalk between steroid receptors in breast and ...
    Jul 22, 2021 · We review the current knowledge of the crosstalk between SRs in breast and prostate cancers. We emphasize how the activity of ER and AR on chromatin can be ...
  86. [86]
    Epidermal growth factor receptor (EGFR) transactivation by estrogen ...
    ... estrogen receptor (ER)-negative human breast cancer cells [Molecular Endocrinology 14 (2000) 1649]. This finding is consistent with a growing body of ...
  87. [87]
    Epidermal growth factor receptor (EGFR) transactivation by estrogen ...
    The first known receptor for estrogen, termed estrogen receptor (ER), was described based on its specific binding activity in extracts prepared from rat uterus ...
  88. [88]
    The glucocorticoid receptor and NFĸB in good times and bad
    This review will focus on GR and NFκB as a means by which to highlight interactions among the endocrine and immune systems and detail the repercussions ...
  89. [89]
    Nuclear Factor-κ B Repression in Antiinflammation and ...
    Because of their potent immunosuppressive action, glucocorticoids are highly effective in the treatment of allergic reactions and in preventing tissue rejection ...
  90. [90]
    Cross-talk between GPER and growth factor signaling - ScienceDirect
    In particular, the cross-talk between GPCRs and growth factor receptors has recently emerged as a signaling mechanism involved in cancer growth, angiogenesis ...
  91. [91]
    Antagonizing effects of membrane-acting androgens on the ... - Nature
    Mar 14, 2017 · ... beta-Catenin, CREB (cAMP response element-binding protein). Blotted ... Membrane androgen receptor activation induces apoptotic ...
  92. [92]
    Nuclear Integration of Glucocorticoid Receptor and Nuclear Factor ...
    Here we show that increased levels of CBP relieves the inhibition of glucocorticoid-mediated repression of NF-κB activity and the NF-κB-mediated repression of ...
  93. [93]
    Autoregulation of glucocorticoid receptor by cortisol in rainbow trout ...
    Together, our results suggest a negative feedback loop for GR gene regulation by cortisol in trout hepatocytes. The autoregulation of GR may be a crucial step ...
  94. [94]
    Overview of the relevance of PI3K pathway in HR-positive breast ...
    The regulatory crosstalk between PI3K and ER is bidirectional as not only upregulation of ER by PI3K inhibition mediates resistance to PI3K inhibitors but also ...
  95. [95]
    Glucocorticoid activation of anti-inflammatory macrophages protects ...
    Apr 20, 2023 · Macrophage glucocorticoid receptor regulates adipose tissue inflammation during obesity. With GR in macrophages being predicted as a major ...
  96. [96]
    Article DNA Binding of the Glucocorticoid Receptor Is Not Essential ...
    At composite elements (e.g., in the proliferin gene), the GR has to contact DNA together with another transcription factor whereas at tethering elements ...
  97. [97]
    Glucocorticoid Receptor Transcriptional Activity Determined by ...
    The first two types are typically induced (simple and paired elements) or repressed (tethering elements) by the receptor, whereas the third, the cGRE, can ...
  98. [98]
    Estrogen-receptor positive breast cancer: Diagnosis, response to ...
    Estrogen receptor–positive breast cancer is the most prevalent subtype, accounting for 70–80% of cases. The overall breast cancer incidence is about 86 cases ...Missing: overexpression prevalence
  99. [99]
    The future of androgen receptor targeting in prostate cancer
    Sep 29, 2025 · A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer ...
  100. [100]
    Glucocorticoid Resistance in Acute Lymphoblastic Leukemia: BIM ...
    Dec 10, 2018 · Numerous aspects of glucocorticoid receptor function and regulation have been implicated in glucocorticoid resistance. These include ...
  101. [101]
    Different types of androgen receptor mutations in patients with ...
    To date, over 500 unique mutations in the AR gene causing androgen insensitivity syndrome had been reported from more than 850 patients (http://androgendb.
  102. [102]
    Endometrial progesterone resistance and PCOS
    Jan 9, 2014 · Several studies have shown that the endometrial PR expression is altered in women with PCOS. See text for details. The normal endometrium is ...
  103. [103]
    Glucocorticoid receptor polymorphisms modulate cardiometabolic ...
    Feb 13, 2016 · Association of glucocorticoid receptor polymorphism A3669G with decreased risk of developing diabetes in patients with Cushing's syndrome.
  104. [104]
    Orphan Nuclear Receptors in Metabolic Dysfunction-associated ...
    Jun 19, 2025 · This review specifically focuses on elucidating the critical contributions of various ONRs to the pathogenesis of metabolic dysfunction- ...
  105. [105]
    G-protein coupled estrogen receptor 1, amyloid-β, and tau tangles in ...
    May 15, 2024 · Accumulation of amyloid-β (Aβ) and tau tangles are hallmarks of Alzheimer's disease (AD). It is hypothesized that Aβ accelerates tau ...
  106. [106]
    Review Neurosteroids and their receptors in ischemic stroke
    Neurosteroids receptors play important roles in neuroprotection mediated by these hormones. Membrane and intracellular receptors are both involved in the ...
  107. [107]
    Estrogen receptor alpha in human breast cancer - PubMed
    Over half of all breast cancers overexpress ER alpha and around 70% of these respond to anti-estrogen (for example tamoxifen) therapy.Missing: prevalence | Show results with:prevalence
  108. [108]
    The Effect of Selective Estrogen Receptor Modulators (SERMs ... - NIH
    Tamoxifen, the prototypical SERM, is extensively used for targeted therapy of ER positive breast cancers. For almost three decades, this drug has been as a ...
  109. [109]
    Tamoxifen - StatPearls - NCBI Bookshelf - NIH
    Mar 28, 2025 · Tamoxifen is a selective estrogen receptor modulator (SERM) medication used to treat breast cancer in both men and women and as a prophylactic agent against ...Continuing Education Activity · Indications · Mechanism of Action · Monitoring
  110. [110]
    Enzalutamide: A New Agent for the Prostate Cancer Treatment ... - NIH
    The indication for enzalutamide is treatment of patients with metastatic castration-resistant prostate cancer who have previously received docetaxel (Astellas ...
  111. [111]
    The Role of Glucocorticoids in Inflammatory Diseases - PMC
    Dex efficiently reduced lung inflammation and restored epithelial barrier integrity, effects that were mediated by the dimeric GR in myeloid cells (Table 3).
  112. [112]
    GPER mediates estrogen cardioprotection against epinephrine ...
    May 20, 2021 · In this study, we investigated the effects and mechanism of GPER on epinephrine (Epi)-induced cardiac stress. SCM was developed with a high dose ...Missing: clinical trials
  113. [113]
    GPER in metabolic homeostasis and disease - PubMed
    Aug 26, 2025 · Rapid advancements in GPER agonist and antagonist research have highlighted the therapeutic potential of GPER agonists for treating metabolic ...
  114. [114]
    Study Details | NCT03888612 | Trial of ARV-110 in Patients With ...
    Phase 1/2 dose escalation study to assess the safety and tolerability of ARV-110 in men with mCRPC who have progressed on prior approved systemic therapies ...
  115. [115]
    Study Details | NCT05654623 | ClinicalTrials.gov - ClinicalTrials.gov
    The purpose of this study is to learn about the safety and effects of the study medicine ARV-471 (PF-07850327, vepdegestrant) compared to fulvestrant (FUL)
  116. [116]
    Fresh insights into glucocorticoid-induced diabetes mellitus and new ...
    May 18, 2022 · Long-term use of glucocorticoids is severely hampered by undesirable metabolic complications, including the development of type 2 diabetes mellitus.
  117. [117]
    Selective steroid receptor modulators, degraders and PROTACs
    This review highlights recent advancements in SNuRMs and SNuRDs for managing NRs-associated endocrine/metabolic disorders.
  118. [118]
    Estrogen-Receptor Loss and ESR1 Mutation in Estrogen ... - PubMed
    Aug 30, 2024 · We conclude that ER loss and ESR1 mutation together account for 30% of the resistance to endocrine therapy. Keywords: ESR1 mutation; breast ...Missing: relapse | Show results with:relapse
  119. [119]
    Current uses and resistance mechanisms of enzalutamide ... - PubMed
    Sep 20, 2024 · Enzalutamide is an effective prostate cancer therapy that is currently used in biochemically-recurrent and metastatic disease, and for both castration ...