Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
J Neurosci ; 21(1): 330-9, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11150350

ABSTRACT

Chronic stressors produce changes in hippocampal neurochemistry, neuronal morphology, and hippocampal-dependent learning and memory processes. In rats, stress-induced changes in CA3 apical dendritic structure are mediated by corticosterone (CORT) acting, in part, on excitatory amino acid neurotransmission. CORT also alters GABA-mediated inhibitory neurotransmission, so the GABA(A) receptor system may also contribute to dendritic remodeling and other stress-related changes in hippocampal function. A previous study indicated that chronic CORT treatment produces complex changes in GABA(A) receptor subunit mRNA levels, so we hypothesized that CORT alters the pharmacological properties of hippocampal GABA(A) receptors. To test this, adult male rats were treated with CORT or vehicle pellets for 10 d, after which we quantified [(35)S]t-butylbicyclophosphorothionate ([(35)S]TBPS) and [(3)H]flunitrazepam binding to GABA(A) receptors using in vitro receptor autoradiography. Pharmacological properties of receptors were assessed by examining the allosteric regulation of binding at both sites by GABA and 5alpha-pregnane-3alpha,21-diol-20-one (THDOC), an endogenous anxiolytic steroid. We found striking regional differences in the modulation of [(35)S]TBPS binding, particularly between strata radiatum and strata oriens, suggesting a functional heterogeneity among hippocampal GABA(A) receptors even within the apical versus basal dendrites of pyramidal neurons. Furthermore, we found that CORT treatment decreased the negative modulation of hippocampal [(35)S]TBPS binding by both GABA and THDOC and increased the enhancement of [(3)H]flunitrazepam binding by GABA and THDOC in the dentate gyrus. Together, these data suggest that prolonged exposure to stress levels of corticosteroids may alter hippocampal inhibitory tone by regulating the pharmacological properties of GABA(A) receptors in discrete dendritic subfields.


Subject(s)
Corticosterone/metabolism , Desoxycorticosterone/analogs & derivatives , Hippocampus/metabolism , Receptors, GABA-A/metabolism , Allosteric Regulation/drug effects , Animals , Autoradiography , Binding, Competitive/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Corticosterone/administration & dosage , Dendrites/metabolism , Desoxycorticosterone/pharmacology , Drug Implants , Flunitrazepam/pharmacokinetics , Hippocampus/cytology , Hippocampus/drug effects , Ligands , Male , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
2.
Endocrinology ; 140(3): 1044-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10067823

ABSTRACT

Estrogens regulate the formation of excitatory synaptic connections in the hippocampus of female rats. Because the adult hippocampus has a very low concentration of intracellular estrogen receptors, it is unclear whether a conventional genomic mechanism is involved. Nonsteroidal estrogen antagonists are useful tools to study estrogen action because they can provide pharmacological data in favor of a particular pathway of estrogen action and evidence against other pathways. To investigate the role of intracellular estrogen receptors in the estrogen induction of synapse formation, we took advantage of previous studies in which we had shown that an estrogen antagonist, CI-628, enters the brain and blocks estrogen induction of progestin receptors to study whether the same antagonist would either mimic or block effects of estradiol to induce excitatory spine synapses. Using silver impregnation of neurons by the single section Golgi technique and morphometric analysis, we found that CI-628 effectively prevented estrogen induction of spines on CA1 pyramidal neurons, without having any agonist effects of its own. This result is consistent with an action of estradiol via intracellular estrogen receptors that are known to be expressed by interneurons within the hippocampus.


Subject(s)
Dendrites/drug effects , Estrogen Antagonists/pharmacology , Nitromifene/pharmacology , Pyramidal Cells/drug effects , Receptors, Estrogen/physiology , Animals , Estradiol/pharmacology , Female , Pyramidal Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/physiology
3.
Psychopharmacol Bull ; 34(3): 251-9, 1998.
Article in English | MEDLINE | ID: mdl-9803750

ABSTRACT

Ovarian steroids produce a variety of effects on the brain, influencing diverse nonreproductive processes such as cognitive function, motor activity, seizure susceptibility, and pain sensitivity, as well as pathological processes such as Parkinson's disease and Alzheimer's disease. Studies of ovarian hormone effects on animal brains have revealed a wide array of neurochemical and structural effects of ovarian steroids, which are reviewed in this article. These studies provide a foundation for understanding hormone effects on mood, behavior, and cognition in the menstrual cycle, during reproductive transitions and in depressive illness.


Subject(s)
Gonadal Steroid Hormones/pharmacology , Mental Health , Animals , Humans , Sex Characteristics
4.
Proc Natl Acad Sci U S A ; 95(6): 3281-6, 1998 Mar 17.
Article in English | MEDLINE | ID: mdl-9501254

ABSTRACT

Evidence exists for the localization of the newly identified estrogen receptor beta (ERbeta) within the rat paraventricular nucleus (PVN) and supraoptic nucleus (SON), regions which lack ERalpha. Presently, we investigate whether ERbeta-like-immunoreactivity (-ir) is found within cells of several major neuropeptide systems of these regions. Young adult Sprague-Dawley rats were ovariectomized (OVX), and 1 week later half of the animals received estradiol-17beta (E). Dual-label immunocytochemistry was performed on adjacent sections by using an ERbeta antibody, followed by an antibody to either oxytocin (OT), arginine-vasopressin (AVP), or corticotropin releasing hormone. Nuclear ERbeta-ir was identified within SON and retrochiasmatic SON, and in specific PVN subnuclei: medial parvicellular part, ventral and dorsal zones, dorsal and lateral parvicellular parts, and in the posterior magnocellular part, medial and lateral zones. However, the ERbeta-ir within magnocellular areas was noticeably less intense. OT-/ERbeta-ir colocalization was confirmed in neurons of the parvicellular subnuclei, in both OVX and OVX+E brains ( approximately 50% of OT and 25% of ERbeta-labeled cells between bregma -1.78 and -2.00). In contrast, few PVN parvicellular neurons contained both AVP- and ERbeta-ir. As well, very little overlap was observed in the distribution of cells containing corticotropin releasing hormone- or ERbeta-ir. In the SON, most nuclear ERbeta-ir colocalized with AVP-ir, whereas few OT-/ERbeta-ir dual-labeled cells were observed. These findings suggest that estrogen can directly modulate specific OT and AVP systems through an ERbeta-mediated mechanism, in a tissue-specific manner.


Subject(s)
Arginine Vasopressin/isolation & purification , Corticotropin-Releasing Hormone/isolation & purification , Hypothalamus, Anterior/chemistry , Oxytocin/isolation & purification , Receptors, Estrogen/isolation & purification , Animals , Estrogen Receptor beta , Female , Hypothalamus, Anterior/cytology , Immunohistochemistry , Neurons/chemistry , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/cytology , Rats , Rats, Sprague-Dawley , Supraoptic Nucleus/chemistry , Supraoptic Nucleus/cytology , Tissue Distribution
5.
J Comp Neurol ; 391(3): 322-34, 1998 Feb 16.
Article in English | MEDLINE | ID: mdl-9492203

ABSTRACT

Estradiol and progesterone modulate central serotonergic activity; however, the mechanism(s) of action remain unclear. Recently, estradiol-induced progestin receptors (PRs) have been localized within the majority of serotonin (5-HT) neurons in the female macaque dorsal raphe nucleus (DRN; Bethea [1994] Neuroendocrinology 60:50-61). In the present study, we investigated whether estrogen receptors (ERs) and/or PRs exist within 5-HT and/or non-5-HT cells in the female and male rat DRN and whether estradiol treatment alters the expression of these receptors. Young adult female and male Sprague-Dawley rats were gonadectomized, and 1 week later, half of the animals received a subcutaneous Silastic implant of estradiol-17beta. Animals were transcardially perfused 2 days later with acrolein and paraformaldehyde, and sequential dual-label immunocytochemistry was performed on adjacent sections by using either a PR antibody or an ERalpha antibody. This was followed by an antibody to either the 5-HT-synthesizing enzyme, tryptophan hydroxylase (TPH), or to the astrocytic marker, glial fibrillary acidic protein (GFAP). Cells containing immunoreactivity (ir) for nuclear ERs or PRs were identified within the rat DRN in a region-specific distribution in both sexes. No colocalization of nuclear ER-ir or PR-ir with cytoplasmic TPH-ir or GFAP-ir was observed in either sex or treatment, indicating that the steroid target cells are neither 5-HT neurons nor astrocytes. Females were found to have approximately 30% more PR-labeled cells compared with males throughout the DRN (P < 0.05), but no sex difference was detected in the number of neurons demonstrating ER-ir. In both sexes, 2 days of estradiol exposure decreased the number of cells with ER-ir, whereas it greatly increased the number of cells containing PR-ir in several DRN regions (P < 0.005). Collectively, these findings demonstrate the existence of nonserotonergic cells that contain nuclear ERs or PRs within the female and male rat DRN, including estradiol-inducible PRs. These findings point to a species difference in ovarian steroid regulation of 5-HT activity between the macaque and the rat, perhaps transsynaptically via local neurons in the rat brain.


Subject(s)
Raphe Nuclei/chemistry , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Animals , Female , Immunohistochemistry , Male , Neurons/chemistry , Rats , Rats, Sprague-Dawley , Serotonin/analysis , Species Specificity
6.
J Comp Neurol ; 388(4): 603-12, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9388019

ABSTRACT

Estrogen regulates the synaptic plasticity and physiology of the hippocampus as well as learning behaviors that are mediated by the hippocampus. The density of dendritic spines and synapses, the number of N-methyl-D-aspartate (NMDA) binding sites, the levels of NMDA receptor subunit NR1 protein, muscimol binding to the gamma-amino butyric acid (GABA)A receptor, and levels of glutamic acid decarboxylase message in the CA1 region of the hippocampus are altered with estrogen treatment. In addition, some of these parameters exhibit sex differences in their response to estrogen treatment. To establish that estrogen can have a direct effect on the hippocampus and to determine whether or not sex differences in estrogen responsiveness are due to sex differences in estrogen receptor (ER) levels, we used immunocytochemistry with the AS409 antibody to map the location of ER-immunoreactive (ER-ir) cells in the hippocampus of male and female rats. We found that (1) the ERs appear to be in interneurons rather than pyramidal or granule cell neurons, (2) ER-ir cells are located in greatest concentration in the hilus of the dentate gyrus and the stratum radiatum of the CA1 region, (3) the density of ER-ir cells exhibits a rostral to caudal gradient in the hilus and the CA1 regions, (4) there are no sex differences in either the number or immunostaining intensity of ER-ir cells in the hippocampus, (5) the ER levels are down-regulated by estrogen in both male and female rats, and (6) the mean intensity of staining for the ER-ir cells in the hippocampus is about 25% of that in the ER-ir cells of the hypothalamus. From this, we can conclude that estrogen can have a direct effect on hippocampal neurons and that any sex differences in estrogen responsiveness is due to something other than sex differences in ER levels or function in the hippocampus.


Subject(s)
Estrogens/physiology , Hippocampus/chemistry , Receptors, Estrogen/analysis , Animals , Down-Regulation , Female , Hippocampus/cytology , Interneurons/chemistry , Male , Pyramidal Cells/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/analysis , Receptors, Progesterone/biosynthesis , Sex Characteristics
7.
Neuroscience ; 78(3): 653-62, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9153648

ABSTRACT

Some of the effects of glucocorticoids on the function and neuronal plasticity of the hippocampus are mediated by N-methyl-D-aspartate receptor activation. We tested the hypothesis that chronic corticosterone administration increases N-methyl-D-aspartate receptor expression in the hippocampus of the rat. We used in situ hybridization histochemistry to measure the messenger RNA levels for the NR1, NR2A and NR2B subunits of the N-methyl-D-aspartate receptor and [3H]dizocilpine maleate (a non-competitive antagonist) binding to measure N-methyl-D-aspartate receptor density. Since corticosterone depresses circulating testosterone levels, we also examined whether the effects of corticosterone are mediated by or interact with the effects of testosterone. In the intact animal, corticosterone increased messenger RNA levels for NR2A and NR2B but not NR1 subunits of the N-methyl-D-aspartate receptor in all regions of the hippocampus. Testosterone had no significant effect on messenger RNA levels of any of the subunits. The subunit composition determines the functional and pharmacological properties of the N-methyl-D-aspartate receptor. We used ifenprodil inhibition of [3H]dizocilpine maleate binding, which has been used to distinguish NR2A- from NR2B-containing receptors, to determine whether corticosterone altered the proportion of high- and low-affinity sites for ifenprodil in parallel with the changes in subunit messenger RNA levels. Corticosterone increased the density of [3H]dizocilpine maleate binding sites without changing the dissociation constant for [3H]dizocilpine maleate or the proportion of high- and low-affinity sites for ifenprodil. These data suggest that the effects of corticosterone on hippocampal function are mediated, in part, by parallel increases in NR2A and NR2B subunit levels and the number of receptor channel binding sites.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Corticosterone/pharmacology , Hippocampus/metabolism , RNA, Messenger/biosynthesis , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/biosynthesis , Animals , Binding Sites/drug effects , Binding, Competitive/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Dizocilpine Maleate/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Glucocorticoids/pharmacology , Hippocampus/drug effects , Image Processing, Computer-Assisted , In Situ Hybridization , Male , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Testosterone/pharmacology
8.
Neurology ; 48(5 Suppl 7): S8-15, 1997 May.
Article in English | MEDLINE | ID: mdl-9153161

ABSTRACT

Ovarian steroids have many effects on the brain throughout the lifespan, beginning during gestation and continuing into senescence. These hormones affect areas of the brain that are not primarily involved in reproduction, such as the basal forebrain, hippocampus, caudate putamen, midbrain raphe, and brainstem locus coeruleus. Here we discuss three effects of estrogens and progestins that are especially relevant to memory processes and identify hormonal alterations associated with aging and neurodegenerative diseases. First, estrogens and progestins regulate synaptogenesis in the CA1 region of the hippocampus during the 4- to 5-day estrous cycle of the female rat. Formation of new excitatory synapses is induced by estradiol and involves N-methyl-D-aspartate (NMDA) receptors, whereas synaptic downregulation involves intracellular progestin receptors. Second, there are developmentally programmed sex differences in the hippocampal structure that mat help explain why male and female rats use different strategies to solve spatial navigation problems. During the period of development when testosterone is elevated in the male, aromatase and estrogen receptors are transiently expressed in the hippocampus. Recent data on behavior and synapse induction strongly suggest that this pathway is involved in the masculinization or defeminization of hippocampal structure and function. Third, ovarian steroids have effects throughout the brain, including effects on brainstem and midbrain catecholaminergic neurons, midbrain serotonergic pathways, and the basal forebrain cholinergic system. Regulation of the serotonergic system appears to be linked to the presence of estrogen- and progestin-sensitive neurons in the midbrain raphe, whereas the ovarian steroid influence on cholinergic function involves induction of choline acetyltransferase and acetylcholinesterase according to a sexually dimorphic pattern. Because of these widespread influences on these various neuronal systems, it is not surprising that ovarian steroids produce measurable cognitive effects after ovariectomy and during aging.


Subject(s)
Aging/physiology , Brain/physiology , Cognition/physiology , Estrogens/physiology , Progestins/physiology , Animals , Female , Humans , Male , Sex Characteristics , Synapses/physiology
9.
J Neurosci ; 17(5): 1848-59, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9030643

ABSTRACT

Previous studies have shown that estradiol induces new dendritic spines and synapses on hippocampal CA1 pyramidal cells. We have assessed the consequences of estradiol-induced dendritic spines on CA1 pyramidal cell intrinsic and synaptic electrophysiological properties. Hippocampal slices were prepared from ovariectomized rats treated with either estradiol or oil vehicle. CA1 pyramidal cells were recorded and injected with biocytin to visualize spines. The association of dendritic spine density and electrophysiological parameters for each cell was then tested using linear regression analysis. We found a negative relationship between spine density and input resistance; however, no other intrinsic property measured was significantly associated with dendritic spine density. Glutamate receptor autoradiography demonstrated an estradiol-induced increase in binding to NMDA, but not AMPA, receptors. We then used input/output (I/O) curves (EPSP slope vs stimulus intensity) to determine whether the sensitivity of CA1 pyramidal cells to synaptic input is correlated with dendritic spine density. Consistent with the lack of an estradiol effect on AMPA receptor binding, we observed no relationship between the slope of an I/O curve generated under standard recording conditions, in which the AMPA receptor dominates the EPSP, and spine density. However, recording the pharmacologically isolated NMDA receptor-mediated component of the EPSP revealed a significant correlation between I/O slope and spine density. These results indicate that, in parallel with estradiol-induced increases in spine/synapse density and NMDA receptor binding, estradiol treatment increases sensitivity of CA1 pyramidal cells to NMDA receptor-mediated synaptic input; further, sensitivity to NMDA receptor-mediated synaptic input is well correlated with dendritic spine density.


Subject(s)
Dendrites/physiology , Estradiol/pharmacology , Pyramidal Cells/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Synaptic Transmission/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/drug effects , Animals , Bicuculline/pharmacology , Binding, Competitive , Dendrites/ultrastructure , Drug Synergism , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Glutamic Acid/metabolism , Kynurenic Acid/pharmacology , Ovariectomy , Pyramidal Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Synaptic Transmission/drug effects
10.
Recent Prog Horm Res ; 52: 279-303; discussion 303-5, 1997.
Article in English | MEDLINE | ID: mdl-9238857

ABSTRACT

The menopause marks the permanent end of fertility in women. It was once thought that the exhaustion of ovarian follicles was the single, most important explanation for the transition to the menopause. Over the past decade, this perception has gradually changed with the realization that there are multiple pacemakers of reproductive senescence. We will present evidence that lends credence to the hypothesis that the central nervous system is a critical pacemaker of reproductive aging and that changes at this level contribute to the timing of the menopause. Studies demonstrate that an increasing de-synchronization of the temporal order of neuroendocrine signals may contribute to the accelerated rate of follicular loss that occurs during middle age. We suggest that the dampening and destabilization of the precisely orchestrated ultradian, circadian, and infradian neural signals lead to miscommunication between the brain and the pituitary-ovarian axis. This constellation of hypothalamic-pituitary-ovarian events leads to the inexorable decline of regular cyclicity and heralds menopausal transition.


Subject(s)
Aging , Brain/physiology , Reproduction/physiology , Aged , Circadian Rhythm , Female , Humans , Luteinizing Hormone/metabolism , Menopause , Middle Aged , Ovary/physiology , Periodicity , Pituitary Gland/physiology
11.
J Neurosci ; 16(21): 6830-8, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8824322

ABSTRACT

Estradiol treatment increases the number of NMDA receptor binding sites, and changes evoked synaptic currents in a manner consistent with a steroid-induced functional enhancement of NMDA receptors in rat hippocampus. In this study, we investigate the cellular mechanisms of estradiol-induced NMDA receptor regulation at the protein and mRNA levels in ovariectomized rats treated with ovarian steroids using immunocytochemical and in situ hybridization techniques. Confocal laser scanning microscopy was used to quantify alterations in immunofluorescence intensity levels of NMDAR1 subunit proteins within neuronal somata and dendrites of discrete hippocampal fields, whereas in parallel, in situ hybridization was used to examine NMDAR1 mRNA levels in corresponding hippocampal regions. The data indicate that estradiol treatment in ovariectomized rats significantly increases immunofluorescence intensity levels in comparison with nonsteroid treated ovariectomized rats within the somata and dendrites of CA1 pyramidal cells and, to a lesser extent, within the granule cell somata of the dentate gyrus. In contrast, such alterations in immunofluorescence intensity occur without concomitant changes in mRNA hybridization levels. Thus, these data suggest that estradiol modulates NMDA receptor function via post-transcriptional regulation of the NMDAR1 subunit protein. The increase in immunofluorescence intensity may reflect an increase in the concentration of the subunit protein, which could account for estrogen-induced changes in pharmacological and physiological properties of the NMDA receptor.


Subject(s)
Estradiol/pharmacology , Hippocampus/cytology , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Autoradiography , Blotting, Western , Dendrites/chemistry , Dendrites/physiology , Female , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Immunohistochemistry , In Situ Hybridization , Microscopy, Confocal , Neurons/chemistry , Neurons/physiology , Neurons/ultrastructure , Ovariectomy , Progesterone/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/analysis , Transcription, Genetic/drug effects
12.
Brain Res Mol Brain Res ; 34(1): 29-37, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-8750858

ABSTRACT

Chronic exposure to stress levels of corticosteroids alters many aspects of hippocampal function and may lead to neurodegeneration. Male rats were treated for 10 days with corticosterone (CORT) or vehicle pellets, and mRNA levels for six gamma-aminobutyric acid (GABAA) receptor subunits were measured. Effects of castration on subunit mRNA levels in CORT- and vehicle-treated animals were also examined. In situ hybridization studies demonstrated that mRNA levels for hippocampal GABAA receptor alpha 1, alpha 2, beta 1, beta 2, beta 3, and gamma 2 subunits were differentially altered by CORT treatment. Levels of alpha 1 and alpha 2 mRNA decreased in the dentate gyrus, and beta 1 mRNA levels decreased in CA1 and dentate gyrus of CORT-, compared to vehicle-treated, animals. In contrast, beta 2 subunit levels increased in all hippocampal regions examined, beta 3 levels increased in the dentate gyrus, and gamma 2 levels increased in CA1-CA3. The alpha 1, beta 1, and beta 2 mRNA levels all increased in the cingulate cortex of CORT-treated animals. There was no significant effect of gonadal state on any of the subunits examined, but there was a significant negative correlation between testosterone levels and mRNA levels of alpha 1, alpha 2 and beta 3 in specific regions. These data demonstrate that chronic exposure to stress levels of CORT produces complex changes in the mRNA levels of multiple GABAA receptor subunits, independently of the CORT-induced suppression of circulating testosterone.


Subject(s)
Corticosterone/adverse effects , Hippocampus/drug effects , RNA, Messenger/metabolism , Receptors, GABA-A/drug effects , Stress, Physiological/physiopathology , Analysis of Variance , Animals , Chronic Disease , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Testosterone/blood , Testosterone/pharmacology
13.
Brain Res ; 694(1-2): 29-39, 1995 Oct 02.
Article in English | MEDLINE | ID: mdl-8974657

ABSTRACT

Muscarinic agonists can act through the hypothalamic ventromedial nucleus (VMN) to facilitate lordosis. To elucidate the neuronal mechanism(s) underlying this muscarinic facilitation, effects of muscarinic agents on the single-unit activity of VMN neurons recorded in brain tissue slices of estrogen-primed female rats were analyzed. All the agonists tested, including acetylcholine (ACh), oxotremorine-M (OM), carbachol (CCh) and McN-A-343 (McN), evoked primarily excitation (80-100%), some inhibition (0-20%) and occasional biphasic responses (0-8%). By comparing the response magnitude and the effectiveness in evoking a response, the rank order for evoking excitation, the primary response, was found to be: OM > CCh > ACh approximately McN, which is consistent with that (OM > CCh > McN) for facilitating lordosis reported by others. This consistency and the frequency of its occurrence suggest that the excitatory electric action of the muscarinic agonists is related to their facilitatory behavioral effect. Experiments with antagonists selective for M1 (pirenzepine), M2 (AF-DX 116) and M3 (4-DAMP and p-F-HHSiD) indicate that muscarinic excitations are mediated by M1 and/or M3, but not M2. Since M1 receptors have been shown to be neither sufficient nor necessary to mediate the muscarinic facilitation, M3 receptor may be crucially involved in this behavioral effect. Autoradiographic assays of binding to [3H]4-DAMP with or without pirenzepine and AF-DX 116, also indicate the presence of M3 receptors in the VMN. Quantitative analyses show that the M3 binding was not affected by the in vivo estrogen priming required to permit muscarinic agonists to facilitate lordosis. Thus, while the excitation mediated by M3 is likely to be involved in muscarinic facilitation of lordosis, the regulation of M3 receptor density does not seem to be involved in the permissive


Subject(s)
Parasympathetic Nervous System/physiology , Posture , Receptors, Muscarinic/metabolism , Sexual Behavior, Animal/physiology , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Autoradiography , Cholinergic Agents/pharmacology , Electrophysiology , Female , In Vitro Techniques , Rats , Rats, Sprague-Dawley , Ventromedial Hypothalamic Nucleus/drug effects
14.
Brain Res Mol Brain Res ; 32(2): 271-8, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7500838

ABSTRACT

The ability of ovarian steroids to regulate the excitability of hippocampal neurons may be mediated by alterations in the inhibitory activity of GABA. We assessed the ability of estradiol, progesterone, and 3 alpha-OH-5 alpha-pregnan-20-one (3 alpha-OH-DHP; a metabolite of progesterone) to regulate gene expression of selected GABAA receptor subunits (alpha 1, alpha 2, beta 1, beta 2, and gamma 2). Using in situ hybridization, we found that progesterone, or 3 alpha-OH-DHP, suppressed mRNA levels for the alpha 1 subunit in the CA2, CA3, and the dentate gyrus subfields of the hippocampus in animals that were pretreated with estradiol. Progesterone had a more limited effect on the alpha 2 subunit, suppressing mRNA levels in estradiol-primed animals only in the CA3 region. In contrast, progesterone increased mRNA levels for the gamma 2 subunit in the CA1, CA2, and CA3 regions of the hippocampus, but only in animals that were not estradiol-primed. Estradiol alone had no significant effect on the expression of any subunit examined. Beta 1 and beta 2 subunit mRNA levels were not altered by any of the hormones tested. These data support the conclusion that progesterone and its metabolites may regulate excitability of the hippocampus by modulating the GABAA receptor gene expression; these effects of progesterone are dependent upon the circulating levels of estradiol. Alterations in the gene expression of selective subunits may lead to changes in the density of GABAA receptor protein or to changes in receptor subunit composition which might alter receptor sensitivity to activation by GABA or modulators such as the benzodiazepines and convulsants.


Subject(s)
Hippocampus/drug effects , Progesterone/pharmacology , RNA, Messenger/biosynthesis , Receptors, GABA-A/metabolism , Animals , Estradiol/pharmacology , Female , Gene Expression/drug effects , In Situ Hybridization , Progesterone/metabolism , Radioimmunoassay , Rats , Rats, Inbred Strains , Steroids/pharmacology
15.
J Neuroendocrinol ; 7(5): 341-6, 1995 May.
Article in English | MEDLINE | ID: mdl-7550279

ABSTRACT

Enkephalin appears to modulate several aspects of reproductive function in female rats. The purpose of this study was to determine if lactation influences preproenkephalin gene expression in one or more hypothalamic nuclei known to be involved in maternal or reproductive behavior and prolactin secretion. Lactating rats were killed on day 3 (LAC 3) or day 10 (LAC 10) of lactation. Controls consisted of regular 4-day cycling rats that were killed on diestrous day 1, with 9 to 12 females per group. We used in situ hybridization histochemistry to assess preproenkephalin gene expression in individual cells in the medial preoptic nucleus, anterior, medial and posterior arcuate nucleus, magnocellular and parvocellular aspects of paraventricular nucleus, and ventromedial nucleus. Preproenkephalin mRNA in the anterior arcuate nucleus increased to reach significance (P < 0.05) at day 10 of lactation. Levels in the medial arcuate nucleus increased significantly (P < 0.001) by day 3 of lactation (LAC 3) and remained elevated on day 10 (LAC 10). No significant differences between lactating and control rats were detected in preproenkephalin mRNA levels in the posterior arcuate nucleus, medial preoptic nucleus or in the ventromedial nucleus. Substantial levels of preproenkephalin mRNA were found in the paraventricular nucleus, particularly in a limited region of the magnocellular portion. However, these levels did not change with lactation. These data provide evidence for differential regulation of the preproenkephalin gene during lactation. This change may contribute to lactational hyperprolactinemia and suppressed GnRH secretion, leading to reproductive acyclicity.


Subject(s)
Enkephalins/genetics , Gene Expression , Hypothalamus/metabolism , Lactation/physiology , Protein Precursors/genetics , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Female , Pregnancy , Preoptic Area/metabolism , Rats , Rats, Sprague-Dawley
16.
Brain Res ; 680(1-2): 217-25, 1995 May 22.
Article in English | MEDLINE | ID: mdl-7663979

ABSTRACT

Adrenal steroid and stress effects were determined in hippocampus on levels of dynorphin (DYN) mRNA, expressed in dentate gyrus, and excitatory amino acid receptors, measured in Ammon's horn and dentate gyrus. Adrenalectomy (ADX) decreased DYN mRNA levels in dentate gyrus and replacement with aldosterone (ALDO), a specific type I adrenal steroid receptor agonist, prevented the decrease. Ru28362, a specific type II receptor agonist, had no effect. Likewise, kainate receptor binding to the stratum lucidum and hilus region of dorsal hippocampus was decreased after ADX and this decrease was prevented by ALDO but not by Ru28362 treatment. Similar though smaller effects were found for CNQX binding to AMPA receptors but only in the dentate gyrus molecular or infra- and supragranular layers. Although corticosterone (CORT) treatment of intact rats (40 mg/kg for 3 weeks) elevated DYN mRNA levels in dentate gyrus, up to 14 days of daily restraint stress (1 or 6 h/day) had no significant effect. Neither CORT treatment nor repeated restraint stress altered NMDA and non-NMDA glutamate receptors in hippocampus. The results of this study showing ADX-induced decreases of DYN mRNA and CNQX binding in dentate gyrus and decreased kainate binding in mossy fiber terminal regions are consistent with morphological evidence showing that adrenal steroids maintain normal integrity and structure of dentate gyrus neurons and do so via type I adrenal steroid receptors. These same parameters are apparently not sensitive to chronic restraint stress although the effects of other stressors must be examined.


Subject(s)
Adrenal Glands/physiology , Dynorphins/genetics , RNA, Messenger/metabolism , Steroids/pharmacology , Stress, Physiological/metabolism , 2-Amino-5-phosphonovalerate/analogs & derivatives , 2-Amino-5-phosphonovalerate/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/metabolism , Adrenalectomy , Aldosterone/pharmacology , Androstanols/pharmacology , Animals , Corticosterone/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Restraint, Physical
17.
Ciba Found Symp ; 191: 52-66; discussion 66-73, 1995.
Article in English | MEDLINE | ID: mdl-8582205

ABSTRACT

Oestrogens have numerous effects on the brain, beginning during gestation and continuing on into adulthood. Many of these actions involve areas of the brain that are not primarily involved in reproduction, such as the basal forebrain, hippocampus, caudate putamen, midbrain raphe and brainstem locus coeruleus. This paper describes three actions of oestrogens that are especially relevant to brain mechanisms involved in memory processes and their alterations during ageing and neurodegenerative diseases: (1) the regulation of cholinergic neurons by oestradiol in the rat basal forebrain, involving induction of choline acetyltransferase and acetylcholinesterase according to a sexually dimorphic pattern; (2) the regulation of synaptogenesis in the CA1 region of the hippocampus by oestrogens and progestins during the four- to five-day oestrus cycle of the female rat. Formation of new excitatory synapses is induced by oestradiol and involves N-methyl-D-aspartate receptors; removal of these synapses involves intracellular progestin receptors; (3) sex differences in hippocampal structure, which may help to explain differences in the strategies that male and female rats use to solve spatial navigation problems. During the period of development when testosterone is elevated in the male, aromatase and oestrogen receptors are also elevated, making it likely that this pathway is involved in the masculinization of hippocampal structure.


Subject(s)
Aging/physiology , Estrogens/physiology , Memory/physiology , Nerve Degeneration/physiology , Neuronal Plasticity/physiology , Animals , Female , Humans , Male , Rats , Structure-Activity Relationship
18.
Mol Cell Neurosci ; 5(5): 451-8, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7820368

ABSTRACT

Hippocampal function is modulated by adrenal corticosteroids released in a diurnal rhythm or in response to stress. Hippocampal excitability is also modulated by the activity of gamma-aminobutyric acid (GABA), the major inhibitory neurotransmitter in the mammalian brain. One mechanism of corticosteroid action in the hippocampus is likely to be via regulation of the GABA system, including corticosteroid regulation of GABAA receptor expression. The GABAA receptor is a heterooligomeric complex composed of subunits derived from a number of distinct genes. We examined the effects of short-term adrenalectomy and low-level corticosterone replacement on mRNA levels for 5 GABAA receptor subunits. In situ hybridization studies demonstrated that mRNA levels for GABAA receptor alpha 1, alpha 2, beta 2, and gamma 2 subunits were altered in the hippocampus of female rats by adrenalectomy. Levels of alpha 1 and gamma 2 mRNA increased in CA3, alpha 2 mRNA increased in the dentate gyrus, while beta 2 mRNA decreased in the dentate gyrus and CA2 relative to sham-operated animals following adrenalectomy. These effects were reversed by the addition of 100 micrograms/ml corticosterone to the drinking water. Adrenalectomy had no effect on the levels of beta 1 mRNA and no effect on any subunit examined in CA1 or the cingulate cortex. These data support the conclusion that corticosteroids can modulate hippocampal excitability through the site-specific regulation of the expression of specific GABAA receptor subunits. Corticosterone-induced changes in subunit expression might alter GABAergic synaptic inhibition by altering the density of GABAA receptors or altering the subunit composition and thereby the pharmacological properties of the receptors.


Subject(s)
Adrenalectomy , Hippocampus/metabolism , Receptors, GABA/metabolism , Animals , Corticosterone/pharmacology , Female , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA/classification , Receptors, GABA/genetics , Tissue Distribution
20.
Microsc Res Tech ; 25(1): 40-5, 1993 May 01.
Article in English | MEDLINE | ID: mdl-7688996

ABSTRACT

We have developed an assay that allows one to monitor gene expression in and peptide secretion from individual cells. By combining the reverse hemolytic plaque with in situ hybridization, investigators can quantitate simultaneously the level of gene expression and the level of secretion of a peptide. The method can be used in any system in which an appropriate antibody for the reverse hemolytic plaque assay and probes complementary to the mRNA of interest are available. It can be used to monitor the level of mRNA and secretion of the peptide product, or expression of one gene and the secretion of another peptide. In this paper we will describe the major steps of the method. We have used the pituitary lactotroph as a model to demonstrate the power of this technique. However, we believe that this method may be an important approach to answer many questions regarding the cellular and molecular mechanisms that regulate the coupling of peptide secretion and gene expression at the single cell level.


Subject(s)
Pituitary Gland, Anterior/metabolism , Prolactin/metabolism , RNA, Messenger/metabolism , Animals , Cattle , Gene Expression , Hemolytic Plaque Technique , In Situ Hybridization , Prolactin/genetics , RNA , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...