Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 236
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Eur J Neurosci ; 57(7): 1068-1080, 2023 04.
Article in English | MEDLINE | ID: mdl-36796802

ABSTRACT

By targeting the endocannabinoid system, delta-9-tetrahydrocannabinol (THC) modulates female motivated behaviours, influenced by sex hormones. Both medial preoptic nucleus (MPN) and ventromedial nucleus of the hypothalamus (VMN) are involved in the modulation of female sexual responses. The first triggers proceptivity, whereas the ventrolateral division of the latter (VMNvl) triggers receptivity. These nuclei are modulated by glutamate, which inhibits female receptivity, and GABA, which has a dichotomous action in female sexual motivation. Here, we evaluated the action of THC on the modulation of social and sexual behaviours, on signalling pathways of MPN and VMNvl and how sex hormones influence these parameters. Young ovariectomized female rats, given sex hormones (oestradiol benzoate, EB, and progesterone, P) and THC were used for behavioural testing and for immunofluorescence analyses of vesicular glutamate transporter 2 (VGlut2) and GAD (glutamic acid decarboxylase)67 expression. Results showed that females given EB + P exhibited a higher preference for male partner, as well as higher proceptivity and a higher receptivity than control or females given only EB. Females treated with THC presented similar responses in control or EB + P female rats and even more facilitated behavioural responses in EB females than the ones that did not receive THC. Immunofluorescence results in the MPN exhibited a decreased expression of GAD67 and VGlut2 in EB + THC-treated female rats. Within VMNvl of EB-primed rats no changes in the expression of both proteins were observed after THC exposure. This study demonstrates how the possible outcomes of endocannabinoid system instability within hypothalamic neuron connectivity can modify female rat sociosexual behaviour.


Subject(s)
Dronabinol , Sexual Behavior, Animal , Rats , Animals , Female , Male , Humans , Dronabinol/pharmacology , Sexual Behavior, Animal/physiology , Endocannabinoids , Progesterone , Estradiol/pharmacology , Estradiol/physiology , Hypothalamus , Ovariectomy
2.
J Neuroendocrinol ; 34(6): e13082, 2022 06.
Article in English | MEDLINE | ID: mdl-35000221

ABSTRACT

The development of oestrogen positive feedback is a hallmark of female puberty. Both oestrogen and progesterone signalling are required for the functioning of this neuroendocrine feedback loop but the physiological changes that underlie the emergence of positive feedback remain unknown. Only after puberty does oestradiol (E2) facilitate progesterone synthesis in the rat female hypothalamus (neuroP), an event critical for positive feedback and the LH surge. We hypothesize that prior to puberty, these astrocytes have low levels of membrane oestrogen receptor alpha (ERα), which is needed for facilitation of neuroP synthesis. Thus, we hypothesized that prepubertal astrocytes are unable to respond to E2 with increased neuroP synthesis due a lack of membrane ERα. To test this, hypothalamic tissues and enriched primary hypothalamic astrocyte cultures were acquired from prepubertal (postnatal week 3) and post-pubertal (week 8) female mice. E2-facilitated neuroP was measured in the hypothalamus pre- and post-puberty, and hypothalamic astrocyte responses were measured after treatment with E2. Prior to puberty, E2-facilitated neuroP synthesis did not occur in the hypothalamus, and mERα expression was low in hypothalamic astrocytes, but E2-facilitated neuroP synthesis in the rostral hypothalamus and mERα expression increased post-puberty. The increase in mERα expression in hypothalamic astrocytes corresponded with a post-pubertal increase in caveolin-1 protein, PKA phosphorylation, and a more rapid [Ca2+ ]i flux in response to E2. Together, results from the present study indicate that E2-facilitated neuroP synthesis occurs in the rostral hypothalamus, develops during puberty, and corresponds to a post-pubertal increase in mERα levels in hypothalamic astrocytes.


Subject(s)
Estradiol , Estrogen Receptor alpha , Animals , Astrocytes/metabolism , Estradiol/physiology , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Hypothalamus/metabolism , Mice , Progesterone/metabolism , Rats , Sexual Maturation
3.
Elife ; 92020 12 08.
Article in English | MEDLINE | ID: mdl-33289482

ABSTRACT

Metabolic dysfunction underlies several chronic diseases, many of which are exacerbated by obesity. Dietary interventions can reverse metabolic declines and slow aging, although compliance issues remain paramount. 17α-estradiol treatment improves metabolic parameters and slows aging in male mice. The mechanisms by which 17α-estradiol elicits these benefits remain unresolved. Herein, we show that 17α-estradiol elicits similar genomic binding and transcriptional activation through estrogen receptor α (ERα) to that of 17ß-estradiol. In addition, we show that the ablation of ERα completely attenuates the beneficial metabolic effects of 17α-E2 in male mice. Our findings suggest that 17α-E2 may act through the liver and hypothalamus to improve metabolic parameters in male mice. Lastly, we also determined that 17α-E2 improves metabolic parameters in male rats, thereby proving that the beneficial effects of 17α-E2 are not limited to mice. Collectively, these studies suggest ERα may be a drug target for mitigating chronic diseases in male mammals.


Subject(s)
Estradiol/physiology , Estrogen Receptor alpha/physiology , Longevity , Animals , Female , Gene Expression Regulation/physiology , Hypothalamus/metabolism , Hypothalamus/physiology , Insulin Resistance/physiology , Liver/metabolism , Liver/physiology , Longevity/physiology , Male , Mice , Mice, Knockout , Rats
4.
Sci Rep ; 10(1): 8223, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32427857

ABSTRACT

Hypothalamic neurons show sex differences in neuritogenesis, female neurons have longer axons and higher levels of the neuritogenic factor neurogenin 3 (Ngn3) than male neurons in vitro. Moreover, the effect of 17-ß-estradiol (E2) on axonal growth and Ngn3 expression is only found in male-derived neurons. To investigate whether sex chromosomes regulate these early sex differences in neuritogenesis by regulating the E2 effect on Ngn3, we evaluated the growth and differentiation of hypothalamic neurons derived from the "four core genotypes" mouse model, in which the factors of "gonadal sex" and "sex chromosome complement" are dissociated. We showed that sex differences in neurite outgrowth are determined by sex chromosome complement (XX > XY). Moreover, E2 increased the mRNA expression of Ngn3 and axonal length only in XY neurons. ERα/ß expressions are regulated by sex chromosome complement; however, E2-effect on Ngn3 expression in XY neurons was only fully reproduced by PPT, a specific ligand of ERα, and prevented by MPP, a specific antagonist of ERα. Together our data indicate that sex chromosomes regulate early development of hypothalamic neurons by orchestrating not only sex differences in neuritogenesis, but also regulating the effect of E2 on Ngn3 expression through activation of ERα in hypothalamic neurons.


Subject(s)
Axons , Basic Helix-Loop-Helix Transcription Factors/physiology , Estradiol/physiology , Hypothalamus/metabolism , Nerve Tissue Proteins/physiology , Neurons/metabolism , Sex Chromosomes , Animals , Female , Male , Mice
5.
FASEB J ; 33(6): 7375-7386, 2019 06.
Article in English | MEDLINE | ID: mdl-30866655

ABSTRACT

Polycystic ovary syndrome (PCOS) is a hypothalamic-pituitary-gonadal (HPG) axis disorder. PCOS symptoms most likely result from a disturbance in the complex feedback regulation system of the HPG axis, which involves gonadotrophic hormones and ovarian steroid hormones. However, the nature of this complex and interconnecting feedback regulation makes it difficult to dissect the molecular mechanisms responsible for PCOS phenotypes. Global estrogen receptor α (ERα) knockout (KO) mice exhibit a disruption of the HPG axis, resulting in hormonal dysregulation in which female ERα KO mice have elevated levels of serum estradiol (E2), testosterone, and LH. The ERα KO females are anovulatory and develop cystic hemorrhagic ovaries that are thought to be due to persistently high circulating levels of LH from the pituitary. However, the role of ERα in the pituitary is still controversial because of the varied phenotypes reported in pituitary-specific ERα KO mouse models. Therefore, we developed a mouse model where ERα is reintroduced to be exclusively expressed in the pituitary on the background of a global ERα-null (PitERtgKO) mouse. Serum E2 and LH levels were normalized in PitERtgKO females and were comparable to wild-type serum levels. However, the ovaries of PitERtgKO adult mice displayed a more overt cystic and hemorrhagic phenotype when compared with ERα KO littermates. We determined that anomalous sporadic LH secretion caused the severe ovarian phenotype of PitERtgKO females. Our observations suggest that pituitary ERα is involved in the estrogen negative feedback regulation, whereas hypothalamic ERα is necessary for the precise control of LH secretion. Uncontrolled, irregular LH secretion may be the root cause of the cystic ovarian phenotype with similarities to PCOS.-Arao, Y., Hamilton, K. J., Wu, S.-P., Tsai, M.-J., DeMayo, F. J., Korach, K. S. Dysregulation of hypothalamic-pituitary estrogen receptor α-mediated signaling causes episodic LH secretion and cystic ovary.


Subject(s)
Estrogen Receptor alpha/physiology , Hypothalamus/physiopathology , Luteinizing Hormone/metabolism , Ovary/physiopathology , Pituitary Gland, Anterior/physiopathology , Polycystic Ovary Syndrome/physiopathology , Animals , Disease Models, Animal , Estradiol/physiology , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Feedback, Physiological , Female , Hemorrhage/etiology , Humans , Hypothalamo-Hypophyseal System/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity , Ovary/pathology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/pathology , Recombinant Proteins/metabolism
6.
J Neuroendocrinol ; 30(11): e12646, 2018 11.
Article in English | MEDLINE | ID: mdl-30246441

ABSTRACT

We recently showed that male rats exhibit lower hypophagia and body weight loss compared to female rats following central leptin delivery, suggesting a role for oestradiol in leptin responsiveness. Accordingly, we delivered Ob (leptin) or GFP (control) gene into the brain of male rats that were simultaneously treated with oestradiol or vehicle. In a reciprocal approach, we compared oestradiol-deficient (OVX) with intact females (sham) that received leptin or control vector. Changes in food intake), body weight and body composition were examined. In males, oestradiol and leptin resulted in lower cumulative food intake (15%) and endpoint body weight (5%), although rats receiving dual treatment (oestradiol-leptin) ate 28% less and weighed 22% less than vehicle-control. Changes in food intake were unique to each treatment, with a rapid decrease in vehicle-leptin followed by gradual renormalisation. By contrast, hypophagia in oestradiol-control was of lower amplitude and sporadic. Leptin selectively targeted fat mass and endpoint abdominal fat mass was 65%-80% lower compared to their respective control groups. In females, both leptin groups had lower body weight (endpoint values 20% lower than control groups) with the highest extent in sham animals (endpoint value was 28% less in sham-leptin than in sham-control). OVX rats rapidly started regaining their lost body weight reminiscent of the pattern in males. Leptin rapidly and robustly reduced fat mass with endpoint values 30%-35% less than control treated animals. It appears that leptin and oestradiol decreased food intake and body weight via different mechanisms, with the pattern of oestradiol-leptin being reminiscent of that observed in females and the pattern of OVX-leptin reminiscent of that observed in males. Oestrogen status did not influence initial fat mass loss by leptin. It can be concluded that oestradiol modulates the long-term response to central leptin overexpression, although its actions on energy homeostasis are additive and independent of those of leptin.


Subject(s)
Adipose Tissue/physiology , Eating/physiology , Estradiol/physiology , Hypothalamus/physiology , Leptin/physiology , Adipose Tissue/drug effects , Animals , Appetite Depressants/administration & dosage , Eating/drug effects , Estradiol/administration & dosage , Estrogens/administration & dosage , Estrogens/physiology , Female , Leptin/administration & dosage , Leptin/genetics , Male , Ovariectomy , Rats, Sprague-Dawley , Rats, Transgenic , Sex Characteristics
7.
Hear Res ; 366: 38-49, 2018 09.
Article in English | MEDLINE | ID: mdl-29983289

ABSTRACT

When a social sound category initially gains behavioral significance to an animal, plasticity events presumably enhance the ability to recognize that sound category in the future. In the context of learning natural social stimuli, neuromodulators such as norepinephrine and estrogen have been associated with experience-dependent plasticity and processing of newly salient social cues, yet continued plasticity once stimuli are familiar could disrupt the stability of sensorineural representations. Here we employed a maternal mouse model of natural sensory cortical plasticity for infant vocalizations to ask whether the engagement of the noradrenergic locus coeruleus (LC) by the playback of pup-calls is affected by either prior experience with the sounds or estrogen availability, using a well-studied cellular activity and plasticity marker, the immediate early gene c-Fos. We counted call-induced c-Fos immunoreactive (c-Fos-IR) cells in both LC and physiologically validated fields within the auditory cortex (AC) of estradiol or blank-implanted virgin female mice with either 0 or 5-days prior experience caring for vocalizing pups. Estradiol and pup experience interacted both in the induction of c-Fos-IR in the LC, as well as in behavioral measures of locomotion during playback, consistent with the neuromodulatory center's activity being an online reflection of both hormonal and experience-dependent influences on arousal. Throughout core AC, as well as in a high frequency sub-region of AC and in secondary AC, a main effect of pup experience was to reduce call-induced c-Fos-IR, irrespective of estradiol availability. This is consistent with the hypothesis that sound familiarity leads to less c-Fos-mediated plasticity, and less disrupted sensory representations of a meaningful call category. Taken together, our data support the view that any coupling between these sensory and neuromodulatory areas is situationally dependent, and their engagement depends differentially on both internal state factors like hormones and external state factors like prior experience.


Subject(s)
Auditory Cortex/physiology , Estradiol/physiology , Locus Coeruleus/physiology , Proto-Oncogene Proteins c-fos/physiology , Acoustic Stimulation , Animals , Auditory Cortex/anatomy & histology , Behavior, Animal/physiology , Female , Immunohistochemistry , Learning/physiology , Locus Coeruleus/anatomy & histology , Mice , Mice, Inbred CBA , Neuronal Plasticity/physiology , Norepinephrine/physiology , Recognition, Psychology/physiology , Social Behavior , Vocalization, Animal/physiology
8.
Taiwan J Obstet Gynecol ; 56(6): 765-769, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29241917

ABSTRACT

OBJECTIVE: The notion that the human endometrium may contain a population of stem cells has recently been proposed. The mesenchymal stem cells (MSCs) in the endometrium are believed to be responsible for the remarkable regenerative ability of endometrial cells. Estrogens influence the physiological and pathological processes of several hormone-dependent tissues, such as the endometrium. Pueraria mirifica (PM) is a herbal plant that contains several phytoestrogens, including isoflavones, lignans, and coumestans, and is known to exert an estrogenic effect on animal models. The present study investigated the effects of PM on the proliferation of human endometrial MSCs (hEN-MSCs). MATERIALS AND METHODS: The hEN-MSCs were isolated from human endometrial tissue. The surface markers of these hEN-MSCs were identified through reverse transcription-polymerase chain reaction analysis. The proliferation potential of hEN-MSCs was measured through a cell proliferation assay. Multilineage differentiation ability was confirmed through Oil red O and von Kossa staining. RESULTS: This study demonstrated that 17ß-estradiol-responsive MSCs with Oct-4, CD90, and CD105 gene expression can be derived from the human endometrium and that PM exerts biological effects on hEN-MSCs, specifically, enhanced cell growth rate, through the estrogen receptor. Furthermore, PM at 1500 and 2000 µg/mL significantly increased cell proliferation compared with the vehicle control, and PM concentration at 1000 µg/mL significantly inhibited the enhanced cell growth rate induced by 17ß-estradiol in hEN-MSCs. CONCLUSION: This study provides new insights into the possible biological effects of PM on the proliferation of hEN-MSCs.


Subject(s)
Cell Proliferation/drug effects , Endometrium/cytology , Estradiol/pharmacology , Mesenchymal Stem Cells/drug effects , Phytoestrogens/pharmacology , Pueraria/chemistry , Cell Differentiation/drug effects , Estradiol/physiology , Estrogen Antagonists , Female , Humans
9.
Best Pract Res Clin Endocrinol Metab ; 31(5): 505-519, 2017 10.
Article in English | MEDLINE | ID: mdl-29223284

ABSTRACT

Female ovulation depends on a surge in circulating luteinizing hormone (LH) which occurs at the end of the resting period and requests high circulating estradiol. This fine tuning involves both an estradiol feedback as an indicator of oocyte maturation, and the master circadian clock of the suprachiasmatic nuclei as an indicator of the time of the day. This review describes the mechanisms through which daily time cues are conveyed to reproductive hypothalamic neurons to time the pre-ovulatory surge. In female rodents, neurotransmitters released by the suprachiasmatic nuclei activate the stimulatory kisspeptin neurons and reduce the inhibitory RFRP neurons precisely at the end of the afternoon of proestrus to allow a full surge in LH secretion. From these findings, the impact of circadian disruptions (during shift or night work) on female reproductive performance and fertility should now being investigated in both animal models and humans.


Subject(s)
Circadian Rhythm/physiology , Fertility/physiology , Animals , Estradiol/physiology , Female , Gonadotropin-Releasing Hormone/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/physiology , Ovulation/physiology , Suprachiasmatic Nucleus/physiology
10.
Psychopharmacology (Berl) ; 233(13): 2607-15, 2016 07.
Article in English | MEDLINE | ID: mdl-27137197

ABSTRACT

RATIONALE: The behavioral and electrophysiological responses in a Go/NoGo task are objective measures of executive functioning that may be impaired in clinical conditions. Prior to the wider application of Go/NoGo tasks in clinics, it is tempting to evaluate factors causing modulation of the responses. OBJECTIVE: We aimed to evaluate the effect of different levels of female sex steroids on Go/NoGo task-related ERPs in healthy females. METHODS: Thirty-four young healthy females performed an equiprobable (50/50) auditory Go/NoGo task. Amplitudes and latencies of N2 and P3 peaks from Fz, Cz, and Pz electrodes were evaluated. 17ß-estradiol and progesterone levels in saliva samples were measured. Electrophysiological measures were correlated to 17ß-estradiol and progesterone concentrations. RESULTS: The diverse pattern of modulation of P3 latencies was shown: higher levels of 17ß-estradiol contributed to Go-P3 latency prolongation, and higher levels of progesterone contributed to NoGo-P3 latency shortening. Higher levels of 17ß-estradiol were associated with more negative frontal N2 amplitude in both conditions. CONCLUSIONS: The relationship between electrophysiological correlates of executive functioning to individual hormonal levels points to a broader range of variation sources in healthy subjects which might mask or pronounce between-group differences in clinical studies.


Subject(s)
Auditory Perception/physiology , Estradiol/physiology , Executive Function/physiology , Progesterone/pharmacology , Acoustic Stimulation/methods , Adult , Analysis of Variance , Electroencephalography , Estradiol/analysis , Evoked Potentials/physiology , Female , Humans , Progesterone/analysis , Reaction Time/physiology , Saliva/chemistry , Young Adult
12.
Pacing Clin Electrophysiol ; 39(5): 418-26, 2016 May.
Article in English | MEDLINE | ID: mdl-26842421

ABSTRACT

BACKGROUND: The effects of gender difference on cardiac electrophysiology have been well studied. In this study, we aimed to evaluate the effects of estradiol and progesteron changes occuring in physiological menstrual cycle on ventricular premature beats (VPBs) and cardiac repolarization parameters. METHODS: Women of reproductive age with VPBs were included into the study group and healthy women were recruited as the control group. During the menstruation period, a 12-lead electrocardiography, blood samples, and 24-hour rhythm Holter were applied to the study group. Similarly, all tests were repeated in the estimated ovulation period (12-14 days before menstruation) by all cases. RESULTS: The study group consisted of 20 women patients with VPB, and the control group of 18 healthy women. While the number of VPB in the menstruation period was 210 beats/day (interquartile range [IQR]: 1,144), it decreased to 86 beats/day (IQR: 251) in the ovulation period with statistical significance (P < 0.05). Average heart rate in the menstruation period was 81.4 ± 10 beats/min and it significantly increased to 84.6 ± 8 beats/min in the ovulation period (P < 0.05). There were no differences in cardiac repolarization parameters in both menstruation and ovulation periods between the study and control groups. Comparing the menstruation and the ovulation periods, J-Tpeak interval, which reflects early repolarization, was shorter in the ovulation period (193 ± 27.7 ms and 201.1 ± 28.6 ms, respectively; P < 0.05). Other repolarization parameters did not show any significant difference. CONCLUSION: VPB frequency decreases with estradiol peak in the ovulation period. This suggests that estrogen may have protective effects against ventricular arrhythmias.


Subject(s)
Estradiol/physiology , Heart Rate/physiology , Menstrual Cycle/physiology , Progesterone/physiology , Ventricular Premature Complexes/physiopathology , Adolescent , Adult , Case-Control Studies , Electrocardiography , Electrophysiologic Techniques, Cardiac , Female , Humans , Middle Aged , Young Adult
13.
Stem Cell Res Ther ; 7: 2, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26738566

ABSTRACT

BACKGROUND: Many retinal degenerative diseases are caused by the loss of retinal ganglion cells (RGCs). Autosomal dominant optic atrophy is the most common hereditary optic atrophy disease and is characterized by central vision loss and degeneration of RGCs. Currently, there is no effective treatment for this group of diseases. However, stem cell therapy holds great potential for replacing lost RGCs of patients. Compared with embryonic stem cells, induced pluripotent stem cells (iPSCs) can be derived from adult somatic cells, and they are associated with fewer ethical concerns and are less prone to immune rejection. In addition, patient-derived iPSCs may provide us with a cellular model for studying the pathogenesis and potential therapeutic agents for optic atrophy. METHODS: In this study, iPSCs were obtained from patients carrying an OPA1 mutation (OPA1 (+/-) -iPSC) that were diagnosed with optic atrophy. These iPSCs were differentiated into putative RGCs, which were subsequently characterized by using RGC-specific expression markers BRN3a and ISLET-1. RESULTS: Mutant OPA1 (+/-) -iPSCs exhibited significantly more apoptosis and were unable to efficiently differentiate into RGCs. However, with the addition of neural induction medium, Noggin, or estrogen, OPA1 (+/-) -iPSC differentiation into RGCs was promoted. CONCLUSIONS: Our results suggest that apoptosis mediated by OPA1 mutations plays an important role in the pathogenesis of optic atrophy, and both noggin and ß-estrogen may represent potential therapeutic agents for OPA1-related optic atrophy.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Optic Atrophy, Autosomal Dominant/pathology , Carrier Proteins/physiology , Cell Differentiation , Cells, Cultured , Drug Evaluation, Preclinical , Estradiol/physiology , GTP Phosphohydrolases/genetics , Human Embryonic Stem Cells/physiology , Humans , Optic Atrophy, Autosomal Dominant/drug therapy , Optic Atrophy, Autosomal Dominant/genetics , Retinal Ganglion Cells/physiology
14.
Neuroscience ; 314: 35-46, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26628404

ABSTRACT

This study addressed the hypothesis that dorsomedial hindbrain adenosine 5'-monophosphate-activated protein kinase (AMPK) imposes inherent estradiol-dependent control of hypothalamic AMPK, neuropeptide, and norepinephrine (NE) activity and feeding in the female rat. Estradiol (E)- or oil (O)-implanted ovariectomized rats were injected with the AMPK inhibitor compound c (Cc) or vehicle into the caudal fourth ventricle (CV4) prior to micropunch-dissection of individual hypothalamic metabolic loci or assessment of food intake. Cc decreased hindbrain dorsal vagal complex phosphoAMPK (pAMPK) in both E and O; tissue ATP levels were reduced by this treatment in O only. In E/Cc, pAMPK expression was diminished in the lateral hypothalamic area (LHA) and ventromedial (VMH) and paraventricular (PVH) nuclei; only PVH pAMPK was suppressed by this treatment in O/Cc. Cc decreased PVH corticotropin-releasing hormone and arcuate (ARH) proopiomelanocortin (POMC) and neuropeptide Y in O, but suppressed only POMC in E. O/Cc exhibited both augmented (PVH, VMH) and decreased (LHA, ARH) hypothalamic NE content, whereas Cc treatment of E elevated preoptic and dorsomedial hypothalamic nucleus NE. Cc completely or incompletely repressed feeding in E versus O, respectively. Results implicate dorsomedial hindbrain AMPK in physiological stimulus-induced feeding in females. Excepting POMC, hypothalamic neuropeptide responses to this sensor may be contingent on estrogen. Estradiol likely designates hypothalamic targets of altered NE signaling due to hindbrain AMPK activation. Divergent changes in NE content of hypothalamic loci in O/Cc uniquely demonstrate sensor-induced bimodal catecholamine signaling to those sites.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Eating , Estradiol/physiology , Hypothalamus/enzymology , Neuropeptides/metabolism , Norepinephrine/metabolism , Rhombencephalon/enzymology , Adenosine Triphosphate/metabolism , Animals , Corticotropin-Releasing Hormone/metabolism , Eating/drug effects , Estradiol/administration & dosage , Female , Hypothalamus/drug effects , Injections, Intraventricular , Neuropeptide Y/metabolism , Orexins/metabolism , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley , Rhombencephalon/drug effects , Steroidogenic Factor 1/metabolism
15.
Biol Reprod ; 93(2): 30, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26063871

ABSTRACT

The mammalian gonadotropin-inhibitory hormone (GnIH) ortholog, RFamide-related peptide (RFRP), is considered to act on gonadotropin-releasing hormone (GnRH) neurons and the pituitary to inhibit gonadotropin synthesis and release. However, there is little evidence documenting whether RFamide-related peptide 3 (RFRP-3) plays a primary role in inhibition of the hypothalamo-pituitary-gonadal (HPG) axis prior to the onset of puberty. The present study aimed to understand the functional significance of the neuropeptide on pubertal development. The developmental changes in reproductive-related gene expression at the mRNA level were investigated in the hypothalamus of female mice. The results indicated that RFRP-3 may be an endogenous inhibitory factor for the activation of the HPG axis prior to the onset of puberty. In addition, centrally administered RFRP-3 significantly suppressed plasma luteinizing hormone (LH) levels in prepubertal female mice. Surprisingly, centrally administered RFRP-3 had no effects on plasma LH levels in ovariectomized (OVX) prepubescent female mice. In contrast, RFRP-3 also inhibited plasma LH levels in OVX prepubescent female mice that were treated with 17beta-estradiol replacement. Our study also examined the effects of RFRP-3 on plasma LH release in adult female mice that were ovariectomized at dioestrus, with or without estradiol (E2). Our results showed that the inhibitory effects of RFRP-3 were independent of E2 status. Quantitative real-time PCR and immunohistochemistry analyses showed that RFRP-3 inhibited GnRH expression at both the mRNA and protein levels in the hypothalamus. These data demonstrated that RFRP-3 could effectively suppress pituitary LH release, via the inhibition of GnRH transcription and translation in prepubescent female mice, which is associated with estrogen signaling pathway and developmental stages.


Subject(s)
Estradiol/physiology , Luteinizing Hormone/antagonists & inhibitors , Luteinizing Hormone/metabolism , Neuropeptides/pharmacology , Sexual Maturation/physiology , Animals , Estradiol/pharmacology , Female , Gene Expression/drug effects , Gonadotropin-Releasing Hormone/biosynthesis , Hypothalamo-Hypophyseal System/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Mice , Ovariectomy , Pregnancy , RNA, Messenger/biosynthesis , Signal Transduction/drug effects
16.
Neuroscience ; 299: 125-33, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-25934040

ABSTRACT

Group I metabotropic glutamate receptors (mGluR1 and mGluR5) are functionally linked to estrogen receptors and play a key role in the plasticity of central neurons. Estrogen status strongly influences sensory input from the temporomandibular joint (TMJ) to neurons at the spinomedullary (Vc/C1-2) region. This study tested the hypothesis that TMJ input to trigeminal subnucleus caudalis/upper cervical cord (Vc/C1-2) neurons involved group I mGluR activation and depended on estrogen status. TMJ-responsive neurons were recorded in superficial laminae at the Vc/C1-2 region in ovariectomized (OvX) female rats treated with low-dose estradiol (2 µg/day, LE) or high-dose estradiol (20 µg/day, HE) for 2 days. TMJ-responsive units were activated by adenosine triphosphate (ATP, 1mM) injected into the joint space. Receptor antagonists selective for mGluR1 (CPCCOEt) or mGluR5 (MPEP) were applied topically to the Vc/C1-2 surface at the site of recording 10 min prior to the intra-TMJ ATP stimulus. In HE rats, CPCCOEt (50 and 500 µM) markedly reduced ATP-evoked unit activity. By contrast, in LE rats, a small but significant increase in neural activity was seen after 50 µM CPCCOEt, while 500 µM caused a large reduction in activity that was similar in magnitude as that seen in HE rats. Local application of MPEP produced a significant inhibition of TMJ-evoked unit activity independent of estrogen status. Neither mGluR1 nor mGluR5 antagonism altered the spontaneous activity of TMJ units in HE or LE rats. High-dose MPEP caused a small reduction in the size of the convergent cutaneous receptive field in HE rats, while CPCCOEt had no effect. These data suggest that group I mGluRs play a key role in sensory integration of TMJ nociceptive input to the Vc/C1-2 region and are largely independent of estrogen status.


Subject(s)
Neurons/physiology , Nociception/physiology , Receptors, Metabotropic Glutamate/physiology , Temporomandibular Joint/physiology , Trigeminal Caudal Nucleus/physiology , Adenosine Triphosphate/pharmacology , Animals , Chromones/pharmacology , Estradiol/administration & dosage , Estradiol/physiology , Female , Neurons/drug effects , Nociception/drug effects , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Temporomandibular Joint/drug effects , Temporomandibular Joint/innervation , Trigeminal Caudal Nucleus/drug effects
18.
Anim Sci J ; 86(3): 251-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25410821

ABSTRACT

Growing porcine oocytes from early antral follicles can acquire meiotic and developmental competence under suitable culture conditions, but at lower rates compared to full-grown oocytes. We postulated that estradiol-17ß (E2 ) supported the acquisition of meiotic and developmental competence as well as cumulus-expansion ability during growth culture. Growing oocytes from early antral follicles (1.2 to 1.5 mm in diameter) were grown in vitro for 5 days in a medium containing 0, 10(-7) , 10(-6) , 10(-5) or 10(-4) mol/L E2 ; after in vitro maturation, 35, 58, 47, 74 and 49% of oocytes matured to metaphase II, 25, 79, 77, 90 and 97% acquired cumulus-expansion ability, and 23, 54, 63, 89 and 64% were fully surrounded by cumulus cells, respectively. Following maturation, electro-stimulation was applied to the oocytes grown with 10(-5) mol/L E2 . After 6 days of culture, in vitro-grown oocytes developed to the blastocyst stage at a rate similar to that for full-grown oocytes (31% and 40%, respectively). Therefore, we suggest that the use of E2 during growth culture improves the meiotic and developmental competence of oocytes, cumulus-expansion ability, and cumulus cell attachment to the oocytes.


Subject(s)
Cumulus Cells/physiology , Estradiol/pharmacology , Meiosis/drug effects , Oocytes/cytology , Ovarian Follicle/cytology , Animals , Blastocyst , Cells, Cultured , Electric Stimulation , Estradiol/physiology , Female , Metaphase/drug effects , Oocytes/drug effects , Swine
19.
Ann Endocrinol (Paris) ; 76(6 Suppl 1): S39-50, 2015 Oct.
Article in French | MEDLINE | ID: mdl-26776288

ABSTRACT

Pregnancy is an immunological paradox that implies that a semi-allogeneic fetus is not rejected by the maternal immune system, from implantation of the embryo to delivery. Progesterone (P4), estradiol (E2) and human chorionic gonadotropin (hCG), contribute to the transformation of immune cells in a transient tolerance state, necessary to the maintenance of pregnancy. The effects of pregnancy hormones depend probably of their maternal plasma level. hCG is dangerous at high concentrations because it can stimulate autoantibodies production, whereas in physiological concentrations, hCG, P4 and E2 upregulate immune response expanding regulatory T and B cells, allowing the fetus to grow within the maternal uterus in a protective environment. A second example of fetal-maternal relation found recently is the role of maternal nutrition on development of the fetal hypothalamic neurons. Experiments in mice fed on a high fat diet reveal a critical timing when altered maternal metabolism affect formation of hypothalamic neurocircuits of the offspring and predispose him to long-term metabolic disorders.


Subject(s)
Chorionic Gonadotropin/physiology , Estradiol/physiology , Maternal-Fetal Exchange/physiology , Progesterone/physiology , Animals , Autoimmune Diseases , Female , Humans , Hypothalamus/embryology , Immunity , Mice , Pregnancy
20.
J Mol Endocrinol ; 53(3): 355-66, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25258388

ABSTRACT

EGR1 and PITX1 are transcription factors required for gonadotroph cell Lhb promoter activation. To determine changes in Egr1 and Pitx1 mRNA levels in central and peripheral pituitary stimulations, an in vivo model based on i.c.v. pulsatile (1 pulse/0.5 h over 2 h) GnRH agonist (1.5 nM buserelin) or antagonist (2 nM antide) microinjections was used. The microinjections were given to ovariectomised and 17ß-oestradiol (E2) (3×20 µg), ERA (ESR1) agonist propyl pyrazole triol (PPT) (3×0.5 mg), ERB (ESR2) agonist diarylpropionitrile (DPN) (3×0.5 mg) s.c. pre-treated rats 30 min after last pulse anterior pituitaries were excised. Relative mRNA expression was determined by quantitative RT-PCR (qRT-PCR). Results revealed a gene-specific response for GnRH and/or oestrogenic stimulations in vivo. Buserelin pulses enhanced Egr1 expression by 66% in ovariectomised rats, whereas the oestradiol-supplemented+i.c.v. NaCl-microinjected group showed a 50% increase in Egr1 mRNA expression. The oestrogenic signal was transmitted via ERA (ESR1) and ERB (ESR2) activation as administration of PPT and DPN resulted in 97 and 62%, respectively, elevation in Egr1 mRNA expression. A synergistic action of GnRH agonist and 17ß-oestradiol (E2) stimulation of the Egr1 gene transcription in vivo were found. GnRHR activity did not affect Pitx1 mRNA expression; regardless of NaCl, buserelin or antide i.c.v. pulses, s.c. oestrogenic supplementation (with E2, PPT or DPN) consistently decreased (by -46, -48 and -41% respectively) the Pitx1 mRNA in the anterior pituitary gland. Orchestrated Egr1 and Pitx1 activities depending on specific central and peripheral regulatory inputs could be responsible for physiologically variable Lhb gene promoter activation in vivo.


Subject(s)
Early Growth Response Protein 1/genetics , Estradiol/pharmacology , Luteinizing Hormone, beta Subunit/genetics , Paired Box Transcription Factors/genetics , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Animals , Early Growth Response Protein 1/metabolism , Estradiol/physiology , Female , Gene Expression Regulation/drug effects , Luteinizing Hormone/blood , Luteinizing Hormone, beta Subunit/metabolism , Paired Box Transcription Factors/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL