Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 887
Filter
1.
Sci Rep ; 13(1): 2164, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36750584

ABSTRACT

Progesterone and its receptors (PRs) participate in mating and reproduction, but their role in spatial declarative memory is not understood. Male mice expressed PRs, predominately in excitatory neurons, in brain regions that support spatial memory, such as the hippocampus and entorhinal cortex (EC). Furthermore, segesterone, a specific PR agonist, activates neurons in both the EC and hippocampus. We assessed the contribution of PRs in promoting spatial and non-spatial cognitive learning in male mice by examining the performance of mice lacking this receptor (PRKO), in novel object recognition, object placement, Y-maze alternation, and Morris-Water Maze (MWM) tasks. In the recognition test, the PRKO mice preferred the familiar object over the novel object. A similar preference for the familiar object was also seen following the EC-specific deletion of PRs. PRKO mice were also unable to recognize the change in object position. We confirmed deficits in spatial memory of PRKO mice by testing them on the Y-maze forced alternation and MWM tasks; PR deletion affected animal's performance in both these tasks. In contrast to spatial tasks, PR removal did not alter the response to fear conditioning. These studies provide novel insights into the role of PRs in facilitating spatial, declarative memory in males, which may help with finding reproductive partners.


Subject(s)
Limbic System , Maze Learning , Receptors, Progesterone , Spatial Memory , Animals , Male , Mice , Entorhinal Cortex/physiology , Hippocampus/physiology , Limbic System/physiology , Maze Learning/physiology , Memory Disorders/physiopathology , Progesterone/physiology , Receptors, Progesterone/physiology , Spatial Memory/physiology
2.
J Neuroendocrinol ; 34(1): e13071, 2022 01.
Article in English | MEDLINE | ID: mdl-34904297

ABSTRACT

Neural circuits in female rats are exposed to sequential estradiol and progesterone to regulate the release of luteinizing hormone (LH) and ultimately ovulation. Estradiol induces progesterone receptors (PGRs) in anteroventral periventricular nucleus (AVPV) kisspeptin neurons, and as estradiol reaches peak concentrations, neuroprogesterone (neuroP) synthesis is induced in hypothalamic astrocytes. This local neuroP signals to PGRs expressed in kisspeptin neurons to trigger the LH surge. We tested the hypothesis that neuroP-PGR signaling through Src family kinase (Src) underlies the LH surge. As observed in vitro, PGR and Src are co-expressed in AVPV neurons. Estradiol treatment increased the number of PGR immunopositive cells and PGR and Src colocalization. Furthermore, estradiol treatment increased the number of AVPV cells that had extranuclear PGR and Src in close proximity (< 40 nm). Infusion of the Src inhibitor (PP2) into the AVPV region of ovariectomized/adrenalectomized (ovx/adx) rats attenuated the LH surge in trunk blood collected 53 h post-estradiol (50 µg) injection that induced neuroP synthesis. Although PP2 reduced the LH surge in estradiol benzoate treated ovx/adx rats, activation of either AVPV PGR or Src in 2 µg estradiol-primed animals significantly elevated LH concentrations compared to dimethyl sulfoxide infused rats. Finally, antagonism of either AVPV PGR or Src blocked the ability of PGR or Src activation to induce an LH surge in estradiol-primed ovx/adx rats. These results indicate that neuroP, which triggers the LH surge, signals through an extranuclear PGR-Src signaling pathway.


Subject(s)
Luteinizing Hormone/metabolism , Neurons/metabolism , Progesterone/metabolism , Receptors, Progesterone/physiology , src-Family Kinases/physiology , Animals , Female , Hypothalamus/metabolism , Ovulation/blood , Ovulation/metabolism , Rats , Rats, Long-Evans , Receptors, Progesterone/metabolism , Signal Transduction/physiology , src-Family Kinases/metabolism
3.
Front Endocrinol (Lausanne) ; 12: 727371, 2021.
Article in English | MEDLINE | ID: mdl-34970218

ABSTRACT

Newly emerging evidence has implicated that progesterone receptor component 1 (PGRMC1) plays a novel role not only in the lipid disturbance induced by atypical antipsychotic drugs (AAPD) but also in the deterioration of glucose homoeostasis induced by clozapine (CLZ) treatment. The present study aimed to investigate the role of PGRMC1 signaling on hepatic gluconeogenesis and glycogenesis in male rats following CLZ treatment (20 mg/kg daily for 4 weeks). Recombinant adeno-associated viruses (AAV) were constructed for the knockdown or overexpression of hepatic PGRMC1. Meanwhile, AG205, the specific inhibitor of PGRMC1 was also used for functional validation of PGRMC1. Hepatic protein expressions were measured by western blotting. Meanwhile, plasma glucose, insulin and glucagon, HbA1c and hepatic glycogen were also determined by assay kits. Additionally, concentrations of progesterone (PROG) in plasma, liver and adrenal gland were measured by a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. Our study demonstrated that CLZ promoted the process of gluconeogenesis and repressed glycogenesis, respectively mediated by PI3K-Akt-FOXO1 and GSK3ß signaling via inhibition of PGRMC1-EGFR/GLP1R in rat liver, along with an increase in fasting blood glucose, HbA1c levels and a decrease in insulin and hepatic glycogen levels. Furthermore, through PGRMC1-EGFR/GLP1R-PI3K-Akt pathway, knockdown or inhibition (by AG205) of PGRMC1 mimics, whereas its overexpression moderately alleviates CLZ-induced glucose disturbances. Potentially, the PGRMC1 target may be regarded as a novel therapeutic strategy for AAPD-induced hepatic glucose metabolism disorder.


Subject(s)
Clozapine/pharmacology , Glucose/metabolism , Liver/drug effects , Membrane Proteins/physiology , Receptors, Progesterone/physiology , Animals , Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacology , Carbohydrate Metabolism/drug effects , Carbohydrate Metabolism/genetics , Clozapine/adverse effects , Glucose Metabolism Disorders/chemically induced , Glucose Metabolism Disorders/genetics , Glucose Metabolism Disorders/metabolism , Liver/metabolism , Male , Membrane Proteins/genetics , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Endocrinology ; 162(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34379733

ABSTRACT

Kisspeptin, encoded by Kiss1, stimulates gonadotropin-releasing hormone neurons to govern reproduction. In female rodents, estrogen-sensitive kisspeptin neurons in the rostral anteroventral periventricular (AVPV) hypothalamus are thought to mediate estradiol (E2)-induced positive feedback induction of the preovulatory luteinizing hormone (LH) surge. AVPV kisspeptin neurons coexpress estrogen and progesterone receptors (PGRs) and are activated during the LH surge. While E2 effects on kisspeptin neurons have been well studied, progesterone's regulation of kisspeptin neurons is less understood. Using transgenic mice lacking PGR exclusively in kisspeptin cells (termed KissPRKOs), we previously demonstrated that progesterone action specifically in kisspeptin cells is essential for ovulation and normal fertility. Unlike control females, KissPRKO females did not generate proper LH surges, indicating that PGR signaling in kisspeptin cells is required for positive feedback. However, because PGR was knocked out from all kisspeptin neurons in the brain, that study was unable to determine the specific kisspeptin population mediating PGR action on the LH surge. Here, we used targeted Cre-mediated adeno-associated virus (AAV) technology to reintroduce PGR selectively into AVPV kisspeptin neurons of adult KissPRKO females, and tested whether this rescues occurrence of the LH surge. We found that targeted upregulation of PGR in kisspeptin neurons exclusively in the AVPV is sufficient to restore proper E2-induced LH surges in KissPRKO females, suggesting that this specific kisspeptin population is a key target of the necessary progesterone action for the surge. These findings further highlight the critical importance of progesterone signaling, along with E2 signaling, in the positive feedback induction of LH surges and ovulation.


Subject(s)
Hypothalamus, Anterior/metabolism , Luteinizing Hormone/metabolism , Neurons/metabolism , Receptors, Progesterone/physiology , Animals , Estradiol/pharmacology , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Female , Hypothalamus, Anterior/cytology , Hypothalamus, Anterior/drug effects , Kisspeptins/metabolism , Mice , Mice, Knockout , Neurons/drug effects , Progesterone/pharmacology , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Up-Regulation/drug effects
5.
Sci Rep ; 11(1): 8781, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888830

ABSTRACT

Obesity is implicated in cardiovascular disease and heart failure. When fatty acids are transported to and not adequately oxidized in cardiac cells, they accumulate, causing lipotoxicity in the heart. Since hepatic progesterone receptor membrane component 1 (Pgrmc1) suppressed de novo lipogenesis in a previous study, it was questioned whether cardiac Pgrmc1 protects against lipotoxicity. Hence, we focused on the role of cardiac Pgrmc1 in basal (Resting), glucose-dominant (Refed) and lipid-dominant high-fat diet (HFD) conditions. Pgrmc1 KO mice showed high FFA levels and low glucose levels compared to wild-type (WT) mice. Pgrmc1 KO mice presented low number of mitochondrial DNA copies in heart, and it was concomitantly observed with low expression of TCA cycle genes and oxidative phosphorylation genes. Pgrmc1 absence in heart presented low fatty acid oxidation activity in all conditions, but the production of acetyl-CoA and ATP was in pronounced suppression only in HFD condition. Furthermore, HFD Pgrmc1 KO mice resulted in high cardiac fatty acyl-CoA levels and TG level. Accordingly, HFD Pgrmc1 KO mice were prone to cardiac lipotoxicity, featuring high levels in markers of inflammation, endoplasmic reticulum stress, oxidative stress, fibrosis, and heart failure. In vitro study, it was also confirmed that Pgrmc1 enhances rates of mitochondrial respiration and fatty acid oxidation. This study is clinically important because mitochondrial defects in Pgrmc1 KO mice hearts represent the late phase of cardiac failure.


Subject(s)
Fatty Acids/metabolism , Membrane Proteins/physiology , Mitochondria/metabolism , Myocardium/metabolism , Receptors, Progesterone/physiology , Animals , Mice , Mice, Knockout , Oxidation-Reduction
6.
FASEB J ; 35(5): e21563, 2021 05.
Article in English | MEDLINE | ID: mdl-33818810

ABSTRACT

One of the endogenous estrogens, 17ß-estradiol (E2 ) is a female steroid hormone secreted from the ovary. It is well established that E2 causes biochemical and histological changes in the uterus. However, it is not completely understood how E2 regulates the oviductal environment in vivo. In this study, we assessed the effect of E2 on each oviductal cell type, using an ovariectomized-hormone-replacement mouse model, single-cell RNA-sequencing (scRNA-seq), in situ hybridization, and cell-type-specific deletion in mice. We found that each cell type in the oviduct responded to E2 distinctively, especially ciliated and secretory epithelial cells. The treatment of exogenous E2 did not drastically alter the transcriptomic profile from that of endogenous E2 produced during estrus. Moreover, we have identified and validated genes of interest in our datasets that may be used as cell- and region-specific markers in the oviduct. Insulin-like growth factor 1 (Igf1) was characterized as an E2 -target gene in the mouse oviduct and was also expressed in human fallopian tubes. Deletion of Igf1 in progesterone receptor (Pgr)-expressing cells resulted in female subfertility, partially due to an embryo developmental defect and embryo retention within the oviduct. In summary, we have shown that oviductal cell types, including epithelial, stromal, and muscle cells, are differentially regulated by E2 and support gene expression changes, such as growth factors that are required for normal embryo development and transport in mouse models. Furthermore, we have identified cell-specific and region-specific gene markers for targeted studies and functional analysis in vivo.


Subject(s)
Biomarkers/metabolism , Estradiol/pharmacology , Fallopian Tubes/physiology , Gene Expression Regulation, Developmental/drug effects , Insulin-Like Growth Factor I/physiology , Oviducts/physiology , Single-Cell Analysis/methods , Animals , Estrogens/pharmacology , Fallopian Tubes/cytology , Fallopian Tubes/drug effects , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oviducts/cytology , Oviducts/drug effects , Receptors, Progesterone/physiology
7.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33707208

ABSTRACT

Uterine contractile dysfunction leads to pregnancy complications such as preterm birth and labor dystocia. In humans, it is hypothesized that progesterone receptor isoform PGR-B promotes a relaxed state of the myometrium, and PGR-A facilitates uterine contraction. This hypothesis was tested in vivo using transgenic mouse models that overexpress PGR-A or PGR-B in smooth muscle cells. Elevated PGR-B abundance results in a marked increase in gestational length compared to control mice (21.1 versus 19.1 d respectively, P < 0.05). In both ex vivo and in vivo experiments, PGR-B overexpression leads to prolonged labor, a significant decrease in uterine contractility, and a high incidence of labor dystocia. Conversely, PGR-A overexpression leads to an increase in uterine contractility without a change in gestational length. Uterine RNA sequencing at midpregnancy identified 1,174 isoform-specific downstream targets and 424 genes that are commonly regulated by both PGR isoforms. Gene signature analyses further reveal PGR-B for muscle relaxation and PGR-A being proinflammatory. Elevated PGR-B abundance reduces Oxtr and Trpc3 and increases Plcl2 expression, which manifests a genetic profile of compromised oxytocin signaling. Functionally, both endogenous PLCL2 and its paralog PLCL1 can attenuate uterine muscle cell contraction in a CRISPRa-based assay system. These findings provide in vivo support that PGR isoform levels determine distinct transcriptomic landscapes and pathways in myometrial function and labor, which may help further the understanding of abnormal uterine function in the clinical setting.


Subject(s)
Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/genetics , Receptors, Oxytocin/genetics , Receptors, Progesterone/physiology , TRPC Cation Channels/genetics , Uterine Contraction/genetics , Animals , Female , Mice , Mice, Mutant Strains , Parturition/physiology , Pregnancy , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Transcriptome
8.
Am J Physiol Endocrinol Metab ; 320(3): E453-E466, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33427050

ABSTRACT

Progesterone acts directly on vascular smooth muscle cells (VSMCs) through activation of membrane progesterone receptor α (mPRα)-dependent signaling to rapidly decrease cytosolic Ca2+ concentrations and induce muscle relaxation. However, it is not known whether this progesterone action involves uptake of Ca2+ by the sarco/endoplasmic reticulum (SR) and increased sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity. The present results show that treatment of cultured human VSMCs with progesterone and the selective mPR agonist Org OD-02-0 (OD 02-0) but not with the nuclear PR agonist R5020 increased SERCA protein expression, which was blocked by knockdown of mPRα with siRNA. Moreover, treatments with progesterone and OD 02-0, but not with R5020, increased phospholamban (PLB) phosphorylation, which would result in disinhibition of SERCA function. Progesterone and OD 02-0 significantly increased Ca2+ levels in the SR and caused VSMC relaxation. These effects were blocked by pretreatment with cyclopiazonic acid (CPA), a SERCA inhibitor, and by knockdown of SERCA2 with siRNA, suggesting that SERCA2 plays a critical role in progesterone induction of VSMC relaxation. Treatment with inhibitors of inhibitory G proteins (Gi, NF023), MAP kinase (AZD 6244), Akt/Pi3k (wortmannin), and a Rho activator (calpeptin) blocked the progesterone- and OD 02-0-induced increase in Ca2+ levels in the SR and SERCA expressions. These results suggest that the rapid effects of progesterone on cytosolic Ca2+ levels and relaxation of VSMCs through mPRα involve regulation of the functions of SERCA2 and PLB through Gi, MAP kinase, and Akt signaling pathways and downregulation of RhoA activity.NEW & NOTEWORTHY The rapid effects of progesterone on cytosolic Ca2+ levels and relaxation of VSMCs through mPRα involve regulation of the functions of SERCA2 and PLB through Gi, MAP kinase, and Akt signaling pathways and downregulation of RhoA activity.


Subject(s)
Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Progesterone/pharmacology , Receptors, Progesterone/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Humans , Muscle Relaxation/genetics , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Umbilical Arteries/cytology , Umbilical Arteries/drug effects , Umbilical Arteries/metabolism
9.
Horm Behav ; 127: 104887, 2021 01.
Article in English | MEDLINE | ID: mdl-33166560

ABSTRACT

Progesterone receptor (PR) is expressed in Cajal-Retzius (CR) cells of the dentate gyrus (DG) molecular layer during the postnatal period (P1-28), a critical stage of development for the dentate gyrus and its circuitry. CR cells secrete the glycoprotein, reelin, which is required for typical development of the DG and its connections, particularly afferent input from the perforant path. This pathway regulates the processing of sensory information arriving from entorhinal cortex and integrates this information to form episodic memories. To assess the potential role of PR activity on the development of these connections and associated behavior, rats were treated daily from P1 to 7 with the PR antagonist, RU486. RU486 treatment increased the number of reelin-ir cells, suggesting an accumulation of reelin, and implicating PR in the regulation of a principle developmental function of CR cells. RU486 also altered the synaptic bouton marker, synaptophysin-ir, in a sex-specific manner, suggesting a role for PR activity in the development of perforant path innervation of the molecular layer (MOL). Finally, both control and RU486 treated rats spent significantly more time with a temporally distant object in the Relative Recency task, suggesting an intact associative memory for object identity and temporal order in both groups. In contrast, the same RU486 treated rats were impaired in an episodic-like memory task compared to controls, failing to integrate object identity ('what'), time ('when'), and object position ('where'). These findings reveal a novel role for PR in regulating CR cell function within the MOL, thereby altering development of DG connectivity and behavioral function.


Subject(s)
Dentate Gyrus/drug effects , Memory, Episodic , Mifepristone/pharmacology , Receptors, Progesterone/antagonists & inhibitors , Synaptic Transmission/drug effects , Age Factors , Animals , Animals, Newborn , Cell Adhesion Molecules, Neuronal/metabolism , Dentate Gyrus/physiology , Extracellular Matrix Proteins/metabolism , Female , Hippocampus/drug effects , Hippocampus/metabolism , Immunohistochemistry , Inhibition, Psychological , Male , Nerve Tissue Proteins/metabolism , Progesterone/metabolism , Rats , Receptors, Progesterone/metabolism , Receptors, Progesterone/physiology , Reelin Protein , Serine Endopeptidases/metabolism , Synaptic Transmission/physiology
10.
Domest Anim Endocrinol ; 74: 106527, 2021 01.
Article in English | MEDLINE | ID: mdl-32799038

ABSTRACT

The steroid hormones 17-ß estradiol (E2) and progesterone (P4) can regulate capacitation, hyperactive motility, and the acrosome reaction (AR) during the sperm transit through the female tract. Moreover, exogenous P4 and E2 can induce the AR in ovine spermatozoa, and progesterone receptor (PR) and estrogen receptors (ERα and ERß) are present in these cells. Thus, to investigate whether the effects both steroid hormones in ram sperm capacitation and AR are receptor-mediated, we incubated them with receptor agonists (tanaproget 1 µM and 5 µM for PR or resveratrol 5 µM and 10 µM for ER) or antagonists (mifepristone 4 µM and 40 µM for PR or tamoxifen 5 µM and 10 µM for ER) in capacitating conditions. The addition of receptor modulators did not affect sperm viability or total motility, although changes in progressive motility were detected. The incubation with both receptor agonists increased the percentage of acrosome-reacted spermatozoa, evaluated by chlortetracycline staining, when compared with the capacitated nontreated sample (Cap-C, P < 0.001). Moreover, the ER agonist resveratrol 10 µM provoked a greater AR than E2 (P < 0.01). Furthermore, the incubation with the receptor antagonists prevented the induction of the AR by P4 or E2, as the antagonists-treated spermatozoa presented a similar CTC pattern to that of Cap-C. In conclusion, these results confirm that P4 and E2 can induce the AR in ram spermatozoa and that this effect is receptor-mediated.


Subject(s)
Acrosome Reaction/drug effects , Acrosome Reaction/physiology , Receptors, Estrogen/physiology , Receptors, Progesterone/physiology , Spermatozoa/physiology , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Cell Survival , Cells, Cultured , Estradiol/pharmacology , Estrogens/pharmacology , Male , Progesterone/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Resveratrol/pharmacology , Sheep , Sperm Capacitation/drug effects , Sperm Capacitation/physiology , Sperm Motility , Tamoxifen/pharmacology
11.
Br J Cancer ; 123(8): 1326-1335, 2020 10.
Article in English | MEDLINE | ID: mdl-32704174

ABSTRACT

BACKGROUND: Increased expression of the progesterone receptor membrane component 1 (PGRMC1) has been linked to multiple cancers, including breast cancer. Despite being a regulatory receptor and a potential therapeutic target, the oncogenic potential of PGRMC1 has not been studied. METHODS: The impact of PGRMC1 on breast cancer growth and progression was studied following chemical inhibition and alteration of PGRMC1 expression, and evaluated by using online-based gene expression datasets of human breast cancer tissue. MTS, flow cytometry, qPCR, Western blotting, confocal microscopy and phosphoproteome analysis were performed. RESULTS: We observed higher PGRMC1 levels in both ER-positive ZR-75-1 and TNBC MDA-MB-468 cells. Both chemical inhibition and silencing decreased cell proliferation, induced cell-cycle arrest, promoted apoptosis and reduced the migratory and invasive capabilities of ZR-75-1 and MDA-MB-468 cells. Further, phosphoproteome analysis demonstrated an overall decrease in activation of proteins involved in PI3K/AKT/mTOR and EGFR signalling pathways. In contrast, overexpression of PGRMC1 in non-malignant MCF10A cells resulted in increased cell proliferation, and enhanced activity of PI3K/AKT/mTOR and EGFR signalling pathways. CONCLUSIONS: Our data demonstrate that PGRMC1 plays a prominent role in regulating the growth of cancer cells by altering the PI3K/AKT/mTOR and EGFR signalling mechanisms in both ER-positive and TNBC cells.


Subject(s)
Breast Neoplasms/pathology , Membrane Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphoproteins/metabolism , Proteome , Proto-Oncogene Proteins c-akt/physiology , Receptors, Progesterone/physiology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , ErbB Receptors/physiology , Female , Humans , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology
12.
Cell Rep ; 31(2): 107496, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294429

ABSTRACT

Ovulation is triggered by the gonadotropin surge that induces the expression of two key genes, progesterone receptor (Pgr) and prostaglandin-endoperoxide synthase 2 (Ptgs2), in the granulosa cells of preovulatory follicles. Their gene products PGR and PTGS2 activate two separate pathways that are both essential for successful ovulation. Here, we show that the PGR plays an additional essential role: it attenuates ovulatory inflammation by diminishing the gonadotropin surge-induced Ptgs2 expression. PGR indirectly terminates Ptgs2 expression and PGE2 synthesis in granulosa cells by inhibiting the nuclear factor κB (NF-κB), a transcription factor required for Ptgs2 expression. When the expression of PGR is ablated in granulosa cells, the ovary undergoes a hyperinflammatory condition manifested by excessive PGE2 synthesis, immune cell infiltration, oxidative damage, and neoplastic transformation of ovarian cells. The PGR-driven termination of PTGS2 expression may protect the ovary from ovulatory inflammation.


Subject(s)
Ovary/metabolism , Ovulation/metabolism , Receptors, Progesterone/physiology , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Granulosa Cells/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Ovarian Follicle/metabolism , Progesterone/genetics , Progesterone/metabolism , RNA, Messenger/genetics , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Transcription Factors/metabolism
13.
Front Endocrinol (Lausanne) ; 11: 552805, 2020.
Article in English | MEDLINE | ID: mdl-33505354

ABSTRACT

Progestogens' (e.g., progesterone and its neuroactive metabolite, allopregnanolone), cognitive effects and mechanisms among males are not well-understood. We hypothesized if progestogen's effects on cognitive performance are through its metabolite allopregnanolone, and not actions via binding to traditional progestin receptors (PRs), then progesterone administration would enhance performance in tasks mediated by the hippocampus and cortex, coincident with increasing allopregnanolone concentrations, brain derived neurotrophic factor (BDNF) and/or muscimol binding of PR knock out (PRKO) and wild-type PR replete mice. Experiment 1: Progesterone (4 mg/kg, subcutaneously (SC; n = 12/grp), or oil vehicle control, was administered to gonadally-intact adult male mice PRKO mice and their wild-type counterparts and cognitive behaviors in object recognition, T-maze and water maze was examined. Progesterone, compared to vehicle, when administered post-training increased time investigating novel objects by the PRKO and wild-type mice in the object recognition task. In the T-maze task, progesterone administration to wild-type and PRKO mice had significantly greater number of spontaneous alternations compared to their vehicle-administered counterparts. In the water maze task, PRKO mice administered vehicle spent significantly fewer seconds in the quadrant associated with the escape platform on testing compared to all other groups. Experiment 2: Progesterone administered to wild-type and PRKO mice increased plasma progesterone and allopregnanolone levels (n = 5/group). PRKO mice had higher allopregnanolone levels in plasma and hippocampus, but not cortex, when administered progesterone and compared to wild-type mice. Experiment 3: Assessment of PR binding revealed progesterone administered wild-type mice had significantly greater levels of PRs in the hippocampus and cortex, compared to all other groups (n = 5/group). Wild-type mice administered progesterone, but not vehicle, had increased BDNF levels in the hippocampus, but not the cortex, compared to PRKOs. Wild-type as well as PRKO mice administered progesterone experienced significant increases in maximal GABAA agonist, muscimol, binding in hippocampus and cortex, compared to their vehicle-administered counterparts. Thus, adult male mice can be responsive to progesterone for cognitive performance, and such effects may be independent of PRs trophic actions of BDNF levels in the hippocampus and/or increases in GABAA activity in the hippocampus and cortex.


Subject(s)
Cerebral Cortex/physiology , Hippocampus/physiology , Maze Learning/physiology , Progesterone/administration & dosage , Receptors, GABA-A/physiology , Receptors, Progesterone/physiology , Spatial Memory/physiology , Animals , Brain-Derived Neurotrophic Factor/physiology , GABA-A Receptor Agonists/administration & dosage , Male , Mice, Knockout , Muscimol/administration & dosage , Receptors, Progesterone/genetics
14.
Fukushima J Med Sci ; 65(2): 30-36, 2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31189789

ABSTRACT

Although eye diseases are considered to be relatively less affected by patient sex, it is noteworthy that the presence of hormone receptors have been confirmed in various ocular tissues, which were considered to have few sex-based differences. Female hormone levels are known to change because of menstruation, pregnancy, and menopause. When female hormone levels markedly fluctuate in such situations, the disease state may change.The fluctuations in the levels of sex hormones affect the corneal thickness conditions of uveitis.Estrogen may be a possible therapeutic option for glaucoma because it protects the eyes from damage caused by glaucoma and reduces intraocular pressure;it is particularly promising in the treatment of postmenopausal women with glaucoma.Estrogen is considered to have a prophylactic effect against eye diseases. However, there is a report that female sex is an independent risk factor for the progression of diabetic retinopathy, so it may not always exert a prophylactic effect. Thus, caution should be exercised.Based on recent progression of studies on this field, the importance of treatment according to gender has been recognized in the treatment of eye diseases.


Subject(s)
Eye Diseases/therapy , Diabetic Retinopathy/etiology , Diabetic Retinopathy/therapy , Eye Diseases/etiology , Female , Glaucoma/etiology , Glaucoma/therapy , Gonadal Steroid Hormones/physiology , Humans , Male , Receptors, Estrogen/physiology , Receptors, Progesterone/physiology , Sex Characteristics , Uveitis/etiology , Uveitis/therapy
15.
Exp Cell Res ; 382(1): 111433, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31100306

ABSTRACT

Autophagy is an evolutionary conserved, self-eating process that targets cellular constituents for lysosomal degradation. Transcription factor EB (TFEB) is a master regulator of autophagy by inducing the expression of genes involved in autophagic and lysosomal degradation. In breast cancer, ligand-activated progesterone receptor has been reported to influence cancer development by manipulating the autophagy pathway. However, understanding of the mechanism that underlies this autophagic response remains limited. Herein, we report that prolonged treatment with progestin R5020 upregulates autophagy in MCF-7 human breast cancer cells via a novel interplay between progesterone receptor B (PRB) and TFEB. R5020 upregulates TFEB gene expression and protein levels in a PRB-dependent manner. Additionally, R5020 enhances the co-recruitment of PRB and TFEB to each other to facilitate TFEB nuclear localization. Once in the nucleus, TFEB induces the expression of autophagy and lysosomal genes to potentiate autophagy. Together, our findings highlight a novel functional connection between ligand-activated PRB and TFEB to modulate autophagy in MCF-7 breast cancer cells. As breast cancer development is controlled by autophagy, the progestin-PRB-TFEB transduction pathway warrants future attention as a potential therapeutic target in cancer therapy.


Subject(s)
Adenocarcinoma/genetics , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Breast Neoplasms/genetics , Neoplasm Proteins/physiology , Receptors, Progesterone/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Autophagosomes/metabolism , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysosomes/metabolism , MCF-7 Cells , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Promegestone/pharmacology , Protein Interaction Mapping , Protein Transport/drug effects , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Transcriptional Activation
16.
Respir Physiol Neurobiol ; 263: 55-61, 2019 05.
Article in English | MEDLINE | ID: mdl-30880277

ABSTRACT

The steroid hormone progesterone is well-known for its role in neuroprotection, in the pre- and postnatal brain development, and is also recognized as a potent respiratory stimulant that reduces the frequency of sleep apnea in adult female subjects. Over the past few years, we have used newborn rats or mice to provide convincing evidence that the respiratory effect of progesterone involves a balance between excitation mediated by progesterone receptors, and an inhibition due to the fast conversion of progesterone to allopregnanolone, a positive allosteric modulator of GABAA receptors. This review focuses on the sex- and age- specific roles of nuclear and membrane progesterone receptors (nPR or mPR), and highlight the clinical potential of these receptors for the treatment of apnea of prematurity. We present original data showing that in newborn rats, selective nPR or mPR agonists are more efficient to reduce apnea frequency at postnatal days 12 than at postnatal day 1, and appear more efficient in males than in females. Furthermore, new results obtained by using intra-cisternal injection of specific siRNA targeting mPRα, mPRß (two mPR with high brain expression) or nPR suggest that mPRß regulates the stability of the breathing pattern in males, while effects of nPR appear in females. While several important questions remain to be addressed before a safe clinical use could be proposed, these results highlight the potential role of these drugs as complementary, and sex-specific tools for the treatment of apnea in preterm neonates.


Subject(s)
Animals, Newborn/physiology , Apnea/metabolism , Progesterone/physiology , Receptors, Progesterone/physiology , Respiration , Sex Characteristics , Animals , Animals, Newborn/metabolism , Female , Male , Progesterone/metabolism , Rats , Receptors, Progesterone/metabolism
17.
Reprod Sci ; 26(5): 690-696, 2019 05.
Article in English | MEDLINE | ID: mdl-30654718

ABSTRACT

The prepartum transition from a soft to ripening cervix is an inflammatory process that occurs well before birth when systemic progesterone is near peak concentration. This 2-part study first determined that stromal fibroblasts but not macrophages in the cervix have progesterone receptors (PRs). Neither the number of PR cells in cervix sections nor the relative abundance or ratio of nuclear PR isoforms (PR-A/PR-B) were diminished in mice between day 15 of pregnancy and term. Second in mice lacking PR-B ( Pgrtm20mc), the number of cells that expressed the PR-A isoform were maintained during this period of prepartum cervix remodeling. Thus, progesterone effects to sustain pregnancy, as well as soften and ripen the cervix, are mediated by a stable stromal cell population that expresses PR-A and, through interactions with resident macrophages, are likely to mediate inflammatory ripening processes in preparation for birth.


Subject(s)
Cervical Ripening , Cervix Uteri/physiology , Progesterone/physiology , Receptors, Progesterone/physiology , Stromal Cells/physiology , Animals , Female , Macrophages/physiology , Mice , Mice, Knockout , Pregnancy , Protein Isoforms/physiology , Receptors, Progesterone/genetics
18.
J Pathol ; 248(1): 77-87, 2019 05.
Article in English | MEDLINE | ID: mdl-30632164

ABSTRACT

Previous studies revealed the increasing risk of tubal pregnancy following failure of levonorgestrel (LNG)-induced emergency contraception, which was attributed to the reduced ciliary motility in response to LNG. However, understanding of the mechanism of LNG-induced reduction in the ciliary beat frequency (CBF) is limited. The transient receptor potential vanilloid (TRPV) 4 channel is located widely in the female reproductive tract and generates an influx of Ca2+ following its activation under normal physiological conditions, which regulates the CBF. The present study aimed to explore whether LNG reduced the CBF in the Fallopian tubes by modulating TRPV4 channels, leading to embryo retention in the Fallopian tubes and subsequent tubal pregnancy. The study provided evidence that the expression of TRPV4 was downregulated in the Fallopian tubes among patients with tubal pregnancy and negatively correlated with the serum level of progesterone. LNG downregulated the expression of TRPV4, limiting the calcium influx to reduce the CBF in mouse oviducts. Furthermore, the distribution of ciliated cells and the morphology of cilia did not change following the administration of LNG. LNG-induced reduction in the CBF and embryo retention in the Fallopian tubes and in mouse oviducts were partially reversed by the progesterone receptor antagonist RU486 or the TRPV4 agonist 4α-phorbol 12,13-didecanoate (4α-PDD). The results indicated that LNG could downregulate the expression of TRPV4 to reduce the CBF in both humans and mice, suggesting the possible mechanism of tubal pregnancy. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Contraceptives, Postcoital/adverse effects , Levonorgestrel/adverse effects , Oviducts/drug effects , Pregnancy, Tubal/chemically induced , TRPV Cation Channels/physiology , Animals , Calcium/metabolism , Cell Line , Cilia/drug effects , Cilia/physiology , Cilia/ultrastructure , Contraception, Postcoital/adverse effects , Contraceptive Agents, Hormonal/adverse effects , Contraceptive Agents, Hormonal/pharmacology , Contraceptive Effectiveness , Contraceptives, Postcoital/pharmacology , Down-Regulation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fallopian Tubes/drug effects , Fallopian Tubes/metabolism , Female , Humans , Levonorgestrel/pharmacology , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Oviducts/physiopathology , Oviducts/ultrastructure , Pregnancy , Pregnancy, Tubal/metabolism , Pregnancy, Tubal/physiopathology , Progesterone/blood , Receptors, Progesterone/physiology , TRPV Cation Channels/biosynthesis
19.
Reproduction ; 156(4): 365-373, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30306772

ABSTRACT

To determine whether conditional depletion of progesterone receptor membrane component (PGRMC) 1 and PGRMC2 affected ovarian follicle development, follicle distribution was assessed in ovaries of young (≈3-month-old) and middle-aged (≈6-month-old) control (Pgrmc1/2fl/fl) and double conditional PGRMC1/2-knockout (Pgrmc1/2d/d) mice. This study revealed that the distribution of primary, preantral and antral follicles was not altered in Pgrmc1/2d/d mice, regardless of the age. Although the number of primordial follicles was similar at ≈3 months of age, their numbers were reduced by ≈80% in 6-month-old Pgrmc1/2d/d mice compared to age-matched Pgrmc1/2fl/fl mice. The Pgrmc1/2d/d mice were generated using Pgr-cre mice, so ablation of Pgrmc1 and Pgrmc2 in the ovary was restricted to peri-ovulatory follicles and subsequent corpora lutea (CL). In addition, the vascularization of CL was attenuated in Pgrmc1/2d/d mice, although mRNA levels of vascular endothelial growth factor A (Vegfa) were elevated. Moreover, depletion of Pgrmc1 and Pgrmc2 altered the gene expression profile in the non-luteal component of the ovary such that Vegfa expression, a stimulator of primordial follicle growth, was elevated; Kit Ligand expression, another stimulator of primordial follicle growth, was suppressed and anti-Mullerian hormone, an inhibitor of primordial follicle growth, was enhanced compared to Pgrmc1/2fl/fl mice. These data reveal that luteal cell depletion of Pgrmc1 and 2 alters the expression of growth factors within the non-luteal component of the ovary, which could account for the premature demise of the adult population of primordial follicles. In summary, the survival of adult primordial follicles is dependent in part on progesterone receptor membrane component 1 and 2.


Subject(s)
Membrane Proteins/physiology , Ovarian Follicle/physiology , Receptors, Progesterone/physiology , Age Factors , Animals , Corpus Luteum/blood supply , Female , Mice , Mice, Knockout , Ovarian Follicle/cytology
20.
Genesis ; 56(8): e23223, 2018 08.
Article in English | MEDLINE | ID: mdl-30004627

ABSTRACT

Using a Rosa26 gene targeting strategy in mouse embryonic stem cells, we have generated a new transgenic mouse (Pgr-B LSL ), which is designed to conditionally express the epitope-tagged mouse progesterone receptor-B (PGR-B) isoform when crossed with a specific cre driver mouse. To functionally validate this transgenic mouse, we crossed the Pgr-B LSL mouse with the MMTV-CREA transgenic mouse to create the MMTV-CREA/Pgr-B LSL bigenic (termed PR-B:OE to denote PGR-B overexpressor). As expected, transgene-derived PGR-B protein was specifically targeted to the virgin mammary gland epithelium. At a functional level, the PR-B:OE bigenic exhibited abnormal mammary morphogenesis-dilated epithelial ducts, precocious alveologenesis and lateral side-branching, along with a prominent proliferative signature-that resulted in pregnant PR-B:OE mice unable to exhibit mammary gland terminal differentiation at parturition. Because of this developmental failure, the PR-B:OE mammary gland was incapable of producing milk resulting in early neonatal death of otherwise healthy litters. This first line of analysis demonstrates the utility of the Pgr-B LSL mouse to examine the role of the PGR-B isoform in different physiologic and pathophysiologic systems that are responsive to progesterone.


Subject(s)
Genetic Engineering/methods , Receptors, Progesterone/genetics , Animals , Cell Proliferation , Epithelial Cells/metabolism , Epithelium/metabolism , Female , Male , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Models, Animal , Morphogenesis/genetics , Protein Isoforms , Receptors, Progesterone/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...