Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Dev Biol ; 442(1): 87-100, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29885287

ABSTRACT

The hypothalamic anteroventral periventricular nucleus (AVPV) is the major regulator of reproductive function within the hypothalamic-pituitary-gonadal (HPG) axis. Despite an understanding of the function of neuronal subtypes within the AVPV, little is known about the molecular mechanisms regulating their development. Previous work from our laboratory has demonstrated that Notch signaling is required in progenitor cell maintenance and formation of kisspeptin neurons of the arcuate nucleus (ARC) while simultaneously restraining POMC neuron number. Based on these findings, we hypothesized that the Notch signaling pathway may act similarly in the AVPV by promoting development of kisspeptin neurons at the expense of other neuronal subtypes. To address this hypothesis, we utilized a genetic mouse model with a conditional loss of Rbpj in Nkx2.1 expressing cells (Rbpj cKO). We noted an increase in cellular proliferation, as marked by Ki-67, in the hypothalamic ventricular zone (HVZ) in Rbpj cKO mice at E13.5. This corresponded to an increase in general neurogenesis and more TH-positive neurons. Additionally, an increase in OLIG2-positive early oligodendrocytic precursor cells was observed at postnatal day 0 in Rbpj cKO mice. By 5 weeks of age in Rbpj cKO mice, TH-positive cells were readily detected in the AVPV but few kisspeptin neurons were present. To elucidate the direct effects of Notch signaling on neuron and glia differentiation, an in vitro primary hypothalamic neurosphere assay was employed. We demonstrated that treatment with the chemical Notch inhibitor DAPT increased mKi67 and Olig2 mRNA expression while decreasing astroglial Gfap expression, suggesting Notch signaling regulates both proliferation and early glial fate decisions. A modest increase in expression of TH in both the cell soma and neurite extensions was observed after extended culture, suggesting that inhibition of Notch signaling alone is enough to bias progenitors towards a dopaminergic fate. Together, these data suggest that Notch signaling restricts early cellular proliferation and differentiation of neurons and oligodendrocytes both in vivo and in vitro and acts as a fate selector of kisspeptin neurons.


Subject(s)
Hypothalamus, Anterior/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Receptors, Notch/physiology , Animals , Anterior Hypothalamic Nucleus/metabolism , Arcuate Nucleus of Hypothalamus/cytology , Cell Differentiation/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Female , Hypothalamus/metabolism , Hypothalamus, Anterior/growth & development , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Kisspeptins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Notch/genetics , Signal Transduction/physiology
2.
Dev Biol ; 439(2): 102-111, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29679559

ABSTRACT

Although the hypothalamus functions as a master homeostat for many behaviors, little is known about the transcriptional networks that control its development. To investigate this question, we analyzed mice deficient for the Forkhead domain transcription factor Foxd1. Foxd1 is selectively expressed in neuroepithelial cells of the prethalamus and hypothalamus prior to the onset of neurogenesis, and is later restricted to neural progenitors of the prethalamus and anterior hypothalamus. During early stages of neurogenesis, we observed that Foxd1-deficient mice showed reduced expression of Six3 and Vax1 in anterior hypothalamus, but overall patterning of the prethalamus and hypothalamus is unaffected. After neurogenesis is complete, however, a progressive reduction and eventual loss of expression of molecular markers of the suprachiasmatic, paraventricular and periventricular hypothalamic is observed. These findings demonstrate that Foxd1 acts in hypothalamic progenitors to allow sustained expression of a subset of genes selectively expressed in mature neurons of the anterior hypothalamus.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Animals , Anterior Hypothalamic Nucleus/metabolism , Anterior Hypothalamic Nucleus/physiology , Body Patterning/genetics , Cell Differentiation/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Forkhead Transcription Factors/physiology , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypothalamus/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Stem Cells/metabolism , Stem Cells/physiology , Transcription Factors/metabolism , Homeobox Protein SIX3
3.
Cell Mol Neurobiol ; 39(8): 1139-1149, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31250245

ABSTRACT

We examined the role of the estrogen receptors alpha (ERα) and beta (ERß) in of the preoptic-anterior hypothalamic area (POA-AHA) in the regulation of ovulation in rats. The number of ERα- and ERß-immunoreactive (-ir) cells was determined at 09:00, 13:00, and 17:00 h of each stage of the estrous cycle in intact rats. Additionally, the effects of blocking ERα and ERß on ovulation rate at 09:00 h on diestrus-2 or proestrus day through the microinjection of methyl-piperidino-pyrazole (MPP) or cyclofenil in either side of POA-AHA were evaluated. The number of ERα-ir and ERß-ir cells in POA-AHA varied in each phase of estrous cycle. Either MPP or cyclofenil in the right side of POA-AHA on diestrus-2 day reduced the ovulation rate, while at proestrus day it was decreased in rats treated in either side with MPP, and in those treated with cyclofenil in the left side. MPP or cyclofenil produced a decrease in the surge of luteinizing hormone levels (LH) and an increase in progesterone and follicle stimulating hormone (FSH). Replacement with synthetic luteinizing hormone-releasing hormone in non-ovulating rats treated with MPP or cyclofenil restored ovulation. These results suggest that activation of estrogen receptors on the morning of diestrus-2 and proestrus day asymmetrically regulates ovulation and appropriately regulates the secretion of FSH and progesterone in the morning and afternoon of proestrus day. This ensures that both, the preovulatory secretion of LH and ovulation, occur at the right time.


