Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Neuroscience ; 165(4): 1377-89, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-19961906

ABSTRACT

Retrograde synaptic signaling by endogenous cannabinoids (endocannabinoids) is a recently discovered form of neuromodulation in various brain regions. In hippocampus, it is well known that endocannabinoids suppress presynaptic inhibitory neurotransmitter release in CA1 region. However, endocannabinoid signaling in CA3 region remains to be examined. Here we investigated whether presynaptic inhibition can be caused by activation of postsynaptic group I metabotropic glutamate receptors (mGluRs) and following presynaptic cannabinoid receptor type 1 (CB1 receptor) using mechanically dissociated rat hippocampal CA3 pyramidal neurons with adherent functional synaptic boutons. Application of group I mGluR agonist (RS)-3,5-dihydroxyphenylglycine (DHPG) reversibly suppressed spontaneous inhibitory postsynaptic currents (IPSCs). In the presence of tetrodotoxin (TTX), frequency of miniature IPSCs was significantly reduced by DHPG, while there were no significant changes in minimum quantal size and sensitivity of postsynaptic GABA(A) receptors to the GABA(A) receptor agonist muscimol, indicating that this suppression was caused by a decrease in GABA release from presynaptic nerve terminals. Application of CB1 synthetic agonist WIN55212-2 (mesylate(R)-(+)-[2,3-dihydro-5-methyl-3-[4-morpholino)methyl]pyrrolo-[1,2,3-de]-1,4-benzoxazin-6-yl](1-naphthyl)methanone) or endocannabinoid 2-arachidonoylglycerol also suppressed the spontaneous IPSC. The inhibitory effect of DHPG on spontaneous IPSCs was abolished by SR-141716 (5-(4-chlorophenyl)-1-(2,4-dichloro-phenyl)-4-methyl-N-(piperidin-1-yl)-1H-pyrazole-3-carboxamide), a CB1 receptor antagonist. Furthermore, postsynaptic application of GDP-betaS blocked the DHPG-induced inhibition of spontaneous IPSCs, indicating the involvement of endcannabinoid-mediated retrograde synaptic signaling. These results provide solid evidence for retrograde signaling from postsynaptic group I mGluRs to presynaptic CB1 receptors, which induces presynaptic inhibition of GABA release in rat hippocampal CA3 region.


Subject(s)
CA3 Region, Hippocampal/metabolism , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Pyramidal Cells/physiology , Receptors, Metabotropic Glutamate/metabolism , Synapses/physiology , gamma-Aminobutyric Acid/metabolism , Animals , CA3 Region, Hippocampal/drug effects , GABA-A Receptor Agonists , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Neurons/drug effects , Neurons/physiology , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Pyramidal Cells/drug effects , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Metabotropic Glutamate 5 , Receptors, GABA-A/metabolism , Receptors, Metabotropic Glutamate/agonists , Signal Transduction/drug effects , Sodium Channels/metabolism , Synapses/drug effects
2.
Neuroscience ; 146(1): 190-201, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17317016

ABSTRACT

The high potassium-induced potentiation of spontaneous glycine release in extracellular Ca2+-free conditions was studied in mechanically dissociated rat spinal dorsal horn neurons using whole-cell patch-clamp technique. Elevating extracellular K+ concentration reversibly increased the frequency of spontaneous glycinergic inhibitory postsynaptic currents (IPSCs) in the absence of extracellular Ca2+. Blocking voltage-dependent Na+ channels (tetrodotoxin) and Ca2+ channels (nifedipine and omega-grammotoxin-SIA) had no effect on this potassium-induced potentiation of glycine-release. The high potassium-induced increase in IPSC frequency was also observed in the absence of extracellular Na+, although the recovery back to baseline levels of release was prolonged under these conditions. The action of high potassium solution on glycine release was prevented by BAPTA-AM, by depletion of intracellular Ca2+ stores by thapsigargin and by the phospholipase C inhibitor U-73122. The results suggest that the elevated extracellular K+ concentration causes Ca2+ release from internal stores which is independent of extracellular Na+- and Ca2+-influx, and may reveal a novel mechanism by which the potassium-induced depolarization of presynaptic nerve terminals can regulate intracellular Ca2+ concentration and exocytosis.


Subject(s)
Calcium/pharmacology , Glycine/metabolism , Posterior Horn Cells/cytology , Potassium/pharmacology , Presynaptic Terminals/metabolism , Spinal Cord/cytology , Action Potentials/drug effects , Action Potentials/physiology , Action Potentials/radiation effects , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Glycine Agents/pharmacology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/radiation effects , Lumbosacral Region , Peptides, Cyclic/pharmacology , Posterior Horn Cells/drug effects , Posterior Horn Cells/physiology , Presynaptic Terminals/drug effects , Rats , Strychnine/pharmacology
3.
J Neurochem ; 78(5): 1009-18, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11553675

ABSTRACT

The modulatory effects of Zn(2+) and other divalent cations on the ATP-induced responses of preganglionic neurons acutely dissociated from the rat dorsal motor nucleus of the vagus (DMV) were examined using a nystatin-perforated patch technique under voltage-clamp. DMV neurons were identified by back-filling of DiI placed on the vagal bundle at the neck. Zn(2+) exerts a concentration-dependent effect on P2X receptor-mediated current (I(ATP)): a potentiation by low concentrations of Zn(2+) (< or = 50 microM) and an inhibition by high concentrations (> 50 microM). Inhibition of the ATP response was associated with a prolongation of the rising phase of I(ATP). Cu(2+) mimicked Zn(2+) regarding the biphasic modulation of I(ATP). On the other hand, Ni(2+) potentiated, but failed to inhibit, the ATP response even at a concentration of 3 mM. Quantitative RT-PCR revealed the similarity of P2X(2) mRNA expression between the DMV and superior cervical ganglion (SCG) but not in the dorsal root ganglion (DRG) and hypoglossal nucleus (XII). The results from the electrophysiological and molecular approaches suggest that functional P2X receptors expressed in DMV neurons are characterized mainly by the P2X(2) and P2X(2/6) subtype. DMV neurons possess similar P2X receptor characteristics to SCG neurons.


Subject(s)
Motor Neurons/metabolism , Receptors, Purinergic P2/metabolism , Vagus Nerve/cytology , Zinc/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Autonomic Fibers, Preganglionic/drug effects , Autonomic Fibers, Preganglionic/metabolism , Brain Stem/cytology , Cations, Divalent/pharmacology , DNA Primers , Female , Ganglia, Spinal/cytology , Gene Expression/physiology , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Motor Neurons/cytology , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X2 , Reverse Transcriptase Polymerase Chain Reaction , Superior Cervical Ganglion/cytology
4.
Biochem Biophys Res Commun ; 284(1): 142-9, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11374883

ABSTRACT

Mice that lack caspase-3, which functions in apoptosis, were generated by gene targeting and shown to undergo hearing loss. The ABR threshold of the caspase-3(-/-) mice was significantly elevated compared to that of caspase-3(+/+) mice at 15 days of age and was progressively elevated further by 30 days. Distortion product otoacoustic emissions were not detectable in caspase-3(-/-) mice at 15 days of age. Caspase-3(-/-) mice exhibited marked degeneration of spiral ganglion neurons and a loss of inner and outer hair cells in the cochlea at 30 days of age, although no such changes were apparent at 15 days. The degenerating neurons manifested features, including cytoplasmic vacuolization, distinct from those characteristic of apoptosis. Spiral ganglion neurons and cochlear hair cells thus appear to require caspase-3 for survival but not for initial development. The mapping of both the human caspase-3 gene and the locus responsible for an autosomal dominant, nonsyndromic form of hearing loss (DFNA24) to chromosome 4q35 suggests that the caspase-3(-/-) mice may represent a model of this human condition.


Subject(s)
Caspases/deficiency , Cochlea/innervation , Deafness/genetics , Neurons/pathology , Aging/pathology , Animals , Auditory Threshold , Caspase 3 , Caspases/biosynthesis , Caspases/genetics , Cell Count , Cell Death/genetics , Cochlea/metabolism , Cochlea/pathology , Deafness/congenital , Deafness/pathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Outer/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Otoacoustic Emissions, Spontaneous/genetics , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Vacuoles/pathology
5.
J Neurophysiol ; 84(1): 281-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10899203

ABSTRACT

The reversibility and cation selectivity of the K(+)-Cl(-) cotransporter (KCC), which normally extrudes Cl(-) out of neurons, was investigated in dissociated lateral superior olive neurons of rats using the gramicidin perforated patch technique. Intracellular Cl(-) activity (alpha[Cl(-)](i)) was maintained well below electrochemical equilibrium as determined from the extracellular Cl(-) activity and the holding potential, where the pipette and external solutions contained 150 mM K(+) ([K(+)](pipette)) and 5 mM K(+) ([K(+)](o)), respectively. Extracellular application of 1 mM furosemide or elevated [K(+)](o) increased alpha[Cl(-)](i). When the pipette solution contained 150 mM Cs(+) ([Cs(+)](pipette)), alpha[Cl(-)](i) increased to a value higher than the passive alpha[Cl(-)](i). An increase of alpha[Cl(-)](i) with the [Cs(+)](pipette) was not due to the simple blockade of net KCC by the intracellular Cs(+) since alpha[Cl(-)](i), with the pipette solution containing 75 mM Cs(+) and 75 mM K(+), reached a value between those obtained using the [K(+)](pipette) and the [Cs(+)](pipette). The higher-than-passive alpha[Cl(-)](i) with the [Cs(+)](pipette) was reduced by 1 mM furosemide, but not by 20 microM bumetanide or Na(+)-free external solution, indicating that the accumulation of [Cl(-)](i) in the [Cs(+)](pipette) was mediated by a KCC operating in a reversed mode rather than by Na(+)-dependent, bumetanide-sensitive mechanisms. Replacement of K(+) in the pipette solution with either Li(+) or Na(+) mimicked the effect of Cs(+) on alpha[Cl(-)](i). On the other hand, Rb(+) mimicked K(+) in the pipette solution. These results indicate that K(+) and Rb(+), but not Cs(+), Li(+), or Na(+), can act as substrates of KCC in LSO neurons.


Subject(s)
Carrier Proteins/metabolism , Neurons/metabolism , Symporters , Animals , Biological Transport/drug effects , Biological Transport/physiology , Bumetanide/pharmacology , Cations/metabolism , Cesium/pharmacology , Chlorides/metabolism , Diuretics/pharmacology , Female , Furosemide/pharmacology , Male , Olivary Nucleus/cytology , Patch-Clamp Techniques , Potassium/metabolism , Rats , Rats, Wistar , K Cl- Cotransporters
6.
J Physiol ; 524 Pt 2: 471-83, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10766927

ABSTRACT

1. The ATP action on spontaneous miniature glycinergic inhibitory postsynaptic currents (mIPSCs) was investigated in rat substantia gelatinosa (SG) neurons mechanically dissociated from the 2nd layer of the dorsal horn in which their presynaptic glycinergic nerve terminals remained intact. 2. ATP reversibly facilitated the frequency of the mIPSCs in a concentration-dependent manner without affecting their amplitude distribution. The ATP agonist, 2-methylthioATP (2MeSATP), mimicked the ATP action, while another ATP receptor agonist, alphabeta-methylene-ATP (alpha,beta-meATP), had no effect on mIPSCs. 3. The ATP receptor antagonists, suramin (1 x 10-6 M) and pyridoxal-5-phosphate-6-azophenyl-2',4'-disulphonic acid (PPADS) (1 x 10-5 M), completely blocked the facilitatory effect of ATP on glycine release (102.0 +/- 11.2 % and 99.3 +/- 16.2 %, n = 6, respectively) without altering the current amplitude distributions. 4. N-Ethylmaleimide (NEM), a sulphydryl alkylating agent, suppressed the inhibitory effect of adenosine on mIPSC frequency (111.2 +/- 13. 3 %, n = 4) without altering the current amplitude distribution. However, ATP still facilitated the mIPSC frequency (693.3 +/- 245.2 %, n = 4) even in the presence of NEM. 5. The facilitatory effect of ATP (1 x 10-5 M) on mIPSC frequency was not affected by adding 1 x 10-4 M Cd2+ to normal external solution but was eliminated in a Ca2+-free external solution. 6. These results suggest that ATP enhances glycine release from nerve terminals, presumably resulting in the inhibition of SG neurons which conduct nociceptive signals to the CNS. This presynaptic P2X-type ATP receptor may function to prevent excess excitability in SG neurons, thus preventing an excessive pain signal and/or SG cell death.


Subject(s)
Adenosine Triphosphate/physiology , Excitatory Postsynaptic Potentials/physiology , Glycine/physiology , Interneurons/physiology , Posterior Horn Cells/physiology , Synapses/physiology , Adenosine Triphosphate/agonists , Adenosine Triphosphate/pharmacology , Animals , Calcium/physiology , Electrophysiology , Ethylmaleimide/pharmacology , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Interneurons/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Posterior Horn Cells/drug effects , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Rats , Rats, Wistar , Receptors, Glycine/drug effects , Receptors, Glycine/physiology , Receptors, Presynaptic/drug effects , Synapses/drug effects
7.
Neuroscience ; 96(2): 385-92, 2000.
Article in English | MEDLINE | ID: mdl-10683578

ABSTRACT

The post-traumatic change of the voltage-dependent Mg(2+) block of N-methyl-D-aspartate response was investigated using nystatin perforated patch recording mode under the voltage-clamp condition. Motor neurons of the dorsal motor nucleus of vagus nerve were freshly dissociated from rat brain at 2h to 10 days after receiving axonal crush injuries in vivo at the neck. The reduction of voltage-dependent Mg(2+) block of N-methyl-D-aspartate response became evident at more than 12h after the injury, sustained for at least five days and recovered within 10 days. Other characteristics examined such as reversal potentials, the Hill coefficient and EC(50) of N-methyl-D-aspartate-induced current were not affected by axonal injury. The Mg(2+) block of N-methyl-D-aspartate response was not affected at all by local application of colchicine onto the vagal axon in in vivo condition, suggesting that axonal injury, but not the blockade of the axonal flow, is responsible for the change of the sensitivity of N-methyl-D-aspartate response to extracellular Mg(2+). In addition, the reduction of Mg(2+) block by the nerve injury persisted regardless of the presence of protein kinase C modulators, such as 10(-6)M chelerythrine and 10(-7)M calphostin C. Therefore alteration of protein kinase C activity after axonal injury is not responsible for the maintenance of the reduced Mg(2+) block. These findings suggest that injured neurons acquire immature characteristics of plasticity with respect to the sensitivity of N-methyl-D-aspartate receptors to extracellular Mg(2+) or a long-term increase in the susceptibility to Ca(2+) excitotoxicity.


Subject(s)
Axons/physiology , Magnesium/metabolism , Magnesium/pharmacology , Nerve Crush/adverse effects , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Animals, Newborn , Colchicine/pharmacology , Gout Suppressants/pharmacology , Patch-Clamp Techniques , Rats , Recovery of Function/drug effects , Recovery of Function/physiology
8.
Neuropharmacology ; 39(2): 191-201, 2000 Jan 04.
Article in English | MEDLINE | ID: mdl-10670414

ABSTRACT

The effect of dopamine (DA) was investigated on acutely dissociated rat substantia nigra pars compacta (SNc) neurones by using patch clamp recording. The SNc neurones could be classified into two groups. About 75% of large neurones (>30 microm in diameter) were tyrosine hydroxylase (TH) positive while almost all small neurones (<20 microm) were TH negative. In the large neurones, DA hyperpolarized the membrane, resulting in a reduction of the frequency of spontaneous action potentials in current-clamp mode and induced an inward rectifier K+ current in voltage-clamp mode. Quinpirole, a D2 receptor agonist, mimicked the DA action. S(-)-sulpiride, a D2 receptor antagonist, inhibited the DA-induced current (I(DA)) more effectively than SKF83566, a D1 receptor antagonist. Intracellular application of either guanosine 5'-O-(2-thiodiphosphate) (GDP-betaS) or pertussis toxin (IAP) suppressed I(DA). Guanosine 5'-O-(3-thiotriphosphate) (GTP-gammaS) sustained the DA response. Modulators for cAMP such as forskolin and isobutylmethylxathine, H-89, a protein kinase A inhibitor, and chelerythrine, a protein kinase C inhibitor, had no effect on I(DA). The frequency of DA-induced single channel currents in the inside-out patch configuration, for which the unitary conductance was 56.6pS, was greatly reduced by the replacement of GTP with GDP perfused at the cytosolic side. These results suggest that DA acts on a D2-like receptor and activates directly an IAP-sensitive G protein coupled with inward rectifier K+ channels, resulting in a decrease in the spontaneous firing activities of rat SNc dopaminergic neurones.


Subject(s)
Dopamine/metabolism , Neurons/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Substantia Nigra/metabolism , Animals , Dopamine Agents/pharmacology , Female , GTP-Binding Proteins/metabolism , Male , Neurons/drug effects , Rats , Rats, Wistar , Receptors, Dopamine/drug effects , Receptors, Dopamine/metabolism , Substantia Nigra/cytology , Substantia Nigra/drug effects
9.
J Neurosci ; 20(1): 251-8, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10627602

ABSTRACT

Microglial cells were isolated from rat cerebral cortex, and kainate (KA)-induced inward current was measured at a holding potential of -40 or -60 mV. 6-Cyano-7-nitroquinoxaline-2, 3-dione-sensitive KA-induced currents increased with increasing KA concentration. The half-activation concentration and Hill coefficient were 3.3 x 10(-4) M and 1.4, respectively. Although glutamate (Glu) and AMPA-induced currents were much smaller than that induced by KA, all KA-, Glu-, and AMPA-induced currents were greatly and consistently enhanced in the presence of cyclothiazide (CTZ). On the other hand, KA-induced currents were much less sensitive to potentiation by concanavain A, suggesting that the KA-induced response in rat microglia is predominantly mediated by AMPA-preferring receptors (subunits GluR1-GluR4). The current-voltage relationships of KA- and AMPA-CTZ-induced currents were almost linear or slightly outward rectifying. The reversal potential of KA-induced current shifted to negative potentials (from +4 to -40 mV) on switching from high Na(+) to high Ca(2+) external solution, indicating the low Ca(2+) permeability through the AMPA-KA receptor channel complexes. AMPA-KA receptor expression was studied with immunohistochemistry and reverse transcription-PCR, from which GluR2, GluR3, GluR4, and GluR5 were identified. Lower levels of mRNAs for GluR7 and KA-1-KA-2 were also indicated. Finally, activation of these receptors with KA or Glu significantly enhanced the production of tumor necrosis factor-alpha. These results suggest that primary cultured rat microglia possesses functional Glu receptor, which may mediate neuron to microglia communication in the physiological and pathological states.


Subject(s)
Microglia/chemistry , Receptors, AMPA/analysis , Receptors, Kainic Acid/analysis , Animals , Antihypertensive Agents/pharmacology , Benzothiadiazines/pharmacology , Calcium/pharmacokinetics , Cells, Cultured , Cerebral Cortex/cytology , Concanavalin A/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Glutamic Acid/pharmacology , Kainic Acid/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microglia/cytology , Microglia/physiology , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, AMPA/genetics , Receptors, Kainic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical , Tumor Necrosis Factor-alpha/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
11.
J Biol Chem ; 274(46): 32762-70, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10551836

ABSTRACT

To explore the intracellular pathways activated by vasopressin receptors, the effects of arginine vasopressin (AVP) and its analogues mediating glycine (Gly)-induced Cl(-) currents (I(Gly)) were examined in acutely dissociated rat hippocampal CA1 neurons using the whole-cell patch recording technique. AVP and its analogues inhibited I(Gly) in a concentration-dependent manner. The inhibitory actions of AVP(4-9) (AVP metabolite) and NC-1900 (AVP(4-9) analogue) were reversed by a V(1) receptor antagonist, or pretreatment with 1,2-bis(2-amino-5-fluorophenoxy)ethane-N,N,N', N'-tetraacetic acid. In contrast, these blocking procedures had no effect on the 1-desamino-8-D-AVP (DDAVP; V(2) agonist) action. A V(2) receptor antagonist did not block the inhibitory action of AVP(4-9) or NC-1900, but blocked that of DDAVP. The inhibitory action of AVP was completely blocked by the co-application of the V(1) and V(2) antagonists. The inhibitory action of NC-1900 was not affected by perfusion with a Ca(2+)-free external solution, but was strongly blocked by thapsigargin. The intracellular application of heparin or anti-inositol 1,4,5-triphosphate (IP(3)) also blocked the NC-1900 action. Furthermore, Ca(2+)/calmodulin (CaM) inhibitors blocked the NC-1900 action, while a CaM-dependent kinase II inhibitor and PKC modulators had no effect. 2',5'-Dideoxyadenosine (an adenylate cyclase inhibitor), H-89, and Rp-cAMPS blocked the inhibitory actions of NC-1900 and DDAVP. These results suggest that the activation of the V(1) receptor in the hippocampal neurons induces the production of IP(3), which releases Ca(2+) from the IP(3)-sensitive Ca(2+) storage sites. The Ca(2+) binds to CaM, resulting in the activation of Ca(2+/)CaM-sensitive adenylate cyclases. The activation of protein kinase A through the adenylate cyclase inhibits I(Gly).


Subject(s)
Arginine Vasopressin/metabolism , Neurons/metabolism , Receptors, Vasopressin/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists , Arginine Vasopressin/analogs & derivatives , Benzazepines/pharmacology , Calcium/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Deamino Arginine Vasopressin/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Glycine/pharmacology , Hippocampus/metabolism , Oligopeptides/pharmacology , Patch-Clamp Techniques , Peptide Fragments/pharmacology , Protein Kinase C/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Rats , Receptors, Vasopressin/agonists
13.
J Neurosci ; 19(8): 2843-51, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10191302

ABSTRACT

The regulatory mechanisms of intracellular Cl- concentration ([Cl-]i) were investigated in the lateral superior olive (LSO) neurons of various developmental stages by taking advantage of gramicidin perforated patch recording mode, which enables neuronal [Cl-]i measurement. Responses to glycine changed from depolarization to hyperpolarization during the second week after birth, resulting from [Cl-]i decrease. Furosemide equally altered the [Cl-]i of both immature and mature LSO neurons, indicating substantial contributions of furosemide-sensitive intracellular Cl- regulators; i.e., K+-Cl- cotransporter (KCC) and Na+-K+-Cl- cotransporter (NKCC), throughout this early development. Increase of extracellular K+ concentration and replacement of intracellular K+ with Cs+ resulted in [Cl-]i elevation at postnatal days 13-15 (P13-P15), but not at P0-P2, indicating that the mechanism of neuronal Cl- extrusion is sensitive to both furosemide and K+-gradient and poorly developed in immature LSO neurons. In addition, removal of extracellular Na+ decreased [Cl-]i at P0-P2, suggesting the existence of extracellular Na+-dependent and furosemide-sensitive Cl- accumulation in immature LSO neurons. These data show clearly that developmental changes of Cl- cotransporters alter [Cl-]i and are responsible for the switch from the neonatal Cl- efflux to the mature Cl- influx in LSO neurons. Such maturational changes in Cl- cotransporters might have the important functional roles for glycinergic and GABAergic synaptic transmission and the broader implications for LSO and auditory development.


Subject(s)
Carrier Proteins/metabolism , Chlorides/metabolism , Neurons/metabolism , Olivary Nucleus/physiology , Potassium/metabolism , Sodium/metabolism , Animals , Animals, Newborn , Carrier Proteins/drug effects , Female , Furosemide/pharmacology , Glycine/pharmacology , In Vitro Techniques , Male , Membrane Potentials/drug effects , Neurons/drug effects , Olivary Nucleus/cytology , Olivary Nucleus/drug effects , Patch-Clamp Techniques , Rats , Rats, Wistar , Sodium-Potassium-Chloride Symporters
14.
Neuroscience ; 89(1): 29-41, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10051215

ABSTRACT

The effect of noradrenaline on the glycine response was investigated in neurons acutely dissociated from the rat sacral dorsal commissural nucleus using nystatin perforated patch recording configuration under voltage-clamp conditions. Noradrenaline reversibly potentiated the 10(-5)M glycine-induced Cl- current in a concentration-dependent manner. Single channel recordings in a cell-attached mode revealed that noradrenaline decreased the closing time of the glycine-activated channel activity. Noradrenaline neither changed the reversal potential of the glycine response nor affected the affinity of glycine to its receptor. Clonidine mimicked and yohimbine blocked the noradrenaline action on glycine response. N-[2(methylamino)ethyl]-5-isoquinoline sulfonamide dihydrochloride, protein kinase A inhibitor, mimicked the effect of noradrenaline on glycine response. Noradrenaline failed to affect the glycine response in the presence of these intracellular cyclic AMP and protein kinase A modulators. However, noradrenaline further enhanced the glycine response even in the presence of phorbol-12-myristate-13-acetate and chelerythrine, a protein kinase C inhibitor. Pertussis toxin treatment for 6-8 h blocked the noradrenaline facilitatory effect on the glycine response. In addition, noradrenaline potentiated the strychnine-sensitive postsynaptic currents evoked in a slice preparation of sacral dorsal commissural nucleus. These results suggest that the activation of alpha2-adrenoceptor by noradrenaline coupled with pertussis toxin-sensitive G-proteins reduces intracellular cyclic AMP formation through the inhibition of adenyl cyclase. The reduction of cyclic AMP decreases the protein kinase A activity, thus resulting in the potentiation of the glycinergic inputs to the sacral dorsal commissural neurons. It is thus feasible that the noradrenergic input to the sacral dorsal commissural nucleus modulates such nociceptive signals as pain by intracellular enhancing the glycine response.


Subject(s)
Glycine/metabolism , Neurons, Afferent/chemistry , Neurons, Afferent/enzymology , Receptors, Adrenergic, alpha-2/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Adenylate Cyclase Toxin , Adrenergic alpha-Agonists/pharmacology , Animals , Bucladesine/pharmacology , Clonidine/pharmacology , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Electrophysiology , GTP-Binding Proteins/metabolism , Glycine Agents/pharmacology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Isoproterenol/pharmacology , Lumbosacral Region , Neurons, Afferent/drug effects , Nociceptors/physiology , Norepinephrine/pharmacology , Pertussis Toxin , Phenylephrine/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Rats , Rats, Wistar , Strychnine/pharmacology , Virulence Factors, Bordetella/pharmacology
15.
J Neurochem ; 72(2): 808-14, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9930757

ABSTRACT

The effects of various types of steroids on the nicotinic acetylcholine (ACh) receptor (nAChR)-mediated responses were investigated in superior cervical ganglionic neurons acutely dissociated from rats using nystatin perforated patch recording. ACh induced a peak followed by a gradual decrease in the inward current at a holding potential of -40 mV. Nicotine, but not muscarine, mimicked ACh. Hydrocortisone at a concentration of >10(-6) M reversibly suppressed both the peak and steady-state nicotine-induced currents (Inic) in a noncompetitive manner. The inhibition of Inic by hydrocortisone did not show any voltage dependency and persisted in the presence of either cyclic AMP modulators, forskolin and 3-isobutyl-1-methylxanthine, or a protein kinase A inhibitor, N-[2-(methylamino)ethyl]-5-isoquinolinesulfonamide dihydrochloride (H-89). Beta-estradiol, androsterone, aldosterone, and 17alpha-estradiol mimicked hydrocortisone in its inhibitory action on ACh-induced currents (I(ACh)). The potency for the inhibitory actions on I(ACh) was as follows: androsterone > beta-estradiol > hydrocortisone > or = aldosterone = 17alpha-estradiol. Cholesterol had no effect on the I(ACh). In conclusion, the structural characteristics of a steroid are thus considered to be necessary to block nicotinic I(ACh) in rat superior cervical ganglionic cells, whereas the cholesterol side chain might disturb the inhibitory action of the steroid skeleton on nAChRs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hydrocortisone/pharmacology , Neurons/chemistry , Receptors, Nicotinic/physiology , Superior Cervical Ganglion/cytology , Acetylcholine/pharmacology , Animals , Cholinergic Agents/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Estradiol/analogs & derivatives , Ionophores/pharmacology , Membrane Potentials/drug effects , Muscarine/pharmacology , Muscarinic Agonists/pharmacology , Neurons/enzymology , Nicotine/pharmacology , Nystatin/pharmacology , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Muscarinic/physiology , Superior Cervical Ganglion/chemistry , Vasodilator Agents/pharmacology
16.
J Biol Chem ; 273(18): 11056-61, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9556589

ABSTRACT

Human gamma-aminobutyric acid type A (GABAA) receptors were expressed in the baculovirus/Sf-9 insect cell expression system using recombinant cDNA of alpha1beta2gamma2s subunits. The effect of unsaturated fatty acids on GABAA receptor complexes was investigated electrophysiologically using conventional whole cell recording under voltage clamp. Three distinct effects of docosahexaenoic acid (DHA) on the GABA responses were observed. First, DHA, at a concentration of 10(-7) M or greater, accelerated the desensitization after the peak of the GABA-induced current. Second, DHA (10(-6) M) potentiated the peak amplitude of GABA response. This potentiation by DHA was inhibited in the presence of Zn2+ (10(-5) M); Cu2+ and Ni2+ mimicked the action of Zn2+. Zn2+ (10(-5) M) did not block the GABA response on alpha1beta2gamma2s receptor complexes. Third, DHA, at a concentration of 3 x 10(-6) M or higher, gradually suppressed the peak amplitude of GABA response. A protein kinase A inhibitor, a protein kinase C inhibitor, and a Ca2+ chelator did not modify the effects of DHA on GABA-induced chloride ion current. Six unsaturated fatty acids other than DHA were examined. Arachidonic acid mimicked the effect of DHA while e.g. oleic acid had no effect. The inhibition of the GABA response in the presence of DHA was also observed in cells expressing GABAA receptors of alpha1 and beta2 subunit combinations. The data show that the gamma subunit is essential for DHA and arachidonic acid to potentiate the GABA-induced Cl- channel activity and to affect the desensitization kinetics of the GABAA receptor.


Subject(s)
Docosahexaenoic Acids/metabolism , Receptors, GABA-A/metabolism , Animals , Chloride Channels/drug effects , Humans , Receptors, GABA-A/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera/cytology
17.
J Neurochem ; 70(2): 865-70, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9453584

ABSTRACT

The effects of the proadrenomedullin N-terminal 20-amino acid peptide (PAMP) on the nicotinic acetylcholine (ACh) receptor (nAChR)-mediated inward current were investigated in neurons acutely dissociated from the rat locus coeruleus using whole-cell recording under voltage clamp. Nicotine and cytidine mimicked the ACh response, whereas the maximal response to dimethylphenylpiperazinium was lower in amplitude compared with that to ACh. Nicotine-induced current (I[nic]) was suppressed more effectively by mecamylamine than by hexamethonium. In addition, neither atropine nor alpha-bungarotoxin affected the I(nic). PAMP reversibly and noncompetitively suppressed the peak amplitude of 10(-4) M I(nic). PAMP concentrations for the threshold, half-maximal inhibition, and maximal inhibition of 10(-4) M I(nic) were 10(-8), 2.6 x 10(-7), and 10(-5) M, respectively. The peak amplitudes of 10(-4) M I(nic) elicited at 2-min intervals showed a gradual decline in the presence of 10(-7) M PAMP. This decline in the I(nic) was independent of the period of PAMP pretreatment. The suppression of I(nic) by PAMP did not show any voltage dependency at a holding potential (VH) of <0 mV, although the inhibitory effect was masked by the marked inward rectification of I(nic) at a VH of >0 mV. These results suggest that PAMP could thus be a unique endogenous peptide that antagonizes the nAChR in the CNS.


Subject(s)
Locus Coeruleus/physiology , Neurons/physiology , Nicotine/pharmacology , Peptide Fragments/pharmacology , Peptides , Proteins/pharmacology , Receptors, Nicotinic/physiology , Adrenomedullin , Animals , Atropine/pharmacology , Bungarotoxins/pharmacology , Cells, Cultured , Cytidine/pharmacology , Dimethylphenylpiperazinium Iodide/pharmacology , Female , Male , Mecamylamine/pharmacology , Neurons/drug effects , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Nicotinic/drug effects
19.
Eur J Pharmacol ; 329(2-3): 253-7, 1997 Jun 25.
Article in English | MEDLINE | ID: mdl-9226420

ABSTRACT

Recombinant human GABA(A) receptors were investigated in vitro by coexpression of cDNAs coding for alpha1, beta2, and gamma2 subunits in the baculovirus/Sf-9 insect cell system. We report that a single amino acid exchange (isoleucine 121 to valine 121) in the N-terminal, extracellular part of the alpha1 subunit induces a marked decrease in agonist GABA(A) receptor ligand sensitivity. The potency of muscimol and GABA to inhibit the binding of the GABA(A) receptor antagonist [3H]SR 95531 (2-(3-carboxypropyl)-3-amino-6-(4-methoxyphenyl)pyridazinium bromide) was higher in receptor complexes of alpha1(ile 121) beta2gamma2 than in those of alpha1(val 121) beta2gamma2 (IC50 values were 32-fold and 26-fold lower for muscimol and GABA, respectively). The apparent affinity of the GABA(A) receptor antagonist bicuculline methiodide to inhibit the binding of [3H]SR 95531 did not differ between the two receptor complex variants. Electrophysiological measurements of GABA induced whole-cell Cl- currents showed a ten-fold decrease in the GABA(A) receptor sensitivity of alpha1 (val 121) beta2gamma2 as compared to alpha1(ile 121) beta2gamma2 receptor complexes. Thus, a relatively small change in the primary structure of the alpha1 subunit leads to a decrease selective for GABA(A) receptor sensitivity to agonist ligands, since no changes were observed in a GABA(A) receptor antagonist affinity and benzodiazepine receptor binding.


Subject(s)
GABA-A Receptor Agonists , Isoleucine/genetics , Valine/genetics , Baculoviridae/genetics , Cell Line , DNA Primers , GABA Agonists/pharmacology , GABA Antagonists/metabolism , GABA Antagonists/pharmacology , Humans , Patch-Clamp Techniques , Point Mutation , Pyridazines/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Neuroscience ; 77(3): 661-71, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9070743

ABSTRACT

We investigated the effects of arachidonic acid on K+ channels in freshly dissociated neurons of 10- to 20-day-old rat visual cortex, using a perforated and conventional whole-cell patch-clamp and inside-out excised patch configurations. Arachidonic acid at 5-30 microM induced an outward current in 88.1% of the neurons in whole-cell mode, and evoked channel opening with a conductance of 170-238 pS in 90.5% of neurons under inside-out patch recording. Arachidonic acid-activated K+ channels were partially blocked by extracellular administration of 1 mM tetraethylammonium and 100 nM charybdotoxin. However, Ba2+ completely blocked the channel in all cases. None of the other K+ channel blockers, including 4-aminopyridine, quinidine, apamin and glibenclamide, inhibited the arachidonic acid-activated channels. Intracellular perfusion with Ca2+-free and 5 mM BAPTA in Ca2+-free extracellular perfusate containing 2 mM EGTA in conventional whole-cell recording did not inhibit the K+ channel, implying that the channel is not Ca2+ dependent. Neither guanosine 5'-O-(2-thiodiphosphate) nor staurosporine applied in inside-out mode affected the arachidonic acid-activated channels, indicating that G-protein and protein kinase C are not involved in this phenomenon. In addition, neither indomethacin nor nordihydroguaiaretic acid blocked the channel currents, demonstrating that it is arachidonic acid itself but not its metabolites that induced the effect. Among the fatty acids tested, only cis-unsaturated fatty acids, having more than two double bonds, such as arachidonic acid, docosahexaenoic acid and linolenic acid, activated the K+ channels. These findings suggest that there exists a novel type of K+ channel activated by arachidonic acid which may play a critical role in modulating neuronal excitability in cortical neurons.


Subject(s)
Arachidonic Acid/pharmacology , Neurons/physiology , Potassium Channels/physiology , Visual Cortex/physiology , 4-Aminopyridine/pharmacology , Animals , Apamin/pharmacology , Barium/pharmacology , Charybdotoxin/pharmacology , Docosahexaenoic Acids/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Fatty Acids, Nonesterified/pharmacology , Fatty Acids, Unsaturated/pharmacology , Glyburide/pharmacology , In Vitro Techniques , Indomethacin/pharmacology , Membrane Potentials/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers , Quinidine/pharmacology , Rats , Rats, Sprague-Dawley , Tetraethylammonium , Tetraethylammonium Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...