45 Mechanism of Progesterone Receptor Action in the Brain S K Mani and B W O’Malley, Baylor College of Medicine, Houston, TX, USA ß 2009 Elsevier Inc. All rights reserved.
Chapter Outline 45.1 45.2 45.2.1 45.2.2 45.2.3 45.2.3.1 45.2.4.2 45.2.4 45.2.4.1 45.2.4.2 45.2.4.3 45.2.5 45.2.6 45.3 45.4 45.4.1 45.4.2 45.5 45.5.1 45.5.2 45.5.3 45.6 45.6.1 45.6.2 45.6.3 45.7
Introduction Structure and Function of PRs: An Overview Structural Organization Gene Activation Coactivators and Repressors Coactivators Corepressors NRs and Chromatin Coactivators and acetylation Chromatin-remodeling proteins Corepressors and deacetylation Receptor Activation and Phosphorylation PR Isoforms Ligand-Independent Activation of PRs Cellular Function of Progesterone in the CNS Reproductive Physiology and Behavior Species Variations PRs in the CNS Spatial and Temporal Correlation between PR Induction and Behavior Estrogen-Inducible versus Estrogen-Noninducible PRs PR Isoforms in the Brain Progestin Receptor Activation in the Brain: Relationship to Female Sexual Behavior Genomic Mechanisms Nongenomic Mechanisms Ligand-Independent Mechanism: An Alternate Mechanism of PR Action in the CNS Mechanisms of Action of Progesterone and DA on Female Reproductive Behavior Interactions between Progesterone and Neurotransmitters DA Signaling and PR Pathway Convergence Multi-signal Pathway Reinforcement Coactivators and PRs in the Brain PRs and CNS Drug Actions PRs and Male Sexual Behavior PRs in Development PRs and Other Behavioral Effects Summary and Conclusions
45.7.1 45.7.2 45.7.3 45.7.4 45.8 45.9 45.10 45.11 45.12 References Further Reading
1468 1468 1469 1469 1470 1470 1472 1473 1473 1473 1473 1473 1474 1475 1476 1476 1477 1477 1478 1478 1479 1479 1480 1480 1482 1483 1483 1484 1485 1487 1488 1488 1489 1490 1490 1491 1503
1467
1468
Mechanism of Progesterone Receptor Action in the Brain
Glossary coregulators Molecules that help the transcription factors (proteins) bind to DNA to activate or repress gene transcription. isoform Different forms of a protein that may be produced from different genes, or from the same gene. ligand A molecule, atom, or ion that specifically and reversibly binds to a protein, causing a conformational change in the protein. neuroendocrine Relating to interplay between the endocrine system and the nervous system. neurotransmitter A chemical messenger released from the synaptic terminal of a neuron at a chemical synapse that diffuses across the synaptic cleft and binds to and stimulates the post synaptic membrane. progesterone A progestin synthesized mainly in the ovary. receptors Cellular proteins that recognize and bind to specific chemical messengers (e.g., hormones and neurotransmitters) and transduce the chemical signal into a change in cellular function. signaling pathways The biochemical pathways by which proteins send signals from the cell surface to the nucleus. transcription The process whereby genetic (DNA) information is copied into ribonucleic acid prior to protein synthesis. translation The process of protein synthesis whereby the primary structure of the protein is determined by the nucleotide sequence in mRNA.
45.1 Introduction Progesterone, a 21-carbon steroid with a cyclopentanoperhydrophenanthrene structure, is one of the most biologically active progestins of ovarian origin. Progestin (from pro, meaning favoring and gest gestation, pregnancy) was originally discovered as the mammalian hormone of pregnancy in the rabbit and was shown to be essential for the implantation of embryos and maintenance of pregnancy (Fraenkel and Cohn, 1901; Marshall and Jolly, 1905; Hammond and Marshall, 1925). The initial observations by Corner (1929) that corpus luteal extracts from the rabbit-induced progestational endometrium were
followed by demonstrations (Allen and Corner, 1929, 1930) of their ability to induce implantation of the fertilized egg and to maintain pregnancy in the castrated rabbit. These observations led to the identification and purification of progesterone as the active substance in the extract (Allen et al., 1935). In the decades following these discoveries, the use of isolated progesterone in a multitude of studies defined the multiple functions of progesterone in a variety of species. The results of these studies have led to the current understanding of the coordinating role of progesterone in other interdependent reproductive functions such as ovulation, mammary gland development, and reproductive behavior (FernandezValdivia et al., 2005). The studies also established that prior sensitization of target tissues by the ovarian steroid hormone, estrogen, was essential for the modulation of reproductive function by progesterone (Burrows, 1949; Hisaw et al., 1937). Estrogen priming results in the induction of several proteins including the progestin receptor (PR). PRs mediate progesterone effects in target tissues (O’Malley and Means, 1974; O’Malley et al., 1991). This chapter focuses on the cellular and molecular mechanisms of progesterone action on one target tissue, that is, the central nervous system. To gain a better understanding and appreciation of progesterone action in the brain, we begin by summarizing our current knowledge of the cellular and molecular biology of PRs.
45.2 Structure and Function of PRs: An Overview Progestins including progesterone exert their physiological effects primarily by binding to cognate intracellular PRs ( Jensen et al., 1968; Yamamoto, 1985; Beato, 1991; O’Malley et al., 1991). PRs are ligand (hormone)-inducible members of a superfamily of transcription factors that undergo conformational changes upon hormone binding, leading to their nuclear translocation, dimerization, DNA binding (Denner et al., 1990b; Beato, 1989; Beato et al., 1995; Umesono and Evans, 1989; Carson-Jurica et al., 1990; Chauchereau et al., 1994), and modulation of target gene expression (Klein-Hitpass et al., 1990; Bagchi et al., 1990). Both cytoplasmic and nuclear phosphorylations occur after hormone binding to the intracellular PRs (Denner et al., 1990a; Weigel et al., 1992; Bagchi et al., 1992), followed by a final round of phosphorylation by a DNA-dependent kinase prior to target gene activation (Takimoto et al., 1992; Sheridan et al., 1989).
Mechanism of Progesterone Receptor Action in the Brain
also contains sequences for heat-shock protein association, receptor dimerization, intermolecular silencing, and intramolecular repression (Guichon-Mantel et al., 1989; Vegeto et al., 1992). A unique 164-aminoacid-long third activation function (AF-3) has been also described in the N-terminal segment of human PR (B-form) which, depending on the cell and promoter context, can either function autonomously or synergize with the downstream activation domains to enhance their activity (Sartorius et al., 1994).
45.2.1 Structural Organization Similar to the other members of the nuclear receptor superfamily, PRs have a modular protein structure organized into distinct functional domains: a variable amino (N)-terminal domain, a short and wellconserved cysteine-rich central domain, and a relatively well-conserved carboxy (C)-terminal domain (Figure 1). The N-terminal region (A/B region) contains a transactivation function (AF-1) that modulates the level and promoter specificity of target gene activation by interacting with components of the core transcriptional machinery, coactivator proteins, and other transactivating proteins (Bai and Weigel, 1995). This region is important for determining target gene specificity for PR isoforms that recognize the same response element (Tora et al., 1988; Becquel et al., 1989). The central C region consisting of 66–68 amino acids is the DNA-binding domain (DBD) and is composed of two type II zinc (Zn) fingers which facilitate binding of the receptor to specific cis-acting DNA sequences (Evans, 1988; Luisi et al., 1991; Freedman, 1992). The DBD also has a receptor dimerization function and can contribute to other protein–protein interactions. Downstream of the C region is a small variable hinge region (D), which contains a nuclear localization signal sequence (Guichon-Mantel et al., 1989). The large (250–300 amino acids) and functionally complex ligand-binding domain (LBD or E region) is located in the carboxy-terminal region. This region contains a second activation function (AF-2), which is indispensable for ligand-dependent activation. In addition to the progesterone-binding function, it
1
45.2.2
Gene Activation
PRs are transcription factors that regulate the expression of target genes involved in metabolism, development, and reproduction. In the absence of ligand, the inactive receptors are associated with a large complex of chaperone proteins in the cytoplasm of target cells (Smith et al., 1990). Upon hormone binding, the PRs dissociate from the chaperone proteins, dimerize, translocate to the nucleus, and bind to progesteroneresponsive elements (PREs) in the regulatory regions of target gene DNA (Evans, 1988; Beato et al., 1987; Denner et al., 1989). Activated, DNA-bound PRs stimulate the rate of formation and/or stabilization of a preinitiation complex, consisting of general transcription factors (GTFs), at enhancer-controlled promoters (Klein-Hitpass et al., 1990; Leroy et al., 1991; Kastner et al., 1990a). The preinitiation complex formation involves direct protein–protein interactions with ancillary factors such as coregulators (see Section 45.2.3) and is initiated by the binding of transcription factor-IID (TFIID) to the TATA box of the promoter, a short
ATGA
ATGB
N
1469
165 A/B AF-3
C AF-1
D
E AF-2
C Transactivation DNA binding Ligand-binding translocation Dimerization
Figure 1 Structural domains of progestin receptor (PR). The amino terminal region (A/B) interacts with the transcriptional machinery, coactivators, and other transactivating proteins. This region also contains the two alternate transcription initiation sites (ATG) that give rise to the two isoforms PR-A and PR-B. The C domain is the DNA-binding domain associated with the dimerization function. The ligand-binding domain (E) and the C domain are connected by the variable hinge domain (D), which contains the nuclear localization signal sequence. The activation functions AF-1 and AF-2 are localized to the A/B domain and E domains, respectively. The third activation function AF-3, also termed the B-upstream segment, is located at the N-terminal segment of A/B domain.
1470
Mechanism of Progesterone Receptor Action in the Brain
distance from the transcriptional start site. TFIID functions as a multi-protein complex composed of TATA-binding protein (TBP) and highly conserved TBP-associated factors (TAFIIs). PRs associate with TAFII110 subunit (Schwerk et al., 1995). TFIIDpromoter complex serves as a specialized nucleoprotein complex that allows recruitment of RNA polymerase II and downstream GTFs, or a preassembled holoenzyme to a chromatin template (Roeder, 1996; Hoffman et al., 1997). Whether the preinitiation complex is preformed or recruited sequentially, the rate of assembly of the complexes induced by PRs, in association with their coregulators, ultimately defines the transcriptionally permissive or nonpermissive environment at the hormone-regulated promoters. 45.2.3
Coactivators and Repressors
In the last decade, a number of proteins termed the coregulators that form a functional link between the activated receptors and transcription complex to effect an efficient transcriptional regulation were discovered. These proteins consist of coactivators and corepressors that enhance or inhibit receptordependent target gene transcription. More than 300 coregulators have been identified till date. Primary coregulators directly interact with steroid receptors and exist in large steady-state complexes with multiple secondary partners. Each of the partners may possess multiple enzymatic capabilities such as acetyl transferase, methyltransferase, phosphokinase, ubiquitin ligase, and ATPase activities. Coregulator activity is affected by its phosphorylation, methylation, acetylation, or other modifications, forming a posttranslational modification code. The coregulator’s transcriptional activity and transcription factor preferences are dictated by this code. Thus, the relative expression level of the coactivators and corepressors could determine cell-specific, appropriate, and graded responses to ligand. For a detailed description of coregulators, the reader is referred to several recent reviews and the references therein (McKenna et al., 1999; Lonard et al., 2008; Lonard and O’Malley, 2007; O’Malley, 2007). In this chapter, we summarize those proteins that function as coactivators and corepressors for the progesterone receptors. 45.2.3.1 Coactivators
Coactivators are often rate limiting for receptor activation and achieve their effects through multiple mechanisms, including stabilization of nuclear receptors (NRs) and the basal transcriptional machinery at
the promoter; covalent modification of histones, activators, other coregulators; and, possibly, turnover of activators and other proteins. Their recruitment is usually, but not always, ligand dependent. The first steroid receptor coactivator-1 (SRC-1) was cloned from the human B-lymphocyte cDNA library and characterized by Ona˜te et al. (1995). A series of biochemical and yeast two-hybrid studies in search of other coactivators led to the characterization of an SRC family of three 160-kDa proteins (p160 coactivators or the SRCs). These studies used hormone-bound receptor LBDs as baits since a strong ligand-dependent transactivation function resides in the C-terminus of the AF-2 domain (Figure 1). The SRC family of p160 coactivators can be classified into three distinct, but related categories represented as SRC-1/NcoA-1; TIF2/GRIP1/NcoA2/SRC-2; and p/CIP/ACTR/AIB1/TRAM-1/RAC3/SRC-3 (Ding et al., 1998). In addition to sequence homology, these coactivators share an ability to stimulate liganddependent transactivation by a large number of nuclear steroid receptors, including the PR. A distinctive structural feature of the SRC family of coactivators is the presence of the recurrent pentapeptide LXXLL signature motif (where L is leucine and X any amino acid), which determines their direct binding to the LBD. Three such motifs, also termed NR boxes, are conserved in the members of the SRC family of coactivators. An additional nonconserved NR box present in the C-terminus of SRC-1 enhances hormone-dependent interaction of SRC-1 with PR (Ding et al., 1998). The distinct NR box motifs exhibit differential binding to different NRs (Ding et al., 1998). Distinct LXXXL motifs and adjacent NR boxes exhibit differential binding affinity for different steroid receptors, suggesting that the receptors have a preference for one motif over the other in the same coactivator, leading to a preference of one coactivator molecule over another (Li et al., 2004). This has been confirmed in studies, wherein PR preferentially recruits SRC-1, SRC-3, and CBP, but not SRC-3 or pCAF (Li et al., 2004). 45.2.3.1(i)
SRC-1/NcoA-1
SRC-1 interacts with the PR in a ligand-dependent manner, and the antagonist RU486 prevents this interaction (Ding et al., 1998). In addition to the coactivation of the PR in transient co-transfection assays, SRC-1 interacts with the other members of the NR superfamily (Ding et al., 1998; Wang et al., 1998; Zhu et al., 1996). It also has limited ability to coactivate unrelated transcription factors, like activating protein-1 (AP-1; Lee et al., 1998), serum
Mechanism of Progesterone Receptor Action in the Brain
1471
response factor (Kim et al., 1998), and nuclear factor kappa B (NF-kB; Na et al., 1998). Although SRC-1 interacts with the PR predominantly through the AF2-containing LBD, several studies have demonstrated functional interactions with the N-terminal AF-1 activation function of the PR (Ona˜te et al., 1998). Thus, SRC-1 appears to communicate functional interactions between the AF-1 and AF-2 of the PR to achieve maximal transcription at target genes. SRC-1 contains two activation domains, which interact with other activators or the GTFs (Ona˜te et al., 1998; McInerney et al., 1996), binding domains for cAMP response-element binding (CREB)-binding protein (CBP) and the histone acetyltransferase (HAT) p/CAF, and an intrinsic HAT activity. A role for SRC-1 in chromatin remodeling via histone modification has been confirmed in vitro (Li et al., 2004). A longer form of SRC-1 also has been cloned in the mouse (NCoA-1 or NR coactivator-1/mouse SRC-1) suggesting the existence of splice variants (Kamei et al., 1996), the biological relevance of which is yet to be determined. Cyclin A/Cdk2 phosphorylates SRC-1 and is a component of PR transcriptional complex (Moore et al., 2007). A physiologic role for SRC-1 in progesterone action has been demonstrated in the SRC-1 null mutant mice, which, while viable and fertile, demonstrate a significantly decreased growth of steroid target organs in response to hormone stimulation (Xu et al., 1996). Interestingly, the SRC-1 knockout mouse exhibited full complement of lordosis response, despite the absence of the coactivator. This is presumed to be due to the overexpression of SRC-3, but on SRC-2, in these animals (Apostolakis et al., 2002).
The third member of SRC family, a highly polymorphic protein with homology to SRC-1 and SRC-2, was also isolated independently as p300/CBP cointegrator-associated protein (pCIP), activator of thyroid receptor (ACTR). Receptor-associated coactivator-3 (RAC-3), amplified in breast cancer (AIB-1), thyroid receptor activator molecule 1 (TRAM-I), and SRC-3 (Voegel et al., 1996; Torchia et al., 1997; Walfish et al., 1997; Chen et al., 1997; Li et al., 1997; Anzick et al., 1997; Takeshita et al., 1997; Suen et al., 1998). SRC-3 coactivates several NRs (Chen et al., 1997; Li et al., 1997) including the PR (Li et al., 1997), and other unrelated transcription factors such as those in the cyclic adenosine 30 ,50 monophosphate (cAMP), growth hormone, or cytokine pathways (Torchia et al., 1997). Its role as a post-translational modification-encoded signal integrator in PR regulation has been well established (Lonard and O’Malley, 2007). It is expressed in a tissue-specific fashion and distributed mainly in the oocytes, mammary gland, uterus, pituitary, testis, heart, and skeletal muscle, to low and barely detectable levels in the bone marrow, liver, lung, brain, kidney, stomach, and adrenal glands (Suen et al., 1998). Mice lacking SRC-3 exhibit retarded growth, reduced sexual maturation, decreased reproductive function, and delayed puberty, characteristics not observed in SRC-1 knockout mice, indicating a distinct in vivo role for SRC-3 (Xu et al., 2000).
45.2.3.1(ii)
45.2.3.1(iv)
TIF2/GRIP1/NcoA2/ SRC-2
The second coactivator having sequence homology to SRC-1, referred to as glucocorticoid receptorinteracting protein-1 (GRIP-1/mouse SRC-2), transcription intermediary factor-2 (TIF-2/human SRC-2), and NCoA-2 was independently identified by several laboratories (Voegel et al., 1996; Hong et al., 1996, 1997; Torchia et al., 1997). This coactivator also has been shown to interact with LBD of the PR in a hormone-dependent manner and coactivate the PR in transient cotransfection assays (Voegel et al., 1996; Hong et al., 1997). SRC-2 also contains two autonomous activation domains capable of stimulating transcription. Adaptation among certain SRC family coactivators is evident in the compensatory elevation in the expression of SRC-2 in the SRC-1 null mice (Xu et al., 1998). SRC-2 null
mice have reduced fertility due to gonadal failure (Gehin et al., 2002). 45.2.3.1(iii) p/CIP/ACTR/AIB1/TRAM-1/ RAC3/SRC-3
Cointegrators: CBP/p300
CBP and the related adenovirus E1A-associated 300-kDa protein (p300) function as coactivators for a diverse group of transcription factors, including the PRs (Chakravarti et al., 1996; Voegel et al., 1998; Fronsdal et al., 1998; Smith et al., 1996; Heery et al., 1997; Hanstein et al., 1996; Kraus and Kadonaga, 1998; Eckner et al., 1994; Tetel et al., 1999). CBP interacts with all three SRC family members (Torchia et al., 1997; Chakravarti et al., 1996; Voegel et al., 1998). It has been shown to exist in a stable preformed complex with RNA Pol II (Fronsdal et al., 1998) and does not form a stable complex with SRC-1 (McKenna et al., 1998). It synergizes with SRC-1 to coactivate PR (Smith et al., 1996). CBP/p300 serve as common limiting cointegrators for distinct but convergent signaling pathways, functioning to integrate
1472
Mechanism of Progesterone Receptor Action in the Brain
multiple afferent signals into an appropriate response at promoters containing multiple response elements (Kamei et al., 1996). 45.2.3.1(v) E3 ubiquitin-protein ligases: E6-AP and RPF-1
E6 papillomavirus-associated protein (E6-AP) is an E3 ubiquitin-protein ligase that targets proteins for proteosomal degradation by the ubiquitin pathway. This protein interacts with and coactivates PR and other NRs (Nawaz et al., 1999). The ubiquitin ligase activity of E6-AP is independent of its coactivation function (Nawaz et al., 1999). It is closely related to the E3 ubiquitin-protein ligase RPF-1, which has been shown to enhance PR transactivation (Imhof and McDonnell, 1996). E6-AP and RPF-1 synergistically enhance PR transactivation in mammalian cells (McKenna et al., 1998). Mutations in the E6-AP gene are associated with Angelman syndrome, a mental retardation disorder of humans (Kishino et al., 1997; Matsura et al., 1997). 45.2.3.1(vi)
L7/SPA
L7/SPA, a small (27-kDa) leucine zipper-containing protein, potentiates the partial agonist activity of RU486-bound PR transactivation but has been shown to have no effect on agonist-bound PR or pure antagonist ZK98299-bound PR (Jackson et al., 1997). 45.2.3.1(vii)
HMG-1/2
The high-mobility group (HMG) proteins 1 and 2 enhance the sequence-specific DNA binding of steroid receptors, including the PR. They stabilize the PR– DNA complex and enhance PR-dependent transcription seven- to tenfold in transient transfection assays (Boonyaratanakornkit et al., 1998; Verrijdt et al., 2002). 45.2.3.1(viii)
Steroid receptor RNA activator
Steroid receptor RNA activator (SRA) functions as an RNA transcript to selectively coactivate steroid receptors, including the PR (Lanz et al., 1999). It exists in a multi-protein complex with SRC-1 and mediates transactivation of the receptor via AF-1 region (Lanz et al., 1999). Six RNA motifs present in SRA are important for coactivation (Lanz et al., 2002). SRA has been demonstrated in the brain where it overlaps with the expression of certain steroid receptors. 45.2.3.1(ix) ASC2/TRBP/RAP250/NRC/PRIP/ NCoA6/AIB3/KIAA0181
Activating signal cointegrator 2 (ASC2) is a coactivator involved in PR-mediated transcription. The
C-terminus of ASC2 interacts with SRC-1, CBP/p300 and DRIP130, and basal transcription factors to mediate promotor-specific alternative splicing of gene products (Auboeuf et al., 2004). Various signaling pathways can target ASC2. The resulting post-translational modifications could result in preferential interaction with other coactivators, and permit synthesis of the spliced variant in a given context. The physiological role of ASC2 in progesterone signaling is under investigation. 45.2.4.2 Corepressors
PRs have little DNA-binding activity in the absence of hormone and are sequestered in ternary interactions with heat-shock proteins, hsp90 and hsp70 (Ding et al., 1998). Binding of an agonist or antagonist facilitates DNA–PR interactions. Interestingly, upon binding an antagonist such as RU486, PRs undergo dimerization and DNA binding, but are transcriptionally inactive. The inability of RU-bound PR to activate transcription has been attributed to be a consequence of its inability to associate with coactivators (Shiau et al., 1998), and its consequent availability to recruit corepressor proteins in vitro (Baniahmed et al., 1995). Thus, the agonists and antagonists affect the PR’s ability to activate transcription by inducing different conformational changes within the receptor. A number of studies have demonstrated the interaction of NR corepressors with RU486-bound PRs. These soluble corepressor proteins interact with repressor regions in the receptors (Baniahmed et al., 1995). The existence of a repression domain in the C-terminal region of the PRs was postulated based on studies with PR mutants that lacked a short sequence in this domain and could be activated by RU486 (Zhang et al., 1998; Lanz and Rusconi, 1994). These domains were found to be transferable and, when fused to a heterologous DBD, mediated transcriptional silencing, indicating an interaction with a soluble corepressor (Xu et al., 1996). Several proteins have been identified as potential mediators of repression. NR corepressor (NcoR/ RIP-13) and silencing mediator for retinoid and thyroid hormone receptor (SMRT) were cloned as two closely related molecules and function as corepressors, allowing unliganded retinoid (RXR) and thyroid hormone receptors (TRs) to repress target gene transcription ( Jackson et al., 1997; Smith et al., 1997). Jackson et al. (1997) have shown that NcoR interacted with RU486-bound PR-LB and overexpression of both NCoR and SMRT suppressed the agonist-like
Mechanism of Progesterone Receptor Action in the Brain
transcriptional activity of RU486-bound PR. This suppressive effect was found to be reversible in the presence of overexpressed PR-LBD ( Jackson et al., 1997). Wagner et al. (1998) demonstrated that NcoR and SMRT preferentially associate with RU486bound PR and that the partial agonist activity of RU486-occupied PR can be suppressed by the overexpression of corepressors. The studies also suggest that the inability of NcoR and SMRT to interact with agonist-activated PR could be due to the increased affinity of PR for coactivators, with a subsequent displacement of corepressors. The interactions of corepressors with antagonist-bound PR appear to vary with the isoforms of PR (Section 45.2.5). Several key proteins possessing histone deacetylase activity also have been shown to mediate transcriptional repression and are discussed in Section 45.2.4. 45.2.4
NRs and Chromatin
Eukaryotic chromosomes are organized into a regularly repeating protein DNA unit called the nucleosome, the integrity of which is dependent upon periodic arrays of DNA-bound histone octamers. Low-resolution structure of a nucleosome core particle indicates a left-handed DNA superhelix wrapped around a histone octamer (Richmond et al., 1984). Higher tiers of organization involve the assembly of nucleosomes into chromatin domains. Such an arrangement creates a thermodynamic barrier against the access of transcription factors to their DNA substrate. Binding of the activated receptor to the enhancer region directs modification of the local chromatin structure into a transcriptionally permissive state (derepression), followed by the recruitment of GTFs to form a preinitiation complex at the promoter (activation). Covalent modification of nucleosomal structure is regulated by diametrically opposed activities of histone acetylation, correlated with gene activation, and histone deacetylation, generally associated with gene repression (Wolfe and Pruss, 1996). 45.2.4.1 Coactivators and acetylation
Hyperacetylation of core histones reduces the net positive charge and weakens their interactions with the negatively charged DNA creating an environment accessible to the transcription factors. An additional effect of acetylation of lysine residues in the amino-terminal tails of histones results in nucleosome–nucleosome contacts to disrupt higher-order chromatin structures (Rhodes, 1997).
1473
The discovery that p300, CBP, and a p300/CBPassociated factor (PCAF) contained HAT activity indicated a role for acetylation in transcriptional regulation of steroid receptors, including PRs (Bannister and Kouzarides, 1996; Ogryzko et al., 1996; Yang et al., 1996). Similar HAT activity found in the SRC family members, including SRC-1 and ACTR/SRC-3 (Chen et al., 1997; Spencer et al., 1997), strengthens their role in chromatin transcription. 45.2.4.2 Chromatin-remodeling proteins
In addition to the HATs, several ATPase complexes that effect noncovalent modifications of chromatin domains are important for transcriptional regulation by NRs. Studies have demonstrated that purified SWI/SNF complexes have intrinsic ATPase activity and function by coupling ATP hydrolysis to nucleosomal remodeling at diverse promoters to facilitate interaction of basal transcription factors (Imbalzano et al., 1994). Studies demonstrating the existence of human SWI/SNF homologs (i.e., BR6-1 complex) in a stable complex with SRC-1 (Guyon et al., 2001) suggest that they also are involved in the activation functions of PR. 45.2.4.3 Corepressors and deacetylation
Several distinct proteins, including RPD/SIN3 and RPD-3, have been shown to negatively regulate the transcriptional activity of the PR by deacetylation of core histones (Rundlett et al., 1996; Tauton et al., 1996). Biochemical evidence indicates that SIN3 proteins and histone deacetylases exist in stable preformed complexes in mammalian cells ( Jones et al., 1998; Wade et al., 1998). Unliganded receptors maintain a transcriptionally inactive steady state at the promoter by recruitment of corepressors and their associated histone deacetylase activities. Ligand binding is thought to induce release of corepressors enabling the receptor to recruit PCAF, p300/CBP, and SRC family members to effect histone acetylation and the creation of a transcriptionally permissive environment at the promoter. 45.2.5 Receptor Activation and Phosphorylation PRs are phosphoproteins that are phosphorylated in response to progesterone and the activation of various kinases (Weigel, 1996; Lange, 2004). Phosphopeptide mapping and mass spectrometry studies indicate that PRs exhibit significant amounts of basal phosphorylation at multiple sites in the absence of progesterone
1474
Mechanism of Progesterone Receptor Action in the Brain
and are hyperphosphorylated upon stimulation (Sullivan et al., 1988; Denner et al., 1990a; Rao et al., 1987; Knotts et al., 2001; Moore et al., 2007). A significant level of delayed phosphorylation is also brought about by a nuclear DNA-dependent protein kinase after the activated receptor binds to DNA (Weigel et al., 1992; Bagchi et al., 1992; Takimoto et al., 1992), suggesting that phosphorylation by different kinases at different times plays multiple roles in receptor function. A majority of phosphorylation sites contain serine/threonine-proline sequences (Denner et al., 1990a) and are localized to the amino terminus, a region important for transcriptional activation. Identified phosphorylation sites and their potential role in receptor function have been described in detail in several reviews (Weigel and Moore, 2007; Moore et al., 2007; Lange, 2004). Although the precise role for phosphorylation is not completely understood, functional correlation studies indicate that receptor phosphorylation is important in gene transactivation (Bagchi et al., 1992; Beck et al., 1992; Weigel et al., 1993). The contributions of phosphorylation to PR function have been addressed either by examining the modulatory effects of cellular protein kinase/phosphatase activities on receptor function or by analyzing the function of receptors containing mutated phosphorylation sites. The ability of protein kinase activators such as 8-bromo-cAMP, and phosphatase inhibitors such as okadaic acid and vanadate, to modulate ligand-independent activation of PRs lends credence to the importance of phosphorylation in gene activation (Denner et al., 1990b; Beck et al., 1992; Weigel et al., 1993). In addition, multiple kinases modulate individual phosphorylation sites of PRs in vitro (Davis, 1994; Zhang et al., 1994a, 1995, 1997). Site-directed mutagenesis studies have shown that multiple phosphorylation sites are important for maximal transcriptional activity of the PRs in a cell- and promoter-specific manner (Zhou et al., 1995; Bai et al., 1994; Bai and Weigel, 1996). Recent studies have also identified phosphorylation sites in the PR coactivators like SRC-1 (Rowan et al., 2000a), suggesting a greater degree of complexity in regulation of receptor activation by phosphorylation. Interestingly, cyclinA/cyclindependent kinase 2 has been demonstrated to phosphorylate not only PR, but also its coactivator SRC-1 (Moore et al., 2007). Some of the phosphorylation sites in the PR are conserved across the species, while others are unique to specific species. For example, while the chicken PR contains four serine-proline sites in the regions
common to both PR-A and PR-B isoforms, the human PR has several sites common to both PR-A and PR-B and several phosphorylation sites unique to the PR-B isoform (Denner et al., 1990a; Knotts et al., 2001). Taken together, these studies suggest that phosphorylation of the PR isoforms may regulate multiple PR-mediated functions. A current concept of progesterone action is schematically represented in Figure 2. 45.2.6
PR Isoforms
Multiple PR isoforms are produced from a single gene in various species (Schrader and O’Malley, 1972). The two major isoforms termed PR-A (79–94 kDa) and PR-B (101–120 kDa) are structurally related but functionally distinct proteins, produced by transcription from alternative estrogen-inducible promoters (Kastner et al., 1990b) and alternate translational sites within a single PR gene (Conneely et al., 1987). Both the isoforms have been identified in several species (Conneely et al., 1989a; Schneider et al., 1991; Lessey et al., 1983; Duffy et al., 1997), while only PR-B is expressed in the rabbit (Loosefelt et al., 1984; Savouret et al., 1991). The ratio of the two isoforms has been shown to differ among species, being equimolar in the human endometrium and chicken oviduct (Conneely et al., 1989b; Horwitz and Alexander, 1983) and predominantly of the smaller A-form in rodents (Ilenchuk and Walters, 1987). In addition, the ratio of PR-A to PR-B in reproductive tissues varies as a function of developmental and hormonal status. Studies in rhesus macaques indicate tissue-specific variations in the relative expression of the PR-A and PR-B proteins. While the expression of PR-A predominated in the estrogen-primed endometrium and pituitary, PR-B protein was highly expressed in the hypothalamus (Bethea and Wildmann, 1998). In human endometrium, the ratios of the PR-A and PR-B protein vary during the menstrual cycle (Mangal et al., 1997), with the PR-B isoform undetectable during the preovulatory phase, followed by increased PR-B expression and equimolar ratios of PR-A and PR-B proteins during the ovulatory phase. The ratio of the isoforms also varies in the mammary glands wherein PR-A is the predominant isoform. PR-A and PR-B isoforms differ only at the amino terminus, with PR-B containing an additional stretch of amino acids (128–165) located at the amino terminus of the receptor. This region encodes a transactivation function (AF-3) that is specific to the PR-B protein (Giangrande and McDonnell, 1999), and allows the binding of a subset of coactivators to PR-B
Mechanism of Progesterone Receptor Action in the Brain
1475
CBP/p300 TRAP p56 HSP p29 PR Ligand
Inactive receptor
P
P
PR
PR
Activated receptor
P
Pol II
SRCs PR
PR
Enhancer
GTFs
Promoter
Figure 2 Model for PR activation in target cells. Progestin receptor present in an inactive state undergoes conformational change and dimerization upon progesterone binding. Liganded receptor recruits SRC family of coactivators and other cofactors to effect local nucleosome disruption around the enhancer/promoter region through histone acetyltransferase activity. Subsequent recruitment of GTFs, RNA Pol II, and other basal factors, including coregulators (TRAP/DRIP), leads to the stabilization of a preinitiation complex and the formation of a transcriptionally active complex, leading to gene transcription, translation, and alteration of physiological function.
and not to PR-A. Both A and B proteins can bind to progesterone, undergo dimerization, and interact with the PRE, as well as general transcriptional machinery to regulate gene expression. When expressed in equimolar ratios in cells, both proteins can dimerize and bind DNA as A:A or B:B homodimers or A:B heterodimers. Interestingly, a third isoform, PR-C, has also been identified in the human (Wei et al., 1990). This protein of 60-kDa molecular weight is also an N-terminally truncated isoform that lacks AF-3 and AF-1 and the first zinc finger of DBD, and initiates at methionine 595, relative to PR-A and PR-B. PR-C isoform is thought to modulate the transcriptional activity of PR-A and PR-B (Wei et al., 1996). Tissue culture studies indicate that while PR-A and PR-B have similar DNA- and ligand-binding affinities, they have distinct transcriptional activities and are functionally different (Chalbous and Galtier, 1994; Tzukerman et al., 1994; Vegeto et al., 1993). PR-B has been demonstrated to function as a strong activator of transcription of several PR-dependent promoters in a variety of cell types in which PR-A is less inactive (Horwitz, 1992). While some genes are preferentially activated by PR-B (Tora et al., 1988; Kastner et al., 1990b; Vegeto et al., 1993), others are equally responsive to PR-A and PR-B. The isoforms also respond differently to progestin antagonists (Giangrande and McDonnell, 1999). Studies suggest differential subcellular localization of PR-A and PR-B in a living
cell line (Lim et al., 1999) with unoccupied PR-A being more nuclear in its localization than PR-B. Thus, the differential structure of the PR isoforms confers distinct tissue-specific responses to progesterone, through post-translational modifications, dimerization, and recruitment of cofactor proteins contributing to the differential transactivation properties of each isoform, and leading to the regulation of distinct subsets of progesterone-dependent target genes. Consistent with the distinct tissue- and promoter-specific activities of PR-A and PR-B in vitro, each individual isoform has been found to modulate distinct, but partially overlapping, subsets of reproductive functions by regulation of a diverse subset of target genes as seen in their phenotypic response in the uterus, the ovary, and the mammary gland (Conneely et al., 2003; Mulac-Jericevic and Conneely, 2004).
45.3 Ligand-Independent Activation of PRs Although the conventional model of PR activation assumes that a cognate ligand (progesterone) is required for the activation of the receptor (liganddependent activation), a number of studies in the past decade have shown that under certain circumstances, PRs can be activated by factors other than their cognate ligands. This activation is ligand
1476
Mechanism of Progesterone Receptor Action in the Brain
independent. The earliest observation from our laboratory revealed that progesterone-dependent, receptor-mediated transcription could be mimicked by 8-bromo-cAMP in the absence of progesterone (Denner et al., 1990a), and suggested that other pathways could also activate avian progesterone receptor. Pharmacologic stimulation of cAMP-dependent protein kinase (PKA) mimicked progesterone-dependent, receptor-mediated transcription in the absence of the hormone; this effect could be blocked by inhibition of PKA, suggesting that phosphorylation of the PR or other proteins in the transcription complex can modulate PR-mediated transcription (Denner et al., 1990a; Rowan et al., 2000a,b). The regulatory mechanisms governing a variety of cellular processes in target cells are dependent not only on the state of intracellular phosphorylation of the PR, but also on the equilibrium between protein kinase and phosphatase activities (Weigel, 1996). Epidermal growth factor, heregulin, phorbol myristate acetate, and insulin growth factors have also been shown to activate PRs by increasing the phosphorylation of PRs in vitro (Zhang et al., 1994b; Pierson-Mullany and Lange, 2004). Power et al. (1991a) demonstrated that a neurotransmitter, dopamine (DA), could activate chicken ovalbumin upstream promoter (COUP) transcription factor, a member of the steroid receptor superfamily in a ligand-independent manner. These were followed by other in vitro studies demonstrating ligand-independent activation of PRs by D1 receptor subtype DA agonists and protein phosphatase-1 (PP-1) inhibitor, okadaic acid, resulting in the translocation of the receptor from the cytoplasm to the nucleus (Power et al., 1991b; Philpott and Shaheed, 1996). Studies on luteinizing hormone-releasing hormone (LHRH) activation of PRs (Turgeon and Waring, 1994; Mani et al., 1995; Levine et al., 2001; GarridoGracia et al., 2006) also support the concept of ligandindependent activation in vivo. We have investigated the physiological relevance of ligand-independent mechanisms and the involvement of PR isoforms in the female sexual behavior and the results are described in Sections 45.6.3 and 45.5.3, respectively.
45.4 Cellular Function of Progesterone in the CNS 45.4.1 Reproductive Physiology and Behavior In most female mammals, ovarian steroid hormones coordinate reproductive physiology with behavior to
ensure successful fertilization. The sequential actions of estradiol secreted during the follicular phase of the estrous cycle followed by a surge of progesterone during the periovulatory phase result in a period of behavioral estrus that is linked to ovulation. During this period, the female mammal exhibits a variety of sexual behaviors, thereby maximizing the probability that the male will inseminate a female. For a detailed description of the sexual behaviors exhibited and the regulation of sexual responsiveness, the reader is referred to Chapter 2, Feminine Reproductive Behavior and Physiology in Rodents: Integration of Hormonal, Behavioral, and Environmental Influences. In this section the role of progesterone in the facilitation of female sexual behavior is discussed and the studies that have established the significance of PRs in progesterone action are reviewed. In gonadally intact female rodents, such as rats, mice, hamsters, and guinea pigs, the sequential release of ovarian estradiol and progesterone integrates the appearance of sexual behavior (heat and behavioral estrus) with ovulation (Beach, 1942; Boling and Blandau, 1939; Dempsey et al., 1936). Estradiol and progesterone regulate cellular functions in the central nervous system resulting in alterations in reproductive behavior (Blaustein and Olster, 1989; Pfaff et al., 1994; Blaustein and Mani, 2007). This behavior can be abolished by ovariectomy and restored by timed exogenous treatment with both estradiol and progesterone (Young, 1969) or by very high doses of estradiol alone (Lisk, 1962; Barfield and Chen, 1977; Rubin and Barfield, 1983a). Sequential treatment with estradiol and progesterone maximizes the probability that the female will assume lordosis posture, a primary behavioral component of female sexual behavior when mounted by a conspecific male (Pfaff et al., 1994, 2002; Feder, 1984). The sequential hormonal regimen also allows lower doses of each of these hormones to be used (Collins et al., 1938; Whalen, 1974), resulting in a more predictable onset and termination of the period of sexual behavior (Beach, 1942; Boling and Blandau, 1939; Wallen and Thonton, 1979). Thus, progesterone plays a significant role in the facilitation of sexual behavior in an estradiol-primed female rodent. In addition to the facilitatory effects, progesterone also has inhibitory effects on reproductive behavior in rodents. Following exposure to progesterone, the animals become refractory to further stimulation of sexual behavior by the administration of progesterone alone (Zucker, 1967) or by estradiol and progesterone (Zucker, 1968; Goy et al., 1966; Blaustein and Wade, 1977). This effect, referred to as the postestrous refractoriness
Mechanism of Progesterone Receptor Action in the Brain
(Morin, 1977), sequential inhibitory effect (Blaustein and Wade, 1977), or the biphasic effect (Zucker, 1968) of progesterone, is believed to limit the duration of behavioral estrus to the periovulatory phase of the estrous cycle. Studies suggest that progesterone is important for both initiation and termination of sexual behavior in female rodents, especially guinea pigs, hamsters, and female rats (Sodersten and Enoroth, 1978). 45.4.2
Species Variations
Progesterone has a significant role in the brain and hypophysis, both as a facilitator and an inhibitor of sexual behavior and gonadotropin release in several species, including the female rat (Everett, 1961; Caligaris et al., 1971; Barraclough, 1973; Goldman and Zarrow, 1973), guinea pig (Morin and Feder, 1974), and primates (Odell and Serdloff, 1968; Spies and Niswender, 1972; Karsch et al., 1973; Knobil, 1974). As described in Section 45.4.1, progesterone is required for the expression of sexual behavior in rodents, such as rats, mice, hamsters (with the exception of Phodus campbelli), and guinea pigs. However, ferrets (Baum et al., 1986), rhesus monkeys (Baum et al., 1977b), bonnet monkeys (MacLusky et al., 1980), goats (Phillips et al., 1946), and prairie voles (Richmond and Conaway, 1969) do not require progesterone for facilitation of sexual behavior. Progesterone terminates estrus in the ferrets (Marshall and Hammond, 1945) and reduces estradiol-induced sexual activity of the rhesus monkeys (Ball, 1941; Baum et al., 1977a). In reflex ovulators such as rabbits and cats, estradiol alone appears to determine the quality and duration of sexual responsiveness (Michael and Scott, 1957). However, the addition of progesterone to rabbits that are already in behavioral estrus enhances the quality of receptiveness (Sawyer and Everett, 1959). In the ewe, the sequence of estradiol and progesterone requirement in the induction of sexual behavior is reversed (Robinson, 1955). During breeding season, the sustained elevations in circulating progesterone during the luteal stage of the estrous cycle, followed by a sharp decline, appear to sensitize the brain to respond behaviorally to the preovulatory increase in plasma estradiol levels (Robinson, 1955). These observations have been confirmed in spayed ewes, which do not display reproductive behavior unless treatment with estrogen is preceded by progesterone (Robinson et al., 1956). In dogs, both estradiol and progesterone are required for the facilitation of sexual behavior. However, behavioral estrus persists for a week or more after ovulation (Lebouf, 1970).
1477
The occurrence of increased sexual receptiveness and behavior coinciding with ovulation in the subhuman species can be viewed as biologically purposeful in that it serves in the perpetuation of the species. Gonadal hormones are assumed to play a permissive role in sexual behavior of the human female. Exogenous progestins and endogenous progesterone have been shown to be associated with decreased sexual behavior in men and women (Barry and Ciccone, 1975). The loss of sexual desire and depression in some women using oral contraceptives is thought to be due to the progestin content in the preparation used (Grant and Davies, 1968). While assessment of hormone– behavior correlations in humans is fraught with methodological difficulties, social, cultural, and environmental variables have added to the complexity in studying over-all sexual behavior. Progesterone facilitation of sexual behavior also has been observed in nonmammalian vertebrates. In the male and the female ring doves, progesterone facilitates incubation behavior, and estrogen priming enhances this behavior (Martinez-Vargas et al., 1975). Similarily, progesterone facilitates female behavioral receptivity in estrogen-primed African clawed frog, Xenopus laevis and lizard Anolis carolinensis (McNicol and Crews, 1979). While scant data exist on the mechanisms of progesterone action in nonrodent species, studies in rats, mice, guinea pigs, and hamsters indicate that progesterone action on sexual behavior is mediated by neural PRs.
45.5 PRs in the CNS Studies in recent decades have provided increasing evidence that the mechanism of progesterone action in the brain is not distinctly different from that in the other reproductive tissues. The neural effects of progesterone on sexual behavior are believed to be predominantly genomic, requiring a two-step interaction with cognate, intracellular receptors in the hormonesensitive neurons (Blaustein and Olster, 1989; Blaustein and Mani, 2007). Estradiol acting via ERs in the brain alters the expression of a number of genes, including the PR gene (Simmerly, 1989; Alexander et al., 1989; Baldino et al., 1988; Romano et al., 1989a; Van Tol et al., 1988; Schumacher et al., 1990; Wilcox and Roberts, 1990). The time course for an estradiol-induced increase in PRmRNA levels parallels the increase in the PR levels, indicating the involvement of genomic mechanism in the action of estradiol (Romano et al., 1989b).
1478
Mechanism of Progesterone Receptor Action in the Brain
45.5.1 Spatial and Temporal Correlation between PR Induction and Behavior Spatial, temporal, and functional correlations suggest that estrogen-induced PRs, upon occupation by progesterone, function as transcriptional mediators and regulate transcription of target genes and affect the neural networks involved in the control of sexual behavior (Pfaff et al., 1994, 2002). The time course of activation and termination of sexual behavior parallels the estradiol-induced increase and decline in progestin-binding sites in the ventromedial hypothalamus (VMH) of the brain (Dempsey et al., 1936; Parsons et al., 1980). The increase in the concentration of unoccupied, hypothalamic-cytosol PRs, followed by the accumulation of occupied, nuclear PRs in response to a behaviorally effective dose of progesterone correlates well with the onset of sexual behavior in rodents, indicating that the hormone facilitation of sexual behavior is a two-step intracellular receptor-mediated mechanism (Blaustein and Feder, 1980; Brown et al., 1987b). Furthermore, the inhibition of progesteronefacilitated sexual behavior by the progesterone antagonist RU486 demonstrates that the hormone effects are mediated by PRs (Whalen et al., 1974). Other correlations between neural PRs and sexual behavior are evident in the studies of progesteroneinduced refractory period (Section 45.4.1) to further facilitation by progesterone in guinea pigs (Boling and Blandau, 1939; Meisel and Pfaff, 1984). During refractory period when the animals are hyposensitive to progesterone, the concentration of unoccupied hypothalamic PRs decreased (especially in the ventrolateral region of the VMH). Treatment with progesterone results in low levels of occupied nuclear PRs suggesting that the hyposensitivity (and the resulting heat termination) is due to the failure to accumulate adequate concentration of occupied nuclear PRs in response to progesterone (Blaustein and Feder, 1980). Intrahypothalamic applications of inhibitors to RNA and protein synthesis block estradiol and progesterone-facilitated lordosis, suggesting the involvement of PRs in the control of female reproductive behavior (Whalen et al., 1974; Meisel and Pfaff, 1985; Rainbow et al., 1982). A critical requirement of PRs in the regulation of progesteronefacilitated sexual behavior in female rats and mice has been demonstrated by studies using molecular approaches and gene targeting (Pollio et al., 1993; Ogawa et al., 1994; Mani et al., 1994b, 1996, 2006). These are discussed in Section 45.6.1 dealing with genomic mechanisms of progesterone action.
Most experiments in which intrahypothalamic antihormones, protein synthesis, and RNA synthesis inhibitors have been applied definitively identify the ventrolateral region of the VMH as the most responsive site of progesterone action in the expression of sexual behavior in rodents (Rubin and Barfield, 1983b). Results from a variety of studies using a wide array of techniques demonstrate the presence of estradiol-induced PRs in the anterior hypothalamus, VMH, ventrolateral hypothalamus, preoptic area (POA), arcuate nucleus, amygdala, and midbrain central gray – all sites at which hormones have well-documented effects on behavior and physiology (Pfaff et al., 1994; Meisel and Pfaff, 1984; Blaustein et al., 1988; DonCarlos et al., 1989, 1991; Blaustein, 1992; Moguilewsky and Raynaud, 1977; Powers, 1972; Ward et al., 1975). However, it should be noted that neural progesterone implants in areas like the midbrain reticular formation (Ross et al., 1971), habenula (Tennent et al., 1982), and tegmentum (Tennent et al., 1982; Luttge and Hughes, 1976), some of which appear to lack estradiol-induced PRs, also facilitate the expression of lordosis in estradiolprimed rats. While it is thought that these areas are part of the lordosis circuitry, it is not definitively known how the various regions are interconnected or how they respond to hormones (in the absence of PRs) to facilitate sexual behavior. It is tempting to postulate that some of these effects could be mediated by nongenomic mechanisms of steroid hormones, or interactions with neurotransmitters, neuropeptides, or other sensorimotor components of behavior, some of which are discussed in section 45.7.1.
45.5.2 Estrogen-Inducible versus Estrogen-Noninducible PRs Using the high-affinity progestin, 3H-R5020 as a radioligand, two anatomically distinct classes of progestin-binding sites were identified in the rat (MacLusky and McEwen, 1978) and in the guinea pig (Blaustein and Feder, 1979). One class of receptors are induced by estradiol and are present in the hypothalamus, POA and pituitary of the female rat (MacLusky and McEwen, 1980a), guinea pig (Blaustein and Feder, 1980), and the rabbit (ComachoArroyo et al., 1994). The other class of PRs is insensitive to estradiol priming, and is widely distributed in the cortex, hippocampus, amygdala, caudate-putamen, and cerebellum (Kato and Grouch, 1977). Significant concentrations of uninduced PRs
Mechanism of Progesterone Receptor Action in the Brain
have also been found in areas that contain inducible PRs (Parsons et al., 1982). The time course for estradiol-induced PR activation and decay parallels the increase and decrease of female sexual behavior (McEwen et al., 1982), suggesting that estrogensensitive PRs are involved in the facilitation of sexual behavior. The physiological role of estrogeninsensitive PRs is less clear. They could mediate progestin effects that are not dependent upon prior exposure to estradiol: for example, alterations of cortical electroencephalograph patterns (Arai and Gorski, 1968). Still another possibility could be their role in mediation of progestin effects on negative changes in mood and anxiety as seen during the late luteal phase of the menstrual cycle (when levels of progesterone are high) in some women (Halbreich et al., 1986). The insensitivity of PRs to estradiol regulation in the rat cerebral cortex (but not in the uterus) has been suggested to be due to the presence of an imperfect estrogen-responsive element near the ATG site of the rat PR-B isoform (Kato et al., 1994). Interestingly, recent studies on cortical PRs in rabbit suggest increased PR gene expression in response to estradiol (Comacho-Arroyo et al., 1996). While immunochemical studies by Gre´co et al. (2001) demonstrate the presence of PRs in the cerebral cortex, estradiol was unable to induce PRs. Whether tissue-specific factors, methods of detection, or species differences contribute to the estrogen insensitivity of regional PR gene expression remain to be determined in the future. 45.5.3
PR Isoforms in the Brain
Both the PR-A and PR-B isoforms, but not the PR-C isoform, have been identified in the rodent and primate brains. Kato et al. (1993, 1994) provided evidence for two distinct mRNA transcripts in the brain for PR-A and PR-B that were differentially regulated. Their analyses revealed that the ratio of the isoforms varied in different brain regions during development of the female rat, with a region-specific ontogenic expression of PR-B mRNA from birth to postnatal day 7 in the hypothalamus and POA, switching to the PR-A mRNA expression from postnatal days 8–12. The low PR-B expression in the cerebellum and cortex, however, remained unchanged throughout this period (Kato et al., 1993, 1994). The messages for both isoforms are differentially regulated by estradiol in the rat forebrain, with both PR-A and PR-B being induced in the hypothalamus and only the PR-B isoform in the pituitary (ComachoArroyo et al.,
1479
1998; Szabo et al., 2000). While mRNA transcripts for both PR-A and PR-B isoforms are induced by E2 and downregulated by P in the hypothalamus, only PR-B mRNA expression was hormonally modulated in the POA (ComachoArroyo et al., 1998). PR isoform transcripts also vary with the estrous cycle in a regionspecific manner, with PR-B being predominantly expressed on proestrus in the hypothalamus and on metestrus in the POA (Guerra-Araiza et al., 2001). Studies in rhesus macaques indicate a region-specific regulation of PR isoform proteins, with PR-B protein highly expressed in the hypothalamus (Bethea and Wildmann, 1998). In addition, sex differences are also evident in PR-A and PR-B mRNA expression patterns (Guerra-Araiza et al., 2001, 2002). We have recently examined the individual contributions of the neural PR-A and PR-B isoforms in mediating physiological responses to progesterone and the ligand-independent intracellular signaling pathways using mutant mice in which the expression of PR-A (PRAKO–/–) and PR-B (PRBKO–/–) isoforms had selectively been ablated (Mani et al., 2006). These studies established that neural PR-A isoform was critical in mediating progesterone-facilitated sexual receptive behavior in the female mice. Furthermore, the functional participation of PR-B isoform, was essential for the full complement of the behaviors. In contrast, the ligand-independent activation by DA involved both the isoforms suggesting that progesterone-dependent and ligandindependent pathways involve different isoforms and distinct intracellular signaling pathways to activate PRs (Figure 3). It is plausible that ligand-dependent and -independent activation of murine PRs could involve altered phosphorylation of distinct coactivators, several coactivators, or other phosphorylation sites on the PR isoforms and/or their coactivators (Mani, 2008).
45.6 Progestin Receptor Activation in the Brain: Relationship to Female Sexual Behavior As discussed in Section 45.5, PRs mediate progesterone action in the brain to facilitate female sexual behavior. Activation of PRs in the brain can be achieved by three distinct mechanisms: (1) classical or genomic; (2) nonclassical or nongenomic; and (3) ligand independent. Genomic and nongenomic mechanisms are both ligand (progesterone) dependent while ligand-independent mechanism refers to
1480
Mechanism of Progesterone Receptor Action in the Brain
100 PRAKO −/− PRBKO −/−
80
PR +/+ LQ
60
40
20
0 Veh
EB + P
EB
EB + SKF
Figure 3 Ligand-dependent and -independent activation of sexual receptivity in PR-isoform-specific null mutant mice (PRAKO–/– and PRBKO–/–). Progesterone-facilitated sexual receptivity is abolished in PRAKO–/– mice and is reduced in PRBKO–/– mice. Both PR-A and PR-B isoforms are essential in dopamine agonist (SKF)-facilitated lordosis. Adapted from Mani SK, Reyna AM, Chen JZ, Mulac-Jericevic B, and Conneely OM (2006) Differential response of progesterone receptor isoforms in hormone-dependent and -independent facilitation of female sexual receptivity. Molecular Endocrinology 20: 1322–1332.
the activation of the PRs by factors other than their cognate ligands. Studies in the recent years suggest that these mechanisms are not mutually exclusive, but interact with each other to achieve the behavioral endpoint. 45.6.1
Genomic Mechanisms
As discussed in Section 45.5.1, the primary regulatory action of progesterone in the facilitation of sexual behavior is believed to involve occupation and activation of PRs. This is evidenced by the temporal correlation between the display of sexual behavior and hypothalamic nuclear PR levels in guinea pigs and rats (Blaustein and Feder, 1979; McGinnis et al., 1981). Subsequent studies using protein and RNA synthesis inhibitors emphasized the role of transcription and protein synthesis associated with genomic action of progesterone in the facilitation of sexual behavior (Wallen et al., 1972; Rainbow et al., 1982). The direct evidence for involvement of neural PRs in progesterone facilitation of sexual behavior was provided by studies using the progesterone antagonist RU486 (Brown and Blaustein, 1984). The antagonist not only inhibited progesterone-facilitated sexual behavior in guinea pigs when administered 1 h prior to progesterone, but also appeared to be a competitive inhibitor of PR binding in the hypothalamic– preoptic area. Additional proof for an obligatory requirement for PRs in female sexual behavior came from studies in which antisense oligonucleotides to PR mRNA have
been administered into the brain. Both PR synthesis and progesterone-facilitated sexual behavior were inhibited by the infusion of antisense oligonucleotides to PRmRNA into the third cerebral ventricle (Mani et al., 1994b) or VMH (Pollio et al., 1993; Ogawa et al., 1994). Studies from our laboratory using transgenic mice carrying a null mutation for the PR gene (PR knockouts) provided definitive evidence for the role of intracellular PRs in rodent female sexual behavior (Mani et al., 1996). Female mice carrying a null mutation for the intracellular PR gene failed to display progesterone-facilitated lordosis response (Figure 4). The aggregate results of these studies support a classical genomic mode of activation for neural PRs in sexual behavior of rats and mice. 45.6.2
Nongenomic Mechanisms
While genomic effects have been assumed to be the primary pathway for hormone action in the brain, there are numerous reports of short-latency effects which suggest the involvement of nonclassical effects via putative cell-surface receptors and other mechanisms coupled to second-messenger-signaling cascades. Progesterone is believed to act through membraneassociated nonclassical receptors to activate Xenopus maturation and to rapidly stimulate calcium infux and chloride efflux to trigger acrosome reaction in human sperm (Ferrell, 1999; Blackmore, 1999). Rapid effects of progesterone have been demonstrated in their effects on the release of LHRH (Ramirez et al., 1990), DA and acetylcholine (Meiri, 1986), the
Mechanism of Progesterone Receptor Action in the Brain
1481
100 100 +/+ 80 80
−/−
60
LQ
LQ
60
40
40
20 20
(a)
0
EB
EB + P
EB + S + P
EB + AS + P
(b)
0 Vehicle
EB
EB + P
Figure 4 Requirement of nuclear progestin receptor in progesterone-facilitated female sexual behavior in rats and mice. (a) Administration of antisense oligonucleotides to progestin receptor into the third cerebral ventricle inhibits progesterone-facilitated lordosis response in female rats. (b) Mice with targeted deletion of nuclear progestin receptors (–/–) fail to exhibit progesterone-facilitated lordosis compared to their wild-type littermates (+/+). (a) Adapted from Mani SK, Blaustein JD, Allen JMc, Law SW, O’Malley BW, and Clark JH (1994b) Inhibition of rat sexual behavior by antisense oligonucleotides to the progesterone receptor. Endocrinology 135: 1409–1414. (b) Adapted from Mani SK, Allen JMC, Lydon JP, et al. (1996) Dopamine requires the unoccupied progesterone receptor to induce sexual behavior in mice. Molecular Endocrinology 10: 1728–1737.
release of excitatory amino acids (Smith et al., 1987), and changes in neuronal activity (Havens and Rose, 1988). These rapid effects, which are not blocked by protein-synthesis inhibitors, include direct actions on membrane receptor sites, ion channels, second-messenger systems and can also be mediated by metabolites of progesterone (Schumacher et al., 1999). Hormone effects are also mediated by binding to putative cell-surface membrane receptors (Ramirez et al., 1996) that gate ion channels (Gee et al., 1987; McEwen, 1994) and are coupled to certain second-messenger systems (McEwen, 1991; Kelly et al., 1999). Rapid effects of progesterone on facilitation of lordosis response in estrogen-primed female rats have been reported (Lisk, 1962; KubliGarfias and Whalen, 1977; Meyerson, 1972). Furthermore, metabolites of progesterone having low affinity for intracellular PRs were effective in facilitating lordosis response in estrogen-primed rats and mice (Glaser et al., 1985; Frye and Vongher, 1999). Functional interactions between membrane-mediated progesterone regulated pathways and intracellular PRs have been observed in the facilitation of sexual behavior in female hamsters (DeBold and Frye, 1994a,b), suggesting that both classical and nonclassical mechanisms act in concert rather than independently. A putative nongenomic cell-membrane progesterone-binding protein, that does not have significant homology to the known classical PR, was isolated
from porcine liver membranes (Meyer et al., 1996) and two putative PRs were cloned (Falkenstein et al., 1996, 1998; Gerdes et al., 1998). Homologous proteins were subsequently cloned in the rat (25-Dx; Selmin et al., 1996) and the human (Hpr6.6; Lo¨sel and Wehling, 2003). The 25-Dx protein is also referred to as the progesterone-membrane-receptor component 1 (PGMC1; Peluso et al., 2006). The expression of 25-Dx is upregulated by estradiol and downregulated by progesterone in the VMH of female rat (Krebs et al., 2000). 25-Dx appears to play a genderspecific role, since a sexually dimorphic pattern of expression was observed in the PR knockout mouse. The expression in the females varied with the genotype (PRKO > PR wild types) while it was not the case in the males. While the presence of 25-Dx in the VMN is suggestive of its role in female sexual behavior, its functional role has yet to be established. A novel membrane PR (mPR), unrelated to both the classical PR and 25-Dx, was identified in the teleost (Zhu et al., 2003a,b). This mPR is a transmembrane G-protein-coupled receptor and has been localized to the surface layer of the plasma membrane of the teleost oocytes and demonstrates a high-affinity saturable binding for P and its metabolites. It is related to a family of 13 genes from a variety of vertebrate species, including human, mouse, and pig. It consists of three subtypes (a, b, and g) which have distinct distribution in reproductive, neural, and
1482
Mechanism of Progesterone Receptor Action in the Brain
other tissues (Zhu et al., 2003a; Kazeto et al., 2005). The biological activity of mPR in mediating P effects in the brain and its relationship with the conventional PR remain to be determined. 45.6.3 Ligand-Independent Mechanism: An Alternate Mechanism of PR Action in the CNS Following our earlier in vitro observations on liganddependent activation of PRs (see Section 45.3), we demonstrated the physiological relevance of ligandindependent activation of PRs by DA in female sexual behavior in rats and mice. Intracerebroventricular (ICV) administration of apomorphine, a DA receptor stimulant, or the D1 agonist, SKF 38393, facilitated sexual receptive behavior (quantitated as lordosis quotient) in female rats mimicking the effects of progesterone (Mani et al., 1994a). The facilitatory effect of DA was specific to the D1 receptor subtype confirming and extending an earlier report in which DA agonists were infused into the hypothalamus and POAs (Foreman and Moss, 1979). Interestingly, similar to the facilitation of sexual behavior by progesterone, the facilitatory effect of DA agonist could also be blocked by ICV administration of PR antagonists, D1 receptor antagonist, or antisense oligonucleotides to PR mRNA (Mani et al., 1994a), suggesting that neural PRs were required for the DA activation of female sexual behavior. Importantly, the inability of PR knockout mice to exhibit D1 agonist-facilitated
sexual behavior, while their wild-type littermates responded to the agonist with lordosis (Mani et al., 1996), provides definitive evidence for the obligatory role of PRs as transcriptional mediators for a membrane receptor-dependent pathway initiated by DA, converging on PR, and resulting in sexual behavior (Figure 5). Our recent studies using antisense oligonucleotides to DA receptor subtypes indicate that DA-facilitated, PR-mediated behavioral effects occur via D1B (D5) subtype and not via the D1A subtype (Apostalakis et al., 1996; Mani et al., 2001). Reverse transcriptase-polymerase chain reaction technique (RT-PCR) also demonstrated the expression of D1B mRNA in the ventromedial nucleus of the rat (Zhou et al., 1999). In situ hybridization and immuno histochemcal studies confirm the co-expression of D1A/D1B and PRs in the medial POA, lateral ventromedial nucleus of the hypothalamus, and the arcuate nucleus of female rats (Blaustein et al., 1999; Lonstein and Blaustein, 2004). Collectively, the data suggest crosstalk between the progesterone and DA pathways in the mediation of female sexual behavior in rats and mice. The signaling pathways involved in the crosstalk are discussed in Section 45.7.3. Ligand-independent mechanisms for activation of PRs have been observed in other studies. Behaviorally relevant stimuli such as the vaginal–cervical stimulation (VCS) have been shown to activate neural PRs in the absence of progesterone (Auger et al., 1997, 2000a,b). DA regulation of PRs in developing
100
100
80
80
60
60
+/+
LQ
LQ
(a)
−/−
40
40
20
20
0 EB
EB + P
EB + SKF
EB + S + SKF EB + As + SKF
(b)
0 Vehicle
EB
EB + P
EB + SKF
Figure 5 Ligand-independent activation of progestin receptor by dopamine in female sexual behavior. (a) Antisense oligonucleotides to nuclear progestin receptor inhibit dopamine agonist-facilitated lordosis response in female rats indicating cross-talk between the two signaling systems. (b) Progestin receptor knockout mice fail to exhibit lordosis response upon administration of dopamine agonist (a) Adapted from Mani SK, Allen JMC, Clark JH, Blaustein JD, and O’Malley BW (1994a) Convergent pathways for steroid hormone- and neurotransmitter-induced rat sexual behavior. Science 265: 1246–1249. (b) Adapted from Mani SK, Allen JMC, Lydon JP, et al. (1996) Dopamine requires the unoccupied progesterone receptor to induce sexual behavior in mice. Molecular Endocrinology 10: 1728–1737.
Mechanism of Progesterone Receptor Action in the Brain
rat brain (Olesen et al., 2007) and dopaminergic activation of estrogen receptors in juvenile social-play behaviors has been reported (Olesen et al., 2005). Ligand-independent activation of PRs has also been observed in GnRH self-priming mechanisms in enriched gonadotropes (Turgeon and Waring, 1994; An et al., 2006). Recent studies using null mutant mice for PR suggest a role for PR in the production of GnRH surges (Chappell et al., 1997). Mutant mice in which PRs have been ablated demonstrated no estradiol-induced GnRH surges while their wildtype littermates responded to estradiol treatment with GnRH surges. A role for ligand-independent activation of PRs also has been observed in GnRH facilitation of sexual behavior. Antagonists to progesterone (Gonzalez-Mariscal et al., 1993; Moss et al., 1977) and antisense oligonucleotides to PR (Kato, 1994) inhibit GnRH facilitation of sexual behavior in female rats. Thus, PRs appear to play a generally important role in reproductive physiology in rats and mice. The signaling mechanisms by which DA or LHRH activate PRs appear to involve secondmessenger cascades that regulate phosphorylation of the receptors or some specific coactivator or other messenger system associated with the receptors (O’Malley et al., 1995; Turgeon and Waring, 1992; Chappell et al., 2000).
45.7 Mechanisms of Action of Progesterone and DA on Female Reproductive Behavior 45.7.1 Interactions between Progesterone and Neurotransmitters Accumulating evidence over the recent years suggests the involvement of neurotransmitters/neuropeptides in cellular processes by which steroid hormones influence sexual behavior in female rodents. While acetylcholine, norepinephrine, serotonin (acting via 5-HT2 receptor subtype), DA (D1), and neuropeptides LHRH, TRH, prolactin, oxytocin, substance P, and gamma-aminobutyric acid-A (GABAA) are considered facilitatory, serotonin (acting via receptor subtype 5-HT1A), DA (acting through receptor subtype D2), opioids, CRF, a-MSH, ACTH, b-endorphin, neuropeptide Y, cholecystokinin, and glutamate are considered inhibitory on sexual behavior (Pfaff et al., 1994; Kow et al., 1994). For a detailed discussion on the complex interactions among neurotransmitters, neuropeptides, and steroid hormones in bringing about the facilitation
1483
or inhibition of female reproductive behavior, the reader is referred to several reviews (Pfaff et al., 1994; Nock and Feder, 1981). Steroid hormones have been shown to have an effect on sexual behaviors by altering neurotransmitter biosynthesis and release (Nock and Feder, 1981; Luine et al., 1980), allosteric modulation of membrane receptors (Dohanich et al., 1982), changes in membrane-receptor densities, interactions with G-protein coupling, and subsequent intracellular signaling pathways (Etgen et al., 1992). This steroid hormone–neurotransmitter interaction is not unidirectional. Not only do steroids affect neurotransmission, but also changes in neurotransmission can alter steroid activity. The influence of neurotransmitters on steroid receptor modulation has been investigated using a wide array of techniques, including in vivo steroid uptake, in vivo steroid-binding assays, steroid receptor autoradiography, steroid receptor and neurotransmitter immunocytochemistry, and in situ hybridization. The role of neurotransmitters DA and norepinephrine in the regulation of ER and PR concentrations in the hypothalamus and pituitary gland in rat and guinea pig brain has been documented (Nock and Feder, 1981). A decrease in the concentration of estradiol-induced cytosolic PRs in the guinea pig hypothalamus was observed upon administration of a DA-b-hydroxylase inhibitor or antagonist of a1-adrenergic receptors (Nock and Feder, 1984; Thornton et al., 1986; Vincent and Feder, 1988). This decrease in cytosolic PRs was accompanied by an increase in nuclear PRs (Blaustein, 1985), suggesting that alterations in catecholaminergic activity may cause changes in hypothalamic PRs, similar to the steroid hormone actions in peripheral reproductive tissues (Gorski et al., 1968; Jensen and Jacobson, 1962; Tsai and O’Malley, 1994). To understand the regulation of steroid hormones by neurotransmitters and vice versa, intraneuronal co-localization of ER, PRs, and neurotransmitters has been investigated. Neuroanatomical studies of co-localization focused on the steroid-sensitive regions of the brain, especially the POA and the mediobasal hypothalamus (including the arcuate nucleus and ventromedial and ventrolateral areas), known to have effects on sexual behavior and physiology. Several neuropeptides and neurotransmitters have been found to be present in PR-containing neurons in these regions (Pfaff et al., 1994). Tyrosine hydroxylase immunoreactive neurons (suggestive of the presence of DA) have been shown to be in close association
1484
Mechanism of Progesterone Receptor Action in the Brain
with, and synapsing upon, PR-containing neurons in the hypothalamic/POA in the guinea pig (Blaustein and Turcotte, 1989; Brown et al., 1991), the rat (Horvath et al., 1992), and the monkey (Horvath et al., 1992). Although these studies do not prove neurotransmitter-related regulation of PRs, they are suggestive of modulatory interactions between PRs and neurotransmitters. In the past decade, our laboratory has investigated the interactions of the neurotransmitter DA and PRs in the modulation of female sexual behavior and the results are discussed in Sections 45.7.2 and 45.7.3. 45.7.2 DA Signaling and PR Pathway Convergence Protein phosphorylation is common to the pathways and molecular mechanisms through which neurotransmitters and steroid hormones produce their biological effects. The regulatory mechanisms governing a variety of cellular processes in target cells are not only dependent upon the state of intracellular phosphorylation of the receptor, but are also dependent on the dynamic balance between cellular protein kinases and phosphatases. This has been found to be the case in PR, wherein the equilibrium between transcriptionally active and inactive forms of the receptor is under the regulation of kinases and phosphatases (Denner et al., 1990b). Modulation of protein kinases and PPs in phosphorylation and signal-transduction mechanisms occur in the mammalian brain, a tissue having an abundance of kinases and phosphatases (Nairn et al., 1985; Shenolikar and Nairn, 1991). Neuronal phosphoproteins, such as neurotransmitters and cyclic nucleotides, are components of the signal-transduction pathway in the nervous system (Greengard, 1987, 2001; Greengard et al., 1998, 1999) and can be phosphorylated/dephosphorylated in response to extracellular stimuli; such dynamic covalent modification is evident in the modulation of the activity of PPs 1 and 2 (PP1 and PP2). Several studies have documented that DA signaling through D1 subclass of receptors induces increases in the levels of second-messenger cAMP and activates cAMP-dependent protein kinase (PKA) in the neostriatum. Increased PKA activity leads to the phosphorylation of the neuronal phosphoproteins, DA and cAMP-regulated phosphoprotein-32 (DARPP-32), and/or inhibitor-1 (I-1). The phosphoprotein I-1 is closely related structurally, enzymologically, and functionally to DARPP-32 (Shenolikar and Nairn, 1991). In its phosphorylated state, DARPP-32 and/or I-1, by inhibiting the activity of PP-1,
increases the state of phosphorylation of many substrate proteins, leading to induction of physiological responses. Thus, PR is one of the potential substrate proteins phosphorylated by DARPP-32. To determine whether DARPP-32 and/or Inh-1 could be the downstream mediators in the DA–PR interactions, we investigated their role in the facilitation of sexual behavior in female rats and mice. Antisense oligonucleotides to DARPP-32 administered ICV into the third cerebral ventricle inhibited D1 agonist- and progesterone-facilitated sexual receptivity in estradiol-primed female rats (Mani et al., 2000). D1 agonist- and progesterone-facilitated sexual receptivity also was inhibited in estradiolprimed female mice carrying a null mutation for DARPP-32 gene. However, I-1 null mutant mice exhibited no deleterious defects in D1 agonist- and progesterone-facilitated lordosis response compared to their wild-type littermates, revealing that involvement of the DARPP-32 pathway was specific (Figure 6). Also, increased immunoreactive phospho-DARPP-32 cells in the PR-containing areas of the rat hypothalamus have been observed after VCS, a somatosensory stimulation that increases expression of sexual behavior (Meredith et al., 1998). Similar to DA effects in the neostriatum, D1 agonist, as well as progesterone, significantly increased hypothalamic cAMP levels and PKA activities, and enhanced phosphorylation of DARPP-32 on Threonine-34. D1 agonist-induced increases were inhibited by the D1 subclass DA antagonist, SCH 23390, indicating that the increases were due to the effects of DA initiated at its membrane receptor. Progesterone-induced increases, however, were not inhibited by SCH 23390, suggesting that the observed increases were due to the direct effects of progesterone and not secondary to modulation of DA receptors by progesterone (Mani et al., 2000). Rp-cAMPS, a compound that blocks cAMP signaltransduction cascade by inhibiting PKA, inhibited D1 agonist- and progesterone-facilitated sexual receptivity in estradiol-primed female rats. While the observations indicate that DARPP-32 activation is an obligatory step in PR regulation of sexual receptivity, the sequence of events leading to the activation of PR (from DARPP-32 phosphorylation step) is complicated and remains to be completely defined. It is likely that the mechanisms include not only a direct, decreased dephosphorylation (activation) of PR, but also enhanced phosphorylation of PR-associated coactivators leading to rapid efficient transcriptional activation (Rowan et al., 2000a,b; De Mora and Brown, 2000; Lange, 2004, 2007).
Mechanism of Progesterone Receptor Action in the Brain
1485
100 C57 129
80
+/+ −/−
LQ
60
40
20
0 (a)
Veh
EB
EB + P
EB + SKF
100
C57 129
80
+/+ −/−
LQ
60
40
20
(b)
0 Veh
EB
EB + P
EB + SKF
Figure 6 DARPP-32 requirement for progesterone- and dopamine-facilitated lordosis response in female mice. (a) Null mutant mice for gene encoding DARPP-32 do not exhibit progesterone- and dopamine agonist-facilitated lordosis. (b) Progesterone- and dopamine agonist-facilitated lordosis response in inhibitor-1 knockout mice was not significantly different from their wild-type littermates. The parental strains, C57 and 129SvEv, responded to both progesterone and dopamine agonist treatments, indicating that the strain difference did not account for the behavioral phenotype observed. Adapted from Mani SK, Fienberg AA, O’Callaghan JP, et al. (2000) Requirement for DARPP-32 in progesterone-facilitated sexual receptivity in female rats and mice. Science 287: 1053–1056.
45.7.3 Multi-signal Pathway Reinforcement Steroid hormone–neurotransmitter interactions within the neural circuitry appear to involve substantial crosstalk between rapid membrane and slower
genomic signal-transduction pathways. Studies on ligand-independent activation of PRs by DA suggest that signaling pathways initiated by both DA and progesterone are not mutually exclusive and signals generated from the membrane-coupled receptors
1486
Mechanism of Progesterone Receptor Action in the Brain
converge and enhance gene expression regulated via classical intracellular steroid hormone receptors. Similar interactions between classical and nonclassical mechanisms have also been observed in the facilitation of sexual behavior in female hamsters (Frye and DeBold, 1993a,b). It is well known that steroid hormones, growth factors, and neurotransmitters activate intracellular cascades leading to the differential expression of genes and subsequent changes in behavior. Studies indicate that hypothalamic cAMP levels are increased on the evening of proestrus, concomitant with the exhibition of sexual behavior (Kimura et al., 1980). cAMP analogs and phosphodiesterase inhibitors have also been observed to facilitate sexual behavior in female rats (Kow et al., 1994; Beyer and Gonzalez-Mariscal, 1986; Whalen and Lauber, 1986). In addition, our observations of the involvement of a cAMP/PKA/DARPP-32 cascade in DA- and progesterone-facilitated sexual behaviors shed light on the probable convergence of events that occur in the PR-sensitive areas of the hypothalamus. A more encompassing model for DA–PR interactions in female sexual behavior is presented in the
schematic model (Figure 7) based on our observations and previously published findings. Progesterone initiates activation of PR by stimulating dual pathways. In a nonclassical manner, perhaps via the activation of membrane-bound PR or activation of G-protein-coupled membrane events, it stimulates the activation of a PKA-mediated-signaling cascade. This cascade results in activation of DARPP-32 and a decreased dephosphorylation of the PR and/or its associated coactivators. Simultaneously, progesterone effects are also mediated by the conventional intracellular nuclear receptors, involving allosteric activation of PR that promotes interactions with nuclear coactivators and gene transcription. Synergistic actions between the two pathways promote PR-mediated gene function. Simultaneously, DA released in the hypothalamus reinforces progesterone action through PR activation by ligand-independent mechanism as well as indirectly through the activation of DARPP-32. Thus, convergence and mutual reinforcement of both pathways present a powerful mechanism to achieve the neuroendocrine integration required for complex sexual behavior. Such convergence and reinforcement mechanisms could play
Progesterone Mating stimulus DA
Progesterone
cAMP cAMP P–PR PKA PKA MAPK
DARPP-32 ?
PR/coactivator (SRC-1,3/CBP)
*PR/* coactivator SRC-1,3/CBP
Physiological effects Figure 7 Model for the regulatory effects of dopamine and progesterone and their interdependence in lordosis. See text for discussion.
Mechanism of Progesterone Receptor Action in the Brain
an important role in integration of other neuronal responses to a multitude of signals that influence steroid effects on physiology and behavior. 45.7.4
Coactivators and PRs in the Brain
Effects of steroid hormone on female sexual behavior are mediated by estradiol-induced PRs in the brain. Local regulation of SRCs present in the brain could enhance these steroid receptor-mediated hormone effects, leading to region-specific changes in steroid sensitivity. SRC-1 expression has been reported throughout the brain, including hypothalamus, hippocampus, amygdala, cerebellum, and thalamus (Meijer et al., 2000; Ogawa et al., 2001; De Arrieta et al., 2000; Koibuchi et al., 2001; Misiti et al., 1998, 1999; Nishihara et al., 2004; Tetel et al., 2007). Using in situ hybridization, Meijer et al. (2000) demonstrated a differential expression profile and distribution of coactivators, SRC-1 and SRC-2, in the rat brain. The message for SRC-1 was expressed in many areas of the brain, while that for SRC-2 was found more prominently in the anterior pituitary. Splice variants of SRC-1, namely SRC-1a and SRC1e, were differentially expressed in several brain nuclei. SRC-1a mRNA was found to be expressed at a higher level in steroid-sensitive areas such as the arcuate nucleus, paraventricular and ventromedial nucleus of the hypothalamus, the locus ceruleus, trigeminal motor nucleus, and the anterior pituitary. Modest increases in SRC-1e mRNA expression were observed in the caudal nucleus accumbens, basolateral amygdala, and some thalamic nuclei. Whereas Meijer et al. (2000) were unable to detect the message for SRC-2 in regions other than in the pituitary, other studies have reported its presence in the brain (Xu et al., 1998) and in the cerebellum (De Arrieta et al., 2000). SRC-3 mRNA has been localized to the neurons in the hippocampus and the mitral and granular layers of the olfactory bulb (Xu et al., 2000). Immunocytochemical analyses have also revealed high levels of expression of yet another coactivator protein, CBP, in the hypothalamus, cortex, and cerebellum (Stromberg et al., 1999) and its potential role in brain development (Oike et al., 1999). CBP and SRC-1 proteins are expressed in estradiol-induced PR-containing neurons in the VMH and medial POA (Stromberg et al., 1999). Double label-fluorescent immunocytochemical studies indicate coexpression of coactivators and PRs in the VMH, medial POA, the arcuate nucleus, and medial central gray of the rat brain (Imtiaz et al., 2000; Tetel et al., 2007). While
1487
in vitro studies have clearly demonstrated functional cooperativity between SRC-1 and CBP in enhancing ER and PR transcription (Smith et al., 1996; Tetel et al., 1999; Rowan et al., 2000b), the investigation on their role in potentiating hormone action in brain and regulation of behavior is still in its infancy. Molenda et al. (2002) have demonstrated the functional importance of both coactivators, SRC-1 and CBP, in estrogen-induced PR expression and hormonedependent regulation of female reproductive behavior. Antisense oligonucleotides to SRC-1 or CBP independently infused into the VMH of ovariectomized, estrogen-primed female rats showed no differences in PR expression in the VMH compared with their scrambled oligonucleotide-treated controls. In contrast, antisense oligonucleotides to both SRC-1 and CBP administered together into the VMH decreased estradiol-induced PR expression, compared to the control-treated contralateral VMH. These observations suggest that functional interactions between the coactivators, SRC-1 and CBP, and ovarian steroid receptors could potentiate the hormone-dependent effects on gene expression in the VMH of the brain (Molenda et al., 2002; Tetel, 2000). Interestingly, while the antisense oligonucleotides to SRC-1 and CBP decreased the PR expression in the VMH, their effects on hormone-dependent female reproductive behavior were very distinct. Whereas the overall lordosis intensity of progesterone-facilitated lordosis was altered by the antisense oligonucleotides, they had no effect on the lordosis quotient. Although the above results are consistent with a permissive role for coactivators in the modulation of hormone-dependent sexual behavior, it is unclear whether the behavioral effects are due to a decrease in the coactivator function on the activity of ER and/or PR. It is also possible that the behavioral effects observed could be due to compensation by other coactivators in the VMH (Molenda et al., 2002). In this regard, it is worth noting that mice with targeted deletion of SRC-1 (Xu et al., 1998) expressed high levels of progesterone-facilitated lordosis similar to their wild-type littermates (SK Mani and B.W.O’Malley, unpublished observations). This lordosis response in the SRC-1 knockout mice could, however, be inhibited by infusion of antisense oligonucleotides to SRC-2 (but not SRC-3), suggesting a possible compensatory role for SRC-2 in the absence of SRC-1. The expression of PRs in the VMH was also reduced, suggesting the involvement of SRC-1 and SRC-2 in ER-mediated behavioral effects (Apostolakis et al., 2002) Thus, it is conceivable
1488
Mechanism of Progesterone Receptor Action in the Brain
that differential expression and distribution of the coactivators in the steroid-sensitive regions of the brain may underlie diversity and neuronal specificity of steroid-mediated signals in the brain as has been observed in other in vitro studies (Li et al., 2004). A requirement of SRC-1 in hormone-mediated sexual differentiation of the brain has also been demonstrated in rats (see Section 45.9). Since coactivators such as SRC-1 are also targets for multiple signaling pathways (Rowan et al., 2000a; Lonard and O’Malley, 2006, 2007), it is plausible that they could play a role in ligand-independent activation of PRs in brain and behavior. Future studies will determine the possible functions of the coactivators in the modulation of hormone-mediated events by various environmental stimuli in the CNS.
45.8 PRs and CNS Drug Actions In addition to their functional role in progesteroneand DA-facilitated signaling pathways, PRs also mediate the effects of psychotropic drugs such as cocaine and marijuana. Using antisense oligonucleotides to DA transporters and DA receptor antagonists, Apostolakis et al. (1996) demonstrated that cocaine facilitation of lordosis response in female rats appeared to involve the blockade of presynaptic DA transporters and stimulation of postsynaptic D5 subtype DA receptors. Furthermore, this response was PR dependent since antisense oligonucleotides to PR and progesterone antagonists could block this response. The data suggest that functional interactions between the cocaineinitiated DA-signaling system and PRs comprise the regulatory network, which allows cocaine to modulate reproductive behavior. We also demonstrated the involvement of PRs in tetrahydrocannabinol (THC) facilitation of female sexual behavior in rats (Mani et al., 2001). Using antisense oligonucleotides to D1 B (D5) subtype DA receptor and PRs, a remarkable crosstalk between THC, progesterone, and DAsignaling pathways was demonstrated in this study. THC effects on lordosis were mediated by CB1 receptors, and required functional progesterone and D1 B receptors. An increase in serum progesterone levels and PR activation has been observed in cocainetreated male rats (Walker et al., 2001; Wu et al., 2006). These studies suggest a high level of crosstalk and reinforcement among multiple intracellular signaling pathways for the mediation of hormonedependent behavior in mammals and suggest new avenues for intervention in drug abuse.
45.9 PRs and Male Sexual Behavior The effects of progesterone and the role of PRs on female sexual behavior have been described in detail in the preceding sections. However, the role of progesterone in male mammals in general, and male sexual behavior in particular, has remained uncertain. It is well known that females are more sensitive than males to progesterone effects on sexual behavior. Sex differences in the brain PR system have been attributed to the differential response to progestins (Moguilewsky and Raynaud, 1979). A smaller increase in cytosolic PRs has been reported in the hypothalamus, POA, and septum of estradiol-primed male guinea pigs compared to the females (Blaustein et al., 1980). Administration of progesterone to estradiolprimed male and female guinea pigs in the same study resulted in an increase in nuclear PRs proportional to cytosolic PR levels in both sexes. The study suggested that sex differences in progesterone facilitation of sexual behavior cannot be attributed to sex differences in brain PRs alone and could be due to a distinct set of neurons responding to estradiolpriming with induction of cytosolic PRs, in females and not in males (Kato, 1985). Ryer and Feder (1984b) reported that the sex difference in PR induction by estradiol in guinea pigs was restricted to hypothalamic nuclei and not to the POA. Using quantitative autoradiography, Brown et al. (1996) demonstrated PR binding in paraventricular thalamic nucleus; posterior part (PVP), medial preoptic nucleus (MPO), arcuate nucleus (ARC), ventrolateral hypothalamus (VLH), and ventrolateral hypothalamic nucleus (VLN) with higher levels of binding in PVP, MPO, and VLN of the female compared to the male estradiol-primed guinea pig. Sex differences in PR distribution have also been reported in rats. While differences in receptor levels have been found in the VMN and arcuate nucleus, no such variations were seen in the MPOA (Brown et al., 1987a; Lauber et al., 1991). Regional sex differences in the expression of estrogen receptors have been observed in both guinea pigs and rats, suggesting that sex differences in estrogen response could be due to lower sensitivity linked to receptor numbers (Lauber et al., 1991; Dufourny et al., 1997). However, regional sex differences in the estradiol-induced PRs are only partially concordant with the sex differences in estrogen-receptor levels (Dufourny et al., 1997). Thus, sex differences in progesterone response may involve not only the
Mechanism of Progesterone Receptor Action in the Brain
estrogen-receptor number, but also other factors that lead to enhanced transcriptional activation of PR gene or message stabilization or any of the other factors discussed in Section 45.2. Progesterone has been demonstrated to have both inhibitory and facilitatory effects on androgendependent sexual behavior in males. Pharmacological doses of progesterone have been shown to have inhibitory effects on androgen-dependent sexual behavior in guinea pigs (Connolly and Resko, 1989), mice (Erpino, 1973), quail (Bottoni et al., 1985), ring doves (Erickson et al., 1967), and nonhuman primates (Bonsall et al., 1990). Synthetic progestins (medroxyprogesterone acetate or cyproterone acetate) have been used routinely on human males to suppress the libido of felony sex offenders (Bradford, 1988; Lehne, 1988). In contrast, Debold et al. (1978) reported that progesterone administered simultaneously with testosterone propionate (TP) was more effective in maintaining sexual behavior of castrated hamsters. Progesterone was also found to increase mounting and intromission behavior in intact mice at lower doses, while higher doses resulted in inhibition of behavior (Erpino, 1973). Progesterone is known to stimulate male sexual behavior in whiptail lizard, Cnemidophorus uniparens (Grassman and Crews, 1986; Lindzey and Crews, 1988). A role for progesterone in androgen-dependent sexual behavior in male rats has been demonstrated by Witt et al. (1995). Enhanced mounting and intromission behaviors were seen in castrated, sexually naive male rats that received silastic implants of progesterone and testosterone. The facilitatory effects of progesterone were abolished upon administration of progesterone antagonist, suggesting a role for progesterone and PRs in male sexual behaviors. In studies using PR-null mutant male mice (PRKO), Phelps et al. (1999) demonstrated that naive null mutant males exhibited reduced mount frequencies compared to their wild-type littermates on the first. Furthermore, PR deletion affected its modulation by DA (Wooley et al., 2006). Thus, progesterone acting via its intracellular receptors and crosstalk appears to also facilitate sexual behaviors in male mice.
45.10 PRs in Development Steroid hormones, estrogens and androgens acting via their intracellular receptors, have been known to influence sexual differentiation of the neural substrates for sexual behaviors during the perinatal
1489
period (Phoenix et al., 1959; Goy et al., 1965). During pregnancy, the developing fetuses are exposed to high levels of maternal hormones, including progesterone in utero (Sanyal, 1978). Exposure of the central nervous system to progestational steroids during early life could have effects on brain development and function. PRs have been identified in the rat brain of both males and females close to the day of birth; PRs increased in a region-specific manner, despite low levels of progesterone in the blood (Dohler and Wuttke, 1974). A rapid increase of cerebral cortical PRs in the females (compared to males) was seen on postnatal days 7–10, while stable PR levels were maintained in the hypothalamus– preoptic area (HPOA; Kato et al., 1984). Both cytosol and nuclear PRs have been detected at birth, and increase slightly during postnatal days 1–7 in both HPOA and the cerebral cortex (MacLusky and McEwen, 1980b; Kato and Onouchi, 1983) of both sexes. While cytosol PRs in the HPOA remained at the same level throughout the postnatal period, those in cerebral cortex increased rapidly, reaching a maximal level at day 10 and decreased to low levels by postnatal days 21–28 in female rats. The development of cytosol PRs in male rats followed a pattern similar to the females. However, the HPOA cytosol PR concentration in males was higher than in females at days 14–21 (Kato et al., 1984). The nuclear PRs followed the same trend in the HPOA and cerebral cortex. Thus, the development pattern of functional steroid receptors in neonatal rats suggests that the animals are capable of responding to behavioral effects of steroid homones (or neurotransmitters) early in life. Neonatal male and female rats exhibit cytosol PR induction along with estradiol-induced female sexual behavior (Williams, 1987; Williams and Blaustein, 1988). Furthermore, progesterone was capable of facilitating the expression of sexual behavior in a 6day-old male, but not in female rats. These data suggest that endogenous testicular hormones prime the male brain to respond to exogenous progesterone with sexual behavior, and at some point during development, the males, and not the females, become less responsive to progesterone. Estradiol-induced HPOA and the cortical PRs, although low in concentration, do not differ between males and females in neonatal female guinea pigs (Reyer and Feder, 1984a). These animals do not exhibit steroid-induced female sexual behavior at this stage of development indicating that deficits in the PR function in the brain of immature guinea pigs may be responsible for the absence of steroid-hormone-induced female
1490
Mechanism of Progesterone Receptor Action in the Brain
sexual behavior (Goy et al., 1967; Olster and Blaustein, 1998). PRs in the neonatal mouse brain also reveal sex differences between the male and female preoptic/central hypothalamic regions on days 8–12 (Shughrue et al., 1991b). Greater PR numbers were found in the female medial preoptic nucleus, but not in arcuate and VMH nuclei. The female cortex also contained more PRs than the male cortex (Shughrue et al., 1991a). Wagner et al. (1999) demonstrated the presence of greater levels of PRs in the medial preoptic nucleus of fetal (embryonic day 19–postnatal day 10) and neonatal male rats. Since plasma testosterone levels surge in male fetuses on days 18–19 of gestation (Weisz and Ward, 1980), the findings suggest the possibility of a regulatory role for fetal testosterone (by aromatization to estradiol) in PR expression in the males. The authors postulate that maternal progestins acting via the PRs in the medial preoptic nucleus could influence testosterone effects on sexual differentiation of the brain. Neonatal treatment of females with PR antagonist, RU486, attenuated masculinizing effects of testosterone (Quadros et al., 2002). In addition, there appears to be a developmental switch in the ventromedial nucleus, but not in the medial preoptic nucleus when PR expression changes from estradiol independent to estradiol dependent. The studies suggest that the regulation of PR expression by estradiol is dependent on age, sex, and brain region, suggesting that PR may play a critical, but specific role in the normal development of these reproductively important brain areas (Quadros and Wagner, 2008; Wagner, 2008; Roselli et al., 2006). It would be interesting to examine whether the effects are mediated by PRs per se or by regulation of their coactivators. A role for coactivator SRC-1 and CREB-binding protein in the hormone-dependent development of male sexual behavior and brain morphology has been reported (Auger et al., 2000b; Auger, 2003). Future studies could potentially include progestins, in addition to androgens and estrogens, as modulators of sexual differentiation of the brain.
Progesterone’s effects on inhibition of aggressive behavior have been demonstrated in hamsters (Fraile et al., 1987a; Meisel et al., 1990). While some studies indicate that PRs in the VMH mediate these effects in the hamster brain (Fraile et al., 1987b), others suggest that the progesterone metabolite, pregnenolone, acting through nongenomic mechanisms could mediate these observed effects (Haug et al., 1980). Similar nongenomic mechanisms have been suggested in the soporific, anxiolytic, and anticonvulsant effects of progesterone in rodents and humans (Selye, 1941; Arafat et al., 1993; Bitran et al., 1995; Brot et al., 1997; Frye and Bayon, 1999; Frye and Scalise, 2000). These effects of progesterone and pregnenolone are thought to be mediated by GABAA receptor, suggesting a level of crosstalk between the two systems. Similar crosstalk between progestins and other systems have also been observed in the analgesic and anticonvulsant effects of progesterone. A distinctive role for progesterone in the development and expression of seizure activity has also been implicated in women with catamenial epilepsy (Herkes et al., 1993; Herzog, 1995). In female rats, progesterone increases amygdaloid after-discharge thresholds, slowing down the development of seizures kindled from hippocampus or amygdala with marked anticonvulsive effects (Edwards et al., 1999). The analgesic properties of progesterone metabolites are mediated by mechanisms involving calcium channels, GABAA receptors, and endogenous opioid systems (Kavaliers and Wiebe, 1987). Interestingly, progesterone metabolites have been shown to facilitate learning and memory in rats and mice (Vallee et al., 1997; Flood et al., 1992). A role for PRs in the cognitive function has also been suggested (Wagner, 2008). While the mechanisms involved in these memory-enhancing properties are not known at the current time, it is assumed that the progestins reinforce the neurotransmitter systems in the brain. The interactions of the progesterone metabolites with the other signaling systems and the role of intracellular PRs, if any, in mediating these behavioral effects remain to be explored.
45.11 PRs and Other Behavioral Effects
45.12 Summary and Conclusions
Besides the facilitatory and inhibitory effects on male and female sexual behavior, progesterone and its metabolites mediate a variety of behavioral effects in rodents. Intracellular PRs in the forebrain are thought to mediate the inhibitory effects of progesterone on rat maternal behavior (Numan et al., 1999).
It is becoming abundantly clear that progesterone modulation of the brain and behavior is more complex than previously envisioned. Although there are a few exceptions, most of the studies on progesterone effects on female reproductive behavior are consistent with the ligand-dependent, transcriptional
Mechanism of Progesterone Receptor Action in the Brain
regulation of intracellular PRs in distinct regions of the brain. In a few instances, rapid, nonclassical pathways involving putative cell-surface receptors or ion channels mediate progesterone effects in the central nervous system. Recent developments in the field indicate that in addition to the conventional receptormediated genomic mechanism, PRs are activated in a ligand-independent manner by afferent input by neurotransmitters to facilitate female sexual behavior. In this chapter, we reviewed evidence that PR modulates female sexual behavior by a mechanism that involves convergence of two distinct pathways initiated by DA and progesterone. The cellular and molecular mechanisms involved in the convergence indicate substantial crosstalk between signal-transduction cascades initiated at the membrane leading to activation of intracellular PRs and gene expression. Neuronal kinases and phosphatases play a predominant role in regulation of the equilibrium between transcriptionally active and inactive states of PRs and their coregulators. Furthermore, coregulators appear to act as master regulators to mediate functional interactions of receptors with diverse classes of transcription factors, integrating gene networks. The activation of multiple kinases and coregulators by diverse environmental stimuli could potentially alter the activity of steroid hormone receptors, thereby modulating hormone action in vivo. A strong recurring theme emerging from this review is the importance of crosstalk between various afferent signals which impinge on steroid receptors. Mutual interdependence and convergence of signaling pathways appear to be a reinforcing mechanism to achieve the neuroendocrine integration required for the complex reproductive behavior. We foresee that future studies will yield significant insights into the complexity of signaling as this active field endeavors toward an integrative model of PR action in brain and behavior.
Acknowledgments The authors thank the support of US Public Health Service grants MH57442, MH63954 (S.K.M.), and HD74095 (B.W.O) for some of the research performed in our laboratories described in this chapter.
References Alexander MJ, Dobner PR, Miller MA, Bullock BP, Dorsa DM, and Leeman SE (1989) Estrogen induces neurotensin,
1491
neuromedin N messenger RNA in preoptic nucleus essential for pre-ovulatory surge of luteinizing hormone in the rat. Endocrinology 125: 2111–2117. Allen WM, Butenandt A, Corner GW, and Slotta KH (1935) Nomenclature of corpus luteum hormone. Science 82: 153–154. Allen WM and Corner GW (1929) Physiology of the corpus luteum. III. Normal growth and implantation of embryos after early ablation of ovaries, under the influence of extracts of corpus luteum. American Journal of Physiology 88: 340–345. Allen WM and Corner GW (1930) Physiology of the corpus luteum. VII. Maintenance of pregnancy in rabbit after early castration, by corpus luteal extracts. Proceedings of the Society for Experimental Biology and Medicine 27: 403–410. An B-S, Selva DM, Hammond GL, Rivero-Muller A, Rahman N, and Leung PCK (2006) Steroid receptor coactivator-3 is required for progesterone receptor transactivation of target genes in response to gonadotropin-releasing hormone treatment of pituitary cells. Journal of Biological Chemistry 281: 20817–20824. Anzick SL, Kononen J, Walker RL, et al. (1997) AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science 277: 965–968. Apostolakis EM, Garai J, Clark JH, and O’Malley BW (1996) In vivo regulation of central nervous system progesterone receptors: Cocaine induces steroid-dependent behavior through dopamine transporter modulation of D5 receptors in rats. Molecular Endocrinology 10: 1595–1604. Apostalakis EM, Garai J, Fox C, Smith CL, Watson SJ, Clark JH, and O’Malley BW (1996) Dopaminergic regulation of progesterone receptors: Brain D5 dopamine receptors mediate induction of lordosis by D1-like agonists in rats. Journal of Neuroscience 16: 4823–4834. Apostolakis EM, Ramamurthy M, Zhou D, Ona˜te S, and O’Malley BW (2002) Acute disruption of select steroid coactivators prevents reproductive behavior in rats and unmasks genetic adaptation in knockout mice. Molecular Endocrinology 16: 1511–1523. Arafat ES, Hargrove JT, Maxson WS, Desiderio DM, Wentz AC, and Andersen RN (1993) Sedative and hypnotic effects of oral administration of micronized progesterone may be mediated through its metabolites. American Journal of Obstetrics and Gynecology 159: 1203–1209. Arai Y and Gorski RA (1968) Effect of anti-estrogen on steroid-induced sexual receptivity in ovariectomized rats. Physiology and Behavior 3: 351–353. Auboeuf D, Dowhan DH, Kang YK, et al. (2004) Differential recruitment of nuclear receptor coactivators may determine alternative RNA splice site choice in target genes. Proceedings of the National Academy of Sciences of the United States of America 101: 2270–2274. Auger AP (2003) Sex differences in the developing brain: Crossroads in the phosphorylation of cAMP response element binding protein. Journal of Neuroendocrinology 15: 622–627. Auger AP, LaRiccia LM, Moffatt CA, and Blaustein JD (2000a) Progesterone, but not progesterone-independent activation of progestin receptors by a mating stimulus, rapidly decreases progestin receptor immunoreactivity in female rat brain. Hormones and Behavior 37: 135–144. Auger AP, Moffat CA, and Blaustein JD (1997) Progesterone-independent activation of rat brain progestin receptors by reproductive stimuli. Endocrinology 138: 511–514. Auger AP, Tetel MJ, and McCarthy MM (2000b) Steroid receptor activator-1 (SRC-1) mediates the development of
1492
Mechanism of Progesterone Receptor Action in the Brain
sex-specific brain morphology and behavior. Proceedings of the National Academy of Sciences of the United States of America 97: 7551–7555. Bagchi MK, Tsai SY, Tsai M-J, and O’Malley BW (1990) Identification of a functional intermediate in receptor activation in progesterone-dependent cell-free transcription. Nature 345: 547–550. Bagchi MK, Tsai SY, Tsai M-J, and O’Malley BW (1992) Ligand and DNA-dependent phosphorylation of human progesterone receptor in vitro. Proceedings of the National Academy of Sciences of the United States of America 89: 2664–2668. Bai W, Tullos S, and Weigel NL (1994) Phosphorylation of Ser530 facilitates hormone-dependent transcriptional activation of the chicken progesterone receptor. Molecular Endocrinology 8: 1465–1473. Bai W and Weigel NL (1995) Phosphorylation and steroid hormone action. Vitamins and Hormones 51: 289–313. Bai W and Weigel NL (1996) Phosphorylation of Ser211 in the chicken progesterone receptor modulates its transcriptional activity. Journal of Biological Chemistry 271: 12801–12806. Baldino F, Jr., Fitzpatrick-McElligot S, O’Kane TM, and Gozes I (1988) Hormonal regulation of somatostatin mRNA. Synapse 2: 317–325. Ball J (1941) Effect of progesterone upon sexual excitability in the female monkey. Psychological Bulletin 38: 533. Baniahmed A, Leng X, Burris TP, Tsai SY, Tsai M-J, and O’Malley BW (1995) The t4 activation domain of thyroid hormone receptor is required for release of a putative corepressor(s) necessary for transcriptional silencing. Molecular and Cellular Biology 15: 76–86. Bannister AJ and Kouzarides T (1996) The CBP co-activator is a histone acetyltransferase. Nature 384: 641–643. Barfield RJ and Chen JJ (1977) Activation of estrous behavior in ovariectomized rats by intracerebral implants of estradiol benzoate. Endocrinology 101: 1716–1725. Barraclough CA (1973) Sex steroid regulation of reproductive neuroendocrine processes. In: Greep RO and Astwood EB (eds.) Handbook of Physiology, Section 7, Endocrinology, vol. 2, pp. 29–56. Washington, DC: American Physiological Society. Barry DJ and Ciccone JR (1975) Use of depo-proversa in the treatment of aggressive sexual offenders: Preliminary report of three cases. Bulletin of the Academy of Psychiatry and the Law 3: 179–184. Baum MJ, Everitt BJ, Herbert J, and Keverne EB (1977a) Hormonal basis of proceptivity and receptivity in female primates. Archives of Sexual Behavior 6: 173–192. Baum MJ, Gerlach JL, Krey LC, and McEwen BS (1986) Biochemical and autoradiographic analysis of estrogen-inducible progestin receptors in female ferret brain and pituitary: Correlations with effects of progesterone on sexual behavior and gonadotropin-releasing hormone-stimulated secretion of luteinizing hormone. Brain Research 368: 296–309. Baum MJ, Keverne EB, Everitt BJ, Herbert J, and de Greef (1977b) Effects of progesterone and estradiol on sexual attractivity of female rhesus monkeys. Physiology and Behavior 18: 659–670. Beach FA (1942) Importance of progesterone to induction of sexual receptivity in spayed female rats. Proceedings of the Society for Experimental Biology and Medicine 51: 369–371. Beato M (1989) Gene regulation by steroid hormones. Cell 56: 335–344. Beato M (1991) Transcriptional control by nuclear receptors. FASEB Journal 5: 2044–2051.
Beato M, Arnemann G, Chalepakis ES, and Willman T (1987) Gene regulation by steroid hormones. Journal of Steroid Biochemistry 27: 9–14. Beato M, Herrlich P, and Schutz G (1995) Steroid hormone receptors: Many actors in search of a plot. Cell 83: 851–857. Beck CA, Weigel NL, and Edwards DP (1992) Effects of hormone and cellular modulators of protein phosphorylation on transcriptional activity, DNA binding, and phosphorylation of human progesterone receptors. Molecular Endocrinology 6: 607–620. Becquel MT, Kumar V, Stricker C, Chambon P, and Gronemeyer H (1989) The contribution of the N- and C-terminal regions of steroid receptors to activation of transcription is both receptor and cell-specific. Nucleic Acids Research 17: 2581–2595. Bethea CL and Wildmann AA (1998) Differential expression of progestin receptor isoforms in the hypothalamus, pituitary, and endometrium of rhesus macaques. Endocrinology 139: 677–687. Beyer C and Gonzalez-Mariscal G (1986) Elevation in hypothalamic cAMP as a common factor in the facilitation of lordosis in rodents: A working hypothesis. Annals of the New York Academy of Sciences 474: 270–281. Bitran D, Shiekh M, and McLeod M (1995) Anxiolytic effect of progesterone is mediated by the neurosteroid allopregnanolone at brain GABAA receptors. Journal of Neuroendocrinology 7: 171–177. Blackmore PE (1999) Extragenomic actions of progesterone in human sperm and progesterone metabolites in human platelets. Steroids 64: 149–156. Blaustein JD (1985) Noradrenergic inhibitors cause accumulation of nuclear progestin receptors in guinea pig hypothalamus. Brain Research 325: 89–98. Blaustein JD (1992) Cytoplasmic estrogen receptors in rat brain: Immunocytochemical evidence using three antibodies with distinct epitopes. Endocrinology 131: 1336–1342. Blaustein JD and Feder HH (1979) Cytoplasmic progestin receptors in guinea pig brain: Characteristics and relationship to induction of sexual behavior. Brain Research 169: 481–497. Blaustein JD and Feder HH (1980) Nuclear progestin receptors in guinea pig brain measured by an in vitro exchange assay after hormonal treatments after lordosis. Endocrinology 106: 1061–1069. Blaustein JD, King JC, Toft DO, and Turcotte J (1988) Immunocytochemical localization of estrogen-induced progestin receptors in guinea pig. Brain Research 474: 1–15. Blaustein JD, Lubbers LS, Meredith J, and Wade GN (1999) Dopamine receptor subtypes in progestin receptor (PR)-rich regions of the hypothalamus in female rats. Society of Neuroscience, Abstract 748.14: 1881. Blaustein JD and Mani SK (2007) Feminine sexual behavior from the neuroendocrine and molecular neurobiological perspectives. In: Blaustein JD and Lajtha A (eds.) Hand Book of Neurochemistry and Molecular Neurobiology: Behavioral Neurochemistry and Neuroendocrinology, vol. 21, pp. 95–150. Berlin: Springer. Blaustein JD and Olster DH (1989) Gonadal steroid hormone receptors and social behaviors. In: Balthazart J (ed.) Advances in Comparative and Environmental Physiology, vol. 3, pp. 31–104. Berlin: Springer. Blaustein JD, Reyer HI, and Feder HH (1980) A sex difference in the progestin receptor system of guinea pig brain. Neuroendocrinology 31: 403–409. Blaustein JD and Turcotte J (1989) A small population of tyrosine hydroxylase immunoreactive neurons in the guinea
Mechanism of Progesterone Receptor Action in the Brain
pig arcuate nucleus contain progesterone receptor immunoreactivity. Journal of Neuroendocrinology 1: 333–338. Blaustein JD and Wade GN (1977) Sequential inhibition of sexual behavior by progesterone in female rats: Comparison with a synthetic antiestrogen. Journal of Comparative and Physiological Psychology 91: 752–760. Boling JL and Blandau RJ (1939) The estrogen-progesterone induction of mating responses in the spayed female rat. Endocrinology 25: 359–364. Bonsall R, Rees H, and Michael R (1990) Uptake of medroxyprogesterone acetate. Endocrinology 93: 417–422. Boonyaratanakornkit V, Melvin V, Prendrgast P, et al. (1998) High-mobility group chromatin proteins 1 and 2 functionally interact with steroid hormone receptors to enhance their DNA binding in vitro and transcriptional activity in mammalian cells. Molecular and Cellular Biology 18: 4471–4487. Bottoni L, Lucini V, and Massa R (1985) Effect of progesterone on the sexual behavior of the malejapanese quail. General and Comparative Endocrinology 57: 345–351. Bradford JMW (1988) Treatment of sexual offenders with cyproterone acetate. In: Sitsen JAM (ed.) Handbook of Sexology, vol. 6, pp. 526–536. New York: Elsevier Science. Brown TJ and Blaustein JD (1984) Inhibition of sexual behavior in female guinea pigs by a progestin receptor antagonist. Brain Research 301: 343–349. Brown TJ, Blaustein JD, Hochberg RB, and MacLusky NJ (1991) Estrogen receptor binding in regions of the rat hypothalamus and preoptic area after inhibition of dopamine,b-hydroxylase. Brain Research 549: 260–267. Brown TJ, Clark AS, and MacLusky NJ (1987a) Regional sex differences in progestin receptor induction in the rat hypothalamus: Effect of various doses of estradiol benzoate. Journal of Neuroscience 7: 2529–2536. Brown TJ, Gagnon M, Sharma M, and MacLusky NJ (1996) Sex differences in estrogen receptor and progestin receptor induction in the guinea pig hypothalamus and preoptic area. Brain Research 725: 37–48. Brown TJ, Moore MJ, and Blaustein JD (1987b) Maintenance of progesterone-facilitated sexual behavior in female rats requires continued hypothalamic protein synthesis and nuclear progestin receptor occupation. Endocrinology 121: 298–304. Brot MD, Akwa Y, Purdy RH, Koob GE, and Britton KT (1997) The anxiolytic-Iike effects of the neurosteroid allopregnanolone: Interactions with GABA(A) receptors. European Journal of Pharmacology 325: 1–7. Burrows H (1949) Biological Actions of Sex Hormones. Cambridge: Cambridge University Press. Caligaris L, Astrada JJ, and Taleisnik S (1971) Biphasic effect of progesterone on the release of gonadotropin in rats. Endocrinology 89: 331–337. Carson-Jurica MA, Schrader WT, and O’Malley BW (1990) Steroid receptor family: Structure and functions. Endocrine Reviews 11: 201–220. Chakravarti D, Mamorte VJ, Nelson MC, et al. (1996) Role of CBP/p300 in nuclear receptor signaling. Nature 383: 99–103. Chalbous D and Galtier F (1994) Differential effect of forms A and B of human progesterone receptor on estradiol-dependent transcription. Journal of Biological Chemistry 269: 23007–23012. Chappell PE, Lee J, and Levine JE (2000) Stimulation of gonadotropin-releasing hormone surges by estrogen. II. Role of cyclic adenosine 30 50 -monophosphate. Endocrinology 141: 1486–1492. Chappell PE and Levine JE (2000) Stimulation of gonadotropin-releasing hormone surges by estrogen. I.
1493
Role of hypothalamic progesterone receptors. Endocrinology 141: 1477–1485. Chappell PE, Lydon JP, Conneely OM, O’Malley BW, and Levine JE (1997) Endocrine defects in mice carrying a null mutation for the progesterone receptor gene. Endocrinology 138: 4147–4152. Chauchereau A, Cohen-Solal K, Jollivet A, Bailly A, and Milgrom E (1994) Phosphorylation sites in ligand-induced and ligand independent activation of progesterone receptor. Biochemistry 33: 13295–13303. Chen H, Lin RJ, Schiltz RL, et al. (1997) Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300. Cell 90: 569–580. Collins VJ, Boling JI, Dempsey EW, and Young WC (1938) Quantitative studies of experimentally induced sexual receptivity in the spayed guinea pig. Endocrinology 23: 181–196. Comacho-Arroyo I, Guerra-Araiza C, and Cerbon MA (1998) Progesterone receptor isoforms are differentially regulated by sex steroids in the rat forebrain. NeuroReport 9: 3993–3996. Comacho-Arroyo I, Pasapera AM, and Cerbon MA (1996) Regulation of progesterone receptor gene expression by sex steroid hormones in the hypothalamus and the cerebral cortex of the rabbit. Neuroscience Letters 214: 25–28. Comacho-Arroyo I, Pasapera AM, Perez-Palacios G, and Cerbon MA (1994) Intracellular progesterone receptors are differentially regulated by sex steroid hormones in the hypothalamus and the cerebral cortex of the rabbit. Journal of Steroid Biochemistry and Molecular Biology 50: 299–303. Conneely OM, Kettelberger DM, Tsai M-J, and O’Malley BW (1989a) Promoter specific activating domains of the chicken progesterone receptor. In: Roy AK and Clark JH (eds.) Gene Regulation by Steroid Hormones, vol. 4, pp. 220–231. New York: Springer. Conneely OM, Kettelberger DM, Tsai M-J, Schrader WT, and O’Malley BW (1989b) The chicken progesterone receptor A and B isoforms are products of an alternate translation initiation event. Journal of Biological Chemistry 264: 14062–14064. Conneely OM, Maxwell BL, Toft DO, Schrader WT, and O’Malley BW (1987) The A and B forms of the chicken progesterone receptor arise by alternate initiation of translation of a unique mRNA. Biochemical and Biophysical Research Communications 149: 493–501. Conneely OM, Mulac-Jericevic B, and Lydon JP (2003) Progesterone-dependent regulation of female reproductive activity by two distinct progesterone receptor isoforms. Steroids 68: 771–778. Connolly PB and Resko JA (1989) Progestins affect reproductive behavior and androgen receptor dynamics in male guinea pig brain. Brain Research 305: 312–315. Corner GW (1929) Physiology of the corpus luteum. II. Production of special uterine reaction (progestational proliferation) by extracts of corpus luteum. American Journal of Physiology 88: 326–332. Davis RJ (1994) MAPKs: New JNK expands the group. Trends in Biochemical Science 19: 470–473. De Arrieta M, Koibuchi N, and Chin WW (2000) Coactivator and corepressor gene expression in rat cerebellum during postnatal development and the effect of altered thyroid status. Endocrinology 141: 1693–1698. DeBold JF and Frye CA (1994a) Genomic and nongenomic actions of progesterone in the control of female hamster sexual behavior. Hormones and Behavior 28: 445–453.
1494
Mechanism of Progesterone Receptor Action in the Brain
DeBold JF and Frye CA (1994b) Progesterone and neural mechanisms of hamster sexual behavior. Psychoneuroendocrinology 19: 563–579. DeBold JF, Morris JL, and Clemens LG (1978) The inhibitory actions of progesterone: Effects on male and female sexual behavior of the hamster. Hormones and Behavior 11: 28–41. DeMora JF and Brown M (2000) AIB1 is a conduit for kinase-mediated growth factor signaling to estrogen receptor. Molecular and Cellular Biology 20: 5041–5047. Dempsey EW, Hertz R, and Young WC (1936) The experimental induction of oestrus (sexual receptivity) in the normal and ovariectomized guinea pig. American Journal of Physiology 116: 201–209. Denner LA, Schrader WT, O’Malley BW, and Weigel NL (1990a) Hormonal regulation in identification of chicken progesterone receptor phosphorylation sites. Journal of Biological Chemistry 265: 16548–16555. Denner LA, Weigel NL, Maxwell BL, Schrader WT, and O’Malley BW (1990b) Regulation of progesterone receptor-mediated transcription by phosphorylation. Science 250: 1740–1743. Denner LA, Weigel NL, Schrader WT, and O’Malley BW (1989) Hormone-dependent regulation of chicken progesterone receptor deoxyribonucleic acid binding and phosphorylation. Endocrinology 125: 3051–3058. Ding XF, Anderson CM, Ma H, Hong H, Uht RM, Kushner PJ, and Stallcup MR (1998) Nuclear receptor binding sites of coactivators glucocorticoid receptor interacting protein 1 (GRIP1) and steroid receptor coactivator 1 (SRC-1): Multiple motifs with different binding specificities. Molecular Endocrinology 12: 302–313. Dohanich GP, Witcher JA, Weaver D, and Clemens L (1982) Alteration of muscarinic binding in specific brain areas following estrogen treatment. Brain Research 241: 347–350. Dohler DD and Wuttke W (1974) Serum LH, FSH, prolactin and progesterone from birth to puberty in female and male rats. Endocrinology 94: 1003–1008. DonCarlos LL, Greene GL, and Morrell JL (1989) Estrogen plus progesterone increases progesterone immunoreactivity in the brain of ovariectomized guinea pigs. Neuroendocrinology 50: 613–623. DonCarlos LL, Monroy E, and Morrell JL (1991) Distribution of estrogen receptor-immunoreactive cells in the forebrain of the female guinea pig. Journal of Comparative Neurology 305: 591–612. Duffy DM, Wells TR, Haluska GJ, and Stouffer RL (1997) The ratio of progesterone receptor isoforms changes in the monkey corpus luteum during the luteal phase of the menstrual cycle. Biology of Reproduction 57: 693–699. Dufourny L, Warembourg M, and Jolivet A (1997) Sex differences in estradiol-induced progestin receptor immunoreactivity in the guinea pig preoptic area and hypothalamus. Neuroscience Letters 223: 109–112. Eckner R, Ewen ME, Newsome D, Gerdes M, DeCaprio JA, Lawrence JB, and Livingston DM (1994) Molecular cloning and functional analysis of the adenovirus EIA-associated 300-kD protein (p300) reveals a protein with properties of a transcriptional adaptor. Genes and Development 8: 869–884. Edwards HE, Burnham WM, Mendonca A, Bowlby DA, and MacLusky NJ (1999) Steroid hormones affect limbic afterdischarge thresholds and kindling rates in adult female rats. Brain Research 838: 136–150. Erickson CJ, Bruder RH, Komisaruk BR, and Lehrman DS (1967) Selective inhibition by progesterone of androgeninduced behavior in male ring doves (Streptopelia risoria). Endocrinology 81: 9–44. Erpino MJ (1973) Temporary inhibiton by progesterone of sexual behavior in intact male mice. Hormones and Behavior 4: 335–339.
Etgen AM, Ungar S, and Petitti N (1992) Estradiol and progestreone modulation of norepinephrine neurotransmission: Implications for the regulation of female reproductive behavior. Journal of Neuroendocrinology 4: 255–271. Evans RM (1988) The steroid and thyroid hormone receptor superfamily. Science 240: 885–889. Everett JW (1961) The mammalian female reproductive cycle and its controlling mechanisms. In: Young WC (ed.) Sex and Internal Secretions, 3rd edn., pp. 497–555. Baltimore, MD: Williams & Wilkins. Falkenstein E, Meyer C, Eisen C, Scriba PC, and Wehling M (1996) Full length cDNA sequence of a progesterone membrane-binding protein from porcine vascular smooth muscle cells. Biochemical and Biophysical Research Communications 229: 86–89. Falkenstein E, Schmeiding K, Lange A, et al. (1998) Localization of a putative progesterone membrane binding protein in porcine hepatocytes. Cell and Molecular Biology 44: 571–578. Feder HH (1984) Hormones and sexual behavior. Annual Review of Physiology 35: 165–200. Fernandez-Valdivia R, Mukherjee A, Mulac-Jericevic B, Conneely OM, DeMayo FJ, Amato P, and Lydon JP (2005) Revealing progesterone’s role in uterine and mammary gland biology: Insights from the mouse. Seminars in Reproductive Medicine 23: 22–37. Ferrell JE (1999) Xenopus oocyte maturation: New lessons from a good egg. BioEssays 21: 833–842. Flood JF, Morley JE, and Roberts E (1992) Memory-enhancing effects in male mice of pregnenolone and steroids metabolically derived from it. Proceedings of the National Academy of Sciences of the United States of America 89: 1567–1571. Foreman MM and Moss RL (1979) Role of hypothalamic dopaminergic receptors in the control of lordosis behavior in the female rat. Physiology and Behavior 22: 282–289. Fraenkel L and Cohn F (1901) Experimentalle unrerschung uber den einfluss des corpus luteum auf die insertion des eies. Anatomischer Anzeiger 20: 294. Fraile LG, McEwen BS, and Pfaff DW (1987a) Progesterone inhibition of aggressive behaviors in hamsters. Physiology and Behavior 39: 225–229. Fraile LG, Pfaff DW, and McEwen BS (1987b) Progestin receptors with and without estrogen induction in male and female hamster brain. Neuroendocrinology 45: 487–491. Freedman LP (1992) Anatomy of steroid receptor zinc finger region. Endocrine Reviews 13: 129–145. Fronsdal K, Engedal N, Slagsvold T, and Scaatcioglu F (1998) CREB binding protein is a nuclear coactivator for androgen receptor and mediates cross-talk with AP-1. Journal of Biological Chemistry 273: 31853–31859. Frye CA and Bayon LE (1999) Cyclic withdrawal from endogenous and exogenous progesterone increases kainic acid and perforant pathway induced seizures. Pharmacology Biochemistry and Behavior 62: 315–321. Frye CA and DeBold JF (1993a) 3 Alpha-OH-DHP and 5 alphaTHDOC implants to the ventral tegmental area facilitate sexual receptivity in hamsters after progesterone priming to the ventral medial hypothalamus. Brain Research 612: 130–137. Frye CA and DeBold JF (1993b) P-3-BSA, but not P-11-BSA, implants in the VTA rapidly facilitate receptivity in hamsters after progesterone priming to the VMH. Behavioural Brain Research 53: 167–175. Frye CA and Scalise TJ (2000) Antiseizure effects of progesterone and 3a, 5a-THP in kainic acid and perforant pathway models of epilepsy. Psychoneuroendocrinology 25: 407–420.
Mechanism of Progesterone Receptor Action in the Brain
Frye CA and Vongher JM (1999) Progestins’ rapid facilitation of lordosis when applied to the ventral tegmentum corresponds to efficacy at enhancing GABA(A) receptor activity. Journal of Neuroendocrinology 11: 829–837. Garrido-Gracia JC, Bellido C, Aguilar R, and Sa´nchezCriado JE (2006) Protein kinase C cross-talk with gonadotrope progesterone receptor is involved in GnRHinduced LH secretion. Journal of Physiology and Biochemistry 62: 35–42. Gee KW, Chang WC, Briton RE, and McEwen BS (1987) GABA-dependent modulation of the Cl-ionophore by steroids in rat brain. European Journal of Pharmacology 136: 419–423. Gehin M, Mark M, Dennefeld C, Dierich A, Gronemeyer H, and Chambon P (2002) The function of TIF2/GRIP1 in mouse reproduction is distinct from those of SRC-1 and p/CIP. Molecular and Cellular Biology 22: 5923–5937. Gerdes D, Wehling M, Leube B, and Falkenstein E (1998) Cloning and tissue expression of two putative steroid membrane receptors. Biological Chemistry 379: 907–911. Giangrande PH and McDonnell DP (1999) The A and B isoforms of the human progesterone receptor: Two functionally different transcription factors encoded by a single gene. Recent Progress in Hormone Research 54: 291–313. Glaser JH, Etgen AM, and Barfield RJ (1985) Intrahypothalamic effects of progestin agonists on estrous behavior and progestin receptor binding. Physiology and Behavior 34: 871–878. Goldman BD and Zarrow MX (1973) The physiology of progestins. In: Greep RO and Astwood EB (eds.) Handbook of Physiology, Section 7, Endocrinology, vol. 2, part 1, pp. 547–576. Washington, DC: American Physiological Society. Gonzalez-Mariscal G, Melo AI, and Beyer C (1993) Progesterone, but not LHRH or prostaglandin-E (2) induces sequential inhibition of lordosis to various lordogenic agents. Neuroendocrinology 57: 940–945. Gorski J, Toft D, Shyamala G, and Notides A (1968) Hormone receptors: Studies on the interaction of estrogen with the uterus. Recent Progress in Hormone Research 24: 45–80. Goy RW, Phoenix CH, and Meidinger R (1967) Postnatal development of sensitivity to estrogen and androgen in male, female and pseudo hermaphroditic guinea pigs. Anatomical Record 157: 87–96. Goy RW, Phoenix CH, and Young WC (1965) Role of developing testis in differentiation of neural tissues mediating mating behavior. Journal of Comparative and Physiological 59: 176–182. Goy RW, Phoenix CH, and Young WC (1966) Inhibitory action in the corpus luteum on the hormonal induction of estrous behavior in the guinea pig. General and Comparative Endocrinology 6: 267–275. Grant ECG and Davies JP (1968) Effect of oral contraceptives on depressive mood changes and on endometrial monoamine oxidase and phosphates. British Medical Journal 3: 777–780. Grassman M and Crews D (1986) Progestrone induction of pseudo copulatory behavior and stimulus-response complementarity in all-female lizard species. Hormones and Behavior 20: 327–335. Gre´co B, Allegreto EA, Tetel MJ, and Blaustein JD (2001) Coexpression of ERb with ERa and progestin receptor proteins in the female rat forebrain: Effects of estradiol treatment. Endocrinology 142: 5172–5181. Greengard P (1987) Neuronal phosphoproteins: Mediators of signal transduction. Molecular Neurobiology 1: 81–119. Greengard P (2001) The neurobiology of slow synaptic transmission. Science 294: 1024–1030.
1495
Greengard P, Allen PB, and Nairn AC (1999) Beyond the dopamine receptor: The DARPP-32/protein phosphatase1 cascade. Neuron 23: 435–447. Greengard P, Nairn AC, Girault JA, et al. (1998) The DARPP-32/ protein phosphatase-1 cascade: A model for signal integration. Brain Research – Brain Research Reviews 26: 274–284. Guerra-Araiza C, Coyoy-Salgado A, and Camacho-Arroyo I (2002) Sex differences in the regulation of progesterone receptor isoforms expression in the rat brain. Brain Research Bulletin 59: 105–109. Guerra-Araiza C, Reyna-Neyra A, Salazar AM, Cerbon MA, Morimoto S, and Camacho-Arroyo I (2001) Progesterone receptor isoforms expression in the prepuberal and adult male rat brain. Brain Research Bulletin 54: 13–17. Guichon-Mantel A, Loosefelt H, Lescop P, Sar S, Atger M, Perrot-Applanat M, and Milgrom E (1989) Mechanisms of nuclear localization of the progesterone receptor: Evidence for interaction between monomers. Cell 57: 1147–1154. Guyon JR, Narlikar GJ, Sullivan EK, and Kingston RE (2001) Stability of a human SWI-SNF remodeled nucleosomal array. Molecular Cellular Biology 21: 1132–1144. Halbreich U, Endicott J, Goldstein S, and Nee J (1986) Premenstrual changes and changes in gonadal hormones. Acta Psychiatrica Scandinavica 74: 576–586. Hammond J and Marshall FHA (1925) Reproduction in the Rabbit. London: Oliver and Boyd. Hanstein B, Eckner R, Direnzo J, et al. (1996) P300 is a component of an estrogen receptor coactivator complex. Proceedings of the National Academy of Sciences of the United States of America 93: 11540–11545. Haug M, Simler S, Kim L, and Mandel P (1980) Studies on the involvement of GABA in the aggression directed by groups of intact or gonadectomized male and female mice towards lactating intruders. Pharmacology, Biochemistry, and Behavior 12: 189–193. Havens MD and Rose JD (1988) Estrogen-dependent and estrogen-independent effects of progesterone on the electrophysiological excitability of dorsal midbrain neurons in golden hamsters. Neuroendocrinology 48: 120–129. Heery DM, Kalkhoven E, Hoaare S, and Parker MG (1997) A signature motif in transcriptional coactivators mediates binding to nuclear receptors. Nature 387: 733–736. Herkes GK, Eadie MJ, Sharbrough F, and Moyer T (1993) Patterns of seizure occurrence in catamenial epilepsy. Epilepsy Research 15: 47–52. Herzog AG (1995) Progesterone therapy in women with complex partial and secondary generalized seizures. Neurology 45: 1660–1662. Hisaw FL, Greep RO, and Ferold HL (1937) Effects of progesterone on female genital tract after castration atrophy. Proceedings of the Society for Experimental Biology and Medicine 36: 840–842. Hoffman A, Oelgeschla¨ger T, and Roeder RG (1997) Considerations of transcriptional control mechanisms: Do TFIID-core promoter complexes recapitulate nucleosomelike functions? Proceedings of the National Academy of Sciences of the United States of America 94: 8928–8935. Hong H, Kohli K, Garabedian MJ, and Stallcup MR (1997) GRIPI, a transcriptional coactivator for the AF-2 transactivation domain of steroid, thyroid, retinoid and vitamin D receptors. Molecular and Cellular Biology 17: 2735–2744. Hong H, Kohli K, Trivedi A, Johnson DL, and Stallcup MR (1996) GRIP1, a novel mouse protein that serves as a transcriptional coactivator in yeast for the hormone binding domains of steroid receptors. Proceedings of the National Academy of Sciences of the United States of America 93: 4948–4952.
1496
Mechanism of Progesterone Receptor Action in the Brain
Horvath TL, Naftolin F, and Leranth C (1992) b-Endorphin innervation of dopamine neurons in the rat hypothalamus: A light and electron microscope double immunostaining study. Endocrinology 131: 1547–1555. Horwitz KB (1992) The molecular biology of RU486. Is there a role for antiprogestins in the treatment of breast cancer? Endocrine Reviews 13: 146–163. Horwitz KB and Alexander PS (1983) In situ photo-linked nuclear progesterone receptors of human breast cancer cells: Subunit molecular weights after transformation and translocation. Endocrinology 113: 2195–2201. Ilenchuk TT and Walters MR (1987) Rat uterine progesterone receptor analyzed by [3H]R5020 photoaffinity labeling: Evidence that the A and B subunits are not equimolar. Endocrinology 120: 1449–1456. Imbalzano AN, Kwon H, Green MR, and Kingston RE (1994) Facilitated of TATA-binding protein to nucleosomal DNA. Nature 370: 481–485. Imhof MO and McDonnell DP (1996) Yeast RSP5 and its human homolog hRPFI potentiate hormone-dependent activation of transcription by human progesterone and glucocorticoid receptors. Molecular and Cellular Biology 16: 2594–2605. Imtiaz UM, Wessels SR, and Tetel MJ (2000) Hypothalamic neurons express steroid receptors and nuclear receptor coactivators in female rats. Society of Neuroscience, Abstract 77.6: 211. Jackson TA, Richer JK, Bain DL, Takimoto GS, and Horwitz KB (1997) The partial agonist activity of antagonist-occupied steroid receptors is controlled by a novel hinge domain-binding coactivator L7/SPA and the corepressors N-CoR or SMRT. Molecular Endocrinology 11: 693–705. Jensen EV and Jacobson HI (1962) Basic guides to mechanism of estrogen action. Recent Progress in Hormone Research 18: 387–414. Jensen EV, Suzuki T, Kawasima T, Stumpf WE, Jungblut PW, and de Sombre ER (1968) A two-step mechanism for the interaction of estradiol with rat uterus. Proceedings of the National Academy of Sciences of the United States of America 59: 632–638. Jones PL, Veenstra GJ, Wade PJ, et al. (1998) Methylated DNA and MeCP2 recruit histone acetylase to repress transcription. Nature Genetics 19: 187–191. Kamei Y, Xu L, Heinzel T, et al. (1996) A CBP integrator complex mediates transcriptional activation and AP-l inhibition by nuclear receptors. Cell 85: 403–414. Karsch FJ, Weick RF, Hotchkiss J, Dierschke DJ, and Knobil E (1973) An analysis of the negative feedback control of gonadotropin secretion utilizing chronic implantation of ovarian steroids in ovariectomized rhesus monkeys. Endocrinology 93: 478–486. Kastner P, Krust A, Mendelsohn C, et al. (1990a) Murine isoforms of retinoic acid receptor gamma with specific patterns of expression. Proceedings of the National Academy of Sciences of the United States of America 87: 2700–2704. Kastner P, Krust A, Turcotte U, Strupp L, Tora H, Gronemeyer H, and Chambon P (1990b) Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO Journal 9: 1603–1614. Kato J (1985) Progesterone receptors in the brain physis. In: Ganten D and Pfaff DW (eds.) Current Topics in Neuroendocrinology, pp. 31–81. Berlin: Springer. Kato J (1994) Basic and clinical studies on sex hormone receptors. Nippon Sanka Fujinka Gakkai Zasshi 46: 647–658. Kato J and Grouch T (1977) Specific progesterone receptors in the hypothalamus and anterior hypophysis of rat. Endocrinology 101: 912–928.
Kato J, Hirata S, Nozawa A, and Yamada-Mouri N (1993) The ontogeny of gene expression of progestin receptors in the female rat brain. Journal of Steroid Biochemistry and Molecular Biology 47: 173–182. Kato J, Hirata S, Nozawa A, and Yamada-Mouri N (1994) Gene expression of progesterone receptor isoforms in the rat brain. Hormones and Behavior 28: 454–463. Kato J and Onouchi T (1983) Progestin receptors in female rat brain and hypophysis in the development from fetal to postnatal stages. Endocrinology 113: 29–36. Kato J, Onouchi T, Okinaga S, and Takamatsu M (1984) The ontogeny of cytosol and nuclear progestin receptors in male rat brain and its male–female differences. Journal of Steroid Biochemistry 20: 147–152. Kavaliers M and Wiebe JP (1987) Analgesic effects of the progesterone metabolite, 3-alpha-hydroxy-5-alphapregnan20-one, and possible modes of action in mice. Brain Research 415: 393–398. Kazeto Y, Goto-Kazeto R, Thomas P, and Trant JM (2005) Molecular characterization of three forms of putative membrane-bound progestin receptors and their tissue-distribution in channel catfish. Ictalurus punctatus Journal of Molecular Endocrinology 34: 781–791. Kelly MJ, Lagrange AH, Wagner EJ, and Ronnekleiv O (1999) Rapid effects of estrogens to modulate G protein coupled receptors via activation of protein kinase A and protein kinase C pathways. Steroids 64: 64–75. Kim HJ, Kim JH, and Lee JW (1998) Steroid receptor coactivator-l interacts with serum response factor and coactivates serum response element-mediated transactivations. Journal of Biological Chemistry 273: 28564–28567. Kimura F, Kawakami M, Nakano H, and McCann SM (1980) Changes in adenosine 30 , 50 -monophosphate and guanosine 30 , 50 -mono phosphate concentrations in the pituitary and hypothalamus during the rat estrous cycle and effects of administration of sodium pentobarbital in proestrus. Endocrinology 106: 631–635. Kishino T, Lalande M, and Wagstaff J (1997) UBE3NE6-AP mutations cause Angelman syndrome. Nature Genetics 15: 70–73. Klein-Hitpass L, Tsai SY, Weigel NL, et al. (1990) The progesterone receptor stimulates cell-free transcription by enhancing the formation of a stable preinitiation complex. Cell 60: 247–257. Knobil E (1974) On the control of gonadotropin secretion in rhesus monkey. Progress in Hormone Research 30: 1–36. Knotts TA, Orkiszewski RS, Cook RG, Edwards DP, and Weigel NL (2001) Identification of a phosphorylation site in the hinge region of the human progesterone receptor and additional amino-terminal phosphorylation sites. Journal of Biological Chemistry 276: 8475–8483. Koibuchi N, Yamaoka S, and Chin WW (2001) Effect of altered thyroid status on neurotrophin gene expression during postnatal development of the mouse cerebellum. Thyroid 11: 205–210. Kow L, Mobbs CV, and Pfaff DW (1994) Role of secondmessenger systems and neuronal activity in the regulation of lordosis by neurotransmitters, neuropeptides, and estrogen: A review. Neuroscience and Biobehavioral Reviews 18: 1–18. Kraus WL and Kadonaga JT (1998) P300 and estrogen receptor cooperatively activate transcription via differential enhancement of initiation and reinitiation. Genes and Development 12: 331–345. Krebs CJ, Jarvis ED, Chan J, Lydon JP, Ogawa S, and Pfaff DW (2000) A membrane-associated progesterone-binding protein, 25-Dx, is regulated by progesterone in brain regions involved in female reproductive behaviors. Proceedings of
Mechanism of Progesterone Receptor Action in the Brain
the National Academy of Sciences of the United States of America 97: 12816–12821. Kubli-Garfias C and Whalen RE (1977) Induction of lordosis behavior in female rats by intravenous administration of progesterone. Hormones and Behavior 93: 5925–5930. Lange CA (2004) Making sense of cross-talk between steroid hormone receptors and intracellular signaling pathways: Who will have the last word? Molecular Endocrinology 18: 269–278. Lange CA (2007) Integration of progesterone receptor action with rapid signaling events in breast cancer models. Journal of Steroid Biochemistry and Molecular Biology 108: 203–212. Lanz RB, McKenna NJ, Ona˜te SA, et al. (1999) A steroid receptor coactivator, SRA, functions as an RNA and is present in an SRC-I complex. Cell 97: 17–27. Lanz RB, Razani B, Goldberg AD, and O’Malley BW (2002) Distinct RNA motifs are important for coactivation of steroid hormone receptors by steroid receptor RNA activator (SRA). Proceedings of the National Academy of Sciences of the United States of America 99: 16081–16086. Lanz RB and Rusconi S (1994) A conserved carboxyterminal sub domain is important for ligand interpretation and transactivation by nuclear receptors. Endocrinology 135: 2183–2195. Lauber AH, Romano GJ, and Pfaff DW (1991) Sex difference in estradiol regulation of progestin receptor mRNA in rat mediobasal hypothalamus as demonstrated by in situ hybridization. Neuroendocrinology 53: 608–613. Lebouf BJ (1970) Copulatory and aggressive behavior in the pre-pubertally castrated male dog. Hormones and Behavior 1: 127–136. Lee SK, Kim HJ, Kim TS, Choi HS, Im SY, and Lee JW (1998) Steroid receptor coactivator-I co-activates activating protein-I-mediated transactivations through interaction with the c-Jun and c-Fos subunits. Journal of Biological Chemistry 273: 16651–16654. Lehne GK (1988) Treatment of sexual offenders with cyproterone acetate. In: Sitsen JMA (ed.) Handbook of Sexology, vol. 6, pp. 516–525. New York: Elsevier Science. Leroy P, Krust A, Zelent A, et al. (1991) Multiple isoforms of the mouse retinoic acid receptor alpha are generated by alternative splicing and differential induction by retinoic acid. EMBO Journal 10: 59–69. Lessey BA, Alexander PS, and Horwitz KB (1983) The subunit structure of human breast cancer progesterone receptors: Characterization by chromatography and photoaffinity labeling. Endocrinology 112: 1267–1274. Levine JE, Chappell PE, Schneider JS, Sleiter NC, and Szabo M (2001) Progesterone receptors as neuroendocrine integrators. Frontiers in Neuroendocrinology 22: 69–106. Li H, Gomes PJ, and Chen JD (1997) RAC3, a steroid/nuclear receptor-associated coactivator that is related to SRC-1 and TIF2. Proceedings of the National Academy of Sciences of the United States of America 94: 8478–8484. Li X, Lonard DM, and O’Malley BW (2004) A contemporary understanding of progesterone receptor function. Mechanisms of Ageing and Development 125: 669–678. Lim CS, Baumann CT, Htun H, Xian WJ, Irie M, Smith CL, and Hager GL (1999) Differential localization and activity of the A- and B-forms of the human progesterone receptor using green fluorescent protein chimeras. Molecular Endocrinology 13: 366–375. Lindzey J and Crews D (1988) Effects of progestins on sexual behavior in castrated lizards (Cnemidophorus inornatus). Journal of Endocrinology 119: 265–273. Lisk RD (1962) Diencephalic placement of estradiol and sexual receptivity in the female rat. American Journal of Physiology 203: 493–496.
1497
Lonard DM, Lanz RB, and O’Malley BW (2008) Nuclear receptor coregulators and human disease. Endocrine Reviews 28: 575–587. Lonard DM and O’Malley BW (2006) The expanding cosmos of nuclear recptor coactivators. Cell 125: 411–415. Lonard DM and O’Malley BW (2007) Nuclear receptor coregulators: Judges, juries and executioners of cellular regulation. Molecular Cell 27: 691–700. Lonstein JS and Blaustein JD (2004) Immunocytochemical investigation of nuclear progestin receptor expression within dopaminergic neurons of the female rat brains. Journal of Neuroendocrinology 16: 534–543. Loosefelt H, Logeat F, VuHai MI, and Milgrom E (1984) The rabbit progesterone receptor. Evidence for a single steroid-binding subunit and characterization of receptor mRNA. Journal of Biological Chemistry 259: 14196–14202. Lo¨sel R and Wehling M (2003) Nongenomic actions of steroid hormones. Nature Reviews Molecular Cell Biology 4: 46–56. Luine V, Park D, Reis D, and McEwen BS (1980) Immunochemical demonstration of increased choline acetyltransferase concentration in rat preoptic area after estradiol administration. Brain Research 191: 273–277. Luisi BF, Xu WX, Otwinowski Z, Freedman LP, Yamamoto KR, and Sigler PB (1991) Crystallographic analysis of the interaction of the glucocorticoid receptor with DNA. Nature 352: 497–505. Luttge WG and Hughes JR (1976) Intracerebral implantation of progesterone: Reexamination of the brain sites responsible for facilitation of sexual receptivity in estrogen-primed ovariectomized rats. Physiology and Behavior 17: 771–775. MacLusky NJ, Lieberburg I, Krey LC, and McEwen BS (1980) Progestin receptors in the brain and pituitary of bonnet monkey (Macaca radiata): Differences between the monkey and the rat in the distribution of progestin receptors. Endocrinology 106: 185–191. MacLusky NJ and McEwen BS (1978) Oestrogen modulates progestin receptor concentration in some brain areas and not in others. Nature 274: 276–277. MacLusky NJ and McEwen BS (1980a) Progestin receptors in rat brain: Distribution and properties of cytoplasmic progestin binding sites. Endocrinology 106: 192–202. MacLusky NJ and McEwen BS (1980b) Progestin receptors in the developing rat brain and pituitary. Brain Research 189: 262–268. Mangal RK, Wiehle RD, Poindexter AN, and Weigel NL (1997) Differential expression of uterine progesterone receptor forms A and B during the menstrual cycle. Journal of Steroid Biochemistry and Molecular Biology 63: 195–202. Mani S (2008) Progestin receptor subtypes in the brain: The known and the unknown. Endocrinology 149, doi:10.1210/en.2008-0097. Mani SK, Allen JMC, Clark JH, Blaustein JD, and O’Malley BW (1994a) Convergent pathways for steroid hormone- and neurotransmitter-induced rat sexual behavior. Science 265: 1246–1249. Mani SK, Allen JMC, Clark JH, and O’Malley BW (1995) Progesterone receptor involvement in the LHRH-facilitated sexual behavior in female rats. Society of Neuroscience, Abstract 77: PI–P440. Mani SK, Allen JMC, Lydon JP, et al. (1996) Dopamine requires the unoccupied progesterone receptor to induce sexual behavior in mice. Molecular Endocrinology 10: 1728–1737. Mani SK, Blaustein JD, Allen JMc, Law SW, O’Malley BW, and Clark JH (1994b) Inhibition of rat sexual behavior by antisense oligonucleotides to the progesterone receptor. Endocrinology 135: 1409–1414.
1498
Mechanism of Progesterone Receptor Action in the Brain
Mani SK, Fienberg AA, O’Callaghan JP, et al. (2000) Requirement for DARPP-32 in progesterone-facilitated sexual receptivity in female rats and mice. Science 287: 1053–1056. Mani SK, Mitchell AM, and O’Malley BW (2001) Progesterone receptors and dopamine receptors are required in D9-tetrahydrocannabinal-modulation of sexual receptivity in female rats. Proceedings of the National Academy of Sciences of the United States of America 98: 1249–1254. Mani SK, Reyna AM, Chen JZ, Mulac-Jericevic B, and Conneely OM (2006) Differential response of progesterone receptor isoforms in hormone-dependent and -independent facilitation of female sexual receptivity. Molecular Endocrinology 20: 1322–1332. Marshall FHA and Hammond J (1945) Experimental control by hormone action of the oestrous cycle in the ferret. Journal of Endocrinology 4: 159–168. Marshall FHA and Jolly WA (1905) Contributions to the physiology of mammalian reproduction. II. The ovary as an organ of internal secretion. Philosophical Transactions of the Royal Society (Biology) 198: 123–142. Martinez-Vargas MC, Stumpf WE, and Sar M (1975) Estrogen localization in the dove brain. Phylogenetic considerations and implications for nomenclature. In: Stumpf WE and Grant LD (eds.) Anatomical Neuroendocrinology, pp. 166–175. Basel: Karger. Matsura T, Sutcliffe JS, Fang P, et al. (1997) De novo truncating mutations in E6/AP ubiquitin-protein ligase gene (UBE3A) in Angelman syndrome. Nature Genetics 15: 74–77. McEwen BS (1991) Non-genomic and genomic effects of steroids on neural activity. Trends in Pharmacological Sciences 12: 141–146. McEwen BS (1994) Steroid hormone actions in the brain: When is genome involved? Hormones and Behavior 28: 396–405. McEwen BS, Biegon A, Davis PG, et al. (1982) Steroid hormones: Humoral signals which alter brain cell properties and functions. Recent Progress in Hormone Research 38: 41–92. McGinnis MY, Parsons B, Rainbow TC, and McEwen BS (1981) Temporal relationship between cell nuclear progestin receptor levels and sexual receptivity following intravenous progesterone administration. Brain Research 218: 365–371. McInerney EM, Tsai M-J, O’Malley BW, and Katzenellenbogen BS (1996) Analysis of estrogen receptor transcriptional enhancement by a nuclear hormone receptor coactivator. Proceedings of the National Academy of Sciences of the United States of America 93: 10069–10073. McKenna NJ, Lanz RB, and O’Malley BW (1999) Nuclear receptor coregulators: Cellular and molecular biology. Endocrine Reviews 20: 321–344. McKenna NJ, Nawaz Z, Tsai SY, Tsai M-J, and O’Malley BW (1998) Distinct steady state nuclear receptor coregulator complexes exist in vitro. Proceedings of the National Academy of Sciences of the United States of America 95: 11697–11702. McNicol D and Crews D (1979) Estrogen/progesterone synergy in the control of female sexual receptivity in the lizard. Anolis carolinensis. General and Comparative Endocrinology 38: 68–74. Meijer OC, Steenbergen PJ, and DeKloet ER (2000) Differential expression and regional distribution of steroid receptor co activators SRC-1 and SRC-2 in brain and pituitary. Endocrinology 141: 2192–2199. Meiri H (1986) Is synaptic transmission modulated by progesterone? Brain Research 385: 193–196. Meisel RL, Fraile IG, and Pfaff DW (1990) Hypothalamic sites of progestin action on aggression and sexual behavior in female Syrian hamsters. Physiology and Behavior 47: 219–223.
Meisel RL and Pfaff DW (1984) RNA and protein synthesis inhibitors: Effects on sexual behavior in female rats. Brain Research Bulletin 12: 187–193. Meisel RL and Pfaff DW (1985) Specificity and neural sites of action of anisomycin in the reduction of facilitation of female sexual behavior in rats. Hormones and Behavior 19: 237–251. Meredith JM, Moffatt CA, Auger AP, Snyder GL, Greengard P, and Blaustein JD (1998) Mating-related stimulation induces phosphorylation of dopamine- and cyclic AMP-regulated phosphoprotein-32 in progestin receptorcontaining areas in the female rat brain. Journal of Neuroscience 18: 10189–10195. Meyer C, Schmid R, Scriba PC, and Wehling M (1996) Purification and partial sequencing of high affinity progesterone binding site(s) from porcine liver meembranes. European Journal of Biochemistry 239: 726–731. Meyerson B (1972) Latency between intravenous injection of progestins and the appearance of oestrous behavior in estrogen-treated ovariectomized rats. Hormones and Behavior 3: 1–10. Michael RP and Scott PP (1957) Quantitative studies on mating behavior of spayed female cats stimulated by treatment with oestrogens. Journal of Physiology 138: 46–47. Misiti S, Koibuchi N, Bei M, Farsetti A, and Chin WW (1999) Expression of steroid receptor coactivator-l mRNA in the developing mouse embryo: A possible role in olfactory epithelium development. Endocrinology 140: 1957–1960. Misiti S, Schomburg L, Yen PM, and Chin WW (1998) Expression and hormonal regulation of coactivator and corepressor genes. Endocrinology 139: 2493–2500. Moguilewsky M and Raynaud JP (1977) Progestin binding sites in the rat hypothalamus, pituitary and uterus. Steroids 30: 99–109. Moguilewsky M and Raynaud JP (1979) Estrogen-sensitive progestin-binding sites in the female rat brain and pituitary. Brain Research 164: 165–175. Molenda HA, Griffin AL, Auger AP, McCarthy MM, and Tetel MJ (2002) Nuclear receptor coactivators modulate hormone-dependent gene expression in brain and female reproductive behavior in rats. Endocrinology 143: 436–444. Moore NL, Narayanan R, and Weigel NL (2007) Cyclin-dependent kinase 2 and the regulation of human progesterone receptor activity. Steroids 72: 202–209. Morin LP (1977) Progesterone: Inhibition of rodent sexual behavior. Physiology and Behavior 18: 701–715. Morin LP and Feder HH (1974) Hypothalamic progesterone implants and facilitation of lordosis behavior in estrogen-primed ovariectomized guinea pigs. Brain Research 70: 81–93. Moss RL, Dudley CA, and Schwartz NB (1977) Coitusinduced release of luteinizing hormone in the proestrous rat: Fantasy or fact? Endocrinology 100: 394–397. Mulac-Jericevic B and Conneely OM (2004) Reproductive tissue selective actions of progesterone receptors. Reproduction 128: 139–146. Na SY, Lee SK, Han SJ, Choi HS, Im SY, and Lee JW (1998) Steroid receptor coactivator-l interacts with p50 subunit and coactivates nuclear factor kappaB-mediated transactivations. Journal of Biological Chemistry 23: 10831–10834. Nairn AC, Hemmings HC, Jr., and Greengard P (1985) Protein kinases in the brain. Annual Review of Biochemistry 54: 931–976. Nawaz Z, Lonard DM, Smith CL, et al. (1999) The Angelman syndrome-associated protein, E6-AP, is a coactivator for the nuclear hormone superfamily. Molecular and Cellular Biology 19: 1182–1189.
Mechanism of Progesterone Receptor Action in the Brain
Nishihara E, O’Malley BW, and Xu J (2004) Nuclear receptor coregulators are new players in nervous system development and function. Molecular Neurobiology 30: 307–325. Nock B and Feder HH (1981) Neurotransmitter modulation of steroid action in target cells that mediate reproduction and reproductive behavior. Neuroscience and Biobehavioral Reviews 5: 437–447. Nock B and Feder HH (1984) a1-Noradrenergic transmission and female sexual behavior of guinea pigs. Brain Research 310: 77–85. Numan M, Roach JK, DelCerro MCR, Guillamon A, Segovia S, Sheehan IP, and Numan MJ (1999) Expression of intracellular progesterone receptors in the rat brain during different reproductive states, and in involvement in maternal behavior. Brain Research 830: 358–371. Odell WD and Serdloff RS (1968) Progesterone-induced luteinizing and follicle-stimulating hormone surge in postmenopausal women: A simulated ovulatory peak. Proceedings of the National Academy of Sciences of the United States of America 61: 529–536. Olesen KM, Auger CJ, and Auger AP (2007) Regulation of progestin receptor expression in the developing rat brain by a dopamine D1 receptor antagonist. Journal of Neuroendocrinology 19: 481–488. Olesen KM, Jessen HM, Auger CJ, and Auger AP (2005) Dopaminergic activation of estrogen receptors in neonatal brain alters progestin receptor expressin and juvenile social play behavior. Endocrinology 146: 3706–3712. O’Malley BW (2007) Coregulators: From whence came these master genes. Molecular Endocrinology 21: 1009–1013. O’Malley BW and Means AR (1974) Female steroid hormones and target cell nuclei. Science 183: 610–620. O’Malley BW, Schrader WT, Mani S, Smith CL, Weigel NL, Conneely OM, and Clark JH (1995) An alternative ligand-independent pathway for activation of steroid receptors. Recent Progress in Hormone Research 50: 333–347. O’Malley BW, Tsai SY, Bagchi M, Weigel NL, Schrader WT, and Tsai M-J (1991) Molecular mechanism of action of a steroid hormone receptor. Recent Progress in Hormone Research 47: 1–24. Ogawa H, Nishi M, and Kawata M (2001) Localization of nuclear coactivators p300 and steroid receptor coactivator 1 in the rat hippocampus. Brain Research 890: 197–202. Ogawa S, Olazabal S, and Pfaff DW (1994) Effects of intrahypothalamic administration of antisense DNA for progesterone receptor mRNA on reproductive behavior and progesterone receptor immunoreactivity. Journal of Neuroscience 14: 1766–1774. Ogryzko VV, Schiltz RL, Russanova V, Howard BH, and Nakatani Y (1996) The transcriptional coactivators p300 and CBP are histone acetyl transferases. Cell 87: 953–959. Oike Y, Hata A, Mamiya T, Kaname T, et al. (1999) Truncated CBP protein leads to Rubinstein–Taybi syndrome phenotypes in mice: Implications for a dominant negative mechanism. Human Molecular Genetics 8: 387–396. Olster DH and Blaustein JD (1998) Progesterone facilitation of lordosis in male and female Sprague-Dawley rats following priming with estradiol pulses. Hormones and Behavior 22: 294–304. Ona˜te SA, Boonyaratanakornkit V, Spencer TE, Tsai SY, Tsai M-J, Edwards DP, and O’Malley BW (1998) The steroid receptor coactivator-l contains multiple receptor interacting and activation domains that cooperatively enhance the activation function 1 (AF1) and AF2 domains of steroid receptors. Journal of Biological Chemistry 273: 12101–12108.
1499
Ona˜te SA, Tsai SY, Tsai M-J, and O’Malley BW (1995) Sequence and characterization of a coactivator for the steroid hormone receptor superfamily. Science 270: 1354–1357. Parsons B, MacLusky NJ, Krey LC, Pfaff DW, and McEwen BS (1980) Temporal relationship between estrogen inducible progestin receptors in the female rat brain and the time course of estrogen activation of mating behavior. Endocrinology 107: 774–779. Parsons B, Rainbow TC, MacLusky NJ, and McEwen BS (1982) Progestin receptor levels in rat hypothalamic and limbic nuclei. Journal of Neuroscience 2: 1446–1452. Peluso JJ, Pappalardo A, Losel R, and Wehling M (2006) Progesterone membrane receptor component 1 expression in the immature rat ovary and its role in mediating progesterone’s antiapoptotic action. Endocrinology 147: 3133–3140. Pfaff DW, Ogawa S, Kia K, Vasudevan N, Krebs C, Frohlich J, and Kow LM (2002) Genetic mechanisms in neural and hormonal controls over female reproductive behaviors. In: Pfaff DW, Arnold AP, Etgen AM, Fahrbach SE, and Rubin RT (eds.) Hormones, Brain and Behavior, vol. III, pp. 441–510. San Diego, CA: Academic Press. Pfaff DW, Schwartz-Giblin S, McCarthy MM, and Kow L (1994) Cellular and molecular mechanisms of female reproductive behavior. In: Knobil E and Neill JD (eds.) Physiology of Reproduction, pp. 107–220. New York: Raven. Phelps SM, Lydon JP, O’Malley BW, and Crews D (1999) Regulation of male sexual behavior by progesterone receptor, sexual experience, and androgen. Hormones and Behavior 34: 294–302. Phillips RW, Fraps RM, and Frank AH (1946) Ovulation and estrus in sheep and goats. In: Engle ET (ed.) The Problem of Fertility, pp. 11–48. Princeton, NJ: Princeton University Press. Philpott AJ and Shaheed M (1996) Dopamine-mediated activation of the human progesterone receptor. Cellular and Molecular Neurobiology 16: 417–420. Phoenix CH, Goy RW, Gerall AA, and Young WC (1959) Organizing action of prenatally administered testosterone propiOna˜te on the tissues mediating mating behavior in the female guinea pig. Endocrinology 65: 369–376. Pierson-Mullany LK and Lange CA (2004) Phosphorylation of progesterone receptor serine 400 mediates liganddependent transcriptional activity in response to activation of cyclin-dependent protein kinase 2. Molecular and Cellular Biology 24: 10542–10547. Pollio G, Xue P, Zanisi A, Nicolin A, and Maggi A (1993) Antisense oligonucleotide blocks progesterone-induced lordosis behavior in ovariectomized rats. Molecular Brain Research 19: 135–139. Power RF, Lydon JP, Conneely OM, and O’Malley BW (1991a) Dopamine activation of an orphan of the steroid receptor superfamily. Science 252: 1546–1548. Power RF, Mani SK, Codina J, Conneely OM, and O’Malley BW (1991b) Dopaminergic and ligand independent activation of steroid hormone receptors. Science 254: 1636–1639. Powers JB (1972) Facilitation of lordosis in ovariectomized rats by intracerebral implants. Brain Research 48: 311–325. Quadros PS, Lopez V, DeVries GJ, Chung WC, and Wagner CK (2002) Progesterone receptors and sexual differentiation of the medial preoptic nucleus. Journal of Neurobiology 51: 24–32. Quadros PS and Wagner CK (2008) Regulation of progesterone receptor expression by estradiol is dependent on age, sex and region in the rat brain. Endocrinology PMID: 18308846. Rainbow TC, McGinnis MY, Davis PG, and McEwen BS (1982) Application of anisomysin to the lateral ventromedial nucleus
1500
Mechanism of Progesterone Receptor Action in the Brain
of the hypothalamus inhibits the activation of sexual behavior by estradiol and progesterone. Brain Research 223: 417–423. Ramirez VD, Dluzen DE, and Ke FC (1990) Effects of progesterone and its metabolites on neuronal membranes. Ciba Foundation Symposium 153: 125–144. Ramirez VD, Zheng J, and Siddique KM (1996) Membrane progesterone receptors for estrogen, progesterone and testosterone in the rat brain: Fantasy or reality. Cellular and Molecular Neurobiology 16: 175–198. Rao KV, Peralta WD, Greene GL, and Fox CF (1987) Cellular progesterone receptor phosphorylation in response to ligands activating protein kinases. Biochemical and Biophysical Research Communications 146: 1357–1365. Reyer HI and Feder HH (1984a) Development of steroid receptor systems in guinea pig brain. II. Cytoplasmic progestin receptors. Brain Research 315: 5–21. Rhodes D (1997) The nucleosome core all wrapped up. Nature 389: 231–233. Richmond ME and Conaway CH (1969) Induced ovulation and estrus in Microtus ochrogater. Journal of Reproduction and Fertility 6: 357–376. Richmond TJ, Finch JT, Rushton B, Rhodes D, and Klug A (1984) Structure of the nucleosome core particle at 7A resolution. Nature 311: 532–537. Robinson TJ (1955) Quantitative studies on the hormonal induction of oestrus in spayed ewes. Journal of Endocrinology 12: 163–173. Robinson TJ, Moore NW, and Binet FE (1956) The effect of duration of progesterone pretreatment on the response of the spayed ewe to oestrogen. Journal of Endocrinology 14: 1–7. Roeder RG (1996) The role of general initiation factors in transcription by RNA polymerase II. Trends in Biochemical Sciences 21: 327–335. Romano GJ, Harlan RE, Shivers BB, Howells RD, and Pfaff DW (1989a) Estrogen increases pro-enkephalin mRNA levels in the ventromedial hypothalamus in the rat. Molecular Endocrinology 2: 1320–1328. Romano GJ, Krust A, and Pfaff DW (1989b) Expression and estrogen regulation of progesterone receptor mRNA in neurons of the mediobasal hypothalamus: An in situ hybridization study. Molecular Endocrinology 3: 1295–1300. Roselli CE, Resko JA, and Stormshak F (2006) Expression of steroid hormone receptors in the fetal sheep brain during the critical period for sexual differentiation. Brain Research 1110: 76–80. Ross J, Claybough C, Clemens LG, and Gorski RA (1971) Sort latency of estrous behavior with intracerebral gonadal hormones in ovariectomized rats. Endocrinology 89: 32–38. Rowan BG, Garrison N, Weigel NL, and O’Malley BW (2000b) 8-Bromo-cyclic AMP induces phosphorylation of two sites in SRC-1 that facilitate ligand-independent activation of the chicken progesterone receptor and are critical for functional cooperation between SRC-1 and CREB binding protein. Molecular and Cellular Biology 20: 8720–8730. Rowan BG, Weigel NL, and O’Malley BW (2000a) Phosphorylation of steroid receptor coactivator-1. Identification of the phosphorylation sites and phosphorylation through the mitogen-activated protein kinase pathway. Journal of Biological Chemistry 275: 4475–4483. Rubin BS and Barfield RJ (1983a) Induction of estrous behavior in ovariectomized rats by sequential replacement of estrogen and progesterone to the ventromedial hypothalamus. Neuroendoclinology 37: 218–224. Rubin BS and Barfield RJ (1983b) Priming of estrous responsiveness by implants of 17/3-estradiol in the
ventromedial hypothalamic nucleus of female rats. Endocrinology 106: 504–509. Rundlett SE, Carmen AA, Kobayashi R, Bavykin S, Turner BM, and Grunstein M (1996) HDAI and RPD3 are members of yeast histone deacetylase complexes that regulate silencing and transcription. Proceedings of the National Academy of Sciences of the United States of America 93: 14503–14508. Sanyal MKJ (1978) Secretion of progesterone during gestation in female rat. Endocrinology 79: 179–190. Sartorius CA, Melville MY, Hovland AR, Tung L, Takimoto GS, and Horwitz KB (1994) A third transactivation function (AF3) of human progesterone receptors located in the unique N-terminal segment of the B-isoform. Molecular Endocrinology 8: 1347–1360. Savouret JE, Bailly A, Misrahi M, Rauch C, Redeuilh G, Chaucherau A, and Milgrom E (1991) Characterization of the hormone responsive element involved in the regulation of the progesterone receptor gene. EMBO Journal 10: 1875–1883. Sawyer CH and Everett JW (1959) Stimulatory and inhibitory effects of progesterone on the release of pituitary ovulating hormone in the rabbit. Endocrinology 65: 622–630. Schneider W, Ramachandran C, Satyaswaroop PG, and Shyamala G (1991) Murine progesterone receptor exists predominantly as the 83-kilodalton ‘A’ form. Journal of Steroid Biochemistry and Molecular Biology 38: 285–291. Schrader WT and O’Malley BW (1972) Progesterone-binding components of chick oviduct. IV. Characterization of purified subunits. Journal of Biological Chemistry 247: 51–59. Schumacher M, Coirini H, Pfaff DW, and McEwen BS (1990) Behavioral effects of progesterone associated with rapid modulation of oxytocin receptors. Science 250: 691–694. Schumacher M, Coirini H, Robert E, Guennoun R, and El-Etr M (1999) Genomic and membrane actions of progesterone: Implications for reproductive physiology and behavior. Behavioural Brain Research 105: 37–52. Schwerk C, Klotzbucher M, Sachs M, Ulber V, and Klein-Hitpass L (1995) Identification of a transactivation function in the progesterone receptor that interacts with the TAFII110 subunit of the TFIID complex. Journal of Biological Chemistry 270: 21331–21338. Selmin O, Lucier GW, Clark GC, et al. (1996) Isolation and characterization of a novel gene induced by 2,3,7,8tetrachlorodibenzo-p-dioxin in rat liver. Carcinogenesis 17: 2609–2615. Selye H (1941) Steroid hormones. Proceedings of the Society for Experimental Biology and Medicine 46: 116–121. Shenolikar S and Nairn AC (1991) Protein phosphatases: Recent progress. Advances in Second Messenger and Phosphoprotein Research 23: 1–123. Sheridan PL, Evans RM, and Horwitz KB (1989) Phosphotryptic peptide analysis of human progesterone receptor. New phosphorylated sites formed in nuclei after hormone treatment. Journal of Biological Chemistry 264: 6520–6528. Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard DA, and Greene GL (1998) The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell 95: 927–937. Shughrue PJ, Stumpf WE, Elger W, Schulze PE, and Sar M (1991a) Progestin receptor cells in mouse cerebral cortex during early postnatal development: A comparison with preoptic area and central hypothalamus using autoradiography with [125I] progestin. Brain Research – Developmental Brain Research 59: 143–155. Shughrue PJ, Stumpf WE, Elger W, Schulze PE, and Sar M (1991b) Progestin receptor cells in the 8-day-old male and female mouse cerebral cortex: Autoradiographic evidence for a sexual dimorphism in target cell number. Endocrinology 128: 87–95.
Mechanism of Progesterone Receptor Action in the Brain
Simmerly RB (1989) Hormonal control of the development and regulation of tyrosine hydroxylase expression within a sexually dimorphic population of dopaminergic cells in the hypothalamus. Molecular Brain Research 6: 297–310. Smith CL, Nawaz Z, and O’Malley BW (1997) Coactivator and corepressor regulation of the agonist/antagonist activity of the mixed antiestrogen, 4-hydroxytamoxifen. Molecular Endocrinology 11: 657–666. Smith CL, Ona˜te S, Tsai M-J, and O’Malley BW (1996) CREB binding protein acts synergistically with steroid receptor coactivator-1 to enhance steroid receptordependent transcription. Proceedings of the National Academy of Sciences of the United States of America 93: 8884–8888. Smith DE, Faber LE, and Toft DO (1990) Purification of unactivated progesterone receptor and identification of novel receptor-associated proteins. Journal of Biological Chemistry 265: 3996–4003. Smith SS, Waterhouse BD, and Woodward DJ (1987) Sex steroid effects on extrahypothalamic CNS. II. Progesterone, alone and in combination with estrogen, modulates responses to amino acid neurotransmitters. Brain Research 422: 52–62. Sodersten P and Enoroth P (1978) Evidence that progesterone does not inhibit the induction of sexual receptivity by oestradiol-17b in the rat. Journal of Endocrinology 89: 63–69. Spencer TE, Jenster G, Burcin MM, et al. (1997) Steroid receptor coactivator-I is a histone acetyl transferase. Nature 389: 194–198. Spies HG and Niswender GD (1972) Effect of progesterone and estradiol on LH release and ovulation in rhesus monkeys. Endocrinology 90: 257–261. Stromberg H, Svensson SP, and Hermanson O (1999) Distribution of CREB-binding protein immunoreactivity in the adult rat brain. Brain Research 818: 510–514. Suen CS, Berrodin TJ, Mastroeni R, Cheskis BJ, Lyttle CR, and Frail DE (1998) A transcriptional coactivator, steroid receptor coactivator-3 selectively augments steroid receptor transcriptional activity. Journal of Biological Chemistry 273: 27645–27653. Sullivan WP, Madden BJ, McCormick DJ, and Toft DO (1988) Hormone-dependent phosphorylation of the avian progesterone receptor. Journal of Biological Chemistry 263: 14717–14723. Szabo M, Kilen SM, Nho SJ, and Schwartz NB (2000) Progesterone receptor A and B messenger ribonucleic acid levels in the anterior pituitary of rats are regulated by estrogen. Biology of Reproduction 62: 95–102. Takeshita A, Cardona GR, Koibuchi N, Suen C-S, and Chin WW (1997) TRAM-1, a novel 160-kDa thyroid receptor activator molecule exhibits distinct properties from steroid receptor coactivator-1. Journal of Biological Chemistry 272: 27629–27634. Takimoto GS, Tasset DM, Eppert AC, and Horwitz KB (1992) Hormone-induced progesterone-induced progesterone receptor phosphorylation consists of sequential DNA-independent and DNA-dependent stages: Analysis with zinc finger mutants and the progesterone antagonist ZK98299. Proceedings of the National Academy of Sciences of the United States of America 89: 3050–3054. Tauton J, Hassig CA, and Schreiber SL (1996) A mammalian histone de acetylase related to yeast transcriptional regulator RPD3p. Science 272: 408–411. Tennent BJ, Smith ER, and Davidson JM (1982) Effects of progesterone implants in the habenula and midbrain on proceptive and receptive behavior in the female rat. Hormones and Behavior 16: 352–373.
1501
Tetel MJ (2000) Nuclear receptor coactivators in neuroendocrine function. Journal of Neuroendocrinology 12: 927–932. Tetel MJ, Giangrande PH, Leonhardt SA, McDonnel DP, and Edwards DP (1999) Hormone-dependent interaction between amino- and carboxyl-terminal domains of progesterone receptor in vitro and in vivo. Molecular Endocrinology 13: 910–924. Tetel MJ, Siegel NK, and Murphy SD (2007) Cells in behaviorally relevant brain regions coexpress nuclear receptor coactivators and ovarian steroid receptors. Journal of Neuroendocrinology 19: 262–271. Thornton JE, Nock B, McEwen BS, and Feder HH (1986) Noradrenergic modulation of hypothalamic progestin receptors in female guinea pigs is specific to the ventromedial nucleus. Brain Research 377: 155–159. Tora L, Gronemeyer H, Turcotte B, Gaub MP, and Chambon P (1988) The N-terminal region of the chicken progesterone receptor specifies target gene activation. Nature 333: 185–188. Torchia J, Rose DW, Inostroza J, Kamei Y, Westin S, Glass CK, and Rosenfeld MG (1997) The transcriptional coactivator p/CIP binds CBP and mediates nuclear-receptor function. Nature 387: 677–684. Tsai M-J and O’Malley BW (1994) Molecular mechanism of action of steroid/thyroid receptor superfamily members. Annual Review of Biochemistry 63: 451–486. Turgeon J and Waring DW (1992) A pathway for luteinizing hormone releasing-hormone self-potentiation: Cross-talk with the progesterone receptor. Endocrinology 130: 3275–3282. Turgeon JL and Waring DW (1994) Activation of the progesterone receptor by the gonadotropin-releasing hormone self-priming signaling pathway. Molecular Endocrinology 8: 860–869. Tzukerman MT, Esty A, Santiso-Mere D, et al. (1994) Human estrogen receptor transactivational capacity is determined by both cellular and promoter context and mediated by two functionally distinct intramolecular regions. Molecular Endocrinology 8: 21–30. Umesono K and Evans RM (1989) Determinants of target gene specificity for steroid/thyroid hormone receptors. Cell 57: 1139–1146. Vallee M, Mayo W, Darnaudery M, et al. (1997) Neurosteroids: Deficient cognitive performance in aged rats depends on low pregnene sulfate levels in the hippocampus. Proceedings of the National Academy of Sciences of the United States of America 94: 14865–14870. Van Tol HHM, Bolwerk ELM, Liu B, and Burbach JPH (1988) Oxytocin and vasopressin gene expression in the hypothalamo-neurohypophyseal system of the rat during the estrous cycle, pregnancy and lactation. Endocrinology 122: 945–949. Vegeto E, Allan GE, Schrader WT, Tsai M-J, McDonnell DP, and O’Malley BW (1992) The mechanism of RU486 antagonism is dependent on the conformation of the carboxyterminal tail of the human progesterone receptor. Cell 69: 703–713. Vegeto E, Shahbaz MM, Wen DX, Goldman ME, and O’Malley BW (1993) Human progesterone receptor A form is cell- and promoter-specific repressor of human progesterone receptor B function. Molecular Endocrinology 75: 1244–1255. Vincent PA and Feder HH (1988) Alpha-1- and alpha-2 noradrenergic receptors modulate lordosis behavior in female guinea pigs. Neuroendocrinology 48: 477–481. Voegel JJ, Heine MS, Tini M, Vivat V, Chambon P, and Gronemeyer H (1998) The coactivator TIF2 contains three nuclear receptor-binding motifs and mediates
1502
Mechanism of Progesterone Receptor Action in the Brain
transactivation through CBP binding-dependent and -independent pathways. EMBO Journal 17: 507–519. Voegel JJ, Heine MJ, Zechel C, Chambon P, and Gronemeyer H (1996) TIF2, a 160 kDa transcriptional mediator for the ligand-dependent activation function AF-2 of nuclear receptors. EMBO Journal 15: 3667–3675. Verrijdt G, Haelens A, Schoenmakers E, Rombauts W, and Claessens F (2002) Comparative analysis of the influence of HMG box 1 protein on DNA binding and transcriptional activation by androgen glucocorticoid, progesterone and mineralocorticoid receptors. Biochemical Journal 361: 97–103. Wade PA, Jons PL, Vermaak D, and Wolffe AP (1998) A multiple subunit Mi-2 histone deacetylase from Xenopus laevis co fractionates with an associated Snf2 superfamily ATPase. Current Biology 8: 843–846. Wagner BL, Norris JD, Knotts TA, Weigel NL, and McDonell DP (1998) The nuclear corepressors NcoR and SMRT are key regulators of both ligand- and 8-bromo-cAMP-dependent transcriptional activity of human progesterone receptor. Molecular and Cellular Biology 18: 1369–1378. Wagner CK (2008) Progesterone receptors and neural development: A gap between bench and bedside? Endocrinology PMID: 1808849. Wagner CK, Nakayama AY, and DeVries GJ (1999) Potential role of maternal progesterone in the sexual differentiation of the brain. Endocrinology 139: 3658–3661. Walfish PG, Yoganathan T, Yang YF, Hong H, Butt IR, and Stallcup MR (1997) Yeast hormone response element assays detect and characterize GRIP1 coactivator-dependent activation of transcription by thyroid and retinoid nuclear receptors. Proceedings of the National Academy of Sciences of the United States of America 94: 3697–3702. Wallen K, Goldfoot DA, joslyn WD, and Paris CA (1972) Modification of behavioral estrus in the guinea pig following intracranial hexamide. Physiology and Behavior 8: 221–224. Wallen K and Thonton JE (1979) Progesterone and duration of heat in estrogen treated, ovariectomized guinea pigs. Physiology and Behavior 22: 95–97. Walker QD, Frances J, Cabassa J, and Kuhn CM (2001) Effect of ovarian hormones and estrous cycle on stimulation of the hypothalamo-pituitary–adrenal axis by cocaine. Journal of Pharmacology and Experimental Therapeutics 297: 291–298. Wang JC, Stafford JM, and Granner DK (1998) SRC-1 and GRIP-1 coactivate transcription with hepatocyte nuclear factor 4. Journal of Biological Chemistry 273: 30847–30850. Ward LL, Crowley WR, Zemlan FP, and Margules DL (1975) Monoaminergic mediation of female sexual behavior. Journal of Comparative and Physiological Psychology 88: 53–61. Wei LL, Gonzalez-Aller C, Wood WM, Miller LA, and Horwitz KB (1990) 50 -Heterogeneity in human progesterone receptor transcripts predicts a new amino-terminal truncated ‘‘C’’-receptor and unique A-receptor messages. Molecular Endocrinology 4: 1833–1840. Wei LL, Hawkins P, Baker C, Norris B, Sheridan PL, and Quinn PG (1996) An amino-terminal truncated progesterone receptor isoform, PRc, enhances progestin-induced transcriptional activity. Molecular Endocrinology 10: 1379–1387. Weigel NL (1996) Steroid hormone receptors and their regulation by phosphorylation. Biochemical Journal 3: 657–667. Weigel NL, Carter TH, Schrader WT, and O’Malley BW (1992) Chicken progesterone receptor phosphorylated by a DNA-dependent protein kinase during in vitro transcription assays. Molecular Endocrinology 6: 8–14. Weigel NL, Denner LA, Poletti A, Beck CA, Edwards DP, and Zhang Y (1993) Phosphorylation/dephosphorylation
regulates the activity of progesterone receptors. Advances in Protein Phosphatases 7: 1740–1743. Weigel NL and Moore NL (2007) Kinases and protein phosphorylation as regulators of steroid hormone action. Nuclear Receptor Signaling 5: 1–13. Weisz J and Ward IL (1980) Plasma testosterone and progesterone titers of pregnant rats, their male and female fetuses, and neonatal offspring. Endocrinology 106: 306–316. Whalen RE (1974) Estrogen–progesterone induction of mating in female rats. Hormones and Behavior 5: 157–162. Whalen RE, Gorzalka BB, DeBold JF, Quadagno DM, Ho GK, and Hough JC, Jr. (1974) Studies on the effect of intracerebral actinimycin D implants on estrogen-induced receptivity in rats. Hormones and Behavior 5: 337–343. Whalen RE and Lauber AH (1986) Progesterone substitutes: cGMP mediation. Neuroscience and Biobehavioral Reviews 10: 47–53. Wilcox JN and Roberts JL (1990) Estrogen decreases rat hypothalamic POMC mRNA levels. Endocrinology 117: 2392–2396. Williams CL (1987) Estradiol benzoate facilitates lordosis and ear-wiggling of 6-day old rats. Behavioral Neuroscience 101: 819–826. Williams CL and Blaustein JD (1988) Steroids induce hypothalamic progestin receptors and facilitate female sexual behavior in neonatal rats. Brain Research 449: 403–407. Witt DM, Young LJ, and Crews D (1995) Progesterone modulation of androgen-dependent sexual behavior in male rats. Physiology and Behavior 57: 307–313. Wolfe AP and Pruss D (1996) Targeting chromatin disruption: Transcription regulators that acetylate histones. Cell 84: 817–819. Wooley SC, O’Malley BW, Lydon J, and Crews D (2006) Genotype differences in behavior and tyrosine hydroxylase expression between wild-type and progesterone receptor knockout mice. Behavioural Brain Research 167: 197–204. Wu HK, Fabian S, Jenab S, and Quinones-Jenab V (2006) Progesterone receptors activation after acute cocaine administration. Brain Research 1126: 188–192. Xu J, Liao L, Ning G, Yoshida-Komiya H, Deng C, and O’Malley BW (2000) The steroid receptor coactivator SRC3 (p/ClPIRAC3/AIB1/ACTR/TRAM-1) is required for normal growth, puberty, female reproductive function, and mammary gland development. Proceedings of the National Academy of Sciences of the United States of America 97: 6379–6384. Xu J, Nawaz Z, Tsai SY, Tsai M-J, and O’Malley BW (1996) The extreme C terminus of progesterone receptor contains a transcriptional repressor domain that functions through a putative corepressor domain. Proceedings of the National Academy of Sciences of the United States of America 93: 12195–12199. Xu J, Qiu Y, DeMayo FJ, Tsai SY, Tsai M-J, and O’Malley BW (1998) Partial hormone resistance in mice with disruption of the steroid receptor coactivator-1 (SRC-1) gene. Science 279: 1922–1925. Yamamoto KR (1985) Steroid receptor regulated transcription of specific genes and gene networks. Annual Review of Genetics 19: 209–252. Yang XJ, Ogryzko VV, Nishikawa J, Howard BH, and Nakatani Y (1996) A p300/CBP-associated factor that competes with adenoviral oncoprotein E1A. Nature 382: 319–324. Young WC (1969) Psychobiology of sexual behavior in the guinea pig. In: Lehrman DS, Hinde RA, and Shaw E (eds.) Advances in the Study of Behavior, pp. 1–110. New York: Academic Press.
Mechanism of Progesterone Receptor Action in the Brain
Zhang Y, Beck CA, Poletti A, Edwards DP, and Weigel NL (1994a) Identification of phosphorylation sites unique to the B form of human progesterone receptor. In vitro phosphorylation by casein kinase I. Journal of Biological Chemistry 269: 31034–31040. Zhang Y, Bai W, Allgood VE, and Weigel NL (1994b) Multiple signaling pathways activate the chicken progesterone receptor. Molecular Endocrinology 8: 577–584. Zhang Y, Beck CA, Poletti A, Edwards DP, and Weigel NL (1995) Identification of a group of Ser-Pro motif hormone-inducible phosphorylation sites in the human progesterone receptor. Molecular Endocrinology 9: 1029–1040. Zhang Y, Beck CA, Poletti A, et al. (1997) Phosphorylation of human progesterone receptor by cyclin-dependent kinase 2 on three sites that are authentic basal phosphorylation sites in vitro. Molecular Endocrinology 11: 823–832. Zhang X, Jeyakumar M, Petukhov S, and Bagchi M (1998) A nuclear receptor corepressor modulates transcriptional activity of antagonist-occupied steroid hormone receptor. Molecular Endocrinology 12: 513–524. Zhou D, Apostalakis EM, and O’Malley BW (1999) Distribution of D(5) dopamine receptor mRNA in rat ventromedial hypothalamic nucleus. Biochemical and Biophysical Research Communications 266: 556–559. Zhu Y, Bond J, and Thomas P (2003a) Identification, classificatin and partial characterization of genes in humans and other vertebrates homologous to a fish membrane progestin receptor. Proceedings of the National Academy of Sciences of the United States of America 100: 2237–2242. Zhu Y, Qi C, Calandra C, Rao MS, and Reddy JK (1996) Cloning and identification of mouse steroid receptor coactivator-1 (mSRC-1), as a coactivator of peroxisome proliferator-activated receptor g. Gene Expression 6: 185–195. Zhu Y, Rice CD, Pang Y, Pace M, and Thomas P (2003b) Cloning, expression and characterization of a membrane progestin receptor and evidence it is an intermediary in meitic maturation of fish oocytes. Proceedings of the
1503
National Academy of Sciences of the United States of America 100: 2231–2236. Zhou ZX, Kemppainen JA, and Wilson EM (1995) Identification of three proline-directed phosphorylation sites in the human androgen receptor. Molecular Endocrinology 9: 605–615. Zucker I (1967) Progesterone in the experimental control of the behavioral sex cycle in the female rat. Journal of Endocrinology 38: 269–273. Zucker I (1968) Biphasic effects of progesterone on the sexual receptivity in the female guinea pig. Journal of Comparative and Physiological Psychology 3: 472–478.
Further Reading Herbison AE, Horvath TL, Naftolin F, and Leranth C (1995) Distribution of estrogen receptor-immunoreactive cells in monkey hypothalamus: Relationship to neurons containing luteinizing hormone-releasing hormone and tyrosine hydroxylase. Neuroendocrinology 61: 1–10. Mani SK (2001) Ligand-independent activation of progestin receptors in sexual receptivity. Hormones and Behavior 40: 183–190. Razandi M, Pedram A, Greene GL, and Levin ER (1999) Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: Studies of ERa and ER/3 expressed in Chinese hamster ovary cells. Molecular Endocrinology 13: 307–319. Reyer HI and Feder HH (1984b) Development of steroid receptor systems in guinea pig brain. III. Nuclear progestin receptor. Brain Research 315: 23–27. Robinson TJ (1954) The necessity for progesterone with estrogen for the induction of recurrent estrus in the ovariectomized ewes. Transactions of the Royal Society of London 52: 303–362.
Biographical Sketch
Bert W. O’Malley, MD, is the Tom Thompson Distinguished Service Professor and chair of the Department of Molecular and Cellular Biology at Baylor College of Medicine, Houston. His pioneering work on molecular mechanisms of steroid hormone action and hormone receptors has led to several outstanding contributions to the field of molecular endocrinology. He is a member of the National Academy of Sciences, a fellow of the Italian Academy of Science, the past president of the Endocrine Society, and has served on numerous committees and editorial boards. He is a recipient of several honorary degrees and numerous awards, including the National Medal of Science. He has authored more than 600 scientific papers and holds several patents for techniques and inventions in the field.
Shaila K. Mani, PhD, is an associate professor in the Department of Molecular and Cellular Biology at Baylor College of Medicine. Her major research interests include molecular and cellular mechanisms of steroid hormone action in the brain, ligand-independent activation of progestin receptors, nonclassical mechanisms of progesterone action, and the role of signal-transduction pathways in brain and behavior.