Subject(s)
Anterior Hypothalamic Nucleus/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Ovulation , Preoptic Area/metabolism , Animals , Anterior Hypothalamic Nucleus/drug effects , Estradiol/blood , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/antagonists & inhibitors , Estrous Cycle/drug effects , Female , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/pharmacology , Neurons/drug effects , Neurons/metabolism , Ovulation/drug effects , Ovum/drug effects , Ovum/metabolism , Preoptic Area/drug effects , Progesterone/blood , Rats
4.
Proc Natl Acad Sci U S A ; 113(17): 4759-64, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27071101

ABSTRACT

In the hypothalamic arcuate nucleus (ARC), proopiomelanocortin (POMC) neurons and the POMC-derived peptide α-melanocyte-stimulating hormone (α-MSH) promote satiety. POMC neurons receive orexin-A (OX-A)-expressing inputs and express both OX-A receptor type 1 (OX-1R) and cannabinoid receptor type 1 (CB1R) on the plasma membrane. OX-A is crucial for the control of wakefulness and energy homeostasis and promotes, in OX-1R-expressing cells, the biosynthesis of the endogenous counterpart of marijuana's psychotropic and appetite-inducing component Δ(9)-tetrahydrocannabinol, i.e., the endocannabinoid 2-arachidonoylglycerol (2-AG), which acts at CB1R. We report that OX-A/OX-1R signaling at POMC neurons promotes 2-AG biosynthesis, hyperphagia, and weight gain by blunting α-MSH production via CB1R-induced and extracellular-signal-regulated kinase 1/2 activation- and STAT3 inhibition-mediated suppression of Pomc gene transcription. Because the systemic pharmacological blockade of OX-1R by SB334867 caused anorectic effects by reducing food intake and body weight, our results unravel a previously unsuspected role for OX-A in endocannabinoid-mediated promotion of appetite by combining OX-induced alertness with food seeking. Notably, increased OX-A trafficking was found in the fibers projecting to the ARC of obese mice (ob/ob and high-fat diet fed) concurrently with elevation of OX-A release in the cerebrospinal fluid and blood of mice. Furthermore, a negative correlation between OX-A and α-MSH serum levels was found in obese mice as well as in human obese subjects (body mass index > 40), in combination with elevation of alanine aminotransferase and γ-glutamyl transferase, two markers of fatty liver disease. These alterations were counteracted by antagonism of OX-1R, thus providing the basis for a therapeutic treatment of these diseases.


Subject(s)
Endocannabinoids/metabolism , Neurons/metabolism , Obesity/metabolism , Orexins/metabolism , Pro-Opiomelanocortin/metabolism , Satiety Response , alpha-MSH/metabolism , Adult , Animals , Anterior Hypothalamic Nucleus/metabolism , Anterior Hypothalamic Nucleus/pathology , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Neural Inhibition , Signal Transduction , Up-Regulation
5.
Can J Physiol Pharmacol ; 95(2): 178-184, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27901351

ABSTRACT

This study was performed to explain how the molecular processes governing the biosynthesis of gonadotropin-releasing hormone (GnRH) and GnRH receptor (GnRHR) in the hypothalamic-pituitary unit are reflected by luteinizing hormone (LH) secretion in sheep during anoestrous period and during luteal and follicular phases of the oestrous cycle. Using an enzyme-linked immunosorbent assay (ELISA), we analyzed the levels of GnRH and GnRHR in preoptic area (POA), anterior (AH) and ventromedial hypothalamus (VM), stalk-median eminence (SME), and GnRHR in the anterior pituitary gland (AP). Radioimmunoassay has also been used to define changes in plasma LH concentrations. The study provides evidence that the levels of GnRH in the whole hypothalamus of anoestrous ewes were lower than that in sheep during the follicular phase of the oestrous cycle (POA: p < 0.001, AH: p < 0.001, VM: p < 0.01, SME: p < 0.001) and not always than in luteal phase animals (POA: p < 0.05, SME: p < 0.05). It has also been demonstrated that the GnRHR amount in the hypothalamus-anterior pituitary unit, as well as LH level, in the blood in anoestrous ewes were significantly lower than those detected in animals of both cyclic groups. Our data suggest that decrease in LH secretion during the long photoperiod in sheep may be due to low translational activity of genes encoding both GnRH and GnRHR.


Subject(s)
Anestrus/metabolism , Estrous Cycle/metabolism , Gonadotropin-Releasing Hormone/biosynthesis , Hypothalamo-Hypophyseal System/metabolism , Receptors, LHRH/biosynthesis , Anestrus/blood , Animals , Anterior Hypothalamic Nucleus/metabolism , Estrous Cycle/blood , Female , Luteinizing Hormone/blood , Median Eminence/metabolism , Pituitary Gland/metabolism , Preoptic Area/metabolism , Sheep , Ventromedial Hypothalamic Nucleus/metabolism
6.
Reprod Biol Endocrinol ; 14(1): 75, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27809846

ABSTRACT

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


Subject(s)
Anterior Hypothalamic Nucleus/metabolism , Diestrus/metabolism , Estradiol/metabolism , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/metabolism , Ovulation/metabolism , Preoptic Area/metabolism , Receptors, Muscarinic/metabolism , Animals , Anterior Hypothalamic Nucleus/drug effects , Atropine/pharmacology , Contraceptive Agents/pharmacology , Diestrus/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Gonadotropin-Releasing Hormone/pharmacology , Luteinizing Hormone/drug effects , Muscarinic Antagonists/pharmacology , Ovariectomy , Ovary/drug effects , Ovulation/drug effects , Preoptic Area/drug effects , Proestrus/drug effects , Proestrus/metabolism , Rats , Receptors, Muscarinic/drug effects
7.
Eur J Neurosci ; 42(6): 2322-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26205995

ABSTRACT

While previous studies and brain atlases divide the hypothalamus into many nuclei and areas, uncharacterised regions remain. Here, we report a new region in the mouse anterior hypothalamus (AH), a triangular-shaped perifornical area of the anterior hypothalamus (PeFAH) between the paraventricular hypothalamic nucleus and fornix, that abundantly expresses chondroitin sulfate proteoglycans (CSPGs). The PeFAH strongly stained with markers for chondroitin sulfate/CSPGs such as Wisteria floribunda agglutinin and antibodies against aggrecan and chondroitin 6 sulfate. Nissl-stained sections of the PeFAH clearly distinguished it as a region of comparatively low density compared to neighboring regions, the paraventricular nucleus and central division of the anterior hypothalamic area. Immunohistochemical and DNA microarray analyses suggested that PeFAH contains sparsely distributed calretinin-positive neurons and a compact cluster of enkephalinergic neurons. Neuronal tract tracing revealed that both enkephalin- and calretinin-positive neurons project to the lateral septum (LS), while the PeFAH receives input from calbindin-positive LS neurons. These results suggest bidirectional connections between the PeFAH and LS. Considering neuronal subtype and projection, part of PeFAH that includes a cluster of enkephalinergic neurons is similar to the rat perifornical nucleus and guinea pig magnocellular dorsal nucleus. Finally, we examined c-Fos expression after several types of stimuli and found that PeFAH neuronal activity was increased by psychological but not homeostatic stressors. These findings suggest that the PeFAH is a source of enkephalin peptides in the LS and indicate that bidirectional neural connections between these regions may participate in controlling responses to psychological stressors.


Subject(s)
Anterior Hypothalamic Nucleus/cytology , Anterior Hypothalamic Nucleus/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Septal Nuclei/cytology , Septal Nuclei/metabolism , Aggression/physiology , Animals , Enkephalins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Motor Activity , Nerve Net/cytology , Nerve Net/metabolism , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-fos , Restraint, Physical , Stress, Psychological/metabolism
8.
J Reprod Dev ; 59(6): 563-8, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24047956

ABSTRACT

The aim of the present study was to determine if the estradiol-induced luteinizing hormone (LH) surge is influenced by the constant exposure to TAK-683, an investigational metastin/kisspeptin analog, that had been established to depress the pulsatile gonadotropin-releasing hormone (GnRH) and LH secretion in goats. Ovariectomized goats subcutaneously received TAK-683 (TAK-683 group, n=6) or vehicle (control group, n=6) constantly via subcutaneous implantation of an osmotic pump. Five days after the start of the treatment, estradiol was infused intravenously in both groups to evaluate the effects on the LH surge. Blood samples were collected at 6-min intervals for 4 h prior to the initiation of either the TAK-683 treatment or the estradiol infusion, to determine the profiles of pulsatile LH secretion. They were also collected at 2-h intervals from -4 h to 32 h after the start of estradiol infusion for analysis of LH surges. The frequency and mean concentrations of LH pulses in the TAK-683 group were remarkably suppressed 5 days after the start of TAK-683 treatment compared with those of the control group (P<0.05). On the other hand, a clear LH surge was observed in all animals of both groups. There were no significant differences in the LH concentrations for surge peak and the peak time of the LH surge between the TAK-683 and control groups. These findings suggest that the effects of continuous exposure to kisspeptin or its analog on the mechanism(s) that regulates the pulsatile and surge mode secretion of GnRH/LH are different in goats.


Subject(s)
Drugs, Investigational/administration & dosage , Hypothalamus/drug effects , Kisspeptins/administration & dosage , Luteinizing Hormone/metabolism , Neurons/drug effects , Receptors, G-Protein-Coupled/agonists , Secretory Pathway/drug effects , Animals , Animals, Inbred Strains , Anterior Hypothalamic Nucleus/drug effects , Anterior Hypothalamic Nucleus/metabolism , Drug Implants , Drugs, Investigational/pharmacology , Estradiol/blood , Estradiol/pharmacokinetics , Estradiol/pharmacology , Female , Goats , Hypothalamus/metabolism , Infusions, Subcutaneous , Japan , Jugular Veins , Kisspeptins/pharmacology , Luteinizing Hormone/blood , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Ovariectomy , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Receptors, G-Protein-Coupled/metabolism , Secretory Rate/drug effects
9.
Proc Natl Acad Sci U S A ; 106(39): 16692-7, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19805359

ABSTRACT

Sexually dimorphic brain nuclei underlie gender-specific neural functions and susceptibility to disease, but the developmental basis of dimorphisms is poorly understood. In these studies, we focused on the anteroventral periventricular nucleus (AVPV), a nucleus that is larger in females and critical for the female-typical cyclic surge pattern of luteinizing hormone (LH) release. Sex differences in the size and function of the AVPV result from apoptosis that occurs preferentially in the developing male. To identify upstream pathways responsible for sexual differentiation of the AVPV, we used targeted apoptosis microarrays and in vivo and in vitro follow-up studies. We found that the tumor necrosis factor alpha (TNFalpha)-TNF receptor 2 (TNFR2)-NFkappaB cell survival pathway is active in postnatal day 2 (PND2) female AVPV and repressed in male counterparts. Genes encoding key members of this pathway were expressed exclusively in GABAergic neurons. One gene in particular, TNF receptor-associated factor 2 (TRAF2)-inhibiting protein (trip), was higher in males and it inhibited both TNFalpha-dependent NFkappaB activation and bcl-2 gene expression. The male AVPV also had higher levels of bax and bad mRNA, but neither of these genes was regulated by either TNFalpha or TRIP. Finally, the trip gene was not expressed in the sexually dimorphic nucleus of the preoptic area (SDN-POA), a nucleus in which apoptosis is higher in females than males. These findings form the basis of a new model of sexual differentiation of the AVPV that may also apply to the development of other sexually dimorphic nuclei.


Subject(s)
Brain/physiology , Sex Differentiation , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Animals , Anterior Hypothalamic Nucleus/metabolism , Female , Genes, bcl-2 , Male , Models, Biological , NF-kappa B/genetics , NF-kappa B/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , gamma-Aminobutyric Acid/metabolism
10.
Proc Natl Acad Sci U S A ; 106(45): 19144-9, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19858480

ABSTRACT

After pair-bonding, male prairie voles (Microtus ochrogaster) display aggression toward novel females but not toward their female partner. Here we show that this selective aggression in pair-bonded male prairie voles is associated with increased release of vasopressin (AVP) in the anterior hypothalamus (AH). Pharmacological activation of AVP-V1a receptors (V1aR) in the AH induced selective aggression in sexually naive males, whereas V1aR blockade diminished selective aggression in pair-bonded males. Pair-bonded males also showed an increased density in V1aR binding in the AH compared to their sexually naive counterparts and overexpression of V1aR in the AH, by viral vector-mediated gene transfer, facilitated aggression toward novel females. These data demonstrate that AH-AVP is both necessary and sufficient in the regulation of selective aggression associated with pair-bonding. In the second part of this study, we examined the effects of amphetamine (AMPH) exposure on female-directed aggression and revealed the potential role of AH-AVP underlying this behavior. Repeated AMPH administration in sexually naive male prairie voles enhanced V1aR expression in the AH and induced aggression toward a familiar or unfamiliar female. In addition, this AMPH-induced aggression was blocked by intra-AH administration of a V1aR antagonist. Together, our data reveal a socioneurobiological mechanism, highlighting a critical role of AH-AVP in the regulation of aggression induced by pair-bonding or drug experience in socially monogamous male prairie voles.


Subject(s)
Aggression/physiology , Anterior Hypothalamic Nucleus/metabolism , Arvicolinae/physiology , Pair Bond , Sexual Behavior, Animal/physiology , Vasopressins/metabolism , Aggression/drug effects , Amphetamine/pharmacology , Analysis of Variance , Animals , Antidiuretic Hormone Receptor Antagonists , Arvicolinae/metabolism , Autoradiography , Enzyme-Linked Immunosorbent Assay , Female , Male
11.
Neurosci Lett ; 706: 30-35, 2019 07 27.
Article in English | MEDLINE | ID: mdl-31051224

ABSTRACT

Aggressive interactions usually reveal individual differences in the competitive ability of contest participants. Individuals with higher competitive ability often gain priority access to resources such as food, territory, and/or mates. Individuals with lower competitive ability usually have reduced access to these resources and limited mating opportunities. Despite the importance of contest performance to the reproductive success of individuals, the neuroendocrine factors associated with individual differences in competitive ability have not been fully elucidated. Here, we investigate the relationship between dopamine (DA)-related gene expression and competitive ability during mate competition in male zebra finches. Males demonstrating high competitive ability (HCA) had higher tyrosine hydroxylase mRNA levels in the ventral tegmental area and higher D1 receptor (D1-R) mRNA levels in the preoptic area than low competitive ability (LCA) males. Additionally, HCA males had lower levels of D1-R mRNA in the anterior hypothalamus relative to LCA males. These data suggest that there are dynamic and region-specific changes in DA function that relate to variation in competitive ability during mate competition.


Subject(s)
Decision Making/physiology , Finches/physiology , Mating Preference, Animal/physiology , Receptors, Dopamine D1/genetics , Tyrosine 3-Monooxygenase/genetics , Animals , Anterior Hypothalamic Nucleus/metabolism , Female , Male , Preoptic Area/metabolism , Receptors, Dopamine D1/metabolism , Social Behavior , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/metabolism
12.
Behav Brain Res ; 192(2): 185-90, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18468701

ABSTRACT

The hypothalamus plays especially important roles in various endocrine, autonomic, and behavioral responses that guarantee the survival of both the individual and the species. In the rat, a distinct hypothalamic defensive circuit has been defined as critical for integrating predatory threats, raising an important question as to whether this concept could be applied to other prey species. To start addressing this matter, in the present study, we investigated, in another prey species (the mouse), the pattern of hypothalamic Fos immunoreactivity in response to exposure to a predator (a rat, using the Rat Exposure Test). During rat exposure, mice remained concealed in the home chamber for a longer period of time and increased freezing and risk assessment activity. We were able to show that the mouse and the rat present a similar pattern of hypothalamic activation in response to a predator. Of particular note, similar to what has been described for the rat, we observed in the mouse that predator exposure induces a striking activation in the elements of the medial hypothalamic defensive system, namely, the anterior hypothalamic nucleus, the dorsomedial part of the ventromedial hypothalamic nucleus and the dorsal premammillary nucleus. Moreover, as described for the rat, predator-exposed mice also presented increased Fos levels in the autonomic and parvicellular parts of the paraventricular hypothalamic nucleus, lateral preoptic area and subfornical region of the lateral hypothalamic area. In conclusion, the present data give further support to the concept that a specific hypothalamic defensive circuit should be preserved across different prey species.


Subject(s)
Escape Reaction/physiology , Freezing Reaction, Cataleptic/physiology , Hypothalamus/metabolism , Predatory Behavior/physiology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Anterior Hypothalamic Nucleus/metabolism , Anterior Hypothalamic Nucleus/physiology , Behavior, Animal/physiology , Dorsomedial Hypothalamic Nucleus/metabolism , Dorsomedial Hypothalamic Nucleus/physiology , Fear/physiology , Hypothalamic Area, Lateral/metabolism , Hypothalamic Area, Lateral/physiology , Hypothalamus/physiology , Immunohistochemistry , Male , Mice , Neural Pathways/metabolism , Neural Pathways/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiology , Preoptic Area/metabolism , Preoptic Area/physiology , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Long-Evans , Species Specificity , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/physiology
13.
J Neuroendocrinol ; 30(3): e12573, 2018 03.
Article in English | MEDLINE | ID: mdl-29377458

ABSTRACT

Thyroid hormone (TH) is crucial for brain development and function. This becomes most evident in untreated congenital hypothyroidism, leading to irreversible mental retardation. Likewise, maternal hypothyroxinaemia, a lack of TH during pregnancy, is associated with neurological dysfunction in the offspring, such as autism and reduced intellectual capacity. In the brain, TH acts mainly through TH receptor α1 (TRα1). Consequently, mice heterozygous for a dominant-negative mutation in TRα1 display profound neuroanatomical abnormalities including deranged development of parvalbumin neurones. However, the exact timing and orchestration of TH signalling during parvalbumin neurone development remains elusive. In the present study, we dissect the development of parvalbumin neurones in the anterior hypothalamic area (AHA) in male mice using different mouse models with impaired pre- and postnatal TH signalling in combination with bromodeoxyuridine birth dating and immunohistochemistry. Our data reveal that hypothalamic parvalbumin neurones are born at embryonic day 12 and are first detected in the AHA at postnatal day 8, reaching their full population number at P13. Interestingly, they do not require TH postnatally because their development is not impaired in mice with impaired TH signalling after birth. By contrast, however, these neurones crucially depend on TH through TRα1 signalling in the second half of pregnancy, when the hormone is almost exclusively provided by the mother. For the first time, our findings directly link a maternal hormone to a neuroanatomical substrate in the foetal brain, and underline the importance of proper TH signalling during pregnancy for offspring mental health. Given the role of hypothalamic parvalbumin neurones in the central control of blood pressure, the present study advocates the inclusion of cardiovascular parameters in the current discussion on possible TH substitution in maternal hypothyroxinaemia.


Subject(s)
Anterior Hypothalamic Nucleus/metabolism , Neurogenesis/physiology , Neurons/metabolism , Parvalbumins/metabolism , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormones/metabolism , Animals , Anterior Hypothalamic Nucleus/cytology , Female , Male , Mice , Neurons/cytology , Pregnancy , Signal Transduction/physiology
14.
J Neurosci ; 26(46): 11961-73, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108170

ABSTRACT

The brain controls fertility through release of gonadotropin-releasing hormone (GnRH), but the mechanisms underlying action potential patterning and GnRH release are not understood. We investigated whether GnRH neurons exhibit afterdepolarizing potentials (ADPs) and whether these are modified by reproductive state. Whole-cell current-clamp recordings of GnRH neurons in brain slices from ovariectomized mice revealed a slow ADP (sADP) after action potentials generated by brief current injection. Generating two or four spikes enhanced sADP amplitude and duration. sADP amplitude was not affected by blocking selected neurotransmitter/neuromodulator receptors, delayed-rectifier potassium channels, calcium-dependent cation channels, or hyperpolarization-activated cation channels but was halved by the calcium channel blocker cadmium and abolished by tetrodotoxin. Cadmium also reduced peak latency. Intrinsic mechanisms underlying the sADP were investigated using voltage-clamp protocols simulating action potential waveforms. A single action potential produced an inward current, which increased after double and quadruple stimulation. Cadmium did not affect current amplitude but reduced peak latency. Pretreatment with blockers of calcium-activated potassium currents (I(KCa)) reproduced this shift and blocked subsequent cadmium-induced changes, suggesting cadmium changes latency indirectly by blocking I(KCa). Tetrodotoxin abolished the inward current, suggesting that it is carried by sodium. In contrast, I(KCa) blockers increased the inward current, indicating that I(KCa) may oppose generation of the sADP. Strong sADPs were suprathreshold, generating repetitive spontaneous firing. I(ADP), sADP, and excitability were enhanced by in vivo estradiol, which triggers a preovulatory surge of GnRH release. Physiological feedback modification of this inward current and resulting sADP may modulate action potential firing and subsequent GnRH release.


Subject(s)
Anterior Hypothalamic Nucleus/metabolism , Fertility/physiology , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Preoptic Area/metabolism , Sodium Channels/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anterior Hypothalamic Nucleus/cytology , Anterior Hypothalamic Nucleus/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Estradiol/metabolism , Estrous Cycle/drug effects , Estrous Cycle/physiology , Feedback, Physiological/physiology , Female , Fertility/drug effects , Mice , Neurons/cytology , Neurons/drug effects , Organ Culture Techniques , Ovariectomy , Patch-Clamp Techniques , Preoptic Area/cytology , Preoptic Area/drug effects , Reaction Time/drug effects , Reaction Time/physiology , Receptors, Neurotransmitter/antagonists & inhibitors , Receptors, Neurotransmitter/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Tetrodotoxin/pharmacology
15.
J Comp Neurol ; 502(6): 1109-22, 2007 Jun 20.
Article in English | MEDLINE | ID: mdl-17444499

ABSTRACT

Male prairie voles (Microtus ochrogaster) display mating-induced pair bonding indicated by social affiliation with their female partners and aggression toward unfamiliar conspecifics. In the present study, we characterized their aggression associated with pair bonding and examined the related neuronal activation and neurochemical architecture. Males that were pair-bonded for 2 weeks displayed intense levels of aggression toward a female or male conspecific stranger but maintained a high level of social affiliation with their familiar female partners. These social interactions induced increases in neural activation, indicated by increased density of Fos-immunoreactive staining (Fos-ir) in several brain regions including the bed nucleus of the stria terminalis (BNST), medial preoptic area (MPOA), paraventricular nucleus (PVN), anterior cortical (AcA), and medial nuclei (MeA) of the amygdala. In the anterior hypothalamus (AH), increased density of Fos-ir staining was found specifically to be associated with aggression toward unfamiliar female or male strangers. In addition, higher densities of AH cells that were stained for tyrosine hydroxylase (TH) or vasopressin (AVP) were also labeled with Fos-ir in these males displaying aggression toward a conspecific stranger compared with males displaying social affiliation toward their female partner. Together, our results indicate that dopamine and vasopressin in the AH may be involved in the regulation of enduring aggression associated with pair bonding in male prairie voles.


Subject(s)
Aggression/physiology , Anterior Hypothalamic Nucleus/metabolism , Arvicolinae/physiology , Behavior, Animal/physiology , Limbic System/metabolism , Pair Bond , Animals , Anterior Hypothalamic Nucleus/anatomy & histology , Arvicolinae/anatomy & histology , Brain/anatomy & histology , Brain/metabolism , Catecholamines/metabolism , Female , Immunohistochemistry , Limbic System/anatomy & histology , Male , Neural Pathways/anatomy & histology , Neural Pathways/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Sexual Behavior, Animal/physiology , Tyrosine 3-Monooxygenase/metabolism , Vasopressins/metabolism
16.
J Comp Neurol ; 494(1): 190-214, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16304687

ABSTRACT

The rat ovulatory cycle is dependent on the preoptic region encompassing the gonadotrophin-releasing hormone (GnRH) perikarya and the anteroventral periventricular nucleus (AVPV). Retrograde tract tracing was used to identify and compare the sources of inputs to these sites in female rats. Within the telencephalon and diencephalon, the incidence of retrograde labelling from both sites was moderate to abundant in the ventral lateral septum, posteromedial bed nucleus of the stria terminalis, amygdalohippocampal area and the periventricular, medial preoptic, anterodorsal preoptic, dorsomedial suprachiasmatic, arcuate, and posterior ventrolateral ventromedial hypothalamic nuclei. In these regions, the incidence of retrograde labelling was either greater from the AVPV than from the GnRH perikarya site or similar from both sites. In the medial amygdaloid, parastrial, striohypothalamic, and ventral premammillary nuclei, the retrograde labelling from the AVPV greatly exceeded the sparse incidence from the GnRH perikarya site. In contrast, retrograde labelling from the GnRH perikarya site predominated in the median preoptic, lateroanterior and dorsomedial hypothalamic nuclei, subparaventricular zone, and retrochiasmatic area; it was abundant in the AVPV. Caudal to the diencephalon, retrograde labelling from either site was sparse, except in the lateral parabrachial nucleus, which displayed a particularly high incidence from the GnRH perikarya site. Other mesencephalic regions labelled from either site included the periaqueductal gray and dorsal and median raphe nuclei. The most caudal labelling was found in the ventrolateral medulla and region of the solitary tract nucleus; this was almost exclusively from the GnRH perikarya site. These findings further elucidate the neuroanatomical connections underlying the control of the ovulatory cycle.


Subject(s)
Anterior Hypothalamic Nucleus/cytology , Gonadotropin-Releasing Hormone/metabolism , Neural Pathways/cytology , Neurons/metabolism , Animals , Anterior Hypothalamic Nucleus/metabolism , Brain Mapping , Cholera Toxin/metabolism , Diencephalon/cytology , Diencephalon/metabolism , Female , Immunohistochemistry/methods , Neural Networks, Computer , Neural Pathways/metabolism , Neurons/cytology , Ovariectomy/methods , Rats , Rats, Wistar , Telencephalon/cytology , Telencephalon/metabolism
17.
J Hypertens ; 24(1): 109-16, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16331108

ABSTRACT

BACKGROUND: Centrally applied angiotensin II (Ang II) increases sympathetic nervous activity and mean arterial blood pressure (MAP), but the mediation of these effects is not fully understood. OBJECTIVE: To test the hypothesis that central effects of Ang II are mediated by reduced nicotinamide adenine dinucleotide phosphate [NAD(P)H]-oxidase-dependent production of superoxide in the hypothalamus. METHODS: Under isoflurane anesthesia, male Sprague-Dawley rats were given an intracerebroventricular infusion of either artificial cerebrospinal fluid or apocynin (4 microg/kg per min), a selective inhibitor for NAD(P)H oxidase, for 30 min, followed by Ang II (20 ng) or carbachol (200 ng), while MAP and heart rate were measured at the femoral artery. At the end of the experiments, hydroethidine, a superoxide-sensitive fluorescent dye, was infused intravenously for 10 min, and superoxide production was assessed in the vasoregulatory hypothalamic nuclei using confocal microscopy. RESULTS: Ang II elicited a rapid 11 +/- 2-mmHg increase in MAP and a 16 +/- 2-beats/min decrease in heart rate. Apocynin abolished these effects of Ang II in a specific manner, as carbachol-induced increases in MAP were unaffected by the inhibition of NAD(P)H oxidase (MAP increased by 9 +/- 2 and 8 +/- 1 mmHg in the absence and presence of apocynin, respectively). In response to Ang II, apocynin-sensitive production of superoxide increased significantly in the nuclei of the anterior hypothalamus, in the subfornical organ, and in the paraventricular nucleus of the hypothalamus. CONCLUSION: These findings demonstrate that acute pressor responses of central Ang II are mediated by NAD(P)H-oxidase-dependent production of superoxide in the hypothalamus.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure/drug effects , Hypothalamus/metabolism , NADPH Oxidases/metabolism , Superoxides/metabolism , Acetophenones/pharmacology , Angiotensin II/administration & dosage , Animals , Anterior Hypothalamic Nucleus/drug effects , Anterior Hypothalamic Nucleus/metabolism , Blood Pressure/physiology , Carbachol/pharmacology , Cardiovascular Physiological Phenomena/drug effects , Heart Rate/drug effects , Heart Rate/physiology , Hypothalamus/drug effects , Male , Microscopy, Confocal , Microscopy, Fluorescence , NADPH Oxidases/antagonists & inhibitors , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Sprague-Dawley , Subfornical Organ/drug effects , Subfornical Organ/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology
18.
Brain Res ; 1073-1074: 425-30, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16445890

ABSTRACT

The present study investigated the hypothesis that social isolation increases aggression by increasing the number of V1a vasopressin receptors in the anterior hypothalamus (AH). Male hamsters were randomly assigned to a group that was allowed to interact with a small nonaggressive hamster three times each week for 3 weeks (socially experienced) or to a group that did not interact socially with other hamsters (social isolates). On the final day of the experiment, hamsters in both groups were placed in a neutral arena with a small, nonaggressive intruder, and agonistic behavior was scored for 10 min. In social isolates, the duration of aggression and the number of attacks were significantly greater than in socially experienced hamsters. There were no significant between-group differences in the latency to the onset of aggression, the number of flank marks or in the duration of defensive/submissive, social or nonsocial behavior. The amount of V1a receptor binding was significantly greater in the AH, the paraventricular nucleus of the hypothalamus and the lateral hypothalamus in the social isolates than in the socially experienced hamsters. The amount of V1a receptor binding was significantly greater in the central amygdala of socially experienced hamsters than in socially isolated hamsters. Serum concentrations of testosterone were significantly higher in the socially experienced hamsters than in social isolates. These data support the hypothesis that social isolation increases aggression by increasing the number of V1a vasopressin receptors in the AH.


Subject(s)
Aggression/physiology , Agonistic Behavior/physiology , Anterior Hypothalamic Nucleus/metabolism , Receptors, Vasopressin/physiology , Analysis of Variance , Animals , Behavior, Animal , Cricetinae , Hypothalamic Area, Lateral/metabolism , Male , Mesocricetus , Protein Binding/physiology , Radioimmunoassay/methods , Radioligand Assay/methods , Random Allocation , Social Isolation , Testosterone/blood
19.
Eksp Klin Farmakol ; 69(3): 14-8, 2006.
Article in Russian | MEDLINE | ID: mdl-16878491

ABSTRACT

Amphetamine (1 mg/kg), morphine (1 mg/kg), and ethaminal sodium (5 mg/kg) activated self-stimulation reaction of lateral hypothalamus in rats. In contrast, intraamygdalary injections of astressin (1 microg/microl), which is a nonselective antagonist of corticoliberin receptors, inhibited this reaction. The astressin blockade of extrahypothalamic corticoliberin receptors in the central nucleus of amygdala modified the effects of various narcogens on self-stimulation reaction. On this background, amphetamine did not activate self-stimulation, ethaminal sodium retained significant psychoactivating effect, whereas the effect of morphine switched from stimulant to depressant. Leu-enkephalin exhibited a stable depressant effect, thus potentiating the action of astressin. The astressin-induced enhancement of the inhibiting action of leu-enkephalin on cerebral self-stimulation is probably related to a temporary switch-off of the activating influence of the central nucleus of amygdala on hypothalamus.


Subject(s)
Amphetamine/pharmacology , Analgesics, Opioid/pharmacology , Central Nervous System Stimulants/pharmacology , Corticotropin-Releasing Hormone/pharmacology , Morphine/pharmacology , Neuroprotective Agents/pharmacology , Neurotransmitter Agents/pharmacology , Peptide Fragments/pharmacology , Self Stimulation/drug effects , Action Potentials/drug effects , Animals , Anterior Hypothalamic Nucleus/metabolism , Enkephalin, Leucine , Hypothalamic Area, Lateral/metabolism , Male , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism
20.
Brain Struct Funct ; 221(7): 3521-46, 2016 09.
Article in English | MEDLINE | ID: mdl-26462664

ABSTRACT

Neuropeptide S (NPS) is a regulatory peptide expressed by limited number of neurons in the brainstem. The simultaneous anxiolytic and arousal-promoting effect of NPS suggests an involvement in mood control and vigilance, making the NPS-NPS receptor system an interesting potential drug target. Here we examined, in detail, the distribution of NPS-immunoreactive (IR) fiber arborizations in brain regions of rat known to be involved in the regulation of sleep and arousal. Such nerve terminals were frequently apposed to GABAergic/galaninergic neurons in the ventro-lateral preoptic area (VLPO) and to tyrosine hydroxylase-IR neurons in all hypothalamic/thalamic dopamine cell groups. Then we applied the single platform-on-water (mainly REM) sleep deprivation method to study the functional role of NPS in the regulation of arousal. Of the three pontine NPS cell clusters, the NPS transcript levels were increased only in the peri-coerulear group in sleep-deprived animals, but not in stress controls. The density of NPS-IR fibers was significantly decreased in the median preoptic nucleus-VLPO region after the sleep deprivation, while radioimmunoassay and mass spectrometry measurements showed a parallel increase of NPS in the anterior hypothalamus. The expression of the NPS receptor was, however, not altered in the VLPO-region. The present results suggest a selective activation of one of the three NPS-expressing neuron clusters as well as release of NPS in distinct forebrain regions after sleep deprivation. Taken together, our results emphasize a role of the peri-coerulear cluster in the modulation of arousal, and the importance of preoptic area for the action of NPS on arousal and sleep.


Subject(s)
Arousal , Brain/cytology , Brain/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Animals , Anterior Hypothalamic Nucleus/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Galanin/metabolism , Glutamic Acid/metabolism , Locus Coeruleus/metabolism , Male , Preoptic Area/cytology , Preoptic Area/metabolism , RNA, Messenger , Rats , Rats, Wistar , Receptors, Neuropeptide/metabolism , Sleep , Sleep Deprivation/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL