Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
J Neuroendocrinol ; 34(5): e13141, 2022 05.
Article in English | MEDLINE | ID: mdl-35726373

ABSTRACT

This article highlights key milestones in GnRH research that have occurred in the 50 plus years since the discovery of the decapeptide. It is by no means exhaustive and inevitably reflects our limitations and idiosyncratic perspectives.


Subject(s)
Gonadotropin-Releasing Hormone , Kisspeptins , Neurons
2.
J Clin Endocrinol Metab ; 107(1): e328-e347, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34387319

ABSTRACT

CONTEXT: Recent evidence suggests that vasomotor symptoms (VMS) or hot flashes in the postmenopausal reproductive state and polycystic ovary syndrome (PCOS) in the premenopausal reproductive state emanate from the hyperactivity of Kiss1 neurons in the hypothalamic infundibular/arcuate nucleus (KNDy neurons). OBJECTIVE: We demonstrate in 2 murine models simulating menopause and PCOS that a peripherally restricted kappa receptor agonist (PRKA) inhibits hyperactive KNDy neurons (accessible from outside the blood-brain barrier) and impedes their downstream effects. DESIGN: Case/control. SETTING: Academic medical center. PARTICIPANTS: Mice. INTERVENTIONS: Administration of peripherally restricted kappa receptor agonists and frequent blood sampling to determine hormone release and body temperature. MAIN OUTCOME MEASURES: LH pulse parameters and body temperature. RESULTS: First, chronic administration of a PRKA to bilaterally ovariectomized mice with experimentally induced hyperactivity of KNDy neurons reduces the animals' elevated body temperature, mean plasma LH level, and mean peak LH per pulse. Second, chronic administration of a PRKA to a murine model of PCOS, having elevated plasma testosterone levels and irregular ovarian cycles, suppresses circulating levels of LH and testosterone and restores normal ovarian cyclicity. CONCLUSION: The inhibition of kisspeptin neuronal activity by activation of kappa receptors shows promise as a novel therapeutic approach to treat both VMS and PCOS in humans.


Subject(s)
Hot Flashes/drug therapy , Kisspeptins/antagonists & inhibitors , Menopause/metabolism , Polycystic Ovary Syndrome/drug therapy , Receptors, Opioid, kappa/agonists , Animals , Buprenorphine/administration & dosage , Disease Models, Animal , Female , Hot Flashes/blood , Hot Flashes/etiology , Humans , Kisspeptins/metabolism , Meloxicam/administration & dosage , Menopause/blood , Mice , Neurons/drug effects , Neurons/metabolism , Polycystic Ovary Syndrome/metabolism , Receptors, Opioid, kappa/metabolism , Vasomotor System/drug effects
3.
J Neuroendocrinol ; : e12600, 2018 Apr 14.
Article in English | MEDLINE | ID: mdl-29656508

ABSTRACT

The 3rd World Conference on Kisspeptin, "Kisspeptin 2017: Brain and Beyond" was held March 30-31 at the Rosen Centre Hotel in Orlando, Florida, providing an international forum for multidisciplinary scientists to meet and share cutting-edge research on kisspeptin biology and its relevance to human health and disease. The meeting built upon previous world conferences focused on the role of kisspeptin and associated peptides in the control of gonadotropin-releasing hormone (GnRH) secretion and reproduction. Based on recent discoveries, the scope of this meeting was expanded to include functions of kisspeptin and related peptides in other physiological systems including energy homeostasis, pregnancy, ovarian and uterine function, and thermoregulation. In addition, discussions addressed the translation of basic knowledge of kisspeptin biology to the treatment of disease, with the goal of seeking consensus about the best approaches to improve human health. The two-day meeting featured a non-traditional structure, with each day starting with poster sessions followed by lunch discussions and facilitated large-group sessions with short presentations to maximize the exchange of new, unpublished data. Topics were identified by a survey prior to the meeting, and focused on major unresolved questions, important controversies, and future directions in the field. Finally, career development activities provided mentoring for trainees and junior investigators, and networking opportunities for those individuals with established researchers in the field. Overall, the meeting was rated as a success by attendees and covered a wide range of lively and provocative discussion topics on the changing nature of the field of "kisspeptinology" and its future. This article is protected by copyright. All rights reserved.

4.
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
5.
Menopause ; 22(12): 1328-34, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25988798

ABSTRACT

OBJECTIVE: The etiology of postmenopausal hot flashes is poorly understood, making it difficult to develop and target ideal therapies. A network of hypothalamic estrogen-sensitive neurons producing kisspeptin, neurokinin B and dynorphin-called KNDy neurons-are located adjacent to the thermoregulatory center. KNDy neurons regulate pulsatile secretion of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH). Dynorphin may inhibit this system by binding κ opioid receptors within the vicinity of KNDy neurons. We hypothesize that hot flashes are reduced by KNDy neuron manipulation. METHODS: A double-blind, cross-over, placebo-controlled pilot study evaluated the effects of a κ agonist. Hot flash frequency was the primary outcome. Twelve healthy postmenopausal women with moderate to severe hot flashes (aged 48-60 y) were randomized. Eight women with sufficient baseline hot flashes for statistical analysis completed all three interventions: placebo, standard-dose pentazocine/naloxone (50/0.5 mg), or low-dose pentazocine/naloxone (25/0.25 mg). In an inpatient research setting, each participant received the three interventions, in randomized order, on three separate days. On each day, an intravenous catheter was inserted for LH blood sampling, and skin conductance and Holter monitors were placed. Subjective hot flash frequency and severity were recorded. RESULTS: The mean (SEM) hot flash frequency 2 to 7 hours after therapy initiation was lower than that for placebo (standard-dose κ agonist, 4.75 [0.67] hot flashes per 5 h; low-dose κ agonist, 4.50 [0.57] hot flashes per 5 h; placebo, 5.94 [0.78] hot flashes per 5 h; P = 0.025). Hot flash intensity did not vary between interventions. LH pulsatility mirrored objective hot flashes in some--but not all--women. CONCLUSIONS: This pilot study suggests that κ agonists may affect menopausal vasomotor symptoms.


Subject(s)
Hot Flashes/drug therapy , Pentazocine/therapeutic use , Postmenopause , Receptors, Opioid, kappa/agonists , Analgesics, Opioid , Cross-Over Studies , Double-Blind Method , Dynorphins/biosynthesis , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , Kisspeptins/biosynthesis , Luteinizing Hormone/metabolism , Middle Aged , Neurokinin B/biosynthesis , Neurons/physiology , Pentazocine/adverse effects , Placebos
6.
J Neurosci ; 35(14): 5549-56, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25855171

ABSTRACT

Hypothalamic neuronal populations are central regulators of energy homeostasis and reproductive function. However, the ontogeny of these critical hypothalamic neuronal populations is largely unknown. We developed a novel approach to examine the developmental pathways that link specific subtypes of neurons by combining embryonic and adult ribosome-tagging strategies in mice. This new method shows that Pomc-expressing precursors not only differentiate into discrete neuronal populations that mediate energy balance (POMC and AgRP neurons), but also into neurons critical for puberty onset and the regulation of reproductive function (Kiss1 neurons). These results demonstrate a developmental link between nutrient-sensing and reproductive neuropeptide synthesizing neuronal populations and suggest a potential pathway that could link maternal nutrition to reproductive development in the offspring.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Hypothalamus/cytology , Kisspeptins/metabolism , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Stem Cells/physiology , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Dependovirus/genetics , Embryo, Mammalian , Genetic Vectors/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Kisspeptins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Pro-Opiomelanocortin/genetics , RNA, Messenger/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 111(28): 10335-40, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982149

ABSTRACT

Most central neurons in the mammalian brain possess an appendage called a primary cilium that projects from the soma into the extracellular space. The importance of these organelles is highlighted by the fact that primary cilia dysfunction is associated with numerous neuropathologies, including hyperphagia-induced obesity, hypogonadism, and learning and memory deficits. Neuronal cilia are enriched for signaling molecules, including certain G protein-coupled receptors (GPCRs), suggesting that neuronal cilia sense and respond to neuromodulators in the extracellular space. However, the impact of cilia on signaling to central neurons has never been demonstrated. Here, we show that the kisspeptin receptor (Kiss1r), a GPCR that is activated by kisspeptin to regulate the onset of puberty and adult reproductive function, is enriched in cilia projecting from mouse gonadotropin-releasing hormone (GnRH) neurons. Interestingly, GnRH neurons in adult animals are multiciliated and the percentage of GnRH neurons possessing multiple Kiss1r-positive cilia increases during postnatal development in a progression that correlates with sexual maturation. Remarkably, disruption of cilia selectively on GnRH neurons leads to a significant reduction in kisspeptin-mediated GnRH neuronal activity. To our knowledge, this result is the first demonstration of cilia disruption affecting central neuronal activity and highlights the importance of cilia for proper GPCR signaling.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Reproduction/physiology , Signal Transduction/physiology , Animals , Cell Line , Cilia/genetics , Cilia/metabolism , Female , Gonadotropin-Releasing Hormone/genetics , Kisspeptins/genetics , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/cytology , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1 , Sexual Maturation/physiology
9.
Am J Physiol Endocrinol Metab ; 305(11): E1384-97, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24105416

ABSTRACT

Kisspeptin (Kiss1) neurons in the rostral periventricular area of the third ventricle (RP3V) provide excitatory drive to gonadotropin-releasing hormone (GnRH) neurons to control fertility. Using whole cell patch clamp recording and single-cell (sc)RT-PCR techniques targeting Kiss1-CreGFP or tyrosine hydroxylase (TH)-EGFP neurons, we characterized the biophysical properties of these neurons and identified the critical intrinsic properties required for burst firing in 17ß-estradiol (E2)-treated, ovariectomized female mice. One-fourth of the RP3V Kiss1 neurons exhibited spontaneous burst firing. RP3V Kiss1 neurons expressed a hyperpolarization-activated h-current (Ih) and a T-type calcium current (IT), which supported hyperpolarization-induced rebound burst firing. Under voltage clamp conditions, all Kiss1 neurons expressed a kinetically fast Ih that was augmented 3.4-fold by high (LH surge-producing)-E2 treatment. scPCR analysis of Kiss1 neurons revealed abundant expression of the HCN1 channel transcripts. Kiss1 neurons also expressed a Ni(2+)- and TTA-P2-sensitive IT that was augmented sixfold with high-E2 treatment. CaV3.1 mRNA was also highly expressed in these cells. Current clamp analysis revealed that rebound burst firing was induced in RP3V Kiss1 neurons in high-E2-treated animals, and the majority of Kiss1 neurons had a hyperpolarization threshold of -84.7 mV, which corresponded to the V½ for IT de-inactivation. Finally, Kiss1 neurons in the RP3V were hyperpolarized by µ- and κ-opioid and GABAB receptor agonists, suggesting that these pathways also contribute to rebound burst firing. Therefore, Kiss1 neurons in the RP3V express the critical channels and receptors that permit E2-dependent rebound burst firing and provide the biophysical substrate that drives the preovulatory surge of GnRH.


Subject(s)
Estradiol/pharmacology , Kisspeptins/metabolism , Neurons/physiology , Preoptic Area/metabolism , Animals , Female , Follicular Phase/drug effects , Follicular Phase/metabolism , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kisspeptins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Ovariectomy , Preoptic Area/drug effects , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Third Ventricle/drug effects , Third Ventricle/metabolism
10.
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
11.
Adv Exp Med Biol ; 784: 3-7, 2013.
Article in English | MEDLINE | ID: mdl-23549999

ABSTRACT

Research in the nineteenth and early twentieth century established that the brain awakens reproduction, governs reproductive activity in the adult of virtually all vertebrates. By 1950, nearly 100 years later, scientists realized that the hypothalamus and its neurosecretory products play a key role in regulating gonadal function in both males and females. Another 20 years would be required to reveal the chemical identity of GnRH and establish that neurons producing GnRH represent the final common pathway through which the brain regulates gonadotropin secretion. It had also become clear that GnRH neurons behave more like motor neurons-better perhaps at going than stopping-and are themselves regulated by a complex network of afferent inputs, which guide the tempo of sexual maturation, regulate estrous and menstrual cycles, control seasonal breeding, and stop reproduction under adversity. In 2003, the revelation that kisspeptin and its receptor are critical for reproduction opened a floodgate of research documenting the role of kisspeptin neurons as central processors of reproduction. Today, there is wide consensus that kisspeptin signaling in the brain is essential, providing the impetus to GnRH neurons to awaken at puberty and reigning the activity of these neurons when discretion is advised. We celebrate this watershed moment-with full knowledge that time and discovery will provide context and perspective to even these heady days.


Subject(s)
Brain/physiology , Kisspeptins/metabolism , Menstrual Cycle/physiology , Reproduction/physiology , Signal Transduction/physiology , Adult , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Kisspeptins/history , Male , Neurons/metabolism
12.
Endocrinology ; 153(10): 4883-93, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22893725

ABSTRACT

At puberty, neurokinin B (NKB) and kisspeptin (Kiss1) may help to amplify GnRH secretion, but their precise roles remain ambiguous. We tested the hypothesis that NKB and Kiss1 are induced as a function of pubertal development, independently of the prevailing sex steroid milieu. We found that levels of Kiss1 mRNA in the arcuate nucleus (ARC) are increased prior to the age of puberty in GnRH/sex steroid-deficient hpg mice, yet levels of Kiss1 mRNA in wild-type mice remained constant, suggesting that sex steroids exert a negative feedback effect on Kiss1 expression early in development and across puberty. In contrast, levels of Tac2 mRNA, encoding NKB, and its receptor (NK3R; encoded by Tacr3) increased as a function of puberty in both wild-type and hpg mice, suggesting that during development Tac2 is less sensitive to sex steroid-dependent negative feedback than Kiss1. To compare the relative responsiveness of Tac2 and Kiss1 to the negative feedback effects of gonadal steroids, we examined the effect of estradiol (E(2)) on Tac2 and Kiss1 mRNA and found that Kiss1 gene expression was more sensitive than Tac2 to E(2)-induced inhibition at both juvenile and adult ages. This differential estrogen sensitivity was tested in vivo by the administration of E(2). Low levels of E(2) significantly suppressed Kiss1 expression in the ARC, whereas Tac2 suppression required higher E(2) levels, supporting differential sensitivity to E(2). Finally, to determine whether inhibition of NKB/NK3R signaling would block the onset of puberty, we administered an NK3R antagonist to prepubertal (before postnatal d 30) females and found no effect on markers of pubertal onset in either WT or hpg mice. These results indicate that the expression of Tac2 and Tacr3 in the ARC are markers of pubertal activation but that increased NKB/NK3R signaling alone is insufficient to trigger the onset of puberty in the mouse.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Feedback, Physiological/physiology , Kisspeptins/metabolism , Protein Precursors/metabolism , Sexual Maturation/physiology , Tachykinins/metabolism , Animals , Estradiol/blood , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/genetics , Luteinizing Hormone/blood , Mice , Mice, Knockout , Neurokinin B/genetics , Neurokinin B/metabolism , Neurons/metabolism , Protein Precursors/genetics , Signal Transduction/physiology , Tachykinins/genetics
13.
14.
J Neurosci ; 32(7): 2388-97, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22396413

ABSTRACT

Human genetic studies have revealed that neurokinin B (NKB) and its receptor, neurokinin-3 receptor (NK3R), are essential elements for normal reproduction; however, the precise role of NKB-NK3R signaling in the initiation of puberty remains unknown. We investigated here the regulation of Tac2 and Tacr3 mRNAs (encoding NKB and NK3R, respectively) in female rats and demonstrated that their hypothalamic expression is increased along postnatal maturation. At puberty, both genes were widely expressed throughout the brain, including the lateral hypothalamic area and the arcuate nucleus (ARC)/medial basal hypothalamus, where the expression of Tacr3 increased across pubertal transition. We showed that central administration of senktide (NK3R agonist) induced luteinizing hormone (LH) secretion in prepubertal and peripubertal females. Conversely, chronic infusion of an NK3R antagonist during puberty moderately delayed the timing of vaginal opening (VO) and tended to decrease LH levels. The expression of NKB and its receptor was sensitive to changes in metabolic status during puberty, as reflected by a reduction in Tacr3 (and, to a lesser extent, Tac2) expression in the ARC after a 48 h fast. Yet, acute LH responses to senktide in pubertal females were preserved, if not augmented, under fasting conditions, suggesting sensitization of the NKB-NK3R-gonadotropin-releasing hormone signaling pathway under metabolic distress. Moreover, repeated administration of senktide to female rats with pubertal arrest due to chronic undernutrition rescued VO (in ∼50% of animals) and potently elicited LH release. Altogether, our observations suggest that NKB-NK3R signaling plays a role in pubertal maturation and that its alterations may contribute to pubertal disorders linked to metabolic stress and negative energy balance.


Subject(s)
Metabolome/physiology , Neurokinin B/physiology , Sexual Maturation/physiology , Age Factors , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/metabolism , Energy Metabolism/physiology , Female , Neurokinin B/metabolism , Rats , Rats, Wistar , Receptors, Neurokinin-3/metabolism , Receptors, Neurokinin-3/physiology
15.
Endocrinology ; 152(11): 4298-309, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21933870

ABSTRACT

Neurons that produce kisspeptin play a critical role in reproduction. However, understanding the molecular physiology of kisspeptin neurons has been limited by the lack of an in vivo marker for those cells. Here, we report the development of a Kiss1-CreGFP knockin mouse, wherein the endogenous Kiss1 promoter directs the expression of a Cre recombinase-enhanced green fluorescent protein (GFP) fusion protein. The pattern of GFP expression in the brain of the knockin recapitulates what has been described earlier for Kiss1 in the male and female mouse, with prominent expression in the arcuate nucleus (ARC) (in both sexes) and the anteroventral periventricular nucleus (in females). Single-cell RT-PCR showed that the Kiss1 transcript is expressed in 100% of GFP-labeled cells, and the CreGFP transcript was regulated by estradiol in the same manner as the Kiss1 gene (i.e. inhibited in the ARC and induced in the anteroventral periventricular nucleus). We used this mouse to evaluate the biophysical properties of kisspeptin (Kiss1) neurons in the ARC of the female mouse. GFP-expressing Kiss1 neurons were identified in hypothalamic slice preparations of the ARC and patch clamped. Whole-cell (and loose attached) recordings revealed that Kiss1 neurons exhibit spontaneous activity and expressed both h- (pacemaker) and T-type calcium currents, and hyperpolarization-activated cyclic nucleotide-regulated 1-4 and CaV3.1 channel subtypes (measured by single cell RT-PCR), respectively. N-methyl-D-aspartate induced bursting activity, characterized by depolarizing/hyperpolarizing oscillations. Therefore, Kiss1 neurons in the ARC share molecular and electrophysiological properties of other CNS pacemaker neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Kisspeptins/metabolism , Neurons/metabolism , Reproduction/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Estradiol/pharmacology , Female , Male , Mice , Mice, Transgenic , Neurons/drug effects , Orchiectomy , Ovariectomy , Reproduction/drug effects
16.
Endocrinology ; 152(5): 2020-30, 2011 May.
Article in English | MEDLINE | ID: mdl-21363930

ABSTRACT

Kisspeptin (encoded by the Kiss1 gene) is an important regulator of reproduction. In rodents, Kiss1 is expressed in two hypothalamic regions, the arcuate nucleus and anteroventral periventricular/ periventricular continuum, where it is regulated by sex steroids. However, the distribution, regulation, and functional significance of neural kisspeptin outside of the hypothalamus have not been studied and are poorly understood. Here, we report the expression of Kiss1 in the amygdala, predominantly in the medial nucleus of the amygdala (MeA), a region implicated in social and emotional behaviors as well as various aspects of reproduction. In gonadally intact rats and mice, Kiss1-expressing neurons were identified in the MeA of both sexes, with higher Kiss1 expression levels in adult males than females in diestrus. In rats, Kiss1 expression in the MeA changed as a function of the estrous cycle, with highest levels at proestrus. Next, we tested whether Kiss1 in the MeA is regulated by the circulating sex steroid milieu. Kiss1 levels in the MeA were low in gonadectomized mice and rats of both sexes, and treatment with either testosterone or estradiol amplified Kiss1 expression in this region. Testosterone's inductive effect on Kiss1 expression in the MeA likely occurs via estrogen receptor-dependent pathways, not through the androgen receptor, because dihydrotestosterone (a nonaromatizable androgen) did not affect MeA Kiss1 levels. Thus, in rodents, Kiss1 is expressed and regulated by sex steroids in the MeA of both sexes and may play a role in modulating reproduction or brain functions that extend beyond reproduction.


Subject(s)
Amygdala/drug effects , Gene Expression/drug effects , Proteins/genetics , Steroids/pharmacology , Amygdala/cytology , Amygdala/metabolism , Androgens/pharmacology , Animals , Estradiol/pharmacology , Estrogens/pharmacology , Estrous Cycle/genetics , Female , In Situ Hybridization , Kisspeptins , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Orchiectomy , Ovariectomy , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Testosterone/pharmacology , Up-Regulation/drug effects
17.
Am J Physiol Endocrinol Metab ; 300(1): E202-10, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21045176

ABSTRACT

Neurokinin B (NKB) and its cognate receptor neurokinin 3 (NK3R) play a critical role in reproduction. NKB and NK3R are coexpressed with dynorphin (Dyn) and kisspeptin (Kiss1) genes in neurons of the arcuate nucleus (Arc). However, the mechanisms of action of NKB as a cotransmitter with kisspeptin and dynorphin remain poorly understood. We explored the role of NKB in the control of LH secretion in the female rat as follows. 1) We examined the effect of an NKB agonist (senktide, 600 pmol, administered into the lateral cerebral ventricle) on luteinizing hormone (LH) secretion. In the presence of physiological levels of estradiol (E(2)), senktide induced a profound increase in serum levels of LH and a 10-fold increase in the number of Kiss1 neurons expressing c-fos in the Arc (P < 0.01 for both). 2) We mapped the distribution of NKB and NK3R mRNAs in the central forebrain and found that both are widely expressed, with intense expression in several hypothalamic nuclei that control reproduction, including the Arc. 3) We studied the effect of E(2) on the expression of NKB and NK3R mRNAs in the Arc and found that E(2) inhibits the expression of both genes (P < 0.01) and that the expression of NKB and NK3R reaches its nadir on the afternoon of proestrus (when circulating levels of E(2) are high). These observations suggest that NKB/NK3R signaling in Kiss1/NKB/Dyn-producing neurons in the Arc has a pivotal role in the control of gonadotropin-releasing hormone (GnRH)/LH secretion and its regulation by E(2)-dependent negative feedback in the rat.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Proteins/metabolism , Receptors, Neurokinin-3/metabolism , Signal Transduction , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Estradiol/metabolism , Estrous Cycle/metabolism , Feedback, Physiological , Female , Gene Expression Regulation , Kisspeptins , Luteinizing Hormone/blood , Neurokinin B/agonists , Neurokinin B/genetics , Neurons/drug effects , Organ Specificity , Peptide Fragments/pharmacology , Prosencephalon/cytology , Prosencephalon/metabolism , Proteins/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Neurokinin-3/agonists , Receptors, Neurokinin-3/genetics , Signal Transduction/drug effects , Substance P/analogs & derivatives , Substance P/pharmacology
18.
J Neurosci ; 30(8): 3124-32, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181609

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons in the basal forebrain are the final common pathway through which the brain regulates reproduction. GnRH secretion occurs in a pulsatile manner, and indirect evidence suggests the kisspeptin neurons in the arcuate nucleus (ARC) serve as the central pacemaker that drives pulsatile GnRH secretion. The purpose of this study was to investigate the possible coexpression of kisspeptin, neurokinin B (NKB), and dynorphin A (Dyn) in neurons of the ARC of the goat and evaluate their potential roles in generating GnRH pulses. Using double and triple labeling, we confirmed that all three neuropeptides are coexpressed in the same population of neurons. Using electrophysiological techniques to record multiple-unit activity (MUA) in the medial basal hypothalamus, we found that bursts of MUA occurred at regular intervals in ovariectomized animals and that these repetitive bursts (volleys) were invariably associated with discrete pulses of luteinizing hormone (LH) (and by inference GnRH). Moreover, the frequency of MUA volleys was reduced by gonadal steroids, suggesting that the volleys reflect the rhythmic discharge of steroid-sensitive neurons that regulate GnRH secretion. Finally, we observed that central administration of Dyn-inhibit MUA volleys and pulsatile LH secretion, whereas NKB induced MUA volleys. These observations are consistent with the hypothesis that kisspeptin neurons in the ARC drive pulsatile GnRH and LH secretion, and suggest that NKB and Dyn expressed in those neurons are involved in the process of generating the rhythmic discharge of kisspeptin.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Tumor Suppressor Proteins/metabolism , Action Potentials/physiology , Animals , Biological Clocks/physiology , Electrophysiology , Estrous Cycle/physiology , Female , Goats , Gonadal Steroid Hormones/metabolism , Kisspeptins , Luteinizing Hormone/metabolism , Neurosecretory Systems/physiology , Ovariectomy , Periodicity , Proteins/metabolism , Reproduction/physiology
19.
Am J Physiol Endocrinol Metab ; 298(1): E80-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19861584

ABSTRACT

In female mammals, increased ovarian estradiol (E(2)) secretion triggers GnRH release from neurons in the basal forebrain, which drives LH secretion from the pituitary and subsequently induces ovulation. However, the neural circuits that activate this preovulatory GnRH/LH surge remain unidentified. Neurotensin is expressed in neurons of the anteroventral periventricular nucleus (AVPV), a region thought to be critical for generating the preovulatory GnRH/LH surge. E(2) induces neurotensin (Nts) gene expression in this region, and blockade of neurotensin signaling reduces the LH surge in the rat. We postulated that neurotensin signaling plays a similar role in generating the E(2)-induced GnRH/LH surge in mice. We used in situ hybridization (ISH) to determine whether E(2) induces Nts expression in the mouse and found evidence to support this proposition. Next, we determined that the neurotensin receptor (Ntsr2) is present in many GnRH-expressing neurons. Since the kisspeptin gene (Kiss1) is expressed in the AVPV and is responsive to E(2), we predicted that some neurons in this region express both Kiss1 and Nts; however, by double-label ISH, we observed no coexpression of the two mRNAs. We also postulated that Nts mRNA expression would increase in parallel with the E(2)-induced LH surge and that the central (icv) administration of neurotensin would stimulate LH secretion and activation of GnRH neurons but found no evidence to support either of these hypotheses. Together, these findings suggest that, although neurotensin neurons in the AVPV are targets for regulation by E(2), neurotensin does not appear to play a direct role in generating the GnRH/LH surge in the mouse.


Subject(s)
Feedback, Physiological/physiology , Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/metabolism , Neurons/metabolism , Neurotensin/metabolism , Animals , Cell Communication/physiology , Estradiol/pharmacology , Estrogens/pharmacology , Feedback, Physiological/drug effects , Female , Genes, fos/physiology , Hypothalamus, Middle/cytology , Hypothalamus, Middle/physiology , Immunohistochemistry , Injections, Intraventricular , Kisspeptins , Median Eminence/cytology , Median Eminence/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurotensin/genetics , Ovariectomy , Preoptic Area/cytology , Preoptic Area/physiology , RNA, Messenger/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
Gen Comp Endocrinol ; 166(1): 66-71, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19686750

ABSTRACT

In songbirds, neurons that regulate learned song behavior undergo extensive seasonal plasticity in their number and size in relation to the bird's reproductive status. Seasonal plasticity of these brain regions is primarily regulated by changes in circulating concentrations of testosterone. Androgen receptors are present in all of the major song nuclei, but it is unknown whether levels of androgen receptor mRNA in the telencephalic song regions HVC, the robust nucleus of the arcopallium, and the lateral magnocellular nucleus of the anterior nidopallium change as a function of season in white-crowned sparrows. To determine whether seasonal changes in levels of androgen receptor mRNA are specific to the song control system, we also measured levels of androgen receptor mRNA in a limbic nucleus, the lateral division of the bed nucleus of the stria terminalis (the lateral division of the bed nucleus of the stria terminalis). We found that levels of androgen receptor mRNA were higher in HVC and the lateral division of the bed nucleus of the stria terminalis of birds in the breeding condition compared with the nonbreeding condition; however, we observed no seasonal differences in levels of androgen receptor mRNA in either the robust nucleus of the arcopallium or the lateral magnocellular nucleus of the anterior nidopallium. These results are consistent with previous observations that seasonal plasticity of the song nuclei results from testosterone acting directly on HVC, which then exerts transsynaptic trophic effects on its efferent targets. The seasonal change in the expression of androgen receptor in HVC may be one component of the cellular mechanisms underlying androgenic effects on seasonal plasticity of the song control nuclei.


Subject(s)
Limbic System/metabolism , Receptors, Androgen/genetics , Telencephalon/metabolism , Vocalization, Animal/physiology , Animals , Neural Pathways/physiology , Neuronal Plasticity/physiology , RNA, Messenger , Reproduction/physiology , Seasons , Sparrows
SELECTION OF CITATIONS
SEARCH DETAIL
...