Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
J Physiol ; 591(7): 1823-39, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23359674

ABSTRACT

Gastrin-releasing peptide (GRP) is a bombesin-like peptide with a widespread distribution in mammalian CNS, where it has a role in food intake, circadian rhythm generation, fear memory, itch sensation and sexual behaviour. While it has been established that GRP predominantly excites neurons, details of the membrane mechanism involved in this action remain largely undefined. We used perforated patch clamp recording in acute brain slice preparations to investigate GRP-affected receptors and ionic conductances in neurons of the rat paraventricular thalamic nucleus (PVT). PVT is a component of the midline and intralaminar thalamus that participates in arousal, motivational drives and stress responses, and exhibits a prominence of GRP-like immunoreactive fibres. Exposure of PVT neurons to low nanomolar concentrations of GRP induced sustained TTX-resistant membrane depolarizations that could trigger rhythmic burst discharges or tonic firing. Membrane current analyses in voltage clamp revealed an underlying postsynaptic bombesin type 2 receptor-mediated inward current that resulted from the simultaneous suppression of a Ba(2+)-sensitive inward rectifier K(+) conductance and activation of a non-selective cation conductance with biophysical and pharmacological properties reminiscent of transient receptor potential vanilloid (TRPV) 1. A role for a TRPV1-like conductance was further implied by a significant suppressant influence of a TRPV1 antagonist on GRP-induced membrane depolarization and rhythmic burst or tonic firing. The results provide a detailed picture of the receptor and ionic conductances that are involved in GRP's excitatory action in midline thalamus.


Subject(s)
Gastrin-Releasing Peptide/physiology , Midline Thalamic Nuclei/physiology , Neurons/physiology , Receptors, Bombesin/physiology , Anilides/pharmacology , Animals , Capsaicin/pharmacology , Cinnamates/pharmacology , Male , Midline Thalamic Nuclei/cytology , Potassium Channels, Inwardly Rectifying/physiology , Rats , Rats, Wistar , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/physiology
2.
J Pharmacol Exp Ther ; 336(3): 840-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21139059

ABSTRACT

Drugs that interact with group II metabotropic glutamate receptors (mGluRs) are presently being evaluated for a role in the treatment of anxiety disorders and symptoms of schizophrenia. Their mechanism of action is believed to involve a reduction in excitatory neurotransmission in limbic and forebrain regions commonly associated with these mental disorders. In rodents, the glutamatergic neurons in the midline paraventricular thalamic nucleus (PVT) provide excitatory inputs to the limbic system and forebrain. PVT also displays a high density of group II mGluRs, predominantly the metabotropic glutamate 2 receptor (mGluR2). Because the role of group II mGluRs in regulating cellular and synaptic excitability in this location has yet to be determined, we used whole-cell patch-clamp recording and acute rat brain slice preparations to evaluate PVT neuron responses to a selective group II mGluR agonist, (1R,4R,5S,6R)-4-amino-2-oxabicyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY 379268). LY 379268 consistently induced membrane hyperpolarization and suppressed firing by postsynaptic receptor-mediated activation of a barium-sensitive background K(+) conductance. This effect could be blocked by (2S)-2-amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xanth-9-yl)propanoic acid (LY 341495), a selective group II mGluR antagonist. In addition, LY 379268 acted at presynaptic receptors to reduce ionotropic glutamate receptor-mediated excitatory synaptic transmission. An mGluR2-positive allosteric modulator, 2,2,2-trifluoro-N-[4-(2-methoxyphenoxy)phenyl]-N-(3-pyridinylmethyl)ethanesulfonamide hydrochloride (LY 487379), resulted in leftward shifts of the LY 379268 dose-response curve for both postsynaptic and presynaptic actions. The data demonstrate that activation of postsynaptic and presynaptic group II (presumably mGluR2) mGluRs reduces neuronal excitability in midline thalamus, an action that may contribute to the effectiveness of mGluR2-activating drugs in rodent models of anxiety and psychosis.


Subject(s)
Midline Thalamic Nuclei/metabolism , Neurons/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, Presynaptic/metabolism , Synaptic Potentials/physiology , Animals , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Midline Thalamic Nuclei/drug effects , Neurons/drug effects , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Presynaptic/antagonists & inhibitors , Synaptic Potentials/drug effects
3.
Neuroscience ; 158(4): 1560-70, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19135504

ABSTRACT

Orexin (hypocretin) peptides are known to depolarize rat thalamic paraventricular nucleus (PVT) neurons by suppression of one or more undefined potassium conductances. Here, we investigated a contribution of TWIK-related acid-sensitive K(+) (TASK) channels to the resting membrane potential and orexin-induced depolarization of PVT neurons, using patch clamp recording techniques in brain slice preparations. Upon exposure to an acidic (pH 6.3) extracellular solution, PVT neurons displayed membrane depolarization. Under voltage-clamp and in the presence of tetrodotoxin (TTX, 0.5 microM), low pH solutions induced an inward shift in baseline membrane current, accompanied by a net decrease in membrane conductance, reversing close to the potassium equilibrium potential. By contrast, exposure to alkaline (pH 8.3) solutions resulted in membrane hyperpolarization, induced an outward shift in baseline membrane current and an increase in net conductance that reversed close to the potassium equilibrium potential. A local anesthetic bupivacaine (20-40 microM) and the endocannabinoid anandamide (5-10 microM) mimicked the effects of the acidic solution. Exposure to the volatile anesthetic isoflurane (0.2-0.5 mM) induced changes in resting membrane potential, baseline current and membrane conductance similar to those caused by the alkaline solution. Although responsiveness to orexins was preserved under each of the above conditions, the amplitude of the orexin B (0.5 microM)-induced inward current was depressed in the acidic solution and in the presence of anandamide, remained largely unchanged in the alkaline solution, and was enhanced by isoflurane when compared with that in normal artificial cerebrospinal solution. We conclude that pH-sensitive potassium channels, TASK-1 and TASK-3 channels, contribute substantially to the resting membrane conductance(s) and excitability in PVT neurons. The observations that orexin-induced currents were affected by putative TASK-specific drugs in a manner predictable from their effects on TASK channels also suggest that the orexin-induced excitation in PVT neurons is mediated by closure of TASK channels.


Subject(s)
Intracellular Signaling Peptides and Proteins/pharmacology , Membrane Potentials/drug effects , Midline Thalamic Nuclei/cytology , Neurons/physiology , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Potassium Channels, Tandem Pore Domain/physiology , Acids/pharmacology , Alkalies/pharmacology , Anesthetics, Local/pharmacology , Animals , Animals, Newborn , Arachidonic Acids/pharmacology , Bupivacaine/pharmacology , Calcium Channel Blockers/pharmacology , Electric Stimulation/methods , Endocannabinoids , Extracellular Space/metabolism , Isoflurane/pharmacology , Membrane Potentials/physiology , Nerve Tissue Proteins , Neurons/drug effects , Orexins , Patch-Clamp Techniques , Polyunsaturated Alkamides/pharmacology , Potassium/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Rats , Rats, Wistar , Tetrodotoxin/pharmacology
4.
Science ; 186(4163): 538-40, 1974 Nov 08.
Article in English | MEDLINE | ID: mdl-4469678

ABSTRACT

Sequential blood samples, obtained from freely behaving, nonstressed male rats, showed a pulsatile pattern of growth hormone secretion with a mean interval between peaks of 68 minutes. The bursts of secretion were blocked by lesions of the hypothalamic ventromedial nuclei and by administration of a longacting preparation of synthetic somatostatin.


Subject(s)
Growth Hormone/metabolism , Hypothalamus, Middle/physiology , Hypothalamus/physiology , Somatostatin/pharmacology , Animals , Corticosterone/blood , Growth Hormone/blood , Male , Rats , Time Factors
5.
Science ; 205(4412): 1275-7, 1979 Sep 21.
Article in English | MEDLINE | ID: mdl-224461

ABSTRACT

The microiontophoretic application of thyrotropin-releasing hormone causes a selective reduction in neuronal excitation evoked by L-glutamate but not by acetylcholine in rat cerebral cortex. Thyrotropin-releasing hormone has no influence on the activity of acetylcholinesterase or on choline uptake and release from cerebral synaptosomes. This evidence for a selective interaction between a centrally acting peptide and an excitatory amino acid neurotransmitter may indicate a specific locus of thyrotropin-releasing hormone action at glutamate-activated receptor sites.


Subject(s)
Cerebral Cortex/drug effects , Excitatory Amino Acid Antagonists , Thyrotropin-Releasing Hormone/pharmacology , Acetylcholine/metabolism , Action Potentials/drug effects , Animals , Aspartic Acid/antagonists & inhibitors , Cerebral Cortex/physiology , Male , Rats , Receptors, Neurotransmitter/drug effects , Synaptic Transmission/drug effects
6.
Neuroscience ; 155(4): 1212-20, 2008 Sep 09.
Article in English | MEDLINE | ID: mdl-18674591

ABSTRACT

Subpopulations of neurons in the median preoptic nucleus (MnPO) located within the lamina terminalis contribute to thermoregulatory, cardiovascular and hydromineral homeostasis, and sleep-promotion. MnPO is innervated by lateral hypothalamic neurons that synthesize and secrete the arousal-promoting and excitatory orexin (hypocretin) neuropeptides. To evaluate the hypothesis that orexins modulate the excitability of MnPO neurons, we used patch-clamp recording techniques applied in rat brain slice preparations to assess the effects of exogenously applied orexin A and orexin B peptides on their intrinsic and synaptic properties. Whole cell recordings under current-clamp mode revealed that 11/15 tested MnPO neurons responded similarly to either orexin A or B (500-1000 nM) with a slowly rising, prolonged (10-15 min) and reversible membrane depolarization. Under voltage-clamp mode, orexin applications induced a tetrodotoxin-resistant inward current of -7.2+/-1.6 pA, indicating a direct (postsynaptic) activation, with a time course similar to the observed membrane depolarization. The orexin-induced responses in 4/7 neurons were associated with a significant decrease in membrane conductance and the net orexin-induced current that reversed at -99+/-5 mV, suggesting closure of potassium channels. Orexins did not attenuate the properties of excitatory (n=4) or inhibitory (n=7) postsynaptic currents evoked by subfornical organ stimulation. By contrast, orexins applications induce a significant increase in both frequency and amplitude of spontaneous glutamatergic postsynaptic currents (5/7 cells) but had no influence on spontaneous GABAergic currents (6/6 cells). Thus, in addition to a direct postsynaptic receptor-mediated excitation, orexins can also increase the excitability of MnPO neurons via increasing their excitatory inputs, presumably through an orexin receptor-mediated excitation of local glutamatergic neurons whose axons project to MnPO neurons.


Subject(s)
Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Neurons/drug effects , Neuropeptides/pharmacology , Preoptic Area/cytology , Synapses/drug effects , Afferent Pathways/drug effects , Afferent Pathways/physiology , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Male , Neuropeptides/metabolism , Orexins , Patch-Clamp Techniques/methods , Rats , Rats, Wistar , Synapses/physiology
7.
Neuroscience ; 147(4): 1066-75, 2007 Jul 29.
Article in English | MEDLINE | ID: mdl-17600629

ABSTRACT

The thalamic paraventricular nucleus (PVT) receives a dense innervation from orexin-synthesizing lateral hypothalamic neurons. Since PVT neurons display state-dependent tonic or low threshold spike-driven burst firing patterns, we examined how the response to exogenously applied orexins might modulate these features. Data were obtained with whole-cell patch clamp recording techniques in rat brain slices prepared during the subjective lights-on period. PVT neurons displayed a mean resting membrane potential of -61+/-6 mV and input conductance of 1.3+/-0.1 nS (n=60). The majority (90/107) of cells tested responded to orexin A and/or orexin B peptides (100-1000 nM), each inducing similar slowly rising and prolonged membrane depolarizations. We next evaluated associated changes in firing patterns and action potential frequency. Of 17 spontaneously silent neurons, 5 were induced into tonic firing and 4 into burst firing modes. Of nine spontaneously bursting neurons, three displayed an increase in burst frequency and in the number of action potentials within a burst. By contrast, another six cells were induced into tonic firing mode, with a marked decrease in instantaneous firing frequency and a shift in their excitatory postsynaptic potential-evoked responses from burst firing patterns to single action potentials. Under voltage clamp, orexins induced inward current (-21.8+/-2.4 pA at -60 mV) in 20/22 cells. In 13 cells, current-voltage (I-V) plots revealed a decrease in net conductance and reversal at -110+/-9 mV, while 3 cells displayed an increase in net conductance that reversed at -26+/-8 mV. These observations imply suppression of potassium and/or induction of nonselective cationic conductances in orexin-induced depolarization in PVT neurons, permitting these peptides to modulate intrinsic state-dependent properties. In vivo, such changes in firing patterns and frequency of action potential discharges could influence neurotransmission through PVT and activity-dependent synaptic plasticity at target sites of these neurons.


Subject(s)
Action Potentials/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Midline Thalamic Nuclei/cytology , Neurons/physiology , Neuropeptides/pharmacology , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Neurons/drug effects , Orexins , Patch-Clamp Techniques , Rats , Rats, Wistar
8.
Neuroscience ; 144(3): 815-24, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17137725

ABSTRACT

The role of hypothalamic ATP-sensitive potassium channels in the maintenance of energy homeostasis has been extensively explored. However, how these channels are incorporated into the neuronal networks of the arcuate nucleus remains unclear. Whole-cell patch-clamp recordings from rat arcuate nucleus neurons in hypothalamic slice preparations revealed widespread expression of functional ATP-sensitive potassium channels within the nucleus. ATP-sensitive potassium channels were expressed in orexigenic neuropeptide Y/agouti-related protein (NPY/AgRP) and ghrelin-sensitive neurons and in anorexigenic cocaine-and-amphetamine regulated transcript (CART) neurons. In 70% of the arcuate nucleus neurons recorded, exposure to glucose-free bathing medium induced inhibition of electrical excitability, the response being characterized by membrane hyperpolarization, a reduction in neuronal input resistance and a reversal potential consistent with opening of potassium channels. These effects were reversible upon re-introduction of glucose to the bathing medium or upon exposure to the ATP-sensitive potassium channel blockers tolbutamide or glibenclamide. The potassium channel opener diazoxide, but not pinacidil, also induced a tolbutamide and glibenclamide-sensitive inhibition of electrical excitability. Single-cell reverse transcription-polymerase chain reaction revealed expression of mRNA for sulfonylurea receptor 1 but not sulfonylurea receptor 2 subunits of ATP-sensitive potassium channels. Thus, rat arcuate nucleus neurons, including those involved in functionally antagonistic orexigenic and anorexigenic pathways express functional ATP-sensitive potassium channels which include sulfonylurea receptor 1 subunits. These data indicate a crucial role for these ion channels in central sensing of metabolic and energy status. However, further studies are needed to clarify the differential roles of these channels, the organization of signaling pathways that regulate them and how they operate in functionally opposing cell types.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Agouti-Related Protein , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Ghrelin , Glucose/metabolism , Glucose/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Multidrug Resistance-Associated Proteins/genetics , Neurons/drug effects , Organ Culture Techniques , Patch-Clamp Techniques , Peptide Hormones/metabolism , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Drug , Sulfonylurea Receptors
9.
J Neuroendocrinol ; 29(10)2017 10.
Article in English | MEDLINE | ID: mdl-28834571

ABSTRACT

Obesity and ageing are risk factors for diabetes. In the present study, we investigated the effects of ageing, obesity and fasting on central and peripheral glucose tolerance and on glucose-sensing neuronal function in the arcuate nucleus of rats, with a view to providing insight into the central mechanisms regulating glucose homeostasis and how they change or are subject to dysfunction with ageing and obesity. We show that, following a glucose load, central glucose tolerance at the level of the cerebrospinal fluid (CSF) and plasma is significantly reduced in rats maintained on a high-fat diet (HFD). With ageing, up to 2 years, central glucose tolerance was impaired in an age-dependent manner, whereas peripheral glucose tolerance remained unaffected. Ageing-induced peripheral glucose intolerance was improved by a 24-hour fast, whereas central glucose tolerance was not corrected. Pre-wean, immature animals have elevated basal plasma glucose levels and a delayed increase in central glucose levels following peripheral glucose injection compared to mature animals. Electrophysiological recording techniques revealed an energy-status-dependent role for glucose-excited, inhibited and adapting neurones, along with glucose-induced changes in synaptic transmission. We conclude that ageing affects central glucose tolerance, whereas HFD profoundly affects central and peripheral glucose tolerance and, in addition, glucose-sensing neurones adapt function in an energy-status-dependent manner.


Subject(s)
Aging , Arcuate Nucleus of Hypothalamus/metabolism , Diet, High-Fat , Fasting , Glucose/metabolism , Animals , Blood Glucose , Glucose/administration & dosage , Glucose/cerebrospinal fluid , Homeostasis , Male , Neurons/physiology , Rats, Wistar
10.
Neuroscience ; 141(4): 2059-66, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16797851

ABSTRACT

The hypothalamic suprachiasmatic nucleus uniquely projects to the midline thalamic paraventricular nucleus. To characterize this projection, patch clamp techniques applied in acute rat brain slice preparations examined responses of anterior thalamic paraventricular nucleus neurons to focal suprachiasmatic nucleus stimulation. Whole cell recordings from slices obtained during daytime (n=40) revealed neurons with a mean membrane potential of -66+/-1.2 mV, input conductance of 1.5+/-0.1 nS and state-dependent tonic or burst firing patterns. Electrical stimulation (one or four pulses) in suprachiasmatic nucleus elicited monosynaptic excitatory postsynaptic potentials (mean latency of 12.6+/-0.6 ms; n=12), featuring both AMPA and N-methyl-D-aspartate-glutamate receptor-mediated components, and monosynaptic bicuculline-sensitive inhibitory postsynaptic potentials (mean latency of 16.6+/-0.6 ms; n=7) reversing polarity at -72+/-2.6 mV, close to the chloride equilibrium potential. Glutamate microstimulation of suprachiasmatic nucleus also elicited transient increases in spontaneous excitatory or inhibitory postsynaptic currents in anterior thalamic paraventricular neurons. Recordings from rats under reverse light/dark conditions (n=22) yielded essentially similar responses to electrical stimulation. At depolarized membrane potentials, suprachiasmatic nucleus-evoked excitatory postsynaptic potentials triggered single action potentials, while evoked inhibitory postsynaptic potentials elicited a silent period in ongoing tonic firing. By contrast, after manual adjustment of membrane potentials to hyperpolarized levels, neuronal response to the same "excitatory" stimulus was a low threshold spike and superimposed burst firing, while responses to "inhibitory" stimuli paradoxically elicited excitatory rebound low threshold spikes and burst firing. These data support the existence of glutamatergic and GABAergic efferents from the suprachiasmatic nucleus to its target neurons. Additionally, in thalamic paraventricular nucleus neurons, responses to activation of their suprachiasmatic afferents may vary in accordance with their membrane potential-dependent intrinsic properties, a characteristic typical of thalamocortical neurons.


Subject(s)
Glutamic Acid/metabolism , Midline Thalamic Nuclei/cytology , Neurons/physiology , Suprachiasmatic Nucleus/cytology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Bicuculline/pharmacology , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Neurons/drug effects , Patch-Clamp Techniques/methods , Quinoxalines/pharmacology , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptic Transmission/radiation effects
11.
Neuroscience ; 322: 308-19, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26924019

ABSTRACT

In rat paraventricular thalamic nucleus (PVT) neurons, activation of low-threshold calcium (Ca(2+)) channels triggers a low-threshold spike (LTS) which may be followed by slow afterpotentials that can dramatically influence action potential patterning. Using gluconate-based internal recording solutions, we investigated the properties of a LTS-induced slow afterdepolarization (sADP) observed in a subpopulation of PVT neurons recorded in brain slice preparations. This LTS-induced sADP required T-type Ca(2+) channel opening, exhibited variable magnitudes between neurons and a voltage dependency with a maximum near -50 mV. The area under the sADP remained stable during control monitoring, but displayed gradual suppression in media where strontium replaced Ca(2+). The sADP was suppressed following bath application of 2-APB or ML204, suggesting engagement of transient receptor potential canonical (TRPC)-like channels. Further investigation revealed a reversible suppression during bath applications of membrane permeable cannabinoid receptor (CBR) blockers rimonabant, AM630 or SR144528 suggesting the presence of both CB1Rs and CB2Rs. Similar results were achieved by intracellular, but not bath application of the membrane impermeant CB1R blocker hemopressin, suggesting an intracellular localization of CB1Rs. Data from pharmacologic manipulation of endocannabinoid biosynthetic pathways suggested 2-arachidonlyglycerol (2-AG) as the endogenous cannabinoid ligand, derived via hydrolysis of diacylglycerol (DAG), with the latter formed from the pathway involving phosphatidylcholine-specific phospholipase D and phosphatic acid phosphohydrolase. The sADP suppression observed during recordings with pipettes containing LY294002, a PI3-kinase inhibitor, suggested a role for PI3kinase in the translocation of these TRPC-like channels to the plasma membrane. Drug-induced attenuation of the availability of 2-AG influences the number of action potentials that surmount the LTS evoked in PVT neurons, implying an ongoing intracellular CBR modulation of neuronal excitability during LTS-induced bursting behavior.


Subject(s)
Arachidonic Acids/metabolism , Calcium/metabolism , Endocannabinoids/metabolism , Glycerides/metabolism , Membrane Potentials/physiology , Midline Thalamic Nuclei/physiology , Neurons/physiology , Receptors, Cannabinoid/metabolism , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Diglycerides/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Lipoprotein Lipase/metabolism , Membrane Potentials/drug effects , Midline Thalamic Nuclei/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase D/metabolism , Rats, Wistar , Tissue Culture Techniques
12.
Neuroscience ; 311: 81-91, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26470810

ABSTRACT

In rat thalamic paraventricular nucleus of thalamus (PVT) neurons, activation of thyrotropin-releasing hormone (TRH) receptors enhances excitability via concurrent decrease in G protein-coupled inwardly-rectifying potassium (GIRK)-like and activation of transient receptor potential cation (TRPC)4/5-like cationic conductances. An exploration of intracellular signaling pathways revealed the TRH-induced current to be insensitive to phosphatidylinositol-specific phospholipase C (PI-PLC) inhibitors, but reduced by D609, an inhibitor of phosphatidylcholine-specific PLC (PC-PLC). A corresponding change in the I-V relationship implied suppression of the cationic component of the TRH-induced current. Diacylglycerol (DAG) is a product of the hydrolysis of PC. Studies focused on the isolated cationic component of the TRH-induced response revealed a reduction by RHC80267, an inhibitor of DAG lipase, the enzyme involved in the hydrolysis of DAG to the endocannabinoid 2-arachidonoylglycerol (2-AG). Further investigation revealed enhancement of the cationic component in the presence of either JZL184 or WWL70, inhibitors of enzymes involved in the hydrolysis of 2-AG. A decrease in the TRH-induced response was noted in the presence of rimonabant or SR144528, membrane permeable CB1 and CB2 receptor antagonists, respectively. A decrease in the TRH-induced current by intracellular, but not by bath application of the membrane impermeable peptide hemopressin, selective for CB1 receptors, suggests a postsynaptic intracellular localization of these receptors. The TRH-induced current was increased in the presence of arachidonyl-2'-chloroethylamide (ACEA) or JWH133, CB1 and CB2 receptor agonists, respectively. The PI3-kinase inhibitor LY294002, known to inhibit TRPC translocation, decreased the response to TRH. In addition, a TRH-induced enhancement of the low-threshold spike was prevented by both rimonabant, and SR144528. TRH had no influence on excitatory or inhibitory miniature postsynaptic currents, suggesting presynaptic CB receptors are not involved in this situation. Collectively, the data imply that activation of TRH receptors in these midline thalamic neurons engages novel signaling pathways that include postsynaptic intracellular CB1 and CB2 receptors in the activation of TRPC4/5-like channels.


Subject(s)
Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , TRPC Cation Channels/metabolism , Animals , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Patch-Clamp Techniques , Rats, Wistar , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/metabolism , Tissue Culture Techniques
13.
Endocrinology ; 98(4): 991-6, 1976 Apr.
Article in English | MEDLINE | ID: mdl-1278103

ABSTRACT

Concurrent 5-hour profiles of growth hormone (GH) secretion and sleep phases were obtained in 7 rats chronically implanted with right atrial cannulae, and electroencephalographic electromyographic, and electroculographic electrodes. Hormone profiles confirmed pulsatile secretion of GH. Secretory GH episodes occurred every 3 to 4 hours and peaks generally exceeded 100 ng/ml, and, in 14 of the 21 troughs recorded, GH was unmeasurable (less than 1 ng/ml). The comparison of hormone profiles and concurrent sleep patterns excluded a temporal relationship between episodic GH secretion and sleep cycles, while scattergrams of hormone values plotted against preceding sleep phase durations also failed to demonstrate a relationship.


Subject(s)
Growth Hormone/physiology , Sleep Stages/physiology , Animals , Brain/physiology , Electrodes, Implanted , Electrophysiology , Heart/physiology , Male , Muscles/physiology , Rats , Time Factors
14.
J Comp Neurol ; 340(3): 381-91, 1994 Feb 15.
Article in English | MEDLINE | ID: mdl-8188857

ABSTRACT

GABAergic projections of the suprachiasmatic nucleus (SCN) were demonstrated in a double-labelling ultrastructural study which visualised the efferents of the SCN by PHA-L tracing, diaminobenzidine (DAB) immunocytochemistry, and GABA with immunogold postembedding staining. The results show a strong contralateral projection of the SCN that is partly GABA-containing. In addition, ipsilateral SCN projections to the dorsomedial hypothalamus and periventricular part of the paraventricular nucleus and sub-paraventricular nucleus were shown to contain GABA. The present results indicate that the SCN may utilize this inhibitory neurotransmitter to regulate and organize its own circadian rhythm as well as using GABA to transmit its diurnal information to other regions of the brain.


Subject(s)
Neurons/ultrastructure , Suprachiasmatic Nucleus/ultrastructure , gamma-Aminobutyric Acid/physiology , 3,3'-Diaminobenzidine , Animals , Circadian Rhythm/physiology , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Immunohistochemistry , Male , Microscopy, Electron , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/ultrastructure , Phytohemagglutinins , Rats , Rats, Sprague-Dawley
15.
J Comp Neurol ; 282(1): 15-23, 1989 Apr 01.
Article in English | MEDLINE | ID: mdl-2708591

ABSTRACT

Supraoptic nucleus (SON) neurons receive a prominent gamma-amino-butyric acid (GABA) input. This study evaluated the hypothesis, partly on the basis of recent electrophysiological data, that this innervation might arise from GABAergic neurons located in the ventral diagonal band of Broca area. For retrograde transport studies, pentobarbital-anesthetized male Long-Evans rats received 0.03-0.20-microliter injections of a suspension of rhodamine tagged latex microspheres into the SON. In two cases where such injections were confined to the SON, less than 60 retrogradely labeled neurons were detected in the ipsilateral diagonal band. In three animals where injections extended into the perinuclear zone around the SON, more than 2,000 retrogradely labeled cells were counted in the ipsilateral diagonal band. For anterograde transport studies, another group of animals received either 30% horseradish peroxidase (HRP) in 0.5% poly-L-ornithine (0.05-0.10 microliter injections) or Phaseolus vulgarus (iontophoresed from a 2% solution) into the diagonal band. After survivals of 18-24 hours (HRP) or 5 days (PHAL-L) labeled axon terminals invested the perinuclear zone above the SON. The presence of just a single fiber within the nucleus indicated a minor projection to the SON itself. The HRP-injected material was processed for ultrastructural examination and revealed dense HRP-labeled axon terminals in this perinuclear zone, most often (98%) forming axodendritic appositions. A postembedding colloidal gold technique to visualize GABA-synthesizing terminals revealed that fewer than 5% of these perinuclear HRP-labeled terminals also exhibited GABA-like immunoreactivity. Within the SON, where GABAergic axon terminals are abundant, few (less than 5%) GABAergic terminals contained HRP.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Frontal Lobe/cytology , Supraoptic Nucleus/cytology , gamma-Aminobutyric Acid/metabolism , Animals , Frontal Lobe/metabolism , Frontal Lobe/ultrastructure , Horseradish Peroxidase , Male , Microscopy, Electron , Phytohemagglutinins , Rats , Supraoptic Nucleus/metabolism , Supraoptic Nucleus/ultrastructure
16.
Neuropharmacology ; 41(4): 472-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11543767

ABSTRACT

The median preoptic (MnPO) nucleus, a key CNS site for hydromineral and cardiovascular homeostasis, receives a dense norepinephrine innervation from brainstem autonomic centers. Since norepinephrine is known to influence neuronal excitability by modulating calcium channel function, we applied whole cell patch clamp techniques to study calcium currents in 116 dissociated MnPO neurons, including 30 cells identified by a retrograde label as projecting to the hypothalamic paraventricular nucleus. Norepinephrine (3-50 microM) suppressed high-voltage-activated calcium currents (HVA I(Ca)) in 80% of cells, selectively blockable by yohimbine and mimicked by UK14,304 and clonidine. The norepinephrine effect was relieved by strong prior depolarization, indicating a voltage-dependent component. Intracellular GTP-gamma-S blocked the effect. Blockade by extracellular NEM suggested involvement of pertussis-toxin sensitive G-proteins. Based on pharmacological properties, these HVA I(Ca)s had the following composition: 40-45% N-type (blockable by omega-conotoxin GVIA); 20-25% L-type (blockable by nimodipine); 15-20% P/Q-type (blockable by omega-agatoxin IVA). Since approximately 75% of the norepinephrine effect was blockable with omega-conotoxin GVIA, we conclude that postsynaptic alpha(2) adrenoceptors preferentially suppress N-type calcium channels, revealing a novel mechanism whereby norepinephrine can modulate excitability in MnPO neurons.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Neurons/metabolism , Norepinephrine/pharmacology , Preoptic Area/drug effects , Receptors, Adrenergic, alpha-2/drug effects , Adrenergic alpha-Antagonists/pharmacology , Animals , Electrophysiology , GTP-Binding Proteins/metabolism , Male , Neurons/drug effects , Preoptic Area/cytology , Rats , Rats, Long-Evans , omega-Conotoxin GVIA/pharmacology
17.
Neuroscience ; 56(4): 823-32, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8284036

ABSTRACT

The suprachiasmatic nucleus in the anterior hypothalamus contains a circadian oscillator that is responsible for 24-h rhythms in several behavioral, endocrine and autonomic processes. Efferent suprachiasmatic projections are likely to transmit rhythmic information to brain nuclei controlling these functions. The hypothalamic paraventricular nucleus is considered to be a target of the suprachiasmatic nucleus due to its important role in autonomic and endocrine regulation. The present study applied extracellular electrophysiological techniques to intact animals to look for a possible interaction between suprachiasmatic nucleus efferents and identified neurons in the hypothalamic paraventricular nucleus. Results showed that electrical stimulation of the suprachiasmatic nucleus induced an increase in the excitability of 87% of paraventricular neurons that project to the median eminence and are situated in the medial and dorsal parvocellular subnucleus; neurons with similar projections but located in the periventricular subnucleus displayed a reduction in firing rate following suprachiasmatic stimulation. Electrical activation of the suprachiasmatic nucleus provoked a decrease in excitability in 75% of paraventricular neurons in the posterior magnocellular subnucleus that send axons to the posterior pituitary and in 85% of paraventricular neurons, located in the medial parvocellular subnucleus, that project to the dorsal vagus complex in the brainstem. The data imply that functional and selective neural connections exist between suprachiasmatic nucleus efferents and specific cell groups within the hypothalamic paraventricular nucleus. These projections would be able to convey rhythmic information to certain endocrine and autonomic functions. The anatomical and neurochemical characteristics of the underlying pathways remain to be determined.


Subject(s)
Circadian Rhythm/physiology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Suprachiasmatic Nucleus/physiology , Afferent Pathways/physiology , Animals , Brain Stem/physiology , Electric Stimulation , Male , Neurons/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Rats , Vasopressins/metabolism
18.
Neuroscience ; 83(3): 905-16, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9483573

ABSTRACT

The median preoptic nucleus is recognized as an important forebrain site involved in hydromineral and cardiovascular homeostasis. In the present study, whole cell patch-clamp recordings in parasagittal slices of adult rat brain were used to obtain information on the properties of median preoptic neurons. Lucifer Yellow-labelled cells demonstrated small ovoid somata with two to three aspiny main dendrites and axons that branched sparingly. Median preoptic neurons displayed varying degrees of hyperpolarization-activated time-dependent and/or time-independent inward rectification, and 86% of cells demonstrated low threshold spikes. Median preoptic nucleus is known to receive a prominent noradrenergic innervation from the medulla, and 59% of 156 tested neurons were found to respond to bath applied noradrenaline (1-100 microM). In the majority (n = 62) of cells, the response was an alpha 2 adrenoreceptor-mediated, tetrodotoxin-resistant, membrane hyperpolarization that was associated with a 43 +/- 6% increase in membrane conductance. The net noradrenaline-induced current (5-45 pA) was inwardly rectifying, cesium-resistant but barium sensitive. Current reversal at -102 +/- 4 mV in 3.1 mM [K]o and -62 +/- 3 mV in 10 mM [K]o implied opening of potassium channels. By contrast, a minority (n = 27) of cells responded to noradrenaline with an alpha 1-mediated, tetrodotoxin-resistant membrane depolarization. These observations imply a functional diversity among median preoptic neurons, and the prevalence of hyperpolarizing alpha 2 and, to a lesser extent, depolarizing alpha 1 adrenoreceptors on median preoptic neurons suggests that noradrenergic inputs can exert a prominent influence on their cellular excitability.


Subject(s)
Adrenergic alpha-Agonists/metabolism , Neurons/physiology , Norepinephrine/metabolism , Prosencephalon/physiology , Receptors, Adrenergic, alpha/metabolism , Adrenergic alpha-Agonists/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Cerebral Ventricles , Electrophysiology , Male , Neurons/drug effects , Neurons/metabolism , Norepinephrine/pharmacology , Patch-Clamp Techniques , Prosencephalon/cytology , Prosencephalon/metabolism , Rats , Rats, Inbred Strains
19.
Neuroscience ; 88(3): 939-47, 1999.
Article in English | MEDLINE | ID: mdl-10363829

ABSTRACT

Recent investigations in the rat have implicated a noradrenergic innervation to the horizontal nucleus of the diagonal band of Broca as a critical link in a neural circuit that conveys baroreceptor information centrally to inhibit the firing of vasopressin-secreting neurons in the hypothalamic supraoptic nucleus. In this study we used small intra-diagonal band injections of a retrograde tracer, rhodamine latex microspheres, in combination with tyrosine hydroxylase histochemistry to identify brainstem noradrenergic cells contributing to this innervation. In three cases where tracer injections were limited to the horizontal limb of the diagonal band, we observed 20-50 double-labelled neurons ipsilaterally in the dorsal part of the locus coeruleus (A6) and the caudal nucleus tractus solitarius (A2), and bilaterally in the caudal ventrolateral medulla (A1). Double-labelled neurons were also noted in the ventral tegmental area (dopaminergic A10 cell group). Although all major brainstem noradrenergic cell groups contribute fibers to the horizontal limb of the nucleus of diagonal band, data from physiological studies suggest that the noradrenergic A2 neurons in the nucleus tractus solitarius are the most likely pathway through which it receives this baroreceptor information.


Subject(s)
Medulla Oblongata/anatomy & histology , Medulla Oblongata/physiology , Neurons/cytology , Pons/anatomy & histology , Pons/physiology , Animals , Axonal Transport , Functional Laterality , Image Processing, Computer-Assisted , Immunohistochemistry , Locus Coeruleus/anatomy & histology , Locus Coeruleus/physiology , Male , Models, Neurological , Neural Pathways/anatomy & histology , Neural Pathways/physiology , Neurons/physiology , Norepinephrine/physiology , Pressoreceptors/physiology , Rats , Rats, Long-Evans , Rhodamines , Tyrosine 3-Monooxygenase/analysis
20.
Neuroscience ; 118(1): 49-58, 2003.
Article in English | MEDLINE | ID: mdl-12676136

ABSTRACT

The suprachiasmatic nucleus (SCN), the dominant circadian pacemaker in mammalian brain, sends axonal projections to the hypothalamic paraventricular nucleus (PVN), a composite of magno- and parvocellular neurons. This neural network likely offers SCN output neurons a means to entrain diurnal rhythmicity in various autonomic and neuroendocrine functions. Earlier investigations using patch-clamp recordings in slice preparations have suggested differential innervation by SCN efferents to magnocellular versus parvocellular PVN cells. In magnocellular PVN, cells respond to focal electrical stimulation in SCN with a GABA(A) receptor-mediated postsynaptic inhibition whose magnitude can be modulated by presynaptic GABA(B) receptors. By contrast, SCN-evoked responses in parvocellular PVN neurons typically involve both GABA(A)- and glutamate-receptor-mediated components. In the present patch-clamp study, 69/85 periventricular parvocellular PVN cells displayed SCN-evoked inhibitory and/or excitatory postsynaptic currents (IPSCs; EPSCs). In the presence of selective receptor antagonists, we sought evidence for their modulation by GABA acting at pre- and/or postsynaptic GABA(B) receptors. Cells responded to bath-applied baclofen (5-10 microM) with a tetrodotoxin-resistant membrane hyperpolarization associated with a reduction in input resistance and/or outward current, due to increase in a potassium conductance, blockable with 2-hydroxysaclofen (300 microM). At 1 microM where baclofen had no significant postsynaptic effect, evidence of activation of presynaptic GABA(B) receptors included reduction in SCN-evoked IPSCs and EPSCs with no change in their kinetics, and paired-pulse depression that was sensitive to both baclofen and saclofen. Baclofen also induced significant reductions in frequency but not amplitudes of miniature IPSCs and EPSCs. These observations suggest that levels of synaptically released GABA from the terminals of SCN output neurons can influence the relative contribution of pre- versus postsynaptic GABA(B) receptors in modulating both excitatory and inhibitory SCN innervation to parvocellular PVN neurons.


Subject(s)
Neural Pathways/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Presynaptic Terminals/metabolism , Receptors, GABA-B/metabolism , Suprachiasmatic Nucleus/metabolism , Synaptic Transmission/physiology , Animals , Circadian Rhythm/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , Male , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Pathways/drug effects , Neurosecretory Systems/drug effects , Neurosecretory Systems/physiology , Organ Culture Techniques , Paraventricular Hypothalamic Nucleus/drug effects , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Rats , Rats, Long-Evans , Reaction Time/drug effects , Reaction Time/physiology , Receptors, GABA-B/drug effects , Suprachiasmatic Nucleus/drug effects , Synaptic Membranes/drug effects , Synaptic Membranes/metabolism , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL