Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 186
1.
J Psychopharmacol ; 35(12): 1523-1535, 2021 12.
Article En | MEDLINE | ID: mdl-34872406

BACKGROUND: Acute hypoxia, which is panicogenic in humans, also evokes panic-like behavior in male rats. Panic disorder is more common in women and susceptibility increases during the premenstrual phase of the cycle. AIMS: We here investigated for the first time the impact of hypoxia on the expression of panic-like escape behavior by female rats and its relationship with the estrous cycle. We also evaluated functional activation of the midbrain panic circuitry in response to this panicogenic stimulus and whether short-term, low-dose fluoxetine treatment inhibits the hyper-responsiveness of females in late diestrus. METHODS: Male and female Sprague Dawley rats were exposed to 7% O2. Females in late diestrus were also tested after short-term treatment with fluoxetine (1.75 or 10 mg/kg, i.p.). Brains were harvested and processed for c-Fos and tryptophan hydroxylase immunoreactivity in the periaqueductal gray matter (PAG) and dorsal raphe nucleus (DR). RESULTS: Acute hypoxia evoked escape in both sexes. Overall, females were more responsive than males and this is clearer in late diestrus phase. In both sexes, hypoxia induced functional activation (c-Fos expression) in non-serotonergic cells in the lateral wings of the DR and dorsomedial PAG, which was greater in late diestrus than proestrus (lowest behavioral response to hypoxia). Increased responding in late diestrus (behavioral and cellular levels) was prevented by 1.75, but not 10 mg/kg fluoxetine. DISCUSSION: The response of female rats to acute hypoxia models panic behavior in women. Low-dose fluoxetine administered in the premenstrual phase deserves further attention for management of panic disorders in women.


Behavior, Animal/drug effects , Diestrus/drug effects , Dorsal Raphe Nucleus/drug effects , Fluoxetine/pharmacology , Hypoxia/complications , Panic/drug effects , Periaqueductal Gray/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Sex Characteristics , Animals , Disease Models, Animal , Female , Male , Menstrual Cycle/drug effects , Panic Disorder/drug therapy , Rats , Rats, Sprague-Dawley , Selective Serotonin Reuptake Inhibitors/administration & dosage
2.
Int J Mol Sci ; 23(1)2021 Dec 23.
Article En | MEDLINE | ID: mdl-35008567

Polycystic ovary syndrome (PCOS), which affects 5-10% of women of reproductive age, is associated with reproductive and metabolic disorders, such as chronic anovulation, infertility, insulin resistance, and type 2 diabetes. However, the mechanism of PCOS is still unknown. Therefore, this study used a letrozole-exposed mouse model in which mice were orally fed letrozole for 20 weeks to investigate the effects of letrozole on the severity of reproductive and metabolic consequences and the expression of cysteine-cysteine motif chemokine receptor 5 (CCR5) in letrozole-induced PCOS mice. The letrozole-treated mice showed a disrupted estrous cycle and were arrested in the diestrus phase. Letrozole treatment also increased plasma testosterone levels, decreased estradiol levels, and caused multicystic follicle formation. Furthermore, histological analysis of the perigonadal white adipose tissue (pgWAT) showed no significant difference in the size and number of adipocytes between the letrozole-treated mice and the control group. Further, the letrozole-treated mice demonstrated glucose intolerance and insulin resistance during oral glucose and insulin tolerance testing. Additionally, the expression of CCR5 and cysteine-cysteine motif ligand 5 (CCL5) were significantly higher in the pgWAT of the letrozole-treated mice compared with the control group. CCR5 and CCL5 were also significantly correlated with the homeostasis model assessment of insulin resistance (HOMA-IR). Finally, the mechanisms of insulin resistance in PCOS may be caused by an increase in serine phosphorylation and a decrease in Akt phosphorylation.


Cysteine/metabolism , Letrozole/pharmacology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Receptors, CCR5/metabolism , Receptors, Chemokine/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diestrus/drug effects , Diestrus/metabolism , Disease Models, Animal , Estrous Cycle/drug effects , Estrous Cycle/metabolism , Female , Glucose/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Ovary/drug effects , Ovary/metabolism , Reproduction/drug effects , Reproduction/physiology , Testosterone/metabolism
3.
Behav Brain Res ; 397: 112952, 2021 01 15.
Article En | MEDLINE | ID: mdl-33017640

Low concentrations of ovarian hormones, among other factors, are associated with greater vulnerability to negative effects of environmental stressors and may trigger anxiety symptoms in females. The flavonoid chrysin (5,7-dihydroxyflavone) exerts anxiolytic-like effects in male and ovariectomized female rats, but it is unknown if chrysin could reduce anxiety-like behavior that naturally occurs through the ovarian cycle phases. The present study evaluated the effect of chrysin on anxiety-like behavior associated with the ovarian cycle phases in rats and the participation of γ-aminobutyric acid-A (GABAA) receptors in these actions. The acute effects of chrysin (2 mg/kg) were investigated in female cycling Wistar rats in the elevated plus maze, locomotor activity test, and light/dark test. Diazepam (2 mg/kg) was used as reference anxiolytic drug. The participation of GABAA receptor in the anxiolytic actions of chrysin was explored by pretreating the rats with the noncompetitive GABAA chloride ion channel antagonist picrotoxin (1 mg/kg). Chrysin and diazepam prevented anxiety-like behavior that was associated with the metestrus-diestrus phase in both the elevated plus maze and light/dark test, and these effects were reversed by picrotoxin, with no significant changes in spontaneous locomotor activity. No significant motor effects of chrysin were detected in either behavioral test during proestrus-estrus or metestrus-diestrus phases, whereas diazepam produced motor hypoactivity in the locomotor activity test during proestrus-estrus phase. These results indicate that the flavonoid chrysin prevents anxiety-like behavior that naturally occurs during metestrus-diestrus in two unconditioned models that are used to evaluate anxiety-like behavior, and these effects were mediated by actions on GABAA receptors.


Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Behavior, Animal/drug effects , Diazepam/pharmacology , Estrous Cycle/drug effects , Flavonoids/pharmacology , GABA-A Receptor Antagonists/pharmacology , Animals , Anti-Anxiety Agents/administration & dosage , Diazepam/administration & dosage , Diestrus/drug effects , Estrus/drug effects , Female , Flavonoids/administration & dosage , GABA-A Receptor Antagonists/administration & dosage , Metestrus/drug effects , Picrotoxin/pharmacology , Proestrus/drug effects , Rats , Rats, Wistar
4.
Acta Histochem ; 122(8): 151630, 2020 Dec.
Article En | MEDLINE | ID: mdl-32992122

Visfatin is a crucial adipokine, which also regulates ovarian functions in many animals. Mice estrous cycle is characterized by a dynamic complex physiological process in the reproductive system. Expression of various factors changes during the estrous cycle in the ovary. To the best of our knowledge, no previous study has been conducted on the expression of visfatin in mice ovaries during the estrous cycle. Therefore, we investigated the localization and expression of visfatin protein in the ovary of mice during the estrous cycle. Western blot analysis showed the elevated expression of visfatin in proestrus and lowest in diestrus. Immunohistochemical localization of visfatin showed intense staining in the corpus luteum of proestrus and diestrus ovaries. Thecal cells, granulosa cells, and oocytes also showed the presence of visfatin. Expression of ovarian visfatin was correlated to BCL2 and active caspase3 expression and exhibited a significant positive correlation. Furthermore, in vivo inhibition of visfatin by FK866 in the proestrus ovary down-regulated active caspase3 and PCNA expression, and up-regulated the BCL2 expression. These results suggest the role of visfatin in the proliferation and apoptosis of the follicles and specific localization of visfatin in the corpus luteum also indicate its role in corpus luteum function, which may be in progesterone biosynthesis and regression of old corpus luteum. However, further study is required to support these findings. In conclusion, visfatin may also be regulating follicular growth during the estrous cycle by regulating proliferation and apoptosis.


Corpus Luteum/metabolism , Cytokines/genetics , Granulosa Cells/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Oocytes/metabolism , Theca Cells/metabolism , Acrylamides/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cell Proliferation/drug effects , Corpus Luteum/cytology , Corpus Luteum/drug effects , Corpus Luteum/growth & development , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Diestrus/drug effects , Diestrus/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Developmental , Granulosa Cells/cytology , Granulosa Cells/drug effects , Immunohistochemistry , Mice , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Oocytes/cytology , Oocytes/drug effects , Piperidines/pharmacology , Proestrus/drug effects , Proestrus/genetics , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Theca Cells/cytology , Theca Cells/drug effects
5.
Neurotoxicology ; 79: 164-176, 2020 07.
Article En | MEDLINE | ID: mdl-32407858

Bisphenol-A (BPA) is an estrogenic chemical extensively used in industrial and household applications. The present study was conducted to investigate the effect of chronic exposure to BPA on the adult female neuroendocrine system. Herein, we found that expose of adult female mice to BPA (50 µg/kg) by oral gavage for 60 days (BPA mice) prolonged diestrus and decreased serum 17ß-estradiol (E2) concentration by reducing the number of antral follicles and corpora luteum. In comparison with controls, the levels of serum luteinizing hormone (LH), follicle stimulating hormone (FSH), hypothalamic gonadotrophin releasing hormone (GnRH) and the expression of kisspeptin in anteroventral periventricular nucleus (AVPV) decreased in BPA mice, which could be reversed by injecting kisspeptin-10 (i.c.v.). Treatment with BPA or estrogen receptor α (ERα) antagonist MPP, but not ERß antagonist PHTPP inhibited E2-induced AVPV-kisspeptin expression in ovariectomized mice. Use of ERα agonist PPT rather than ERß agonist DPN enhanced AVPV-kisspepetin expression, which decreased after treatment with BPA. The amplitude of the proestrus LH surge decreased in mice exposed to BPA, but was recovered by administering kisspeptin-10. The present study provides in vivo evidence that chronic exposure to a low dose of BPA suppressed ERα-induced activation of AVPV-kisspeptin neurons, leading to prolonged diestrus and reduced ovulation in adult female mice.


Benzhydryl Compounds/toxicity , Diestrus/drug effects , Endocrine Disruptors/toxicity , Follicular Atresia/drug effects , Hypothalamus, Anterior/drug effects , Kisspeptins/metabolism , Neurons/drug effects , Ovarian Follicle/drug effects , Phenols/toxicity , Animals , Apoptosis/drug effects , Diestrus/metabolism , Down-Regulation , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Female , Hypothalamus, Anterior/metabolism , Mice, Inbred ICR , Neurons/metabolism , Ovarian Follicle/pathology , Ovariectomy , Ovulation/drug effects , Signal Transduction , Time Factors
6.
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
7.
Bull Exp Biol Med ; 168(3): 385-389, 2020 Jan.
Article En | MEDLINE | ID: mdl-31938908

We studied the influence of the estrous cycle on the morphology of preovulatory (germinal vesicle, GV) oocytes in mice and their capacity to meiotic maturation in vitro. After standard injections of eCG gonadotropin (PMSG, Follimag) to females at different stages of the estrous cycle, the maximum levels of GV oocytes (26±1/mouse) were isolated from the ovaries of animals injected with the hormone during estrus. The capacity of isolated GV oocytes to meiotic maturation in vitro decreased in the following order: estrus (75.5±2.3%), metestrus (67.9±3.4%), proestrus (57.8±4.4%), and diestrus (50.6±5.6%); the differences between estrus and diestrus/proestrus were significant (p<0.05). After eCG injections during estrus, GV oocytes differed from other oocytes by lesser total diameter, lesser diameter of cytoplasm, lesser thickness of zona pellucida, and moderately dilated perivitelline space. These signs reflected higher competence of the "estrous" GV oocytes for meiotic maturation in vitro. Hormone stimulation of females with eCG, with consideration for the stage of the estrous cycle, seems to be an effective method for improving the quality of GV oocytes isolated from mouse ovaries.


Estrous Cycle/drug effects , Oocytes/cytology , Oocytes/drug effects , Animals , Diestrus/drug effects , Estrus/drug effects , Female , Gonadotropins/pharmacology , Metestrus/drug effects , Mice , Ovary/cytology , Ovary/drug effects
8.
Endocrinology ; 159(9): 3421-3432, 2018 09 01.
Article En | MEDLINE | ID: mdl-30137285

We have previously mimicked the morphological and functional changes occurring in the oviduct epithelium during the estrous cycle in vitro by using an air-liquid interface (ALI) culture system and basolateral application of 17ß-estradiol (E2) and progesterone (P4). In the current study we aimed to explore the transcriptomic changes elicited by E2 and P4 together during estrous cycle simulation and to dissect the individual effects of E2 and P4 on oviduct epithelium physiology. Primary porcine oviduct epithelial cells (POECs) (N = 6 animals) were cultured at the ALI. After differentiation for 11 days, we sequentially simulated diestrus (10 days) and estrus (2.5 days) by adding serum levels of E2 and P4 to the basolateral compartment either in combination (mix trial) or separately (P4 trial and E2 trial, respectively). Cell response was evaluated by microarray analysis (mix and P4 trials), quantitative RT-PCR, and histomorphometry (all trials). When we compared simulated diestrus with estrus stage in the mix trial, there were 169 (142 upregulated and 27 downregulated) differentially expressed genes (DEGs; fold change ≥1.5). In the P4 trial, 108 DEGs (83 upregulated and 25 downregulated) were detected. Gene enrichment analysis revealed that immune-related pathways were exclusively affected in the mix trial. In both mix and P4 trials, POECs exhibited in vivo-like morphological changes regarding epithelium height and portion of ciliated cells. However, E2 alone did not trigger morphological changes. We deduce that P4 mainly drives structural variations, and E2 is imperative for regulating immune function of the oviduct epithelium during estrous cycle.


Diestrus/drug effects , Epithelial Cells/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Estrus/drug effects , Oviducts/drug effects , Progesterone/pharmacology , Progestins/pharmacology , Animals , Cell Differentiation/drug effects , Culture Techniques , Diestrus/metabolism , Epithelial Cells/metabolism , Epithelium , Estrous Cycle/drug effects , Estrous Cycle/metabolism , Estrus/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , In Vitro Techniques , Oviducts/cytology , Oviducts/metabolism , Sus scrofa , Swine
9.
Toxicol Sci ; 165(2): 475-486, 2018 10 01.
Article En | MEDLINE | ID: mdl-29939337

Perfluorooctane sulfonate (PFOS) is used extensively in industrial and household applications. High exposure to PFOS has been associated with increased odds of irregular and long menstrual cycles in women. However, the underlying mechanisms remain to be elucidated. Herein, we show that adult female mice appeared prolongation of diestrus and reduction of corpora luteum within a week of oral administration of PFOS (10 mg/kg), which are associated with decreases in the levels of serum progesterone, LH and hypothalamic GnRH. The number of AVPV-kisspeptin neurons and the AVPV-kisspeptin expression were increased in proestrus mice or OVX-mice treated with high-dose estradiol benzoate (0.05 mg/kg), which were suppressed by the administration of PFOS. The administration of PFOS or GPR54 antagonist P234 prevented the generation of LH-surge in OVX-mice treated with high-dose E2. In hypothalamic slices incubated in 100 nM E2 for 4 h, the AVPV-kisspeptin expression was significantly enhanced, which was inhibited by PFOS in a dose-dependent manner or estrogen receptor α (ERα) antagonist MPP, but not ERß antagonist PHTPP. The incubation of ERα agonist PPT rather than ERß agonist DPN could increase the level of AVPV-kisspeptin expression, which was sensitive to the treatment with PFOS. The administration of GPR54 agonist kisspeptin-10 in PFOS-mice could correct the prolongation of diestrus and reduction of corpora luteum, and recover the LH-surge and the levels of LH and GnRH. The results indicate that exposure to PFOS suppressed ERα-induced activation of AVPV-kisspeptin neurons leads to diestrus prolongation and ovulation reduction.


Alkanesulfonic Acids/toxicity , Endocrine Disruptors/toxicity , Estrogen Receptor alpha/metabolism , Fluorocarbons/toxicity , Kisspeptins/metabolism , Neurons/drug effects , Reproduction/drug effects , Alkanesulfonic Acids/blood , Animals , Diestrus/drug effects , Endocrine Disruptors/blood , Female , Fluorocarbons/blood , Gonadotropin-Releasing Hormone/blood , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/metabolism , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Mice , Mice, Inbred ICR , Neurons/metabolism , Ovariectomy , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Ovulation/drug effects
10.
Pharmacol Biochem Behav ; 167: 50-59, 2018 04.
Article En | MEDLINE | ID: mdl-29486222

BACKGROUND: The mu-opioid antagonist, naltrexone (NTX), is a FDA-approved treatment for alcohol use disorder (AUD); however, the data on whether it differentially affects males vs. females are mixed. NTX increases hypothalamic-pituitary-adrenal (HPA) axis activity that associates with subjective responses to alcohol and craving in individuals with AUD. The present study tested for sex differences in the ability of NTX to decrease appetitive and consummatory behaviors in rats in operant alcohol self-administration. Because the opioid system and HPA axis are sexually dimorphic, we examined NTX's effect on adrenocorticotropic hormone (ACTH) and corticosterone (CORT) levels. METHODS: Male and female Sprague-Dawley rats (n's = 6-8) were trained to lever press for alcohol (10% v/v) under a fixed-ratio 2 schedule of reinforcement. NTX doses (0, 0.1-10 mg/kg) were assessed in tests conducted under a progressive ratio schedule of reinforcement. Separate groups of alcohol and water drinking rats (n's = 8) were used to assess NTX's (10 mg/kg) effects on HPA axis hormones. RESULTS: NTX decreased consummatory behaviors for alcohol in a dose-related manner, but not appetitive behaviors in males. In females, NTX decreased appetitive behaviors for alcohol in a dose-dependent manner, but only decreased consummatory behaviors at the highest (10 mg/kg) NTX dose. NTX increased ACTH levels in alcohol drinking females in diestrus, but not in other groups. However, NTX increased CORT levels for longer durations in alcohol drinking males relative to alcohol drinking females in diestrus. CONCLUSIONS: Our findings suggest that NTX selectively reduces consummatory behaviors for alcohol in males and appetitive behaviors in females, while also showing differential sex effects on HPA hormones.


Conditioning, Operant/drug effects , Ethanol/administration & dosage , Ethanol/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Naltrexone/pharmacology , Pituitary-Adrenal System/drug effects , Sex Characteristics , Adrenocorticotropic Hormone/blood , Animals , Appetitive Behavior/drug effects , Consummatory Behavior/drug effects , Corticosterone/blood , Diestrus/drug effects , Dose-Response Relationship, Drug , Female , Male , Rats , Reinforcement Schedule , Self Administration
11.
Reprod Fertil Dev ; 29(11): 2175-2182, 2017 Oct.
Article En | MEDLINE | ID: mdl-28442026

The aims of the present study were to determine: (1) whether oestradiol (E2) in the superior mesenteric ganglion (SMG) modifies the release of ovarian progesterone (P4), androstenedione (A2) and E2, the activity and gene expression of 3ß-hydroxysteroid dehydrogenase (3ß-HSD) and 20α-HSD and the expression of P450 aromatase (Cyp19a1) and (2) whether any such modifications are related to changes in ovarian nitric oxide (NO) and noradrenaline (NA) levels during dioestrus II. Using an ex vivo SMG-ovarian nervous plexus-ovary system, ovarian P4 release was measured following the addition E2 plus tamoxifen (Txf) (10-6M) to the ganglion, whereas A2, E2, NA and NO were measured following the addition of E2 alone. Steroids were measured by radioimmunoassay, NA concentrations were determined by HPLC and gene expression was evaluated using reverse transcription-polymerase chain reaction. Oestradiol in the ganglion decreased ovarian P4, E2 and NA release, as well as 3ß-HSD activity, but increased the release of A2 and nitrites, as well as the 20α-HSD expression and its activity. No changes were observed in Cyp19a1 gene expression. The addition of E2 plus Txf to the ganglion reversed the effects of E2 alone. The action of oestradiol in SMG favours the beginning of functional luteolysis, due to an increase in NO release and a decrease in NA in the ovary. These results may help elucidate the role of E2 in hormone-dependent pathologies in women.


Diestrus/drug effects , Estradiol/pharmacology , Ganglia, Sympathetic/drug effects , Nitric Oxide/metabolism , Ovary/drug effects , Progesterone/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Androstenedione/metabolism , Animals , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Diestrus/metabolism , Female , Ganglia, Sympathetic/metabolism , Norepinephrine/metabolism , Ovary/metabolism , Rats , Rats, Sprague-Dawley
12.
Brain Res ; 1659: 142-147, 2017 03 15.
Article En | MEDLINE | ID: mdl-28137424

Progesterone (P) binding to the intracellular progesterone receptors (PRs) plays a key role in epilepsy via modulation of GABA-A receptor plasticity in the brain. This is thought to occur via conversion of P to neurosteroids such as allopregnanolone, an allosteric modulator of GABA-A receptors. In the female brain, the composition of GABA-A receptors is not static and undergoes dynamic spatial changes in response to fluctuations in P and neurosteroid levels. Synaptic α2-containing GABA-A receptors contribute to phasic neuronal excitability and seizure susceptibility. However, the mechanisms underlying α2-subunit plasticity remain unclear. Here, we utilized the neurosteroid synthesis inhibitor finasteride and PR knockout mice to investigate the role of PRs in α2-subunit in the hippocampus. α2-Subunit expression was significantly upregulated during the high-P state of diestrous stage and with P treatment in wildtype and PR knockout mice. In contrast, there was no change in α2-subunit expression when metabolism of P into neurosteroids was blocked by finasteride in both genotypes. These findings suggest that ovarian cycle-related P and neurosteroids regulate α2-GABA-A receptor expression in the hippocampus via a non-PR pathway, which may be relevant to menstrual-cycle related brain conditions.


Diestrus/metabolism , Hippocampus/metabolism , Neurotransmitter Agents/metabolism , Progesterone/metabolism , Receptors, GABA-A/metabolism , Receptors, Progesterone/metabolism , Animals , Diestrus/drug effects , Female , Hippocampus/drug effects , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurotransmitter Agents/administration & dosage , Neurotransmitter Agents/antagonists & inhibitors , Progesterone/administration & dosage , RNA, Messenger/metabolism , Receptors, GABA-A/genetics , Receptors, Progesterone/genetics , Up-Regulation/drug effects , Up-Regulation/physiology
13.
Anim Reprod Sci ; 177: 12-24, 2017 Feb.
Article En | MEDLINE | ID: mdl-27939589

Lipopolysaccharides (LPS) from Gram negative bacteria are involved in the pathogeny of uterine diseases in cows. This study aimed to investigate LPS effects on the growth of bovine endometrial epithelial cells (bEEC) and relationships between LPS response and tissue characteristics. Uteri from 35 females were characterized for parity and stage of oestrous cycle. Densities of glandular tissue (dGT), CD11b+ cells and Ki67+ cells were measured in the endometrial tissue. Cells from 13 dioestrus cows were exposed to 0, 2, 4, 8, 12, 16 or 24µg/mL LPS. Effects of parity and stage of the oestrous cycle on tissue characteristics and effects of LPS dosage, cow and tissue characteristics on changes in cell numbers were analyzed by ANOVA. The dGT was higher in metoestrus and dioestrus samples than in pro-oestrus ones whereas densities of CD11b+ and Ki67+ cells were higher at pro-oestrus (p<0.05-p<0.01). LPS influenced bEEC populations in a dose related manner. An increase in number of live cells was observed for dosages ranging from 2 to 12µg/mL LPS (p<0.0001 vs controls). No effect was found on numbers and frequencies of dead cells. With higher dosages, the numbers of live cells did not increase but the numbers of dead did increase. No relationships were observed between cow or tissue characteristics and growth patterns or frequencies of viable bEEC in controls nor in the response to LPS. To conclude this model is suitable for further studies on dysregulations induced by LPS in endometrial tissue.


Cattle , Diestrus/drug effects , Endometrium/drug effects , Epithelial Cells/drug effects , Lipopolysaccharides/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endometrium/cytology , Epithelial Cells/physiology , Female , Pregnancy
14.
Anim Reprod Sci ; 176: 64-69, 2017 Jan.
Article En | MEDLINE | ID: mdl-27908671

The objective of the present study was to investigate the effect of reproductive hormones (GnRH, hCG, LH and progesterone) on the regulation of corpus luteum (CL) and ovarian blood flow. Diestrous mares received a single treatment of saline, 100µg gonadorelin (GnRH), or 1500IU hCG 10days after ovulation. Plasma LH and progesterone concentrations, resistance index (RI) for ovarian artery blood-flow, and percentage of corpus luteum (CL) with color-Doppler signals of blood flow were determined immediately before treatment (hour 0) and at hours 0.25, 0.5, 1, 1.5, 2, 3, 4, 5, and 6. In the GnRH group, LH increased (P<0.0001) between hours 0 and 0.25 and then progressively decreased; concentration of LH was not affected in the saline and hCG groups. Progesterone concentration was not different among groups. In the GnRH group, RI tended (P<0.07) to decrease between hours 0 and 1.5 and increased (P<0.01) between hours 1.5 and 4. In the hCG group, two transient RI decreases (P<0.05) occurred before hour 2. The percentage change from hour 0 in the percentage of CL with blood-flow signals was greater at hour 0.5 in the GnRH group than in the saline group and was intermediate in the hCG group. The similarity among groups in progesterone concentration indicated that changes in progesterone were not involved in the GnRH and hCG stimulation of ovarian vascular perfusion. Effects of treatment might have been mediated through LH; however, since hCG biological activity is primarily LH-like, the differences in timing and degree of ovarian and luteal blood flow changes after GnRH or hCG administration in the present study suggest that GnRH might have a direct effect on ovarian blood vessels and vascular control.


Diestrus/drug effects , Gonadotropin-Releasing Hormone/pharmacology , Horses , Ovary/blood supply , Progesterone/blood , Animals , Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/pharmacology , Diestrus/blood , Diestrus/physiology , Female , Gonadotropin-Releasing Hormone/administration & dosage , Luteinizing Hormone , Ovary/physiology , Progesterone/metabolism
15.
Domest Anim Endocrinol ; 58: 30-38, 2017 01.
Article En | MEDLINE | ID: mdl-27639459

Ovarian antral follicles in the ewe grow in an orderly succession, producing 3 to 4 waves per estrous cycle. In prolific sheep, some large antral follicles from the second-to-last wave of the estrous cycle are added to the ovulatory follicles emerging just before estrus to give a higher ovulation rate; it is feasible that regression of these follicles is prevented by an increase in serum concentrations of FSH or LH pulsatility at proestrus. Prolific sheep tend to have a shorter luteal phase than nonprolific ewes and there is a great deal of evidence that luteal progesterone (P4), in addition to regulating LH release, may govern the secretion of FSH heralding the emergence of follicular waves. The specific purpose of this study was to determine whether or not extending the duration of the luteal phase in prolific sheep to that typically seen in nonprolific breeds would alter the follicle wave dynamics and ovulation rate. In 2 separate experiments, exogenous P4 (7.5 mg per ewe intramuscularly) was administered on day 11 at PM and day 12 at AM (day 0 = first ovulation of the interovulatory interval studied) in moderately prolific Rideau Arcott × Polled Dorset ewes (experiment 1, n = 8) and highly prolific Olkuska ewes (experiment 2, n = 7; TRT), whereas the equinumerous groups of animals served as controls (CTR). Transrectal ovarian ultrasonography was performed daily, and jugular blood samples were drawn twice a day from day 9 until the next ovulation. Progesterone injections resulted in relatively uniform increments in serum P4 levels, but the mean duration of the interovulatory interval did not differ (P > 0.05) between TRT and CTR groups of ewes in either experiment. The mean ovulation rate post-treatment was 1.6 ± 0.2 vs 3.2 ± 0.4 (experiment 1, P < 0.001) and 3.2 ± 0.8 vs 4.0 ± 1.0 (experiment 2, P > 0.05) in TRT vs CTR, respectively. The number and percentage of ovulating follicles from the penultimate wave of the interovulatory interval studied was 0.25 ± 0.16 vs 1.75 ± 0.45 (P < 0.01) and 25.0 ± 16.4% vs 75.0 ± 16.4% (P < 0.05) in experiment 1, and 0.50 ± 0.30 vs 1.60 ± 0.40 (P < 0.05) and 13.8 ± 9.0% vs 53.4 ± 16.7% (P < 0.05) in experiment 2, for TRT vs CTR, respectively. In summary, administration of P4 at the end of diestrus decreased the incidence of ovulations from the penultimate wave of the estrous cycle in both the moderately and highly prolific strains of sheep, but it reduced the ovulation rate only in moderately prolific ewes.


Ovulation/physiology , Progesterone/physiology , Sheep/physiology , Animals , Breeding , Diestrus/drug effects , Female , Follicle Stimulating Hormone/blood , Luteal Phase/physiology , Luteinizing Hormone/blood , Ovarian Follicle/physiology , Ovary/diagnostic imaging , Ovulation/drug effects , Progesterone/administration & dosage , Progesterone/blood , Ultrasonography/veterinary
16.
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
17.
Biochem Pharmacol ; 116: 162-75, 2016 Sep 15.
Article En | MEDLINE | ID: mdl-27453435

P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance protein 4 (MRP4) are three efflux transporters that play key roles in the pharmacokinetics of antiretroviral drugs used in the pre-exposure prophylaxis of HIV sexual transmission. In this study, we investigated the expression, regulation, and function of these transporters in cervicovaginal tissues of a mouse model. Expression and regulation were examined using real-time RT-PCR and immunohistochemical staining, in the mouse tissues harvested at estrus and diestrus stages under natural cycling or after hormone synchronization. The three transporters were expressed at moderate to high levels compared to the liver. Transporter proteins were localized in various cell types in different tissue segments. Estrous cycle and exogenous hormone treatment affected transporter mRNA and protein expression, in a tissue- and transporter-dependent manner. Depo-Provera-synchronized mice were dosed vaginally or intraperitoneally with (3)H-TFV, with or without MK571 co-administration, to delineate the function of cervicovaginal Mrp4. Co-administration of MK571 significantly increased the concentration of vaginally-administered TFV in endocervix and vagina. MK571 increased the concentration of intraperitoneally-administered TFV in the cervicovaginal lavage and vagina by several fold. Overall, P-gp, Bcrp, and Mrp4 were positively expressed in mouse cervicovaginal tissues, and their expression can be regulated by the estrous cycle or by exogenous hormones. In this model, the Mrp4 transporter impacted TFV distribution in cervicovaginal tissues.


ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Anti-HIV Agents/pharmacokinetics , Cervix Uteri/metabolism , Gene Expression Regulation , Multidrug Resistance-Associated Proteins/metabolism , Vagina/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Anti-HIV Agents/metabolism , Cell Line , Cervix Uteri/cytology , Cervix Uteri/drug effects , Diestrus/drug effects , Diestrus/metabolism , Estrus/drug effects , Estrus/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Mice , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/genetics , Organ Specificity , Propionates/pharmacology , Quinolines/pharmacology , Rabbits , Reproductive Control Agents/pharmacology , Species Specificity , Tenofovir/metabolism , Tenofovir/pharmacokinetics , Tissue Distribution/drug effects , Vagina/cytology , Vagina/drug effects , Vaginal Creams, Foams, and Jellies/metabolism , Vaginal Creams, Foams, and Jellies/pharmacokinetics
18.
Endocrinology ; 157(3): 1187-99, 2016 Mar.
Article En | MEDLINE | ID: mdl-26697722

Stress elicits activation of the hypothalamic-pituitary-adrenal axis, which leads to enhanced circulating glucocorticoids, as well as impaired gonadotropin secretion and ovarian cyclicity. Here, we tested the hypothesis that elevated, stress-levels of glucocorticoids disrupt ovarian cyclicity by interfering with the preovulatory sequence of endocrine events necessary for the LH surge. Ovarian cyclicity was monitored in female mice implanted with a cholesterol or corticosterone (Cort) pellet. Cort, but not cholesterol, arrested cyclicity in diestrus. Subsequent studies focused on the mechanism whereby Cort stalled the preovulatory sequence by assessing responsiveness to the positive feedback estradiol signal. Ovariectomized mice were treated with an LH surge-inducing estradiol implant, as well as Cort or cholesterol, and assessed several days later for LH levels on the evening of the anticipated surge. All cholesterol females showed a clear LH surge. At the time of the anticipated surge, LH levels were undetectable in Cort-treated females. In situ hybridization analyses the anteroventral periventricular nucleus revealed that Cort robustly suppressed the percentage of Kiss1 cells coexpressing cfos, as well as reduced the number of Kiss1 cells and amount of Kiss1 mRNA per cell, compared with expression in control brains. In addition, Cort blunted pituitary expression of the genes encoding the GnRH receptor and LHß, indicating inhibition of gonadotropes during the blockage of the LH surge. Collectively, our findings support the hypothesis that physiological stress-levels of Cort disrupts ovarian cyclicity, in part, through disruption of positive feedback mechanisms at both the hypothalamic and pituitary levels which are necessary for generation of the preovulatory LH surge.


Anti-Inflammatory Agents/pharmacology , Corticosterone/pharmacology , Estrous Cycle/drug effects , Kisspeptins/drug effects , Luteinizing Hormone/drug effects , Neurons/drug effects , Pituitary Gland/drug effects , Animals , Brain/drug effects , Brain/metabolism , Diestrus/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Estrous Cycle/metabolism , Female , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/metabolism , In Situ Hybridization , Kisspeptins/genetics , Kisspeptins/metabolism , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Mice , Neurons/metabolism , Ovariectomy , Ovary , Pituitary Gland/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Real-Time Polymerase Chain Reaction , Receptors, LHRH/drug effects , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological , Stress, Psychological/metabolism
19.
Toxins (Basel) ; 7(11): 4668-83, 2015 Nov 09.
Article En | MEDLINE | ID: mdl-26569305

Growing evidence has revealed the deleterious influence of environmental and food contaminants on puberty onset and development in both animals and children, provoking an increasing health concern. T-2 toxin, a naturally-produced Type A trichothecene mycotoxin which is frequently found in cereal grains and products intended for human and animal consumption, has been shown to impair the reproduction and development in animals. Nevertheless, whether this trichothecene mycotoxin can disturb the onset of puberty in females remains unclear. To clarify this point, infantile female rats were given a daily intragastric administration of vehicle or 187.5 µg/kg body weight of T-2 toxin for five consecutive days from postnatal day 15 to 19, and the effects on puberty onset were evaluated in the present study. The results revealed that the days of vaginal opening, first dioestrus, and first estrus in regular estrous cycle were delayed following prepubertal exposure to T-2 toxin. The relative weights of reproductive organs uterus, ovaries, and vagina, and the incidence of corpora lutea were all diminished in T-2 toxin-treated rats. Serum levels of gonadotropins luteinizing hormone, follicle-stimulating hormone, and estradiol were also reduced by T-2 toxin treatment. The mRNA expressions of hypothalamic gonadotropin-releasing hormone (GnRH) and pituitary GnRH receptor displayed significant reductions following exposure to T-2 toxin, which were consistent with the changes of serum gonadotropins, delayed reproductive organ development, and delayed vaginal opening. In conclusion, the present study reveals that prepubertal exposure to T-2 toxin delays the onset of puberty in immature female rats, probably by the mechanism of disturbance of hypothalamic-pituitary-gonadal (HPG) axis function. Considering the vulnerability of developmental children to food contaminants and the relative high level of dietary intake of T-2 toxin in children, we think the findings of the present study provide valuable information for the health risk assessment in children.


Sexual Maturation/drug effects , T-2 Toxin/toxicity , Animals , Body Weight/drug effects , Corpus Luteum/drug effects , Diestrus/drug effects , Estrous Cycle/drug effects , Estrus/drug effects , Female , Gonadotropin-Releasing Hormone/blood , Intubation, Gastrointestinal , Organ Size , Ovary/drug effects , Ovary/growth & development , Rats , Rats, Sprague-Dawley , Receptors, LHRH/biosynthesis , Receptors, LHRH/drug effects , T-2 Toxin/administration & dosage , Uterus/drug effects , Uterus/growth & development , Vagina/drug effects , Vagina/growth & development
20.
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
...