Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Eur J Neurosci ; 46(10): 2596-2607, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28973792

ABSTRACT

During mammalian embryonic development, GnRH neurones differentiate from the nasal placode and migrate through the nasal septum towards the forebrain. We previously showed that a category of glial cells, the olfactory ensheathing cells (OEC), forms the microenvironment of migrating GnRH neurones. Here, to characterize the quantitative and qualitative importance of this glial, we investigated the spatiotemporal maturation of glial cells in situ and the role of maturing glia in GnRH neurones development ex vivo. More than 90% of migrating GnRH neurones were found to be associated with glial cells. There was no change in the cellular microenvironment of GnRH neurones in the regions crossed during embryonic development as glial cells formed the main microenvironment of these neurones (53.4%). However, the phenotype of OEC associated with GnRH neurones changed across regions. The OEC progenitors immunoreactive to brain lipid binding protein formed the microenvironment of migrating GnRH neurones from the vomeronasal organ to the telencephalon and were also present in the diencephalon. However, during GnRH neurone migration, maturation of OEC to [GFAP+] state (glial fibrillary acid protein) was only observed in the nasal septum. Inducing depletion of OEC in maturation, using transgenic mice expressing herpes simplex virus thymidine kinase driven by the GFAP promoter, had no impact on neurogenesis or on triggering GnRH neurones migration in nasal explant culture. Nevertheless, depletion of [GFAP+] cells decreased GnRH neurites outgrowth by 57.4%. This study suggests that specific maturation of OEC in the nasal septum plays a role in morphological differentiation of GnRH neurones.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurites/physiology , Neuroglia/physiology , Neuronal Outgrowth , Neurons/physiology , Olfactory Bulb/growth & development , Animals , Cell Movement , Mice , Mice, Transgenic , Nasal Septum/growth & development , Neural Stem Cells/physiology , Neuroglia/metabolism , Neurons/metabolism , Olfactory Bulb/metabolism , Organ Culture Techniques , Stem Cells , Vomeronasal Organ/growth & development
2.
J Med Chem ; 58(8): 3459-70, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25811530

ABSTRACT

New potent and selective KISS1R agonists were designed using a combination of rational chemical modifications of the endogenous neuropeptide kisspeptin 10 (KP10). Improved resistance to degradation and presumably reduced renal clearance were obtained by introducing a 1,4-disubstituted 1,2,3-triazole as a proteolysis-resistant amide mimic and a serum albumin-binding motif, respectively. These triazololipopeptides are highly potent full agonists of KISS1R and are >100 selective over the closely related NPFF1R. When injected in ewes with a quiescent reproductive system, the best compound of our series induced a much prolonged increase of luteinizing hormone release compared to KP10 and increased follicle-stimulating hormone plasma concentration. Hence, this KISS1R agonist is a new valuable pharmacological tool to explore the potential of KP system in reproduction control. Furthermore, it represents the first step to develop drugs treating reproductive system disorders due to a reduced activity of the hypothalamo-pituitary-gonadal axis such as delayed puberty, hypothalamic amenorrhea, and hypogonadotropic hypogonadism.


Subject(s)
Follicle Stimulating Hormone/metabolism , Kisspeptins/chemistry , Kisspeptins/pharmacology , Luteinizing Hormone/metabolism , Triazoles/chemistry , Triazoles/pharmacology , Acetylation , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cricetulus , Female , Follicle Stimulating Hormone/blood , HEK293 Cells , Humans , Kisspeptins/blood , Kisspeptins/metabolism , Luteinizing Hormone/blood , Peptide Hydrolases/metabolism , Protein Binding , Serum Albumin/metabolism , Sheep , Triazoles/blood , Triazoles/metabolism
3.
Biol Reprod ; 92(2): 46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505201

ABSTRACT

Increased body weight (BW) gain during the juvenile period leads to early maturation of the reproductive neuroendocrine system. We investigated whether a nutritional regimen that advances the onset of puberty leads to alterations in the hypothalamic neuropeptide Y (NPY) circuitry that are permissive for enhanced gonadotropin-releasing hormone (GnRH) secretion. It was hypothesized that NPY mRNA and NPY projections to GnRH and kisspeptin neurons are reduced in heifers that gain BW at an accelerated rate, compared with a lower one, during the juvenile period. Heifers were weaned at approximately 4 mo of age and fed diets to promote relatively low (0.5 kg/day; low gain [LG]) or high (1.0 kg/day; high gain [HG]) rates of BW gain until 8.5 mo of age. Heifers that gained BW at a higher rate exhibited greater circulating concentrations of leptin and reduced overall NPY expression in the arcuate nucleus. The proportion of GnRH neurons in close apposition to NPY fibers and the magnitude of NPY projections to GnRH neurons located in the mediobasal hypothalamus were reduced in HG heifers. However, no differences in NPY projections to kisspeptin neurons in the arcuate nucleus were detected between HG and LG heifers. Results indicate that a reduction in NPY innervation of GnRH neurons, particularly at the level of the mediobasal hypothalamus, occurs in response to elevated BW gain during the juvenile period. This functional plasticity may facilitate early onset of puberty in heifers.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurons/physiology , Neuropeptide Y/metabolism , Weight Gain/physiology , Animals , Cattle , Female , Kisspeptins/metabolism , Leptin/blood , Sexual Maturation/physiology
4.
Biol Reprod ; 90(2): 36, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24429215

ABSTRACT

Kisspeptin has emerged as the most potent gonadotropin-releasing hormone (GnRH) secretagogue and appears to represent the penultimate step in the central control of reproduction. In the sheep, we showed that kisspeptin could be used to manipulate gonadotropin secretion and control ovulation. Prompted by these results, we decided to investigate whether kisspeptin could be used as an ovulation-inducing agent in another photoperiodic domestic mammal, the horse. Equine kisspeptin-10 (eKp10) was administered intravenously as bolus injections or short- to long-term perfusions to Welsh pony mares, either during the anestrus season or at various stages of the cycle during the breeding season. In all the experimental conditions, eKp10 reliably increased peripheral concentrations of both luteinizing hormone and follicle-stimulating hormone. The nature of the response to eKp10 was consistent across experimental conditions and physiological states: the increase in gonadotropins was always rapid and essentially transient even when eKp10 was perfused for prolonged periods. Furthermore, eKp10 consistently failed to induce ovulation in the mare. To gain insights into the underlying mechanisms, we used acute injections or perfusions of GnRH. We also cloned the equine orthologues of the kisspeptin precursor and Kiss1r; this was justified by the facts that the current equine genome assembly predicted an amino acid difference between eKp10 and Kp10 in other species while an equine orthologue for Kiss1r was missing altogether. In light of these findings, potential reasons for the divergence in the response to kisspeptin between ewe and mare are discussed. Our data highlight that kisspeptin is not a universal ovulation-inducing agent.


Subject(s)
Gonadotropins/metabolism , Horses , Kisspeptins/administration & dosage , Ovulation Induction/veterinary , Ovulation/drug effects , Animals , Cloning, Molecular , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Horses/physiology , Kisspeptins/genetics , Kisspeptins/metabolism , Ovulation Induction/methods , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/isolation & purification , Treatment Failure
5.
Biol Reprod ; 90(2): 28, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24389874

ABSTRACT

RFamide-related peptide 3 (RFRP3), the mammalian homologue of avian gonadotropin-inhibitory hormone, has been shown to negatively regulate the secretion of LH and may contribute to reproductive seasonality in some species. Herein, we examined the presence and potential role of the RFRP3-signaling system in regulating LH secretion in the mare during the breeding and nonbreeding seasons. Hypothalamic NPVF mRNA (the precursor mRNA for RFRP3) was detected at the level of the dorsomedial nucleus and paraventricular nucleus, but expression did not change with season. A greater number of RFRP3-expressing cells was observed throughout the rostral-caudal extension of the dorsomedial nucleus. Furthermore, adenohypophyseal expression of the RFRP3 receptor (NPFFR1) during the winter anovulatory season did not differ from that during either the follicular or luteal phases of the estrous cycle. When tested in primary adenohypophyseal cell culture or in vivo during both the breeding and nonbreeding seasons, neither equine nor ovine peptide sequences for RFRP3 suppressed basal or GnRH-mediated release of LH. However, infusion of RF9, an RFRP3 receptor-signaling antagonist, into seasonally anovulatory mares induced a robust increase in secretion of LH both before and following continuous treatment with GnRH. The results indicate that the cellular machinery associated with RFRP3 function is present in the equine hypothalamus and adenohypophysis. However, evidence for functionality of the RFRP3-signaling network was only obvious when an antagonist RF9 was employed. Because GnRH-induced release of LH was not affected by RF9, its actions may occur upstream from the gonadotrope to stimulate or disinhibit secretion of GnRH.


Subject(s)
Horses , Hypothalamus/metabolism , Neuropeptides/metabolism , Pituitary Gland, Anterior/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Reproduction/physiology , Animals , Breeding , Cells, Cultured , Female , Horses/genetics , Horses/metabolism , Hypothalamus/drug effects , Neuropeptides/genetics , Neuropeptides/pharmacology , Pituitary Gland, Anterior/drug effects , Seasons , Tissue Distribution
6.
Mol Cell Endocrinol ; 382(1): 387-399, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-24145132

ABSTRACT

The hypothalamus integrates endogenous and exogenous inputs to control the pituitary-gonadal axis. The ultimate hypothalamic influence on reproductive activity is mediated through timely secretion of GnRH in the portal blood, which modulates the release of gonadotropins from the pituitary. In this context neurons expressing the RF-amide neuropeptide kisspeptin present required features to fulfill the role of the long sought-after hypothalamic integrative centre governing the stimulation of GnRH neurons. Here we focus on the intracellular signaling pathways triggered by kisspeptin through its cognate receptor KISS1R and on the potential role of proteins interacting with this receptor. We then review evidence implicating both kisspeptin and RFRP3--another RF-amide neuropeptide--in the temporal orchestration of both the pre-ovulatory LH surge in female rodents and the organization of seasonal breeding in photoperiodic species.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Neurosecretory Systems/metabolism , Animals , Humans , Neurons/metabolism , Signal Transduction , Time Factors
7.
Neuroendocrinology ; 98(4): 281-9, 2013.
Article in English | MEDLINE | ID: mdl-24080803

ABSTRACT

Kisspeptin (KP) neurones in the rostral periventricular area of the third ventricle (RP3V) and arcuate nucleus (Arc) are important elements in the neuronal circuitry regulating gonadotropin-releasing hormone (GnRH) secretion. KP and co-synthesised neuropeptides/neurotransmitters act directly on GnRH perikarya and processes. GnRH neurones not only form the final output pathway regulating the reproductive functions of the anterior pituitary gland, but also provide neuronal input to sites within the hypothalamus. The current double-label immunohistochemical studies investigated whether GnRH-immunoreactive (IR) projections to the RP3V and/or Arc establish morphological connections with KP-IR neurones at these sites. To optimise visualisation of KP immunoreactivity in, respectively, the RP3V and Arc, ovariectomised (OVX) oestrogen-treated and OVX oil-treated female mice were studied. Confocal laser microscopic analysis of immunofluorescent specimens revealed GnRH-IR axon varicosities in apposition to approximately 25% of the KP-IR neurones in the RP3V and 50% of the KP-IR neurones in the Arc. At the ultrastructural level, GnRH-IR neurones were seen to establish asymmetric synaptic contacts, which usually reflect excitatory neurotransmission, with KP-IR neurones in both the RP3V and Arc. Together with previous data, these findings indicate reciprocal connectivity between both of the KP cell populations and the GnRH neuronal system. The functional significance of the GnRH-IR input to the two separate KP cell populations requires electrophysiological investigation.


Subject(s)
Brain/cytology , Brain/metabolism , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Dendrites/metabolism , Estrogens/metabolism , Female , Fluorescent Antibody Technique , Imaging, Three-Dimensional , Immunohistochemistry , Mice , Mice, Inbred Strains , Microscopy, Confocal , Microscopy, Electron , Neural Pathways/cytology , Neural Pathways/metabolism , Ovariectomy , Synapses/metabolism , Third Ventricle
8.
Endocrinology ; 154(8): 2784-94, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23736293

ABSTRACT

Kisspeptin (Kiss1) signaling to GnRH neurons is widely acknowledged to be a prerequisite for puberty and reproduction. Animals lacking functional genes for either kisspeptin or its receptor exhibit low gonadotropin secretion and infertility. Paradoxically, a recent study reported that genetic ablation of nearly all Kiss1-expressing neurons (Kiss1 neurons) does not impair reproduction, arguing that neither Kiss1 neurons nor their products are essential for sexual maturation. We posited that only minute quantities of kisspeptin are sufficient to support reproduction. If this were the case, animals having dramatically reduced Kiss1 expression might retain fertility, testifying to the redundancy of Kiss1 neurons and their products. To test this hypothesis and to determine whether males and females differ in the required amount of kisspeptin needed for reproduction, we used a mouse (Kiss1-CreGFP) that has a severe reduction in Kiss1 expression. Mice that are heterozygous and homozygous for this allele (Kiss1(Cre/+) and Kiss1(Cre/Cre)) have ∼50% and 95% reductions in Kiss1 transcript, respectively. We found that although male Kiss1(Cre/Cre) mice sire normal-sized litters, female Kiss1(Cre/Cre) mice exhibit significantly impaired fertility and ovulation. These observations suggest that males require only 5% of normal Kiss1 expression to be reproductively competent, whereas females require higher levels for reproductive success.


Subject(s)
Kisspeptins/metabolism , Neurons/metabolism , Reproduction/physiology , Signal Transduction/physiology , Animals , Dynorphins/genetics , Female , Fertility/genetics , Fertility/physiology , Gene Expression , Genotype , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Kisspeptins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Precursors/genetics , Receptors, Neurokinin-3/genetics , Reproduction/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sex Characteristics , Sex Factors , Sexual Maturation/genetics , Sexual Maturation/physiology , Signal Transduction/genetics , Tachykinins/genetics
9.
Adv Exp Med Biol ; 784: 411-30, 2013.
Article in English | MEDLINE | ID: mdl-23550017

ABSTRACT

Wild and domesticated species display seasonality in reproductive function, controlled predominantly by photoperiod. Seasonal alterations in breeding status are caused by changes in the secretion of gonadotropin-releasing hormone (GnRH) that are mediated by upstream neuronal afferents that regulate the GnRH cells. In particular, kisspeptin appears to play a major role in seasonality of reproduction, transducing the feedback effect of gonadal steroids as well as having an independent (nonsteroid dependent) circannual rhythm. A substantial body of data on this issue has been obtained from studies in sheep and hamsters and this is reviewed here in detail. Kisspeptin function is upregulated during the breeding season in sheep, stimulating reproductive function, but contradictory data are found in Siberian and Syrian hamsters. The relative quiescence of kisspeptin cells in the nonbreeding season can be counteracted by administration of the peptide, leading to activation of reproductive function. Although there is a major role for melatonin in the transduction of photoperiod to the reproductive system, kisspeptin cells do not appear to express the melatonin receptor, so the means by which seasonality changes the level of kisspeptin activity remains unknown.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Receptors, Melatonin/metabolism , Reproduction/physiology , Seasons , Animals , Cricetinae , Mesocricetus , Sheep
10.
J Comp Neurol ; 521(13): 3003-29, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23504961

ABSTRACT

In naked mole-rat (NMR) colonies, breeding is monopolized by the queen and her consorts. Subordinates experience gonadal development if separated from the queen. To elucidate the neuroendocrine factors underlying reproductive suppression/development in NMRs, we quantified plasma gonadal steroids and GnRH-1- and kisspeptin-immunoreactive (ir) neurons in subordinate adults and in those allowed to develop into breeders, with or without subsequent gonadectomy. In males and females, respectively, plasma testosterone and progesterone are higher in breeders than in subordinates. No such distinction occurs for plasma estradiol; its presence after gonadectomy and its positive correlation with adrenal estradiol suggest an adrenal source. Numbers of GnRH-1-ir cell bodies do not differ between gonad-intact breeders and subordinates within or between the sexes. As in phylogenetically related guinea pigs, kisspeptin-ir processes pervade the internal and external zones of the median eminence. Their distribution is consistent with actions on GnRH-1 neurons at perikaryal and/or terminal levels. In previously investigated species, numbers of kisspeptin-ir cell bodies vary from substantial to negligible according to sex and/or reproductive state. NMRs are exceptional: irrespective of sex, reproductive state, or presence of gonads, substantial numbers of kisspeptin-ir cell bodies are detected in the rostral periventricular region of the third ventricle (RP3V) and in the anterior periventricular (PVa), arcuate, and dorsomedial hypothalamic nuclei. Nevertheless, the greater number in the RP3V/PVa of female breeders compared with female subordinates or male breeders suggests that emergence from a hypogonadotrophic state in females may involve kisspeptin-related mechanisms similar to those underlying puberty or seasonal breeding in other species.


Subject(s)
Brain/cytology , Cooperative Behavior , Kisspeptins/metabolism , Neurons/metabolism , Receptors, LHRH/metabolism , Sexual Behavior, Animal/physiology , Animals , Body Weight , Castration , Cell Count , Estradiol/blood , Female , Male , Mole Rats , Progesterone/blood , Radioimmunoassay , Testosterone/blood
11.
J Comp Neurol ; 521(13): 3030-41, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23504980

ABSTRACT

RFamide-related peptide-3 (RFRP-3) neurons have been shown to inhibit gonadotropin-releasing hormone (GnRH) neuronal activity and hence reproduction in birds and eutherian mammals. They have also been proposed to have a direct hypophysiotropic effect on pituitary gonadotropin release. We used a new RFRP-3 antibody to characterize the cell body distribution and fiber projections of RFRP-3 neurons in the adult female brushtail possum brain. RFRP-3-immunoreactive cell bodies were found scattered within the dorsomedial hypothalamus and the dorsomedial half of the ventromedial hypothalamus, while GnRH neurons were observed scattered rostrocaudally along the lateral septum, rostral to the medial septum. There was a significant 2-fold increase in the RFRP-3 cell body number during the nonbreeding season (summer) compared to the breeding season (winter). Immunoreactive RFRP-3 fibers were distributed throughout the thalamus, preoptic area, and hypothalamus. Very few fibers were observed in the median eminence, especially in the external zone. Intraperitoneal injection of the retrograde tracer Fluoro-Gold resulted in the labeling of 40% of hypophysiotropic tuberoinfundibular dopaminergic (tyrosine hydroxylase-positive) neurons; however, <10% of zona incerta dopaminergic neurons (which are not hypophysiotropic) or RFRP-3 neurons were labeled with this tracer. These observations suggest that RFRP-3 exhibits a seasonal fluctuation in cell numbers, as seen in sheep and birds, which is consistent with an increased inhibitory tone during the nonbreeding season. The lack of RFRP-3 fibers in the median eminence and of Fluoro-Gold uptake from the periphery imply that the actions of this peptide occur primarily centrally rather than at the anterior pituitary gland.


Subject(s)
Breeding , Gene Expression Regulation/physiology , Hypothalamus/cytology , Neurons/metabolism , Neuropeptides/metabolism , Seasons , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Nerve Fibers/metabolism , Neural Pathways/physiology , Stilbamidines/metabolism , Trichosurus , Tyrosine 3-Monooxygenase/metabolism
12.
J Clin Endocrinol Metab ; 97(12): E2221-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015653

ABSTRACT

OBJECTIVE: Kisspeptins were recently identified as hypothalamic neuropeptides that control GnRH release at pubertal onset and in adults via the activation of KISS-1 receptor (KISS1R). Here, we have tested whether the fetal activation of the gonadotropic axis is related to the hypothalamic expression of kisspeptins and KISS1R. DESIGN AND METHODS: LH and FSH levels were measured in fetal blood from the 15th week of gestation (WG) to birth. Immunohistochemistry was performed on the hypothalamus and pituitary at different developmental stages. RESULTS: Immunostaining for kisspeptins and KISS1R appeared for both proteins in the hypothalamus as early as 15 WG and subsequently increased until 30-31 WG. In the meantime, serum LH and FSH levels decreased from postmenopausal levels in females or adult levels in males to very low levels. At full term, kisspeptin and KISS1R staining was still observed in the paraventricular, supraoptic, and ventromedial hypothalamic nuclei but not in the arcuate nucleus or median eminence. Hypothalamic GnRH staining was observed at 15 WG and did not vary after the first trimester. In an arhinencephalic fetus of 23 WG, very few GnRH neurons were observed in the hypothalamus, but serum FSH and LH levels were postmenopausal. CONCLUSION: Serum LH and FSH levels are independent from GnRH and kisspeptins at midgestation, and then GnRH progressively controls LH and FSH release. A shift from kisspeptin-independent to kisspeptin-dependent GnRH-induced LH and FSH release seems to occur after 30-31 WG. In addition to their function in adults, kisspeptins are also the master regulators of the gonadotropic axis activation in the fetus.


Subject(s)
Follicle Stimulating Hormone/metabolism , Hypothalamus/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Pregnancy Trimester, Second/metabolism , Pregnancy Trimester, Third/metabolism , Receptors, G-Protein-Coupled/metabolism , Autopsy , Down-Regulation , Female , Fetal Blood/chemistry , Fetal Blood/metabolism , Fetus/metabolism , Follicle Stimulating Hormone/blood , Gestational Age , HEK293 Cells , Humans , Hypothalamus/embryology , Hypothalamus/pathology , Luteinizing Hormone/blood , Male , Models, Biological , Pregnancy , Pregnancy Trimester, Second/blood , Pregnancy Trimester, Third/blood , Receptors, Kisspeptin-1
13.
J Clin Invest ; 122(10): 3791-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23006326

ABSTRACT

Hyperprolactinemia is the most common cause of hypogonadotropic anovulation and is one of the leading causes of infertility in women aged 25-34. Hyperprolactinemia has been proposed to block ovulation through inhibition of GnRH release. Kisspeptin neurons, which express prolactin receptors, were recently identified as major regulators of GnRH neurons. To mimic the human pathology of anovulation, we continuously infused female mice with prolactin. Our studies demonstrated that hyperprolactinemia in mice induced anovulation, reduced GnRH and gonadotropin secretion, and diminished kisspeptin expression. Kisspeptin administration restored gonadotropin secretion and ovarian cyclicity, suggesting that kisspeptin neurons play a major role in hyperprolactinemic anovulation. Our studies indicate that administration of kisspeptin may serve as an alternative therapeutic approach to restore the fertility of hyperprolactinemic women who are resistant or intolerant to dopamine agonists.


Subject(s)
Anovulation/drug therapy , Hyperprolactinemia/drug therapy , Kisspeptins/therapeutic use , Animals , Anovulation/etiology , Anovulation/physiopathology , Disease Models, Animal , Drug Evaluation, Preclinical , Estrous Cycle/drug effects , Female , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/metabolism , Gonadotropins, Pituitary/biosynthesis , Gonadotropins, Pituitary/blood , Gonadotropins, Pituitary/metabolism , Hyperprolactinemia/chemically induced , Hyperprolactinemia/complications , Hyperprolactinemia/physiopathology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Infusion Pumps, Implantable , Kisspeptins/biosynthesis , Kisspeptins/genetics , Male , Mice , Prolactin/administration & dosage , Prolactin/toxicity , Pulsatile Flow , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
14.
Brain Res ; 1467: 1-9, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22668987

ABSTRACT

An intact hypothalamic kiss1/kisspeptin/kiss1r complex is a prerequisite for reproductive competence, and kisspeptin treatment could be a practical therapeutic approach to some problems of infertility. One such disorder is polycystic ovarian syndrome (PCOS), a common cause of infertility affecting more than 100 million women. A rodent model of PCOS is the prepubertal female rat treated for a prolonged period with dihydrotestosterone (DHT), which induces many of the metabolic characteristics of the syndrome. We hypothesized that hypothalamic kiss1 mRNA levels, and kisspeptin immunoreactivity (ir), would be abnormal in these rats. Prepubertal female rats were exposed to DHT for 60 days. Rats were killed in two groups: at 26 and 60 days of DHT exposure. Kiss1 mRNA was quantified in hypothalamus, pituitary, ovary and visceral adipose tissue. Separate groups of rats provided brain tissue for immunohistochemical analysis of kisspeptin-ir. At 26 days of DHT exposure, hypothalamic kiss1 mRNA was severely depleted. In contrast DHT had no effect on pituitary kiss1 expression but it significantly increased levels of kiss1 mRNA in fat (+9-fold; p<0.01) and in ovary (+3-fold; p<0.05). At 60days, kiss1 expression had reverted to normal in hypothalamus and ovary but remained elevated in fat (+4-fold; p<0.05). Immunohistochemical analysis revealed that after 26 days of exposure to DHT, kisspeptin-ir was almost completely absent in the arcuate nucleus and a large depletion in kisspeptin +ve fibers was also seen in the paraventricular nucleus, supraoptic nucleus and in the anteroventral periventricular area. At 60 days, despite restored normal levels of kiss1 mRNA, hypothalamic kisspeptin-ir remained depleted in the treated rats. In summary Kiss1 gene expression is differentially affected in various tissues by chronic exposure to dihydrotestosterone in a rat model of polycystic ovary syndrome. In hypothalamus, specifically, kiss1 mRNA, and levels of kisspeptin immunoreactivity, are significantly reduced. Since these rats exhibit many of the characteristics of polycystic ovary syndrome, we suggest that atypical kiss1 expression may contribute to the multiple tissue abnormalities observed in women with this disorder. However, and of some importance, our data do not appear to be consistent with the elevated levels of LH seen in women with PCOS; i.e. reduced levels of hypothalamic kiss1 mRNA and kisspeptin immunoreactivity observed in DHT-treated rats are unlikely to produce elevated LH secretion.


Subject(s)
Hypothalamus/metabolism , Kisspeptins/biosynthesis , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , RNA, Messenger/biosynthesis , Adipose Tissue/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/physiology , Dihydrotestosterone/pharmacology , Energy Metabolism/physiology , Female , Gonadal Steroid Hormones/metabolism , Immunohistochemistry , Ovary/metabolism , Pituitary Gland/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
15.
Neuroendocrinology ; 95(4): 305-16, 2012.
Article in English | MEDLINE | ID: mdl-22286004

ABSTRACT

OBJECTIVE: Gonadotropin-inhibitory hormone (GnIH)-3 is a neuropeptide that plays a major role in the regulation of reproduction and feeding in mammals. MATERIALS AND METHODS: We measured endocrine and behavioural parameters of reproduction in sheep, and sexual behaviour in sheep, mice and cynomolgus monkeys. In addition, GnIH gene expression (in situ hybridization) was examined in ewes, and effects of GnIH-3 on food intake and energy expenditure were measured in various species. GnIH-3 was infused (i.v.) into ewes after an i.m. injection of estradiol benzoate to determine whether the peptide blocks the surge in luteinizing hormone (LH) secretion. RESULTS: GnIH gene expression was reduced in the preovulatory period in ewes. Infusion (i.v.) of GnIH-3 blocked the estrogen-induced LH surge (in ewes). Intracerebroventricular infusion had no effect on female or male sexual behaviour in each of the three species, but increased food intake. There were no effects on energy expenditure in sheep or rats. GnIH increased fos protein (immunohistochemistry) was seen in orexigenic neurons (in sheep and rats), but also in anorexigenic neurons (in sheep). CONCLUSIONS: GnIH-3 reduces reproductive hormone levels and increases food intake in mammals without reducing energy expenditure. There is minimal effect on reproductive behaviour. The dual effect on reproduction and feeding suggests that GnIH-3 provides a molecular switch between these two functions. Blockade of the positive feedback effect of estrogen with parenteral infusion indicates that this peptide may have utility as a blocker of reproductive function in mammals.


Subject(s)
Feeding Behavior/physiology , Glycoproteins/physiology , Hypothalamic Hormones/physiology , Reproduction , Animals , Drug Evaluation, Preclinical , Eating/drug effects , Eating/genetics , Eating/physiology , Feeding Behavior/drug effects , Female , Genes, Switch/physiology , Glycoproteins/genetics , Glycoproteins/pharmacology , Hypothalamic Hormones/genetics , Hypothalamic Hormones/pharmacology , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Neuropeptides/genetics , Neuropeptides/pharmacology , Neuropeptides/physiology , Rats , Reproduction/drug effects , Reproduction/genetics , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology , Sheep
16.
J Chem Neuroanat ; 43(1): 14-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21983419

ABSTRACT

In rodents, the neuropeptide galanin (Gal) is involved in controlling the release of gonadotrophin-releasing hormone (GnRH). In the female, this peptide is colocalized in a subpopulation of GnRH neurones and its expression is stimulated by oestradiol. In the ewe, the morphofunctional relationship between these two neuronal peptides is poorly understood. The morphological interaction between Gal and GnRH was studied in ewes treated with oestradiol or with colchicine and in control animals. Five ewes were treated for 6h with oestradiol implants, a treatment known to induce a preovulatory surge of GnRH, and compared with five control animals. In addition, four animals received an intracerebroventricular injection of colchicine known to increase the intracellular level of galanin immunoreactivity. The morphological relationship between the two peptides was investigated by immunofluorescence using specific antibodies on the same sections, and the results were analysed using confocal microscopy. In colchicine-treated ewes, numerous Gal-immunoreactive neurones were found in the preoptic area in the vicinity of GnRH-immunoreactive neurones, but the two peptides were never observed in the same neurone. In all animals, Gal-ir fibres were observed to be in apposition to GnRH-containing perikarya in the preoptic area and these appositions were more numerous in oestradiol-treated ewes than in control animals. In contrast with rodents, galanin was not colocalized with GnRH in the neurones of the preoptic area of ewes, but this peptide could control GnRH neuronal secretion through axosomatic interactions. However, the presence of synaptic contacts between galanin terminals and GnRH perikarya needs to be confirmed by electron microscopy. As in rodents and primates, galanin could mediate the positive feedback of oestradiol on GnRH neurones during the preovulatory surge in ewes.


Subject(s)
Estradiol/pharmacology , Galanin/metabolism , Gonadotropin-Releasing Hormone/metabolism , Nerve Net/metabolism , Neurons/metabolism , Preoptic Area/metabolism , Animals , Female , Nerve Net/drug effects , Neurons/drug effects , Preoptic Area/drug effects , Progesterone/pharmacology , Sheep
17.
J Comp Neurol ; 519(17): 3456-69, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21800299

ABSTRACT

It is now well established that the kisspeptin neurons of the hypothalamus play a key role in regulating the activity of gonadotropin-releasing hormone (GnRH) neurons. The population of kisspeptin neurons residing in the rostral periventricular region of the third ventricle (RP3V), encompassing the anteroventral periventricular (AVPV) and periventricular preoptic nuclei (PVpo), are implicated in the generation of the preovulatory GnRH surge mechanism and puberty onset in female rodents. The present study examined whether these kisspeptin neurons may express other neuropeptides in the adult female mouse. Initially, the distribution of galanin, neurotensin, met-enkephalin (mENK), and cholecystokinin (CCK)-immunoreactive cells was determined within the RP3V of colchicine-treated mice. Subsequent experiments, using a new kisspeptin-10 antibody raised in sheep, examined the relationship of these neuropeptides to kisspeptin neurons. No evidence was found for expression of neurotensin or CCK by RP3V kisspeptin neurons, but subpopulations of kisspeptin neurons were observed to express galanin and mENK. Dual-labeled RP3V kisspeptin/galanin cells represented 7% of all kisspeptin and 21% of all galanin neurons whereas dual-labeled kisspeptin/mENK cells represented 28-38% of kisspeptin neurons and 58-68% of the mENK population, depending on location within the AVPV or PVpo. Kisspeptin neurons in the arcuate nucleus were also found to express galanin but not mENK. These observations indicate that, like the kisspeptin population of the arcuate nucleus, kisspeptin neurons in the RP3V also co-express a range of neuropeptides. This pattern of co-expression should greatly increase the dynamic range with which kisspeptin neurons can modulate the activity of their afferent neurons.


Subject(s)
Enkephalin, Methionine/biosynthesis , Galanin/biosynthesis , Gene Expression Regulation , Hypothalamus/metabolism , Kisspeptins/biosynthesis , Neurons/metabolism , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Arcuate Nucleus of Hypothalamus/metabolism , Female , Hypothalamus/chemistry , Mice , Neurons/chemistry , Third Ventricle/chemistry , Third Ventricle/metabolism
18.
Endocrinology ; 152(4): 1527-40, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21325050

ABSTRACT

Kisspeptins are new actors in the neuroendocrine regulation of reproduction. In vertebrates, the number of kiss genes varies from none to three. Zebrafish have two kiss genes, kiss1 and kiss2, and two kiss receptors (GPR54), kiss1r and kiss2r. To provide detailed information on the organization of the kiss systems in zebrafish, antibodies were raised against the C terminus of zebrafish preproKiss1 and preproKiss2. Immunohistochemistry fully confirmed in situ hybridization data, showing that kiss1-expressing neurons are only located in the habenular nucleus, while kiss2-expressing neurons are found in the dorsal and ventral hypothalamus. Kiss1-expressing cells project only to the interpeduncular and raphe nuclei and strongly expressed the kiss1r receptor. In contrast, kiss2-expressing cells are mostly present in the dorsal and ventral hypothalamus and project widely into the subpallium, the preoptic area, the thalamus, the ventral and caudal hypothalamus, and the mesencephalon. All these regions strongly expressed the kiss2r messengers. Kiss2 fibers profusely innervate the ventral forebrain and notably made close apposition with GnRH3 neurons. Estrogen treatment of juvenile fish with estradiol causes increase in kiss2 and kiss2r expression. In the pituitary gland, no proKiss2- positive fibers were detected, while positive cells were observed in the pars intermedia. In addition to proposing a successful strategy to develop antibodies to kisspeptins, these data indicate that the kiss2 systems of zebrafish are implicated in reproductive events, while the kiss1 gene would play other functions that remain to be established.


Subject(s)
Brain/metabolism , Receptors, G-Protein-Coupled/metabolism , Tumor Suppressor Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Brain/drug effects , Estrogens/pharmacology , Evolution, Molecular , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/metabolism , Habenula/drug effects , Habenula/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Kisspeptins , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Polymerase Chain Reaction , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Receptors, Kisspeptin-1 , Zebrafish
19.
Endocrinology ; 152(1): 214-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21047947

ABSTRACT

We tested the working hypothesis that Fos will identify the critical population of kisspeptin neurons that accompanies the LHRH surge using a synchronized follicular phase model in intact cycling ewes. The model generates an LH surge that starts within a defined 2-h window in a 20-d synchronized cycle. With a modified push-pull cannula in vivo LHRH release from the median eminence was sampled in luteal phase ewes, ewes undergoing an LH surge for 2-4 h, and postsurge animals whose LH surge peaked 10-12 h earlier. In vivo release of LHRH was lower in the luteal and follicular phases than in animals undergoing an LH surge (P < 0.01); it fell to presurge levels after the LH surge. Ewes killed 2-4 h after the surge started, expressed Fos in a large portion of preoptic area (POA) kisspeptin (53.90 ± 4.69%, P < 0.01) and LHRH neurons (48.20 ± 4.49%, P < 0.0001) compared with animals euthanized at any of the other times tested (under <5% of the cells activated). Little Fos activation (under 5%) was observed during any of the times sampled in arcuate (Arc) kisspeptin neurons. The relationship between the number of LHRH neurons and the POA kisspeptin neurons stimulated showed a striking positive correlation with r(2) = 0.68, P = 0.0003, reinforcing the evidence that POA kisspeptin neurons actively participate in the stimulation of LHRH surges.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/metabolism , Median Eminence/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Sheep/physiology , Tumor Suppressor Proteins/metabolism , Animals , Gene Expression Regulation/physiology , Neurons/physiology , Preoptic Area , Tumor Suppressor Proteins/genetics
20.
Eur J Neurosci ; 32(12): 2031-41, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21143658

ABSTRACT

The neurohormone gonadotropin-releasing hormone (GnRH) is critical for all the aspects of reproductive life in vertebrates. GnRH is secreted by a small number of neurons dispersed within the preoptic-hypothalamic region. These neurons are derived from the embryonic olfactory pit. They then migrate along olfactory, vomeronasal and terminal nerves to their final destination. Classical approaches to study the regulation of GnRH secretion during the reproductive cycle have focused on the various neuronal inputs on GnRH neurons and their regulation by ovarian steroids. However, it is well known that steroids will change the microenvironment of neuronal networks and can induce plasticity and functional changes. In this review, we will focus on the intimate relationship of developing and adult GnRH neurons with the polysialylated form of neural cell adhesion molecule (PSA-NCAM), a major molecular actor in the morphogenesis and adult plasticity of the nervous system. We will first recapitulate the spatiotemporal relationship between PSA-NCAM and migrating GnRH neurons during embryogenesis of various vertebrate species and discuss its importance for GnRH neuron development as shown by various loss of function studies. In the adult, we will review the relationships between PSA-NCAM and GnRH neurons across various physiological states, and open the discussion to the use of new model systems that can help to unravel the function and mechanism of action of PSA-NCAM on GnRH neuronal network activity and GnRH release.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neuronal Plasticity/physiology , Neurons/physiology , Sialic Acids/metabolism , Animals , Cell Movement , Humans , Hypothalamus/anatomy & histology , Hypothalamus/growth & development , Hypothalamus/physiology , Models, Neurological , Neurons/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...