Skip to main content
Molecular Human Reproduction logoLink to Molecular Human Reproduction
. 2023 Jun 8;29(8):gaad022. doi: 10.1093/molehr/gaad022

Progesterone signaling in the regulation of luteal steroidogenesis

Kayhan Yakin 1,2,, Francesko Hela 3,4, Ozgur Oktem 5,6
PMCID: PMC10631818  PMID: 37289566

Abstract

The corpus luteum is the major source of progesterone, the essential hormone for female reproductive function. While progesterone activity has been the subject of extensive research for decades, characterization of non-canonical progesterone receptor/signaling pathways provided a new perspective for understanding the complex signal transduction mechanisms exploited by the progesterone hormone. Deciphering these mechanisms has significant implications in the management of luteal phase disorders and early pregnancy complications. The purpose of this review is to highlight the complex mechanisms through which progesterone-induced signaling mediates luteal granulosa cell activity in the corpus luteum. Here, we review the literature and discuss the up-to-date evidence on how paracrine and autocrine effects of progesterone regulate luteal steroidogenic activity. We also review the limitations of the published data and highlight future research priorities.

Keywords: progesterone, corpus luteum, pregnancy, luteotrophin, cholesterol, steroidogenesis, granulosa cell, apoptosis, angiogenesis

Introduction

This review article summarizes the current state of knowledge regarding the different facets of progesterone signaling in corpus luteum activity, covering its hormonal, paracrine, and autocrine actions. The notion that progesterone modulates its own secretion dates back to observations in the 1980s (Rothchild, 1981). Since then, progesterone activity in different tissues has become a subject of extensive research. Elegant studies have unraveled many of the complex signal transduction mechanisms exploited by the progesterone hormone. Collectively, these data indicate that the biological functions of progesterone are more versatile than previously envisioned. Both in vivo and in vitro studies in luteal cells as well as in different tissues are strongly supportive of the regulatory effect of progesterone.

Progesterone hormone is essential for female reproductive function. Deciphering its mechanism of action in target tissues can provide us with invaluable information to understand the pathophysiology of menstrual disorders, implantation failure, and early pregnancy complications.

Methods

Our goal was to review the published data on progesterone activity in steroidogenic tissues. A comprehensive search for peer-reviewed articles in English was implemented based on articles available in Scopus, Web of Science, and PubMed databases, with no limits placed on time. The main terms ‘progesterone’, ‘progestin’, ‘granulosa cell’, ‘luteal cell’, ‘ovary’, ‘luteotrophin’, ‘autocrine’, and ‘paracrine’ were searched using Boolean operators ‘AND’, ‘OR’, and ‘NOT’ in various combinations. Experimental studies on human and animal tissues/cells, reviews, editorials, and commentaries were included. Duplicates or unrelated articles were excluded. Additional studies addressed by primary references and results from our own studies were included where relevant. The search strategy and selection steps are described in Fig. 1.

Figure 1.

Figure 1.

Schematic representation of literature search strategy and selection of relevant publications.

Progesterone-induced signaling

Progesterone is synthesized from the parent molecule cholesterol through a series of complex chemical reactions catalyzed by steroid cytochrome P450 and hydroxysteroid dehydrogenases (Miller and Auchus, 2011). The source of cholesterol for steroidogenesis in ovarian luteal cells depends on circulating plasma lipoproteins, de novo synthesis, and utilization of intracellular cholesterol ester stores. Steroidogenic acute regulatory protein (StAR) facilitates the transport of cholesterol from the outer to the inner mitochondrial membrane, serving as the rate-limiting step in progesterone synthesis. P450 cholesterol side-chain cleavage enzyme (CYP11A) catalyzes the conversion of cholesterol to pregnenolone, which passes into the smooth endoplasmic reticulum where it is converted to progesterone by 3β-hydroxysteroid isomerase (3β-HSD). Progesterone then diffuses out of the luteal cell to be transported to the target tissues.

Progesterone-induced gene transcription is mediated through different receptor/signal transduction pathways including classical (nuclear or canonical) receptors and nonclassical (membrane and membrane-associated or non-canonical) receptors (Fig. 2). Progesterone shows tissue-specific, context-specific, and menstrual phase-dependent activity. This context-dependent activity is achieved through the expression pattern and relative concentration of different types of progesterone receptors (PRs), interactions with co-regulatory factors (co-activators and co-inhibitors), cooperating with other transcriptional factors, and chromatin-dependent access to target genes. Moreover, while ligand binding and post-receptor signal transduction cascades vary and are complex, they seem to interact at several points but also have independent activities that lead to cell- and context-specific responses to progesterone (Peluso, 2006; Sueldo et al., 2015).

Figure 2.

Figure 2.

Schematic summary of progesterone-induced receptor-signal transduction systems. Canonical (or classical) progesterone receptors include progesterone receptor A (PR-A), B (PR-B), and C (PR-C). Non-canonical (nonclassical) progesterone signaling functions through both progestin and adiponectin Q receptors (MPRα, MPRβ, MPRγ, MPRð, and MPRɛ) and/or membrane-associated progesterone receptors such as the progesterone receptor membrane component 1 (PGRMC1) and 2 (PGRMC2). While ligand binding and downstream signal transduction cascades of canonical and non-canonical receptors differ in a tissue- and context-dependent manner, they seem to interact through secondary messengers (image created with Biorender.com). 20α-HSD: 20α-hydroxysteroid dehydrogenase; Bcl-2: B-cell lymphoma protein 2; Bad: Bcl-2-associated death promoter; INSIG1: insulin-induced gene 1; LCN-2: Lipocalin 2; LDL: low-density lipoprotein; MPR: membrane progesterone receptor; P450SCC: P450 cholesterol side-chain cleavage enzyme; PGRMC: progesterone receptor membrane component; PR: progesterone receptor; SCAP: sterol regulatory element-binding protein cleavage-activating protein; SRE: sterol regulatory element; SREBP: sterol regulatory element-binding protein; StAR: steroidogenic acute regulatory protein; VEGF: vascular endothelial growth factor.

Nuclear (classical) PRs

The PRs belong to the nuclear/steroid hormone receptor family of ligand-dependent transcription factors. Following engagement with its ligand, a PR regulates the activity of a multitude of downstream effectors and the expression of target genes. Signaling through PRs induces a steadily growing and sustained response (Medina-Laver et al., 2021).

Three major receptor isoforms, namely PR-A, PR-B, and PR-C, are encoded by the same gene but they are transcribed from different promoters (Misrahi et al., 1993; Grimm et al., 2016). An additional B upstream segment in the amino-terminal binding domain in PR-B includes an extra transactivation function domain, endowing PR-B a greater transcriptional activity than PR-A. The common N-terminal domain possesses a proline-rich sequence including a binding site for the Src homology 3 (SH3) domain of the Src tyrosine kinase. The third form, PR-C (45–50 kDa), which has been shown to be expressed in the pregnant uterus, can bind to the ligand but, owing to the lack of a DNA-binding domain, cannot bind to the progesterone response elements (PREs) of the target genes (Wei et al., 1996).

The classical nuclear PRs are localized in the cytoplasm and the nucleus. In the absence of ligand, PR resides in complexes with chaperone molecules including heat shock proteins (HSP70 and HSP90). Ligand binding triggers a conformational change in the PR that leads to its dissociation from the HSP complex. Dimerized PR is rapidly transported to the nucleus and binds the chromatin-embedded genome where it acts as a transcription factor to regulate PREs of the responsive genes. Recruitment of several co-activators or co-inhibitors, including activator protein 1 (AP1), specificity protein1 (Sp1), and signal transducer and activator of transcription proteins (STATs), regulate the subsequent progesterone-induced target gene expression (Owen et al., 1998). Moreover, progesterone modulates gene promoters that lack complete canonical PRE-binding sequences in association with other DNA-bound transcription factors (Faivre et al., 2008) and cross-talk with the estrogen receptors (ER) (Migliaccio et al., 1998). PR–chromatin interactions demonstrate remarkable tissue-specificity in terms of binding sites and interactions with other transcription factors (Dinh et al., 2019).

In conjunction with their transcriptional activity, PRs also utilize the growth factor receptor signaling pathways. Ligand binding facilitates the association between proline-rich regions of the N-terminal domain and Src tyrosine kinases to activate the Src/Ras/Raf/Mek/Erk-1 cascade (Migliaccio et al., 1998; Garg et al., 2017). PR activity is controlled through phosphorylation by several kinases such as the cyclin-dependent kinase 1/2 (CDK1/2), mitogen-activated protein kinase (MAPK), protein kinase A (PKA), DNA-dependent protein kinase (DNA-PK), and casein kinase II (CK2) (Grimm et al., 2016).

Progestin and adiponectin Q receptors

Membrane progesterone receptors (MPRs) are seven-transmembrane G protein-coupled receptors with their N-termini facing toward the extracellular surface (Zhu et al., 2003). They belong to the family of progestin and adiponectin Q receptors (PAQR). The expression of five isoforms (MPRα, β, γ, ð, and ɛ or PAQR7, PAQR8, PAQR5, PAQR6, and PAQR9) is cell-specific and hormone-dependent (Medina-Laver et al., 2021).

MPRs bind progesterone with high affinity (Kd of 20–40 nM) but do not bind to synthetic progesterone agonists or antagonists such as mifepristone. Ligand activation increases tyrosine kinase and MAPK activities and decreases adenylate cyclase activity. Moreover, MPRs participate in other signal transduction pathways including extracellular signal-regulated kinases 1/2 (Erk1/2) or p38 MAPKs. (Charles et al., 2010).

Membrane-associated PRs

The membrane-associated PR family comprises four homologous proteins: progesterone receptor membrane component 1 (PGRMC1), progesterone receptor membrane component 2 (PGRMC2), neudesin, and neuferricin (Kimura et al. 2012). These proteins possess a similar non-covalent heme/steroid-binding domain, which is related to the cytochrome b5, a crucial component of microsomal cytochrome P450 monooxygenase systems.

The PGRMC1 protein comprises a single transmembrane domain, a short extracellular domain, and a cytoplasmic tail (Gerdes et al.1998; Cahill, 2007; Peluso, 2007). It binds and interacts with plasminogen activator inhibitor RNA binding protein-1 (PAIRBP-1) (Peluso et al., 2005, 2006). PGRMC2 harbors a structurally similar cytochrome b5 domain to PGRMC1 but a different N-terminal transmembrane domain. Both PGRMC1 and PGRMC2 possess several Src homology target sequences for molecular interactions with other intracellular proteins that contain SH2 and SH3 domains (Cahill, 2007; Cahill et al., 2016).

The two other members of this receptor family, neudesin and neuferricin, are involved in the regulation of neural activity but no functions related with progesterone have been reported to date (Ryu et al., 2017).

Unlike nuclear PRs, which mainly modify transcriptional activity to build a gradually emerging cellular response, non-canonical signaling induces an array of secondary messengers and signal transduction pathways that lead to an immediate response upon ligand binding (Medina-Laver et al., 2021).

Unlike PR-A and PR-A, PGRMC1 and MPRs do not bind to RU 486 (Peluso, 2006). Upon ligand binding, PGRMC1 interacts with PGRMC2 and functions as a membrane receptor complex (Peluso, 2006). As PGRMC1 interacts with both steroidogenic and drug-metabolizing cytochrome P450 enzymes, it is localized in various locations that correspond to its proposed function including the mitochondria, smooth endoplasmic reticulum, plasma membrane, cytoplasm, and nucleus (Piel et al., 2016). PGRMC2 mainly localizes to the cytoplasm (Peluso et al., 2019). Recent studies on breast tissue reveal a crosstalk between PGRMC1 and ERα. Silencing PGRMC1 attenuated ERα expression in breast cancer cells (Pedroza et al., 2021). Progestins, including norethisterone and dydrogesterone, facilitated the interaction between PGRMC1 and prohibitins which, in turn, decreased binding of prohibitins to ERα and increased ERα activation (Bai et al., 2021).

The characteristics of receptors involved in progesterone-induced signaling in ovarian cells are summarized in Table 1. The expression profile of these receptors varies among species. While PRA and PRB expression is not detectable in rat corpus luteum, both isoforms appear in the granulosa cells of preovulatory follicles in humans and Rhesus monkey and continue throughout the luteal phase of the cycle, in correlation with the serum progesterone level. In contrast, in the Rhesus monkey, PRA protein levels gradually decline during the luteal phase while PRB levels remain relatively constant (Misao et al., 1998; Ottander et al., 2000; Peluso et al., 2006).

Table 1.

The characteristics of receptors involved in progesterone-induced signaling in ovarian cells and their expression profile through the menstrual cycle.

Receptor family Isoform Gene locus (chromosome) Molecular weight (kDa) Progesterone binding constant (nM) Expression in granulosa cells
Follicular Periovulatory follicular Luteal
Nuclear progesterone receptors A 11 94 1–5 +*
B 11 114 1–5
Progestin and adiponectin Q receptors MPRα 1 40 20–40 ND +** +
MPRβ 6 40 20–40
MPRγ 15 40 20–40
Membrane -associated progesterone receptors PGRMC1 X 22 35 + + +
PGRMC2 4 24

MPR: membrane progesterone receptor, PGRMC: progesterone receptor membrane component,.

*

Species-specific, not detected in rats.

**

Detected in Medaka fish: all other data are from human. ND: not determined.

PGRMC2 and PAIRBP1 are expressed in granulosa cells throughout the menstrual cycle in humans (Sueldo et al., 2015) and their expression level was reported to be increased in the luteal phase in a rat model (Peluso et al., 2006; Griffin et al., 2014). While MPRα, MPRβ, and MPRγ expression was shown in rat corpus luteum and granulosa cells of Medaka fish preovulatory follicles (Hagiwara et al., 2016), human data are lacking (Cai and Stocco, 2005).

Human luteal granulosa cells express PR-A, PR-B, PGRMC1, PGRMC2, and MPRα, β, γ, and ð at different levels (Peluso et al., 2009; Pietrowski et al., 2014; Sueldo et al., 2015). While significant patient variation has been detected in their relative expression, PGRMC1 is the most abundantly expressed receptor, followed by MPRα, PGRMC2, and PR, when compared to actin expression (Sueldo et al. 2015).

Progesterone-modulated mechanisms in luteal granulosa cell activity

There is considerable evidence showing that progesterone supports luteal granulosa cell survival and regulates its own synthesis through different mechanisms (Fig. 3). Table 2 provides a summary of the key experimental studies related to the proposed mechanisms of progesterone’s luteotrophic activity.

Figure 3.

Figure 3.

Progesterone modulates luteal activity on granulosa cells through different mechanisms. The different mechanisms are: (1) suppression of apoptosis and supporting the survival of granulosa cells, (2) supporting its own synthesis through induction of steroidogenic enzymes, (3) regulating de novo cholesterol synthesis through interaction of PGRMC1 with INSIG1 and SCAP, (4*) supporting luteal vascularization through angiogenic modulators such as VEGF and lipocalin, (5*) promoting lipoprotein uptake, (6) suppressing its own catabolism through inhibition of 20α-hydroxysteroid dehydrogenase, and (7) increasing its own receptor expression. * denotes functions yet to be demonstrated in ovarian granulosa cells (image created with Biorender.com). AC: adenylyl cyclase; AKT: Ak strain transforming protein; cAMP: cyclic adenosine monophosphate; ER: endoplasmic reticulum; ERK: extracellular signal-regulated kinases; HSP: heat shock protein; INSIG1: insulin-induced gene 1; MAPK: mitogen-activated protein kinase; MPR: membrane progesterone receptor; NFκB: nuclear factor kappa-light-chain-enhancer of activated B cell; PGRMC1: progesterone receptor membrane component 1; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; PR: progesterone receptor; PRE: progesterone response elements; SCAP: sterol regulatory element-binding protein cleavage-activating protein; VEGF: vascular endothelial growth factor

Table 2.

Summary of the key experimental studies related to proposed mechanisms involved in the luteotrophic activity of progesterone.

Reference Species Cell/tissue Study outcome
Progesterone inhibits apoptosis and supports survival of granulosa/luteal cells Makrigiannakis et al. (2000) Human Granulosa cells Progesterone reduces and RU 486 increases the rate of apoptosis. RU 486 inhibits the protective effect of progesterone on survival.
Chaffin et al. (2001) Macaque Granulosa cells Suppression of progesterone levels through inhibition of 3βHSD by trilostane increases apoptosis which can be reversed by progesterone supplementation
Mussche and D’Herde (2001) Quail Granulosa cells Blocking progesterone synthesis by aminoglutethimide or progesterone receptors by RU 486 causes an increase in the rate of apoptosis
Shao et al. (2003) Mouse Granulosa cells RU 486 treatment augments caspase-3 activation
Rung et al. (2005) Rat and human Granulosa cells Inhibition of progesterone signaling and/or synthesis increases apoptosis, which is reversed by progesterone administration
Peluso et al. (2005) Rat Luteal/granulosa cells PAIRBP1 mediates progesterone’s antiapoptotic activity and this activity might involve interaction with PGRMC1
Liszewska et al. (2005) Cow Luteal cells Progesterone treatment increases the expression of the anti-apoptotic factor Bcl-2
Peluso et al. (2006) Rat Luteal/granulosa cells PGRMC1 interacts with PAIRBP1 to form a complex that is required to exert its anti-apoptotic action
Engmann et al. (2006) Human Luteal/granulosa cells Progesterone suppresses the rate of apoptosis
Peluso et al. (2007) Rat Granulosa cells Progesterone promotes survival through regulation of PKG and Mek/Erk activities, as induction of PKG activity by 8-br-cGMP supports and inhibition of PKG by delta-tocotrienol suppresses progesterone’s anti-apoptotic effects
Peluso et al. (2008) Rat Granulosa cells siRNA-induced knockdown of PGRMC1 prevents progesterone from inhibiting apoptosis
Peluso et al. (2009) Human Granulosa cells Progesterone activates a PGRMC1-dependent mechanism to promote survival
Peluso et al. (2010) Rat Granulosa cells Progesterone supports cell survival by increasing the Bcl21d to Bad ratio
Peluso et al. (2012) Human Granulosa cells Progesterone downregulates caspase-3 expression
Quirk et al. (2013) Cow Luteal cells Progesterone treatment decreases Fas-ligand induced apoptosis
Pietrowski et al. (2014) Human Luteal cells Progesterone treatment downregulates Fas expression
Griffin et al. (2014) Rat Granulosa cells Progesterone slows down the rate of apoptosis through PGRMC2
Peluso et al. (2014) Rat Granulosa cells siRNA-induced depletion of Pgrmc1 or antibody-induced disruption of PGRMC1/2 complex increases the rate of apoptosis
Progesterone augments its own synthesis Uchida et al. (1970) Rat Granulosa cells Exogenous progesterone augmented progesterone secretion
Fortune and Vincent (1983) Mice Granulosa cells Progesterone regulates in vitro steroidogenic activity
Fanjul et al. (1983) Rat Granulosa cells Synthetic progestin R5020 increases gonadotropin-induced progesterone synthesis
Ruiz de Galarreta et al. (1985) Rat Granulosa cells Natural and synthetic progestins augment pregnenolone and progesterone production via increasing 3β-HSD activity
Hibbert et al. (1996) Monkey Luteal cells Inhibition of 3β-HSD induces structural changes similar to luteal regression, which is reversed by addition of the synthetic progesterone R5020
VandeVoort et al. (2000) Human Luteal/granulosa cells Progesterone antagonists RU 486 and HRP 2000 suppress ovarian steroid synthesis
Ottander et al. (2000) Human Luteal/granulosa cells Progesterone exerts a stimulatory action on steroid synthesis through its receptors
Yang et al. (2002) Human Kidney cells PGRMC1/SCAP/INSIG1 complex activates transcriptional activity of SREBP and induces the expression of steroidogenic genes, including StAR
Suchanek et al. (2005) Monkey Kidney cells PGRMC1 interacts with SCAP and INSIG1
Rekawiecki et al. (2005) Cow Luteal/granulosa cells Progesterone increases mRNA expression of StAR, 3β-HSD, and CYP19A1
Peluso et al. (2009) Human Granulosa cells Synthetic progesterone R5020 stimulates progesterone secretion
Yao et al. (2020) Sheep Luteal/granulosa cells Progesterone increases mRNA expression of StAR, 3β-HSD, and CYP19A1
Progesterone modulates de novo cholesterol synthesis Rung et al. (2005) Rat and human Granulosa cells Progesterone receptor antagonists Org 31710 and RU 486 suppress de novo cholesterol synthesis which is reversed by addition of progesterone
Hughes et al. (2007) Human Kidney cells PGRMC1 binds to INSIG1 and modulates CYP51A1 family members that control cholesterol synthesis
Progesterone promotes luteal vascularization Shimizu and Miyamoto (2007) Cow Granulosa and theca cells Progesterone augments the expression of VegfA120
Peluso et al. (2018) Mice Luteal/granulosa cells Vascularization of corpus luteum is attenuated in PGRMC1/2 deleted mice
Peluso et al. (2018) Mice Luteal/granulosa cells Inhibition of PGRMC1/2 activity is associated with reduction in the number of vascular endothelial cells
Nichols et al. (2019) Cattle Granulosa and theca cells Progesterone augments the expression of Vegf-A
Narimani et al. (2020) Mice Granulosa and theca cells Progesterone augments the expression of Vegf and angiogenic parameters including vascular density and area
Progesterone regulates lipoprotein uptake Furuhata et al. (2020) Mice Adipocytes PGRMC1 interacts with VLDL-R and LDL-R or GLUT4 to facilitate the uptake of lipoproteins
Oh et al. (2013) Mice Not specified Progesterone upregulates low-density lipoprotein receptor-related protein 2
Riad et al. (2018) Human HeLa cells PGRMC1 forms a complex with TMEM97 and LDL-R to assist the uptake of LDL and apolipoprotein E
Progesterone suppresses its own catabolism Sugino et al. (1997) Rodent Luteal cells Progesterone suppresses its own catabolism by downregulating the expression of 20α-HSD
Fortune and Vincent (1983) Rodent Granulosa cells Progesterone suppresses the conversion of progesterone to estrogen by inhibiting aromatase activity
Beranic et al. (2011) Human Endometrium Progesterone inhibits aldo-ketoreductases 1C1 and 1C3 which convert progesterone to the less potent metabolite 20α-hydroxyprogesterone
Progesterone modulates the expression of progesterone receptors Misao et al. (1998) Human Luteal cells High concentration of progesterone induces PR-A expression, which suppresses PR-B expression and subsequently decreases progesterone activity
Chaffin et al. (1999) Macaque Granulosa cells Steroid depletion reduces progesterone receptor expression in periovulatory granulosa cells which is reversed by progesterone replacement
Guerra-Araiza et al. (2002) Rat Brain tissues Progesterone shows a gender-dependent and tissue-specific effect on the expression of PR isoforms
Gordon et al. (2015) Rat Brain tissues Progesterone shows a time- and dose-dependent effect on the expression of PR isoforms
Diep et al. (2016) Human Breast, endometrial, and ovarian cancer cell lines High doses of synthetic progestin R5020 upregulate PR expression which is reversed by the addition of an antiprogestin RU 486

3β-HSD: 3β-hydroxysteroid isomerase; 8-br-cGMP: 8-bromo-cyclic GMP; 20α-HSD: 20α-hydroxysteroid dehydrogenase; CYP19A1: cytochrome P450 family 19 subfamily A member 1; CYP51A1: cytochrome P450 family 51 subfamily A member 1; GLUT4: glucose transporter type 4; INSIG1: insulin-induced gene 1; LDL: low-density lipoprotein receptor; PAIRBP-1: plasminogen activator inhibitor RNA binding protein -1; PGRMC1: progesterone receptor membrane component 1; PGRMC2: progesterone receptor membrane component 2; PKG: cyclic GMP-dependent kinase; PR-A: progesterone receptor A, PR-B: progesterone receptor-B; SCAP: sterol regulatory element binding protein cleavage- activating protein; SERBP-1: serpine 1 mRNA binding protein; SREBP: sterol regulatory element binding protein; StAR: steroidogenic acute regulatory protein; TMEM97: sigma-2 receptor/transmembrane protein 97; VEGF-A: vascular endothelial growth factor-A; VLDL: very low-density lipoprotein receptor.

Progesterone suppresses apoptosis and supports the survival of granulosa cells

Several studies indicate that progesterone promotes luteal activity by supporting the survival of granulosa/luteal cells and by inhibiting their apoptosis. In vitro studies in animal and human periovulatory granulosa cells show that inhibition of progesterone signaling and/or synthesis increases apoptosis, which can be reversed by administration of progesterone (Makrigiannakis et al., 2000; Mussche and D’Herde, 2001; Rung et al., 2005). Progesterone’s anti-apoptotic activity might occur through different mechanisms. For example, progesterone treatment decreases Fas ligand-induced apoptosis in cow corpus luteum (Quirk et al., 2013) and in luteal granulosa cells recovered from infertile women (Pietrowski et al., 2014). In a human granulosa cell line (hGL5 cells), progesterone suppresses the expression of the apoptosis activator caspase-3 (Peluso et al., 2012) while in mouse granulosa cells, RU-486 treatment was associated with an increase in caspase-3 activation (Shao et al., 2003). Progesterone treatment increased the expression of anti-apoptotic B-cell-lymphoma-2 protein (Bcl-2) in bovine luteal cells (Liszewska et al., 2005) and favored cell survival in spontaneously immortalized granulosa cells (SIGC) by increasing the Bcl21d to Bcl-2 associated death promoter ratio (Peluso et al., 2010). In freshly recovered human luteal granulosa cells and spontaneously immortalized rat granulosa cells, progesterone promotes survival through regulation of cyclic GMP-dependent kinase (PKG) and Mek/Erk activities, as induction of PKG activity by 8-br-cGMP supports, and inhibition of PKG by delta-tocotrienol suppresses, progesterone’s anti-apoptotic effects (Peluso et al., 2006, 2007).

The mechanism of how progesterone attenuates apoptosis and promotes cell survival, even in cells that do not express classical PRs, is explained by the outstanding mechanistic analyses reported by Peluso’s team, who demonstrated that PGRMC1 is responsible for the pro-survival effects of progesterone in human and rat granulosa/luteal cells (Peluso et al., 2005, 2008, 2009, 2012, 2014; Griffin et al., 2014). While progesterone favored cell survival in SIGC (Peluso et al., 2010), siRNA-induced depletion of Pgrmc1 or antibody-induced disruption of the PGRMC1/2 complex was associated with an inappropriate entry into the cell cycle, which resulted in an increased rate of apoptosis (Peluso et al., 2014).

Progesterone increases its own synthesis by StAR-induced steroidogenesis

Research on the characterization of progesterone’s role in the formation and maintenance of the corpus luteum dates back to the early 1970s. Administration of exogenous progesterone augmented progesterone secretion from rat ovaries (Uchida et al., 1970). In addition, progesterone was shown to regulate in vitro steroidogenic activity of the granulosa cells of mice (Fortune and Vincent, 1983). The synthetic progestin R5020 increased the gonadotrophin-induced progesterone synthesis in cultured rat granulosa cells (Fanjul et al., 1983). Likewise, in the same setting, both natural and synthetic progestins augmented pregnenolone and progesterone production via increasing 3β-HSD activity (Ruiz de Galarreta et al., 1985). Inhibiting progesterone synthesis by the 3βHSD inhibitor trilostane induced structural changes similar to those seen in luteal regression in Rhesus monkeys, which was reversed by the addition of R5020 (Hibbert et al., 1996). Based on these data in 1996, Rothchild proposed that intraovarian progesterone might function as ‘the universal luteotropin’, to promote its own secretion (Rothchild, 1996). Subsequent studies provided further molecular evidence to explain Rothchild’s proposal for the autocrine effects of progesterone to stimulate its own synthesis by the corpus luteum (VandeVoort et al., 2000; Ottander et al., 2000; Stouffer, 2003; Rung et al., 2005; Peluso et al., 2009). Progesterone increased the mRNA expression of StAR, 3β-HSD, and CYP19A1 in cow (Rekawiecki et al., 2005) and sheep (Yao et al., 2020) luteal/granulosa cells.

The identification of non-canonical progesterone signaling in granulosa cells helped to explain the molecular mechanism of progesterone enhancing its own synthesis in luteal cells, which are known to be devoid of classical PRs (Peluso and Pru, 2014). Elegant studies demonstrated that PGRMC1 is likely to be involved in progesterone-induced augmentation of steroid synthesis (Peluso and Pru, 2014). PGRMC1 interacts with two critical proteins that regulate steroidogenesis; sterol regulatory element binding protein cleavage-activating protein (SCAP) and insulin-induced gene 1 (INSIG1) (Suchanek et al., 2005). This complex activates transcriptional activity of sterol regulatory element binding protein (SREBP) to induce the expression of various genes involved in steroidogenesis, including the key enzyme StAR (Christenson and Strauss, 2001; Yang et al., 2002).

Interestingly, the capacity of luteal granulosa cells to secrete progesterone was not altered when any of the classical and nonclassical PRs were selectively depleted through siRNA treatment (Sueldo et al., 2015). The most plausible explanation is that, despite sharing one or more common signaling pathways, each of these modulators regulates separate and independent transduction pathways to maintain progesterone secretion.

Another interesting topic that warrants further research is the association of PRs with autophagy modulators. In cancer cell lines, PGRMC1/2 was shown to bind microtubule-associated protein 1A/1B light chain 3B, a critical protein that is necessary for the formation of autophagosomes (Mir et al., 2013). Autophagy is known to have an important role in the steroidogenic activity of steroid-producing cells (Tang et al., 2019). Further research is necessary to dissect the role of PGRMC in the autophagic machinery of luteal granulosa cells.

Progesterone signaling is associated with de novo cholesterol synthesis in granulosa cells

Several lines of evidence suggest that progesterone is involved in cholesterol sensing and synthesis mechanisms in the ovarian granulosa cells. For instance, the PR antagonists Org 31710 and RU 486 suppressed de novo cholesterol synthesis in rat and human granulosa cells (Rung et al., 2005) and this could be reversed by the addition of progesterone. As the genes that regulate cholesterol synthesis do not contain functional PREs, these findings were proposed to be related to SREBP-2. The most convincing evidence regarding progesterone’s role in cholesterol metabolism is derived from a study that showed that PGRMC1 binds to INSIG1 and modulates CYP51A1 family members that control cholesterol synthesis (Hughes et al., 2007). Based on the interaction of PGRMC1 with INSIG1 and SCAP, as well as circumstantial evidence from a non-peer-reviewed study, Cahill and Medlock suggested that PGRMC1 might be involved in cholesterol sensing and trafficking through different compartments of the cell by acting as a cholesterol-chaperone (Warnick, 2008; Cahill and Medlock, 2017). PGRMC1 appears to function as a versatile protein in cholesterol trafficking and steroid synthesis, and many of its functions remain to be studied in detail.

Progesterone promotes luteal vascularization

In the midluteal phase of the menstrual cycle, the corpus luteum has the capacity to augment progesterone synthesis by several orders of magnitude (Taraborrelli, 2015), maintaining a progesterone output of around 40 mg per day (MacDonald et al., 1991). This requires a significant increase in the availability of cholesterol for steroid synthesis. An increase in luteal vascularization would facilitate lipid uptake from the circulation and support the increased demand. Progesterone seems to support its own synthesis by inducing luteal vascularization (Curry and Nothnick, 1996; Pall et al., 2000; Stouffer, 2003; Peluso et al., 2018). Progesterone is known to increase the expression of the vascular endothelial growth factor (VEGF) gene in ovarian tissue in mice (Narimani et al., 2020), granulosa and theca cells in ewes (Narimani et al., 2020), cows (Shimizu and Miyamoto, 2007), and cattle (Nichols et al., 2019). Inhibition of PGRMC1/2 activity is associated with a reduction in the number of vascular endothelial cells within the mouse corpus luteum (Peluso et al., 2018), This effect seems to be independent of VEGF-A but rather related to another novel angiogenesis regulator, lipocalin 2, as its expression in corpus luteum is dependent on the presence of PGRMC1/2. In different target tissues other than the ovaries, progesterone exploits multiple signaling pathways to regulate neovascularization, including basic fibroblast growth factor, angiopoietin 1 and 2, hypoxia inducible factor α, and nitric oxide (Xia et al., 2021). However, involvement of these factors in the augmented vascularization of the corpus luteum has yet to be determined.

Progesterone regulates lipoprotein uptake

No less important than its role in amplified steroidogenic enzyme activity, progesterone may also increase the availability of lipoprotein-derived cholesterol substrate for steroid synthesis. In adipocytes PGRMC1 interacts with low-density lipoprotein receptors (VLDL-R and LDL-R) or glucose transport protein 4 (GLUT4) to facilitate the uptake of lipoproteins (Furuhata et al., 2020). In murine uterus, high-density DNA microarray analysis indicated that progesterone upregulates LDL receptor-related protein 2 (Lrp2) (Oh et al., 2013). CRISPR-induced gene knockout studies in HeLa cells revealed that PGRMC1 forms a complex with sigma-2 receptor/transmembrane protein 97 (TMEM97) and LDL-R to assist the uptake of LDL and apolipoprotein E (Riad et al., 2018). The intriguing question of whether progesterone promotes lipoprotein uptake in luteal granulosa cells has not yet been studied.

Progesterone suppresses its own catabolism

Progesterone seems to regulate its own catabolism through different mechanisms. The human aldo-ketoreductase (AKR) subfamily member 20α-hydroxysteroid dehydrogenase (20α-HSD) reduces progesterone to its inactive metabolites, 20α-hydroxyprogesterone, 20α-hydroxy-4-pregnen-3-one, and other weak progesterones (Penning et al., 2004; Chu et al., 2022). In rodent luteal cells, progesterone was shown to suppress its own catabolism by downregulating the expression of 20α-HSD enzyme (Sugino et al., 1997). In rodent granulosa cells, progesterone suppresses the conversion of progesterone to estrogen by inhibiting aromatase activity (Fortune and Vincent, 1983). In human endometrium, progesterone and its derivatives (dydrogesterone, 20α-hydroxydydrogesterone, medroxyprogesterone acetate, desogestrel, norethindrone, and levonorgestrel) were shown to inhibit AKR1C1 and AKR1C3 (Beranic et al., 2011).

Progesterone modulates the expression of PRs

The current understanding is that progesterone downregulates the expression of PRs (Park and Mayo, 1991). However, Stouffer challenged this concept, at least for the granulosa cell, suggesting that recovery of PR expression following progesterone treatment in macaque granulosa cells after blockage with trilostane plus hCG shows that progesterone may upregulate PR expression, at least in early luteinization (Chaffin et al., 1999; Stouffer, 2003). The idea is supported by another study, which showed that a high concentration of progesterone in luteal cells induced PR-A expression, which suppresses PR-B expression and subsequently decreased progesterone activity (Misao et al., 1998). In the brain tissues of gonadectomized rats, progesterone showed a gender-dependent and tissue-specific (Guerra-Araiza et al., 2002) as well as time- and dose-dependent (Gordon et al., 2015) effect on the expression of PR isoforms. In PR+ERα+ breast, endometrial, and ovarian cancer cell lines, high doses of the progestin R5020 upregulated PR expression at the transcriptional and translational levels and the effect was reversed by the addition of the antagonist RU 486 (Diep et al., 2016). These data indicate that the impact of progesterone on PR gene expression is more complex than has been reported. It is of note that the available data are concentrated on the expression of classical (nuclear) PRs. The effect of progesterone on the expression of membrane and membrane-associated receptors in the luteal granulosa cells warrants further research.

Conclusions

A functional corpus luteum is essential for endometrial preparation, implantation, and the maintenance of early gestation. Despite extensive research, many questions regarding the intricate mechanism(s) that control luteinization, luteolysis, and the luteo-placental shift remain to be answered. Studies exploiting the advantages of progesterone antagonists and gene knockout/knockdown technologies and characterization of non-canonical receptor/signal transduction pathways have helped the research on progesterone gain momentum in the last two decades. Readers are referred to the excellent reviews on classical and nonclassical progesterone signal transduction, chromatin interactions, and regulation of transcription for further details (Peluso and Pru, 2014; Grimm et al., 2016; Garg et al., 2017; Cahill and Medlock, 2017; Ryu et al., 2017; Dinh et al., 2019; Medina-Laver et al., 2021). Recent data provide further evidence and reiterate the long-held claim that the corpus luteum is not only the major site of progesterone synthesis but also a target tissue where progesterone regulates luteal granulosa cell activity through diverse mechanisms (Rothchild, 1996). Some of the mechanisms have been well-characterized in human corpus luteum. However, many have been studied in animal models or in tissues other than the ovaries. Caution should be exercised when extrapolating findings from in vitro studies, and species-specific, tissue/cell-specific, and menstrual phase-dependent differences in progesterone activity need to be taken into consideration. A significant majority of studies on human granulosa cells were based on luteal granulosa cells isolated from the follicular fluid obtained during oocyte retrieval in women who had undergone ART, unless otherwise specified. These mural luteinized granulosa cells of the early stage-corpus luteum may not truly represent pure natural cycles where the mid-cycle surge of endogenous LH triggers the ovulation. In ART, women are subjected to gonadotrophin treatment for ovarian stimulation and hCG or a GnRH analog for induction of final oocyte maturation. Both ovarian stimulation and the mode of triggering of oocyte maturation are associated with significant alterations in the viability and steroidogenic capacity of luteal granulosa cells (Bildik et al., 2019). Moreover, cholesterol uptake/trafficking and progesterone synthesis by luteal granulosa cells are also related to maternal age and ovarian response to gonadotrophin stimulation (Bildik et al., 2022). These findings should be considered when interpreting the findings of such studies.

There is a pressing need for studies to demonstrate the molecular mechanisms of how progesterone manages to control the time-, site-, and context-dependent gene expression in target cells. There are major gaps in our knowledge regarding progesterone-induced signaling and progesterone’s luteotrophic activity. The overwhelming majority of published data is derived from animal models and they remain to be replicated in the human ovary. Figure 4 illustrates a list of research questions that necessitate further molecular and translational studies. However, the list is by no means exhaustive. A better understanding of the molecular mechanisms of progesterone-mediated functions is key to understanding luteal physiology, to illuminate the pathophysiology of luteal disorders, and to develop safe and effective strategies for their clinical management.

Figure 4.

Figure 4.

Major gaps in our understanding and future research priorities regarding progesterone signaling in the regulation of luteal steroidogenesis.

Contributor Information

Kayhan Yakin, Graduate School of Health Sciences, Koç University, Istanbul, Turkey; School of Medicine, Department of Obstetrics and Gynecology, Koç University, Istanbul, Turkey.

Francesko Hela, Graduate School of Health Sciences, Koç University, Istanbul, Turkey; Harvard Medical School, Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, USA.

Ozgur Oktem, Graduate School of Health Sciences, Koç University, Istanbul, Turkey; School of Medicine, Department of Obstetrics and Gynecology, Koç University, Istanbul, Turkey.

Data availability

No new data were generated or analyzed in support of this article.

Authors’ roles

K.Y.: conceived the idea of the article, contributed to the construction of the design and interpretation of the data, and drafted the article; F.H.: contributed to the interpretation of the data and construction of the figures and revised the article for important intellectual content; O.O.: revised the article for important intellectual content; and all authors approved the final version of the article.

Funding

No specific funding was obtained for this review.

Conflict of interest

The authors have no relevant financial or non-financial interests to disclose.

References

  1. Bai Y, Ludescher M, Poschmann G, Stuhler K, Wyrich M, Oles J, Franken A, Rivandi M, Abramova A, Reinhardt F. et al. PGRMC1 promotes progestin-dependent proliferation of breast cancer cells by binding prohibitins resulting in activation of ERalpha signaling. Cancers (Basel) 2021;13:5635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Beranic N, Gobec S, Rizner TL.. Progestins as inhibitors of the human 20-ketosteroid reductases, AKR1C1 and AKR1C3. Chem Biol Interact 2011;191:227–233. [DOI] [PubMed] [Google Scholar]
  3. Bildik G, Akin N, Seyhan A, Esmaeilian Y, Yakin K, Keles I, Balaban B, Ata B, Urman B, Oktem O.. Luteal granulosa cells from natural cycles are more capable of maintaining their viability, steroidogenic activity and LH receptor expression than those of stimulated IVF cycles. Hum Reprod 2019;34:345–355. [DOI] [PubMed] [Google Scholar]
  4. Bildik G, Esmaeilian Y, Hela F, Akin N, Iltumur E, Yusufoglu S, Yildiz CS, Yakin K, Oktem O.. Cholesterol uptake or trafficking, steroid biosynthesis, and gonadotropin responsiveness are defective in young poor responders. Fertil Steril 2022;117:1069–1080. [DOI] [PubMed] [Google Scholar]
  5. Cahill MA. Progesterone receptor membrane component 1: an integrative review. J Steroid Biochem Mol Biol 2007;105:16–36. [DOI] [PubMed] [Google Scholar]
  6. Cahill MA, Jazayeri JA, Kovacevic Z, Richardson DR.. PGRMC1 regulation by phosphorylation: potential new insights in controlling biological activity. Oncotarget 2016;7:50822–50827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Cahill MA, Medlock AE.. Thoughts on interactions between PGRMC1 and diverse attested and potential hydrophobic ligands. J Steroid Biochem Mol Biol 2017;171:11–33. [DOI] [PubMed] [Google Scholar]
  8. Cai Z, Stocco C.. Expression and regulation of progestin membrane receptors in the rat corpus luteum. Endocrinology 2005;146:5522–5532. [DOI] [PubMed] [Google Scholar]
  9. Chaffin CL, Schwinof KM, Stouffer RL.. Gonadotropin and steroid control of granulosa cell proliferation during the periovulatory ınterval in rhesus monkeys. Biol Reprod 2001;65:755–762. [DOI] [PubMed] [Google Scholar]
  10. Chaffin CL, Stouffer RL, Duffy DM.. Gonadotropin and steroid regulation of steroid receptor and aryl hydrocarbon receptor messenger ribonucleic acid in macaque granulosa cells during the periovulatory interval. Endocrinology 1999;140:4753–4760. [DOI] [PubMed] [Google Scholar]
  11. Charles NJ, Thomas P, Lange CA.. Expression of membrane progesterone receptors (mPR/PAQR) in ovarian cancer cells: implications for progesterone-induced signaling events. Horm Cancer 2010;1:167–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Christenson LK, Strauss JF 3rd. Steroidogenic acute regulatory protein: an update on its regulation and mechanism of action. Arch Med Res 2001;32:576–586. [DOI] [PubMed] [Google Scholar]
  13. Chu X, He S, Liu Y, Liu Y, Feng F, Guo Q, Zhao L, Sun H.. Overview of human 20 alpha-hydroxysteroid dehydrogenase (AKR1C1): functions, regulation, and structural insights of inhibitors. Chem Biol Interact 2022;351:109746. [DOI] [PubMed] [Google Scholar]
  14. Curry TE Jr., Nothnick WB.. Mifepristone and ovarian function. Clin Obstet Gynecol 1996;39:486–497. [DOI] [PubMed] [Google Scholar]
  15. Diep CH, Ahrendt H, Lange CA.. Progesterone induces progesterone receptor gene (PGR) expression via rapid activation of protein kinase pathways required for cooperative estrogen receptor alpha (ER) and progesterone receptor (PR) genomic action at ER/PR target genes. Steroids 2016;114:48–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Dinh DT, Breen J, Akison LK, DeMayo FJ, Brown HM, Robker RL, Russell DL.. Tissue-specific progesterone receptor-chromatin binding and the regulation of progesterone-dependent gene expression. Sci Rep 2019;9:11966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Engmann L, Losel R, Wehling M, Peluso JJ.. Progesterone regulation of human granulosa/luteal cell viability by an RU486-independent mechanism. J Clin Endocrinol Metab 2006;91:4962–4968. [DOI] [PubMed] [Google Scholar]
  18. Faivre EJ, Daniel AR, Hillard CJ, Lange CA.. Progesterone receptor rapid signaling mediates serine 345 phosphorylation and tethering to specificity protein 1 transcription factors. Mol Endocrinol 2008;22:823–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Fanjul LF, Ruiz de Galarreta CM, Hsueh AJ.. Progestin augmentation of gonadotropin-stimulated progesterone production by cultured rat granulosa cells. Endocrinology 1983;112:405–407. [DOI] [PubMed] [Google Scholar]
  20. Fortune JE, Vincent SE.. Progesterone inhibits the induction of aromatase activity in rat granulosa cells in vitro. Biol Reprod 1983;28:1078–1089. [DOI] [PubMed] [Google Scholar]
  21. Furuhata R, Kabe Y, Kanai A, Sugiura Y, Tsugawa H, Sugiyama E, Hirai M, Yamamoto T, Koike I, Yoshikawa N. et al. Progesterone receptor membrane associated component 1 enhances obesity progression in mice by facilitating lipid accumulation in adipocytes. Commun Biol 2020;3:479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Garg D, Ng SSM, Baig KM, Driggers P, Segars J.. Progesterone-mediated non-classical signaling. Trends Endocrinol Metab 2017;28:656–668. [DOI] [PubMed] [Google Scholar]
  23. Gerdes D, Wehling M, Leube B, Falkenstein E.. Cloning and tissue expression of two putative steroid membrane receptors. Biol Chem 1998;379:907–911. [DOI] [PubMed] [Google Scholar]
  24. Gordon A, Garrido-Gracia JC, Aguilar R, Sanchez-Criado JE.. Understanding the regulation of pituitary progesterone receptor expression and phosphorylation. Reproduction 2015;149:615–623. [DOI] [PubMed] [Google Scholar]
  25. Griffin D, Liu X, Pru C, Pru JK, Peluso JJ.. Expression of progesterone receptor membrane component-2 within the immature rat ovary and its role in regulating mitosis and apoptosis of spontaneously immortalized granulosa cells. Biol Reprod 2014;91:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Grimm SL, Hartig SM, Edwards DP.. Progesterone receptor signaling mechanisms. J Mol Biol 2016;428:3831–3849. [DOI] [PubMed] [Google Scholar]
  27. Guerra-Araiza C, Coyoy-Salgado A, Camacho-Arroyo I.. Sex differences in the regulation of progesterone receptor isoforms expression in the rat brain. Brain Res Bull 2002;59:105–109. [DOI] [PubMed] [Google Scholar]
  28. Hagiwara A, Ogiwara K, Takahashi T.. Expression of membrane progestin receptors (mPRs) in granulosa cells of medaka preovulatory follicles. Zool Sci 2016;33:98–105. [DOI] [PubMed] [Google Scholar]
  29. Hibbert ML, Stouffer RL, Wolf DP, Zelinski-Wooten MB.. Midcycle administration of a progesterone synthesis inhibitor prevents ovulation in primates. Proc Natl Acad Sci U S A 1996;93:1897–1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Hughes AL, Powell DW, Bard M, Eckstein J, Barbuch R, Link AJ, Espenshade PJ.. Dap1/PGRMC1 binds and regulates cytochrome P450 enzymes. Cell Metab 2007;5:143–149. [DOI] [PubMed] [Google Scholar]
  31. Kimura I, Nakayama Y, Konishi M, Terasawa K, Ohta M, Itoh N, Fujimoto M.. Functions of MAPR (membrane-associated progesterone receptor) family members as heme/steroid-binding proteins. Curr Protein Pept Sci 2012;13:687–696. [DOI] [PubMed] [Google Scholar]
  32. Liszewska E, Rekawiecki R, Kotwica J.. Effect of progesterone on the expression of bax and bcl-2 and on caspase activity in bovine luteal cells. Prostaglandins Other Lipid Mediat 2005;78:67–81. [DOI] [PubMed] [Google Scholar]
  33. MacDonald PC, Dombroski RA, Casey ML.. Recurrent secretion of progesterone in large amounts: an endocrine/metabolic disorder unique to young women? Endocr Rev 1991;12:372–401. [DOI] [PubMed] [Google Scholar]
  34. Makrigiannakis A, Coukos G, Christofidou-Solomidou M, Montas S, Coutifaris C.. Progesterone is an autocrine/paracrine regulator of human granulosa cell survival in vitro. Ann N Y Acad Sci 2000;900:16–25. [DOI] [PubMed] [Google Scholar]
  35. Medina-Laver Y, Rodríguez-Varela C, Salsano S, Labarta E, Domínguez F.. What Do We Know about Classical and Non-Classical Progesterone Receptors in the Human Female Reproductive Tract? A Review. IJMS 2021;22:11278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Migliaccio A, Piccolo D, Castoria G, Di Domenico M, Bilancio A, Lombardi M, Gong W, Beato M, Auricchio F.. Activation of the Src/p21ras/Erk pathway by progesterone receptor via cross-talk with estrogen receptor. EMBO J 1998;17:2008–2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Miller WL, Auchus RJ.. The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr Rev 2011;32:81–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Mir SU, Schwarze SR, Jin L, Zhang J, Friend W, Miriyala S, St Clair D, Craven RJ.. Progesterone receptor membrane component 1/Sigma-2 receptor associates with MAP1LC3B and promotes autophagy. Autophagy 2013;9:1566–1578. [DOI] [PubMed] [Google Scholar]
  39. Misao R, Nakanishi Y, Iwagaki S, Fujimoto J, Tamaya T.. Expression of progesterone receptor isoforms in corpora lutea of human subjects: correlation with serum oestrogen and progesterone concentrations. Mol Hum Reprod 1998;4:1045–1052. [DOI] [PubMed] [Google Scholar]
  40. Misrahi M, Venencie PY, Saugier-Veber P, Sar S, Dessen P, Milgrom E.. Structure of the human progesterone receptor gene. Biochim Biophys Acta 1993;1216:289–292. [DOI] [PubMed] [Google Scholar]
  41. Mussche S, D’Herde K.. Contribution of progesterone, follicle stimulating hormone and glucocorticoids in survival of serum-free cultured granulosa cell explants. J Endocrinol 2001;169:321–331. [DOI] [PubMed] [Google Scholar]
  42. Narimani L, Boroujeni NB, Gholami M, Anbari K, Alavi SER, Ahmadi SAY, Boroujeni MB.. Pre-ımplantation effects of progesterone administration on ovarian angiogenesis after ovarian stimulation: a histological, hormonal, and molecular analysis. JBRA Assist Reprod 2020;24:289–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Nichols JA, Perego MC, Schutz LF, Hemple AM, Spicer LJ.. Hormonal regulation of vascular endothelial growth factor A (VEGFA) gene expression in granulosa and theca cells of cattle1. J Anim Sci 2019;97:3034–3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Oh SJ, Kim TH, Lim JM, Jeong JW.. Progesterone induces expression of Lrp2 in the murine uterus. Biochem Biophys Res Commun 2013;441:175–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Ottander U, Hosokawa K, Liu K, Bergh A, Ny T, Olofsson JI.. A putative stimulatory role of progesterone acting via progesterone receptors in the steroidogenic cells of the human corpus luteum. Biol Reprod 2000;62:655–663. [DOI] [PubMed] [Google Scholar]
  46. Owen GI, Richer JK, Tung L, Takimoto G, Horwitz KB.. Progesterone regulates transcription of the p21(WAF1) cyclin-dependent kinase inhibitor gene through Sp1 and CBP/p300. J Biol Chem 1998;273:10696–10701. [DOI] [PubMed] [Google Scholar]
  47. Pall M, Mikuni M, Mitsube K, Brannstrom M.. Time-dependent ovulation inhibition of a selective progesterone-receptor antagonist (Org 31710) and effects on ovulatory mediators in the in vitro perfused rat ovary. Biol Reprod 2000;63:1642–1647. [DOI] [PubMed] [Google Scholar]
  48. Park OK, Mayo KE.. Transient expression of progesterone receptor messenger RNA in ovarian granulosa cells after the preovulatory luteinizing hormone surge. Mol Endocrinol 1991;5:967–978. [DOI] [PubMed] [Google Scholar]
  49. Pedroza DA, Subramani R, Tiula K, Do A, Rashiraj N, Galvez A, Chatterjee A, Bencomo A, Rivera S, Lakshmanaswamy R.. Crosstalk between progesterone receptor membrane component 1 and estrogen receptor alpha promotes breast cancer cell proliferation. Lab Invest 2021;101:733–744. [DOI] [PubMed] [Google Scholar]
  50. Peluso JJ. Multiplicity of progesterone's actions and receptors in the mammalian ovary. Biol Reprod 2006;75:2–8. [DOI] [PubMed] [Google Scholar]
  51. Peluso JJ. Non-genomic actions of progesterone in the normal and neoplastic mammalian ovary. Semin Reprod Med 2007;25:198–207. [DOI] [PubMed] [Google Scholar]
  52. Peluso JJ, DeCerbo J, Lodde V.. Evidence for a genomic mechanism of action for progesterone receptor membrane component-1. Steroids 2012;77:1007–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Peluso JJ, Griffin D, Liu X, Horne M.. Progesterone receptor membrane component-1 (PGRMC1) and PGRMC-2 interact to suppress entry into the cell cycle in spontaneously immortalized rat granulosa cells. Biol Reprod 2014;91:104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Peluso JJ, Liu X, Gawkowska A, Johnston-MacAnanny E.. Progesterone activates a progesterone receptor membrane component 1-dependent mechanism that promotes human granulosa/luteal cell survival but not progesterone secretion. J Clin Endocrinol Metab 2009;94:2644–2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Peluso JJ, Liu X, Gawkowska A, Lodde V, Wu CA.. Progesterone inhibits apoptosis in part by PGRMC1-regulated gene expression. Mol Cell Endocrinol 2010;320:153–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Peluso JJ, Liu X, Romak J.. Progesterone maintains basal intracellular adenosine triphosphate levels and viability of spontaneously immortalized granulosa cells by promoting an interaction between 14-3-3sigma and ATP synthase beta/precursor through a protein kinase G-dependent mechanism. Endocrinology 2007;148:2037–2044. [DOI] [PubMed] [Google Scholar]
  57. Peluso JJ, Liu X, Uliasz T, Pru CA, Kelp NC, Pru J.. PGRMC1/2 promotes luteal vascularization and maintains the primordial follicles of mice. Reproduction 2018;156:365–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Peluso JJ, Pappalardo A, Losel R, Wehling M.. Expression and function of PAIRBP1 within gonadotropin-primed immature rat ovaries: PAIRBP1 regulation of granulosa and luteal cell viability. Biol Reprod 2005;73:261–270. [DOI] [PubMed] [Google Scholar]
  59. Peluso JJ, Pappalardo A, Losel R, Wehling M.. Progesterone membrane receptor component 1 expression in the immature rat ovary and its role in mediating progesterone's antiapoptotic action. Endocrinology 2006;147:3133–3140. [DOI] [PubMed] [Google Scholar]
  60. Peluso JJ, Pru CA, Liu X, Kelp NC, Pru JK.. Progesterone receptor membrane component 1 and 2 regulate granulosa cell mitosis and survival through a NFKappaB-dependent mechanism. Biol Reprod 2019;100:1571–1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Peluso JJ, Pru JK.. Non-canonical progesterone signaling in granulosa cell function. Reproduction 2014;147:R169–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Peluso JJ, Romak J, Liu X.. Progesterone receptor membrane component-1 (PGRMC1) is the mediator of progesterone's antiapoptotic action in spontaneously immortalized granulosa cells as revealed by PGRMC1 small interfering ribonucleic acid treatment and functional analysis of PGRMC1 mutations. Endocrinology 2008;149:534–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Penning TM, Jin Y, Steckelbroeck S, Lanisnik Rizner T, Lewis M.. Structure-function of human 3 alpha-hydroxysteroid dehydrogenases: genes and proteins. Mol Cell Endocrinol 2004;215:63–72. [DOI] [PubMed] [Google Scholar]
  64. Piel RB 3rd, Shiferaw MT, Vashisht AA, Marcero JR, Praissman JL, Phillips JD, Wohlschlegel JA, Medlock AE.. A novel role for Progesterone Receptor Membrane Component 1 (PGRMC1): a partner and regulator of ferrochelatase. Biochemistry 2016;55:5204–5217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Pietrowski D, Gong Y, Mairhofer M, Gessele R, Sator M.. Effects of progesterone and its metabolites on human granulosa cells. Horm Metab Res 2014;46:133–137. [DOI] [PubMed] [Google Scholar]
  66. Quirk SM, Cowan RG, Harman RM.. Role of the cell cycle in regression of the corpus luteum. Reproduction 2013;145:161–175. [DOI] [PubMed] [Google Scholar]
  67. Rekawiecki R, Nowik M, Kotwica J.. Stimulatory effect of LH, PGE2 and progesterone on StAR protein, cytochrome P450 cholesterol side chain cleavage and 3beta hydroxysteroid dehydrogenase gene expression in bovine luteal cells. Prostaglandins Other Lipid Mediat 2005;78:169–184. [DOI] [PubMed] [Google Scholar]
  68. Riad A, Zeng C, Weng CC, Winters H, Xu K, Makvandi M, Metz T, Carlin S, Mach RH.. Sigma-2 receptor/TMEM97 and PGRMC-1 ıncrease the rate of ınternalization of LDL by LDL receptor through the formation of a ternary complex. Sci Rep 2018;8:16845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Rothchild I. The regulation of the mammalian corpus luteum. Recent Prog Horm Res 1981;37:183–298. [DOI] [PubMed] [Google Scholar]
  70. Rothchild I. The corpus luteum revisited: are the paradoxical effects of RU486 a clue to how progesterone stimulates its own secretion? Biol Reprod 1996;55:1–4. [DOI] [PubMed] [Google Scholar]
  71. Ruiz de Galarreta CM, Fanjul LF, Hsueh AJ.. Progestin regulation of progesterone biosynthetic enzymes in cultured rat granulosa cells. Steroids 1985;46:987–1002. [DOI] [PubMed] [Google Scholar]
  72. Rung E, Friberg PA, Shao R, Larsson DG, Nielsen E, Svensson PA, Carlsson B, Carlsson LM, Billig H.. Progesterone-receptor antagonists and statins decrease de novo cholesterol synthesis and increase apoptosis in rat and human periovulatory granulosa cells in vitro. Biol Reprod 2005;72:538–545. [DOI] [PubMed] [Google Scholar]
  73. Ryu CS, Klein K, Zanger UM.. Membrane associated progesterone receptors: promiscuous proteins with pleiotropic functions—focus on ınteractions with cytochromes P450. Front Pharmacol 2017;8:159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Shao R, Markstrom E, Friberg PA, Johansson M, Billig H.. Expression of progesterone receptor (PR) A and B isoforms in mouse granulosa cells: stage-dependent PR-mediated regulation of apoptosis and cell proliferation. Biol Reprod 2003;68:914–921. [DOI] [PubMed] [Google Scholar]
  75. Shimizu T, Miyamoto A.. Progesterone induces the expression of vascular endothelial growth factor (VEGF) 120 and Flk-1, its receptor, in bovine granulosa cells. Anim Reprod Sci 2007;102:228–237. [DOI] [PubMed] [Google Scholar]
  76. Stouffer RL. Progesterone as a mediator of gonadotrophin action in the corpus luteum: beyond steroidogenesis. Hum Reprod Update 2003;9:99–117. [DOI] [PubMed] [Google Scholar]
  77. Suchanek M, Radzikowska A, Thiele C.. Photo-leucine and photo-methionine allow identification of protein-protein interactions in living cells. Nat Methods 2005;2:261–267. [DOI] [PubMed] [Google Scholar]
  78. Sueldo C, Liu X, Peluso JJ.. Progestin and AdipoQ Receptor 7, Progesterone Membrane Receptor Component 1 (PGRMC1), and PGRMC2 and their role in regulating progesterone's ability to suppress human granulosa/luteal cells from entering into the cell cycle. Biol Reprod 2015;93:63. [DOI] [PubMed] [Google Scholar]
  79. Sugino N, Telleria CM, Gibori G.. Progesterone inhibits 20alpha-hydroxysteroid dehydrogenase expression in the rat corpus luteum through the glucocorticoid receptor. Endocrinology 1997;138:4497–4500. [DOI] [PubMed] [Google Scholar]
  80. Tang Z, Zhang Z, Zhang H, Wang Y, Zhang Y, Zhao J, Yang H, Wang Z.. Autophagy attenuation hampers progesterone synthesis during the development of pregnant corpus luteum. Cells 2019;9:71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Taraborrelli S. Physiology, production and action of progesterone. Acta Obstet Gynecol Scand 2015;94:8–16. [DOI] [PubMed] [Google Scholar]
  82. Uchida K, Kadowaki M, Miyake T.. Further studies on the effect of exogenous progesterone and related compounds on the preovulatory progesterone secretion in the rat. Endocrinol Jpn 1970;17:99–106. [DOI] [PubMed] [Google Scholar]
  83. VandeVoort CA, Overstreet JW, Lasley BL, Stewart DR.. Effects of progesterone receptor blockers on human granulosa-luteal cell culture secretion of progesterone, estradiol, and relaxin. Biol Reprod 2000;62:200–205. [DOI] [PubMed] [Google Scholar]
  84. Warnick DC. Role of PGRMC1 in intracellular sterol transport. Ph.D. Thesis. The University of Utah, J. Willard Marriott Digital Library, 2008, 170.
  85. Wei LL, Hawkins P, Baker C, Norris B, Sheridan PL, Quinn PG.. An amino-terminal truncated progesterone receptor isoform, PRc, enhances progestin-induced transcriptional activity. Mol Endocrinol 1996;10:1379–1387. [DOI] [PubMed] [Google Scholar]
  86. Xia Z, Xiao J, Chen Q.. Solving the puzzle: what ıs the role of progestogens in neovascularization? Biomolecules 2021;11:1686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Yang T, Espenshade PJ, Wright ME, Yabe D, Gong Y, Aebersold R, Goldstein JL, Brown MS.. Crucial step in cholesterol homeostasis: sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell 2002;110:489–500. [DOI] [PubMed] [Google Scholar]
  88. Yao X, Wang Z, Gao X, Li X, Yang H, Ei-Samahy MA, Bao Y, Xiao S, Meng F, Wang F.. Unconservative_15_2570409 suppresses progesterone receptor expression in the granulosa cells of Hu sheep. Theriogenology 2020;157:303–313. [DOI] [PubMed] [Google Scholar]
  89. Zhu Y, Bond J, Thomas P.. Identification, classification, and partial characterization of genes in humans and other vertebrates homologous to a fish membrane progestin receptor. Proc Natl Acad Sci U S A 2003;100:2237–2242. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No new data were generated or analyzed in support of this article.


Articles from Molecular Human Reproduction are provided here courtesy of Oxford University Press

RESOURCES