1
|
Single-Cell Gene Profiling Reveals Social Status-Dependent Modulation of Nuclear Hormone Receptors in GnRH Neurons in a Male Cichlid Fish. Int J Mol Sci 2020; 21:ijms21082724. [PMID: 32326396 PMCID: PMC7215790 DOI: 10.3390/ijms21082724] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/17/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is essential for the initiation and maintenance of reproductive functions in vertebrates. To date, three distinct paralogue lineages, GnRH1, GnRH2, and GnRH3, have been identified with different functions and regulatory mechanisms. Among them, hypothalamic GnRH1 neurons are classically known as the hypophysiotropic form that is regulated by estrogen feedback. However, the mechanism of action underlying the estrogen-dependent regulation of GnRH1 has been debated, mainly due to the coexpression of low levels of estrogen receptor (ER) genes. In addition, the role of sex steroids in the modulation of GnRH2 and GnRH3 neurons has not been fully elucidated. Using single-cell real-time PCR, we revealed the expression of genes for estrogen, androgen, glucocorticoid, thyroid, and xenobiotic receptors in GnRH1, GnRH2, and GnRH3 neurons in the male Nile tilapia Oreochromis niloticus. We further quantified expression levels of estrogen receptor genes (ERα, ERβ, and ERγ) in three GnRH neuron types in male tilapia of two different social statuses (dominant and subordinate) at the single cell level. In dominant males, GnRH1 mRNA levels were positively proportional to ERγ mRNA levels, while in subordinate males, GnRH2 mRNA levels were positively proportional to ERβ mRNA levels. These results indicate that variations in the expression of nuclear receptors (and possibly steroid sensitivities) among individual GnRH cells may facilitate different physiological processes, such as the promotion of reproductive activities through GnRH1 neurons, and the inhibition of feeding and sexual behaviors through GnRH2 neurons.
Collapse
|
2
|
Struik D, Sanna F, Fattore L. The Modulating Role of Sex and Anabolic-Androgenic Steroid Hormones in Cannabinoid Sensitivity. Front Behav Neurosci 2018; 12:249. [PMID: 30416437 PMCID: PMC6212868 DOI: 10.3389/fnbeh.2018.00249] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/05/2018] [Indexed: 02/01/2023] Open
Abstract
Cannabis is the most commonly used illicit drug worldwide. Although its use is associated with multiple adverse health effects, including the risk of developing addiction, recreational and medical cannabis use is being increasing legalized. In addition, use of synthetic cannabinoid drugs is gaining considerable popularity and is associated with mass poisonings and occasional deaths. Delineating factors involved in cannabis use and addiction therefore becomes increasingly important. Similarly to other drugs of abuse, the prevalence of cannabis use and addiction differs remarkably between males and females, suggesting that sex plays a role in regulating cannabinoid sensitivity. Although it remains unclear how sex may affect the initiation and maintenance of cannabis use in humans, animal studies strongly suggest that endogenous sex hormones modulate cannabinoid sensitivity. In addition, synthetic anabolic-androgenic steroids alter substance use and further support the importance of sex steroids in controlling drug sensitivity. The recent discovery that pregnenolone, the precursor of all steroid hormones, controls cannabinoid receptor activation corroborates the link between steroid hormones and the endocannabinoid system. This article reviews the literature regarding the influence of endogenous and synthetic steroid hormones on the endocannabinoid system and cannabinoid action.
Collapse
Affiliation(s)
- Dicky Struik
- Department of Biomedical Sciences, University of Cagliari - Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, University of Cagliari - Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Liana Fattore
- CNR Institute of Neuroscience-Cagliari, National Research Council, Rome, Italy
| |
Collapse
|
3
|
Argiolas A, Melis MR. Neuropeptides and central control of sexual behaviour from the past to the present: a review. Prog Neurobiol 2013; 108:80-107. [PMID: 23851261 DOI: 10.1016/j.pneurobio.2013.06.006] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 06/28/2013] [Accepted: 06/29/2013] [Indexed: 12/23/2022]
Abstract
Of the numerous neuropeptides identified in the central nervous system, only a few are involved in the control of sexual behaviour. Among these, the most studied are oxytocin, adrenocorticotropin, α-melanocyte stimulating hormone and opioid peptides. While opioid peptides inhibit sexual performance, the others facilitate sexual behaviour in most of the species studied so far (rats, mice, monkeys and humans). However, evidence for a sexual role of gonadotropin-releasing hormone, corticotropin releasing factor, neuropeptide Y, galanin and galanin-like peptide, cholecystokinin, substance P, vasoactive intestinal peptide, vasopressin, angiotensin II, hypocretins/orexins and VGF-derived peptides are also available. Corticotropin releasing factor, neuropeptide Y, cholecystokinin, vasopressin and angiotensin II inhibit, while substance P, vasoactive intestinal peptide, hypocretins/orexins and some VGF-derived peptide facilitate sexual behaviour. Neuropeptides influence sexual behaviour by acting mainly in the hypothalamic nuclei (i.e., lateral hypothalamus, paraventricular nucleus, ventromedial nucleus, arcuate nucleus), in the medial preoptic area and in the spinal cord. However, it is often unclear whether neuropeptides influence the anticipatory phase (sexual arousal and/or motivation) or the consummatory phase (performance) of sexual behaviour, except in a few cases (e.g., opioid peptides and oxytocin). Unfortunately, scarce information has been added in the last 15 years on the neural mechanisms by which neuropeptides influence sexual behaviour, most studied neuropeptides apart. This may be due to a decreased interest of researchers on neuropeptides and sexual behaviour or on sexual behaviour in general. Such a decrease may be related to the discovery of orally effective, locally acting type V phosphodiesterase inhibitors for the therapy of erectile dysfunction.
Collapse
Affiliation(s)
- Antonio Argiolas
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| | | |
Collapse
|
4
|
Burel D, Li JH, Do-Rego JL, Wang AF, Luu-The V, Pelletier G, Tillet Y, Taragnat C, Kwon HB, Seong JY, Vaudry H. Gonadotropin-releasing hormone stimulates the biosynthesis of pregnenolone sulfate and dehydroepiandrosterone sulfate in the hypothalamus. Endocrinology 2013; 154:2114-28. [PMID: 23554453 DOI: 10.1210/en.2013-1095] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The sulfated neurosteroids pregnenolone sulfate (Δ(5)PS) and dehydroepiandrosterone sulfate (DHEAS) are known to play a role in the control of reproductive behavior. In the frog Pelophylax ridibundus, the enzyme hydroxysteroid sulfotransferase (HST), responsible for the biosynthesis of Δ(5)PS and DHEAS, is expressed in the magnocellular nucleus and the anterior preoptic area, two hypothalamic regions that are richly innervated by GnRH1-containing fibers. This observation suggests that GnRH1 may regulate the formation of sulfated neurosteroids to control sexual activity. Double labeling of frog brain slices with HST and GnRH1 antibodies revealed that GnRH1-immunoreactive fibers are located in close vicinity of HST-positive neurons. The cDNAs encoding 3 GnRH receptors (designated riGnRHR-1, -2, and -3) were cloned from the frog brain. RT-PCR analyses revealed that riGnRHR-1 is strongly expressed in the hypothalamus and the pituitary whereas riGnRHR-2 and -3 are primarily expressed in the brain. In situ hybridization histochemistry indicated that GnRHR-1 and GnRHR-3 mRNAs are particularly abundant in preoptic area and magnocellular nucleus whereas the concentration of GnRHR-2 mRNA in these 2 nuclei is much lower. Pulse-chase experiments using tritiated Δ(5)P and DHEA as steroid precursors, and 3'-phosphoadenosine 5'-phosphosulfate as a sulfonate moiety donor, showed that GnRH1 stimulates, in a dose-dependent manner, the biosynthesis of Δ(5)PS and DHEAS in frog diencephalic explants. Because Δ(5)PS and DHEAS, like GnRH, stimulate sexual activity, our data strongly suggest that some of the behavioral effects of GnRH could be mediated via the modulation of sulfated neurosteroid production.
Collapse
Affiliation(s)
- Delphine Burel
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institut National de la Sante´ et de la Recherche Me´ dicale U982, Research Institute for Biomedecine (IRIB), International Associated Laboratory Samuel de Champlain, University of Rouen, 76821 Mont-Saint Aignan, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Bosch MA, Tonsfeldt KJ, Rønnekleiv OK. mRNA expression of ion channels in GnRH neurons: subtype-specific regulation by 17β-estradiol. Mol Cell Endocrinol 2013; 367:85-97. [PMID: 23305677 PMCID: PMC3570747 DOI: 10.1016/j.mce.2012.12.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 11/22/2012] [Accepted: 12/11/2012] [Indexed: 12/31/2022]
Abstract
Burst firing of neurons optimizes neurotransmitter release. GnRH neurons exhibit burst firing activity and T-type calcium channels, which are vital for burst firing activity, are regulated by 17β-estradiol (E2) in GnRH neurons. To further elucidate ion channel expression and E2 regulation during positive and negative feedback on GnRH neurosecretion, we used single cell RT-PCR and real-time qPCR to quantify channel mRNA expression in GnRH neurons. GFP-GnRH neurons expressed numerous ion channels important for burst firing activity. E2-treatment sufficient to induce an LH surge increased mRNA expression of HCN1 channels, which underlie the pacemaker current, the calcium-permeable Ca(V)1.3, Ca(V)2.2, Ca(V)2.3 channels, and TRPC4 channels, which mediate the kisspeptin excitatory response. E2 also decreased mRNA expression of SK3 channels underlying the medium AHP current. Therefore, E2 exerts fundamental changes in ion channel expression in GnRH neurons, to prime them to respond to incoming stimuli with increased excitability at the time of the surge.
Collapse
Affiliation(s)
- Martha A. Bosch
- Department of Physiology and Pharmacology, Oregon Health and Sciences University, Portland, OR 97239 USA
| | - Karen J. Tonsfeldt
- Department of Physiology and Pharmacology, Oregon Health and Sciences University, Portland, OR 97239 USA
| | - Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health and Sciences University, Portland, OR 97239 USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, OR 97005 USA
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Sciences University, Portland, OR 97239 USA
| |
Collapse
|
6
|
Parhar I, Ogawa S, Kitahashi T. RFamide peptides as mediators in environmental control of GnRH neurons. Prog Neurobiol 2012; 98:176-96. [DOI: 10.1016/j.pneurobio.2012.05.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 11/25/2022]
|
7
|
Kokay IC, Petersen SL, Grattan DR. Identification of prolactin-sensitive GABA and kisspeptin neurons in regions of the rat hypothalamus involved in the control of fertility. Endocrinology 2011; 152:526-35. [PMID: 21177834 DOI: 10.1210/en.2010-0668] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
High levels of circulating prolactin are known to cause infertility, but the precise mechanisms by which prolactin influences the neuroendocrine axis are yet to be determined. We used dual-label in situ hybridization to investigate whether prolactin-receptor (PRLR) mRNA is expressed in GnRH neurons. In addition, because γ-aminobutyric acidergic and kisspeptin neurons in the rostral hypothalamus are known to regulate GnRH neurons and, hence, might mediate the actions of prolactin, we investigated whether these neurons coexpress PRLR mRNA. (35)S-labeled RNA probes to detect PRLR mRNA were hybridized together with digoxigenin-labeled probes to detect either GnRH, Gad1/Gad2, or Kiss1 mRNA in the rostral hypothalamus of ovariectomized (OVX), estradiol-treated rats. Additional sets of serial sections were cut through the arcuate nucleus of OVX rats, without estradiol replacement, to examine coexpression of PRLR mRNA in the arcuate population of kisspeptin neurons. PRLR mRNA was highly expressed throughout the rostral preoptic area, particularly in periventricular regions surrounding the third ventricle, and there was a high degree of colocalization of PRLR mRNA in both Gad1/Gad2 and Kiss1 mRNA-containing cells (86 and 85.5%, respectively). In contrast, only a small number of GnRH neurons (<5%) was found to coexpress PRLR mRNA. In the arcuate nucleus of OVX rats, the majority of Kiss1 mRNA-containing cells also coexpressed PRLR mRNA. These data are consistent with the hypothesis that, in addition to a direct action on a small subpopulation of GnRH neurons, prolactin actions on GnRH neurons are predominantly mediated indirectly, through known afferent pathways.
Collapse
Affiliation(s)
- Ilona C Kokay
- Centre for Neuroendocrinology and Department of Anatomy and Structural Biology, University of Otago, Dunedin 9016, New Zealand
| | | | | |
Collapse
|
8
|
Dalvi PS, Nazarians-Armavil A, Tung S, Belsham DD. Immortalized neurons for the study of hypothalamic function. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1030-52. [PMID: 21248304 DOI: 10.1152/ajpregu.00649.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The hypothalamus is a vital part of the central nervous system: it harbors control systems implicated in regulation of a wide range of homeostatic processes, including energy balance and reproduction. Structurally, the hypothalamus is a complex neuroendocrine tissue composed of a multitude of unique neuronal cell types that express a number of neuromodulators, including hormones, classical neurotransmitters, and specific neuropeptides that play a critical role in mediating hypothalamic function. However, neuropeptide and receptor gene expression, second messenger activation, and electrophysiological and secretory properties of these hypothalamic neurons are not yet fully defined, primarily because the heterogeneity and complex neuronal architecture of the neuroendocrine hypothalamus make such studies challenging to perform in vivo. To circumvent this problem, our research group recently generated embryonic- and adult-derived hypothalamic neuronal cell models by utilizing the novel molecular techniques of ciliary neurotrophic factor-induced neurogenesis and SV40 T antigen transfer to primary hypothalamic neuronal cell cultures. Significant research with these cell lines has demonstrated their value as a potential tool for use in molecular genetic analysis of hypothalamic neuronal function. Insights gained from hypothalamic immortalized cells used in conjunction with in vivo models will enhance our understanding of hypothalamic functions such as neurogenesis, neuronal plasticity, glucose sensing, energy homeostasis, circadian rhythms, and reproduction. This review discusses the generation and use of hypothalamic cell models to study mechanisms underlying the function of individual hypothalamic neurons and to gain a more complete understanding of the overall physiology of the hypothalamus.
Collapse
Affiliation(s)
- Prasad S Dalvi
- Dept. of Physiology, University of Toronto, 1 Kings College Circle, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
9
|
Franceschini I, Desroziers E, Caraty A, Duittoz A. The intimate relationship of gonadotropin-releasing hormone neurons with the polysialylated neural cell adhesion molecule revisited across development and adult plasticity. Eur J Neurosci 2010; 32:2031-41. [DOI: 10.1111/j.1460-9568.2010.07517.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
10
|
Krsmanovic LZ, Hu L, Leung PK, Feng H, Catt KJ. The hypothalamic GnRH pulse generator: multiple regulatory mechanisms. Trends Endocrinol Metab 2009; 20:402-8. [PMID: 19740674 PMCID: PMC2769988 DOI: 10.1016/j.tem.2009.05.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 05/04/2009] [Accepted: 05/05/2009] [Indexed: 12/30/2022]
Abstract
Pulsatile secretion of gonadotropin-releasing hormone (GnRH) release is an intrinsic property of hypothalamic GnRH neurons. Pulse generation has been attributed to multiple specific mechanisms, including spontaneous electrical activity of GnRH neurons, calcium and cAMP signaling, a GnRH receptor autocrine regulatory component, a GnRH concentration-dependent switch in GnRH receptor (GnRH-R) coupling to specific G proteins, the expression of G protein-coupled receptors (GPCRs) and steroid receptors, and homologous and heterologous interactions between cell membrane receptors expressed in GnRH neurons. The coexistence of multiple regulatory mechanisms for pulsatile GnRH secretion provides a high degree of redundancy in maintaining this crucial component of the mammalian reproductive process. These studies provide insights into the basic cellular and molecular mechanisms involved in GnRH neuronal function.
Collapse
Affiliation(s)
- Lazar Z Krsmanovic
- Section on Hormonal Regulation, Program on Developmental Endocrinology and Genetics, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
11
|
Sergeeva A, Jansen HT. Neuroanatomical plasticity in the gonadotropin-releasing hormone system of the ewe: seasonal variation in glutamatergic and gamma-aminobutyric acidergic afferents. J Comp Neurol 2009; 515:615-28. [PMID: 19496167 DOI: 10.1002/cne.22087] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Temperate zone animals time the onset of reproductive events to coincide with specific portions of the sidereal year. Although the neural mechanisms involved remain poorly understood, a marked annual variation in the brain's sensitivity to estradiol negative feedback is thought to mediate many of the changes in neuroendocrine hormone secretion, especially that of the gonadotropin-releasing hormone (GnRH) neurons, via neural afferents. The aim of the present study was to determine whether glutamatergic inputs to GnRH neurons in sheep vary seasonally and to expand our previous observations of seasonal changes in gamma-aminobutyric acid (GABA)-ergic inputs. Brains from adult sheep were collected during the breeding season (N = 8) or the nonbreeding season (anestrus; N = 7). Confocal microscopy and optical sectioning were used to quantify the density of labeled VGLUT2 and VGAT immunoreactivity onto GnRH neurons. The results reveal a significantly greater number of VGLUT2-ir inputs to GnRH dendrites during the breeding season vs. the nonbreeding season but no seasonal changes on GnRH cell somas. The number of VGAT-ir terminals onto GnRH dendrites was reduced in the breeding season compared with the nonbreeding season. GnRH neurons were also found to receive dual-phenotype (VGLUT + VGAT) inputs; these varied with season in a manner similar to VGAT inputs. Morphologically, the numbers of branches of proximal dendrites increased significantly in a subset of GnRH neurons located near the midline. Together these results reveal a dynamic seasonal reorganization of identified inputs onto GnRH neurons and lend additional support to the overall hypothesis that seasonal modulation of GnRH neurons involves glutamatergic and GABAergic neural plasticity.
Collapse
Affiliation(s)
- Anna Sergeeva
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, Pullman, WA 99164-6520, USA
| | | |
Collapse
|
12
|
Canosa LF, Stacey N, Peter RE. Changes in brain mRNA levels of gonadotropin-releasing hormone, pituitary adenylate cyclase activating polypeptide, and somatostatin during ovulatory luteinizing hormone and growth hormone surges in goldfish. Am J Physiol Regul Integr Comp Physiol 2008; 295:R1815-21. [DOI: 10.1152/ajpregu.00166.2008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In goldfish, circulating LH and growth hormone (GH) levels surge at the time of ovulation. In the present study, changes in gene expression of salmon gonadotropin-releasing hormone (sGnRH), chicken GnRH-II (cGnRH-II), somatostatin (SS) and pituitary adenylate cyclase activating polypeptide (PACAP) were analyzed during temperature- and spawning substrate-induced ovulation in goldfish. The results demonstrated that increases in PACAP gene expression during ovulation are best correlated with the GH secretion profile. These results suggest that PACAP, instead of GnRH, is involved in the control of GH secretion during ovulation. Increases of two of the SS transcripts during ovulation are interpreted as the activation of a negative feedback mechanism triggered by high GH levels. The results showed a differential regulation of sGnRH and cGnRH-II gene expression during ovulation, suggesting that sGnRH controls LH secretion, whereas cGnRH-II correlates best with spawning behavior. This conclusion is further supported by the finding that nonovulated fish induced to perform spawning behavior by prostaglandin F2α treatment increased cGnRH-II expression in both forebrain and midbrain, but decreased sGnRH expression in the forebrain.
Collapse
|
13
|
|
14
|
Renn SCP, Aubin-Horth N, Hofmann HA. Fish and chips: functional genomics of social plasticity in an African cichlid fish. J Exp Biol 2008; 211:3041-56. [PMID: 18775941 PMCID: PMC3728697 DOI: 10.1242/jeb.018242] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Behavior and physiology are regulated by both environment and social context. A central goal in the study of the social control of behavior is to determine the underlying physiological, cellular and molecular mechanisms in the brain. The African cichlid fish Astatotilapia burtoni has long been used as a model system to study how social interactions regulate neural and behavioral plasticity. In this species, males are either socially dominant and reproductively active or subordinate and reproductively suppressed. This phenotypic difference is reversible. Using an integrative approach that combines quantitative behavioral measurements, functional genomics and bioinformatic analyses, we examine neural gene expression in dominant and subordinate males as well as in brooding females. We confirm the role of numerous candidate genes that are part of neuroendocrine pathways and show that specific co-regulated gene sets (modules), as well as specific functional gene ontology categories, are significantly associated with either dominance or reproductive state. Finally, even though the dominant and subordinate phenotypes are robustly defined, we find a surprisingly high degree of individual variation in the transcript levels of the very genes that are differentially regulated between these phenotypes. The results of the present study demonstrate the molecular complexity in the brain underlying social behavior, identify novel targets for future studies, validate many candidate genes and exploit individual variation in order to gain biological insights.
Collapse
Affiliation(s)
- Susan C P Renn
- Harvard University, Bauer Center for Genomics Research, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | | | | |
Collapse
|
15
|
Hormonal regulation of clonal, immortalized hypothalamic neurons expressing neuropeptides involved in reproduction and feeding. Mol Neurobiol 2008; 35:182-94. [PMID: 17917107 DOI: 10.1007/s12035-007-0010-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Revised: 10/03/2006] [Accepted: 11/09/2006] [Indexed: 12/27/2022]
Abstract
The hypothalamus has been particularly difficult to study at the molecular level because of the inherent cellular heterogeneity and complexity of neuronal circuits within. We have generated a large number of immortalized, clonal cell lines through retroviral gene transfer of the oncogene SV40 T-Ag into primary murine hypothalamic neuronal cell cultures. A number of these neuronal cell lines express neuropeptides linked to the control of feeding behavior and reproduction, including neuropeptide Y (NPY) and neurotensin (NT). We review recent studies on the direct regulation of NPY gene expression by estrogen, and the leptin-mediated control of signal transduction pathways and NT transcription. These studies provide new insights into the direct control of neuropeptide synthesis by hormones and nutrients at a mechanistic level in the individual neuron, not yet possible in the whole brain. Using these novel cell models, we expect to contribute substantially to the understanding of how individual neuronal cell types control overall endocrine function, especially with regard to two of the most well-known roles of distinct peptidergic neurons; these being the control of reproduction and energy homeostasis.
Collapse
|
16
|
Shibata M, Friedman RL, Ramaswamy S, Plant TM. Evidence that down regulation of hypothalamic KiSS-1 expression is involved in the negative feedback action of testosterone to regulate luteinising hormone secretion in the adult male rhesus monkey (Macaca mulatta). J Neuroendocrinol 2007; 19:432-8. [PMID: 17504437 DOI: 10.1111/j.1365-2826.2007.01549.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In the male monkey, luteinising hormone (LH) secretion is regulated by a negative feedback action of testicular testosterone that is exerted indirectly at the hypothalamic level to decelerate pulsatile gonadotrophin-releasing hormone release (GnRH). The purpose of the present experiment was to investigate whether the kisspeptin-G protein-coupled receptor 54 (GPR54) signalling pathway is involved in mediating the action of testosterone to suppress GnRH release in the monkey, as has been indicated by studies of nonprimates. To this end, 12 castrated adult male rhesus monkeys were implanted with either testosterone containing or empty Silastic capsules. Testosterone treatment produced a square wave increment in circulating testosterone levels within the physiologic range. After suppression of LH and follicle-stimulating hormone secretion was established at 5-6 weeks of testosterone exposure, the animals were killed and expression of the genes encoding for kisspeptin, GPR54 and GnRH determined in the mediobasal hypothalamus and preoptic area of both treated and control animals using RNase protection assays. The suppression in pituitary gonadotrophin secretion was associated with a reduction in kisspeptin mRNA levels in the mediobasal hypothalamus, but not the preoptic area. GPR54 mRNA levels, on the other hand, were not influenced by testosterone treatment. These results are consistent with those previously reported for the rodent, and suggest that the neurobiology of the negative feedback action of testicular testosterone on LH secretion in the monkey, a representative higher primate, may be mediated by kisspeptinergic neurones upstream to the GnRH network.
Collapse
Affiliation(s)
- M Shibata
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | |
Collapse
|
17
|
Cottone E, Campantico E, Guastalla A, Aramu S, Polzonetti-Magni AM, Franzoni M. Are the cannabinoids involved in bony fish reproduction? Ann N Y Acad Sci 2006; 1040:273-6. [PMID: 15891041 DOI: 10.1196/annals.1327.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Following the discovery of two CB1 genes in the fish Fugu rubripes, investigations on the phylogeny of endocannabinoids have indicated that this system is highly conserved. Our study demonstrated that CB1 receptors are expressed in the CNS and gonads of two teleosts, Carassius auratus and Pelvicachromis pulcher, and they show a high percentage of sequence identity with Fugu rubripes CB(1A) and Danio rerio CB1. By means of immunohistochemistry for CB1, sGnRH, and TH, we found a codistribution of these signaling molecules in the basal telencephalon/preoptic area, which are key centers for gonadotropic regulation. We therefore suggest that endocannabinoids are possibly involved in modulating fish reproduction at both the central and peripheral levels.
Collapse
Affiliation(s)
- Erika Cottone
- Dipartimento di Biologia Animale e dell'Uomo, Università degli Studi di Torino, Italy
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Comprehensive studies have provided a clear understanding of the effects of gonadal steroids on the secretion of gonadotropin releasing hormone (GnRH), but some inconsistent results exist with regard to effects on synthesis. It is clear that regulation of both synthesis and the secretion of GnRH are effected by neurotransmitter systems in the brain. Thus, steroid regulation of GnRH synthesis and secretion can be direct, but the predominant effects are transmitted through steroid-responsive neuronal systems in various parts of the brain. There is also emerging evidence of direct effects on GnRH cells. Overriding effects on synthesis and secretion of GnRH can be observed during aging, in undernutrition and under stressful situations; these involve various neuronal systems, which may have serial or parallel effects on GnRH cells. The effect of aging is accompanied by changes in GnRH synthesis, but comprehensive studies of synthesis during undernutrition and stress are less well documented. Altered GnRH and gonadotropin secretion that occurs in seasonal breeding animals and during the pubertal transition is not generally accompanied by changes in GnRH synthesis. Secretion of GnRH from the brain is a reflection of the inherent function of GnRH cells and the inputs that integrate all of the central regulatory elements. Ultimately, the pattern of secretion dictates the reproductive status of the organism. In order to fully understand the central mechanisms that control reproduction, more extensive studies are required on the neuronal circuitry that provides input to GnRH cells.
Collapse
Affiliation(s)
- Iain J Clarke
- Prince Henry's Institute of Medical Research, P.O. Box 5152, Clayton 3168, Australia.
| | | |
Collapse
|
19
|
Ugrumov MV, Sapronova AY, Melnikova VI, Proshlyakova EV, Adamskaya EI, Lavrentieva AV, Nasirova DI, Babichev VN. Brain is an important source of GnRH in general circulation in the rat during prenatal and early postnatal ontogenesis. Comp Biochem Physiol A Mol Integr Physiol 2005; 141:271-9. [PMID: 16009589 DOI: 10.1016/j.cbpb.2005.04.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 04/07/2005] [Accepted: 04/07/2005] [Indexed: 10/25/2022]
Abstract
This study was aimed to test our hypothesis that, in contrast to adult rats, in fetuses and neonates, a large amount of the brain-derived GnRH is delivered to the general circulation. The GnRH concentration and content were estimated in general circulation and in the forebrain in rats on the 18th embryonic day (E18), E21, 3rd postnatal day (P3) and P30-36. Moreover, the GnRH concentration was measured in general circulation on E21 following microsurgical lesion on E18 of the forebrain containing most GnRH neurons. The concentration and content of GnRH in plasma on E18, E21 and P3 enormously exceeded those on P30-36. Reverse was true for the ontogenetic dynamics of the GnRH concentration in the forebrain. The lesion of the forebrain resulted in a drop of the GnRH concentration in plasma. The above data strongly suggest that the forebrain is the principal source of GnRH in general circulation in fetal and neonatal rats. Thus, the brain-derived GnRH is delivered to the general circulation in fetal and neonatal rats in amounts likely sufficient to influence the potential peripheral targets.
Collapse
Affiliation(s)
- Michael V Ugrumov
- Laboratory of Hormonal Regulations, Institute of Developmental Biology RAS, Moscow, Russia.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Ottinger MA, Abdelnabi M, Li Q, Chen K, Thompson N, Harada N, Viglietti-Panzica C, Panzica GC. The Japanese quail: a model for studying reproductive aging of hypothalamic systems. Exp Gerontol 2005; 39:1679-93. [PMID: 15582284 DOI: 10.1016/j.exger.2004.06.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Accepted: 06/14/2004] [Indexed: 10/26/2022]
Abstract
During aging, the decline of neuroendocrine, endocrine, and behavioral components of reproduction ultimately leads to reproductive failure. These studies considered both neuroendocrine and behavioral aspects of reproductive aging in Japanese quail, using chronological age and reproductive status to separate animals into experimental groups. In Study I, age-related changes in the gonadotropin releasing hormone (GnRH-I) system were investigated and a sharp decrease was observed in GnRH-I concentration in the median eminence of aging animals of both sexes, whereas preoptic-lateral septal region GnRH-I concentrations declined only in aging males. Immunohistochemistry confirmed these findings since aging females retained, whereas males lost GnRH-I cells. Functional changes were assessed by in vitro incubation of parasaggittal hypothalamic slices collected from young and old inactive males and females. Results showed reduced baseline GnRH-I release and diminished response to norepinephrine (NE). Deteriorating fertility also correlated with decreased male sexual behavior and loss of aromatase immunoreactive (AROM-ir) neurons in the medial, but not lateral preoptic nucleus (POA). Sexual behavior and AROM-ir were restored with exogenous testosterone, which was associated with increased cell size in the medial POA. Comparison of cell size and number of AROM-ir cells showed that aged sexually active males had fewer, larger AROM-ir cells when compared to young males, suggesting neuroplasticity of specific neural systems and a critical role of estradiol in maintaining reproductive function.
Collapse
Affiliation(s)
- Mary Ann Ottinger
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Giampietro C, Luzzati F, Gambarotta G, Giacobini P, Boda E, Fasolo A, Perroteau I. Stathmin expression modulates migratory properties of GN-11 neurons in vitro. Endocrinology 2005; 146:1825-34. [PMID: 15625246 DOI: 10.1210/en.2004-0972] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Expression of stathmin, a microtubule-associated cytoplasmic protein, prominently localized in neuroproliferative zones and neuronal migration pathways in brain, was investigated in the GnRH neuroendocrine system in vivo and the function was analyzed using an in vitro approach. Here we present novel data demonstrating that GnRH migrating neurons in nasal regions and basal forebrain areas of mouse embryos express stathmin protein. In addition, this expression pattern is dependent on location, as GnRH neurons reaching the hypothalamus are stathmin negative. Immortalized GN-11 cells, which retain many characteristics of migrating GnRH neurons, strongly express stathmin mRNA and protein. The role of stathmin in GnRH migratory properties was evaluated using GN-11 cell line. We up-regulated [stathmin-transfected clones (STMN)+] and down-regulated (STMN-) the expression of stathmin in GN-11 cells, and we investigated changes in cell morphology and motility in vitro. Cells overexpressing stathmin assume a spindle-shaped morphology and their proliferation, as well as their motility, is higher with respect to parental cells. Furthermore, they do not aggregate and express low levels of cadherins compared with control cells. STMN- GN-11 cells are endowed with multipolar processes, and they show a decreased motility and express high levels of cadherin protein. Our findings suggest that stathmin plays a permissive role in GnRH cell motility, possibly via modulation of cadherins expression.
Collapse
Affiliation(s)
- Costanza Giampietro
- Department of Human and Animal Biology, University of Torino, Via Accademia Albertina 13, 10123 Torino, Italy
| | | | | | | | | | | | | |
Collapse
|
22
|
Russell JM, Murphree E, Janik J, Callahan P. Effect of steroids and nitric oxide on pituitary hormone release in ovariectomized, peripubertal rats. Reproduction 2005; 129:497-504. [PMID: 15798025 DOI: 10.1530/rep.1.00472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The purpose of this study was to determine the effects of the duration of steroid depletion on the steroid-induced luteinizing hormone and prolactin surges in ovariectomized, peripubertal female rats. Additionally, the role of nitric oxide (NO) in mediating the surge responses was determined. Peripubertal, 6-week-old, female Sprague-Dawley rats were ovariectomized. One or three weeks later, animals were injected with 17β-estradiol (50 μg, sc) followed 48 h later by progesterone (2.5 mg, sc). Effects of NO were examined by administeringl-arginine (300 mg/kg, ip). The response of ovariectomized, adult females to steroid treatment was also determined.One and three weeks after ovariectomy, steroid replacement produced an LH and prolactin surge in peripubertal animals. However, both the magnitude and duration of the LH surge was greater 3 weeks after ovariectomy. Whilel-arginine significantly enhanced the magnitude of the LH surge 1 week after ovariectomy, by 3 weeksl-arginine caused a decrease in the duration, but not the magnitude of the surge. In contrast,l-arginine did not affect either the magnitude or duration of the prolactin surge one week after ovariectomy, but diminished the magnitude after 3 weeks of steroid depletion. In adults, steroids induced significant increases in both LH and prolactin. These results demonstrate that sensitivity to NO stimulation of LH, but not prolactin secretion, is modulated by the duration of gonadal steroid hormone depletion. The differences in the responsiveness of LH and prolactin to steroid-induced stimulation in peripubertal animals demonstrate that these hormones are regulated by NO through different mechanisms.
Collapse
Affiliation(s)
- Jill M Russell
- Center for Neuroscience, Department of Zoology, Miami University, Oxford, Ohio 45056, USA
| | | | | | | |
Collapse
|
23
|
An BS, Choi JH, Choi KC, Leung PCK. Differential role of progesterone receptor isoforms in the transcriptional regulation of human gonadotropin-releasing hormone I (GnRH I) receptor, GnRH I, and GnRH II. J Clin Endocrinol Metab 2005; 90:1106-13. [PMID: 15562029 DOI: 10.1210/jc.2004-0318] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Hypothalamic GnRH is a decapeptide that plays a pivotal role in mammalian reproduction by stimulating the synthesis and secretion of gonadotropins via binding to the GnRH receptor on the pituitary gonadotropins. It is hypothesized that sex steroids may regulate GnRH I (a classical form of GnRH), GnRH II (a second form of GnRH), and GnRH I receptor (GnRHRI) at the transcriptional level in target tissues. Thus, in the present study a role for progesterone (P4) in the regulation of GnRH I, GnRH II, and GnRHRI was investigated using a human neuronal medulloblastoma cell line (TE671) as an in vitro model. The cells were transfected with human GnRHRI promoter-luciferase constructs, and promoter activities were analyzed after P4 treatment by luciferase and beta-galactosidase assay. The mRNA levels of GnRH I and GnRH II were analyzed by RT-PCR. Treatment of TE671 cells with P4 resulted in a decrease in GnRHRI promoter activity compared with the control level in a dose- and time-dependent manner. Cotreatment of these cells with RU486, an antagonist of P4, reversed P4-induced inhibition of GnRHRI promoter activity, suggesting that the P4 effect is mediated by P4 receptor (PR). In the cells transfected with a full-length of PR A- or PR B-expressing vector, overexpression of PR A increased the sensitivity toward P4 in an inhibition of GnRHRI promoter, whereas PR B increased transcriptional activity of GnRHRI promoter in the presence of P4. However, PR B itself did not act as a transcriptional activator of GnRHRI promoter. Because TE671 cells have been recently demonstrated to express and synthesize two forms of GnRHs, we also investigated the regulation of GnRH mRNAs by P4. In the present study, P4 increased GnRH I mRNA levels in a time- and dose-dependent manner. This stimulatory effect of P4 in the regulation of GnRH I mRNAs was significantly attenuated by RU486, whereas no significant difference in the expression level of GnRH II was observed with P4 or RU496. Interestingly, although the expression level of PR B was low compared with that of PR A, P4 action on the GnRH I gene was mediated by PR B. In conclusion, these results indicate that P4 is a potent regulator of GnRHRI at the transcriptional level as well as GnRH I mRNA. This distinct effect of P4 on the GnRH system may be derived from different pathways through PR A or PR B.
Collapse
Affiliation(s)
- Beum-Soo An
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada V6H 3V5
| | | | | | | |
Collapse
|
24
|
Sandoval-Guzmán T, Rance NE. Central injection of senktide, an NK3 receptor agonist, or neuropeptide Y inhibits LH secretion and induces different patterns of Fos expression in the rat hypothalamus. Brain Res 2005; 1026:307-12. [PMID: 15488494 DOI: 10.1016/j.brainres.2004.08.026] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2004] [Indexed: 11/22/2022]
Abstract
Arcuate neurokinin B (NKB) neurons express estrogen receptor-alpha and are strongly modulated by gonadal steroids. Although numerous studies suggest that NKB neurons participate in the reproductive axis, there is no information on the regulation of luteinizing hormone (LH) secretion by NKB or its receptor, NK3. In the present study, we determined if central injection of senktide, a selective NK3 receptor agonist, would alter serum LH in ovariectomized, estrogen-primed rats. The effects of senktide were compared to neuropeptide Y (NPY), a well-characterized modulator of LH secretion. Saline, senktide, or NPY was injected into the lateral ventricle of unanesthetized rats and serial blood samples were collected for LH radioimmunoassay. The rats were sacrificed 90 min after injection and the brains were removed and processed for Fos immunocytochemistry. A significant inhibition of serum LH was observed from 30 to 90 min after injection of senktide relative to saline controls. In the senktide-injected rats, the inhibition of serum LH was accompanied by increased Fos expression in the medial preoptic area and arcuate nucleus--two reproductive control centers. Senktide also induced Fos in the paraventricular nuclei (PVN) and supraoptic nuclei (SON). Injection of NPY also inhibited serum LH but increased Fos expression only in the PVN and SON. This study provides the first demonstration of alterations in LH secretion by an NK3 receptor agonist. These data, combined with the induction of Fos in medial preoptic and arcuate neurons, strongly support the hypothesis that NKB neurons play a role in the regulation of gonadotropin secretion.
Collapse
|
25
|
Belsham DD, Lovejoy DA. Gonadotropin‐Releasing Hormone: Gene Evolution, Expression, and Regulation. VITAMINS & HORMONES 2005; 71:59-94. [PMID: 16112265 DOI: 10.1016/s0083-6729(05)71003-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The gonadotropin-releasing hormone (GnRH) gene is a superb example of the diverse regulation that is required to maintain the function of an evolutionarily conserved and fundamental gene. Because reproductive capacity is critical to the survival of the species, physiological homeostasis dictates optimal conditions for reproductive success, and any perturbation from this balance may affect GnRH expression. These disturbances may include alterations in signals dictated by stress, nutritional imbalance, body weight, and neurological problems; therefore, changes in other neuroendocrine systems may directly influence the hypothalamic-pituitary-gonadal axis through direct regulation of GnRH. Thus, to maintain optimal reproductive capacity, the regulation of the GnRH gene is tightly constrained by a number of diverse signaling pathways and neuromodulators. In this review, we summarize what is currently known of GnRH gene structure, the location and function of the two isoforms of the GnRH gene, some of the many hormones and neuromodulators found to affect GnRH expression, and the molecular mechanisms responsible for the regulation of the GnRH gene. We also discuss the latest models used to study the transcriptional regulation of the GnRH gene, from cell models to evolving in vivo technologies. Although we have come a long way in the last two decades toward uncovering the intricacies behind the control of the GnRH neuron, there remain vast distances to cover before direct therapeutic manipulation of the GnRH gene to control reproductive competence is possible.
Collapse
Affiliation(s)
- Denise D Belsham
- Department of Physiology, University of Toronto, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | | |
Collapse
|
26
|
Heideman PD. Top-down Approaches to the Study of Natural Variation in Complex Physiological Pathways Using the White-footed Mouse (Peromyscus leucopus) as a Model. ILAR J 2004; 45:4-13. [PMID: 14752203 DOI: 10.1093/ilar.45.1.4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Variation in complex physiological pathways has important effects on human function and medical treatment. Complex pathways involve cells at multiple locations, which serve different functions regulated by many genes and include complex neuroendocrine pathways that regulate physiological function. One of two competing hypotheses regarding the effects of selection on complex pathways predicts that variability should be common within complex pathways. If this hypothesis is correct, then we should expect wide variation in neuroendocrine function to be typical within natural populations. To test this hypothesis, a complex neuroendocrine pathway that regulates photoperiod-dependent changes in fertility in a natural population of white-footed mice (Peromyscus leucopus) was used to test for natural genetic variability in multiple components of the pathway. After testing only six elements in the photoperiod pathway in P. leucopus, genetic variation in the following four of these elements was evident: the circadian clock, melatonin receptor abundance or affinity, sensitivity of the reproductive axis to steroid negative feedback, and gonadotropin-releasing hormone neuronal activity. If this result can be extended to humans, the prediction would be that significant variation at multiple loci in complex neuroendocrine pathways is common among humans, and that variation would exist even in human populations from a common genetic background. This finding could only be drawn from an "exotic" animal model derived from a natural source population, confirming the continuing importance of nontraditional models alongside the standard laboratory species.
Collapse
|
27
|
Thanky NR, Slater R, Herbison AE. Sex differences in estrogen-dependent transcription of gonadotropin-releasing hormone (GnRH) gene revealed in GnRH transgenic mice. Endocrinology 2003; 144:3351-8. [PMID: 12865313 DOI: 10.1210/en.2001-211342] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The mechanisms through which gonadal steroids exert feedback actions on the activity of the GnRH neurons are not understood. Using a series of GnRH-LacZ transgenic mice we have examined the manner in which gonadal steroids suppress GnRH mRNA expression in male and female mice. The long-term gonadectomy of 5.5-GNZ-3.5 transgenic mice resulted in significant increases in cellular GnRH mRNA expression (P < 0.05) and plasma LH concentrations (P < 0.01) in both sexes. However, cellular levels of LacZ mRNA and beta-galactosidase, which provide an index of GnRH gene transcription, were only elevated in males after gonadectomy. This sexually differentiated response was also observed in mice gonadectomized for 2 wk. Estrogen replacement in gonadectomized males returned transgene expression to intact levels. Experiments in transgenic mice with 3' and 5' deleted GnRH-LacZ constructs revealed that the suppressive influence of estrogen on LacZ transcription in the male required a critical element located between -5.2 and -1.7 kb of the GnRH promoter. These studies show that the suppression of GnRH mRNA expression by estrogen in the male involves a decrease in GnRH gene transcription that is dependent on a distal GnRH promoter element. The same mechanism does not exist in females, indicating that gonadal steroids suppress GnRH mRNA levels in a sexually dimorphic manner.
Collapse
Affiliation(s)
- Niren R Thanky
- Laboratory of Neuroendocrinology, The Babraham Institute, Cambridge CB2 4AT, United Kingdom
| | | | | |
Collapse
|
28
|
Peter RE, Prasada Rao PD, Baby SM, Illing N, Millar RP. Differential brain distribution of gonadotropin-releasing hormone receptors in the goldfish. Gen Comp Endocrinol 2003; 132:399-408. [PMID: 12849963 DOI: 10.1016/s0016-6480(03)00084-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present study describes the differential distributions in the brain of the two goldfish gonadotropin-releasing hormone (GnRH) receptors, using both immunohistochemistry and in situ hybridization approaches. The goldfish GnRH GfA and GfB receptors are variant forms of the same receptor subtype, although with distinct differences in ligand binding characteristics, and differential distributions in the pituitary and body tissues [Proc. Natl. Acad. Sci. USA 96 (1999) 2526]. The goldfish GnRH GfA receptor was found to be widespread throughout the brain, with neurons showing immunoreactivity in the olfactory bulbs, telencephalon, preoptic region, ventro-basal hypothalamus, thalamus, midbrain, motor neurons of the fifth, seventh, and tenth cranial nerves, reticular formation, cerebellum, and motor zone of the vagal lobes. The tracts in the posterior commissure, optic tectum, and motor zone of the vagal lobes also demonstrated immunoreactivity. While the brain was not systematically surveyed for in situ hybridization, hybridization was found in similar locations in the telencephalon, preoptic region, ventro-basal hypothalamus, cerebellum, and optic tectum. Hybridization was additionally found in the medial hypothalamus. The goldfish GnRH GfB receptor was found to have a more restricted distribution in the brain, with neurons showing immunoreactivity in the telencephalon, preoptic region, and ventro-basal hypothalamus. In situ hybridization demonstrated a somewhat wider distribution of expression of the receptor, with hybridization occurring in the preoptic region, ventro-basal and medial hypothalamus, as well as in the thalamus, epithalamus, and optic tectum. The widespread distribution of GnRH GfA receptor, and in particular its localization in the midbrain tegmentum in the region of the GnRH-II neurons, suggests that this receptor may be involved in the behavioral actions of GnRH peptides in the goldfish.
Collapse
Affiliation(s)
- Richard E Peter
- Department of Biological Sciences, University of Alberta, Alta., T6G 2E9 Edmonton, Canada.
| | | | | | | | | |
Collapse
|
29
|
Gore AC. Gonadotropin-releasing hormone (GnRH) neurons: gene expression and neuroanatomical studies. PROGRESS IN BRAIN RESEARCH 2003; 141:193-208. [PMID: 12508571 DOI: 10.1016/s0079-6123(02)41094-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Affiliation(s)
- Andrea C Gore
- Kastor Neurobiology of Aging Laboratories, Fishberg Research Center for Neurobiology, and Brookdale Department of Geriatrics and Development, Box 1639, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029, USA.
| |
Collapse
|
30
|
McCarthy MM, Amateau SK, Mong JA. Steroid modulation of astrocytes in the neonatal brain: implications for adult reproductive function. Biol Reprod 2002; 67:691-8. [PMID: 12193373 DOI: 10.1095/biolreprod.102.003251] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
There is a growing appreciation for the importance of astrocytes, a type of nonneuronal glial cell, to overall brain functioning. The ability of astrocytes to respond to gonadal steroid hormones with changes in morphology has been well documented in the adult brain. It is also apparent that astrocytes of the developing brain are permanently differentiated by the neonatal hormonal milieu, in particular by estradiol, resulting in sexually dimorphic cell morphology, synaptic patterning, and density in males and females. The mechanisms of hormonally mediated astrocyte differentiation are likely to be region specific. In the arcuate nucleus of the hypothalamus, neuron-to-astrocyte signaling appears to play a critical role in estradiol-induced astrocyte differentiation during the first few days of life. Gamma aminobutyric acid (GABA) is an amino acid neurotransmitter that is synthesized and released exclusively by neurons. The levels of GABA are increased in the arcuate nucleus of neonatal males versus females. Preventing the increase in males or mimicking GABA action in females modulates astrocytes accordingly. Speculation about and evidence in support of the functional significance of this dimorphism to adult reproductive functioning is the topic of this review.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Physiology, University of Maryland, Baltimore, Maryland 21201-1559, USA.
| | | | | |
Collapse
|
31
|
Melcangi RC, Martini L, Galbiati M. Growth factors and steroid hormones: a complex interplay in the hypothalamic control of reproductive functions. Prog Neurobiol 2002; 67:421-49. [PMID: 12385863 DOI: 10.1016/s0301-0082(02)00060-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The mechanisms through which LHRH-secreting neurons are controlled still represent a crucial and debated field of research in the neuroendocrine control of reproduction. In the present review, we have specifically considered two potential signals reaching these hypothalamic neurons: steroid hormones and growth factors. Examples of the relevant physiological role of the interactions between these two families of biologically acting molecules have been provided. In many cases, these interactions occur at the level of hypothalamic astrocytes, which are presently accepted as functional partners of the LHRH-secreting neurons. On the basis of the observations here summarized, we have formulated the hypothesis that a functional co-operation of steroid hormones and growth factors occurring in the hypothalamic astrocytic compartment represents a key factor in the neuroendocrine control of reproductive functions.
Collapse
Affiliation(s)
- Roberto C Melcangi
- Department of Endocrinology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, 20133, Milan, Italy.
| | | | | |
Collapse
|
32
|
Gore AC. Organochlorine pesticides directly regulate gonadotropin-releasing hormone gene expression and biosynthesis in the GT1-7 hypothalamic cell line. Mol Cell Endocrinol 2002; 192:157-70. [PMID: 12088877 DOI: 10.1016/s0303-7207(02)00010-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Environmental toxicants profoundly affect growth and developmental processes. In the present study, we hypothesized that hypothalamic gonadotropin-releasing hormone (GnRH) neurons, which regulate the reproductive axis, are targets of environmental endocrine disrupting chemicals. Two organochlorine pesticides (methoxychlor and chlorpyrifos) were tested for their effects on GnRH gene expression and biosynthesis in the immortalized hypothalamic GT1-7 cells, which synthesize and secrete GnRH. GT1-7 cells were treated with methoxychlor or chlorpyrifos for 24 h in dose-response experiments, and GnRH gene expression and peptide levels were quantified. In order to examine whether these pesticides affect GnRH biosynthesis through the estrogen receptor (ER), in other experiments their effects were compared to those of estrogen, or they were co-administered with the ER antagonist, ICI 182,780 (ICI). Both methoxychlor and chlorpyrifos had significant effects on GnRH gene transcription and GnRH mRNA levels. These effects were not consistently blocked by ICI, nor did the effects of these pesticides consistently mimic those of estrogen, suggesting a mechanism independent of the ER. Chlorpyrifos and methoxychlor slightly stimulated peptide levels, and this effect was blocked by ICI, suggesting that the ER may mediate effects of pesticides on GnRH release. These results indicate that chlorpyrifos and methoxychlor alter GnRH biosynthesis in this hypothalamic cell line in vitro, suggesting that they may have endocrine disrupting effects on GnRH neurons in vivo.
Collapse
Affiliation(s)
- Andrea C Gore
- Kastor Neurobiology of Aging Laboratories, Fishberg Research Center for Neurobiology, and Brookdale Department of Geriatrics and Adult Development, Mount Sinai School of Medicine, Box 1639, New York, NY 10029, USA.
| |
Collapse
|
33
|
Episodic bursting activity and response to excitatory amino acids in acutely dissociated gonadotropin-releasing hormone neurons genetically targeted with green fluorescent protein. J Neurosci 2002. [PMID: 11896170 DOI: 10.1523/jneurosci.22-06-02313.2002] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The gonadotropin-releasing hormone (GnRH) system, considered to be the final common pathway for the control of reproduction, has been difficult to study because of a lack of distinguishing characteristics and the scattered distribution of neurons. The development of a transgenic mouse in which the GnRH promoter drives expression of enhanced green fluorescent protein (EGFP) has provided the opportunity to perform electrophysiological studies of GnRH neurons. In this study, neurons were dissociated from brain slices prepared from prepubertal female GnRH-EGFP mice. Both current- and voltage-clamp recordings were obtained from acutely dissociated GnRH neurons identified on the basis of EGFP expression. Most isolated GnRH-EGFP neurons fired spontaneous action potentials (recorded in cell-attached or whole-cell mode) that typically consisted of brief bursts (2-20 Hz) separated by 1-10 sec. At more negative resting potentials, GnRH-EGFP neurons exhibited oscillations in membrane potential, which could lead to bursting episodes lasting from seconds to minutes. These bursting episodes were often separated by minutes of inactivity. Rapid application of glutamate or NMDA increased firing activity in all neurons and usually generated small inward currents (<15 pA), although larger currents were evoked in the remaining neurons. Both AMPA and NMDA receptors mediated the glutamate-evoked inward currents. These results suggest that isolated GnRH-EGFP neurons from juvenile mice can generate episodes of repetitive burst discharges that may underlie the pulsatile secretion of GnRH, and glutamatergic inputs may contribute to the activation of endogenous bursts.
Collapse
|
34
|
Grattan DR. The actions of prolactin in the brain during pregnancy and lactation. PROGRESS IN BRAIN RESEARCH 2001; 133:153-71. [PMID: 11589128 DOI: 10.1016/s0079-6123(01)33012-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The vital role played by prolactin during pregnancy and lactation is emphasized by the physiological adaptations that occur in the mother to maintain a prolonged state of hyperprolactinemia. In many species the placenta provides a source of lactogenic hormones in the circulation, ensuring the continued presence of a hormone capable of activating the prolactin receptor throughout pregnancy. In addition, the tuberoinfundibular dopamine neurons, which normally maintain a tonic inhibitory influence over prolactin secretion, show a reduced ability to respond to prolactin during late pregnancy and lactation, allowing high levels of prolactin to be maintained unopposed by a regulatory feedback mechanisms. There is clear evidence that systemic prolactin gains access to the cerebrospinal fluid, from where it can diffuse to numerous brain regions. Prolactin receptors are expressed in several hypothalamic nuclei, including the medial preoptic and arcuate nuclei, and we have observed marked increases in expression of prolactin receptors in these nuclei during lactation. Moreover, a number of hypothalamic nuclei, including the paraventricular, supraoptic and ventromedial nuclei, in which prolactin receptors were not detected in diestrous rats, were found to express significant amounts of prolactin receptor during lactation. These observations have important implications for the variety of documented actions of prolactin on the brain. Prolactin has been reported to influence numerous brain functions, including maternal behavior, feeding and appetite, oxytocin secretion, and ACTH secretion in response to stress. In light of the high circulating levels of prolactin during pregnancy and lactation and the increased expression of prolactin receptors in the hypothalamus, many of these effects of prolactin may be enhanced or exaggerated during lactation. Hence, prolactin may be a key player in the coordination of neuroendocrine and behavioral adaptations of the maternal brain.
Collapse
Affiliation(s)
- D R Grattan
- Department of Anatomy and Structural Biology, School of Medical Sciences, Neuroscience Research Centre, University of Otago, P.O. Box 913, Dunedin, New Zealand.
| |
Collapse
|
35
|
Grattan DR, Pi XJ, Andrews ZB, Augustine RA, Kokay IC, Summerfield MR, Todd B, Bunn SJ. Prolactin receptors in the brain during pregnancy and lactation: implications for behavior. Horm Behav 2001; 40:115-24. [PMID: 11534971 DOI: 10.1006/hbeh.2001.1698] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Numerous studies have documented prolactin regulation of a variety of brain functions, including maternal behavior, regulation of oxytocin neurons, regulation of feeding and appetite, suppression of ACTH secretion in response to stress, and suppression of fertility. We have observed marked changes in expression of prolactin receptors in specific hypothalamic nuclei during pregnancy and lactation. This has important implications for neuronal functions regulated by prolactin. In light of the high circulating levels of prolactin during pregnancy and lactation and the increased expression of prolactin receptors in the hypothalamus, many of these functions may be enhanced or exaggerated in the maternal brain. The adaptations of the maternal brain allow the female to exhibit the appropriate behavior to feed and nurture her offspring, to adjust to the nutritional and metabolic demands of milk production, and to maintain appropriate hormone secretion to allow milk synthesis, secretion, and ejection. This review aims to summarize the evidence that prolactin plays a key role in regulating hypothalamic function during lactation and to discuss the hypothesis that the overall role of prolactin is to organize and coordinate this wide range of behavioral and neuroendocrine adaptations during pregnancy and lactation.
Collapse
Affiliation(s)
- D R Grattan
- Department of Anatomy and Structural Biology, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Spergel DJ, Krüth U, Shimshek DR, Sprengel R, Seeburg PH. Using reporter genes to label selected neuronal populations in transgenic mice for gene promoter, anatomical, and physiological studies. Prog Neurobiol 2001; 63:673-86. [PMID: 11165000 DOI: 10.1016/s0301-0082(00)00038-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This review summarizes recent work on the use of reporter genes to label selected neuronal populations in transgenic mice, with particular emphasis on gonadotropin-releasing hormone (GnRH) neurons. Reporter genes discussed are the lacZ, green fluorescent protein (GFP), luc, and bla genes, which encode the reporter proteins beta-galactosidase, GFP, luciferase, and beta-lactamase, respectively. Targeted transgenic expression of these reporter proteins is obtained by fusing the corresponding reporter gene, with or without a subcellular localization signal, to a cell type- or brain region-specific gene promoter. Mice carrying GnRH promoter-driven reporter genes have proven useful for revealing the promoter elements required for cell type-specific expression of GnRH, the full anatomical profile of the GnRH neuronal network, and its electrophysiological activity, suggesting that similar approaches will assist in elucidating the properties of other neuronal populations as well.
Collapse
Affiliation(s)
- D J Spergel
- Department of Molecular Neuroscience, Max-Planck-Institute for Medical Research, Jahnstrasse 29, 69120, Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
37
|
Pi X, Voogt JL. Mechanisms for suckling-induced changes in expression of prolactin receptor in the hypothalamus of the lactating rat. Brain Res 2001; 891:197-205. [PMID: 11164823 DOI: 10.1016/s0006-8993(00)03212-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The present study aimed to investigate whether increased expression of prolactin receptor (PRL-R) during lactation is caused by suckling-induced hyperprolactinemia or the suckling stimulus itself. Three groups (n=7) of mid-lactating rats were used. Each rat received 3 days of s.c. injection of vehicle or drug before sacrifice on lactation day 10. Rats in the control group received vehicle only and were suckled by pups. The second group received bromocriptine to suppress PRL levels and were suckled by pups. The third group of rats received haloperidol (high PRL) and were deprived of pups. Plasma PRL levels were measured. Animals were perfused with 2% paraformaldehyde for immunofluorescent study. Results showed that PRL-R immunoreactivity in the ventrolateral preoptic, ventromedial preoptic, and ventromedial hypothalamic nuclei was significantly increased in the bromocriptine-treated group compared to the control group, indicating PRL-R expression in these areas may be inhibited by hyperprolactinemia in the presence of the suckling stimulus. The PRL-R in the lateroanterior, ventrolateral and paraventricular nuclei was significantly decreased in the haloperidol-treated group compared to the control group, suggesting that the PRL-R in these areas is most likely regulated by the suckling stimulus itself. The PRL-R in the arcuate nucleus was significantly increased in bromocriptine-treated rats and decreased in haloperidol-treated rats, suggesting that the PRL-R in this nucleus is regulated by mechanisms related to both the stimulus of suckling itself and suckling-induced hyperprolactinemia. These results support the hypothesis that expression of PRL-R in discrete hypothalamic nuclei is differentially regulated by either PRL and/or suckling.
Collapse
Affiliation(s)
- X Pi
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160-7401, USA
| | | |
Collapse
|
38
|
Gore AC, Oung T, Yung S, Flagg RA, Woller MJ. Neuroendocrine mechanisms for reproductive senescence in the female rat: gonadotropin-releasing hormone neurons. Endocrine 2000; 13:315-23. [PMID: 11216643 DOI: 10.1385/endo:13:3:315] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2000] [Revised: 06/06/2000] [Accepted: 06/23/2000] [Indexed: 11/11/2022]
Abstract
Reproductive aging in female rats is characterized by profound alterations in the neuroendocrine axis. The preovulatory luteinizing hormone (LH) surge is attenuated, and preovulatory expression of the immediate early gene fos in gonadotropin-releasing hormone (GnRH) neurons is substantially reduced in middle-aged compared with young rats. We tested the hypothesis that alterations in GnRH gene expression may be correlated with the attenuation of the LH surge and may be a possible mechanism involved in neuroendocrine senescent changes. Sprague-Dawley rats ages 4 to 5 mo (young), 12-14 mo (middle-aged), or 25 to 26 mo (old) were killed at 10:00 AM or 3:00 PM on proestrus, the day of the LH surge, or diestrus I in cycling rats, and on persistent estrus or persistent diestrus in acyclic rats. RNase protection assays of GnRH mRNA and GnRH primary transcript were performed. GnRH mRNA levels increased significantly with age, whereas GnRH primary transcript levels, an index of GnRH gene transcription, decreased in old compared to young and middle-aged rats. This latter result suggests that an age-related change in GnRH mRNA levels occurs independently of a change in gene transcription, indicating a potential posttranscriptional mechanism. On proestrus, GnRH mRNA levels increased significantly from 10:00 AM to 3:00 PM in young rats. This was in contrast to proestrous middle-aged rats, in which this afternoon increase in GnRH mRNA levels was not observed. Thus, the normal afternoon increase in GnRH mRNA levels on proestrus is disrupted by middle age and may represent a substrate for the attenuation of the preovulatory GnRH/LH surge that occurs in rats of this age, prior to reproductive failure.
Collapse
Affiliation(s)
- A C Gore
- Kastor Neurobiology of Aging Laboratories, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | | | |
Collapse
|
39
|
Yoo MJ, Searles RV, He JR, Shen WB, Grattan DR, Selmanoff M. Castration rapidly decreases hypothalamic gamma-aminobutyric acidergic neuronal activity in both male and female rats. Brain Res 2000; 878:1-10. [PMID: 10996131 DOI: 10.1016/s0006-8993(00)02600-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The postcastration LH response is greater and somewhat more rapid in male than female rats. We have previously demonstrated that hypothalamic gamma-aminobutyric acid (GABA)ergic neuronal activity decreases following gonadectomy in male rats. To investigate whether these same hypothalamic GABA neurons decrease their activity postcastration in female rats, and whether more rapid and or greater postcastration decreases occur in male rats, we determined the timing and magnitude of the postcastration decreases in GABA turnover which are associated with the sexually dimorphic postcastration LH response. Adult male and 4-day cycling female rats were castrated between 0800 and 1000 h (females ovariectomized on diestrus day 1). Serum LH levels increased significantly by 12 h postcastration in both males and females with the magnitude of the increases being 6.2-fold in males and 2.8-fold in females. GABA turnover was determined in 16 microdissected brain structures by the GABA transaminase inhibition method at 0 h (sham-operated controls), 6 h, 12 h and 1, 2, 4 and 6 days postcastration. In male rats, in the diagonal band of Broca at the level of the organum vasculosum of the lamina terminalis [DBB(ovlt)], the rate of GABA turnover decreased significantly already by 6 h postcastration compared with the 0 h controls, and remained suppressed through 6 days. This rapid down regulation of DBB(ovlt) GABAergic neurons also occurred in female rats, however, the duration of the decrease was not as prolonged as in male rats. Similar changes occurred in the tuberoinfundibular GABAergic (TIGA) neurons projecting to the median eminence in both males and females. Down regulation of these GABAergic neurons precedes or is coincident with increased postcastration LH secretion in both sexes, and the duration of the decreases is consistent with the less robust postcastration LH response in female rats. In addition, the rate of GABA turnover decreased after castration in the interstitial (bed) nucleus of the stria terminalis, ventral aspect (INSTv), the medial preoptic nucleus, dorsomedial aspect (MPNdm) and the ventromedial nucleus, ventrolateral aspect (VMNvl) in male rats, and in the INSTv and VMNvl of female rats, while there was no effect of castration in other hypothalamic regions or control structures. The result in the female VMNvl is consistent with reports that GABA facilitates lordosis behavior in this hypothalamic structure. These findings are consistent with the hypothesis that discrete hypothalamic populations of sex steroid-sensitive GABAergic neurons mediate the postcastration LH responses in both male and female rats, and may underlie other sexually dimorphic adult phenotypes such as sex behavior.
Collapse
Affiliation(s)
- M J Yoo
- Center for Studies in Reproduction, Department of Physiology, University of Maryland, School of Medicine, Baltimore, MD 21201-1559, USA
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
GnRH is the key neuropeptide controlling reproductive function in all vertebrate species. Two different neuroendocrine mechanisms have evolved among female mammals to regulate the mediobasal hypothalamic (MBH) release of GnRH leading to the preovulatory secretion of LH by the anterior pituitary gland. In females of spontaneously ovulating species, including rats, mice, guinea pigs, sheep, monkeys, and women, ovarian steroids secreted by maturing ovarian follicles induce a pulsatile pattern of GnRH release in the median eminence that, in turn, stimulates a preovulatory LH surge. In females of induced ovulating species, including rabbits, ferrets, cats, and camels, the preovulatory release of GnRH, and the resultant preovulatory LH surge, is induced by the receipt of genital somatosensory stimuli during mating. Induced ovulators generally do not show "spontaneous" steroid-induced LH surges during their reproductive cycles, suggesting that the positive feedback actions of steroid hormones on GnRH release are reduced or absent in these species. By contrast, mating-induced preovulatory surges occasionally occur in some spontaneously ovulating species. Most research in the field of GnRH neurobiology has been performed using spontaneous ovulators including rat, guinea pig, sheep, and rhesus monkey. This review summarizes the literature concerning the neuroendocrine mechanisms controlling GnRH biosynthesis and release in females of several induced ovulating species, and whenever possible it contrasts the results with those obtained for spontaneously ovulating species. It also considers the adaptive, evolutionary benefits and disadvantages of each type of ovulatory control mechanism. In females of induced ovulating species estradiol acts in the brain to induce aspects of proceptive and receptive sexual behavior. The primary mechanism involved in the preovulatory release of GnRH among induced ovulators involves the activation of midbrain and brainstem noradrenergic neurons in response to genital-somatosensory signals generated by receipt of an intromission from a male during mating. These noradrenergic neurons project to the MBH and, when activated, promote the release of GnRH from nerve terminals in the median eminence. In contrast to spontaneous ovulators, there is little evidence that endogenous opioid peptides normally inhibit MBH GnRH release among induced ovulators. Instead, the neural signals that induce a preovulatory LH surge in these species seem to be primarily excitatory. A complete understanding of the neuroendocrine control of ovulation will only be achieved in the future by comparative studies of several animal model systems in which mating-induced as well as spontaneous, hormonally stimulated activation of GnRH neurons drives the preovulatory LH surge.
Collapse
Affiliation(s)
- J Bakker
- Department of Biology, Boston University, 5 Cummington Street, Boston, Massachusetts, 02215, USA
| | | |
Collapse
|
41
|
Parhar IS, Soga T, Sakuma Y. Thyroid hormone and estrogen regulate brain region-specific messenger ribonucleic acids encoding three gonadotropin-releasing hormone genes in sexually immature male fish, Oreochromis niloticus. Endocrinology 2000; 141:1618-26. [PMID: 10803569 DOI: 10.1210/endo.141.5.7460] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The present study was undertaken to determine whether T3, estrogen, and 11-ketotestosterone could alter a specific population of GnRH-containing neurons, as indicated by a change in messenger RNA (mRNA) levels in sexually immature male tilapia, Oreochromis niloticus. Two weeks after castration, fish were assigned to four treatment groups. One group served as the control (sesame oil); a single ip injection of (T3; 5 microg/g), estradiol benzoate (EB; 5 microg/g), or 11-ketotestosterone (KT; 5 microg/g) was administered to the remaining three groups. Twenty-four hours after the injection, brains were collected and processed for in situ hybridization histochemistry using 35S-labeled 30-mer antisense oligonucleotide probes complementary to the GnRH-coding region of chicken II, salmon, and seabream GnRH. Computerized image analysis was performed to quantify mRNA concentrations, neuronal numbers, and neuronal size of the terminal nerve-nucleus olfactoretinalis, preoptic, and midbrain GnRH neurons. KT had no effect on any of the above neuronal parameters examined for salmon or seabream GnRH. Neither T3, EB, nor KT was effective to induce changes in midbrain chicken GnRH II mRNA concentrations, neuronal numbers, and neuronal size, indicating that an as yet unknown regulatory mechanism may operate midbrain GnRH neurons. T3 specifically suppressed the concentration of terminal nerve salmon GnRH mRNA, and EB significantly increased preoptic seabream GnRH neuronal numbers. These results are consistent with the hypothesis that thyroid hormone, by suppressing terminal nerve GnRH expression, promotes inhibition of sexual maturation. Furthermore, the failure of KT, a nonaromatizable androgen, to influence preoptic GnRH neurons emphasizes that an estrogenic pathway, at the onset of sexual maturation, is responsible for the recruitment of additional preoptic GnRH neurons that are fundamental to reproduction and behavior.
Collapse
Affiliation(s)
- I S Parhar
- Department of Physiology, Nippon Medical School, Tokyo, Japan.
| | | | | |
Collapse
|
42
|
El Majdoubi M, Ramaswamy S, Sahu A, Plant TM. Effects of orchidectomy on levels of the mRNAs encoding gonadotropin-releasing hormone and other hypothalamic peptides in the adult male rhesus monkey (Macaca mulatta). J Neuroendocrinol 2000; 12:167-76. [PMID: 10718912 DOI: 10.1046/j.1365-2826.2000.00433.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The testicular regulation of luteinizing hormone (LH) secretion in the adult rhesus monkey is mediated by an indirect action of testosterone to decelerate pulsatile gonadotrophin releasing hormone (GnRH) release. Whether this negative feedback action of testosterone involves regulation of GnRH gene expression is unknown. Therefore, the effect of bilateral orchidectomy on hypothalamic levels of the mRNA encoding this hypophysiotropic factor was examined. The feedback action of testosterone is generally considered to be mediated through non-GnRH cells, and the present experiment provided the opportunity to also examine testicular influences on mRNAs encoding putative hypothalamic factors implicated in the testicular regulation of LH secretion. Adult male rhesus monkeys were orchidectomized (n=5) or sham-orchidectomized (n=5) and killed 6 weeks later, after a castration-induced hypersecretion of LH was established. Separate preoptic and mediobasal hypothalamus containing areas were collected, and levels of GnRH mRNA, as well as those of mRNAs encoding pro-opiomelanocortin (POMC), the gamma-aminobutyric acid (GABA) synthesizing enzymes (glutamic acid decarboxylase 65 and 67; GAD65 and GAD67, respectively), neuropeptide Y, galanin and transforming growth factor (TGF)alpha, were quantified using RNase protection assay. Values were expressed in terms of optical density relative to that of cyclophilin mRNA levels. Bilateral orchidectomy produced a significant increase in GnRH mRNA levels that was restricted to the mediobasal hypothalamus and that was associated with a significant decrease in POMC, GAD65 and GAD67 mRNA levels in this region of the hypothalamus. In contrast, neuropeptide Y, galanin and TGFalpha mRNA levels were not affected by castration. These results indicate that, in the monkey, the deceleration of pulsatile GnRH release that is imposed by the testis, and presumably mediated by testosterone, is associated with a concomitant down regulation of GnRH gene expression in the mediobasal hypothalamus. They also support the notion that this hypothalamic feedback action may be mediated by POMC-and GABA-producing neurones in the mediobasal hypothalamus.
Collapse
Affiliation(s)
- M El Majdoubi
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | |
Collapse
|
43
|
Volkoff H, Peter RE. Actions of two forms of gonadotropin releasing hormone and a GnRH antagonist on spawning behavior of the goldfish Carassius auratus. Gen Comp Endocrinol 1999; 116:347-55. [PMID: 10603273 DOI: 10.1006/gcen.1999.7377] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The central effects of two native forms of gonadotropin-releasing hormone (GnRH), salmon GnRH (sGnRH) and chicken GnRH-II (cGnRH-II), and a GnRH antagonist, ¿Ac-delta 3-Pro(1), 4FD-Phe(2), D-Trp(3, 6)mGnRH (analog E), on the spawning behavior of sexually recrudescent female goldfish were investigated. The effects of analog E were also observed in mature males. Female spawning behavior was induced by intramuscular injection of females with prostaglandin F(2alpha) and placing them in the presence of mature males. Behavioral responses were quantified by recording the numbers of spawning acts performed by each pair of fish for 2 h following brain intracerebroventricular (icv) injection of different dosages of peptide or saline as control. For males, the time spent courting the female was recorded. Each pair of fish was pretested to determine their level of spawning behavior, for comparison to spawning behavior following icv treatment. Icv injection of analog E caused a significant decrease in the number of spawning acts performed by females, suggesting a role of endogenous GnRH in modulating female spawning behavior. icv injection of 0.5 ng/g of sGnRH or cGnRH-II significantly stimulated female spawning behavior, whereas doses of 1 ng/g and higher resulted in an almost complete inhibition of spawning, reflecting a down-regulation as a result of the excessive dosages. Analog E suppressed the actions of exogenous sGnRH and cGnRH-II on spawning behavior, as both the sGnRH- and cGnRH-II-induced increases in the number of spawning acts were inhibited by concomitant treatment with analog E. Analog E-injected males showed no alteration in courtship behavior. These results indicate that GnRH peptides play a major role in the control of female reproductive behavior in goldfish, but have little or no role in the control of male behavior.
Collapse
Affiliation(s)
- H Volkoff
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | | |
Collapse
|
44
|
Abstract
Many neuropeptides are involved in the control of sexual behaviour at the central level. Among these, the most studied are adrenocorticotropin, alpha-melanocyte stimulating hormone, oxytocin and opioid peptides. This attempt to review old and new neuropharmacological, biochemical and psychobiological studies in this field, shows that all these neuropeptides apparently facilitate sexual behaviour, except for opioid peptides, which inhibit sexual performance, in most of the species studied so far (rats, mice, monkeys and humans). However, gonadotropin-releasing hormone, corticotropin releasing factor, neuropeptide Y, galanin, cholecystokinin, substance P and vasoactive intestinal peptide may be also involved in the control of sexual behaviour. Apparently, corticotropin releasing factor, neuropeptide Y and cholecystokinin inhibit, while substance P and vasoactive intestinal peptide facilitate, sexual behaviour. In contrast, gonadotropin-releasing hormone has been reported to exert a facilitative, inhibitory or no effect at all on sexual behaviour. Galanin was also shown either to facilitate or inhibit sexual behaviour. The above-mentioned putative role of the neuropeptides in sexual behaviour derives mainly from studies done in rats. In these studies, neuropeptides, their antisera or drugs that act as agonists or antagonists of neuropeptide receptors, were tested for their effect on sexual behaviour after systemic, intracerebroventricular, or intracerebral administration. The latter were infused into brain areas relevant for sexual behaviour, such as the medial preoptic area, and the ventromedial and paraventricular nuclei of the hypothalamus. The above studies show that little information is available on the mechanisms by which neuropeptides influence sexual behaviour. Also unclear is whether the above neuropeptides influence the anticipatory phase (sexual arousal and/or motivation) or the consummatory phase (performance) of sexual behaviour, except for opioid peptides. New information about the role of neuropeptides may come from the application of molecular biology and genetic manipulation techniques to the study of sexual behaviour. Of these, FOS protein determination, antisense oligonucleotides aimed at the neutralisation of neuropeptide and/or neuropeptide receptor mRNAs in specific brain areas, and gene ablation seem the most promising. Although still in the early stages, it is likely that these methodologies will provide new insights into the role of neuropeptides in the control of sexual behaviour.
Collapse
Affiliation(s)
- A Argiolas
- Bernard B. Brodie Department of Neuroscience and Centre for Neuropharmacology, National Research Council, Cagliari, Italy.
| |
Collapse
|
45
|
Roy D, Angelini NL, Belsham DD. Estrogen directly respresses gonadotropin-releasing hormone (GnRH) gene expression in estrogen receptor-alpha (ERalpha)- and ERbeta-expressing GT1-7 GnRH neurons. Endocrinology 1999; 140:5045-53. [PMID: 10537130 DOI: 10.1210/endo.140.11.7117] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Estrogen has wide-ranging and complex effects on the reproductive axis, which are often difficult to interpret from in vivo studies. Estrogen negatively regulates tonic GnRH synthesis and also plays a pivotal role in the positive regulation of GnRH necessary for the preovulatory surge. To dissect the mechanisms by which these divergent effects occur, we attempted to observe the direct action of estrogen on the regulation of GnRH messenger RNA (mRNA) levels using the well characterized, GnRH-secreting, hypothalamic cell line, GT1-7. Using RT-PCR, we first investigated estrogen receptor transcript expression in GT1-7 neurons. We found that the GT1-7 cells express both estrogen receptor-alpha (ERalpha) and the recently described ERbeta mRNAs. We also detected the presence of both receptor subtypes in the GT1-7 neurons by Western blot analysis using specific ER antibodies. By Northern blot analysis of total GT1-7 RNA, we found that 17beta-estradiol (1 nM) down-regulates GnRH mRNA levels to approximately 55% of basal levels over a 48-h time course. This effect appears to occur specifically through an ER-mediated mechanism, as ICI 182,780, a complete ER antagonist, blocks the repression of GnRH mRNA levels by estradiol. The recently reported ERalpha-specific agonist/ERbeta-specific antagonist 2,2-bis-(p-hydroxyphenyl-1,1,1-trichloroethane (HPTE), a methoxychlor metabolite, also down-regulated GnRH gene expression. The repression of GnRH mRNA levels appears to occur at the transcriptional level, as simian virus 40 T antigen mRNA expression, which is under the control of 2.3 kb of the rat GnRH 5'-regulatory region, mimics the down-regulation of GnRH after treatment with estradiol. As the rat GnRH regulatory region in GT1-7 neurons does not appear to harbor a classic estrogen response element, the mechanism involved in the repression of GnRH has yet to be determined. These results suggest that estradiol directly regulates GnRH gene expression at the level of the GnRH neuron and may exert its neuroendocrine control through direct interaction with specific receptors expressed in these cells.
Collapse
Affiliation(s)
- D Roy
- Institute for Medical Sciences, University of Toronto, Ontario, Canada
| | | | | |
Collapse
|
46
|
Fitzpatrick SL, Berrodin TJ, Jenkins SF, Sindoni DM, Deecher DC, Frail DE. Effect of estrogen agonists and antagonists on induction of progesterone receptor in a rat hypothalamic cell line. Endocrinology 1999; 140:3928-37. [PMID: 10465261 DOI: 10.1210/endo.140.9.7006] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogen is essential in the hypothalamus for the central regulation of reproduction. To understand the molecular mechanism(s) of estrogen action in the hypothalamus, immortalized rat embryonic hypothalamic cell lines were characterized for steroid receptors and subcloned. Scatchard analysis of the D12 subclone demonstrated one high affinity estrogen receptor-binding site (Kd = 31.3+/-1.9 pM) with a Bmax of 30.8+/-0.8 fmol/mg. Estrogen receptor-alpha protein was identified by Western blot and gel shift analyses. Treatment with estradiol (48 h) stimulated progesterone receptor (PR) messenger RNA expression and binding to [3H]R5020, a synthetic progestin. Because the agonist or antagonist activity of estrogen mimetics can be cell type dependent, the activities of various estrogen mimetics were determined in D12 cells. ICI 182,780 (IC50 = 0.63 nM), raloxifene (IC50 = 1 nM), enclomiphene (IC50 = 77 nM), and tamoxifen (IC50 = 174 nM) inhibited the induction of PR by estradiol, and none of these compounds significantly stimulated PR when given alone. In contrast, 17alpha-ethynyl estradiol (EC50 = 0.014 nM), zuclomiphene (EC50 = 100 nM), and genistein (EC50 = 17.5 nM) functioned as estrogen agonists in these cells. In addition, the estrogen-induced progesterone receptor activated a progesterone response element reporter construct in response to progestins. Thus, the D12 rat hypothalamic cell line provides a useful model for characterizing tissue-selective estrogenic compounds, identifying estrogen- and progesterone-regulated hypothalamic genes, and understanding the molecular mechanisms of steroid action in various physiological processes mediated by the hypothalamus.
Collapse
Affiliation(s)
- S L Fitzpatrick
- Women's Health Research Institute, Wyeth-Ayerst Research, Radnor, Pennsylvania 19087, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Prolactin (PRL) exerts numerous effects in the brain including induction of maternal behaviour, increased food intake, and inhibition of GnRH secretion. Knowledge about the distribution of PRL receptors (PRL-R) in the brain will be critical for investigating mechanisms of PRL-brain interactions during lactation. The present study aimed to investigate the distribution of PRL-R in specific hypothalamic nuclei of lactating rats by immunohistochemistry and to compare this distribution with that in dioestrous rats. Rats were perfused with 2% paraformaldehyde and brains were cut into coronal sections (18 microm) for immunostaining. Immunoreactivity was detected by the avidin biotin complex method using mouse monoclonal antibody U5. In dioestrous rats, PRL-R immunoreactivity was observed in the choroid plexus, three hypothalamic nuclei: medial preoptic, periventricular and arcuate, and in the median eminence. The number of labelled profiles per section in the medial preoptic and arcuate nuclei increased significantly (P<0.05) in lactating rats (days 7-10 to post partum) when compared with dioestrous rats. Furthermore, in lactating rats, PRL-R immunoreactive neurons were identified in the cerebral cortex, substantia nigra and numerous additional hypothalamic nuclei including the ventromedial preoptic, ventrolateral preoptic, lateroanterior hypothalamic, ventrolateral hypothalamic, paraventricular hypothalamic, supraoptic, suprachiasmatic, and ventromedial hypothalamic nuclei. These observations assist our understanding of the multiple sites of PRL effects on brain function during lactation.
Collapse
Affiliation(s)
- X J Pi
- Department of Anatomy and Structural Biology, School of Medical Sciences and Neuroscience Research Centre, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
48
|
Pi X, Grattan DR. Expression of prolactin receptor mRNA is increased in the preoptic area of lactating rats. Endocrine 1999; 11:91-8. [PMID: 10668647 DOI: 10.1385/endo:11:1:91] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/1999] [Revised: 05/12/1999] [Accepted: 06/01/1999] [Indexed: 11/11/2022]
Abstract
This study investigated expression of prolactin receptor (PRL-R) mRNA in the preoptic area in midlactating rats compared with diestrous rats. Tissues from specific nuclei were micropunched from 300-microm thick rat brain sections with 300- or 500-microm diameter needles. After total RNA was extracted, the two forms of PRL-R mRNA were evaluated by reverse transcriptase polymerase chain reaction and Southern hybridization. The results showed that levels of long-form PRL-R mRNA in the ventrolateral preoptic nucleus and lateroanterior nucleus in lactating rats were significantly higher (p < 0.05) than in diestrous rats. The ventromedial and medial preoptic nuclei in lactating rats also expressed moderately high levels of long-form mRNA when compared with (p = 0.0547) diestrous rats. The ventromedial and ventrolateral preoptic nuclei, and ventrolateral hypothalamic nucleus in lactating rats expressed significantly higher levels of short-form mRNA than in diestrous rats. The increased expression of both forms of PRL-R mRNA helps explain numerous effects of PRL on brain functions during lactation.
Collapse
Affiliation(s)
- X Pi
- Department of Anatomy and Structural Biology, School of Medical Sciences and Neuroscience Research Centre, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
49
|
Adams MM, Flagg RA, Gore AC. Perinatal changes in hypothalamic N-methyl-D-aspartate receptors and their relationship to gonadotropin-releasing hormone neurons. Endocrinology 1999; 140:2288-96. [PMID: 10218982 DOI: 10.1210/endo.140.5.6749] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During the neonatal period, the brain is subject to profound alterations in neuronal circuitry due to high levels of synaptogenesis and gliogenesis. In neuroendocrine regions such as the preoptic area-anterior hypothalamus (POA-AH), the site of GnRH perikarya, these changes could affect the maturation of GnRH neurons. Because the GnRH system is developmentally regulated by glutamatergic neurons, we hypothesized that changes in the N-methyl-D-aspartate (NMDA) receptor system begin early in postnatal development, before the onset of puberty, thereby playing a role in establishing the appropriate environment for the subsequent maturation of GnRH neurons. To this end, we determined developmental changes in NMDA receptors, alterations in GnRH gene expression, and the regulation of GnRH neurons by the NMDA receptor system in developing male and female rats. In Exp I, NMDA receptor subunit (NR) 1 mRNA levels in the POA-AH were found to increase significantly (approximately 5-fold) from E18 through P10 in both males and females. NR2b mRNA increased significantly between P0 and P5 in both males and females. In contrast, NR2a subunit mRNA, which was in very low abundance in both males and females, increased only in males between P10 and P15. In Exp II we determined that GnRH gene expression changes differentially in developing male and female rats, with increases from P0 to P5 in males, and decreases from P5 to P10 in females. This latter effect in females is attributed to a change in GnRH gene transcription because GnRH primary transcript RNA levels paralleled changes in GnRH mRNA levels. In Exp III, we tested effects of treatment with an NMDA receptor analog on GnRH mRNA levels and found that only P5 and P10 male rats responded to NMDA receptor activation with an increase in GnRH mRNA levels, via a posttranscriptional mechanism. This greater responsiveness of males to NMDA receptor stimulation may be due to differences in the composition and levels of NMDA receptor subunits. Exp IV examined the localization of NR1 in the POA-AH during neonatal development. No GnRH neurons were immunopositive for NR1, indicating that effects of glutamate on GnRH neurons are mediated by interneurons or other glutamate receptor subunits or types. Taken together, these data indicate that glutamatergic inputs to the POA-AH change dramatically during the early postnatal period, before puberty and before the GnRH system is fully responsive to glutamate, consistent with the hypothesis that the maturation of inputs to GnRH neurons, and the establishment of the proper neurotransmitter "milieu" enabling the activation of GnRH neurons, occurs before the onset of puberty.
Collapse
Affiliation(s)
- M M Adams
- Neurobiology of Aging Laboratories and Fishberg Research Center for Neurobiology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | |
Collapse
|
50
|
Bakker J, Rubin BS, Baum MJ. Changes in mediobasal hypothalamic gonadotropin-releasing hormone messenger ribonucleic acid levels induced by mating or ovariectomy in a reflex ovulator, the ferret. Endocrinology 1999; 140:595-602. [PMID: 9927282 DOI: 10.1210/endo.140.2.6519] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ferret is a reflex-ovulating species in which receipt of an intromission induces a prolonged (+/- 12 h) preovulatory LH surge in the estrous female. This LH surge is probably stimulated by a large release of GnRH from the mediobasal hypothalamus (MBH). In Exp 1 we asked whether GnRH messenger RNA (mRNA) levels increase in response to mating so as to replenish the MBH GnRH stores needed to sustain the preovulatory LH surge. Estrous females were killed 0, 0.25, 0.5, 1, 3, 6, 14, or 24 h after the onset of a 10-min intromission from a male. Coronal brain sections ranging from the rostral preoptic area caudally to the posterior hypothalamus were processed for in situ hybridization using a 35S-labeled oligoprobe complementary to the human GnRH-coding region. We found no evidence of increased MBH GnRH mRNA levels during the ferret's mating-induced preovulatory LH surge. Instead, the number of GnRH mRNA-expressing cells dropped significantly in the arcuate region beginning 6 h after onset of intromission and remained low thereafter. Furthermore, cellular GnRH mRNA levels decreased in the arcuate region toward the end of the preovulatory LH surge. In Exp 2 we asked whether ovarian hormones regulate MBH GnRH mRNA levels in the female ferret. Ovariectomy of estrous females significantly reduced the number of GnRH mRNA-expressing cells in the arcuate region. This decrease was probably not due to the absence of circulating estradiol. Gonadally intact anestrous females had levels of MBH GnRH mRNA similar to those in estrous females even though plasma estradiol levels were equally low in anestrous females and ovariectomized females. Ovarian hormones other than estradiol may stimulate MBH GnRH mRNA levels in anestrous and estrous females.
Collapse
Affiliation(s)
- J Bakker
- Department of Biology, Boston University, Massachusetts 02215, USA.
| | | | | |
Collapse
|