Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 188: 106328, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37852390

ABSTRACT

Like a handful of other neuronal types in the brain, cholinergic neurons (CNs) in the pedunculopontine nucleus (PPN) are lost during Parkinson's disease (PD). Why this is the case is unknown. One neuronal trait implicated in PD selective neuronal vulnerability is the engagement of feed-forward stimulation of mitochondrial oxidative phosphorylation (OXPHOS) to meet high bioenergetic demand, leading to sustained oxidant stress and ultimately degeneration. The extent to which this trait is shared by PPN CNs is unresolved. To address this question, a combination of molecular and physiological approaches were used. These studies revealed that PPN CNs are autonomous pacemakers with modest spike-associated cytosolic Ca2+ transients. These Ca2+ transients were partly attributable to the opening of high-threshold Cav1.2 Ca2+ channels, but not Cav1.3 channels. Cav1.2 channel signaling through endoplasmic reticulum ryanodine receptors stimulated mitochondrial OXPHOS to help maintain cytosolic adenosine triphosphate (ATP) levels necessary for pacemaking. Inhibition of Cav1.2 channels led to the recruitment of ATP-sensitive K+ channels and the slowing of pacemaking. A 'side-effect' of Cav1.2 channel-mediated stimulation of mitochondria was increased oxidant stress. Thus, PPN CNs have a distinctive physiological phenotype that shares some, but not all, of the features of other neurons that are selectively vulnerable in PD.


Subject(s)
Parkinson Disease , Humans , Cholinergic Neurons , Signal Transduction , Adenosine Triphosphate , Oxidants
2.
bioRxiv ; 2023 Aug 07.
Article in English | MEDLINE | ID: mdl-37609299

ABSTRACT

Like a handful of other neuronal types in the brain, cholinergic neurons (CNs) in the pedunculopontine nucleus (PPN) are lost in the course of Parkinson's disease (PD). Why this is the case is unknown. One neuronal trait implicated in PD selective neuronal vulnerability is the engagement of feed-forward stimulation of mitochondrial oxidative phosphorylation (OXPHOS) to meet high bioenergetic demand, leading to sustained oxidant stress and ultimately degeneration. The extent to which this trait is shared by PPN CNs is unresolved. To address this question, a combination of molecular and physiological approaches were used. These studies revealed that PPN CNs are autonomous pacemakers with modest spike-associated cytosolic Ca 2+ transients. These Ca 2+ transients were attributable in part to the opening of high-threshold Cav1.2 Ca 2+ channels, but not Cav1.3 channels. Nevertheless, Cav1.2 channel signaling through endoplasmic reticulum ryanodine receptors stimulated mitochondrial OXPHOS to help maintain cytosolic adenosine triphosphate (ATP) levels necessary for pacemaking. Inhibition of Cav1.2 channels led to recruitment of ATP-sensitive K + channels and slowing of pacemaking. Cav1.2 channel-mediated stimulation of mitochondria increased oxidant stress. Thus, PPN CNs have a distinctive physiological phenotype that shares some, but not all, of the features of other neurons that are selectively vulnerable in PD.

3.
J Physiol ; 581(Pt 3): 941-60, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17379638

ABSTRACT

We determined the expression of Kv2 channel subunits in rat somatosensory and motor cortex and tested for the contributions of Kv2 subunits to slowly inactivating K+ currents in supragranular pyramidal neurons. Single cell RT-PCR showed that virtually all pyramidal cells expressed Kv2.1 mRNA and approximately 80% expressed Kv2.2 mRNA. Immunocytochemistry revealed striking differences in the distribution of Kv2.1 and Kv2.2 subunits. Kv2.1 subunits were clustered and located on somata and proximal dendrites of all pyramidal cells. Kv2.2 subunits were primarily distributed on large apical dendrites of a subset of pyramidal cells from deep layers. We used two methods for isolating currents through Kv2 channels after excluding contributions from Kv1 subunits: intracellular diffusion of Kv2.1 antibodies through the recording pipette and extracellular application of rStromatoxin-1 (ScTx). The Kv2.1 antibody specifically blocked the slowly inactivating K+ current by 25-50% (at 8 min), demonstrating that Kv2.1 subunits underlie much of this current in neocortical pyramidal neurons. ScTx (300 nM) also inhibited approximately 40% of the slowly inactivating K+ current. We observed occlusion between the actions of Kv2.1 antibody and ScTx. In addition, Kv2.1 antibody- and ScTx-sensitive currents demonstrated similar recovery from inactivation and voltage dependence and kinetics of activation and inactivation. These data indicate that both agents targeted the same channels. Considering the localization of Kv2.1 and 2.2 subunits, currents from truncated dissociated cells are probably dominated by Kv2.1 subunits. Compared with Kv2.1 currents in expression systems, the Kv2.1 current in neocortical pyramidal cells activated and inactivated at relatively negative potentials and was very sensitive to holding potential.


Subject(s)
Neocortex/metabolism , Potassium/metabolism , Pyramidal Cells/metabolism , Shab Potassium Channels/metabolism , Animals , Antibodies , Dendrites/metabolism , Gene Expression , In Vitro Techniques , Kinetics , Membrane Potentials , Models, Neurological , Neocortex/cytology , Neocortex/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Protein Subunits/metabolism , Pyramidal Cells/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Shab Potassium Channels/antagonists & inhibitors , Shab Potassium Channels/genetics , Shab Potassium Channels/immunology , Spider Venoms/pharmacology , Tetraethylammonium/pharmacology
4.
Neuroscience ; 146(2): 537-54, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17324523

ABSTRACT

Somatostatin is synthesized and released by aspiny GABAergic interneurons of the neostriatum, some of them identified as low threshold spike generating neurons (LTS-interneurons). These neurons make synaptic contacts with spiny neostriatal projection neurons. However, very few somatostatin actions on projection neurons have been described. The present work reports that somatostatin modulates the Ca(2+) activated K(+) currents (K(Ca) currents) expressed by projection cells. These actions contribute in designing the firing pattern of the spiny projection neuron; which is the output of the neostriatum. Small conductance (SK) and large conductance (BK) K(Ca) currents represent between 30% and 50% of the sustained outward current in spiny cells. Somatostatin reduces SK-type K(+) currents and at the same time enhances BK-type K(+) currents. This dual effect enhances the fast component of the after hyperpolarizing potential while reducing the slow component. Somatostatin then modifies the firing pattern of spiny neurons which changed from a tonic regular pattern to an interrupted "stuttering"-like pattern. Semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) tissue expression analysis of dorsal striatal somatostatinergic receptors (SSTR) mRNA revealed that all five SSTR mRNAs are present. However, single cell RT-PCR profiling suggests that the most probable receptor in charge of this modulation is the SSTR2 receptor. Interestingly, aspiny interneurons may exhibit a "stuttering"-like firing pattern. Therefore, somatostatin actions appear to be the entrainment of projection neurons to the rhythms generated by some interneurons. Somatostatin is then capable of modifying the processing and output of the neostriatum.


Subject(s)
Action Potentials/physiology , Corpus Striatum/cytology , Dendritic Spines/metabolism , Neurons , Potassium Channels, Calcium-Activated/physiology , Somatostatin/metabolism , 4-Aminopyridine/pharmacology , Action Potentials/drug effects , Anesthetics, Local/pharmacology , Animals , Apamin/pharmacology , Calcitonin/pharmacology , Dendritic Spines/drug effects , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Gene Expression/drug effects , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Peptide Fragments/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Receptors, Somatostatin/classification , Receptors, Somatostatin/metabolism , Somatostatin/pharmacology , Tetrodotoxin/pharmacology
5.
J Physiol ; 571(Pt 2): 371-89, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16373387

ABSTRACT

Potassium channels are extremely diverse regulators of neuronal excitability. As part of an investigation into how this molecular diversity is utilized by neurones, we examined the expression and biophysical properties of native Kv1 channels in layer II/III pyramidal neurones from somatosensory and motor cortex. Single-cell RT-PCR, immunocytochemistry, and whole cell recordings with specific peptide toxins revealed that individual pyramidal cells express multiple Kv1 alpha-subunits. The most abundant subunit mRNAs were Kv1.1 > 1.2 > 1.4 > 1.3. All of these subunits were localized to somatodendritic as well as axonal cell compartments. These data suggest variability in the subunit complexion of Kv1 channels in these cells. The alpha-dendrotoxin (alpha-DTX)-sensitive current activated more rapidly and at more negative potentials than the alpha-DTX-insensitive current, was first observed at voltages near action potential threshold, and was relatively insensitive to holding potential. The alpha-DTX-sensitive current comprised about 10% of outward current at steady-state, in response to steps from -70 mV. From -50 mV, this percentage increased to approximately 20%. All cells expressed an alpha-DTX-sensitive current with slow inactivation kinetics. In some cells a transient component was also present. Deactivation kinetics were voltage dependent, such that deactivation was slow at potentials traversed by interspike intervals during repetitive firing. Because of its kinetics and voltage dependence, the alpha-DTX-sensitive current should be most important at physiological resting potentials and in response to brief stimuli. Kv1 channels should also be important at voltages near threshold and corresponding to interspike intervals.


Subject(s)
Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/metabolism , Kv1.3 Potassium Channel/metabolism , Kv1.4 Potassium Channel/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Animals , Delayed Rectifier Potassium Channels , Elapid Venoms/pharmacology , Immunohistochemistry , Ion Channel Gating/drug effects , Motor Cortex/metabolism , Neocortex/cytology , Neocortex/physiology , Potassium Channel Blockers/pharmacology , Pyramidal Cells/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/metabolism
6.
J Neurosci ; 21(7): 2268-77, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11264302

ABSTRACT

Dopamine (DA) is a well established modulator of prefrontal cortex (PFC) function, yet the cellular mechanisms by which DA exerts its effects in this region are controversial. A major point of contention is the consequence of D(1) DA receptor activation. Several studies have argued that D(1) receptors enhance the excitability of PFC pyramidal neurons by augmenting voltage-dependent Na(+) currents, particularly persistent Na(+) currents. However, this conjecture is based on indirect evidence. To provide a direct test of this hypothesis, we combined voltage-clamp studies of acutely isolated layer V-VI prefrontal pyramidal neurons with single-cell RT-PCR profiling. Contrary to prediction, the activation of D(1) or D(5) DA receptors consistently suppressed rapidly inactivating Na(+) currents in identified corticostriatal pyramidal neurons. This modulation was attenuated by a D(1)/D(5) receptor antagonist, mimicked by a cAMP analog, and blocked by a protein kinase A (PKA) inhibitor. In the same cells the persistent component of the Na(+) current was unaffected by D(1)/D(5) receptor activation-suggesting that rapidly inactivating and persistent Na(+) currents arise in part from different channels. Single-cell RT-PCR profiling showed that pyramidal neurons coexpressed three alpha-subunit mRNAs (Nav1.1, 1.2, and 1.6) that code for the Na(+) channel pore. In neurons from Nav1.6 null mice the persistent Na(+) currents were significantly smaller than in wild-type neurons. Moreover, the residual persistent currents in these mutant neurons-which are attributable to Nav1.1/1.2 channels-were reduced significantly by PKA activation. These results argue that D(1)/D(5) DA receptor activation reduces the rapidly inactivating component of Na(+) current in PFC pyramidal neurons arising from Nav1.1/1.2 Na(+) channels but does not modulate effectively the persistent component of the Na(+) current that is attributable to Nav1.6 Na(+) channels.


Subject(s)
Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Receptors, Dopamine D1/physiology , Sodium Channels/physiology , Sodium/physiology , Animals , Mice , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D5
7.
J Neurosci ; 20(24): 8987-95, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11124974

ABSTRACT

In spite of the recognition that striatal D(2) receptors are critical determinants in a variety of psychomotor disorders, the cellular mechanisms by which these receptors shape neuronal activity have remained a mystery. The studies presented here reveal that D(2) receptor stimulation in enkephalin-expressing medium spiny neurons suppresses transmembrane Ca(2+) currents through L-type Ca(2+) channels, resulting in diminished excitability. This modulation is mediated by G(beta)(gamma) activation of phospholipase C, mobilization of intracellular Ca(2+) stores, and activation of the calcium-dependent phosphatase calcineurin. In addition to providing a unifying mechanism to explain the apparently divergent effects of D(2) receptors in striatal medium spiny neurons, this novel signaling linkage provides a foundation for understanding how this pivotal receptor shapes striatal excitability and gene expression.


Subject(s)
Calcineurin/metabolism , Calcium Channels, L-Type/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Isoenzymes/metabolism , Neurons/metabolism , Receptors, Dopamine D2/metabolism , Type C Phospholipases/metabolism , Action Potentials/drug effects , Adenylyl Cyclase Inhibitors , Animals , Barium/pharmacology , Calcineurin Inhibitors , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine/metabolism , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Ion Transport/drug effects , Isoenzymes/antagonists & inhibitors , Isoenzymes/pharmacology , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Phospholipase C beta , Rats , Receptors, Dopamine D2/agonists , Signal Transduction/physiology , Sulpiride/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/pharmacology
8.
J Neurosci ; 20(2): 579-88, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10632587

ABSTRACT

A-type K(+) currents are key determinants of repetitive activity and synaptic integration. Although several gene families have been shown to code for A-type channel subunits, recent studies have suggested that Kv4 family channels are the principal contributors to A-type channels in the somatodendritic membrane of mammalian brain neurons. If this hypothesis is correct, there should be a strong correlation between Kv4 family mRNA and A-type channel protein or aggregate channel currents. To test this hypothesis, quantitative single-cell reverse transcription-PCR analysis of Kv4 family mRNA was combined with voltage-clamp analysis of A-type K(+) currents in acutely isolated neurons. These studies revealed that Kv4.2 mRNA abundance was linearly related to A-type K(+) current amplitude in neostriatal medium spiny neurons and cholinergic interneurons, in globus pallidus neurons, and in basal forebrain cholinergic neurons. In contrast, there was not a significant correlation between estimates of Kv4.1 or Kv4.3 mRNA abundance and A-type K(+) current amplitudes. These results argue that Kv4.2 subunits are major constituents of somatodendritic A-type K(+) channels in these four types of neuron. In spite of this common structural feature, there were significant differences in the voltage dependence and kinetics of A-type currents in the cell types studied, suggesting that other determinants may create important functional differences between A-type K(+) currents.


Subject(s)
Basal Ganglia/physiology , Dendrites/physiology , Neurons/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium Channels/physiology , Prosencephalon/physiology , RNA, Messenger/genetics , Animals , Cell Membrane/physiology , Choline O-Acetyltransferase/analysis , Corpus Striatum/physiology , Electrophysiology/methods , Globus Pallidus/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neostriatum/physiology , Neurons/cytology , Organ Specificity , Polymerase Chain Reaction , Potassium Channels/classification , RNA, Messenger/metabolism , Rats , Shal Potassium Channels , Tetrodotoxin/pharmacology
9.
J Neurophysiol ; 83(1): 322-32, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634875

ABSTRACT

Adenosine is a potent regulator of acetylcholine release in the striatum, yet the mechanisms mediating this regulation are largely undefined. To begin to fill this gap, adenosine receptor expression and coupling to voltage-dependent Ca(2+) channels were studied in cholinergic interneurons by combined whole cell voltage-clamp recording and single-cell reverse transcription-polymerase chain reaction. Cholinergic interneurons were identified by the presence of choline acetyltransferase mRNA. Nearly all of these interneurons (90%, n = 28) expressed detectable levels of A(1) adenosine receptor mRNA. A(2a) and A(2b) receptor mRNAs were less frequently detected. A(3) receptor mRNA was undetectable. Adenosine rapidly and reversibly reduced N-type Ca(2+) currents in cholinergic interneurons. The A(1) receptor antagonist 8-cyclopentyl-1, 3-dimethylxanthine completely blocked the effect of adenosine. The IC(50) of the A(1) receptor selective agonist 2-chloro-N6-cyclopentyladenosine was 45 nM, whereas it was near 30 microM for the A(2a) receptor agonist CGS-21680. Dialysis with GDPbetaS or brief exposure to the G protein (G(i/o)) alkylating agent N-ethylmaleimide also blocked the adenosine modulation. The reduction in N-type currents was partially reversed by depolarizing prepulses. A membrane-delimited pathway mediated the modulation, because it was not seen in cell-attached patches when agonist was applied to the bath. Activation of protein kinase C attenuated the adenosine modulation. Taken together, our results argue that activation of A(1) adenosine receptors in cholinergic interneurons reduces N-type Ca(2+) currents via a membrane-delimited, G(i/o) class G-protein pathway that is regulated by protein kinase C. These observations establish a cellular mechanism by which adenosine may serve to reduce acetylcholine release.


Subject(s)
Calcium Channels, N-Type/physiology , Corpus Striatum/physiology , Interneurons/physiology , Receptors, Purinergic P1/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Calcium Channels, N-Type/drug effects , Choline O-Acetyltransferase/genetics , Ethylmaleimide/pharmacology , GTP-Binding Proteins/metabolism , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , In Vitro Techniques , Interneurons/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Phenethylamines/pharmacology , RNA, Messenger/analysis , Rats , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Theophylline/analogs & derivatives , Theophylline/pharmacology , Thionucleotides/pharmacology
10.
Nature ; 401(6750): 272-6, 1999 Sep 16.
Article in English | MEDLINE | ID: mdl-10499584

ABSTRACT

Acute exposure to cocaine transiently induces several Fos family transcription factors in the nucleus accumbens, a region of the brain that is important for addiction. In contrast, chronic exposure to cocaine does not induce these proteins, but instead causes the persistent expression of highly stable isoforms of deltaFosB. deltaFosB is also induced in the nucleus accumbens by repeated exposure to other drugs of abuse, including amphetamine, morphine, nicotine and phencyclidine. The sustained accumulation of deltaFosB in the nucleus accumbens indicates that this transcription factor may mediate some of the persistent neural and behavioural plasticity that accompanies chronic drug exposure. Using transgenic mice in which deltaFosB can be induced in adults in the subset of nucleus accumbens neurons in which cocaine induces the protein, we show that deltaFosB expression increases the responsiveness of an animal to the rewarding and locomotor-activating effects of cocaine. These effects of deltaFosB appear to be mediated partly by induction of the AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazole) glutamate receptor subunit GluR2 in the nucleus accumbens. These results support a model in which deltaFosB, by altering gene expression, enhances sensitivity to cocaine and may thereby contribute to cocaine addiction.


Subject(s)
Cocaine/pharmacology , Nucleus Accumbens/drug effects , Proto-Oncogene Proteins c-fos/physiology , Animals , Gene Transfer Techniques , Genetic Vectors , Male , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Nucleus Accumbens/physiology , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Receptors, AMPA/genetics , Receptors, AMPA/physiology , Simplexvirus/genetics
11.
J Neurosci ; 19(17): 7268-77, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10460233

ABSTRACT

In brain neurons, P- and Q-type Ca(2+) channels both appear to include a class A alpha1 subunit. In spite of this similarity, these channels differ pharmacologically and biophysically, particularly in inactivation kinetics. The molecular basis for this difference is unclear. In heterologous systems, alternative splicing and ancillary beta subunits have been shown to alter biophysical properties of channels containing a class A alpha1 subunit. To test the hypothesis that similar mechanisms are at work in native systems, P- and Q-type currents were characterized in acutely isolated rat neostriatal, medium spiny neurons and cortical pyramidal neurons using whole-cell voltage-clamp techniques. Cells were subsequently aspirated and subjected to single-cell RT-PCR (scRT-PCR) analysis of calcium channel alpha(1) and beta (beta(1-4)) subunit expression. In both cortical and neostriatal neurons, P- and Q-type currents were found in cells expressing class A alpha(1) subunit mRNA. Although P-type currents in cortical and neostriatal neurons were similar, Q-type currents differed significantly in inactivation kinetics. Notably, Q-type currents in neostriatal neurons were similar to P-type currents in inactivation rate. The variation in Q-type channel biophysics was correlated with beta subunit expression. Neostriatal neurons expressed significantly higher levels of beta(2a) mRNA and lower levels of beta(1b) mRNA than cortical neurons. These findings are consistent with the association of beta(2a) and beta(1b) subunits with slow and fast inactivation, respectively. Analysis of alpha(1A) splice variants in the linker between domains I and II failed to provide an alternative explanation for the differences in inactivation rates. These findings are consistent with the hypothesis that the biophysical properties of Q-type channels are governed by beta subunit isoforms and are separable from toxin sensitivity.


Subject(s)
Calcium Channels, N-Type , Calcium Channels/physiology , Cerebral Cortex/physiology , Gene Expression Regulation , Neostriatum/physiology , Neurons/physiology , Pyramidal Cells/physiology , Transcription, Genetic , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/genetics , In Vitro Techniques , Kinetics , Macromolecular Substances , Neurons/drug effects , Peptides/pharmacology , Pyramidal Cells/drug effects , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tetrodotoxin/pharmacology , omega-Conotoxin GVIA
12.
J Neurosci ; 19(15): 6394-404, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10414968

ABSTRACT

The symptoms of Parkinson disease are thought to result in part from increased burst activity in globus pallidus neurons. To gain a better understanding of the factors governing this activity, we studied delayed rectifier K(+) conductances in acutely isolated rat globus pallidus (GP) neurons, using whole-cell voltage-clamp and single-cell RT-PCR techniques. From a holding potential of -40 mV, depolarizing voltage steps in identified GP neurons evoked slowly inactivating K(+) currents. Analysis of the tail currents revealed rapidly and slowly deactivating currents of similar amplitude. The fast component of the current deactivated with a time constant of 11. 1 +/- 0.8 msec at -40 mV and was blocked by micromolar concentrations of 4-AP and TEA (K(D) approximately 140 microM). The slow component of the current deactivated with a time constant of 89 +/- 10 microseconds at -40 mV and was less sensitive to TEA (K(D) = 0.8 mM) and 4-AP (K(D) approximately 6 mM). Organic antagonists of Kv1 family channels had little or no effect on somatic currents. These properties are consistent with the hypothesis that the rapidly deactivating current is attributable to Kv3.1/3.2 channels and the slowly deactivating current to Kv2.1-containing channels. Semiquantitative single-cell RT-PCR analysis of Kv3 and Kv2 family mRNAs supported this conclusion. An alteration in the balance of these two channel types could underlie the emergence of burst firing after dopamine-depleting lesions.


