Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 410
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Neurosci Lett ; 773: 136518, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35150776

ABSTRACT

In normal hormonal conditions, increased neuronal activity in the ventromedial hypothalamus (VMH) induces lordosis whereas activation of the preoptic area (POA) exerts an opposite effect. In the present work, we explored the effect of bilateral infusion of different doses of the apelin-13 (0.37, 0.75, 1.5, and 15 µg) in both brain areas on the expression of lordosis behavior. Lordosis quotient and lordosis reflex score were performed at 30, 120, and 240 min. Weak lordosis was observed following the 0.37 µg dose of apelin-13 at 30 min in the VMH of EB-primed rats; however, the rest of the doses induced significant lordosis relative to the control group. At 120 min, all doses induced lordosis behavior, while at 240 min, the highest dose of 15 µg did not induce significant differences. Interestingly, only the 0.75 µg infusion of apelin in the POA induced significant lordosis at 120 and 240 min. These results indicate that apelin-13 acts preferably in HVM and slightly in POA to initiate lordosis behavior in estrogen-primed rats.


Subject(s)
Intercellular Signaling Peptides and Proteins , Lordosis , Preoptic Area , Animals , Estradiol/pharmacology , Estrogens/pharmacology , Hypothalamus/drug effects , Hypothalamus/pathology , Intercellular Signaling Peptides and Proteins/pharmacology , Lordosis/chemically induced , Preoptic Area/drug effects , Preoptic Area/pathology , Progesterone/pharmacology , Rats , Sexual Behavior, Animal/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/pathology
2.
Biomed Pharmacother ; 146: 112301, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34915415

ABSTRACT

Treatment of sleep disorders promotes the long-term use of commercially available sleep inducers that have several adverse effects, including addiction, systemic fatigue, weakness, loss of concentration, headache, and digestive problems. Therefore, we aimed to limit these adverse effects by investigating a natural product, the extract of the Hibiscus syriacus Linnaeus flower (HSF), as an alternative treatment. In the electric footshock model, we measured anxiety and assessed the degree of sleep improvement after administering HSF extract. In the restraint model, we studied the sleep rate using PiezoSleep, a noninvasive assessment system. In the pentobarbital model, we measured sleep improvement and changes in sleep-related factors. Our first model confirmed the desirable effects of HSF extract and its active constituent, saponarin, on anxiolysis and Wake times. HSF extract also increased REM sleep time. Furthermore, HSF extract and saponarin increased the expression of cortical GABAA receptor α1 (GABAAR α1) and c-Fos in the ventrolateral preoptic nucleus (VLPO). In the second model, HSF extract and saponarin restored the sleep rate and the sleep bout duration. In the third model, HSF extract and saponarin increased sleep maintenance time. Moreover, HSF extract and saponarin increased cortical cholecystokinin (CCK) mRNA levels and the expression of VLPO c-Fos. HSF extract also increased GABAAR α1 mRNA level. Our results suggest that HSF extract and saponarin are effective in maintaining sleep and may be used as a novel treatment for sleep disorder. Eventually, we hope to introduce HSF and saponarin as a clinical treatment for sleep disorders in humans.


Subject(s)
Apigenin/therapeutic use , Glucosides/therapeutic use , Hibiscus , Plant Extracts/therapeutic use , Sleep Wake Disorders/drug therapy , Sleep/drug effects , Animals , Apigenin/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Corticosterone/blood , Disease Models, Animal , Electroencephalography , Glucosides/pharmacology , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Pentobarbital , Plant Extracts/pharmacology , Preoptic Area/drug effects , Preoptic Area/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Sleep Aids, Pharmaceutical , Sleep Wake Disorders/blood , Sleep Wake Disorders/genetics , Sleep Wake Disorders/physiopathology , Stress, Psychological/blood , Stress, Psychological/complications , Stress, Psychological/genetics , Stress, Psychological/physiopathology
3.
Int J Mol Sci ; 22(8)2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33918982

ABSTRACT

Lithium (Li+) salt is widely used as a therapeutic agent for treating neurological and psychiatric disorders. Despite its therapeutic effects on neurological and psychiatric disorders, it can also disturb the neuroendocrine axis in patients under lithium therapy. The hypothalamic area contains GABAergic and glutamatergic neurons and their receptors, which regulate various hypothalamic functions such as the release of neurohormones, control circadian activities. At the neuronal level, several neurotransmitter systems are modulated by lithium exposure. However, the effect of Li+ on hypothalamic neuron excitability and the precise action mechanism involved in such an effect have not been fully understood yet. Therefore, Li+ action on hypothalamic neurons was investigated using a whole-cell patch-clamp technique. In hypothalamic neurons, Li+ increased the GABAergic synaptic activities via action potential independent presynaptic mechanisms. Next, concentration-dependent replacement of Na+ by Li+ in artificial cerebrospinal fluid increased frequencies of GABAergic miniature inhibitory postsynaptic currents without altering their amplitudes. Li+ perfusion induced inward currents in the majority of hypothalamic neurons independent of amino-acids receptor activation. These results suggests that Li+ treatment can directly affect the hypothalamic region of the brain and regulate the release of various neurohormones involved in synchronizing the neuroendocrine axis.


Subject(s)
GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Lithium/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Synapses/drug effects , Synapses/metabolism , Animals , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Inhibitory Postsynaptic Potentials/drug effects , Patch-Clamp Techniques , Preoptic Area/drug effects , Preoptic Area/metabolism , Receptors, Amino Acid/metabolism , Synaptic Transmission/drug effects
4.
Neuropeptides ; 84: 102096, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33059245

ABSTRACT

Leptin mediates the interaction between reproductive function and energy balance. However, leptin receptors are not expressed in neurons that produce gonadotropin-releasing hormone (GnRH), likely indicating an indirect action through interneurons. Among likely neurons that modulate the secretion of GnRH are NO (nitric oxide) neurons. We assessed whether estradiol and feeding conditions modulate a possible interaction between leptin and NO in brain areas related to the control of reproductive function. Estradiol-treated and untreated ovariectomized rats were normally fed or fasted for 48 h. Then, saline (control) or leptin (3 µg/1 µl) intracerebroventricular microinjections were administered, and after thirty minutes, the brains collected subsequent to the decapitation or transcardially perfusion. Leptin and estradiol increased NO synthase (nNOS) gene expression (RT-PCR) and content (Western blotting) in the medial preoptic area (MPOA) and medial basal hypothalamus (MBH) only in fasted rats. Leptin increased: 1-phosphorylated-signal transducer and activator of transcription-3(pSTAT3) (immunohistochemistry) in the MPOA and various hypothalamic nuclei [arcuate (ARC); ventromedial (VMH); dorsal/ventral dorsomedial (dDMH/vDMH); premammilar ventral (PMV)], effects potentiated by estradiol/fasting interaction; 2- nNOS/pSTAT3 coexpression in the MPOA only in estradiol-treated, fasted rats; 3- nNOS-immunoreactive cell expression in the VMH, DMH and PMV (areas related to reproductive function control) of estradiol -treated rats. Thus, when leptin is reduced during fasting, leptin replacement effectively increased the expression of nitric oxide, which activated the HPG axis only in the presence of estradiol. Estradiol modulates the nitrergic system, leptin sensitivity and consequently leptin's effects on the nitrergic system in hypothalamus and in particular vDMH and PMV.


Subject(s)
Estradiol/metabolism , Gonadotropin-Releasing Hormone/metabolism , Leptin/metabolism , Neurons/metabolism , Animals , Female , Hypothalamus/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism
5.
J Neuroendocrinol ; 32(11): e12908, 2020 11.
Article in English | MEDLINE | ID: mdl-33034148

ABSTRACT

In addition to its critical role in lactation, the anterior pituitary hormone prolactin also influences a broad range of other physiological processes. In particular, widespread expression of prolactin receptor (Prlr) in the brain has highlighted pleiotropic roles for prolactin in regulating neuronal function, including maternal behaviour, reproduction and energy balance. Research into the central actions of prolactin has predominately focused on effects on gene transcription via the canonical JAK2/STAT5; however, it is evident that prolactin can exert rapid actions to stimulate activity in specific populations of neurones. We aimed to investigate how widespread these rapid actions of prolactin are in regions of the brain with large populations of prolactin-sensitive neurones, and whether physiological state alters these responses. Using transgenic mice where the Cre-dependent calcium indicator, GCaMP6f, was conditionally expressed in cells expressing the long form of the Prlr, we monitored changes in levels of intracellular calcium ([Ca2+ ]i ) in ex vivo brain slice preparations as a surrogate marker of cellular activity. Here, we surveyed hypothalamic regions implicated in the diverse physiological functions of prolactin such as the arcuate (ARC) and paraventricular nuclei of the hypothalamus (PVN), as well as the medial preoptic area (MPOA). We observed that, in the ARC of males and in both virgin and lactating females, prolactin can exert rapid actions to stimulate neuronal activity in the majority of Prlr-expressing neurones. In the PVN and MPOA, we found a smaller subset of cells that rapidly respond to prolactin. In these brain regions, the effects we detected ranged from rapid or sustained increases in [Ca2+ ]i to inhibitory effects, indicating a heterogeneous nature of these Prlr-expressing populations. These results enhance our understanding of mechanisms by which prolactin acts on hypothalamic neurones and provide insights into how prolactin might influence neuronal circuits in the mouse brain.


Subject(s)
Hypothalamus/drug effects , Hypothalamus/metabolism , Neurons/metabolism , Prolactin/pharmacology , Receptors, Prolactin/drug effects , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Calcium Signaling , Female , Humans , Hypothalamus/cytology , Immunohistochemistry , Lactation , Male , Mice , Mice, Transgenic , Neurons/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Receptors, Prolactin/genetics
6.
J Exp Zool A Ecol Integr Physiol ; 333(8): 550-560, 2020 10.
Article in English | MEDLINE | ID: mdl-32798281

ABSTRACT

The developing brain is highly sensitive to the hormonal milieu, with gonadal steroid hormones involved in neurogenesis, neural survival, and brain organization. Limited available evidence suggests that endocrine-disrupting chemicals (EDCs) may perturb these developmental processes. In this study, we tested the hypothesis that prenatal exposure to a mixture of polychlorinated biphenyls (PCBs), Aroclor 1221, would disrupt the normal timing of neurogenesis in two hypothalamic regions: the ventromedial nucleus (VMN) and the preoptic area (POA). These regions were selected because of their important roles in the control of sociosexual behaviors that are perturbed in adulthood by prenatal EDC exposure. Pregnant Sprague-Dawley rats were exposed to PCBs from Embryonic Day 8 (E8) to E18, encompassing the period of neurogenesis of all hypothalamic neurons. To determine the birth dates of neurons, bromo-2-deoxy-5-uridine (BrdU) was administered to dams on E12, E14, or E16. On the day after birth, male and female pups were perfused, brains immunolabeled for BrdU, and numbers of cells counted. In the VMN, exposure to PCBs significantly advanced the timing of neurogenesis compared to vehicle-treated pups, without changing the total number of BrdU+ cells. In the POA, PCBs did not change the timing of neurogenesis nor the total number of cells born. This is the first study to show that PCBs can shift the timing of neurogenesis in the hypothalamus, specifically in the VMN but not the POA. This result has implications for functions controlled by the VMN, especially sociosexual behaviors, as well as for sexual selection more generally.


Subject(s)
Endocrine Disruptors/pharmacology , Hypothalamus/drug effects , Neurogenesis/drug effects , Animals , Aroclors/pharmacology , Female , Fetus/drug effects , Neurons/drug effects , Polychlorinated Biphenyls/pharmacology , Pregnancy , Preoptic Area/cytology , Preoptic Area/drug effects , Rats , Rats, Sprague-Dawley , Sexual Behavior/drug effects , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/drug effects
7.
Horm Behav ; 119: 104637, 2020 03.
Article in English | MEDLINE | ID: mdl-31783026

ABSTRACT

Phytoestrogens comprise biologically active constituents of human and animal diet that can impact on systemic and local estrogen functions in the brain. Here we report on the importance of dietary phytoestrogens for maintaining activity in a brain circuit controlling aggressive and social behavior of male mice. After six weeks of low-phytoestrogen chronic diet (diadzein plus genistein <20 µg/g) a reduction of intermale aggression and altered territorial marking behavior could be observed, compared to littermates on a standard soy-bean based diet (300 µg/g). Further, mice on low-phyto diet displayed a decrease in sociability and a reduced preference for social odors, indicating a general disturbance of social behavior. Underlying circuits were investigated by analysing the induction of the activity marker c-Fos upon social encounter. Low-phyto diet led to a markedly reduced c-Fos induction in the medial as well as the cortical amygdala, the lateral septum, medial preoptic area and bed nucleus of the stria terminalis. No difference between groups was observed in the olfactory bulb. Together our data suggest that dietary phytoestrogens critically modulate social behavior circuits in the male mouse brain.


Subject(s)
Aggression/drug effects , Nerve Net/drug effects , Phytochemicals/pharmacology , Phytoestrogens/pharmacology , Social Behavior , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Corticomedial Nuclear Complex/cytology , Corticomedial Nuclear Complex/drug effects , Corticomedial Nuclear Complex/metabolism , Diet , Isoflavones/pharmacology , Male , Mice , Mice, Inbred C57BL , Nerve Net/physiology , Preoptic Area/cytology , Preoptic Area/drug effects , Preoptic Area/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Territoriality
8.
Acta Physiol (Oxf) ; 228(3): e13373, 2020 03.
Article in English | MEDLINE | ID: mdl-31483934

ABSTRACT

AIM: Whereas some patients have important changes in body core temperature (Tb) during systemic inflammation, others maintain a normal Tb, which is intrinsically associated to immune paralysis. One classical model to study immune paralysis is the use of repeated administration of lipopolysaccharide (LPS), the so-called endotoxin tolerance. However, the neuroimmune mechanisms of endotoxin tolerance remain poorly understood. Hydrogen sulphide (H2 S) is a gaseous neuromodulator produced in the brain by the enzyme cystathionine ß-synthase (CBS). The present study assessed whether endotoxin tolerance is modulated by hypothalamic H2 S. METHODS: Rats with central cannulas (drug microinjection) and intraperitoneal datalogger (temperature record) received a low-dose of lipopolysaccharide (LPS; 100 µg kg-1 ) daily for four consecutive days. Hypothalamic CBS expression and H2 S production rate were assessed, together with febrigenic signalling. Tolerant rats received an inhibitor of H2 S synthesis (AOA, 100 pmol 1 µL-1 icv) or its vehicle in the last day. RESULTS: Antero-ventral preoptic area of the hypothalamus (AVPO) H2 S production rate and CBS expression were increased in endotoxin-tolerant rats. Additionally, hypothalamic H2 S inhibition reversed endotoxin tolerance reestablishing fever, AVPO and plasma PGE2 levels without altering the absent plasma cytokines surges. CONCLUSION: Endotoxin tolerance is not simply a reflection of peripheral reduced cytokines release but actually results from a complex set of mechanisms acting at multiple levels. Hypothalamic H2 S production modulates most of these mechanisms.


Subject(s)
Dinoprostone/biosynthesis , Endotoxins/pharmacology , Hydrogen Sulfide/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Animals , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cytokines/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Disease Models, Animal , Drug Tolerance , Fever/drug therapy , Fever/metabolism , Lipopolysaccharides/pharmacology , Male , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats , Rats, Wistar
9.
Psychoneuroendocrinology ; 102: 84-94, 2019 04.
Article in English | MEDLINE | ID: mdl-30529907

ABSTRACT

Estrogens play important roles in regulating brain development, brain function, and behavior. Many studies have evaluated these effects using ovariectomized (OVX) rats or mice with different doses of estrogen replacement, assuming that estradiol levels in all regions of the brain are the same as levels achieved in the serum. It is well known, however, that the brain contains all the enzymes necessary to produce estrogens, and that estrogen levels in the brain are determined by both systemic and local production and are region-specific. The present study conducted a detailed analysis of the relationship between systemic levels of 17-ß-estradiol (E2) achieved by estrogen replacement and levels achieved in specific regions of the brain. Levels of E2 were measured in both brain and serum in OVX rats treated with different doses of estradiol benzoate (EB) using a novel and recently validated UPLC-MS/MS method. Results confirmed significantly higher levels of E2 in the brain than in serum in brain regions known to contain aromatase (ARO) activity, both in OVX controls and in rats treated with physiological doses of EB. Additional studies compared the level of E2 and testosterone (T) in the brain and serum between testosterone propionate (TP) treated OVX and male. This demonstrated higher levels of E2 in certain brain regions of males than in TP treated OVX females even though T levels in the brain and serum were similar between the two groups. Studies also demonstrated that the differences between serum and brain levels of E2 can be eliminated by letrozole (ARO inhibitor) treatment, which indicates that the differences are due to local ARO activity. Collectively the results provide a detailed analysis of brain region-specific E2 concentrations in OVX, E2-, and T-treated rats and demonstrate the degree to which these concentrations are ARO-dependent.


Subject(s)
Brain/metabolism , Estradiol/analysis , Estradiol/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Aromatase/metabolism , Aromatase Inhibitors/metabolism , Brain/drug effects , Cerebellum/drug effects , Cerebellum/metabolism , Chromatography/methods , Chromatography, Liquid/methods , Estradiol/pharmacology , Estrogen Replacement Therapy , Estrogens , Female , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Letrozole/pharmacology , Male , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry/methods , Testosterone/pharmacology , Testosterone Propionate
10.
Psychopharmacology (Berl) ; 236(4): 1293-1301, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30539267

ABSTRACT

RATIONALE: Lycium barbarum polysaccharide (LBP) is known to promote reproductive functions. However, its role in noncontact erection (NCE) of penis initiated by brain regions including medial preoptic area (MPOA) and paraventricular nucleus (PVN) regions responsible for sexual behavior has not been investigated. OBJECTIVES: Therefore, this study initially investigated the effects of LBP on male sexual function, and subsequently, the mechanistic insight was investigated through assessing the expression of neuronal nitric oxide synthase (nNOS) in the MPOA and PVN. METHODS: The adult male rats were treated with 100 mg/kg of LBP or vehicle by oral gavage. Before and after 14 days of treatment, copulatory behavior and noncontact erection (NCE) were recorded. After the last behavioral test, the brain was isolated to measure nNOS expression in the MPOA and PVN. RESULTS: Data showed that LBP treatment significantly increased both the frequencies of intromission as well as ejaculation, compared to the control group. Whereas, a reduced post-ejaculatory interval was observed compared to same group on day 0. Furthermore, the treatment led to an increased intromission ratio, inter-intromission interval, and the number of MPOA nNOS-immunoreactive cells (nNOS-ir). Additionally, a significantly positive correlation between ejaculation frequency and MPOA nNOS-ir cells was recorded. Of note, LBP treatment had no effects on NCE and PVN nNOS-ir expression. CONCLUSION: These findings suggest that LBP enhances sexual behavior through increased nNOS expression in the MPOA in male rats.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Nitric Oxide Synthase Type I/metabolism , Penile Erection/drug effects , Preoptic Area/drug effects , Sexual Behavior, Animal/drug effects , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Male , Neurons/drug effects , Neurons/enzymology , Nitric Oxide , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/enzymology , Penile Erection/physiology , Preoptic Area/enzymology , Rats , Rats, Long-Evans , Sexual Behavior, Animal/physiology , Testis/drug effects , Testis/enzymology
11.
Mediators Inflamm ; 2018: 9150207, 2018.
Article in English | MEDLINE | ID: mdl-30402044

ABSTRACT

The study was designed to examine whether the administration of neostigmine (0.5 mg/animal), a peripheral inhibitor of acetylcholinesterase (AChE), during an immune/inflammatory challenge provoked by intravenous injection of bacterial endotoxin-lipopolysaccharide (LPS; 400 ng/kg)-attenuates the synthesis of proinflammatory cytokines in the ovine preoptic area (POA), the hypothalamic structure playing an essential role in the control of the reproduction process, and in the choroid plexus (CP), a multifunctional organ sited at the interface between the blood and cerebrospinal fluid in the ewe. Neostigmine suppressed (p < 0.05) LPS-stimulated synthesis of cytokines such as interleukin- (IL-) 1ß, IL-6, and tumor necrosis factor (TNF) α in the POA, and this effect was similar to that induced by the treatment with systemic AChE inhibitor-donepezil (2.5 mg/animal). On the other hand, both AChE inhibitors did not influence the gene expression of these cytokines and their corresponding receptors in the CP. It was found that this structure seems to not express the neuronal acetylcholine (ACh) receptor subunit alpha-7, required for anti-inflammatory action of ACh. The mechanism of action involves inhibition of the proinflammatory cytokine synthesis on the periphery as well as inhibition of their de novo synthesis rather in brain microvessels and not in the CP. In conclusion, it is suggested that the AChE inhibitors incapable of reaching brain parenchyma might be used in the treatment of neuroinflammatory processes induced by peripheral inflammation.


Subject(s)
Choroid Plexus/metabolism , Cytokines/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Neostigmine/therapeutic use , Preoptic Area/metabolism , Animals , Cholinesterase Inhibitors/therapeutic use , Choroid Plexus/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Preoptic Area/drug effects , Sheep , Tumor Necrosis Factor-alpha/metabolism
12.
J Endocrinol ; 231(2): 167-180, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27613338

ABSTRACT

Water deprivation (WD) induces changes in plasma volume and osmolality, which in turn activate several responses, including thirst, the activation of the renin-angiotensin system (RAS) and vasopressin (AVP) and oxytocin (OT) secretion. These systems seem to be influenced by oestradiol, as evidenced by the expression of its receptor in brain areas that control fluid balance. Thus, we investigated the effects of oestradiol treatment on behavioural and neuroendocrine changes of ovariectomized rats in response to WD. We observed that in response to WD, oestradiol treatment attenuated water intake, plasma osmolality and haematocrit but did not change urinary volume or osmolality. Moreover, oestradiol potentiated WD-induced AVP secretion, but did not alter the plasma OT or angiotensin II (Ang II) concentrations. Immunohistochemical data showed that oestradiol potentiated vasopressinergic neuronal activation in the lateral magnocellular PVN (PaLM) and supraoptic (SON) nuclei but did not induce further changes in Fos expression in the median preoptic nucleus (MnPO) or subfornical organ (SFO) or in oxytocinergic neuronal activation in the SON and PVN of WD rats. Regarding mRNA expression, oestradiol increased OT mRNA expression in the SON and PVN under basal conditions and after WD, but did not induce additional changes in the mRNA expression for AVP in the SON or PVN. It also did not affect the mRNA expression of RAS components in the PVN. In conclusion, our results show that oestradiol acts mainly on the vasopressinergic system in response to WD, potentiating vasopressinergic neuronal activation and AVP secretion without altering AVP mRNA expression.


Subject(s)
Dehydration/physiopathology , Estradiol/therapeutic use , Estrogens/therapeutic use , Neurons/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Supraoptic Nucleus/drug effects , Water-Electrolyte Imbalance/prevention & control , Animals , Arginine Vasopressin/agonists , Arginine Vasopressin/analysis , Arginine Vasopressin/metabolism , Behavior, Animal/drug effects , Dehydration/therapy , Drinking/drug effects , Estrogen Replacement Therapy , Female , Fluid Therapy , Gene Expression Regulation/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Ovariectomy/adverse effects , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Preoptic Area/drug effects , Preoptic Area/metabolism , Preoptic Area/pathology , Rats, Wistar , Subfornical Organ/drug effects , Subfornical Organ/metabolism , Subfornical Organ/pathology , Supraoptic Nucleus/metabolism , Supraoptic Nucleus/pathology , Vestibular Nucleus, Lateral/drug effects , Vestibular Nucleus, Lateral/metabolism , Vestibular Nucleus, Lateral/pathology , Water-Electrolyte Imbalance/blood , Water-Electrolyte Imbalance/etiology , Water-Electrolyte Imbalance/physiopathology
13.
Ann Anat ; 208: 31-39, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27562857

ABSTRACT

Perimenopausal syndromes begin as ovarian function ceases and the most common symptoms are hot flushes. Data indicate that the projections of serotonin to hypothalamus may be involved in the mechanism of hot flushes. Therefore, the aim of this study is to investigate the potential role of the serotonin dorsal raphe-preoptic hypothalamus pathway for hot flushes in an animal model of menopause. We determined the changes in serotonin expression in the dorsal raphe (DR) and preoptic anterior hypothalamus (POAH) in ovariectomized rats. We also explored the therapeutical effects of estradiol valerate and Remifemin in this model. Eighty female Sprague-Dawley rats were randomly assigned to sham-operated (SHAM) group, ovariectomy (OVX) group with vehicle, ovariectomy with estradiol valerate treatment (OVX+E) group and ovariectomy with Remifemin (OVX+ICR) group. Serotonin expression was evaluated in the DR and POAH using immunofluorescence and quantified in the DR using an enzyme-linked immunosorbent assay (ELISA). Apoptosis was analyzed in the DR by TUNEL assay. The number of serotonin immunoreactive neurons and the level of serotonin expression in the DR decreased significantly following OVX compared to the SHAM group. No TUNEL-positive cells were detected in the DR in any group. In addition, following OVX, the number of serotonin-positive fibers decreased significantly in the ventromedial preoptic nucleus (VMPO), especially in the ventrolateral preoptic nucleus (VLPO). Treatment with either estradiol or Remifemin for 4 weeks countered the OVX-induced decreases in serotonin levels in both the DR and the hypothalamus, with levels in the treated rats similar to those in the SHAM group. A fluorescently labeled retrograde tracer was injected into the VLPO at the 4-week time point. A significantly lower percentage of serotonin with CTB double-labeled neurons in CTB-labeled neurons was demonstrated after ovariectomy, and both estradiol and Remifemin countered this OVX-induced decrease. We conclude that serotonin pathway is changed after ovariectomy, including the serotonin synthesis in DR and serotonin fibers in PO/AH, both E and Remifemin have an equivalent therapeutic effect on it.


Subject(s)
Dorsal Raphe Nucleus/metabolism , Estradiol/analogs & derivatives , Hot Flashes/metabolism , Hypothalamus/metabolism , Plant Extracts/administration & dosage , Preoptic Area/metabolism , Serotonin/metabolism , Animals , Cimicifuga , Dorsal Raphe Nucleus/cytology , Dorsal Raphe Nucleus/drug effects , Down-Regulation/drug effects , Estradiol/administration & dosage , Female , Hot Flashes/drug therapy , Hypothalamus/cytology , Hypothalamus/drug effects , Menopause/drug effects , Menopause/metabolism , Neural Pathways/cytology , Neural Pathways/metabolism , Ovariectomy , Preoptic Area/cytology , Preoptic Area/drug effects , Rats , Rats, Sprague-Dawley , Serotonergic Neurons/cytology , Serotonergic Neurons/metabolism , Tissue Distribution
14.
Brain Res ; 1650: 21-30, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27566061

ABSTRACT

The time course effects of ovarian steroids on kisspeptin and GnRH/LH systems is not totally clarified. We investigated the temporal relationship among kisspeptin and GnRH mRNA and kisspeptin content in the preoptic area (POA), GnRH content and release in the medial basal hypothalamus (MBH) and plasma LH levels under different steroid treatments. Ovariectomized rats treated with oil (OVOO), oil plus single dose of estradiol (OVOE), oil plus single dose of progesterone (OVOP), estradiol for 3 days plus oil (OVEO) or estradiol for 3 days plus progesterone (OVEP) were hourly decapitated from 10:00 to 17:00 or had the MBH microdialyzed from 09:00 to 19:00. Estradiol and progesterone acutely increased POA kisspeptin content without altering POA kisspeptin mRNA levels. Short-term exposure to both hormones stimulated MBH GnRH content, although no GnRH/LH surges had occurred. Chronic estradiol-treatment increased both kisspeptin mRNA levels and content in the POA, demonstrating that long exposure to estradiol is required to activate the whole kisspeptin synthesis machinery. This was followed by the peak in the GnRH/LH release. In estradiol-primed rats, progesterone further increased POA kisspeptin content, amplified and advanced GnRH/LH surges, with no additional change on POA kisspeptin mRNA. The data show an estradiol-induced temporal association between kisspeptin increase in the POA and GnRH/LH surges. Interestingly, the classic action of progesterone in amplifying and accelerating the GnRH/LH surges seems to occur by a mechanism which involves POA kisspeptin system.


Subject(s)
Estradiol/metabolism , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/metabolism , Animals , Estradiol/pharmacology , Female , Gonadotropins , Hypothalamus/drug effects , Hypothalamus, Middle/drug effects , Kisspeptins/drug effects , Kisspeptins/metabolism , Luteinizing Hormone/blood , Ovariectomy , Preoptic Area/drug effects , Progesterone/metabolism , Progesterone/pharmacology , Rats , Rats, Wistar , Spatio-Temporal Analysis
15.
J Neuroendocrinol ; 28(3): 12357, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26716764

ABSTRACT

The adipocyte-derived hormone leptin plays a critical role in the control of reproduction via signalling in hypothalamic neurones. The drivers of the hypothalamic-pituitary-gonadal axis, the gonadotrophin-releasing hormone (GnRH) neurones, do not have the receptors for leptin. Therefore, intermediate leptin responsive neurones must provide leptin-to-GnRH signalling. We investigated the populations of leptin responsive neurones that provide input to the rostral preoptic area (rPOA) where GnRH cell bodies reside. Fluorescent retrograde tracer beads (RetroBeads; Lumafluor Inc., Naples, FL, USA) were injected into the rPOA of transgenic leptin receptor enhanced green fluorescent protein (Lepr-eGFP) reporter mice. Uptake of the RetroBeads by Lepr-eGFP neurones was assessed throughout the hypothalamus. RetroBead uptake was most evident in the medial arcuate nucleus (ARC), the dorsomedial nucleus (DMN) and the ventral premammillary nucleus (PMV) of the hypothalamus. The uptake of RetroBeads specifically by Lepr-eGFP neurones was highest in the medial ARC (18% of tracer-labelled neurones Lepr-eGFP-positive). Because neurones that are both leptin responsive and GABAergic play a critical role in the regulation of fertility by leptin, we next focussed on the location of these populations. To address whether GABAergic neurones in leptin-responsive hypothalamic regions project to the rPOA, the experiment was repeated in GABA neurone reporter mice (Vgat-tdTomato). Between 10% and 45% of RetroBead-labelled neurones in the ARC were GABAergic, whereas uptake of tracer by GABAergic neurones in the DMN and PMV was very low (< 5%). These results show that both leptin responsive and GABAergic neurones from the ARC project to the region of the GnRH cell bodies. Our findings suggest that LEPR-expressing GABA neurones from the ARC may be mediators of leptin-to-GnRH signalling.


Subject(s)
GABAergic Neurons/drug effects , Leptin/pharmacology , Preoptic Area/drug effects , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Cell Tracking/methods , Female , GABAergic Neurons/physiology , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Preoptic Area/cytology , Receptors, Leptin/genetics
16.
Life Sci ; 141: 1-7, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26409312

ABSTRACT

AIMS: Administration of estradiol or compounds with estrogenic activity to newborn female rats results in irreversible masculinization as well as defeminization in the brain and the animals exhibit altered reproductive behavior as adults. The cellular and molecular mechanism involved in inducing the irreversible changes is largely unknown. In the present study, we have monitored the changes in the expression of selected synaptogenesis related genes in the sexually dimorphic brain regions such as POA, hypothalamus and pituitary following 17ß-estradiol administration to neonatal female rats. MAIN METHODS: Female Wistar rats which were administered 17ß-estradiol on day 2 and 3 after birth were sacrificed 120days later and the expression levels of genes implicated in synaptogenesis were monitored by semi-quantitative reverse transcription PCR. Since estradiol induced up-regulation of COX-2 in POA is a marker for estradiol induced masculinization as well as defeminization, in the present study only animals in which the increase in expression of COX-2 gene was observed in POA were included in the study. KEY FINDINGS: Down-regulation of genes such as NMDA-2B, NETRIN-1, BDNF, MT-5 MMP and TNF-α was observed in the pre-optic area of neonatally E2 treated female rat brain but not in hypothalamus and pituitary compared to the vehicle- treated controls as assessed by RT-PCR and Western blot analysis. SIGNIFICANCE: Our results suggest a possibility that down-regulation of genes associated with synaptogenesis in POA, may be resulting in disruption of the cyclical regulation of hormone secretion by pituitary the consequence of which could be infertility and altered reproductive behavior.


Subject(s)
Animals, Newborn/physiology , Brain/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Neurogenesis/genetics , Synapses/drug effects , Animals , Brain/growth & development , Cyclooxygenase 2/biosynthesis , Down-Regulation/drug effects , Female , Feminization , Gene Expression/drug effects , Hypothalamus/drug effects , Hypothalamus/growth & development , Male , Neurogenesis/drug effects , Pituitary Gland/drug effects , Pituitary Gland/growth & development , Polymerase Chain Reaction , Pregnancy , Preoptic Area/drug effects , Preoptic Area/growth & development , Rats , Sex Differentiation/drug effects
17.
Neuroendocrinology ; 101(4): 331-46, 2015.
Article in English | MEDLINE | ID: mdl-25791982

ABSTRACT

Elevated levels of thyroid hormones (TH) reduce estradiol (E2)-dependent female sexual behavior. E2 stimulates progesterone receptor (Pgr) and oxytocin receptor (Oxtr) within the ventromedial hypothalamus and preoptic area, critical hypothalamic nuclei for sexual and maternal behavior, respectively. Here, we investigated the impact of TH on E2-dependent transcriptional mechanisms in female mice. First, we observed that triiodothyronine (T3) inhibited the E2 induction of Pgr and Oxtr. We hypothesized that differences in histone modifications and receptor recruitment could explain the influence of TH on E2-responsive Pgr and Oxtr expression. We observed that histone H3 acetylation (H3Ac) and methylation (H3K4me3) was gene and brain-region specific. We then analyzed the recruitment of estrogen receptor α (ERα) and TH receptor α (TRα) on the putative regulatory sequences of Pgr and Oxtr. Interestingly, T3 inhibited E2-induced ERα binding to a specific Pgr enhancer site, whereas TRα binding was not affected, corroborating our theory that the competitive binding of TRα to an ERα binding site can inhibit ERα transactivation and the subsequent E2-responsive gene expression. On the Oxtr promoter, E2 and T3 worked together to modulate ERα and TRα binding. Finally, the E2-dependent induction of cofactors was reduced by hypothyroidism and T3. Thus, we determined that the Pgr and Oxtr promoter regions are responsive to E2 and that T3 interferes with the E2 regulation of Pgr and Oxtr expression by altering the recruitment of receptors to DNA and changing the availability of cofactors. Collectively, our findings provide insights into molecular mechanisms of response to E2 and TH interactions controlling sex behavior in the hypothalamus.


Subject(s)
Estrogen Receptor alpha/metabolism , Hypothalamus/metabolism , Preoptic Area/metabolism , Receptors, Oxytocin/metabolism , Receptors, Progesterone/metabolism , Acetylation/drug effects , Animals , DNA Methylation/drug effects , DNA Methylation/physiology , Estradiol/metabolism , Female , Histones/drug effects , Histones/metabolism , Hormones/pharmacology , Hypothalamus/drug effects , Mice , Preoptic Area/drug effects , Promoter Regions, Genetic , Receptors, Oxytocin/genetics , Receptors, Progesterone/genetics , Thyroid Hormone Receptors alpha/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Triiodothyronine/pharmacology
18.
Neuroendocrinology ; 101(2): 120-32, 2015.
Article in English | MEDLINE | ID: mdl-25613345

ABSTRACT

AIMS: We investigated the norepinephrine pathway changes from the locus coeruleus (LC) to the preoptic area of the hypothalamus (POAH) in the brain of ovariectomized rats under low estrogen levels and explored the therapeutic effects of estradiol valerate (E2) and Remifemin (ICR) on these changes. METHODS: 40 female Sprague-Dawley rats were randomly divided into the following groups: surgery with vehicle (SHAM), ovariectomy surgery with vehicle (OVX), ovariectomy with E2 treatment (OVX + E2), and ovariectomy with Remifemin (OVX + ICR). After 4 weeks of treatment, we observed the changes by immunohistochemistry. RESULTS: (1) The average optical density of DBH-ir fibers and the number of α1-adrenoreceptor- and estrogen receptor (ER)α-positive neurons in the main nuclei of POAH were all reduced in OVX rats compared with the SHAM group. The above changes were normalized in all nuclei of the POAH in the E2 group, while they were normalized in some nuclei in the ICR group. Coexpression of ERα and α1-adrenoreceptor was observed in the POAH. (2) The number of DBH- and ERα-positive neurons in the LC decreased in the OVX group compared with the SHAM group and increased after treatment with E2 and ICR. Coexpression of ERα and DBH was observed in the LC. CONCLUSION: Low estrogen (OVX) altered norepinephrine synthesis in the LC, the projection of norepinephrine fibers and α1-adrenoreceptor expression in the POAH. Both E2 and ICR normalized the norepinephrine pathway, but E2 achieved greater effects than ICR. ICR had different effects in different nuclei in the POAH and its therapeutic effect was better in the LC.


Subject(s)
Estradiol/analogs & derivatives , Estrogens/pharmacology , Locus Coeruleus/drug effects , Norepinephrine/metabolism , Plant Extracts/pharmacology , Preoptic Area/drug effects , Animals , Cimicifuga , Dopamine beta-Hydroxylase/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Immunohistochemistry , Locus Coeruleus/metabolism , Locus Coeruleus/pathology , Neuroanatomical Tract-Tracing Techniques , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Ovariectomy , Preoptic Area/metabolism , Preoptic Area/pathology , Random Allocation , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction/drug effects
19.
Neurosci Lett ; 583: 194-8, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25281790

ABSTRACT

The role of the serotonergic system in regulating the expression of estrogen receptor (ER) α in the hypothalamus was investigated in ovariectomized rats by injecting a serotonin synthesis inhibitor, parachlorophenylalanine (PCPA), or by destroying the dorsal raphe nucleus (DR). The number of ERα-immunoreactive (ir) cells was counted in the anteroventral periventricular nucleus in the preoptic area (AVPV), ventrolateral ventromedial hypothalamic nucleus (vlVMN), and arcuate nucleus (ARCN). Seven days after ovariectomy, 100mg/kg PCPA or saline was injected daily for 4 days. Alternatively, radiofrequency lesioning of the DR (DRL) or sham lesions were made on the same time of ovariectomy. One-day after the last injection of PCPA or 7 days after brain surgery, the brain was fixed for immunostaining of ERα and the number of ERα-ir cell were counted in the nuclei of interest. The mean number of ERα-ir cells/mm(3) (density) in the AVPV of the PCPA or DRL groups was statistically higher than that in the saline or sham group. In the vlVMN and ARCN of the PCPA or DRL groups, the mean density of ERα-ir cells was comparable to the saline or sham groups. These results suggest that the serotonergic system of the DR plays an inhibitory role on the expression of ERα in the AVPV, but not in the vlVMN and ARCN.


Subject(s)
Estrogen Receptor alpha/metabolism , Fenclonine/pharmacology , Hypothalamus/metabolism , Serotonin Antagonists/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Dorsal Raphe Nucleus/physiology , Female , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus, Anterior/cytology , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/metabolism , Ovariectomy , Preoptic Area/cytology , Preoptic Area/drug effects , Preoptic Area/metabolism , Rats, Wistar , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/metabolism
20.
J Neuroendocrinol ; 26(11): 776-84, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25040132

ABSTRACT

Neurokinin B (NKB) is essential for human reproduction and has been shown to stimulate luteinising hormone (LH) secretion in several species, including sheep. Ewes express the neurokinin-3 receptor (NK3R) in the retrochiasmatic area (RCh) and there is one report that placement of senktide, an NK3R agonist, therein stimulates LH secretion that resembles an LH surge in ewes. In the present study, we first confirmed that local administration of senktide to the RCh produced a surge-like increase in LH secretion, and then tested the effects of this agonist in two other areas implicated in the control of LH secretion and where NK3R is found in high abundance: the preoptic area (POA) and arcuate nucleus (ARC). Bilateral microimplants containing senktide induced a dramatic surge-like increase in LH when given in the POA similar to that seen with RCh treatment. By contrast, senktide treatment in the ARC resulted in a much smaller but significant increase in LH concentrations suggestive of an effect on tonic secretion. The possible role of POA and RCh NK3R activation in the LH surge was next tested by treating ewes with SB222200, an NK3R antagonist, in each area during an oestradiol-induced LH surge. SB222200 in the RCh, but not in the POA, reduced the LH surge amplitude by approximately 40% compared to controls, indicating that NK3R activation in the former region is essential for full expression of the pre-ovulatory LH surge. Based on these data, we propose that the actions of NKB in the RCh are an important component of the pre-ovulatory LH surge in ewes.


Subject(s)
Hypothalamus/drug effects , Luteinizing Hormone/blood , Ovulation/drug effects , Peptide Fragments/pharmacology , Receptors, Neurokinin-3/agonists , Substance P/analogs & derivatives , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Female , Hypothalamus/metabolism , Neurons/drug effects , Neurons/metabolism , Ovulation/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Quinolines/pharmacology , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/metabolism , Sheep , Substance P/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL