Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 157
1.
J Neuroendocrinol ; 32(3): e12836, 2020 03.
Article En | MEDLINE | ID: mdl-32062869

The present study aimed to determine whether an i.c.v. administration of allopregnanolone (ALLO) rapidly modifies the hypothalamic and ovarian 3ß-hydroxysteroid dehydrogenase (3ß-HSD) enzymatic activity and gene expression in in vivo and ex vivo systems in pro-oestrus (PE) and dioestrus I (DI) rats. Animals were injected with vehicle, ALLO, bicuculline or bicuculline plus ALLO and were then killed. In the in vivo experiment, the hypothalamus, ovaries and serum were extracted and analysed. In the ex vivo experiment, the superior mesenteric ganglion - ovarian nerve plexus - ovary system was extracted and incubated during 120 minutes at 37 ºC. The serum and ovarian compartment fluids were used to determine progesterone by radioimmunoanalysis. In the in vivo experiments, ALLO caused a decrease in hypothalamic and ovarian 3ß-HSD enzymatic activity during PE. During DI, ALLO increased hypothalamic and ovarian 3ß-HSD activity and gene expression. The ovarian 3ß-HSD activity increased in both stages in the ex vivo system; gene expression increased only during DI. ALLO induced an increase in serum progesterone only in D1 and in the ovarian incubation liquids in both stages. All findings were reversed by an injection of bicuculline before ALLO. Ovarian steroidogenic changes could be attributed to signals coming from ganglion neurones, which are affected by the acute central neurosteroid stimulation. The i.c.v. administration of ALLO via the GABAergic system altered 3ß-HSD activity and gene expression, modulating the neuroendocrine axis. The present study reveals the action that ALLO exerts on the GABAA receptor in both the central and peripheral nervous system and its relationship with hormonal variations. ALLO is involved in the "fine tuning" of neurosecretory functions as a potent modulator of reproductive processes in female rats.


3-Hydroxysteroid Dehydrogenases/metabolism , Hypothalamus/drug effects , Neurosteroids/administration & dosage , Ovary/drug effects , Pregnanolone/administration & dosage , Animals , Diestrus/drug effects , Diestrus/metabolism , Female , Gene Expression/drug effects , Hypothalamus/enzymology , Injections, Intraventricular , Ovary/metabolism , Proestrus/drug effects , Proestrus/metabolism , Progesterone/blood , Rats
2.
Exp Physiol ; 104(8): 1179-1189, 2019 08.
Article En | MEDLINE | ID: mdl-31241201

NEW FINDINGS: What is the central question of this study? What is the role of the nicotinic system of the suprachiasmatic nucleus (SCN) in the regulation of follicular growth and ovulation? What is the main finding and its importance? The stimulation of the nicotinic system of the pro-oestrus rat SCN results in an increase in the number of ova shed, in the number of growing ovarian follicles and in the secretion of oestradiol. ABSTRACT: The timing of the preovulatory luteinizing hormone surge that leads to ovulation depends to a large extent on a functional circadian clock that is localized in the suprachiasmatic nucleus (SCN). The activities of the SCN are regulated by several neurotransmitter systems, including the muscarinic system. Given that acetylcholine binds to muscarinic (mAChRs) and nicotinic (nAChRs) receptors, in the present study, we analysed the effects of unilaterally stimulating nAChRs in the left or right SCN. Stimulation treatment was administered in rats in pro-oestrus at 09.00 or 19.00 h by injecting 0.3 µl of a nicotine solution (200 µm). The effects of the stimulation were assessed by evaluating the number of ova shed, the number of ovarian follicles, and the levels of oestradiol and progesterone in serum 24 h after treatment. We observed that regardless of the time (4 h after lights on, 09.00 h, or immediately after lights off, 19.00 h) or the side of the SCN treated, the unilateral microinjection of nicotine resulted in a higher number of ova shed and higher number of growing follicles in the ovaries as well as higher oestradiol serum levels. When the nicotine microinjection treatment failed to reach the SCN, the oestradiol levels in serum were similar to those of animals treated with vehicle solution. Based on the current results, we suggest that during pro-oestrus, the nicotinic neuronal information in the SCN modulates follicular growth and ovulation in a stimulatory manner.


Ovarian Follicle/metabolism , Ovary/metabolism , Receptors, Nicotinic/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Estradiol/metabolism , Estrus/metabolism , Female , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/metabolism , Ovulation/metabolism , Proestrus/metabolism , Progesterone/metabolism , Rats
3.
J Steroid Biochem Mol Biol ; 185: 225-236, 2019 01.
Article En | MEDLINE | ID: mdl-30227242

Visfatin is an adipokine which has an endocrine effect on reproductive functions and regulates ovarian steroidogenesis. There is scant information about the expression, regulation, and functions of visfatin in the mammalian uterus. The present study examined expression and localization of visfatin in the mouse uterus at various stages of the natural estrous cycle, effects of estrogen and progesterone on localization and expression of visfatin in the ovariectomised mouse uterus and effect of visfatin inhibition by a specific inhibitor, FK866 on proliferation and apoptosis in the uterus. Western blot analysis of visfatin showed high expression in proestrus and metestrus while it declined in estrus and diestrus. Immulocalization study also showed strong immunostaining in the cells of endometrium, myometrium, luminal and glandular epithelium during proestrus and metestrus that estrus and diestrus. The uterine visfatin expression closely related to the increased estrogen levels in proestrus and suppressed when progesterone rose to a high level in diestrus. The treatment with estrogen to ovariectomised mice up-regulates visfatin, PCNA, and active caspase3 whereas progesterone up-regulates PCNA and down-regulates visfatin and active caspase3 expression in mouse uterus. The co-treatment with estrogen and progesterone up-regulates visfatin and down-regulates PCNA and active caspase3. In vitro study showed endogenous visfatin inhibition by FK866 increased expression of PCNA and BCL2 increased catalase activity while FK866 treatment decreased expression of active caspase3 and BAX with decreased SOD and GPx activity. BrdU labeling showed that inhibition of visfatin modulates the uterine proliferation. This study showed that expression of visfatin protein is steroid dependent in mouse uterus which is involved in the regulation of proliferation and apoptosis via modulating antioxidant system in the uterus of mice during the reproductive cycle.


Apoptosis/physiology , Cell Proliferation/physiology , Endometrium/metabolism , Estrogens/metabolism , Estrous Cycle/metabolism , Myometrium/metabolism , Nicotinamide Phosphoribosyltransferase/biosynthesis , Progesterone/metabolism , Acrylamides/pharmacology , Animals , Caspase 3/biosynthesis , Catalase/biosynthesis , Diestrus/metabolism , Estrus/metabolism , Female , Glutathione Peroxidase/biosynthesis , Mice , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Piperidines/pharmacology , Proestrus/metabolism , Proliferating Cell Nuclear Antigen/biosynthesis , Superoxide Dismutase/biosynthesis , bcl-2-Associated X Protein/biosynthesis
4.
eNeuro ; 5(4)2018.
Article En | MEDLINE | ID: mdl-30079374

Surge release of gonadotropin-releasing hormone (GnRH) is essential in the activation of pituitary gonadal unit at proestrus afternoon preceded by the rise of serum 17ß-estradiol (E2) level during positive feedback period. Here, we describe a mechanism of positive estradiol feedback regulation acting directly on GnRH-green fluorescent protein (GFP) neurons of mice. Whole-cell clamp and loose patch recordings revealed that a high physiological dose of estradiol (200 pM), significantly increased firing rate at proestrus afternoon. The mPSC frequency at proestrus afternoon also increased, whereas it decreased at metestrus afternoon and had no effect at proestrus morning. Inhibition of the estrogen receptor ß (ERß), intracellular blockade of the Src kinase and phosphatidylinositol 3 kinase (PI3K) and scavenge of nitric oxide (NO) inside GnRH neurons prevented the facilitatory estradiol effect indicating involvement of the ERß/Src/PI3K/Akt/nNOS pathway in this fast, direct stimulatory effect. Immunohistochemistry localized soluble guanylate cyclase, the main NO receptor, in both glutamatergic and GABAergic terminals innervating GnRH neurons. Accordingly, estradiol facilitated neurotransmissions to GnRH neurons via both GABAA-R and glutamate/AMPA/kainate-R. These results indicate that estradiol acts directly on GnRH neurons via the ERß/Akt/nNOS pathway at proestrus afternoon generating NO that retrogradely accelerates GABA and glutamate release from the presynaptic terminals contacting GnRH neurons. The newly explored mechanism might contribute to the regulation of the GnRH surge, a fundamental prerequisite of the ovulation.


Estradiol/metabolism , Glutamic Acid/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Proestrus/metabolism , Signal Transduction/physiology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
J Cell Physiol ; 233(9): 6591-6603, 2018 09.
Article En | MEDLINE | ID: mdl-29115663

Annexin A1 (AnxA1) is a glucocorticoid-regulated anti-inflammatory protein secreted by phagocytes and other specialised cells. In the endocrine system, AnxA1 controls secretion of steroid hormones and it is abundantly expressed in the testis, ovaries, placenta and seminal fluid, yet its potential modulation of fertility has not been described. Here, we observed that AnxA1 knockout (KO) mice delivered a higher number of pups, with a higher percentage of female offsprings. This profile was not dependent on the male features, as sperm from KO male mice did not present functional alterations, and had an equal proportion of Y and X chromosomes, comparable to wild type (WT) male mice. Furthermore, mismatched matings of male WT mice with female KO yielded a higher percentage of female pups per litter, a phenomenon which was not observed when male KO mice mated with female WT animals. Indeed, AnxA1 KO female mice displayed several differences in parameters related to gestation including (i) an arrested estrous cycle at proestrus phase; (ii) increased sites of implantation; (iii) reduced pre- and post-implantation losses; (iv) exacerbated features of the inflammatory reaction in the uterine fluid during implantation phase; and (v) enhanced plasma progesterone in the beginning of pregnancy. In summary, herein we highlight that AnxA1 pathway as a novel determinant of fundamental non-redundant regulatory functions during early pregnancy.


Annexin A1/metabolism , Embryo Implantation/physiology , Animals , Estrous Cycle/metabolism , Estrous Cycle/physiology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Animal , Pregnancy , Proestrus/metabolism , Proestrus/physiology , Sex Ratio , Uterus/metabolism , Uterus/physiology , X Chromosome/metabolism , X Chromosome/physiology , Y Chromosome/metabolism , Y Chromosome/physiology
6.
Theriogenology ; 93: 71-77, 2017 Apr 15.
Article En | MEDLINE | ID: mdl-28257870

The aim of this study was to determine the mRNA LHR and LHR protein expression pattern in the canine ovarian follicles at different stage of development throughout the estrous cycle. Dog ovaries were obtained from 1-6y bitches at proestrus/estrus, anestrus and diestrus stages following ovariohysterectomy. Follicular cells were mechanically recovered from follicles distributed into four types (preantral, small antral, medium antral and large antral). Total RNA extraction was performed and the evaluation of gene expression levels was achieved by relative quantification q-PCR analysis. Intrafollicular amounts of LHR were assessed by western blot method. All results were evaluated by ANOVA. The expression levels of mRNA LHR in follicular cells were observed in every stage of development, however this gene expression varied over the estrous cycle. LHR transcripts increased (P < 0.05) from preantral to antral stage. There were not differences in LHR gene expression among follicles at preantral stages; however, at antral stages the lowest (P < 0.05) LHR mRNA expression was found at anestrus and the highest (P < 0.05) at proestrus/estrus. The LHR protein was also detected in dog follicles in all reproductive phases with patterns varying with stage of follicular development over the reproductive cycle. The antibody against human LHR revealed two bands at ∼90 and ∼67 kDa, probably representing the matured protein and its precursor respectively. Both bands LHR appeared already at preantral follicles increasing (P < 0.05) with growth. A high proportion of LHR was presented as immature forms in all follicles stages during different phases of the estrous cycle. In conclusion, the gene and protein of LHR are differentially expressed in dog follicles over the estrous cycle, increasing with growth and the precursor protein is the most predominant LHR form present in canine follicles.


Dogs/metabolism , Estrous Cycle/metabolism , Ovarian Follicle/chemistry , Receptors, LH/analysis , Receptors, LH/genetics , Anestrus/metabolism , Animals , Blotting, Western/veterinary , Diestrus/metabolism , Estrus/metabolism , Female , Gene Expression , Hysterectomy/veterinary , Ovariectomy/veterinary , Proestrus/metabolism , RNA, Messenger/analysis
7.
Morphologie ; 101(332): 39-46, 2017 Mar.
Article En | MEDLINE | ID: mdl-27746040

Tanycytes are special ependymal cells located in the ventrolateral wall and floor of the third ventricle having processes extending nuclei that regulate reproductive functions and around of vessels in median eminance. The aquaporins (AQPs) are a family of transmembrane proteins that transport water and glycerol. AQP-7 and -9 are permeable to other small molecules as glycerol and therefore called aquaglyceroporins. In this study, we aimed to show localization of AQP-7 and -9 in epithelial cells of choroid plexus and tanycytes during female mouse estrus cycle. AQP-7 and -9 proteins were detected in α2 and ß1 tanycytes in prœstrus stage. Interestingly, there is no staining in estrus stage in any type of tanycytes. We observed weak immunoreactivity in α1, α2 and ß1 tanycyte cells in metestrus stage for AQP-7 and α1 for AQP-9 protein. AQP-7 and -9 showed intense immunoreactivity in α2, ß1 and ß2 tanycyte cells during diestrus stage. Consequently, AQP-7 and -9 showed differential staining pattern in different stages of mouse estrus cycle. In the light of our findings and other recent publications, we suggest that AQP-7 and -9-mediated glycerol transport in tanycyte cells might be under hormonal control to use glycerol as a potential energy substrate during mouse estrus cycle.


Aquaporins/metabolism , Choroid Plexus/metabolism , Ependymoglial Cells/metabolism , Estrous Cycle/metabolism , Animals , Biological Transport , Choroid Plexus/cytology , Epithelial Cells/metabolism , Estrus/metabolism , Female , Glycerol/metabolism , Metestrus/metabolism , Mice , Mice, Inbred BALB C , Proestrus/metabolism , Third Ventricle/cytology
8.
Reprod Fertil Dev ; 29(10): 1971-1981, 2017 Sep.
Article En | MEDLINE | ID: mdl-27997334

Kisspeptin is crucial for the generation of the circadian-gated preovulatory gonadotrophin-releasing hormone (GnRH)-LH surge in female rodents, with expression in the anteroventral periventricular nucleus (AVPV) peaking in the late afternoon of pro-oestrus. Given kisspeptin expression is established before puberty, the aim of the present study was to investigate kisspeptin and clock gene rhythms during the neonatal period. Anterior and posterior hypothalami were collected from C57BL/6J mice on Postnatal Days (P) 5, 15 and 25, at six time points across 24h, for analysis of gene expression by reverse transcription-quantitative polymerase chain reaction. Expression of aryl hydrocarbon receptor nuclear translocator-like gene (Bmal1) and nuclear receptor subfamily 1, group D, member 2 (Rev-erbα) in the anterior hypothalamus (containing the suprachiasmatic nucleus) was not rhythmic at P5 or P15, but Bmal1 expression exhibited rhythmicity in P25 females, whereas Rev-erbα expression was rhythmic in P25 males. KiSS-1 metastasis-suppressor (Kiss1) expression did not exhibit time-of-day variation in the anterior (containing the AVPV) or posterior (containing the arcuate nucleus) hypothalami in female and male mice at P5, P15 or P25. The data indicate that the kisspeptin circadian peak in expression observed in the AVPV of pro-oestrous females does not manifest at P5, P15 or P25, likely due to inadequate oestrogenic stimuli, as well as incomplete development of clock gene rhythmicity before puberty.


CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Gene Expression Regulation, Developmental , Hypothalamus/metabolism , Kisspeptins/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/genetics , Female , Kisspeptins/genetics , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Proestrus/genetics , Proestrus/metabolism , Receptors, Kisspeptin-1/genetics , Receptors, Kisspeptin-1/metabolism , Sex Factors
9.
J Physiol Pharmacol ; 67(5): 653-666, 2016 Oct.
Article En | MEDLINE | ID: mdl-28011946

This study was conducted to determine whether two estrus phases (proestrus and diestrus) in female rats may influence the metabolic response to a high-fat diet and/or stress, focusing on pancreatic insulin secretion and content. Animals were divided into high-fat and normal diet groups, then each group was subdivided into stress and non-stress groups, and finally, each one of these was divided into proestrus and diestrus subgroups. At the end of high-fat diet treatment, foot-shock stress was applied to the animals. Then, blood samples were taken to measure plasma factors. Finally, the pancreas was removed for determination of glucose transporter 2 (GLUT2) protein levels and assessment of insulin content and secretion of the isolated islets. In the normal and high-fat diet groups, stress increased plasma corticosterone concentration in both phases. In both study phases, high-fat diet consumption decreased estradiol and increased leptin plasma levels. In the high-fat diet group in response to high glucose concentration, a reduction in insulin secretion was observed in the proestrus phase compared with the same phase in the normal diet group in the presence and absence of stress. Also, high-fat diet decreased the insulin content of islets in the proestrus phase compared with the normal diet. High-fat diet and/or stress caused a reduction in islet GLUT2 protein levels in both phases. In conclusion, it seems possible that high-fat diet alone or combined with foot-shock, predispose female rats to impaired insulin secretion, at least in part, by interfering with estradiol levels in the proestrus phase and decreasing pancreatic GLUT2 protein levels.


Diet, High-Fat , Estradiol/blood , Glucose Transporter Type 2/metabolism , Insulin/metabolism , Pancreas/metabolism , Proestrus/metabolism , Animals , Blood Glucose/analysis , Corticosterone/blood , Diestrus/blood , Diestrus/metabolism , Electric Stimulation , Female , Foot , Insulin/blood , Insulin Secretion , Islets of Langerhans/metabolism , Proestrus/blood , Rats, Wistar , Stress, Physiological , Stress, Psychological/blood , Stress, Psychological/metabolism
10.
Reprod Biol Endocrinol ; 14(1): 75, 2016 Nov 04.
Article En | MEDLINE | ID: mdl-27809846

BACKGROUND: Muscarinic receptors (mAChRs) of the preoptic and anterior hypothalamus areas (POA-AHA) regulate ovulation in an asymmetric manner during the estrous cycle. The aims of the present study were to analyze the effects of a temporal blockade of mAChRs on either side of the POA-AHA performed in diestrus-2 rats on ovulation, the levels of estradiol, follicle stimulating hormone (FSH) and luteinizing hormone (LH) and the mechanisms involved in changes in ovulation. METHODS: Cyclic rats on diestrus-2 day were anesthetized and randomly assigned to the following groups: 1) microinjection of 1 µl of saline or atropine solution (62.5 ng) in the left or right POA-AHA; 2) removal (unilateral ovariectomty, ULO) of the left (L-ULO) or right (R-ULO) ovary, and 3) rats microinjected with atropine into the left or right POA-AHA plus L-ULO or R-ULO. The ovulation rate and the number of ova shed were measured during the predicted estrus, as well as the levels of estradiol, FSH and LH during the predicted proestrus and the effects of injecting synthetic LH-releasing hormone (LHRH) or estradiol benzoate (EB). RESULTS: Atropine in the left POA-AHA decreased both the ovulation rate and estradiol and LH levels on the afternoon of proestrus, also LHRH or EB injection restored ovulation. L- or R-ULO resulted in a lower ovulation rate and smaller number of ova shed, and only injection of LHRH restored ovulation. EB injection at diestrus-2 restored ovulation in animals with L-ULO only. The levels of estradiol, FSH and LH in rats with L-ULO were higher than in animals with unilateral laparotomy. In the group microinjected with atropine in the left POA-AHA, ovulation was similar to that in ULO rats. In contrast, atropine in the right POA-AHA of ULO rats blocked ovulation, an action that was restored by either LHRH or EB injection. CONCLUSIONS: These results indicated that the removal of a single ovary at noon on diestrus-2 day perturbed the neuronal pathways regulating LH secretion, which was mediated by the muscarinic system connecting the right POA-AHA and the ovaries.


Anterior Hypothalamic Nucleus/metabolism , Diestrus/metabolism , Estradiol/metabolism , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/metabolism , Ovulation/metabolism , Preoptic Area/metabolism , Receptors, Muscarinic/metabolism , Animals , Anterior Hypothalamic Nucleus/drug effects , Atropine/pharmacology , Contraceptive Agents/pharmacology , Diestrus/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Gonadotropin-Releasing Hormone/pharmacology , Luteinizing Hormone/drug effects , Muscarinic Antagonists/pharmacology , Ovariectomy , Ovary/drug effects , Ovulation/drug effects , Preoptic Area/drug effects , Proestrus/drug effects , Proestrus/metabolism , Rats , Receptors, Muscarinic/drug effects
11.
Comp Med ; 66(3): 235-40, 2016.
Article En | MEDLINE | ID: mdl-27298249

Insulin resistance occurs during various stages of the estrus cycle in dogs. To quantify the effects of proestrus-estrus (PE) and determine whether PE affects liver insulin sensitivity, 11 female mongrel dogs were implanted with sampling and intraportal infusion catheters. Five of the dogs (PE group) entered proestrus after surgery; those remaining in anestrus were controls. The dogs were fasted overnight, [3-(3)H]glucose and somatostatin were infused through peripheral veins, and glucagon was infused intraportally. Insulin was infused intraportally, with the rate adjusted to maintain arterial plasma glucose at basal levels (PE, 294±25 µU/kg/min; control, 223±21 µU/kg/min). Subsequently the insulin infusion rate was increased by 0.2 mU/kg/min for 120 min (P1) and then to 1.5 mU/kg/min for the last 120 min (P2); glucose was infused peripherally as needed to maintain euglycemia. Insulin concentrations did not differ between groups at any time; they increased 3 µU/mL over baseline during P1 and to 3 times baseline during P2. The glucose infusion rate in PE dogs during P2 was 63% of that in control dogs. Net hepatic glucose output and the endogenous glucose production rate declined 40% to 50% from baseline in both groups during P1; during P2, both groups exhibited a low rate of net hepatic glucose uptake with full suppression of endogenous glucose production. The glucose disappearance rate during P1 and P2 was 35% greater in control than PE dogs. Therefore, PE in canines is associated with loss of nonhepatic (primarily muscle) but not hepatic insulin sensitivity.


Dogs/metabolism , Estrus/metabolism , Insulin/metabolism , Liver/metabolism , Proestrus/metabolism , Animals , Insulin Resistance
12.
Reprod Biol Endocrinol ; 14(1): 34, 2016 Jun 16.
Article En | MEDLINE | ID: mdl-27306649

BACKGROUND: The suprachiasmatic nucleus (SCN) and the cholinergic system of various regions of the hypothalamus participate in the regulation of gonadotropin-releasing hormone (GnRH) and gonadotropin secretion, which are necessary for the occurrence of ovulation. In the present study, our goal was to analyse the effects of unilaterally blocking the muscarinic receptors in the SCN on ovulation and steroid secretion. METHODS: Cyclic rats were randomly allotted to one of the experimental groups. Groups of 8-14 rats were anaesthetized and microinjected with 0.3 µl of saline or a solution of atropine (62.5 ng in 0.3 µl of saline) into the left or right SCN at 09.00 or 19.00 h during diestrus-1 or on the proestrus day. The rats were euthanized on the predicted day of oestrus, and evaluated ovulation and levels of progesterone and oestradiol. Other groups of 10 rats were microinjected with atropine into the left or right SCNs at 09.00 h on the proestrus day, were euthanized eight h later, and luteinizing hormone (LH) was measured. RESULTS: At 09.00 or 19.00 h during diestrus-1, atropine microinjections into the SCNs on either side did not modify ovulation. The atropine microinjections performed at 09.00 h of proestrus into either side of the SCN blocked ovulation (right SCN: 1/9 ovulated vs. 9/10 in the saline group; left SCN: 8/14 ovulated vs. 10/10 in the saline group). The LH levels at 17.00 h in the rats that were microinjected with atropine at 09.00 h of proestrus were lower than those of the controls. In the non-ovulating atropine-treated rats, the injection of synthetic LH-releasing hormone (LHRH) restored ovulation. Atropine treatment at 19.00 h of proestrus on either side of the SCN did not modify ovulation, while the progesterone and oestradiol levels were lower. CONCLUSION: Based on the present results, we suggest that the cholinergic neural information arriving on either side of the SCN is necessary for the pre-ovulatory secretion of LH to induce ovulation. Additionally, the regulation of progesterone and oestradiol secretion by the cholinergic innervation of the SCN varies with the time of day, the day of the cycle, and the affected SCN.


Atropine/pharmacology , Luteinizing Hormone/blood , Muscarinic Antagonists/pharmacology , Ovulation/drug effects , Proestrus/drug effects , Suprachiasmatic Nucleus/drug effects , Animals , Female , Ovary/drug effects , Proestrus/metabolism , Rats , Suprachiasmatic Nucleus/metabolism
13.
Endocrinology ; 157(2): 820-30, 2016 Feb.
Article En | MEDLINE | ID: mdl-26653570

The neuropeptide kisspeptin (Kiss1) is integral to the advent of puberty and the generation of cyclical LH surges. Although many complex actions of Kiss1 are known, the mechanisms governing the processing/regulation of this peptide have not been unveiled. The metallo enzyme, endopeptidase 24.15 (thimet oligopeptidase), has been demonstrated to play a key role in the processing and thus the duration of action of the reproductive neuropeptide, GnRH, which signals downstream of Kiss1. Initial in silico modeling implied that Kiss1 could also be a putative substrate for EP24.15. Coincubation of Kiss1 and EP24.15 demonstrated multiple cleavages of the peptide predominantly between Arg29-Gly30 and Ser47-Phe48 (corresponding to Ser5-Phe6 in Kiss-10; Kiss-10 as a substrate had an additional cleavage between Phe6-Gly7) as determined by mass spectrometry. Vmax for the reaction was 2.37±0.09 pmol/min · ng with a Km of 19.68 ± 2.53µM, which is comparable with other known substrates of EP24.15. EP24.15 immunoreactivity, as previously demonstrated, is distributed in cell bodies, nuclei, and processes throughout the hypothalamus. Kiss1 immunoreactivity is localized primarily to cell bodies and fibers within the mediobasal and anteroventral-periventricular hypothalamus. Double-label immunohistochemistry indicated coexpression of EP24.15 and Kiss1, implicating that the regulation of Kiss1 by EP24.15 could occur in vivo. Further studies will be directed at determining the precise temporal sequence of EP24.15 effects on Kiss1 as it relates to the control of reproductive hormone secretion and treatment of fertility issues.


Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/enzymology , Kisspeptins/metabolism , Metalloendopeptidases/metabolism , Animals , Computer Simulation , Escherichia coli , Female , Hypothalamus/metabolism , Immunohistochemistry , Male , Mass Spectrometry , Metestrus/metabolism , Proestrus/metabolism , Rats
14.
Endocrinology ; 157(3): 1082-93, 2016 Mar.
Article En | MEDLINE | ID: mdl-26677881

Gonadotropes in the anterior pituitary gland are of particular importance within the hypothalamic-pituitary-gonadal axis because they provide a means of communication and thus a functional link between the brain and the gonads. Recent results indicate that female gonadotropes may be organized in the form of a network that shows plasticity and adapts to the altered endocrine conditions of different physiological states. However, little is known about functional changes on the molecular level within gonadotropes during these different conditions. In this study we capitalize on a binary genetic strategy in order to fluorescently label murine gonadotrope cells. Using this mouse model allows to produce an enriched gonadotrope population using fluorescence activated cell sorting to perform mRNA sequencing. By using this strategy, we analyze and compare the expression profile of murine gonadotropes in different genders and developmental and hormonal stages. We find that gonadotropes taken from juvenile males and females, from cycling females at diestrus and at proestrus, from lactating females, and from adult males each have unique gene expression patterns with approximately 100 to approximately 500 genes expressed only in one particular stage. We also demonstrate extensive gene-expression profile changes with up to approximately 2200 differentially expressed genes when comparing female and male development, juveniles and adults, and cycling females. Differentially expressed genes were significantly enriched in the GnRH signaling, calcium signaling, and MAPK signaling pathways by Kyoto Encyclopedia of Genes and Genomes analysis. Our data provide an unprecedented molecular view of the primary gonadotropes and reveal a high degree of molecular plasticity within the gonadotrope population.


Cell Plasticity/genetics , Estrous Cycle/genetics , Gonadotrophs/metabolism , Lactation/genetics , RNA, Messenger/genetics , Sexual Maturation/genetics , Transcriptome , Animals , Calcium Signaling/genetics , Diestrus/genetics , Diestrus/metabolism , Estrous Cycle/metabolism , Female , Flow Cytometry , Gonadotropin-Releasing Hormone/metabolism , Lactation/metabolism , MAP Kinase Signaling System/genetics , Male , Mice , Proestrus/genetics , Proestrus/metabolism , Sequence Analysis, RNA , Signal Transduction/genetics
15.
Biol Reprod ; 93(1): 15, 2015 Jul.
Article En | MEDLINE | ID: mdl-25995272

Variations in mRNA levels and sources of metastin/kisspeptin, neurokinin B (NKB), dynorphin, and kisspeptin receptor GPR54 were examined in the ovaries of cycling rats. Kisspeptin and dynorphin mRNAs dramatically increased at 2000 h of the proestrous day. NKB mRNA also increased, but the peak was delayed by 6 h. GPR54 mRNA declined inversely with kisspeptin. Whole-ovary expressions of kisspeptin and dynorphin mRNAs, but not of NKB mRNA, were augmented by the administration of human chorionic gonadotropin (hCG). By means of laser-capture microdissection, kisspeptin mRNA was shown mostly in follicles at 2000 h of proestrus, whereas NKB and dynorphin were expressed mainly in interstitial tissues. GPR54 mRNA was detected equally in follicles, corpora lutea, and interstitial tissues. The hCG stimulated the follicular expression of kisspeptin and interstitial tissue expression of dynorphin mRNA. In primary cultures of granulosa cells prepared from equine chorionic gonadotropin-pretreated immature rats, hCG stimulated the expression of kisspeptin, dynorphin, and NKB mRNAs. Distortion of the corpus luteum and surrounding tissue borders was sometimes seen after intra-ovarian bursa administration of kisspeptin antagonist p234 for 3 days from proestrus. Progesterone production stimulated by hCG in granulosa cell culture was suppressed by p234. These data demonstrate that significant amounts of kisspeptin are synthesized in granulosa cells and dynorphin in interstitial tissues, in response to the proestrous luteinizing hormone surge, whereas granulosa cells also contain dynorphin and NKB, suggesting at least a role for kisspeptin in the luteinization of granulosa cells.


Gene Expression Regulation , Granulosa Cells/metabolism , Kisspeptins/metabolism , Luteinization/metabolism , Luteinizing Hormone/metabolism , Proestrus/metabolism , Animals , Corpus Luteum/drug effects , Corpus Luteum/metabolism , Dynorphins/genetics , Dynorphins/metabolism , Female , Granulosa Cells/drug effects , Kisspeptins/genetics , Luteinization/genetics , Peptides/pharmacology , Proestrus/drug effects , Proestrus/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1
16.
Endocrinology ; 156(8): 2999-3011, 2015 Aug.
Article En | MEDLINE | ID: mdl-25993523

The kisspeptin (Kp) neurons in the anteroventral periventricular nucleus (AVPV) are essential for the preovulatory LH surge, which is gated by circulating estradiol (E2) and the time of day. We investigated whether AVPV Kp neurons in intact female mice may be the site in which both E2 and daily signals are integrated and whether these neurons may host a circadian oscillator involved in the timed LH surge. In the afternoon of proestrous day, Kp immunoreactivity displayed a marked and transient decrease 2 hours before the LH surge. In contrast, Kp content was stable throughout the day of diestrus, when LH levels are constantly low. AVPV Kp neurons expressed the clock protein period 1 (PER1) with a daily rhythm that is phase delayed compared with the PER1 rhythm measured in the main clock of the suprachiasmatic nuclei (SCN). PER1 rhythm in the AVPV, but not in the SCN, exhibited a significant phase delay of 2.8 hours in diestrus as compared with proestrus. Isolated Kp-expressing AVPV explants from PER2::LUCIFERASE mice displayed sustained circadian oscillations of bioluminescence with a circadian period (23.2 h) significantly shorter than that of SCN explants (24.5 h). Furthermore, in AVPV explants incubated with E2 (10 nM to 1 µM), the circadian period was lengthened by 1 hour, whereas the SCN clock remained unaltered. In conclusion, these findings indicate that AVPV Kp neurons display an E2-dependent daily rhythm, which may possibly be driven by an intrinsic circadian clock acting in combination with the SCN timing signal.


Anterior Hypothalamic Nucleus/metabolism , Circadian Clocks/genetics , Kisspeptins/genetics , Animals , Diestrus/drug effects , Diestrus/genetics , Diestrus/metabolism , Estradiol/metabolism , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Hypothalamus/metabolism , Kisspeptins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Proestrus/drug effects , Proestrus/genetics , Proestrus/metabolism , Suprachiasmatic Nucleus/metabolism
17.
Pharmacol Biochem Behav ; 122: 20-9, 2014 Jul.
Article En | MEDLINE | ID: mdl-24650589

The capacity to form progesterone (P4)'s 5α-reduced metabolite, 5α-pregnan-3α-ol-20-one (3α,5α-THP; a.k.a. allopregnanolone), in the brain may be related to facilitation of lordosis among estrogen-primed (E2) mice. We investigated this idea further by comparing effects of endogenous and exogenous progestogens in mice that are deficient in the Type One 5α-reductase enzyme (5α-reductase knockout mice; 5α-RKO), and their wildtype counterparts for sexual behavior. Comparisons were made following administration of progestogens that are expected to increase 3α,5α-THP or not. Sexual receptivity of 5α-RKO mice and their wildtype counterparts was examined when mice were naturally-cycling (Experiment 1); ovariectomized (OVX), E2-primed (10 µg, subcutaneous; SC) and administered P4 (0, 125, 250, or 500 µg SC; Experiment 2); and OVX, E2-primed and administered P4, medroxyprogesterone acetate (MPA, 4 mg/kg, SC, which does not convert to 3α,5α-THP) or 3α,5α-THP (4 mg/kg, SC; Experiment 3). The percentage of mounts that elicited lordosis (lordosis quotient) or aggression/rejection behavior (aggression quotient), as well as the quality of lordosis (lordosis rating), was scored. Wildtype, but not 5α-RKO, mice in behavioral estrus demonstrated significantly greater lordosis quotients and lordosis ratings, but similar aggression quotients, compared to their diestrous counterparts. Among OVX and E2-primed mice, P4 facilitated lordosis of wildtype, but not 5α-RKO, mice. MPA neither facilitated lordosis of wildtype, nor 5α-RKO mice. 3α,5α-THP administered to wildtype or 5α-RKO mice increased lordosis quotients and lordosis ratings and decreased aggression quotients. 3α,5α-THP levels in the midbrain, one brain region important for sexual behavior, were increased during behavioral estrus, with P4 administered to WT, but not 5α-RKO mice, and 3α,5α-THP administered to WT and 5α-RKO mice. MPA did not increase 3α,5α-THP. Thus, deletion of Type One 5α-reductase among female mice may attenuate reproductive responding during the estrous cycle and after hormone-priming.


Cholestenone 5 alpha-Reductase/deficiency , Proestrus/metabolism , Progesterone/pharmacology , Reproduction/physiology , Sexual Behavior, Animal/physiology , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/deficiency , Animals , Female , Male , Membrane Proteins/deficiency , Mice , Mice, Knockout , Proestrus/drug effects , Reproduction/drug effects , Sexual Behavior, Animal/drug effects
18.
J Endocrinol ; 219(3): 269-78, 2013 Dec.
Article En | MEDLINE | ID: mdl-24109089

Urocortin 2 (Ucn2) is a member of the corticotropin-releasing factor peptide family and is expressed by various tissues, including reproductive tissues such as the uterus, ovary, and placenta. However, the regulatory mechanisms of Ucn2 expression and the physiological significance of Ucn2 in these tissues remain unclear. We previously showed that passive immunization of immature female rats by i.p. injection of anti-Ucn2 IgG induces earlier onset of puberty. Therefore, this study was designed to clarify the site and regulatory mechanisms of Ucn2 expression in the uterus. Expression levels of Ucn2 mRNA in the uterus were higher in immature (2- and 4-week-old) and aged (17-month-old) rats than in mature (9-week-old) rats in the proestrus phase. In 9-week-old rats, mRNA expression levels and contents in the uterus were lower in the proestrus phase than in the diestrus phase, while plasma Ucn2 concentrations did not differ between the two phases. Ucn2-like immunoreactivitiy was detected in the endometrial gland epithelial cells of the uterus. S.c. injection of estradiol benzoate or an estrogen receptor α (ERα) agonist significantly reduced mRNA expression levels and contents of Ucn2 in the uterus when compared with vehicle-injected ovariectomized rats. By contrast, estradiol benzoate increased Ucn2 mRNA expression levels in the lung. Thus, estrogens downregulate Ucn2 expression in the uterus in a tissue-specific manner, and Ucn2 may play a role in the regulatory mechanisms of maturation of the uterus through ERα and estrous cycle.


Aging , Corticotropin-Releasing Hormone/metabolism , Down-Regulation , Estrogens/metabolism , Proestrus/metabolism , Urocortins/metabolism , Uterus/metabolism , Animals , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/blood , Corticotropin-Releasing Hormone/genetics , Down-Regulation/drug effects , Endometrium/cytology , Endometrium/drug effects , Endometrium/growth & development , Endometrium/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Estrogens/agonists , Estrogens/blood , Estrogens/pharmacology , Estrous Cycle/blood , Estrous Cycle/metabolism , Female , Organ Specificity , Ovariectomy , Proestrus/blood , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Up-Regulation/drug effects , Urocortins/antagonists & inhibitors , Urocortins/blood , Urocortins/genetics , Uterus/cytology , Uterus/drug effects , Uterus/growth & development
19.
Am J Physiol Lung Cell Mol Physiol ; 305(11): L878-89, 2013 Dec.
Article En | MEDLINE | ID: mdl-24097558

Female sex predisposes individuals to poorer outcomes during respiratory disorders like cystic fibrosis and influenza-associated pneumonia. A common link between these disorders is dysregulation of alveolar fluid clearance via disruption of epithelial sodium channel (ENaC) activity. Recent evidence suggests that female sex hormones directly regulate expression and activity of alveolar ENaC. In our study, we identified the mechanism by which estradiol (E2) or progesterone (P4) independently regulates alveolar ENaC. Using cell-attached patch clamp, we measured ENaC single-channel activity in a rat alveolar cell line (L2) in response to overnight exposure to either E2 or P4. In contrast to P4, E2 increased ENaC channel activity (NPo) through an increase in channel open probability (Po) and an increased number of patches with observable channel activity. Apical plasma membrane abundance of the ENaC α-subunit (αENaC) more than doubled in response to E2 as determined by cell surface biotinylation. αENaC membrane abundance was approximately threefold greater in lungs from female rats in proestrus, when serum E2 is greatest, compared with diestrus, when it is lowest. Our results also revealed a significant role for the G protein-coupled estrogen receptor (Gper) to mediate E2's effects on ENaC. Overall, our results demonstrate that E2 signaling through Gper selectively activates alveolar ENaC through an effect on channel gating and channel density, the latter via greater trafficking of channels to the plasma membrane. The results presented herein implicate E2-mediated regulation of alveolar sodium channels in the sex differences observed in the pathogenesis of several pulmonary diseases.


Alveolar Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Estradiol/physiology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cells, Cultured , Epithelial Sodium Channels/genetics , Estradiol Congeners/pharmacology , Female , Ion Channel Gating , Membrane Potentials , Nitriles/pharmacology , Proestrus/metabolism , Protein Transport , Rats , Rats, Wistar
20.
Brain Res ; 1535: 71-7, 2013 Oct 16.
Article En | MEDLINE | ID: mdl-23994211

In rodents, the display of reproductive behavior occurs during the proestrus-estrus transition of the estrus cycle. This behavior is regulated by estradiol and progesterone mainly via their intracellular receptors. Two isoforms of the progesterone receptor have been described (A and B), and they have different promoters for their regulation. It has been demonstrated that the mRNA for both isoforms changes during the proestrus-estrus transition. It has been recently established that DNA methylation can be transient and cyclical in gene promoters, however, these changes have only been reported in vitro but not in physiological models. The aim of this study was to analyze the pattern of DNA methylation in the PR (A and B) promoter regions during the proestrus-estrus transition in the rat hypothalamus and its correlation with the regulation of mRNA expression. The results demonstrated a differential mRNA expression of the progesterone receptor (A and B) isoforms. The expression of total PR did not change significantly during the proestrus day, while the expression of isoform B increased significantly at 17:00 h, followed by a significant decrease at 21:00 h of the proestrus day. Interestingly, we also found that the isoform A promoter was mainly unmethylated at all studied time points. In contrast, the isoform B promoter showed a transient methylation increase during the evening of proestrus. The overall results indicate that there is a switch of progesterone receptor isoforms expression during the evening of proestrus that is related to the differential gene methylation patterns of their promoter regions, mainly for the isoform B promoter.


DNA Methylation/physiology , Hypothalamus/metabolism , Proestrus/metabolism , Promoter Regions, Genetic/physiology , RNA, Messenger/genetics , Receptors, Progesterone/genetics , Animals , Female , Gene Expression Regulation , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Progesterone/metabolism , Time Factors
...