Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
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
2.
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
3.
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
4.
J Neurosci ; 29(38): 11859-66, 2009 Sep 23.
Article in English | MEDLINE | ID: mdl-19776272

ABSTRACT

Kisspeptin is encoded by the Kiss1 gene, and kisspeptin signaling plays a critical role in reproduction. In rodents, kisspeptin neurons in the arcuate nucleus (Arc) provide tonic drive to gonadotropin-releasing hormone (GnRH) neurons, which in turn supports basal luteinizing hormone (LH) secretion. Our objectives were to determine whether preprodynorphin (Dyn) and neurokinin B (NKB) are coexpressed in Kiss1 neurons in the mouse and to evaluate its physiological significance. Using in situ hybridization, we found that Kiss1 neurons in the Arc of female mice not only express the Dyn and NKB genes but also the NKB receptor gene (NK3) and the Dyn receptor [the kappa opioid receptor (KOR)] gene. We also found that expression of the Dyn, NKB, KOR, and NK3 in the Arc are inhibited by estradiol, as has been established for Kiss1, and confirmed that Dyn and NKB inhibit LH secretion. Moreover, using Dyn and KOR knock-out mice, we found that long-term disruption of Dyn/KOR signaling compromises the rise of LH after ovariectomy. We propose a model whereby NKB and dynorphin act autosynaptically on kisspeptin neurons in the Arc to synchronize and shape the pulsatile secretion of kisspeptin and drive the release of GnRH from fibers in the median eminence.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Protein Precursors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Dynorphins/genetics , Estradiol/metabolism , Female , In Situ Hybridization , Kisspeptins , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Ovariectomy , Protein Precursors/genetics , RNA, Messenger/metabolism , Receptors, Neurokinin-3/agonists , Receptors, Neurokinin-3/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , Signal Transduction
5.
J Neurosci ; 29(29): 9390-5, 2009 Jul 22.
Article in English | MEDLINE | ID: mdl-19625529

ABSTRACT

Kisspeptin is a product of the Kiss1 gene and is expressed in the forebrain. Neurons that express Kiss1 play a crucial role in the regulation of pituitary luteinizing hormone secretion and reproduction. These neurons are the direct targets for the action of estradiol-17beta (E(2)), which acts via the estrogen receptor alpha isoform (ER alpha) to regulate Kiss1 expression. In the arcuate nucleus (Arc), where the dynorphin gene (Dyn) is expressed in Kiss1 neurons, E(2) inhibits the expression of Kiss1 mRNA. However, E(2) induces the expression of Kiss1 in the anteroventral periventricular nucleus (AVPV). The mechanism for differential regulation of Kiss1 in the Arc and AVPV by E(2) is unknown. ER alpha signals through multiple pathways, which can be categorized as either classical, involving the estrogen response element (ERE), or nonclassical, involving ERE-independent mechanisms. To elucidate the molecular basis for the action of E(2) on Kiss1 and Dyn expression, we studied the effects of E(2) on Kiss1 and Dyn mRNAs in the brains of mice bearing targeted alterations in the ER alpha signaling pathways. We found that stimulation of Kiss1 expression by E(2) in the AVPV and inhibition of Dyn in the Arc required an ERE-dependent pathway, whereas the inhibition of Kiss1 expression by E(2) in the Arc involved ERE-independent mechanisms. Thus, distinct ER alpha signaling pathways can differentially regulate the expression of identical genes across different brain regions, and E(2) can act within the same neuron through divergent ER alpha signaling pathways to regulate different neurotransmitter genes.


Subject(s)
Brain/drug effects , Dynorphins/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Proteins/metabolism , Animals , Anterior Thalamic Nuclei/drug effects , Anterior Thalamic Nuclei/metabolism , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Brain/metabolism , Dynorphins/genetics , Female , Gene Expression Regulation/drug effects , Gene Knock-In Techniques , Kisspeptins , Luteinizing Hormone/blood , Mice , Mice, Transgenic , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/metabolism , Neurons/drug effects , Neurons/metabolism , Proteins/genetics , RNA, Messenger/metabolism , Signal Transduction
6.
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
7.
Am J Physiol Endocrinol Metab ; 297(5): E1212-21, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19755669

ABSTRACT

In mammals, puberty onset typically occurs earlier in females than in males, but the explanation for sexual differentiation in the tempo of pubertal development is unknown. Puberty in both sexes is a brain-dependent phenomenon and involves alterations in the sensitivity of neuronal circuits to gonadal steroid feedback as well as gonadal hormone-independent changes in neuronal circuitry. Kisspeptin, encoded by the Kiss1 gene, plays an essential but ill-defined role in pubertal maturation. Neurokinin B (NKB) is coexpressed with Kiss1 in the arcuate nucleus (ARC) and is also important for puberty. We tested whether sex differences in the timing of pubertal development are attributable to sexual differentiation of gonadal hormone-independent mechanisms regulating hypothalamic Kiss1/NKB gene expression. We found that, in juvenile females, gonadotropin secretion and expression of Kiss1 and NKB in the ARC increased immediately following ovariectomy, suggesting that prepubertal females have negligible gonadal hormone-independent restraint on their reproductive axis. In contrast, in similarly aged juvenile males, no changes occurred in LH levels or Kiss1 or NKB expression following castration, suggesting that gonadal hormone-independent mechanisms restrain kisspeptin/NKB-dependent activation of the male reproductive axis before puberty. Notably, adult mice of both sexes showed comparable rapid increases in Kiss1/NKB expression and LH secretion following gonadectomy, signifying that sex differences in the regulation of ARC Kiss1/NKB neurons are manifest only during peripubertal development. Our findings demonstrate that the mechanisms controlling pubertal activation of reproduction in mice are different between the sexes and suggest that gonadal hormone-independent central restraint on pubertal timing involves Kiss1/NKB neurons in the ARC.


Subject(s)
Kisspeptins/biosynthesis , Kisspeptins/genetics , Neurokinin B/biosynthesis , Neurokinin B/genetics , Neurons/physiology , Sexual Maturation/genetics , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Data Interpretation, Statistical , Female , Gene Expression Regulation/genetics , Gonadal Steroid Hormones/physiology , In Situ Hybridization , Luteinizing Hormone/blood , Male , Mice , Mice, Inbred C57BL , Orchiectomy , Ovariectomy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sex Characteristics , Testosterone/blood
8.
Trends Neurosci ; 30(10): 504-11, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17904653

ABSTRACT

Neurons that produce gonadotropin-releasing hormone (GnRH) drive the reproductive axis, but the molecular and cellular mechanisms by which hormonal and environmental signals regulate GnRH secretion remain poorly understood. Kisspeptins are products of the Kiss1 gene, and the interaction of kisspeptin and its receptor GPR54 plays a crucial role in governing the onset of puberty and adult reproductive function. This review discusses the latest ideas about kisspeptin-GPR54 signaling in the neuroendocrine regulation of reproduction, with special emphasis on the role of Kiss1 and kisspeptin in the negative and positive feedback control of gonadotropin secretion by sex steroids, timing of puberty onset, sexual differentiation of the brain and photoperiodic regulation of seasonal reproduction.


Subject(s)
Neurosecretory Systems/metabolism , Receptors, G-Protein-Coupled/metabolism , Reproduction/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/physiology , Animals , Humans , Kisspeptins , Receptors, Kisspeptin-1
9.
Peptides ; 30(1): 4-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18644415

ABSTRACT

The cancer suppressor gene, KISS1, was initially described as having an important role in inhibiting cancer metastasis. Since then, KISS1 and its receptor, KISS1R, have been shown to play a key role in controlling the onset of puberty of reproductive physiology in the human and other species. Recent studies have also linked KISS1/kisspeptin/KISS1R to other processes, such as vasoconstriction, aging, adipocyte physiology, and perhaps as a molecular conduit linking metabolism and reproduction. This article highlights the history of KISS1/kisspeptin/KISS1R biology and proposes a consensus for nomenclature of the key molecules in this signaling pathway.


Subject(s)
Receptors, G-Protein-Coupled , Terminology as Topic , Tumor Suppressor Proteins , Animals , Humans , Hypothalamus/cytology , Hypothalamus/metabolism , Kisspeptins , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Reproduction/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
J Neurosci ; 27(44): 12088-95, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17978050

ABSTRACT

The Kiss1 gene codes for kisspeptin, which binds to GPR54, a G-protein-coupled receptor. Kisspeptin and GPR54 are expressed in discrete regions of the forebrain, and they have been implicated in the neuroendocrine regulation of reproduction. Kiss1-expressing neurons are thought to regulate the secretion of gonadotropin-releasing hormone (GnRH) and thus coordinate the estrous cycle in rodents; however, the precise role of kisspeptin-GPR54 signaling in the regulation of gonadotropin secretion is unknown. In this study, we used female mice with deletions in the GPR54 gene [GPR54 knock-outs (KOs)] to test the hypothesis that kisspeptin-GPR54 signaling provides the drive necessary for tonic GnRH/luteinizing hormone (LH) release. We predicted that tonic GnRH/LH secretion would be disrupted in GPR54 KOs and that such animals would be incapable of showing a compensatory rise in LH secretion after ovariectomy. As predicted, we found that GPR54 KO mice do not exhibit a postovariectomy rise in LH, suggesting that tonic GnRH secretion is disrupted in the absence of kisspeptin-GPR54 signaling. We also postulated that kisspeptin-GPR54 signaling is critical for the generation of the estradiol (E)-induced GnRH/LH surge and thus E should be incapable of inducing an LH surge in the absence of GPR54. However, we found that E induced Fos expression in GnRH neurons and produced a GnRH-dependent LH surge in GPR54 KOs. Thus, in mice, kisspeptin-GPR54 signaling is required for the tonic stimulation of GnRH/LH secretion but is not required for generating the E-induced GnRH/LH surge.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Proteins/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Animals , Behavior, Animal , Brain/cytology , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/genetics , Kisspeptins , Luteinizing Hormone/blood , Mice , Mice, Knockout , Neurons/metabolism , Oligopeptides/pharmacology , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Ovariectomy/methods , Proteins/genetics , Radioimmunoassay/methods , Receptors, G-Protein-Coupled/deficiency , Receptors, Kisspeptin-1
11.
J Neurosci ; 27(33): 8826-35, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699664

ABSTRACT

GPR54 is a G-protein-coupled receptor, which binds kisspeptins and is widely expressed throughout the brain. Kisspeptin-GPR54 signaling has been implicated in the regulation of pubertal and adulthood gonadotropin-releasing hormone (GnRH) secretion, and mutations or deletions of GPR54 cause hypogonadotropic hypogonadism in humans and mice. Other reproductive roles for kisspeptin-GPR54 signaling, including the regulation of developmental GnRH secretion or sexual behavior in adults, have not yet been explored. Using adult wild-type (WT) and GPR54 knock-out (KO) mice, we first tested whether kisspeptin-GPR54 signaling is necessary for male and female sexual behaviors. We found that hormone-replaced gonadectomized GPR54 KO males and females displayed appropriate gender-specific adult sexual behaviors. Next, we examined whether GPR54 signaling is required for proper display of olfactory-mediated partner preference behavior. Testosterone-treated WT males preferred stimulus females rather than males, whereas similarly treated WT females and GPR54 KO males showed no preference for either sex. Because olfactory preference is sexually dimorphic and organized during development by androgens, we assessed whether GPR54 signaling is essential for sexual differentiation of other sexually dimorphic traits. Interestingly, adult testosterone-treated GPR54 KO males displayed "female-like" numbers of tyrosine hydroxylase-immunoreactive and Kiss1 mRNA-containing neurons in the anteroventral periventricular nucleus and likewise possessed fewer motoneurons in the spino-bulbocavernosus nucleus than did WT males. Our findings indicate that kisspeptin-GPR54 signaling is not required for male or female copulatory behavior, provided there is appropriate adulthood hormone replacement. However, GPR54 is necessary for proper male-like development of several sexually dimorphic traits, likely by regulating GnRH-mediated androgen secretion during "critical windows" in perinatal development.


Subject(s)
Brain/metabolism , Receptors, G-Protein-Coupled/physiology , Sex Differentiation/physiology , Sexual Behavior, Animal/physiology , Signal Transduction/physiology , Analysis of Variance , Animals , Behavior, Animal/physiology , Brain/cytology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/metabolism , Humans , Kisspeptins , Luteinizing Hormone/blood , Male , Mice , Mice, Knockout , Neurons/classification , Neurons/drug effects , Neurons/metabolism , Proteins/genetics , Proteins/metabolism , Receptors, G-Protein-Coupled/deficiency , Receptors, Kisspeptin-1 , Sex Characteristics , Sex Differentiation/drug effects , Sex Differentiation/genetics , Sexual Behavior, Animal/drug effects , Signal Transduction/drug effects , Testosterone/pharmacology , Tumor Suppressor Proteins/pharmacology , Tyrosine 3-Monooxygenase/metabolism
12.
Mol Cell Biol ; 25(11): 4804-11, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15899880

ABSTRACT

Galanin is a neuropeptide implicated in the regulation of feeding, reproduction, cognition, nociception, and seizure susceptibility. There are three known galanin receptor (GALR) subtypes (GALR1, GALR2, and GALR3), which bind to galanin with different affinities and have their own unique distributions, signaling mechanisms, and putative functions in the brain and peripheral nervous system. To gain further insight into the possible physiological significance of GALR2, we created mutant mice that were deficient in GALR2 and compared their phenotype to that of wild-type (WT) littermate or age-matched controls, with respect to basic motor and sensory function, feeding behavior, reproduction, mood, learning and memory, and seizure susceptibility. Phenotypic analysis revealed that animals bearing a deletion of GALR2 did not differ significantly from their WT controls in any of the measured variables. We conclude that either GALR2 plays no role in these physiological functions or through redundancy or compensation these mutant animals can adapt to the congenital absence of GALR2. It is also conceivable that GALR2 plays only a subtle role in some of these functions and that the impact of its loss could not be detected by the analytical procedures used here.


Subject(s)
Phenotype , Receptor, Galanin, Type 2/physiology , Animals , Body Weight/genetics , Feeding Behavior , Female , Gene Deletion , Learning , Male , Memory , Mice , Mice, Knockout , Receptor, Galanin, Type 2/deficiency , Receptor, Galanin, Type 2/genetics , Reproduction/genetics , Seizures/genetics , Sex Factors
13.
J Neurosci ; 26(25): 6687-94, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16793876

ABSTRACT

Kisspeptins are neuropeptides encoded by the Kiss1 gene, which have been implicated in the neuroendocrine regulation of gonadotropin-releasing hormone (GnRH) secretion. The goal of this study was to test the hypothesis that activation of Kiss1 neurons in the anteroventral periventricular nucleus (AVPV) is linked to the induction of the preovulatory luteinizing hormone (LH) surge in the rat. First, we determined that levels of Kiss1 mRNA in the AVPV peaked during the evening of proestrus, whereas Kiss1 mRNA in the arcuate nucleus (Arc) was at its nadir. Second, we corroborated this observation by demonstrating that Kiss1 mRNA is increased in the AVPV at the time of an estrogen (E)- and progesterone-induced LH surge in ovariectomized animals, whereas in the Arc, the expression of Kiss1 mRNA was decreased. Third, we found that most Kiss1 neurons in the AVPV coexpress the immediate early gene Fos coincidently with the LH surge, but virtually none coexpressed Fos on diestrus. In contrast, Kiss1 neurons in the Arc were Fos negative at the time of the LH surge as well as on diestrus. Finally, we found that most Kiss1 neurons in the AVPV and Arc express estrogen receptor alpha mRNA, suggesting that E acts directly on these neurons. These results suggest that Kiss1 neurons in the AVPV play an active role in mediating the effects of E on the generation of the preovulatory GnRH/LH surge on proestrus.


Subject(s)
Gene Expression Regulation/physiology , Luteinizing Hormone/metabolism , Neurons/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Prosencephalon/cytology , Proteins/metabolism , Analysis of Variance , Animals , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrous Cycle/metabolism , Female , Gene Expression Regulation/drug effects , In Situ Hybridization/methods , Kisspeptins , Neurons/drug effects , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Ovariectomy/methods , Proteins/genetics , RNA, Messenger/metabolism , Radioimmunoassay/methods , Rats , Rats, Sprague-Dawley
14.
Neuroendocrinology ; 86(3): 175-82, 2007.
Article in English | MEDLINE | ID: mdl-17898535

ABSTRACT

Fertility is gated by nutrition and the availability of stored energy reserves, but the cellular and molecular mechanisms that link energy stores and reproduction are not well understood. Neuropeptides including galanin-like peptide (GALP), neuropeptide Y (NPY), products of the proopiomelanocortin (POMC; e.g., alpha-MSH and beta-endorphin), and kisspeptin are thought to be involved in this process for several reasons. First, the neurons that express these neuropeptides all reside in the hypothalamic arcuate nucleus, a critical site for the regulation of both metabolism and reproduction. Second, these neuropeptides are all targets for regulation by metabolic hormones, such as leptin and insulin. And third, these neuropeptides have either direct or indirect effects on feeding and metabolism, as well as on the secretion of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH). As the target for the action of metabolic hormones and sex steroids, these neuropeptides serve as molecular motifs integrating the control of metabolism and reproduction.


Subject(s)
Metabolic Networks and Pathways/physiology , Neuropeptides/physiology , Reproduction/physiology , Signal Transduction/physiology , Animals , Humans
15.
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
16.
J Neurosci ; 25(49): 11349-56, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16339030

ABSTRACT

We examined the role of kisspeptin and its receptor, the G-protein-coupled receptor GPR54, in governing the onset of puberty in the mouse. In the adult male and female mouse, kisspeptin (10-100 nM) evoked a remarkably potent, long-lasting depolarization of >90% of gonadotropin-releasing hormone (GnRH)-green fluorescent protein neurons in situ. In contrast, in juvenile [postnatal day 8 (P8) to P19] and prepubertal (P26-P33) male mice, kisspeptin activated only 27 and 44% of GnRH neurons, respectively. This developmental recruitment of GnRH neurons into a kisspeptin-responsive pool was paralleled by an increase in the ability of centrally administered kisspeptin to evoke luteinizing hormone secretion in vivo. To learn more about the mechanisms through which kisspeptin-GPR54 signaling at the GnRH neuron may change over postnatal development, we performed quantitative in situ hybridization for kisspeptin and GPR54 transcripts. Approximately 90% of GnRH neurons were found to express GPR54 mRNA in both juvenile and adult mice, without a detectable difference in the mRNA content between the age groups. In contrast, the expression of KiSS-1 mRNA increased dramatically across the transition from juvenile to adult life in the anteroventral periventricular nucleus (AVPV; p < 0.001). These results demonstrate that kisspeptin exerts a potent depolarizing effect on the excitability of almost all adult GnRH neurons and that the responsiveness of GnRH neurons to kisspeptin increases over postnatal development. Together, these observations suggest that activation of GnRH neurons by kisspeptin at puberty reflects a dual process involving an increase in kisspeptin input from the AVPV and a post-transcriptional change in GPR54 signaling within the GnRH neuron.


Subject(s)
Action Potentials/physiology , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Neurosecretory Systems/physiology , Proteins/physiology , Sexual Maturation/physiology , Age Factors , Animals , Animals, Newborn , Anterior Thalamic Nuclei/metabolism , Anterior Thalamic Nuclei/physiology , Female , Kisspeptins , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Mutant Strains , Neurons/physiology , Neurosecretory Systems/growth & development , Proteins/pharmacology , Tumor Suppressor Proteins
17.
Endocrinology ; 147(3): 1154-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16373418

ABSTRACT

The Kiss1 gene encodes a family of peptides called kisspeptins, which bind to the G protein-coupled receptor GPR54. Kisspeptin(s) and its receptor are expressed in the forebrain, and the discovery that mice and humans lacking a functional GPR54 fail to undergo puberty and exhibit hypogonadotropic hypogonadism implies that kisspeptin signaling plays an essential role in reproduction. Studies in several mammalian species have shown that kisspeptins stimulate the secretion of gonadotropins from the pituitary by stimulating the release of GnRH from the forebrain after the activation of GPR54, which is expressed by GnRH neurons. Kisspeptin is expressed abundantly in the arcuate nucleus (Arc) and the anteroventral periventricular nucleus (AVPV) of the forebrain. Both estradiol and testosterone regulate the expression of the Kiss1 gene in the Arc and AVPV; however, the response of the Kiss1 gene to these steroids is exactly opposite between these two nuclei. Estradiol and testosterone down-regulate Kiss1 mRNA in the Arc and up-regulate its expression in the AVPV. Thus, kisspeptin neurons in the Arc may participate in the negative feedback regulation of gonadotropin secretion, whereas kisspeptin neurons in the AVPV may contribute to generating the preovulatory gonadotropin surge in the female. Hypothalamic levels of Kiss1 and GPR54 mRNA increase dramatically at puberty, suggesting that kisspeptin signaling could mediate the neuroendocrine events that trigger the onset of puberty. Together, these observations demonstrate that kisspeptin-GPR54 signaling in the brain serves as an important conduit for controlling GnRH secretion in the developing and adult animal.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Proteins/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Down-Regulation , Estradiol/metabolism , Gene Expression Regulation , Humans , Kisspeptins , Mice , Midline Thalamic Nuclei/metabolism , Models, Biological , Pituitary Gland/metabolism , Prosencephalon/metabolism , Puberty , RNA, Messenger/metabolism , Rats , Receptors, G-Protein-Coupled , Receptors, Galanin/metabolism , Receptors, Kisspeptin-1 , Signal Transduction , Testosterone/metabolism , Tumor Suppressor Proteins , Up-Regulation
18.
Diabetes ; 54(8): 2471-6, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16046316

ABSTRACT

Galanin-like peptide (GALP) is expressed in the hypothalamic arcuate nucleus and is regulated by leptin and insulin. Centrally administered GALP stimulates gonadotropin secretion and sexual behavior in the rat. Type 1 diabetes is associated with reduced expression of GALP, as well as an overall decline in reproductive function. We postulated that tonic activity of GALP in the brain is required to sustain normal reproductive activity. To test this hypothesis, we examined whether central (intracerebroventricular) immunoblockade of GALP would reduce sexual behaviors and serum levels of luteinizing hormone (LH) in normal adult male rats. We found that GALP antibody reversibly reduced serum levels of LH and abolished male sexual behaviors (P < 0.05 and 0.001, respectively). Second, we tested whether intracerebroventricular GALP could restore normal plasma LH levels and sexual behavior in diabetic animals. We compared groups of diabetic rats that received intracerebroventricular GALP or vehicle and found that GALP increased serum levels of LH and sexual behavior. Third, we examined whether intracerebroventricular administration of affinity-purified GALP antibody could block the effect of insulin and leptin in reversing the effects of diabetes on LH and sexual behavior. We found that treatment of diabetic animals with insulin and leptin nearly normalized LH levels and sexual behaviors; however, this effect was attenuated by intracerebroventricular administration of GALP antibody (P < 0.05). These observations demonstrate that endogenous GALP provides trophic support to the neuroendocrine reproductive axis, including sexual behavior.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Galanin-Like Peptide/physiology , Reproduction/physiology , Animals , Antibodies/administration & dosage , Blood Glucose/analysis , Body Weight , Eating , Female , Galanin-Like Peptide/administration & dosage , Galanin-Like Peptide/immunology , Injections, Intraventricular , Insulin/administration & dosage , Leptin/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Sexual Behavior, Animal/physiology
19.
Trends Endocrinol Metab ; 16(6): 249-50, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16002301

ABSTRACT

The Kiss1 gene encodes a family of neuropeptides named kisspeptins, which bind to a (former orphan) G-protein-coupled receptor called GPR54. Recent investigations suggest that kisspeptins play a vital role in regulating the secretion of gonadotropin-releasing hormone (GnRH). New evidence confirms that kisspeptins act through GPR54 to stimulate GnRH secretion. Kisspeptins and GPR54 are crucial for pubertal maturation in the primate. Kiss1 mRNA is differentially regulated by sex steroids in distinct hypothalamic nuclei.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Proteins/metabolism , Puberty/metabolism , Receptors, Neuropeptide/metabolism , Humans , Kisspeptins , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Tumor Suppressor Proteins
20.
Endocrinology ; 146(9): 3686-92, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15919741

ABSTRACT

The Kiss1 gene encodes a family of neuropeptides called kisspeptins, which activate the receptor G protein-coupled receptor-54 and play a role in the neuroendocrine regulation of GnRH secretion. We examined whether estradiol (E2) regulates KiSS-1 in the forebrain of the female mouse by comparing KiSS-1 mRNA expression among groups of ovary-intact (diestrus), ovariectomized (OVX), and OVX plus E2-treated mice. In the arcuate nucleus (Arc), KiSS-1 expression increased after ovariectomy and decreased with E2 treatment. Conversely, in the anteroventral periventricular nucleus (AVPV), KiSS-1 expression was reduced after ovariectomy and increased with E2 treatment. To determine whether the effects of E2 on KiSS-1 are mediated through estrogen receptor (ER)alpha or ERbeta, we evaluated the effects of E2 in OVX mice that lacked functional ERalpha or ERbeta. In OVX mice that lacked functional ERalpha, KiSS-1 mRNA did not respond to E2 in either the Arc or AVPV, suggesting that ERalpha is essential for mediating the inhibitory and stimulatory effects of E2. In contrast, KiSS-1 mRNA in OVX mice that lacked functional ERbeta responded to E2 exactly as wild-type animals. Double-label in situ hybridization revealed that virtually all KiSS-1-expressing neurons in the Arc and AVPV coexpress ERalpha, suggesting that the effects of E2 are mediated directly through KiSS-1 neurons. We conclude that KiSS-1 neurons in the Arc, which are inhibited by E2, may play a role in the negative feedback regulation of GnRH secretion, whereas KiSS-1 neurons in the AVPV, which are stimulated by E2, may participate in the positive feedback regulation of GnRH secretion.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Paraventricular Hypothalamic Nucleus/physiology , Proteins/genetics , Animals , Body Weight , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Estrogens/blood , Estrogens/pharmacology , Female , Kisspeptins , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Ovariectomy , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL