Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Neuroendocrinol ; 33(12): e13061, 2021 12.
Article in English | MEDLINE | ID: mdl-34786775

ABSTRACT

Oxytocin (OXT) neurons of the hypothalamus are at the center of several physiological functions, including milk ejection, uterus contraction, and maternal and social behavior. In lactating females, OXT neurons show a pattern of burst firing and inter-neuron synchronization during suckling that leads to pulsatile release of surges of OXT into the bloodstream to stimulate milk ejection. This pattern of firing and population synchronization may be facilitated in part by hypothalamic glutamatergic circuits, as has been observed in vitro using brain slices obtained from male rats and neonates. However, it remains unknown how hypothalamic glutamatergic circuits influence OXT cell activity outside the context of lactation. In this review, we summarize the in vivo and in vitro studies that describe the synchronized burst firing pattern of OXT neurons and the implication of hypothalamic glutamate in this pattern of firing. We also make note of the few studies that have traced glutamatergic afferents to the hypothalamic paraventricular and supraoptic nuclei. Finally, we discuss the genetic findings implicating several glutamatergic genes in neurodevelopmental disorders, including autism spectrum disorder, thus underscoring the need for future studies to investigate the impact of these mutations on hypothalamic glutamatergic circuits and the OXT system.


Subject(s)
Glutamic Acid/metabolism , Hypothalamus/metabolism , Neurodevelopmental Disorders/etiology , Neurons/physiology , Oxytocin/metabolism , Animals , Cell Communication/physiology , Female , Humans , Male , Nerve Net/metabolism , Nerve Net/physiology , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/physiopathology , Neurons/metabolism , Rats
2.
J Neuroendocrinol ; 32(4): e12826, 2020 04.
Article in English | MEDLINE | ID: mdl-31917875

ABSTRACT

Hypothalamic magnocellular neuroendocrine cells have unique electrical properties and a remarkable capacity for morphological and synaptic plasticity. Their large somatic size, their relatively uniform and dense clustering in the supraoptic and paraventricular nuclei, and their large axon terminals in the neurohypophysis make them an attractive target for direct electrophysiological interrogation. Here, we provide a brief review of significant recent findings in the neuroplasticity and neurophysiological properties of these neurones that were presented at the symposium "Electrophysiology of Magnocellular Neurons" during the 13th World Congress on Neurohypophysial Hormones in Ein Gedi, Israel in April 2019. Magnocellular vasopressin (VP) neurones respond directly to hypertonic stimulation with membrane depolarisation, which is triggered by cell shrinkage-induced opening of an N-terminal-truncated variant of transient receptor potential vanilloid type-1 (TRPV1) channels. New findings indicate that this mechanotransduction depends on actin and microtubule cytoskeletal networks, and that direct coupling of the TRPV1 channels to microtubules is responsible for mechanical gating of the channels. Vasopressin neurones also respond to osmostimulation by activation of epithelial Na+ channels (ENaC). It was shown recently that changes in ENaC activity modulate magnocellular neurone basal firing by generating tonic changes in membrane potential. Both oxytocin and VP neurones also undergo robust excitatory synapse plasticity during chronic osmotic stimulation. Recent findings indicate that new glutamate synapses induced during chronic salt loading express highly labile Ca2+ -permeable GluA1 receptors requiring continuous dendritic protein synthesis for synapse maintenance. Finally, recordings from the uniquely tractable neurohypophysial terminals recently revealed an unexpected property of activity-dependent neuropeptide release. A significant fraction of the voltage-dependent neurohypophysial neurosecretion was found to be independent of Ca2+ influx through voltage-gated Ca2+ channels. Together, these findings provide a snapshot of significant new advances in the electrophysiological signalling mechanisms and neuroplasticity of the hypothalamic-neurohypophysial system, a system that continues to make important contributions to the field of neurophysiology.


Subject(s)
Hypothalamus/metabolism , Neuroendocrine Cells/metabolism , Neurons/metabolism , Oxytocin/metabolism , Vasopressins/metabolism , Animals , Neuronal Plasticity/physiology , Synapses/metabolism
3.
Cell Rep ; 29(13): 4349-4361.e4, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875545

ABSTRACT

In addition to their support role in neurotransmitter and ion buffering, astrocytes directly regulate neurotransmission at synapses via local bidirectional signaling with neurons. Here, we reveal a form of neuronal-astrocytic signaling that transmits retrograde dendritic signals to distal upstream neurons in order to activate recurrent synaptic circuits. Norepinephrine activates α1 adrenoreceptors in hypothalamic corticotropin-releasing hormone (CRH) neurons to stimulate dendritic release, which triggers an astrocytic calcium response and release of ATP; ATP stimulates action potentials in upstream glutamate and GABA neurons to activate recurrent excitatory and inhibitory synaptic circuits to the CRH neurons. Thus, norepinephrine activates a retrograde signaling mechanism in CRH neurons that engages astrocytes in order to extend dendritic volume transmission to reach distal presynaptic glutamate and GABA neurons, thereby amplifying volume transmission mediated by dendritic release.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Astrocytes/drug effects , Dendrites/drug effects , GABAergic Neurons/drug effects , Norepinephrine/pharmacology , Synaptic Transmission/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Cell Communication , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Dendrites/metabolism , Dendrites/ultrastructure , GABAergic Neurons/metabolism , GABAergic Neurons/ultrastructure , Gene Expression Regulation , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Male , Mice , Mice, Transgenic , Microtomy , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/physiology , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
4.
Physiol Rep ; 7(8): e14047, 2019 04.
Article in English | MEDLINE | ID: mdl-31008554

ABSTRACT

Hypothalamic magnocellular neurosecretory cells (MNCs) undergo dramatic structural reorganization during lactation in female rats that is thought to contribute to the pulsatile secretion of oxytocin critical for milk ejection. MNCs from male rats generate robust bursts of GABAergic synaptic currents, a subset of which are onset-synchronized between MNC pairs, but the functional role of the IPSC bursts is not known. To determine the physiological relevance of IPSC bursts, we compared MNCs from lactating and non-lactating female rats using whole-cell recordings in brain slices. We recorded a sixfold increase in the incidence of IPSC bursts in oxytocin (OT)-MNCs from lactating rats compared to non-lactating rats, whereas there was no change in IPSC bursts in vasopressin (VP)-MNCs. Synchronized bursts of IPSCs were observed in pairs of MNCs in slices from lactating rats. Our data indicate, therefore, that IPSC bursts are upregulated specifically in OT-MNCs during lactation, and may, therefore, contribute via rebound depolarization to the spike trains in OT neurons that lead to reflex milk ejection.


Subject(s)
Inhibitory Postsynaptic Potentials , Lactation/physiology , Neuroendocrine Cells/physiology , Oxytocin/metabolism , Animals , Female , Hypothalamus/cytology , Hypothalamus/metabolism , Hypothalamus/physiology , Lactation/metabolism , Neuroendocrine Cells/metabolism , Rats , Rats, Wistar , Vasopressins/metabolism
5.
Steroids ; 142: 55-64, 2019 02.
Article in English | MEDLINE | ID: mdl-29242167

ABSTRACT

Glucocorticoid binding to the intracellular glucocorticoid receptor (GR) stimulates the translocation of the GR from the cytosol to the nucleus, which leads to the transactivation or transrepression of gene transcription. However, multiple lines of evidence suggest that glucocorticoid signaling can also be initiated from the plasma membrane. Here, we provide evidence for membrane-initiated glucocorticoid signaling by a membrane-impermeant dexamethasone-bovine serum albumin (Dex-BSA) conjugate, which induced GR nuclear trafficking in hypothalamic neurons in vitro and in vivo. The GR nuclear translocation induced by a membrane-impermeant glucocorticoid suggests trafficking of an unliganded GR. The membrane-initiated GR trafficking was not blocked by inhibiting ERK MAPK, p38 MAPK, PKA, Akt, Src kinase, or calcium signaling, but was inhibited by Akt activation. Short-term exposure of hypothalamic neurons to dexamethasone (Dex) activated the glucocorticoid response element (GRE), suggesting transcriptional transactivation, whereas exposure to the Dex-BSA conjugate failed to activate the GRE, suggesting differential transcriptional activity of the liganded compared to the unliganded GR. Microarray analysis revealed divergent transcriptional regulation by Dex-BSA compared to Dex. Together, our data suggest that signaling from a putative membrane glucocorticoid receptor induces the trafficking of unliganded GR to the nucleus, which elicits a pattern of gene transcription that differs from that of the liganded receptor. The differential transcriptional signaling by liganded and unliganded receptors may contribute to the broad range of genetic regulation by glucocorticoids, and may help explain some of the different off-target actions of glucocorticoid drugs.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Dexamethasone/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Neurons/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cattle , Cells, Cultured , Dexamethasone/chemistry , Male , Mice , Mice, Inbred C57BL , Rats , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism
6.
Steroids ; 142: 2-5, 2019 02.
Article in English | MEDLINE | ID: mdl-28939328

ABSTRACT

Cellular effects of glucocorticoids can be separated into classical transcriptional regulation via activation of the canonical nuclear glucocorticoid receptor and rapid actions mediated by activation of one or more putative membrane-associated glucocorticoid receptors that regulate both transcriptional and non-transcriptional signaling. Dexamethasone-bovine serum albumin (Dex-BSA) is one of several membrane-limited steroid receptor agonists. Dex-BSA and other steroid conjugates such as corticosterone-, estradiol- and testosterone-BSA have been used to study rapid steroid effects initiated by putative membrane receptors. The purity and stability of the steroid-BSA conjugate is crucial, therefore, since any steroid that is not bound to or that dissociates from the BSA conjugate could penetrate into the intracellular compartment and confound the experiment. We used fluorine NMR to determine if free Dex could be detected in a commercially available Dex-BSA dissolved in H2O. Non-covalently bound Dex was detected in the Dex-BSA solution, but the level of free Dex remained constant over time and with increasing temperature, indicating that the free Dex was not a result of instability of the Dex-BSA conjugate. The free Dex was lost when the Dex-BSA was denatured and subjected to dialysis, which suggested that it was trapped in the Dex-BSA three-dimensional structure and not covalently bound to the BSA. The purified, renatured Dex-BSA retained its rapid activity, which confirmed that the observed effects of Dex-BSA are not caused by non-covalently-bound Dex. Therefore, the Dex contaminant found in the Dex-BSA solution is likely to be tightly, but non-covalently, bound to BSA, and the Dex-BSA activity remains membrane-limited. Our findings indicate that Dex-BSA remains a suitable membrane-restricted glucocorticoid receptor agonist, but suggest that denaturing purification is a useful control for the study of membrane-initiated steroid-BSA actions.


Subject(s)
Cell Membrane/drug effects , Cell Nucleus/drug effects , Dexamethasone/chemistry , Dexamethasone/pharmacology , Drug Contamination , Hypothalamus/drug effects , Receptors, Glucocorticoid/agonists , Serum Albumin, Bovine/chemistry , Animals , Cattle , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Dexamethasone/analysis , Drug Stability , Hypothalamus/metabolism , Mice , Rats , Receptors, Glucocorticoid/metabolism , Serum Albumin, Bovine/analysis
7.
Steroids ; 114: 33-40, 2016 10.
Article in English | MEDLINE | ID: mdl-27327842

ABSTRACT

In parallel with their well-characterized delayed genomic effects, steroid hormones exhibit rapid, non-genomic effects at molecular, cellular and behavioral levels. We have proposed a model of rapid, non-genomic glucocorticoid inhibition of hypothalamic neuroendocrine cells through a putative membrane-associated glucocorticoid receptor (GR). Here we tested for plasma membrane GR immunoreactivity and binding in the hypothalamic supraoptic and paraventricular nuclei. Selective cross-linking of membrane proteins with membrane-impermeant BS3 and subsequent Western blot analysis with a monoclonal GR antibody revealed a reduction in the intensities of a ∼98kDa immunoreactive band and a ∼64kDa band in the rat paraventricular and supraoptic nuclei, and of a 64kDa band in hippocampal tissue, which suggested that these proteins are associated with the membrane. Saturation binding of [3H]-corticosterone and [3H]-dexamethasone in rat and mouse hypothalamic tissue revealed a Kd 4-24-fold lower and a Bmax 4-7-fold lower for the membrane-associated GR compared to the intracellular GR, suggesting a lower affinity and abundance of the glucocorticoid binding sites in the membrane than in the cytosol. Together, these findings suggest the presence of a low-affinity, low-abundance membrane-associated GR in the hypothalamus that shares homology with the intracellular GR, and are consistent with physiological evidence of rapid, non-genomic glucocorticoid actions in hypothalamic neuroendocrine cells that are GR dependent.


Subject(s)
Glucocorticoids/metabolism , Hypothalamus/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Corticosterone/metabolism , Cytosol/metabolism , Dexamethasone/metabolism , In Vitro Techniques , Male , Mice , Protein Binding , Rats
8.
Int Rev Neurobiol ; 125: 163-201, 2015.
Article in English | MEDLINE | ID: mdl-26638767

ABSTRACT

The hypothalamus is a part of the brain that is critical for sustaining life through its homeostatic control and integrative regulation of the autonomic nervous system and neuroendocrine systems. Neuroendocrine function in mammals is mediated mainly through the control of pituitary hormone secretion by diverse neuroendocrine cell groups in the hypothalamus. Cannabinoid receptors are expressed throughout the hypothalamus, and endocannabinoids have been found to exert pronounced regulatory effects on neuroendocrine function via modulation of the outputs of several neuroendocrine systems. Here, we review the physiological regulation of neuroendocrine function by endocannabinoids, focusing on the role of endocannabinoids in the neuroendocrine regulation of the stress response, food intake, fluid homeostasis, and reproductive function. Cannabis sativa (marijuana) has a long history of recreational and/or medicinal use dating back to ancient times. It was used as an analgesic, anesthetic, and antianxiety herb as early as 2600 B.C. The hedonic, anxiolytic, and mood-elevating properties of cannabis have also been cited in ancient records from different cultures. However, it was not until 1964 that the psychoactive constituent of cannabis, Δ(9)-tetrahydrocannabinol, was isolated and its chemical structure determined (Gaoni & Mechoulam, 1964).


Subject(s)
Endocannabinoids/physiology , Neurosecretory Systems/physiology , Endocannabinoids/pharmacology , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism , Neurosecretory Systems/drug effects
9.
Endocrinology ; 156(8): 2843-53, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26046806

ABSTRACT

Glucocorticoids act rapidly at the paraventricular nucleus (PVN) to inhibit stress-excitatory neurons and limit excessive glucocorticoid secretion. The signaling mechanism underlying rapid feedback inhibition remains to be determined. The present study was designed to test the hypothesis that the canonical glucocorticoid receptors (GRs) is required for appropriate hypothalamic-pituitary-adrenal (HPA) axis regulation. Local PVN GR knockdown (KD) was achieved by breeding homozygous floxed GR mice with Sim1-cre recombinase transgenic mice. This genetic approach created mice with a KD of GR primarily confined to hypothalamic cell groups, including the PVN, sparing GR expression in other HPA axis limbic regulatory regions, and the pituitary. There were no differences in circadian nadir and peak corticosterone concentrations between male PVN GR KD mice and male littermate controls. However, reduction of PVN GR increased ACTH and corticosterone responses to acute, but not chronic stress, indicating that PVN GR is critical for limiting neuroendocrine responses to acute stress in males. Loss of PVN GR induced an opposite neuroendocrine phenotype in females, characterized by increased circadian nadir corticosterone levels and suppressed ACTH responses to acute restraint stress, without a concomitant change in corticosterone responses under acute or chronic stress conditions. PVN GR deletion had no effect on depression-like behavior in either sex in the forced swim test. Overall, these findings reveal pronounced sex differences in the PVN GR dependence of acute stress feedback regulation of HPA axis function. In addition, these data further indicate that glucocorticoid control of HPA axis responses after chronic stress operates via a PVN-independent mechanism.


Subject(s)
Gene Deletion , Hypothalamus/metabolism , Neurosecretory Systems/physiology , Receptors, Glucocorticoid/genetics , Animals , Circadian Rhythm/genetics , Feedback, Physiological , Female , Gene Knockdown Techniques , Hypothalamo-Hypophyseal System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pituitary-Adrenal System/physiology , Receptors, Glucocorticoid/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism
10.
Endocrinology ; 156(8): 2831-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26061727

ABSTRACT

Corticosteroids act classically via cognate nuclear receptors to regulate gene transcription; however, increasing evidence supports rapid, nontranscriptional corticosteroid actions via activation of membrane receptors. Using whole-cell patch clamp recordings in hypothalamic slices from male mouse genetic models, we tested for nongenomic glucocorticoid actions at glutamate and gamma aminobutyric acid (GABA) synapses in hypothalamic neuroendocrine cells, and for their dependence on the nuclear glucocorticoid receptor (GR). In enhanced green fluorescent protein-expressing CRH neurons of the paraventricular nucleus (PVN) and in magnocellular neurons of the PVN and supraoptic nucleus (SON), dexamethasone activated postsynaptic membrane-associated receptors and G protein signaling to elicit a rapid suppression of excitatory postsynaptic inputs, which was blocked by genetic deletion of type I cannabinoid receptors and a type I cannabinoid receptor antagonist. In magnocellular neurons, dexamethasone also elicited a rapid nitric oxide-dependent increase in inhibitory postsynaptic inputs. These data indicate a rapid, synapse-specific glucocorticoid-induced retrograde endocannabinoid signaling at glutamate synapses and nitric oxide signaling at GABA synapses. Unexpectedly, the rapid glucocorticoid effects on both excitatory and inhibitory synaptic transmission were lost with conditional deletion of GR in the PVN and SON in slices from a single minded-1-cre-directed conditional GR knockout mouse. Thus, the nongenomic glucocorticoid actions at glutamate and GABA synapses on PVN and SON neuroendocrine cells are dependent on the nuclear GR. The nuclear GR, therefore, is responsible for transducing the rapid steroid response at the membrane, or is either a critical component in the signaling cascade or regulates a critical component of the signaling cascade of a distinct membrane GR.


Subject(s)
Glucocorticoids/pharmacology , Hypothalamus/drug effects , Neuroendocrine Cells/drug effects , Receptors, Glucocorticoid/physiology , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroendocrine Cells/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Glucocorticoid/genetics , Supraoptic Nucleus/metabolism , Synaptic Transmission/genetics , Time Factors
11.
J Neurosci ; 33(46): 18331-42, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24227742

ABSTRACT

Cannabinoid receptors are functionally operant at both glutamate and GABA synapses on hypothalamic magnocellular neuroendocrine cells; however, retrograde endocannabinoid actions are evoked at only glutamate synapses. We tested whether the functional targeting of evoked retrograde endocannabinoid actions to glutamate, and not GABA, synapses on magnocellular neurons is the result of the spatial restriction of extracellular endocannabinoids by astrocytes. Whole-cell GABA synaptic currents were recorded in magnocellular neurons in rat hypothalamic slices following manipulations to reduce glial buffering of extracellular signals. Depolarization- and glucocorticoid-evoked retrograde endocannabinoid suppression of synaptic GABA release was not detected under normal conditions, but occurred in both oxytocin and vasopressin neurons under conditions of attenuated glial coverage and depressed glial metabolic function, suggesting an emergent endocannabinoid modulation of GABA synapses with the loss of astrocyte function. Tonic endocannabinoid suppression of GABA release was insensitive to glial manipulation. Blocking cannabinoid transport mimicked, and increasing the extracellular viscosity reversed, the effect of suppressed glial buffering on the endocannabinoid modulation of GABA release. Evoked, but not tonic, endocannabinoid modulation of GABA synapses was mediated by 2-arachidonoylglycerol. Therefore, depolarization- and glucocorticoid-evoked 2-arachidonoylglycerol release from magnocellular neurons is spatially restricted to glutamate synapses by astrocytes, but spills over onto GABA synapses under conditions of reduced astrocyte buffering; tonic endocannabinoid modulation of GABA release, in contrast, is likely mediated by anandamide and is insensitive to astrocytic buffering. Astrocytes, therefore, provide dynamic control of stimulus-evoked 2-arachidonoylglycerol, but not tonic anandamide, regulation of GABA synaptic inputs to magnocellular neuroendocrine cells under different physiological conditions.


Subject(s)
Endocannabinoids/physiology , Hypothalamus/physiology , Neuroendocrine Cells/physiology , Neuroglia/physiology , Synapses/physiology , Animals , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
12.
PLoS One ; 6(12): e28152, 2011.
Article in English | MEDLINE | ID: mdl-22164236

ABSTRACT

ProSAAS is the precursor of a number of peptides that have been proposed to function as neuropeptides. Because proSAAS mRNA is highly expressed in the arcuate nucleus of the hypothalamus, we examined the cellular localization of several proSAAS-derived peptides in the mouse hypothalamus and found that they generally colocalized with neuropeptide Y (NPY), but not α-melanocyte stimulating hormone. However, unlike proNPY mRNA, which is upregulated by food deprivation in the mediobasal hypothalamus, neither proSAAS mRNA nor proSAAS-derived peptides were significantly altered by 1-2 days of food deprivation in wild-type mice. Furthermore, while proSAAS mRNA levels in the mediobasal hypothalamus were significantly lower in Cpe(fat/fat) mice as compared to wild-type littermates, proNPY mRNA levels in the mediobasal hypothalamus and in other subregions of the hypothalamus were not significantly different between wild-type and Cpe(fat/fat) mice. Intracerebroventricular injections of antibodies to two proSAAS-derived peptides (big LEN and PEN) significantly reduced food intake in fasted mice, while injections of antibodies to two other proSAAS-derived peptides (little LEN and little SAAS) did not. Whole-cell patch clamp recordings of parvocellular neurons in the hypothalamic paraventricular nucleus, a target of arcuate NPY projections, showed that big LEN produced a rapid and reversible inhibition of synaptic glutamate release that was spike independent and abolished by blocking postsynaptic G protein activity, suggesting the involvement of a postsynaptic G protein-coupled receptor and the release of a retrograde synaptic messenger. Taken together with previous studies, these findings support a role for proSAAS-derived peptides such as big LEN as neuropeptides regulating food intake.


Subject(s)
Eating/genetics , Gene Expression Regulation , Neuropeptide Y/chemistry , Peptides/chemistry , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Feeding Behavior , GTP-Binding Proteins/metabolism , Green Fluorescent Proteins/metabolism , Hypothalamus/metabolism , Immunohistochemistry/methods , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence/methods , Nerve Tissue Proteins/genetics , Neuropeptides , Patch-Clamp Techniques , RNA, Messenger/metabolism
13.
J Neurosci ; 30(45): 14980-6, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21068301

ABSTRACT

Endocannabinoid signaling is distributed throughout the brain, regulating synaptic release of both excitatory and inhibitory neurotransmitters. The presence of endocannabinoid signaling within stress-sensitive nuclei of the hypothalamus, as well as upstream limbic structures such as the amygdala, suggests it may play an important role in regulating the neuroendocrine and behavioral effects of stress. The evidence reviewed here demonstrates that endocannabinoid signaling is involved in both activating and terminating the hypothalamic-pituitary-adrenal axis response to both acute and repeated stress. In addition to neuroendocrine function, however, endocannabinoid signaling is also recruited by stress and glucocorticoid hormones to modulate cognitive and emotional processes such as memory consolidation and extinction. Collectively, these data demonstrate the importance of endocannabinoid signaling at multiple levels as both a regulator and an effector of the stress response.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Hypothalamus/physiopathology , Stress, Psychological/physiopathology , Synapses/metabolism , Synaptic Transmission/physiology , Animals , Glucocorticoids/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/metabolism , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Stress, Psychological/metabolism
14.
Endocrinology ; 151(10): 4811-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20702575

ABSTRACT

Glucocorticoid hormones are secreted in response to stimuli that activate the hypothalamo-pituitary-adrenocortical (HPA) axis and self-regulate through negative feedback. Negative feedback that occurs on a rapid time scale is thought to act through nongenomic mechanisms. In these studies, we investigated fast feedback inhibition of HPA axis stress responses by direct glucocorticoid action at the paraventricular nucleus of the hypothalamus (PVN). Local infusion of dexamethasone or a membrane-impermeant dexamethasone-BSA conjugate into the PVN rapidly inhibits restraint-induced ACTH and corticosterone release in a manner consistent with feedback actions at the cell membrane. The dexamethasone fast feedback response is blocked by the cannabinoid CB1 receptor antagonist AM-251, suggesting that fast feedback requires local release of endocannabinoids. Hypothalamic tissue content of the endocannabinoid 2-arachidonoyl glycerol is elevated by restraint stress, consistent with endocannabinoid action on feedback processes. These data support the hypothesis that glucocorticoid-induced fast feedback inhibition of the HPA axis is mediated by a nongenomic signaling mechanism that involves endocannabinoid signaling at the level of the PVN.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Feedback, Physiological/drug effects , Glucocorticoids/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Animals , Cannabinoid Receptor Modulators/pharmacology , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Feedback, Physiological/physiology , Glucocorticoids/administration & dosage , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/metabolism , Injections, Intraventricular , Male , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiology , Rats , Rats, Sprague-Dawley , Restraint, Physical/physiology , Restraint, Physical/psychology , Signal Transduction/drug effects , Signal Transduction/physiology , Stress, Psychological/metabolism , Time Factors
15.
J Physiol ; 588(Pt 6): 939-51, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20123785

ABSTRACT

Spike-independent miniature postsynaptic currents are generally stochastic and are therefore not thought to mediate information relay in neuronal circuits. However, we recorded endogenous bursts of IPSCs in hypothalamic magnocellular neurones in the presence of TTX, which implicated a coordinated mechanism of spike-independent GABA release. IPSC bursts were identical in the absence of TTX, although the burst incidence increased 5-fold, indicating that IPSC bursts were composed of miniature IPSCs (mIPSCs), and that the probability of burst generation increased with action potential activity. IPSC bursts required extracellular calcium, although they were not dependent on calcium influx through voltage-gated calcium channels or on calcium mobilization from intracellular stores. Current injections simulating IPSC bursts were capable of triggering and terminating action potential trains. In 25% of dual recordings, a subset of IPSC bursts were highly synchronized in onset in pairs of magnocellular neurones. Synchronized IPSC bursts displayed properties that were consistent with simultaneous release at GABA synapses shared between pairs of postsynaptic magnocellular neurones. Synchronized bursts of inhibitory synaptic inputs represent a novel mechanism that may contribute to the action potential burst generation, termination and synchronization responsible for pulsatile hormone release from neuroendocrine cells.


Subject(s)
Action Potentials/physiology , Inhibitory Postsynaptic Potentials/physiology , Neurons/physiology , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Animals , Calcium/metabolism , Calcium Channels/physiology , Electrophysiological Phenomena/physiology , Hypothalamus/cytology , Inhibitory Postsynaptic Potentials/drug effects , Male , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
16.
J Neurosci ; 29(2): 393-401, 2009 Jan 14.
Article in English | MEDLINE | ID: mdl-19144839

ABSTRACT

Glucocorticoids exert an opposing rapid regulation of glutamate and GABA synaptic inputs to hypothalamic magnocellular neurons via the activation of postsynaptic membrane-associated receptors and the release of retrograde messengers. Glucocorticoids suppress synaptic glutamate release via the retrograde release of endocannabinoids and facilitate synaptic GABA release via an unknown retrograde messenger. Here, we show that the glucocorticoid facilitation of GABA inputs is due to the retrograde release of neuronal nitric oxide and that glucocorticoid-induced endocannabinoid synthesis and nitric oxide synthesis are mediated by divergent G-protein signaling mechanisms. While the glucocorticoid-induced, endocannabinoid-mediated suppression of glutamate release is dependent on activation of the G(alpha)s G-protein subunit and cAMP-cAMP-dependent protein kinase activation, the nitric oxide facilitation of GABA release is mediated by G(beta)gamma signaling that leads to activation of neuronal nitric oxide synthase. Our findings indicate, therefore, that glucocorticoids exert opposing rapid actions on glutamate and GABA release by activating divergent G-protein signaling pathways that trigger the synthesis of, and glutamate and GABA synapse-specific retrograde actions of, endocannabinoids and nitric oxide, respectively. The simultaneous rapid stimulation of nitric oxide and endocannabinoid synthesis by glucocorticoids has important implications for the impact of stress on the brain as well as on neural-immune interactions in the hypothalamus.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Enzyme Inhibitors/pharmacology , Glucocorticoids/pharmacology , Glutamic Acid/metabolism , Nitric Oxide/metabolism , Signal Transduction/drug effects , Synapses/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Arginine/pharmacology , Cannabinoid Receptor Modulators/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dexamethasone/pharmacology , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , Hypothalamus/cytology , In Vitro Techniques , Male , Neurons/drug effects , Neurons/physiology , Nitric Oxide Donors/pharmacology , Patch-Clamp Techniques/methods , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Piperidines/pharmacology , Pyrans/pharmacology , Pyrazoles/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Rimonabant , Synapses/physiology , Synaptic Transmission/physiology , Thionucleotides/pharmacology , Valine/analogs & derivatives , Valine/pharmacology
17.
Prog Brain Res ; 170: 379-88, 2008.
Article in English | MEDLINE | ID: mdl-18655897

ABSTRACT

Glucocorticoids secreted in response to stress activation of the hypothalamic-pituitary-adrenal axis feed back onto the hypothalamus to rapidly suppress neuroendocrine activation, including oxytocin and vasopressin secretion. Here we provide a brief review focused on our recent findings of a rapid glucocorticoid-induced opposing regulation of glutamate and gamma-aminobutyric acid (GABA) inputs to magnocellular neurons via the release of distinct retrograde messengers. The stress hormone corticosterone and its synthetic analogue dexamethasone elicit the rapid retrograde release of endocannabinoids by activating a novel membrane-associated, G protein-coupled receptor in parvocellular and magnocellular neuroendocrine cells of the hypothalamic paraventricular and supraoptic nuclei. Glucocorticoids also cause the rapid retrograde release of an unknown messenger that facilitates presynaptic GABA release onto magnocellular neuroendocrine cells. These finding suggest that there is a strict synapse-specific segregation of the opposing actions of the two retrogradely released messengers. Thus, the combined actions of glucocorticoids cause a rapid synaptic inhibition of the magnocellular neurons and would be expected, therefore, to mediate a rapid feedback inhibition of the secretion of oxytocin and vasopressin during stress activation of the hypothalamic-pituitary-adrenal axis.


Subject(s)
Basal Nucleus of Meynert/physiology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Neurons/physiology , Synapses/physiology , Animals , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Glutamic Acid/physiology , Homeostasis/drug effects , Homeostasis/physiology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/drug effects , Hypothalamus/physiology , Mammals , Neurons/drug effects , Norepinephrine/pharmacology , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Synapses/drug effects , TRPV Cation Channels/drug effects , TRPV Cation Channels/physiology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/physiology
18.
Endocrinology ; 146(10): 4292-301, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15994343

ABSTRACT

Glucocorticoids secreted in response to stress activation of the hypothalamic-pituitary-adrenal axis feed back onto the brain to rapidly suppress neuroendocrine activation, including oxytocin and vasopressin secretion. Here we show using whole-cell patch clamp recordings that glucocorticoids elicit a rapid, opposing action on synaptic glutamate and gamma-aminobutyric acid (GABA) release onto magnocellular neurons of the hypothalamic supraoptic nucleus and paraventricular nucleus, suppressing glutamate release and facilitating GABA release by activating a putative membrane receptor. The glucocorticoid effect on both glutamate and GABA release was blocked by inhibiting postsynaptic G protein activity, suggesting a dependence on postsynaptic G protein signaling and the involvement of a retrograde messenger. Biochemical analysis of hypothalamic slices treated with dexamethasone revealed a glucocorticoid-induced rapid increase in the levels of the endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG). The glucocorticoid suppression of glutamate release was blocked by the type I cannabinoid receptor cannabinoid receptor antagonist, AM251, and was mimicked and occluded by AEA and 2-AG, suggesting it was mediated by retrograde endocannabinoid release. The glucocorticoid facilitation of GABA release was also blocked by AM251 but was not mimicked by AEA, 2-AG, or a synthetic cannabinoid, WIN 55,212-2, nor was it blocked by vanilloid or ionotropic glutamate receptor antagonists, suggesting that it was mediated by a retrograde messenger acting at an AM251-sensitive, noncannabinoid/nonvanilloid receptor at presynaptic GABA terminals. The combined, opposing actions of glucocorticoids mediate a rapid inhibition of the magnocellular neuroendocrine cells, which in turn should mediate rapid feedback inhibition of the secretion of oxytocin and vasopressin by glucocorticoids during stress activation of the hypothalamic-pituitary-adrenal axis.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Cholesterol/pharmacology , Corticosterone/pharmacology , Endocannabinoids , Glutamic Acid/pharmacology , Hypothalamus/physiology , Neurons/physiology , gamma-Aminobutyric Acid/pharmacology , Animals , Dexamethasone/pharmacology , Excitatory Postsynaptic Potentials , Hypothalamus/drug effects , In Vitro Techniques , Male , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
19.
J Neurosci ; 23(12): 4850-7, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12832507

ABSTRACT

Glucocorticoid negative feedback in the brain controls stress, feeding, and neural-immune interactions by regulating the hypothalamic-pituitary-adrenal axis, but the mechanisms of inhibition of hypothalamic neurosecretory cells have never been elucidated. Using whole-cell patch-clamp recordings in an acute hypothalamic slice preparation, we demonstrate a rapid suppression of excitatory glutamatergic synaptic inputs to parvocellular neurosecretory neurons of the hypothalamic paraventricular nucleus (PVN) by the glucocorticoids dexamethasone and corticosterone. The effect was maintained with dexamethasone conjugated to bovine serum albumin and was not seen with direct intracellular glucocorticoid perfusion via the patch pipette, suggesting actions at a membrane receptor. The presynaptic inhibition of glutamate release by glucocorticoids was blocked by postsynaptic inhibition of G-protein activity with intracellular GDP-beta-S application, implicating a postsynaptic G-protein-coupled receptor and the release of a retrograde messenger. The glucocorticoid effect was not blocked by the nitric oxide synthesis antagonist N(G)-nitro-L-arginine methyl ester hydrochloride or by hemoglobin but was blocked completely by the CB1 cannabinoid receptor antagonists AM251 [N-(piperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide] and AM281 [1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-4-morpholinyl-1H-pyrazole-3-carboxamide] and mimicked and occluded by the cannabinoid receptor agonist WIN55,212-2 [(beta)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone mesylate], indicating that it was mediated by retrograde endocannabinoid release. Several peptidergic subtypes of parvocellular neuron, identified by single-cell reverse transcripton-PCR analysis, were subject to rapid inhibitory glucocorticoid regulation, including corticotropin-releasing hormone-, thyrotropin-releasing hormone-, vasopressin-, and oxytocin-expressing neurons. Therefore, our findings reveal a mechanism of rapid glucocorticoid feedback inhibition of hypothalamic hormone secretion via endocannabinoid release in the PVN and provide a link between the actions of glucocorticoids and cannabinoids in the hypothalamus that regulate stress and energy homeostasis.


Subject(s)
Fatty Acids, Unsaturated/metabolism , Feedback, Physiological/physiology , Glucocorticoids/physiology , Guanosine Diphosphate/analogs & derivatives , Hypothalamus/metabolism , Animals , Cannabinoid Receptor Modulators , Corticosterone/pharmacology , Dexamethasone/pharmacology , Endocannabinoids , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Feedback, Physiological/drug effects , GABA Antagonists/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Glucocorticoids/pharmacology , Glutamic Acid/metabolism , Guanosine Diphosphate/pharmacology , Hypothalamus/drug effects , In Vitro Techniques , Male , Neurons/classification , Neurons/drug effects , Neurons/metabolism , Neurosecretory Systems/drug effects , Neurosecretory Systems/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Patch-Clamp Techniques , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid , Receptors, Drug/antagonists & inhibitors , Receptors, Drug/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Thionucleotides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL