Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 251
Filter
1.
Reprod Domest Anim ; 50(1): 68-75, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25399917

ABSTRACT

The aim of this study was to investigate how acute insulin-induced hypoglycaemia (IIH) alters the activity of cells containing oestradiol receptor α (ERα) or somatostatin (SST) in the arcuate nucleus (ARC) and ventromedial nucleus (VMN), and ERα cells in the medial preoptic area (mPOA) of intact ewes. Follicular phases were synchronized with progesterone vaginal pessaries. Control animals were killed at 0 h or 31 h (n = 5 and 6, respectively) after progesterone withdrawal (PW; time zero). At 28 h, five other animals received insulin (INS; 4 iu/kg) and were subsequently killed at 31 h. Hypothalamic sections were immunostained for ERα or SST each with c-Fos, a marker of neuronal transcriptional activation. Insulin did not alter the percentage of activated ERα cells in the ARC; however, it appeared visually that two insulin-treated animals (INS responders, with no LH surge) had an increase in the VMN (from 32 to 78%) and a decrease in the mPOA (from 40 to 12%) compared to no increase in the two INS non-responders (with an LH surge). The percentage of activated SST cells in the ARC was greater in all four insulin-treated animals (from 10 to 60%), whereas it was visually estimated that activated SST cells in the VMN increased only in the two insulin responders (from 10 to 70%). From these results, we suggest that IIH stimulates SST activation in the ARC as part of the glucose-sensing mechanism but ERα activation is unaffected in this region. We present evidence to support a hypothesis that disruption of the GnRH/LH surge may occur in insulin responders via a mechanism that involves, at least in part, SST cell activation in the VMN along with decreased ERα cell activation in the mPOA.


Subject(s)
Estrogen Receptor alpha/analysis , Hypothalamus/chemistry , Insulin/pharmacology , Proto-Oncogene Proteins c-fos/analysis , Sheep/metabolism , Somatostatin/analysis , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Blood Glucose/analysis , Estradiol/blood , Female , Follicular Phase/metabolism , Gene Expression , Hydrocortisone/blood , Luteinizing Hormone/blood , Preoptic Area/chemistry , Progesterone/blood , Ventromedial Hypothalamic Nucleus/chemistry
2.
Neuroendocrinology ; 94(4): 323-32, 2011.
Article in English | MEDLINE | ID: mdl-22116451

ABSTRACT

Kisspeptin has been thought to play pivotal roles in the control of both pulse and surge modes of gonadotropin-releasing hormone (GnRH) secretion. To clarify loci of kisspeptin action on GnRH neurons, the present study examined the morphology of the kisspeptin system and the associations between kisspeptin and GnRH systems in gonadally intact and castrated male goats. Kisspeptin-immunoreactive (ir) and Kiss1-positive neurons were found in the medial preoptic area of intact but not castrated goats. Kisspeptin-ir cell bodies and fibers in the arcuate nucleus (ARC) and median eminence (ME) were fewer in intact male goats compared with castrated animals. Apposition of kisspeptin-ir fibers on GnRH-ir cell bodies was very rare in both intact and castrated goats, whereas the intimate association of kisspeptin-ir fibers with GnRH-ir nerve terminals was observed in the ME of castrated animals. Neurokinin B immunoreactivity colocalized not only in kisspeptin-ir cell bodies in the ARC but also in kisspeptin-ir fibers in the ME, suggesting that a majority of kisspeptin-ir fibers projecting to the ME originates from the ARC. A dual immunoelectron microscopic examination revealed that nerve terminals containing kisspeptin-ir vesicles made direct contact with GnRH-ir nerve terminals at the ME of castrated goats. There was no evidence for the existence of the typical synaptic structure between kisspeptin- and GnRH-ir fibers. The present results suggest that the ARC kisspeptin neurons act on GnRH neurons at the ME to control (possibly the pulse mode of) GnRH secretion in males.


Subject(s)
Gonadotropin-Releasing Hormone/analysis , Kisspeptins/analysis , Median Eminence/ultrastructure , Neurons/chemistry , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Goats , Hypothalamus/chemistry , Immunohistochemistry , Male , Median Eminence/chemistry , Median Eminence/cytology , Microscopy, Immunoelectron , Neurokinin B/analysis , Neurons/ultrastructure , Preoptic Area/chemistry
3.
Neuroendocrinology ; 89(1): 27-37, 2009.
Article in English | MEDLINE | ID: mdl-18758154

ABSTRACT

Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) play key roles in vertebrate gametogenesis and steroidogenesis. They are mainly synthesized in the pituitary gland. While investigating the ontogeny of FSH and LH cells in the cichlid fish Cichlasoma dimerus by immunohistochemistry (IHC), we unexpectedly found immunoreactive neurons in the preoptic area, sending their projections through different brain areas and neurohypophysis. Our previous work using Western blot and IHC techniques applied to the adult brain confirmed these findings. To further demonstrate the extrapituitary expression of these hormones, we performed RT-PCR detecting sequences coding for beta-FSH and beta-LH subunits in the C. dimerus pituitary and brain (preoptic-hypothalamic area). The expression of these transcripts in both organs was consistent with their peptide expression showing a high sequence homology when compared with other phylogenetically related fish. An individual pituitary in vitro culture system was utilized to study the possible modulatory effect of brain-derived gonadotropins on pituitary hormone secretion. Pituitary explants were cultured with different concentrations of LH or FSH, and the culture media were analyzed by Western blot. Exogenous LH produced a dose-dependent increase in pituitary beta-LH, beta-FSH and somatolactin (SL) releases. No effect was observed on growth hormone (GH). The effect on prolactin (PRL) was not consistent among treatments. Exogenous FSH produced an inhibition in beta-LH release, dose-dependent increases in beta-FSH and SL releases, and no effect on PRL and GH releases. These findings support the concept of regulation of pituitary trophic hormones by brain-derived gonadotropins.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/analysis , Follicle Stimulating Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/analysis , Luteinizing Hormone, beta Subunit/genetics , Pituitary Gland/metabolism , Preoptic Area/chemistry , RNA, Messenger/analysis , Amino Acid Sequence , Animals , Cichlids , Dose-Response Relationship, Drug , Female , Fish Proteins/metabolism , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression , Glycoproteins/metabolism , Growth Hormone/metabolism , Immunohistochemistry , Luteinizing Hormone/pharmacology , Luteinizing Hormone, beta Subunit/metabolism , Male , Molecular Sequence Data , Organ Culture Techniques , Pituitary Hormones/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
4.
Article in English | MEDLINE | ID: mdl-18664387

ABSTRACT

We studied the expression of sGnRH mRNA in the neurons of the nucleus preopticus (NPO) of the Indian major carp, Cirrhinus cirrhosus, and their correlation with the reproductive status of the fish. Non-radioisotopic in situ hybridization histochemistry protocol employing biotinylated-oligonucleotide probes complementary to salmon GnRH, cichlid GnRH I, catfish GnRH, chicken GnRH II (from cichlid and catfish), and mammalian GnRH, were applied to the sections through the POA of the female Indian major carp Cirrhinus cirrhosus. Incubation with the probe complimentary to salmon GnRH (sGnRH) mRNA from salmon, produced distinct hybridization signal in the cytosol of several neurosecretory neurons of the magnocellular and parvocellular subdivisions of the NPO of the fish collected during February-April (preparatory phase) and May-June (prespawning phase). However, no signal was detected in the NPO of fish collected during July-August (spawning phase). Application of other antisense probes, or sense probe for salmon GnRH mRNA, produced no signal. We suggest that NPO neurons in C. cirrhosus may express sGnRH mRNA, produce GnRH peptide, and play a role in regulation of pituitary-ovary axis.


Subject(s)
Cyprinidae/physiology , Gonadotropin-Releasing Hormone/metabolism , Neuropeptides/metabolism , Neurosecretory Systems/physiology , Preoptic Area/metabolism , RNA, Messenger/metabolism , Reproduction/physiology , Animals , Cell Nucleus/metabolism , Cyprinidae/genetics , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/genetics , Neurons/cytology , Neurons/metabolism , Neuropeptides/chemistry , Ovary/physiology , Pituitary-Adrenal System/physiology , Preoptic Area/chemistry , RNA, Messenger/genetics , Salmon/genetics , Salmon/metabolism , Time Factors
5.
Nat Neurosci ; 3(5): 472-5, 2000 May.
Article in English | MEDLINE | ID: mdl-10769387

ABSTRACT

The two genes coding for thyroid hormone receptors (TR) alpha 1 and beta have opposite effects on female sex behaviors. Deletion of TRalpha 1 reduced them, whereas deletion of TRbeta actually increased them. These results could not be attributed to altered levels of hormones in the blood, general alterations in estrogen responsiveness or altered general activity. Instead, they indicate a previously unknown molecular mechanism upon which the two TR genes exert opposite influences.


Subject(s)
Estrogens/pharmacology , Gene Deletion , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/physiology , Sexual Behavior, Animal/drug effects , Animals , Body Weight/drug effects , Estradiol/analogs & derivatives , Estradiol/blood , Estradiol/pharmacology , Estrogens/blood , Female , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Organ Size/drug effects , Oxytocin/analysis , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/cytology , Posture , Preoptic Area/chemistry , Preoptic Area/cytology , Progesterone/pharmacology , Receptors, Estrogen/analysis , Thyroxine/blood , Triiodothyronine/blood , Uterus/drug effects , Uterus/growth & development , Vasopressins/analysis
6.
Mol Neurobiol ; 55(1): 156-163, 2018 01.
Article in English | MEDLINE | ID: mdl-28840477

ABSTRACT

The neurovascular unit (NVU) can be conceptualized as a functional entity consisting of neurons, astrocytes, pericytes, and endothelial and smooth muscle cells that operate in concert to affect blood flow to a very circumscribed area. Although we are currently in a "golden era" of bioengineering, there are, as yet, no living NVUs-on-a-chip modules available and the development of a neural chip that would mimic NVUs is a seemingly lofty goal. The sexually dimorphic nucleus of the preoptic area (SDN-POA) is a tiny brain structure (between 0.001~0.007 mm3 in rats) with an assessable biological function (i.e., male sexual behavior). The present effort was undertaken to determine whether there are identifiable NVUs in the SDN-POA by assessing its vasculature relative to its known neural components. First, a thorough and systematic review of thousands of histologic and immunofluorescent images from 201 weanling and adult rats was undertaken to define the characteristics of the vessels supplying the SDN-POA: its primary supply artery/arteriole and capillaries are physically inseparable from their neural elements. A subsequent immunofluorescent study targeting α-smooth muscle actin confirmed the identity of an artery/arteriole supplying the SDN-POA. In reality, the predominant components of the SDN-POA are calbindin D28k-positive neurons that are comingled with tyrosine hydroxylase-positive projections. Finally, a schematic of an SDN-POA NVU is proposed as a working model of the basic building block of the CNS. Such modules could serve the study of neurovascular mechanisms and potentially inform the development of next generation bioengineered neural transplants, i.e., the construct of an NVU neural chip.


Subject(s)
Nerve Net/blood supply , Nerve Net/chemistry , Neurons/chemistry , Preoptic Area/blood supply , Preoptic Area/chemistry , Sex Characteristics , Animals , Female , Male , Nerve Net/cytology , Preoptic Area/cytology , Rats , Rats, Sprague-Dawley
7.
Neuron ; 98(1): 192-207.e10, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29621487

ABSTRACT

Maternal behaviors are essential for the survival of the young. Previous studies implicated the medial preoptic area (MPOA) as an important region for maternal behaviors, but details of the maternal circuit remain incompletely understood. Here we identify estrogen receptor alpha (Esr1)-expressing cells in the MPOA as key mediators of pup approach and retrieval. Reversible inactivation of MPOAEsr1+ cells impairs those behaviors, whereas optogenetic activation induces immediate pup retrieval. In vivo recordings demonstrate preferential activation of MPOAEsr1+ cells during maternal behaviors and changes in MPOA cell responses across reproductive states. Furthermore, channelrhodopsin-assisted circuit mapping reveals a strong inhibitory projection from MPOAEsr1+ cells to ventral tegmental area (VTA) non-dopaminergic cells. Pathway-specific manipulations reveal that this projection is essential for driving pup approach and retrieval and that VTA dopaminergic cells are reliably activated during those behaviors. Altogether, this study provides new insight into the neural circuit that generates maternal behaviors.


Subject(s)
Hypothalamus/metabolism , Maternal Behavior/physiology , Mesencephalon/metabolism , Preoptic Area/metabolism , Ventral Tegmental Area/metabolism , Animals , Estrogen Receptor alpha/biosynthesis , Female , Hypothalamus/chemistry , Maternal Behavior/psychology , Mesencephalon/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/chemistry , Neural Pathways/metabolism , Organ Culture Techniques , Preoptic Area/chemistry , Ventral Tegmental Area/chemistry
8.
Endocrinology ; 148(3): 1376-83, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17122073

ABSTRACT

Hot flushes and night sweats, referred to as vasomotor symptoms (VMS), are presumed to be a result of declining hormone levels and are the principal menopausal symptoms for which women seek medical treatment. To date, estrogens and/or some progestins are the most effective therapeutics for alleviating VMS; however, these therapies may not be appropriate for all women. Therefore, nonhormonal therapies are being evaluated. The present study investigated a new reuptake inhibitor, desvenlafaxine succinate (DVS), in animal models of temperature dysfunction. Both models used are based on measuring changes in tail-skin temperature (TST) in ovariectomized (OVX) rats. The first relies on naloxone-induced withdrawal in morphine-dependent (MD) OVX rats, resulting in an acute rise in TST. The second depends on an OVX-induced loss of TST decreases during the dark phase as measured by telemetry. An initial evaluation demonstrated abatement of the rise in TST with long-term administration of ethinyl estradiol or with a single oral dose of DVS (130 mg/kg) in the MD model. Further evaluation showed that orally administered DVS acutely and dose dependently (10-100 mg/kg) abated a naloxone-induced rise in TST of MD rats and alleviated OVX-induced temperature dysfunction in the telemetry model. Oral administration of DVS to OVX rats caused significant increases in serotonin and norepinephrine levels in the preoptic area of the hypothalamus, a key region of the brain involved in temperature regulation. These preclinical studies provide evidence that DVS directly impacts thermoregulatory dysfunction in OVX rats and may have utility in alleviating VMS associated with menopause.


Subject(s)
Body Temperature Regulation/drug effects , Cyclohexanols/pharmacology , Ovariectomy , Administration, Oral , Adrenergic alpha-Antagonists/pharmacology , Animals , Cyclohexanols/administration & dosage , Desvenlafaxine Succinate , Drug Evaluation, Preclinical , Ethinyl Estradiol/pharmacology , Female , Models, Animal , Morphine Dependence/pathology , Norepinephrine/antagonists & inhibitors , Preoptic Area/chemistry , Preoptic Area/drug effects , Rats , Serotonin Antagonists/pharmacology , Telemetry
9.
Ideggyogy Sz ; 60(3-4): 94-6, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17451047

ABSTRACT

Atrial natriuretic peptide-synthesizing neurons in the hypothalamic paraventricular nucleus constitute the major sources of ANP in the three lobes of the pituitary gland. Complete transection of the pituitary stalk eliminated 93% of ANP from the intermediate lobe, 47 and 77% from the anterior and the posterior lobes, respectively. Meantime, increased levels of immunoreactive ANP were measured in the median eminence, due to the accumulation of the peptide in the transected axons centrally to the transected stalk and in the paraventricular nucleus. It is likely that ANP neurons in the paraventricular nucleus innervate the pituitary, but those in the periventricular (median) preoptic nucleus and the organum vasculosum laminae terminalis may not contribute to the ANP innervation of the pituitary gland.


Subject(s)
Atrial Natriuretic Factor/analysis , Hypothalamus/surgery , Neurons/chemistry , Paraventricular Hypothalamic Nucleus/chemistry , Animals , Diabetes Insipidus/metabolism , Diabetes Insipidus/pathology , Drinking , Male , Median Eminence/chemistry , Microdissection , Paraventricular Hypothalamic Nucleus/anatomy & histology , Preoptic Area/chemistry , Radioimmunoassay , Rats , Rats, Wistar
10.
Endocrinology ; 158(10): 3259-3268, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28531316

ABSTRACT

Vasomotor symptoms (VMS; or hot flashes) plague millions of reproductive-aged men and women who have natural or iatrogenic loss of sex steroid production. Many affected individuals are left without treatment options because of contraindications to hormone replacement therapy and the lack of equally effective nonhormonal alternatives. Moreover, development of safer, more effective therapies has been stymied by the lack of an animal model that recapitulates the hot-flash phenomenon and enables direct testing of hypotheses regarding the pathophysiology underlying hot flashes. To address these problems, we developed a murine model for hot flashes and a comprehensive method for measuring autonomic and behavioral thermoregulation in mice. We designed and constructed an instrument called a thermocline that produces a thermal gradient along which mice behaviorally adapt to a thermal challenge to their core body temperature set point while their thermal preference over time is tracked and recorded. We tested and validated this murine model for VMS by administration of a TRPV1 agonist and a neurokinin B receptor agonist, capsaicin and senktide, respectively, to unrestrained mice and observed their autonomic and behavioral responses. Following both treatments, the mice exhibited a VMS-like response characterized by a drop in core body temperature and cold-seeking behavior on the thermocline. Senktide also caused a rise in tail skin temperature and increased Fos expression in the median preoptic area, a hypothalamic temperature control center. This dynamic model may be used to fully explore the cellular and molecular bases for VMS and to develop and test new therapeutic options.


Subject(s)
Adaptation, Physiological/physiology , Hot Flashes/chemically induced , Hot Flashes/physiopathology , Peptide Fragments/pharmacology , Receptors, Neurokinin-3/agonists , Receptors, Neurokinin-3/physiology , Substance P/analogs & derivatives , Animals , Behavior, Animal/physiology , Body Temperature , Capsaicin/pharmacology , Disease Models, Animal , Female , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Preoptic Area/chemistry , Preoptic Area/physiopathology , Proto-Oncogene Proteins c-fos/analysis , Skin Temperature , Substance P/pharmacology
11.
Mol Nutr Food Res ; 50(4-5): 451-5, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16534751

ABSTRACT

The innate immune system of mammals is able to detect bacteria when they infect local tissue or enter the blood stream, and initiate an immediate immune response. Prostaglandin (PG) E2 is considered as the most important link between the peripheral immune system and the brain. Due to four PGE2 receptors (EP receptors) and their differential expression in various areas of the hypothalamus and brain stem, PGE2 mediates different components of the acute phase reaction. A fever model is discussed in which the preoptic area contains the mechanisms for both hyperthermic and hypothermic responses and EP receptors in the median preoptic area (MnPO) modulate the thermogenic system. The neuron-specific modulation of EP receptors in the MnPO can be critically tested by using Cre-recombinase-mediated DNA recombination in genetically engineered mice. A concept for mice with conditional expression of EP3R and EP4R to investigate the different roles of those receptors in lipopolysaccharide (LPS)-induced fever is presented.


Subject(s)
Dinoprostone/physiology , Fever/physiopathology , Receptors, Prostaglandin E/physiology , Acute-Phase Reaction/physiopathology , Animals , Brain Stem/chemistry , Brain Stem/physiopathology , Gene Deletion , Hypothalamus/chemistry , Hypothalamus/physiopathology , Hypothermia/physiopathology , Integrases , Mice , Mice, Knockout , Preoptic Area/chemistry , Preoptic Area/physiopathology , Receptors, Prostaglandin E/deficiency , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP3 Subtype , Receptors, Prostaglandin E, EP4 Subtype
12.
J Steroid Biochem Mol Biol ; 163: 113-20, 2016 10.
Article in English | MEDLINE | ID: mdl-27113434

ABSTRACT

Aromatase (ARO) is a cytochrome P450 enzyme that accounts for local estrogen production in the brain. The goal of this study was to develop a microsomal based assay to sensitively and reliably detect the low levels of ARO activity in different brain regions. Enzyme activity was detected based on the conversion of testosterone to estradiol. Quantity of estradiol was measured using ultra performance liquid chromatography-mass spectrometry. Detection was linear over a range of 2.5-200pg/ml estradiol, and was reproducible with intra- and inter-assay coefficients of variation (CV) <15%. Estradiol production using isolated microsomes was linear with time up to 30min as well as linearly related to amount of microsome. Substrate concentration curves revealed enzymatic kinetics (hippocampus: Vmax and Km: 0.57pmol estradiol/h per mg microsome and 48.58nM; amygdala: Vmax and Km: 1.69pmol estradiol/h per mg microsome and 48.4nM; preoptic area: Vmax and Km: 0.96pmol estradiol/h per mg microsome and 44.31nM) with testosterone used at a saturating concentration of 400nM. Anastrozole treatment blocked ARO activity in hippocampal and ovarian microsomes, indicating that the assay is specific for ARO. Also, we showed that the distribution of the long form ARO mRNA (CYP19A1) in different regions of the brain is correlated with ARO activity, with highest levels in the amygdala, followed by preoptic area and hippocampus. In the frontal cortex, very little long form ARO mRNA, and little to no ARO activity, were detected. These findings demonstrate that the microsomal incubation (MIB) assay is a sensitive and reliable method for quantifying ARO activity in discrete brain regions.


Subject(s)
Amygdala/enzymology , Aromatase/analysis , Chromatography, High Pressure Liquid/methods , Hippocampus/enzymology , Preoptic Area/enzymology , Amygdala/chemistry , Anastrozole , Animals , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , Brain Chemistry , Cytochrome P-450 CYP1A1/metabolism , Estradiol/metabolism , Female , Hippocampus/chemistry , Kinetics , Limit of Detection , Male , Microsomes/chemistry , Nitriles/pharmacology , Ovary/chemistry , Ovary/enzymology , Preoptic Area/chemistry , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Testosterone/metabolism , Triazoles/pharmacology
13.
J Neurosci ; 23(10): 4386-93, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12764128

ABSTRACT

Many neuropeptide systems subserving sex-typical behavior are dependent on sex steroids for both their organization early in life and activation during maturity. The arginine vasopressin/vasotocin (AVP/AVT) system is strongly androgen dependent in many species and critically mediates responses to sociosexual stimuli. The bluehead wrasse is a teleost fish that exhibits a female-to-male sex change in response to social cues, and neither the development nor the maintenance of male-typical behavior depends on the presence of gonads. To examine social and gonadal inputs on the AVP/AVT system in the preoptic area (POA) of the hypothalamus, we conducted three field experiments. In the first experiment, we found that AVT mRNA abundance is higher in sex-changing females that attain social dominance and display dominant male behavior than in subordinate females, regardless of whether the dominant females were intact or ovariectomized. However, AVT-immunoreactive (IR) soma size in the gigantocellular POA (gPOA), but not in the magnocellular or parvocellular POA, increased only when females were displaying both dominant male behavior and had developed testes. In the second experiment, castration of dominant terminal-phase males had no effect on AVT mRNA abundance or any behavior we measured but did increase gPOA AVT-IR soma size compared with sham-operated controls. In the third experiment, 11-ketotestosterone implants in socially subordinate, ovariectomized females had no effect on either AVT mRNA abundance or AVT-IR soma size compared with controls. These results demonstrate that the AVT neural phenotype in the bluehead wrasse can be strongly influenced by social status, and that these social influences can be manifested independent of gonads.


Subject(s)
Arginine Vasopressin/physiology , Sex Determination Processes , Social Behavior , Testosterone/analogs & derivatives , Vasotocin/physiology , Animals , Arginine Vasopressin/genetics , Arginine Vasopressin/metabolism , Dominance-Subordination , Drug Implants/pharmacology , Female , Fishes , Male , Perciformes , Phenotype , Preoptic Area/chemistry , Preoptic Area/drug effects , Preoptic Area/physiology , RNA, Messenger/metabolism , Sex Characteristics , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology , Testosterone/metabolism , Testosterone/pharmacology , Vasotocin/genetics , Vasotocin/metabolism
14.
J Neurosci ; 24(37): 8097-105, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15371511

ABSTRACT

It is generally assumed that the inhibitory neurotransmitter GABA and the stimulatory neurotransmitter glutamate are released from different neurons in adults. However, this tenet has made it difficult to explain how the same afferent signals can cause opposite changes in GABA and glutamate release. Such reciprocal release is a central mechanism in the neural control of many physiological processes including activation of gonadotropin-releasing hormone (GnRH) neurons, the neural signal for ovulation. Activation of GnRH neurons requires simultaneous suppression of GABA and stimulation of glutamate release, each of which occurs in response to a daily photoperiodic signal, but only in the presence of estradiol (E2). In rodents, E2 and photoperiodic signals converge in the anteroventral periventricular nucleus (AVPV), but it is unclear how these signals differentially regulate GABA and glutamate secretion. We now report that nearly all neurons in the AVPV of female rats express both vesicular glutamate transporter 2 (VGLUT2), a marker of hypothalamic glutamatergic neurons, as well as glutamic acid decarboxylase and vesicular GABA transporter (VGAT), markers of GABAergic neurons. These dual-phenotype neurons are the main targets of E2 in the region and are more than twice as numerous in females as in males. Moreover, dual-phenotype synaptic terminals contact GnRH neurons, and at the time of the surge, VGAT-containing vesicles decrease and VGLUT2-containing vesicles increase in these terminals. Thus, we propose a new model for ovulation that includes dual-phenotype GABA/glutamate neurons as central transducers of hormonal and neural signals to GnRH neurons.


Subject(s)
Glutamic Acid/analysis , Neurons/classification , Ovulation/physiology , Preoptic Area/cytology , Sex Characteristics , gamma-Aminobutyric Acid/analysis , Amino Acid Transport Systems/analysis , Animals , Biomarkers , Castration , Circadian Rhythm/physiology , Drug Implants , Estradiol/pharmacology , Estradiol/physiology , Estrogen Receptor Modulators/administration & dosage , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/analysis , Glutamate Decarboxylase/biosynthesis , Glutamate Decarboxylase/genetics , Glutamic Acid/metabolism , Gonadotropin-Releasing Hormone/analysis , In Situ Hybridization , Male , Membrane Transport Proteins/analysis , Models, Biological , Nerve Endings/chemistry , Nerve Endings/ultrastructure , Nerve Tissue Proteins/analysis , Neurons/chemistry , Neurons/metabolism , Neurons/physiology , Phenotype , Preoptic Area/chemistry , Preoptic Area/physiology , Rats , Rats, Sprague-Dawley , Vesicular Glutamate Transport Protein 2 , Vesicular Inhibitory Amino Acid Transport Proteins , gamma-Aminobutyric Acid/metabolism
15.
J Endocrinol ; 184(2): 435-45, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15684351

ABSTRACT

Neonatal handling induces anovulatory estrous cycles and decreases sexual receptivity in female rats. The synchronous secretion of hormones from the gonads (estradiol (E2) and progesterone (P)), pituitary (luteinizing (LH) and follicle-stimulating (FSH) hormones) and hypothalamus (LH-releasing hormone (LHRH)) are essential for the reproductive functions in female rats. The present study aimed to describe the plasma levels of E2 and P throughout the estrous cycle and LH, FSH and prolactin (PRL) in the afternoon of the proestrus, and the LHRH content in the medial preoptic area (MPOA), median eminence (ME) and medial septal area (MSA) in the proestrus, in the neonatal handled rats. Wistar pup rats were handled for 1 min during the first 10 days after delivery (neonatal handled group) or left undisturbed (nonhandled group). When they reached adulthood, blood samples were collected through a jugular cannula and the MPOA, ME and MSA were microdissected. Plasma levels of the hormones and the content of LHRH were determined by RIA. The number of oocytes counted in the morning of the estrus day in the handled rats was significantly lower than in the nonhandled ones. Neonatal handling reduces E2 levels only on the proestrus day while P levels decreased in metestrus and estrus. Handled females also showed reduced plasma levels of LH, FSH and PRL in the afternoon of the proestrus. The LHRH content in the MPOA was significantly higher than in the nonhandled group. The reduced secretion of E2, LH, FSH and LHRH on the proestrus day may explain the anovulatory estrous cycle in neonatal handled rats. The reduced secretion of PRL in the proestrus may be related to the decreased sexual receptiveness in handled females. In conclusion, early-life environmental stimulation can induce long-lasting effects on the hypothalamus-pituitary-gonad axis.


Subject(s)
Animals, Newborn/physiology , Handling, Psychological , Reproduction/physiology , Animals , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/analysis , Luteinizing Hormone/blood , Median Eminence/chemistry , Preoptic Area/chemistry , Proestrus/blood , Progesterone/blood , Prolactin/blood , Radioimmunoassay/methods , Rats , Rats, Wistar , Septum of Brain/chemistry
16.
J Endocrinol ; 186(1): 123-30, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16002542

ABSTRACT

The objectives of this study were to characterize rainbow trout (Oncorhynchus mykiss) corticotropin-releasing factor (CRF)-binding protein (CRF-BP) cDNA and to examine the variations in CRF-BP and CRF mRNA levels in response to different intensities of stress. Trout were physically disturbed by a single or three consecutive periods of chasing until exhaustion followed by 2 h of recovery. The pituitary CRF-BP and preoptic area CRF1 mRNA contents were significantly increased only after repeated chasing events. Physical disturbance increased plasma cortisol levels with the largest change occurring in the group of trout that were exposed to repeated chasing events. Trout were also individually isolated in 120 l tanks or confined to 1.5 l boxes for 4, 24 or 72 h. CRF-BP mRNA levels in confined fish were greater than those of isolated fish at 72 h although there were no differences compared with the control group. CRF1 mRNA levels in the preoptic area were greater and remained elevated for a longer period in confined compared with isolated trout. Isolation led to a transient increase in plasma cortisol levels, but the higher cortisol values developed in the confined fish suggest that this treatment was more stressful than isolation. These results demonstrate that the intensity and duration of stress are important factors regulating CRF and CRF-BP mRNA levels in rainbow trout. We hypothesize that pituitary CRF-BP is involved in regulating the activity of the stress axis, possibly by reducing access to CRF1 receptors in the corticotropes.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Oncorhynchus mykiss/metabolism , Pituitary Gland/metabolism , RNA, Messenger/metabolism , Stress, Psychological , Animals , Base Sequence , DNA, Complementary/analysis , Female , Hydrocortisone/blood , Molecular Sequence Data , Physical Exertion , Pituitary Gland/chemistry , Preoptic Area/chemistry , Preoptic Area/metabolism , RNA, Messenger/analysis , Social Isolation , Time Factors
17.
Mol Endocrinol ; 4(8): 1205-10, 1990 Aug.
Article in English | MEDLINE | ID: mdl-2293025

ABSTRACT

Neuropeptides are central to the regulation of normal sexual development and reproduction. Two new hypothalamic cDNAs have been identified by Northern blot analysis and molecular cloning. Each potentially encodes a precursor for a unique GnRH-like decapeptide. Northern blot analysis reveals tissue-specific transcripts from each gene in the hypothalamus and testis. These preoptic area regulatory factor genes, PORF-1 and PORF-2, may thus encode members of a family of GnRH-related peptides which have been described in these tissues.


Subject(s)
Cloning, Molecular , DNA/genetics , Gonadotropin-Releasing Hormone/genetics , Hypothalamus/chemistry , Preoptic Area/chemistry , Testis/chemistry , Amino Acid Sequence , Animals , Base Sequence , DNA/isolation & purification , Male , Molecular Sequence Data , Nucleic Acid Hybridization , RNA, Messenger/analysis , Rats , Rats, Inbred Strains , Regulatory Sequences, Nucleic Acid , Transcription, Genetic
18.
Mol Endocrinol ; 16(12): 2872-91, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12456806

ABSTRACT

Gamma-aminobutyric acid (GABA), acting through GABA(A) receptors (GABA(A)R), is hypothesized to suppress reproduction by inhibiting GnRH secretion, but GABA actions directly on GnRH neurons are not well established. In green fluorescent protein-identified adult mouse GnRH neurons in brain slices, gramicidin-perforated-patch-clamp experiments revealed the reversal potential (E(GABA)) for current through GABA(A)Rs was depolarized relative to the resting potential. Furthermore, rapid GABA application elicited action potentials in GnRH neurons but not controls. The consequence of GABA(A)R activation depends on intracellular chloride levels, which are maintained by homeostatic mechanisms. Membrane proteins that typically extrude chloride (KCC-2 cotransporter, CLC-2 channel) were absent from the GT1-7 immortalized GnRH cell line and GnRH neurons in situ or were not localized to the proper cell compartment for function. In contrast, GT1-7 cells and some GnRH neurons expressed the chloride-accumulating cotransporter, NKCC-1. Patch-clamp experiments showed that blockade of NKCC hyperpolarized E(GABA) by lowering intracellular chloride. Regardless of reproductive state, rapid GABA application excited GnRH neurons. In contrast, bath application of the GABA(A)R agonist muscimol transiently increased then suppressed firing; suppression persisted 4-15 min. Rapid activation of GABA(A)R thus excites GnRH neurons whereas prolonged activation reduces excitability, suggesting the physiological consequence of synaptic activation of GABA(A)R in GnRH neurons is excitation.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurons/physiology , Receptors, GABA-A/physiology , Action Potentials/drug effects , Animals , Cell Line, Transformed , Chlorides/analysis , Electric Conductivity , Female , Gene Expression , Gonadotropin-Releasing Hormone/analysis , Gonadotropin-Releasing Hormone/genetics , Gramicidin , Green Fluorescent Proteins , Homeostasis , Immunohistochemistry , In Situ Hybridization , Luminescent Proteins/genetics , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Transgenic , Muscimol/pharmacology , Neurons/chemistry , Patch-Clamp Techniques , Preoptic Area/chemistry , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/analysis , Sodium Potassium Chloride Symporter Inhibitors , Sodium-Potassium-Chloride Symporters/analysis , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2 , Symporters , gamma-Aminobutyric Acid/administration & dosage , K Cl- Cotransporters
19.
Anat Histol Embryol ; 44(4): 308-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25186451

ABSTRACT

This study describes the distribution of galanin (Gal) and galanin receptor 2 (GalR2) in the pre-optic area (POA) of the female guinea pig. Frozen sections were undergone for a routine immunofluorescence labelling. Gal and GalR2 display immunoreactivity in all parts of the pre-optic area. Gal shows reactivity both in perikarya and fibres, whereas GalR2 was observed only in perikarya. Gal- and GalR2-immunoreactive (-ir) perikarya were the most numerous in the medial pre-optic area (MPA) with the highest reactivity in its dorsal part. In the median pre-optic nucleus (MPN) and periventricular pre-optic nucleus (PPN), only single Gal- and GalR2-ir neurons were observed. The highest density of Gal-ir fibres was revealed in the PPN and the lowest in the lateral pre-optic area (LPA). The results of this study indicate that the distribution pattern of Gal containing neurons overlaps well with the distribution pattern of GalR2-positive neurons, especially in the MPA. This may suggest GalR2-dependent activity in this brain region.


Subject(s)
Galanin/analysis , Guinea Pigs/metabolism , Preoptic Area/chemistry , Receptor, Galanin, Type 2/analysis , Animals , Dendrites/chemistry , Female , Fluorescent Antibody Technique/veterinary , Frozen Sections/veterinary , Neurons/chemistry , Preoptic Area/metabolism
20.
Endocrinology ; 138(1): 230-6, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8977408

ABSTRACT

Growth retardation induced by dietary restriction in the lamb results in a low GnRH pulse frequency, and thus, puberty is delayed. In our experimental model, in which ovariectomized lambs are maintained at weaning weight (approximately 20 kg BW), hypothalamic GnRH is present and releasable, suggesting that central mechanisms limit the release of GnRH during chronic growth restriction. Our study compared the number and distribution of GnRH-containing neurons in growth-restricted (n = 5) and rapidly growing (n = 5) ovariectomized prepubertal female lambs at 24 weeks of age (normal age of puberty is about 30 weeks). Immunoreactive cells were labeled using LR-1 antiserum (R. Benoit) and an avidin-biotin-immunoperoxidase procedure. GnRH neurons were localized in 60-micron coronal sections from the level of the diagonal band of Broca to the mammillary bodies. The estimated total number of GnRH neurons in the growth-restricted and rapidly growing lambs was similar (3364.8 +/- 513.8 vs. 3151.2 +/- 279.8, respectively). In addition, the percent distributions of GnRH neurons in the diagonal band of Broca, the anterior hypothalamus, the lateral hypothalamus, and the posterior hypothalamus were not different. A trend (P = 0.07) toward a smaller percent distribution in the preoptic area was noted in growth-restricted lambs (30.6 +/- 3.6) compared to rapidly growing lambs (44.0 +/- 5.2). By contrast, the percent distribution of GnRH neurons in the medial basal hypothalamus was significantly greater in the growth-restricted lambs compared with the rapidly growing lambs (17.7 +/- 2.2 vs. 6.7 +/- 1.4, respectively; P < 0.005). It is of interest that the percent distribution of GnRH-containing neurons in the medial basal hypothalamus of the hypogonadotropic growth-restricted lamb is similar to that observed in the fetal lamb, whereas the eugonadotropic rapidly growing lamb is more similar to the adult female. In this context, decreased GnRH secretion and delayed puberty during diet-induced growth restriction may arise from alterations in the GnRH neurosecretory system.


Subject(s)
Gonadotropin-Releasing Hormone/analysis , Growth Disorders/metabolism , Hypothalamus/chemistry , Neurons/chemistry , Preoptic Area/chemistry , Animals , Female , Hypothalamus/cytology , Luteinizing Hormone/blood , Preoptic Area/cytology , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL