Fact-checked by Grok 2 weeks ago

Progesterone receptor

The progesterone receptor (PR) is a member of the nuclear receptor superfamily of ligand-activated transcription factors that binds the steroid hormone progesterone to modulate gene expression in target cells, playing a central role in reproductive development, menstrual cycle regulation, and pregnancy maintenance. Expressed primarily in female reproductive tissues such as the uterus, ovaries, and mammary glands, PR also functions in the brain, bone, and cardiovascular system, influencing processes like neuroprotection, bone formation, and vascular tone. Upon binding progesterone, which diffuses across the cell membrane due to its lipophilic nature, the receptor undergoes a conformational change, dissociates from chaperone proteins, dimerizes, and translocates to the nucleus where it binds to progesterone response elements (PREs) in DNA to activate or repress transcription. This genomic action is complemented by rapid non-genomic effects, such as activation of signaling pathways like MAPK and PI3K/Akt, which contribute to progesterone's diverse physiological impacts. Structurally, PR consists of an N-terminal domain containing activation function-1 (AF-1), a central (DBD) with zinc fingers for DNA recognition, a region for flexibility, and a C-terminal ligand-binding domain (LBD) that includes activation function-2 (AF-2) and nuclear localization signals. Humans express two primary isoforms, -A (94 ) and -B (114 ), arising from alternative promoter usage of the single PGR gene on chromosome 11q22; -B includes an additional 164-amino-acid N-terminal extension that confers unique transcriptional activities, while -A often acts as a transrepressor of -B, , and other nuclear receptors. A truncated isoform, -C, lacks the DBD and AF-1 but can modulate full-length activity. These isoforms exhibit tissue-specific expression and functions: -B predominantly drives proliferation and differentiation in mammary glands, whereas -A predominates in the to inhibit estrogen-induced growth and ensure proper endometrial receptivity. PR's regulatory mechanisms involve extensive post-translational modifications, including by kinases such as CDK2 and MAPK, which fine-tune sensitivity, DNA binding, and interactions with coactivators (e.g., SRC-1) or corepressors (e.g., NCoR). In reproductive contexts, PR orchestrates critical events like , implantation, and through networks involving factors such as and Hand2. Dysregulation of PR signaling contributes to pathologies including , uterine fibroids, and hormone-dependent cancers like breast and endometrial , where loss of PR expression or isoform imbalance correlates with poor prognosis. Therapeutically, selective progesterone receptor modulators (SPRMs) like exploit PR's structural dynamics to antagonize or partially agonize its activity for conditions such as and hormone-receptor-positive .

Discovery and Genetics

Historical Discovery

The discovery of the progesterone receptor (PR) in the 1960s built upon the pioneering methods developed for the by Elwood V. Jensen, who used tritium-labeled to identify specific in uterine and tissues. Applying similar radiolabeled progesterone assays, researchers identified high-affinity, saturable sites in target organs such as the chick oviduct and mammalian . In 1970, Bert W. O'Malley and colleagues reported the first evidence of a specific progesterone- in the and of chick oviduct cells, a progesterone-responsive tissue analogous to the mammalian , using sucrose to characterize the receptor complex. These studies demonstrated that progesterone was retained in target tissues longer than in non-target tissues, suggesting a receptor-mediated uptake mechanism. Early experiments further established PR as a . Autoradiographic studies in the early 1970s, such as those by Walter E. Stumpf and Madhabananda Sar, showed that injected [3H]-progesterone localized predominantly to the nuclei of epithelial cells in the rat and , with minimal binding in non-target tissues like muscle. This nuclear concentration occurred rapidly, even at low temperatures, indicating a direct association with nuclear components rather than passive diffusion. Concurrently, functional assays linked PR to progesterone's biological effects; for instance, O'Malley demonstrated that progesterone induced specific RNA and protein synthesis (e.g., ) in chick , and this induction was blocked by inhibitors of receptor binding, confirming the receptor's role in . These findings solidified PR's identity as a nuclear factor mediating progesterone's genomic actions in reproductive tissues. A major milestone came in 1987 with the of the human PR , led by teams including Marcelle Misrahi and Milgrom, who isolated cDNA from T47D cells and sequenced the full-length coding region, revealing a 933-amino-acid protein. This work demonstrated PR's membership in the superfamily, sharing structural homology with , , and receptors, particularly in the DNA-binding and ligand-binding domains. The enabled subsequent studies on PR's and isoforms, transforming understanding of its molecular basis. The was localized to 11q22 by . Independently, Pierre Loosfelt and colleagues cloned the PR cDNA in 1986, revealing high and confirming PR's evolutionary conservation. These advances highlighted PR's evolutionary conservation and its central role in hormone-responsive .

Gene Structure and Expression

The PGR gene, which encodes the progesterone receptor, is located on the long arm of human chromosome 11 at position 11q22.1 and spans approximately 90 kb of genomic DNA, comprising eight exons and seven introns. This organization allows for the transcription of multiple mRNA variants through alternative splicing, though isoforms primarily arise from differential promoter usage and translational starts. Expression of the PGR gene is tightly regulated by promoter regions upstream of the transcription start sites, involving interactions with various transcription factors. induces PGR transcription primarily through (ERα), which binds to composite estrogen response elements (EREs) overlapping Sp1 sites in the proximal promoter, rather than classical EREs; this mechanism facilitates cooperative activation by ERα and Sp1 proteins. Additional regulation occurs via other factors, such as AP-1 and C/EBP, which modulate basal and hormone-responsive expression in target cells. PGR exhibits tissue-specific expression patterns, with high levels in reproductive tissues including the , , and , where it supports progesterone-mediated functions like implantation and . Moderate to high expression is also observed in the (e.g., and ) and , contributing to and skeletal maintenance, respectively, while lower levels occur in tissues such as the and cardiovascular system. These patterns are influenced by hormonal cues and developmental stages, ensuring context-appropriate receptor availability. Epigenetic mechanisms, including and modifications, play a critical role in modulating PGR expression, particularly in pathological contexts like cancer. In normal tissues, the PGR promoter maintains low levels, permitting robust transcription; however, hypermethylation of CpG islands in the promoter is frequent in , endometrial, and other solid tumors, leading to transcriptional silencing and reduced receptor levels associated with poor prognosis. Similarly, repressive modifications, such as increased and decreased acetylation, correlate with downregulated PGR in cancer cells compared to normal counterparts, whereas (HDAC) inhibitors can restore expression by alleviating these marks. These epigenetic alterations highlight PGR as a and potential therapeutic target in hormone-responsive malignancies.

Protein Structure and Isoforms

Overall Architecture and Domains

The progesterone receptor (PR) is a member of the nuclear receptor superfamily characterized by a modular architecture consisting of four principal domains: the N-terminal domain (NTD), DNA-binding domain (DBD), hinge region, and C-terminal ligand-binding domain (LBD). The NTD, spanning residues 1–553 in the full-length PR-B isoform, encompasses activation function 1 (AF-1), an intrinsically disordered region that mediates ligand-independent transcriptional activation through interactions with coregulators. The DBD, located centrally (residues ~554–630), contains two zinc-finger motifs that recognize and bind to progesterone response elements (PREs) in target gene promoters via major groove interactions. The hinge region (residues ~631–688) serves as a flexible linker, facilitating conformational changes and containing a carboxyl-terminal extension (CTE) that interacts with the minor groove of DNA. The LBD (residues ~689–933), which houses activation function 2 (AF-2), adopts a helical sandwich fold typical of nuclear receptors, enabling ligand binding and coactivator recruitment. Within the LBD, the progesterone binding pocket is a hydrophobic cavity formed by 12 α-helices, where key residues such as Gln725 and Arg766 form hydrogen bonds with the 3-keto group of progesterone, while Asn719 contributes to water-mediated interactions stabilizing the ligand. Dimerization interfaces are prominent in both the DBD, which supports DNA-dependent dimer formation through direct and water-mediated contacts in the zinc-finger regions, and the LBD, where residues 885–922 mediate ligand-independent homodimerization essential for cooperative DNA binding. Nuclear localization signals (NLS) are distributed across domains, including a constitutive monopartite NLS in the hinge region (residues 638–642, homologous to the SV40 large T antigen sequence) and a ligand-inducible bipartite NLS overlapping the DBD and hinge, enabling active transport via importin-mediated nuclear import. Phosphorylation sites, primarily serine residues in the NTD (at least seven, including Ser81, Ser162, Ser294, and Ser345), regulate PR activity by modulating stability, DNA binding, and coactivator interactions; for instance, MAPK-mediated phosphorylation at Ser294 promotes ligand-induced ubiquitination and turnover. Recent structural studies using hydrogen-deuterium exchange mass spectrometry (HDX-MS) and cross-linking mass spectrometry (XL-MS) have provided high-resolution insights into full-length PR dynamics, revealing that progesterone binding induces repositioning of helix 12 in the LBD, which reshapes the AF-2 surface for coactivator binding and elongates the overall dimer structure upon DNA engagement. These techniques, achieving amino acid-level resolution, demonstrate sequential priming where ligand binding first stabilizes the LBD dimer interface, followed by DNA-induced conformational rigidification that reduces NTD-LBD proximity, contrasting with lower-resolution cryo-EM models (~10 Å) of PR-coregulator-DNA complexes that highlight domain flexibility but lack atomic details. Such findings underscore the allosteric interplay between domains, with the hinge and CTE playing pivotal roles in transmitting signals from ligand binding to DNA recognition.

Isoforms and Variants

The progesterone receptor (PR) exists primarily as two isoforms, PR-A and PR-B, which are transcribed from a single (PGR) located on 11q22 in humans. PR-B is the full-length isoform, consisting of 933 , while PR-A is a shorter variant lacking the first 164 at the , resulting in a protein of approximately 769 . This N-terminal region in PR-B, known as the B-upstream segment (BUS), contains an additional (AF-3) that enhances its transcriptional activity. These isoforms are generated through the use of alternative promoters and distinct transcription start sites within the PGR gene. PR-A is initiated from an upstream promoter (promoter A), which includes sequences from -711 to +31 relative to the translation start site, whereas PR-B arises from a downstream promoter (promoter B) spanning +464 to +1105. This alternative promoter usage allows for tissue-specific regulation of isoform expression, often influenced by signaling. Functionally, PR-B acts as a potent transcriptional activator, promoting progesterone-responsive , while PR-A exhibits repressive properties and can inhibit PR-B-mediated transcription on certain target genes, thereby fine-tuning progesterone responses. Beyond these main isoforms, several truncated and variants of PR have been identified, contributing to diverse signaling outcomes. PR-C is a notable truncated variant, approximately 60 kDa in size, that initiates translation at 595 and lacks both the N-terminal (including AF-1 and AF-3) and the (DBD), rendering it incapable of direct DNA binding to progesterone response elements (PREs). This isoform is generated via alternative translation start sites and has been implicated in inhibitory roles, potentially modulating full-length PR activity, though its physiological prevalence remains debated. PR-C may associate with membranes, suggesting involvement in non-genomic signaling. Splice variants such as PR-Δ (including subtypes like PR-Δ4, PR-Δ6, and PR-Δ4/6) arise from or deletion events in the PGR gene, often lacking the DBD due to removal of exons 4 and/or 6, which encode critical DNA-contacting residues. These variants retain ligand-binding capability but exhibit altered or absent genomic activity, potentially acting as dominant negatives or influencing non-genomic pathways. Other reported splice variants include PR-S, PR-T, and PR-M (a mitochondrial form lacking NTD and DBD), though their functions are less well-characterized and vary by context. The ratio of PR-A to PR-B is tightly regulated and tissue-specific, influencing overall progesterone responsiveness. In normal , the isoforms are typically expressed at near-equal levels (approximately 1:1), but in , PR-A often predominates, correlating with aggressive phenotypes and altered transcriptional outcomes where PR-A represses PR-B-driven activation of proliferation-associated genes. This imbalance can shift the balance toward inhibitory effects on signaling and other pathways.

Mechanism of Action

Genomic Signaling

The genomic signaling pathway of the progesterone receptor () represents the classical mechanism by which progesterone exerts its transcriptional effects. Upon binding to progesterone, the ligand-binding domain of undergoes a conformational change, leading to dissociation from inhibitory heat shock proteins (such as ), exposure of nuclear localization signals, and subsequent nuclear translocation of the receptor. This process facilitates homodimerization (or heterodimerization with other steroid receptors) through interactions in the , enabling the receptor complex to interact with . In the nucleus, the dimeric PR binds directly to progesterone response elements (PREs) in the promoter regions of target genes. The consensus PRE sequence is typically 5'-AGAACAnnnTGTTCT-3', where the central nnn represents variable nucleotides, allowing for some flexibility in binding affinity. This direct DNA binding recruits the general transcriptional machinery, including , to initiate gene transcription. Additionally, PR can indirectly regulate transcription through tethering mechanisms, such as interaction with (ER) bound to estrogen response elements, thereby facilitating cross-talk between progesterone and estrogen signaling pathways. PR-mediated transcription involves the recruitment of co-regulatory proteins to modulate accessibility and . Ligand-bound engages co-activators, such as steroid receptor co-activator 1 (SRC-1), via its function-2 (AF-2) domain in the ligand-binding region; SRC-1 possesses intrinsic activity that promotes and H4 acetylation, resulting in decompaction and enhanced transcriptional . Conversely, in the absence of ligand or under antagonistic conditions, can recruit co-repressors (e.g., NCoR or SMRT) that facilitate histone deacetylation via associated histone deacetylases, repressing gene . This dynamic balance of and deacetylation is crucial for fine-tuning transcriptional output. In physiological contexts like the , PR genomic signaling exhibits cyclical regulation, with estrogen-induced PR expression peaking in the late proliferative phase, followed by progesterone activation in the to drive endometrial differentiation and . This temporal orchestration ensures coordinated reproductive events, such as preparation for implantation, through sustained transcription of genes involved in and secretory transformation.

Non-Genomic Signaling

Non-genomic signaling by progesterone refers to rapid cellular responses initiated at the plasma membrane or , occurring within seconds to minutes and independent of transcription. These actions are mediated by membrane-associated progesterone receptors, including the distinct progestin and AdipoQ receptor (PAQR) members (mPRs) and membrane-localized forms of the classical PR isoforms. mPRs are characterized by seven transmembrane domains and G-protein capabilities, with isoforms including mPRα, mPRβ, mPRγ, mPRδ, and mPRε expressed across various tissues. Membrane-associated classical PR can initiate signaling via palmitoylation-dependent localization, activating pathways like . Upon progesterone binding, mPRs, particularly mPRα, trigger rapid activation of intracellular signaling cascades. This includes the /extracellular signal-regulated kinase (MAPK/ERK) pathway and the / (PI3K/Akt) pathway, often within minutes of exposure, leading to downstream effects such as cell survival and modulation. These pathways are frequently coupled through G-protein subtypes, with mPRα and mPRβ associating with inhibitory G_i proteins to inhibit , while others like mPRε link to stimulatory G_s proteins. Key effects of this non-genomic signaling include modulation of ion channels and cytoskeletal dynamics. In the brain, progesterone enhances GABA_A receptor function by potentiating chloride conductance, contributing to and neuroprotective responses, often via metabolites or direct mPR activation. In reproductive cells, such as oocytes and , mPR signaling induces cytoskeletal rearrangements, including polymerization that promotes oocyte maturation, meiotic resumption, and hypermotility. Non-genomic actions also exhibit cross-talk with classical nuclear PR, where membrane-initiated kinases phosphorylate nuclear PR, thereby enhancing its transcriptional activity and integrating rapid signals with slower genomic responses. For instance, Src/ERK activation by membrane PR can lead to phosphorylation of nuclear PR-B, amplifying progesterone's overall cellular effects without direct DNA binding.

Ligands and Modulation

Agonists

The primary natural agonist of the progesterone receptor (PR) is progesterone itself, which binds with high affinity (Kd ≈ 1 nM) to both PR-A and PR-B isoforms, initiating conformational changes that enable transcriptional activation. This endogenous steroid serves as the reference ligand for PR activation in reproductive tissues. Synthetic progestins, developed to mimic progesterone's effects with improved oral and potency, are classified structurally as - or 19-nortestosterone-derived, with the latter often further subclassified into generations based on chemical modifications and introduction timeline for use in oral contraceptives. -derived progestins include (), which exhibits high , with approximately 115% relative binding affinity (RBA) compared to progesterone. Among 19-nortestosterone-derived progestins, first-generation compounds such as norethindrone are derived from structures and exhibit moderate to high . Second-generation progestins, such as and , enhance progestational activity through structural modifications like the 17α-ethinyl group, which increases binding potency and reduces metabolism while conferring some androgenic properties ( RBA ≈ 50% relative to progesterone). Third-generation progestins, including and norgestimate, feature gonane backbones with further refinements at C-13 and C-17, yielding high selectivity and minimal estrogenic cross-reactivity. Fourth-generation progestins, such as , incorporate spiro ring structures derived from , providing strong alongside antimineralocorticoid activity to counter fluid retention. Structure-activity relationships among synthetic progestins highlight how modifications to the core influence PR binding and selectivity; for instance, the addition of a 17α-ethinyl to testosterone-derived scaffolds markedly boosts progestational potency by stabilizing receptor interactions in the ligand-binding domain, often exceeding that of progesterone. These compounds generally bind both PR isoforms, though some, like dydrogesterone, display preferential toward PR-B, potentially modulating isoform-specific transcriptional outcomes. In clinical applications, synthetic progestins are widely employed in contraception—via oral pills, implants, injections, and intrauterine devices—and hormone replacement therapy (HRT) to oppose estrogen effects in postmenopausal women, with choices like MPA and drospirenone selected for their favorable PR activation profiles in these contexts.

Antagonists and Mixed Modulators

Antagonists of the progesterone receptor (PR) are compounds that competitively bind to the ligand-binding domain (LBD) and inhibit PR-mediated transcriptional activity, primarily by repositioning helix 12 to occlude the co-activator binding groove and prevent recruitment of co-activators such as SRC-1. Mifepristone (RU-486), a prototypical pure antagonist, binds with higher affinity to the PR LBD than progesterone (approximately 2- to 3-fold higher, with relative binding affinities of 100% for mifepristone vs. 43% for progesterone), stabilizing a conformation that promotes PR dimerization but blocks transcriptional activation through genomic pathways. Structurally, mifepristone's 11β-dimethylaminophenyl side chain induces steric hindrance, leading to a flexible or displaced helix 12 that favors corepressor interactions over co-activator binding, as revealed by X-ray crystallography at 1.95 Å resolution. This mechanism contrasts with agonists, which position helix 12 to form a stable hydrophobic cleft for co-activator docking. Selective progesterone receptor modulators (SPRMs) exhibit tissue-specific mixed profiles, acting as antagonists in reproductive tissues like the while displaying partial agonistic effects in bone and cardiovascular systems. , an SPRM approved for and treatment, binds potently to PR with minimal antiglucocorticoid activity, modulating 12 to variably recruit co-regulators and reduce PR-B isoform levels while increasing the PR-A/PR-B ratio. In endometrial cells, it inhibits by downregulating progesterone-responsive genes like IL-15 and , leading to antiproliferative effects and reduced fibroid volume by 21-42%. Similarly, asoprisnil binds to PR with 3-fold higher affinity than progesterone, inducing a mixed conformation that suppresses endometrial proliferation and accumulation in uterine cells, though clinical development was halted due to risks. PR antagonists are classified into Type I and Type II based on their impact on DNA binding: Type I antagonists, such as onaprisone (ZK 98.299), impair PR dimerization and DNA association, preventing gene transcription without promoting receptor nuclear translocation; Type II antagonists, like mifepristone, allow DNA binding but induce a transcriptionally inactive conformation. This distinction arises from differential effects on the DNA-binding domain, with Type II compounds stabilizing abnormal receptor shapes that recruit corepressors. Tanaproget, while primarily a selective PR agonist, demonstrates partial agonistic activity in certain assays as a nonsteroidal modulator, contributing to its classification among mixed-profile compounds with reduced maximal transcriptional efficacy compared to full agonists. These modulators are utilized in emergency contraception by delaying ovulation and inhibiting endometrial receptivity, with mifepristone and ulipristal acetate exemplifying their role in blocking PR-dependent processes.

Molecular Interactions

Protein-Protein Interactions

The progesterone receptor (PR) functions primarily as a dimer, with its two major isoforms, PR-A and PR-B, capable of forming homodimers (PR-A/PR-A or PR-B/PR-B) or heterodimers (PR-A/PR-B). These dimeric complexes are stabilized upon and occur predominantly through interfaces in the (LBD), where specific helical regions facilitate inter-receptor contacts essential for DNA and . Studies in cell lines have demonstrated that these homo- and heterodimers differentially regulate target gene subsets, with PR-B homodimers often promoting activation while PR-A homodimers or heterodimers can exert inhibitory effects on PR-B activity. PR engages in cross-talk with other nuclear receptors, notably the (ER) and (AR), through direct or indirect protein-protein interactions that modulate their activities. Co-expression of PR and ER can lead to squelching, a competitive mechanism where both receptors vie for shared coactivators, resulting in mutual transcriptional inhibition; for instance, progesterone-activated PR sequesters ER complexes, redirecting them away from estrogen-responsive elements to suppress ER-driven in models. Similarly, interactions between PR and AR influence chromatin binding and in and breast cancers, with PR often reprogramming AR occupancy at shared response elements, highlighting a broader network of steroid receptor antagonism. Scaffold proteins such as 14-3-3 isoforms interact with PR to influence its localization and function. Specifically, 14-3-3τ, a progesterone-responsive gene product, binds PR in uterine cells to enhance its transcriptional activity, potentially by stabilizing receptor conformation or facilitating co-regulator recruitment. This interaction underscores the role of 14-3-3 proteins in modulating PR signaling within reproductive tissues. Post-translational modifications like SUMOylation critically regulate PR activity through interactions with protein inhibitor of activated STAT (PIAS) family members. SUMO-1 conjugation at specific residues in the PR N-terminal domain, mediated by PIAS3, promotes transcriptional repression by inhibiting coactivator binding and altering interactions; this modification is particularly prominent in PR-B and contributes to fine-tuning ligand-dependent responses in and endometrial cells.

Co-Regulators and Chromatin Effects

The progesterone receptor (PR) interacts with various co-activators to enhance its transcriptional activity, primarily through histone modifications and recruitment of the basal transcription machinery. Key among these are the steroid receptor coactivator (SRC) family members (SRC-1, SRC-2, SRC-3), which bind ligand-activated PR via LXXLL motifs in their nuclear receptor boxes and serve as adapter proteins to recruit secondary coactivators. Recent structural studies using hydrogen-deuterium exchange mass spectrometry (HDX-MS) and cross-linking have revealed a sequential priming mechanism: SRC-3 first binds to stabilize PR homodimers and induce conformational changes, followed by recruitment of p300 for histone acetylation and chromatin opening. Co-activators such as p300 and CREB-binding protein (CBP) function as histone acetyltransferases (HATs), acetylating histones to promote chromatin decondensation and facilitate PR access to target promoters. For instance, the PR N-terminal and ligand-binding domains bind p300/CBP-associated factor (PCAF), another HAT, independent of ligand status, enabling synergistic activation of progesterone-responsive genes like p21(WAF1) via Sp1 sites. Additionally, the Mediator complex, particularly its subunit MED1 (also known as TRAP220), is recruited by ligand-bound PR to bridge the receptor with RNA polymerase II (Pol II), stabilizing the pre-initiation complex and promoting Pol II phosphorylation for efficient transcription elongation. In contrast, co-repressors such as corepressor (NCoR) and silencing mediator for and hormone receptors (SMRT) inhibit PR-mediated transcription by recruiting deacetylases (HDACs) that compact and block activator access. The PR-A isoform exhibits a higher affinity for SMRT than PR-B, mediated by its inhibitory domain, which enables PR-A to actively repress and counteract PR-B activation on shared targets. Antagonist-bound PR, such as with RU486, similarly favors co-repressor recruitment over co-activators, shifting the co-regulator balance to promote repression and effects depending on cellular context. This recruitment is stabilized by direct interactions between the PR ligand-binding domain and the co-repressor interaction domains, often enhanced in the absence of agonists. PR also influences higher-order structure by inducing long-range looping interactions between enhancers and promoters, facilitated by the architectural protein and . Ligand-activated PR promotes CTCF/-mediated loops that position distal enhancers near progesterone-responsive , such as Ihh and Fst in the , enabling coordinated activation essential for tissue maturation. Disruption of impairs these loops, selectively reducing expression of PR-dependent without affecting global organization. During the , co-regulator dynamics in the shift in response to progesterone fluctuations, contributing to tissue remodeling. Progesterone , as occurs at the cycle's end or term , leads to a decline in co-activator levels (e.g., SRC-2/3 and CBP) and reduced , effectively switching the balance toward co-repressors like NCoR and SMRT to antagonize residual function and facilitate endometrial shedding or labor initiation. This co-regulator switch is evident in cyclic variations of NCoR and SMRT expression, which peak in the secretory phase to fine-tune progesterone's decidualizing effects before promotes inflammatory and contractile responses.

Physiological Functions

Reproductive and Developmental Roles

The progesterone receptor (PR) plays a pivotal role in preparing the for implantation through genomic signaling that induces , the transformation of endometrial stromal cells into decidual cells essential for supporting . This process involves PR-mediated , including the activation of such as HOXA10 and HOXA11, which are critical for endometrial receptivity and proper uterine patterning during implantation. In PR knockout mice, disruption of this signaling leads to defective decidualization and , underscoring PR's necessity for successful attachment and early establishment. In development, PR drives lobuloalveolar growth during , promoting the and of alveolar epithelial cells to form milk-producing structures. Progesterone acting via PR stimulates , including the expression of and , which coordinate ductal branching and alveolar in coordination with . Studies in PR-null mice reveal severely impaired lobuloalveolar development and reduced capacity, with glands exhibiting limited alveolar budding and insufficient secretory , leading to pup post-partum. PR contributes to ovarian function by modulating follicle maturation and through both inhibitory and inductive mechanisms. In s, progesterone via PR slows granulosa cell mitosis, thereby regulating follicle growth and preventing premature maturation, which maintains ovarian cyclicity. Conversely, PR is indispensable for , as it transcriptionally activates genes like Adamts1 and Has2 in preovulatory follicles in response to the surge, facilitating cumulus expansion and follicular rupture; PR-deficient mice are completely anovulatory. At the , progesterone mediates to prevent maternal immune rejection of the , primarily through non-genomic signaling pathways that rapidly modulate immune cell activity via non-classical membrane progesterone receptors (mPRs). Progesterone binding to these membrane receptors inhibits T-cell proliferation and production by blocking channels and , fostering a tolerogenic environment at the maternal-fetal interface. This non-genomic action, observed in decidual natural killer cells and T lymphocytes, promotes regulatory T-cell expansion and reduces pro-inflammatory responses, essential for placental maintenance and viability.

Behavioral and Neurological Effects

The progesterone receptor (PR) plays a significant role in , particularly in models of , where of PR by progesterone reduces neuronal damage and promotes functional . In experimental models, such as occlusion in , progesterone administration via PR signaling pathways mitigates infarct size and by modulating anti-inflammatory responses and enhancing cell survival mechanisms, including upregulation of (BDNF). For instance, selective PR agonists like nestorone have demonstrated long-term neuroprotective effects against permanent focal cerebral ischemia in both adult and aged male rats, highlighting PR's direct involvement in reducing and neuronal loss. These effects are PR-dependent, as evidenced by studies showing diminished neuroprotection in PR knockout models under similar ischemic conditions. In the realm of sexual behavior, in the is crucial for regulating reproductive behaviors, notably in female rodents. Activation of in the ventromedial by progesterone facilitates , a reflexive essential for mating, through transcriptional regulation of downstream genes that enhance behavioral receptivity following priming. Studies in female mice demonstrate that genetic or pharmacological blockade of impairs the development and expression of , underscoring its role in integrating hormonal signals for estrous behavior. In humans, fluctuations in progesterone levels across the , mediated by , are associated with mood variations, where luteal-phase elevations correlate with symptoms such as irritability and emotional lability, potentially through modulation of hypothalamic stress responses. PR isoforms, particularly PR-A, contribute to effects and influence and . Activation of PR by progesterone reduces anxiety-like behaviors in models, as shown in elevated plus-maze tests where progesterone administration increases time spent in open arms, an effect blocked by PR antagonists. studies in mice reveal that PR deficiency leads to reduced , such as lack of infanticidal behavior in males, indicating PR's role in promoting aggressive responses via signaling in limbic regions. Cognitively, PR signaling supports , with progesterone enhancing hippocampal function in tasks like spatial learning, though these effects are more pronounced in females during hormonal cycles. Non-genomic actions of progesterone enable rapid modulation of receptors, contributing to effects primarily through its metabolite . This directly enhances receptor sensitivity to , increasing influx and hyperpolarization in neurons, which promotes sedation and reduces excitability within minutes of progesterone exposure. This rapid pathway, distinct from nuclear transcription, is implicated in the calming effects observed in stress models, where progesterone-derived neurosteroids like amplify inhibition for anxiolysis and sleep induction.

Genetic Variations and Pathophysiology

Functional Polymorphisms

Functional polymorphisms in the progesterone receptor () gene represent common genetic variants that modulate receptor expression, isoform balance, interactions, and transcriptional activity without typically causing overt . These variants contribute to subtle differences in progesterone signaling efficiency across individuals, as evidenced by assays demonstrating variations in potential and isoform ratios. Key examples include the PROGINS , the Val660Leu coding variant, and promoter region polymorphisms such as +331G/A, each with distinct biochemical consequences. However, associations with disease risks, such as cancers or , remain inconsistent across studies. The PROGINS haplotype (also known as the A2 ) arises from an Alu insertion/deletion in 7 of the PGR gene, accompanied by linked single nucleotide polymorphisms (SNPs) including rs1042838 (Val660Leu) in exon 4 and rs1042839 (His770His, silent) in exon 5. This results in a higher PR-A to PR-B isoform ratio due to altered splicing efficiency and reduced overall PR transcript levels. In functional studies using luciferase reporter assays in mammalian cell lines, PROGINS-containing PR exhibits diminished activity in response to progesterone, with approximately 20-30% reduced transcriptional compared to wild-type PR, reflecting lower responsiveness to stimulation. The Val660Leu polymorphism (rs1042838), situated in the region adjacent to the LBD, is a core component of the PROGINS and influences interactions independently in some contexts. This missense variant substitutes for at position 660, leading to conformational changes. Although isolated expression of Val660Leu shows transcriptional properties largely similar to wild-type in transactivation reporter systems, its linkage within the PROGINS amplifies functional deficits, including mildly impaired sensitivity and altered receptor stability. Promoter variants, exemplified by the +331G/A polymorphism (rs10885030), directly impact PR isoform expression by altering transcriptional initiation. The A introduces a novel start site approximately 67 upstream, preferentially driving PR-B transcription and increasing the PR-B/PR-A ratio. Quantitative RT-PCR and analyses reveal a 17.8% elevation in PR-B mRNA and corresponding protein levels for the variant, while promoter assays demonstrate 2.1-fold higher basal and hormone-induced transcriptional activity. This polymorphism is associated with modified sensitivity and has been linked to increased risk through enhanced PR-B-mediated signaling. In vitro functional assays across these polymorphisms consistently highlight modest quantitative differences, such as 20-30% variations in efficiency for certain SNPs in systems, underscoring their role in fine-tuning responsiveness rather than abolishing function. These effects are typically assessed in cell models like or Ishikawa lines transiently transfected with variant PR constructs and progesterone-responsive reporters.

Mutations and Disease Associations

Loss-of-function mutations in the progesterone receptor gene (PGR) are rare in humans, with limited reports of frameshift or nonsense variants directly causing reproductive disorders such as or amenorrhea. However, animal models provide critical insights into the phenotypes associated with PR ablation. In progesterone receptor (PRKO) mice, complete loss of PR function results in characterized by failure to ovulate, impaired transport, uterine , and defective endometrial necessary for implantation. These mice also display increased mammary tumorigenesis, highlighting PR's tumor-suppressive role in reproductive tissues. Somatic alterations in PGR, including mutations and epigenetic silencing leading to reduced PR expression, are implicated in cancer progression. In endometrial cancer, loss of PR expression correlates with advanced invasive disease, higher tumor grade, and resistance to progestin-based therapies, as PR normally mediates progesterone's antiproliferative effects on endometrial cells. This reduction often arises from promoter hypermethylation or transcriptional repression rather than coding mutations. In , variants in PGR occur in approximately 67% of metastatic receptor-positive tumors, though the majority are single polymorphisms with uncertain functional impact; deleterious variants, such as those in the ligand-binding (e.g., Y890C), may impair PR activity and contribute to endocrine . Studies of primary PR-positive tumors report mutations in PGR at low frequencies, approximately 2%, often co-occurring with alterations in other signaling pathways. These changes can lead to ligand-independent dysregulation, though activating gain-of-function mutations in PGR remain infrequently documented compared to those in the .

Clinical Significance

Role in Cancers

The progesterone receptor (PR) plays a significant role in , where its expression status serves as a key prognostic indicator. Approximately 70% of breast cancers express PR, and PR-positive tumors are generally associated with a more favorable outcome compared to PR-negative ones, including improved overall survival and reduced risk of recurrence. In estrogen receptor-positive breast cancers, PR positivity correlates with longer median overall survival, reflecting a more differentiated tumor responsive to hormonal influences. Loss of the PR-B isoform, often due to an imbalanced PRA/PRB ratio favoring PRA overexpression, promotes tumor invasiveness and by enhancing and altering profiles that support epithelial-to-mesenchymal transition. In endometrial and ovarian cancers, PR expression is crucial for the efficacy of progestin-based therapies, particularly in hormone receptor-high tumors. High PR levels predict better responses to progestins such as , which inhibit tumor by downregulating signaling and inducing arrest in early-stage endometrial cancers. However, resistance to progestin frequently arises from isoform imbalances, where PRA dominance suppresses PR-B-mediated inhibition, leading to persistent despite . In ovarian cancers, similar patterns occur, with PR-high tumors showing initial sensitivity to progestins, though isoform dysregulation contributes to progression in advanced cases. Mechanistically, PR contributes to tumorigenesis through cross-talk with other signaling pathways. In , PR interacts with (ER) to modulate transcriptional responses, where PR loss enhances ER-driven and endocrine resistance. Additionally, PR signaling intersects with the PI3K/AKT pathway, activating downstream effectors that promote cell survival and invasion in hormone-responsive tumors. Non-genomic actions of PR further drive by rapidly activating MAPK and PI3K pathways independent of nuclear transcription, facilitating cytoskeletal rearrangements and enhanced motility in breast and cells. Epidemiologically, higher PR expression levels are linked to superior responses to across breast cancers, as evidenced by post-2020 meta-analyses demonstrating reduced recurrence risk and improved in PR-high versus PR-low cohorts. These findings underscore PR as a for therapy stratification, with low PR expression indicating poorer endocrine therapy outcomes and higher metastatic potential.

Therapeutic Applications

Progestins, which act as agonists at the (), are routinely combined with in () for postmenopausal women with an intact to mitigate the risk of induced by unopposed exposure. This approach counteracts 's proliferative effects on the by promoting secretory changes and reducing incidence, with supplementation recommended for at least 10-14 days per cycle in continuous or sequential regimens. Clinical evidence demonstrates that progestin addition resolves in approximately 94% of cases associated with -only , establishing it as a standard preventive measure. In contraception, synthetic progestins such as , a agonist, are key components of combined oral contraceptive pills, where they inhibit , thicken cervical mucus, and alter endometrial receptivity to prevent pregnancy. These formulations, typically pairing with , achieve high efficacy rates exceeding 99% with perfect use by suppressing release and stabilizing the . antagonists, including , serve as selective progesterone receptor modulators (SPRMs) for medical termination of pregnancy, blocking PR-mediated and sensitizing the uterus to prostaglandins like for effective evacuation. , administered at 200 mg followed by , completes in over 95% of cases up to 10 weeks , offering a non-surgical alternative with reduced complications compared to aspiration alone. For cancer therapy, has been explored as a antagonist in treatment due to frequent expression in these tumors, with early studies showing tumor stabilization in up to 60% of unresectable cases during long-term administration at 200 mg daily. However, a phase III randomized trial (SWOG S9005) failed to demonstrate superiority over in for inoperable meningiomas, limiting its routine use despite tolerability and occasional radiographic responses in diffuse or recurrent subtypes. In breast cancer, onapristone, a type I antiprogestin, was investigated in the phase II trial, where combination with yielded clinical benefit rates of approximately 40% based on data in ER-positive, HER2-negative metastatic disease previously treated with endocrine therapy and CDK4/6 inhibitors. Emerging therapeutic strategies target PR isoforms PRA and PRB, which exhibit distinct transcriptional activities, with recent research highlighting isoform-specific modulators to overcome resistance in hormone-dependent cancers. For instance, high PRA:PRB ratios correlate with poor prognosis and resistance in , prompting of agents that selectively enhance PRB-mediated anti-proliferative effects or degrade PRA-dominant complexes. Preclinical models demonstrate that isoform-biased SPRMs can restore progesterone responsiveness in endometrial and breast tumors by modulating enhancer activation and coregulator recruitment, paving the way for precision therapies in PR-positive malignancies. Additionally, proteolysis-targeting chimeras (PROTACs) designed against receptors show promise for degrading PR in resistant tumors, though PR-specific degraders remain in early discovery phases with off-target PR effects observed in PROTAC studies.

References

  1. [1]
    Physiology, Progesterone - StatPearls - NCBI Bookshelf - NIH
    May 1, 2023 · Progesterone increases the process of bone formation by stimulating P-4-receptor mediated osteoblastic growth during the initiation of bone ...
  2. [2]
    Progestins - StatPearls - NCBI Bookshelf - NIH
    Jan 10, 2024 · When unbound, the progesterone receptor exists as a monomer. After binding progesterone, the receptor undergoes a conformational change and ...
  3. [3]
    Ancient and modern mechanisms compete in progesterone receptor ...
    The progesterone receptor (PR) belongs to the steroid receptor family of ligand-regulated transcription factors, controlling genes important for development, ...
  4. [4]
    Progesterone Receptors, their Isoforms and Progesterone ... - NIH
    This review discusses mechanisms by which progesterone receptors (PR) regulate transcription. We examine available data in different species and tissues.
  5. [5]
    Progesterone Receptor Signaling Mechanisms - PubMed - NIH
    Progesterone receptor (PR) is a master regulator in female reproductive tissues that controls developmental processes and proliferation and differentiation.
  6. [6]
    Progesterone "receptors" in the cytoplasm and nucleus of ... - PubMed
    This report demonstrates that the chick oviduct, a specific target organ for progesterone, contains both cytoplasmic and nuclear macromolecules which bind ...Missing: 1967 | Show results with:1967
  7. [7]
    Complete amino acid sequence of the human progesterone receptor ...
    Complete amino acid sequence of the human progesterone receptor deduced from cloned cDNA ... Biochem Biophys Res Commun. 1987 Mar 13;143(2):740-8.
  8. [8]
    The human progesterone receptor shows evidence of adaptive ...
    Structure of Progesterone Receptor. The gene encoding human PR has eight exons and is located on chromosome 11q22.1. PR is characterized by alternatively ...
  9. [9]
    5241 - Gene ResultPGR progesterone receptor [ (human)] - NCBI
    Sep 5, 2025 · Data suggest expression of PGR in endometrium in women with endometriosis may be biomarker for subsequent fertility; PGR levels in normal range ...
  10. [10]
    Differential regulation of the human progesterone receptor gene ...
    Our studies provide evidence that effective regulation of transcription through the +571 ERE/Sp1 site requires the binding of ERalpha and Sp1 to their ...Missing: PGR | Show results with:PGR
  11. [11]
    Progesterone induces progesterone receptor gene (PGR ... - NIH
    Progesterone Receptors (PRs) are critical effectors of estrogen receptor (ER) signaling required for mammary gland development and reproductive proficiency.1. Introduction · 3. Results · 4. Discussion
  12. [12]
    regulation by progesterone receptor (PR) isoform expression ...
    Dec 31, 2009 · Progesterone is a critical regulator of normal female reproductive function, with diverse tissue-specific effects in the human.Missing: PGR | Show results with:PGR
  13. [13]
    Tissue expression of PGR - Summary - The Human Protein Atlas
    Summary of PGR (NR3C3, PR) expression in human tissue. Selective nuclear expression in female reproductive system and smooth muscle.
  14. [14]
    Loss of progesterone receptor through epigenetic regulation is ... - NIH
    Loss of progesterone receptor through epigenetic regulation is associated with poor prognosis in solid tumors.
  15. [15]
    Association of PRA Promoter Methylation with Worse Outcome in ...
    Other epigenetic regulations, such as histone modifications, are likely to be involved, given that treatment with HDAC inhibitor induces re-expression of the PR ...
  16. [16]
    Epigenetic modification restores functional PR expression ... - PubMed
    Our findings reveal that epigenetic modulators can restore endogenous functional PR expression in endometrial cancer cells and suggest that strategies to re- ...Missing: PGR | Show results with:PGR
  17. [17]
    Structural proteomics defines a sequential priming mechanism for ...
    May 12, 2025 · The progesterone receptor (PR) is a steroid-responsive nuclear receptor with two isoforms: PR-A and PR-B. Disruption of PR-A:PR-B signaling ...
  18. [18]
    Structural and Functional Analysis of Domains of the Progesterone ...
    This review focuses on studies of the CTE and NTD of progesterone receptor (PR), as well as related work with other steroid/nuclear receptors.2. Ligand Binding Domain... · 3. Dna Binding Domain (dbd) · 6. Amino-Terminal Domain...
  19. [19]
    Structure of the Progesterone Receptor-Deoxyribonucleic Acid ... - NIH
    The DNA binding domain (DBD) of nuclear hormone receptors contains a highly conserved globular domain and a less conserved carboxyl-terminal extension (CTE).Missing: phosphorylation | Show results with:phosphorylation
  20. [20]
    X-ray Structures of Progesterone Receptor Ligand Binding Domain ...
    Residues forming a hydrophobic pocket surrounding the 17α group are also shown. No direct or indirect polar interaction exists between the ligand and Asn719.Missing: Gly- 722 727
  21. [21]
    Nucleocytoplasmic shuttling of the progesterone receptor - PMC - NIH
    The nuclear localization of the progesterone receptor is mediated by two signal sequences: one is constitutive and lies in the hinge region.Missing: NLS | Show results with:NLS
  22. [22]
    Phosphorylation of human progesterone receptors at serine-294 by ...
    Thus, specific phosphorylation of PR on serine-294 by MAPK appears to be required for progestin-induced PR ubiquitination and degradation by the 26S proteasome.
  23. [23]
  24. [24]
  25. [25]
  26. [26]
    [Progesterone receptor isoforms: function and regulation] - PubMed
    PR-B acts as a transcriptional activator in different cellular contexts whereas PR-A functions as a strong inhibitor of transcriptional activity. PR isoforms ...
  27. [27]
    New insights into the functions of progesterone receptor (PR ...
    Progesterone receptors were first cloned in 1986 using the same strategies, simultaneously in the laboratories of Pierre Chambon, Edwin Milgrom, and Bert O' ...
  28. [28]
    PGR (progesterone receptor)
    Feb 1, 2017 · The third isoform is the splice variant PR-C. PR-C can bind the ligand, but in the absence of the DBD would not be able to bind to PREs of ...
  29. [29]
  30. [30]
    Integration of Progesterone Receptor Action with Rapid Signaling ...
    After binding to progesterone, the receptors undergoe restructuring, dimerization, and hsp dissociation. ... Following ligand binding, PR undergoes rapid ...
  31. [31]
    Progesterone receptor signaling in the initiation of pregnancy and ...
    Therefore, the AF-2 functional domain is responsible for ligand binding, dimerization, and also the translocation of the dimer into the nucleus. The presence of ...
  32. [32]
    Tissue-specific progesterone receptor-chromatin binding and the ...
    Aug 19, 2019 · In uterus PGR showed far more tendency to bind intergenic chromatin regions and low evidence of interaction with other transcription factors.
  33. [33]
    Steroid Receptor Coactivators 1, 2, and 3: Critical Regulators of ...
    SRC-1 overexpression enhances ligand-induced transcriptional activation by progesterone receptor ... co-activator SRC-1. EMBO J. 2007;26:3534–3544. doi: 10.1038/ ...
  34. [34]
    Dynamics of coactivator recruitment and chromatin modifications ...
    ... co-activators and co-repressors ... Aoyagi S, Archer TK. Dynamic histone acetylation/deacetylation with progesterone receptor-mediated transcription.Introduction · Nuclear Receptor Mediated... · Future Directions
  35. [35]
    Progesterone and Estrogen Signaling in the Endometrium - MDPI
    ... menstrual cycle and promote an embryo-receptive state to allow implantation during the window of receptivity ... progesterone receptor (PGR) and estrogen ...2. Steroid Hormone... · 2.1. Progesterone Receptors... · 3.1. Progesterone Resistance<|control11|><|separator|>
  36. [36]
  37. [37]
  38. [38]
  39. [39]
  40. [40]
  41. [41]
  42. [42]
  43. [43]
    Analysis of Progesterone Receptor Binding in the Ovine Uterus
    The dissociation constant (Kd) measured for the 3H-P4 binding was 1.60 x 10-9 M indicating that the 3H-P4 binding was of high affinity.
  44. [44]
    Recent advances in structure of progestins and their binding to ...
    The aim of this review is to describe the basic structure of PR agonists and antagonists as well as the recent treatments for illness associated with the ...
  45. [45]
    Progestins Related to Progesterone and Testosterone Elicit ... - MDPI
    Even subtle changes in progesterone structure or synthetic progestins with progestational activity and varied binding affinities for PR and other steroid ...
  46. [46]
    The progestin revolution 2 - NIH
    Oct 9, 2023 · There have been four generations of progestins. Each generation has different qualities that have additional benefits and some added risks ...
  47. [47]
    The X-ray Structure of RU486 Bound to the Progesterone Receptor ...
    ... progesterone receptor (PR). The structure was obtained by taking a ... A, overview of the total LBD; B and C, detailed views of the ligand binding pocket.
  48. [48]
    Selective Progesterone Receptor Modulators—Mechanisms and ...
    Ulipristal acetate has strong antagonistic and partial agonistic effects at PR in humans (235). Once this SPRM binds to PR, it reduces the binding capacity of ...
  49. [49]
    Progesterone receptor and the mechanism of action of ... - PubMed
    This paper discusses both published and unpublished data supporting the concept of two types of progestin antagonists.
  50. [50]
    New human breast cancer cells to study progesterone receptor ...
    In human breast cancer cells, homodimers of progesterone-occupied PR-A or PR-B regulate different gene subsets. To study PR homo- and heterodimers, we ...
  51. [51]
    DNA bending is induced by binding of the glucocorticoid receptor ...
    The PR-B:B and PR-A:A homodimers and the PR-A:B heterodimers all induced similar DNA bending angles of 72-77 degrees. The substantially greater DNA bend induced ...
  52. [52]
    ERα/PR crosstalk is altered in the context of the ERα Y537S ... - Nature
    Nov 30, 2023 · However, the complex relationship between ERα and the progesterone receptor (PR), known as ERα/PR crosstalk, has yet to be characterized in the ...
  53. [53]
    Renewed interest in the progesterone receptor in breast cancer
    Sep 22, 2016 · In essence, progesterone was able to redirect ER-mediated transcription via sequestration of the ER complex to inhibit breast tumour growth ...<|separator|>
  54. [54]
    Genome-wide crosstalk between steroid receptors in breast and ...
    Jul 22, 2021 · Especially, progesterone (PR) and glucocorticoid receptor (GR) can reprogram chromatin binding of ER and gene programs in breast cancer cells.
  55. [55]
    The progesterone-responsive gene 14-3-3τ enhances the ...
    The progesterone-responsive gene 14-3-3τ enhances the transcriptional activity of progesterone receptor in uterine cells ... The 14-3-3 proteins form a ...
  56. [56]
    PIAS3 induction of PRB sumoylation represses PRB transactivation ...
    Sumoylation of the progesterone receptor and of the steroid receptor coactivator SRC-1. ... PIASy-mediated repression of the androgen receptor is ...
  57. [57]
    Steroid receptor induction of gene transcription: a two-step model
    Jul 22, 1997 · Here we show that the human PR also interacts with p300/CBP-associated factor in vitro. Recruitment of multiple HATs to target promoters ...
  58. [58]
    Progesterone regulates transcription of the p21(WAF1) cyclin
    Apr 24, 1998 · Progesterone regulates transcription of the p21(WAF1) cyclin- dependent kinase inhibitor gene through Sp1 and CBP/p300. J Biol Chem. 1998 Apr ...
  59. [59]
    Progesterone and Glucocorticoid Receptors Recruit Distinct ...
    This study indicates that differential recruitment of coactivators by nuclear receptors determines the assembly of coactivator complexes on target promoters.
  60. [60]
    The opposing transcriptional activities of the two isoforms ... - PubMed
    The opposing transcriptional activities of the two isoforms of the human progesterone receptor are due to differential cofactor binding · Authors · Affiliation.
  61. [61]
    The partial agonist activity of antagonist-occupied steroid receptors ...
    Our data suggest that the direction of transcription by antagonist-occupied steroid receptors can be controlled by the ratio of coactivators to corepressors ...
  62. [62]
    The Nuclear Corepressors NCoR and SMRT Are Key ... - PubMed
    The Nuclear Corepressors NCoR and SMRT Are Key Regulators of Both Ligand- And 8-bromo-cyclic AMP-dependent Transcriptional Activity of the Human Progesterone ...
  63. [63]
    Chromatin architectural factor CTCF is essential for progesterone ...
    Receptors for estrogen and progesterone frequently interact, via Cohesin/CTCF loop extrusion, at enhancers distal from regulated genes.<|separator|>
  64. [64]
    A decline in the levels of progesterone receptor coactivators in the ...
    Aug 6, 2025 · Many mechanisms have been proposed to underlie functional progesterone withdrawal, including local breakdown of progesterone in the endometrium ...<|control11|><|separator|>
  65. [65]
    Cyclic Changes in the Expression of Steroid Receptor Coactivators ...
    Our study also demonstrated the expression of corepressors NCoR and SMRT in the endometrium. ... progesterone receptor(s);. SDS,. sodium dodecyl sulfate ...
  66. [66]
    Progesterone Receptor Signaling in the Uterus Is Essential for ... - NIH
    Apr 27, 2022 · Progesterone signaling through PR controls endometrial homeostasis and directs the transcriptional program during decidualization [30]. Studies ...
  67. [67]
    HOXA10 regulates endometrial GABAA π receptor expression and ...
    Estrogen and progesterone regulate endometrial expression of HOXA10. HOXA10 expression rises dramatically at the time of implantation, suggesting a role for ...
  68. [68]
    The Biology of Progesterone Receptor in the Normal Mammary ...
    This paper reviews work on progesterone and the progesterone receptor (PR) in the mouse mammary gland that has been used extensively as an experimental model.
  69. [69]
    Key stages in mammary gland development - The alveolar switch
    The progesterone receptor (Pgr) knockout mouse demonstrated that Pg is required for alveolar morphogenesis, and epithelial recombination experiments ...
  70. [70]
    Progesterone receptors - animal models and cell signaling in breast ...
    The progesterone receptor knockout mouse demonstrated progesterone's importance to parity-induced mammary tertiary branching and lobuloalveologenesis.
  71. [71]
    Progesterone Signaling and Mammalian Ovarian Follicle Growth ...
    May 13, 2022 · As previously discussed, progesterone inhibits ovarian follicle growth by slowing the rate at which granulosa cells undergo mitosis. Defining ...
  72. [72]
    Control of ovulation in mice by progesterone receptor-regulated ...
    Progesterone receptor (PGR), which mediates the biological effects of the steroid hormone progesterone, has emerged as a key regulator of ovulation in mice.
  73. [73]
    Control of oocyte release by progesterone receptor-regulated gene ...
    Dec 31, 2009 · PGR is a key specific regulator of ovulation across species. It is induced by the LH surge specifically in granulosa cells of mature preovulatory follicles.
  74. [74]
    The Effects of Progesterone on Immune Cellular Function at the ...
    These studies suggest that progesterone exhibits immunosuppressive functions not only through direct effects on decidual immune cells but also by the promotion ...
  75. [75]
    Progesterone promotes maternal–fetal tolerance by reducing human ...
    Progesterone is a steroid hormone essential for the maintenance of human pregnancy, and its actions are thought to include promoting maternal immune tolerance.
  76. [76]
    Inhibition of K+ Channels, Ca2+ Signaling, and Gene Expression in ...
    Abstract. The mechanism by which progesterone causes localized suppression of the immune response during pregnancy has remained elusive.
  77. [77]
    Progesterone Receptors: A Key for Neuroprotection in Experimental ...
    These results show that 1) PR are linked to signaling pathways that influence susceptibility to stroke, and 2) PR are direct key targets for both endogenous ...
  78. [78]
    Progesterone Receptor Agonist, Nestorone, Exerts Long-Term ...
    These results suggested that nestorone exerts long-term neuroprotective effects against permanent focal cerebral ischemia in adult and aged male rats. Nestorone ...
  79. [79]
    Progesterone and Neuroprotection - PMC - PubMed Central - NIH
    Progesterone has been shown to exert neuroprotective effects, both in cell models, animal models and in humans.
  80. [80]
    The Progestin Receptor Interactome in the Female Mouse ... - eNeuro
    Sep 11, 2017 · Progesterone binds to the progestin receptor (PR) isoforms, PR-A and PR-B, in the brain to profoundly influence female reproduction, brain ...
  81. [81]
    Potential contribution of progesterone receptors to the development ...
    Taken together, these results suggest a role for PR and thus perhaps progesterone in the development of lordosis behavior in female mice. By contrast, no ...
  82. [82]
    Progesterone, reproduction, and psychiatric illness - PMC
    Some found that cyclical increases in progesterone were associated with increased mood symptoms premenstrually, but only in women with a history of physical ...
  83. [83]
    Progestin Receptor-Mediated Reduction of Anxiety-Like Behavior in ...
    Here we present evidence suggesting that the anxiolytic-like effects of progesterone in male rats can be mediated, in part, by progestin receptors, as these ...Missing: aggression | Show results with:aggression
  84. [84]
    Progesterone receptors mediate male aggression toward infants
    We have found that male progesterone receptor knockout (PRKO) mice exhibit no infanticidal behavior and little aggression toward young.Missing: anxiolytic effects studies
  85. [85]
    Progesterone facilitates exploration, affective and social behaviors ...
    Sep 15, 2013 · Progesterone (P4) facilitates exploration, anxiety and social behaviors in estrogen (E2)-primed mice. Some of these effects may be due to ...
  86. [86]
    Non-genomic mechanisms of progesterone action in the brain - NIH
    Sep 19, 2013 · Here, we review various non-nuclear (ie, non-genomic) signaling mechanisms that progesterone can recruit to elicit its effects.
  87. [87]
    Progesterone Modulates Neuronal Excitability Bidirectionally - PMC
    Progesterone acts on neurons directly by activating its receptor and through metabolic conversion to neurosteroids.
  88. [88]
    The PROGINS polymorphism of the human progesterone receptor ...
    Our results indicate that the PROGINS variant of PR is less responsive to progestin compared with the most common PR because of (i) reduced amounts of gene ...
  89. [89]
    Progesterone receptor variants associated with the PROGINS ...
    Progesterone receptor variants associated with the PROGINS haplotype exhibit functional properties similar to those of wild-type progesterone receptor.
  90. [90]
  91. [91]
    Mice lacking progesterone receptor exhibit pleiotropic reproductive ...
    However, the adult female PR mutant displayed significant defects in all reproductive tissues. These included an inability to ovulate, uterine hyperplasia and ...
  92. [92]
    Consequences of Loss of Progesterone Receptor Expression in ...
    In endometrial cancer, loss of PR expression is associated with late-stage disease in which the tumor no longer responds to progesterone treatment. In this ...Abstract · INTRODUCTION · MATERIALS AND METHODS · RESULTS
  93. [93]
    Progesterone Receptor Gene Variants in Metastatic Estrogen ...
    This study expands the existing literature by identification of PGR variants in patients with ER+ metastatic breast cancer. In contrast to ESR1, the clinical ...
  94. [94]
    Changing Trends in Estrogen Receptors/Progesterone Receptors ...
    Apr 18, 2022 · ER/PR/HER2 were 77.0%/72.4%./23.8%, respectively. Triple-negative breast cancers were 10.1%. In comparison with previous results of 2006, the ...
  95. [95]
    Progesterone Receptor Expression Level Predicts Prognosis ... - NIH
    Jun 30, 2023 · Patients with PR-positive cancers also have a longer median OS than patients with PR-negative tumors in ER-positive tumors [7]. Previous studies ...
  96. [96]
    Clinical association of progesterone receptor isoform A with breast ...
    Jun 3, 2020 · PR-B directly supports L-BCa invasion and metastasis and also inhibits tumor growth, both only at high progesterone levels. As public datasets ...<|control11|><|separator|>
  97. [97]
    Progesterone receptor isoforms PRA and PRB differentially ...
    Mar 13, 2013 · Although most breast cancer metastases lack estrogen receptor (ER) and PR expression, PR has been reported to facilitate metastasis evolution ...
  98. [98]
    Hormone Therapy for Endometrial Cancer - American Cancer Society
    Feb 28, 2025 · The progestins used to manage endometrial cancer are: Medroxyprogesterone acetate (Provera®), which can be given as an injection or as a pill.
  99. [99]
    Redefining the Position of Hormonal Therapy in Endometrial Cancer ...
    Oct 3, 2023 · Mechanistically, progestin therapy inhibits estrogen-driven tumor growth by binding to progesterone receptor. The resultant progestin-PR complex ...
  100. [100]
    Progesterone receptor isoforms A and B: new insights into ... - ecancer
    In vitro studies using human endometrial cancer cells indicate that decreased PRB expression in endometrial cancer cells is likely responsible for progesterone ...
  101. [101]
    Advances in the molecular mechanisms underlying progestin ...
    Sep 24, 2023 · Typically, it is assumed that if PR expression is strong, progestin therapy will be effective. However, there are two anomalies observed in ...
  102. [102]
    PI3K inhibition results in enhanced estrogen receptor function and ...
    PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor-positive breast cancer.
  103. [103]
    the growing complexities of extra-nuclear progesterone signaling
    Jun 16, 2010 · In fact, non-genomic PR signaling appears to promote proliferation of breast cancer cell lines under some conditions [3], although the ...
  104. [104]
    Progestin nongenomic actions regulate cell proliferation and ...
    Apr 15, 2006 · Recently, we demonstrated that progesterone regulates proliferation and proteases activity via rapid activation of MAPKs and PI-3K/Akt pathways ...
  105. [105]
    Association of progesterone receptor status with breast cancer ...
    Oct 2, 2025 · This meta-analysis enrolled 35 studies on the impact of progesterone receptor (PR) status on breast cancer (BC) prognosis and illustrated that ...
  106. [106]
    The clinical value of progesterone receptor expression in luminal ...
    Jan 18, 2023 · Our results indicate that loss of PR predicts endocrine resistance, and this suggests that PR is a key marker of prediction of endocrine ...
  107. [107]
    Hormone therapy in postmenopausal women and risk of ...
    Hormone therapy for postmenopausal women with an intact uterus should comprise both estrogen and progestogen to reduce the risk of endometrial hyperplasia.
  108. [108]
    [PDF] Management of Endometrial Hyperplasia - RCOG
    Progestogens have been advocated to treat endometrial hyperplasia because they modify the proliferative effects of estrogen on the endometrium. Treatment with ...
  109. [109]
    Estrogen Replacement Therapy and Endometrial Hyperplasia
    In 94% (34/36) of postmenopausal women who developed endometrial hyperplasia in association with estrogen-only replacement, progestin therapy was effective.
  110. [110]
    Oral Contraceptive Pills - StatPearls - NCBI Bookshelf - NIH
    The most commonly prescribed pill is the COC pill. Progesterone is the hormone that prevents pregnancy, and the estrogen component controls menstrual bleeding.
  111. [111]
    Combined estrogen-progestin oral contraceptives: Patient selection ...
    Apr 22, 2024 · There may be a slightly increased risk with newer progestins (gestodene, desogestrel, and drospirenone) compared with levonorgestrel; however, ...Missing: termination | Show results with:termination
  112. [112]
    Selective progesterone receptor modulators and ... - Oxford Academic
    Mifepristone is used to terminate pregnancy, and as such is commercially available in many countries. The negative abortion-related image of mifepristone has ...
  113. [113]
    Promising selective progesterone receptor modulators: what's new ...
    Mifepristone combined with misoprostol is the most effective and popular form of medication abortion. Mifepristone and ulipristal acetate (UPA) demonstrate ...Missing: termination | Show results with:termination<|separator|>
  114. [114]
    Treatment of unresectable meningiomas with the antiprogesterone ...
    Long-term therapy with mifepristone is a new therapeutic option that may have efficacy in cases of unresectable benign meningioma.
  115. [115]
    SWOG S9005: Double blind phase III randomized trial of the ... - ASCO
    Background: Progesterone receptors are expressed on approximately 70 percent of meningiomas. Mifepristone is an oral anti-progestational agent that was ...
  116. [116]
    A Phase II Trial of Onapristone and Fulvestrant for Patients With ER+ ...
    The SMILE study is a multi-institutional phase II clinical trial to determine the efficacy and safety of an antiprogestin, onapristone, in combination with ...
  117. [117]
    OATH trial: A phase II clinical trial evaluating the combination of ...
    May 31, 2023 · The aim of OATH is to investigate the potential of dual hormonal blockade through the combination of antiprogesterone and anti-estrogen therapy.
  118. [118]
    9 biotech companies advancing treatment for breast cancer in 2024
    Nov 14, 2024 · The company's lead candidate, onapristone extended release (ONA-XR), is ... The company plans to initiate phase 3 clinical trials in 2025.
  119. [119]
    [PDF] Isoform-Specific Gene Regulation by Progesterone Receptors ...
    Jun 4, 2025 · modulators or chromatin-targeting therapies represents an exciting avenue for improving treatment efficacy in PR+ breast cancer. METHODS.Missing: emerging | Show results with:emerging
  120. [120]
    Progesterone receptor isoforms, agonists and antagonists ...
    Jan 12, 2018 · Priority Research Papers: Progesterone receptor isoforms, agonists and antagonists differentially reprogram estrogen signaling. PDF | HTML | ...
  121. [121]
    Progesterone receptor isoform modulation via enhancer activation ...
    Aug 20, 2025 · Progesterone receptor isoform modulation via enhancer activation regulates progesterone signaling in endometrial stromal cells. Skylar G.
  122. [122]
    Abstract 6577: Chemoproteomic evaluation of PROTACs targeting ...
    Mar 22, 2024 · Global proteomic analysis unveiled off-target protein degradation of progesterone receptor (PR) and phosphodiesterase 6D (PDE6D) by ER targeted PROTACs.