Subject(s)
Globus Pallidus/physiology , Neurons/physiology , Neuropeptides/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , 4-Aminopyridine/pharmacology , Animals , Delayed Rectifier Potassium Channels , Electric Conductivity , Globus Pallidus/cytology , Globus Pallidus/metabolism , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Potassium Channel Blockers , Potassium Channels/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Shab Potassium Channels , Shaw Potassium Channels , Tetraethylammonium/pharmacology
13.
J Neurosci ; 18(17): 6650-61, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9712637

ABSTRACT

Inwardly rectifying K+ (IRK) channels are critical for shaping cell excitability. Whole-cell patch-clamp and single-cell RT-PCR techniques were used to characterize the inwardly rectifying K+ currents found in projection neurons of the rat nucleus accumbens. Inwardly rectifying currents were highly selective for K+ and blocked by low millimolar concentrations of Cs+ or Ba2+. In a subset of neurons, the inwardly rectifying current appeared to inactivate at hyperpolarized membrane potentials. In an attempt to identify this subset, neurons were profiled using single-cell RT-PCR. Neurons expressing substance P mRNA exhibited noninactivating inward rectifier currents, whereas neurons expressing enkephalin mRNA exhibited inactivating inward rectifier currents. The inactivation of the inward rectifier was correlated with the expression of IRK1 mRNA. These results demonstrate a clear physiological difference in the properties of medium spiny neurons and suggest that this difference could influence active state transitions driven by cortical and hippocampal excitatory input.


Subject(s)
Nucleus Accumbens/physiology , Peptide Fragments/physiology , Potassium Channels/physiology , Animals , Nucleus Accumbens/cytology , Patch-Clamp Techniques , Polymerase Chain Reaction/methods , Rats , Transcription, Genetic
14.
Neuroreport ; 9(9): 1935-9, 1998 Jun 22.
Article in English | MEDLINE | ID: mdl-9674570

ABSTRACT

Semi-quantitative single cell RT-PCR techniques were used to determine the expression of mRNAs related to GABAergic and cholinergic neurotransmission in neurons of the rat globus pallidus and adjacent basal forebrain region. Neurons of the globus pallidus expressed relatively high levels of GAD67 and GABA vesicular transporter mRNA but undetectable levels of ChAT or ACh vesicular transporter mRNA. In contrast, nominally basal forebrain neurons co-expressed ChAT and GAD67 mRNAs and mRNAs for both ACh and GABA vesicular transporters. These results suggest that the neurons along the medial border of the globus pallidus may co-release GABA and ACh.


Subject(s)
Globus Pallidus/metabolism , Membrane Transport Proteins , Neurons/metabolism , Organic Anion Transporters , Parasympathetic Nervous System/metabolism , Prosencephalon/metabolism , RNA, Messenger/biosynthesis , Vesicular Transport Proteins , gamma-Aminobutyric Acid/physiology , Animals , Biomarkers , Carrier Proteins/biosynthesis , Choline O-Acetyltransferase/biosynthesis , DNA/biosynthesis , DNA/genetics , GABA Plasma Membrane Transport Proteins , Globus Pallidus/cytology , Glutamate Decarboxylase/biosynthesis , Membrane Proteins/biosynthesis , Parasympathetic Nervous System/drug effects , Parasympathetic Nervous System/enzymology , Polymerase Chain Reaction , Prosencephalon/cytology , Rats , Vesicular Acetylcholine Transport Proteins
15.
J Neurosci ; 18(9): 3124-37, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9547221

ABSTRACT

Unlike other neostriatal neurons, cholinergic interneurons exhibit spontaneous, low-frequency, repetitive firing. To gain an understanding of the K+ channels regulating this behavior, acutely isolated adult rat cholinergic interneurons were studied using whole-cell voltage-clamp and single-cell reverse transcription-PCR techniques. Cholinergic interneurons were identified by the presence of choline acetyltransferase (ChAT) mRNA. Depolarization-activated potassium currents in cholinergic interneurons were dominated by a rapidly inactivating, K+-selective A current that became active at subthreshold potentials. Depolarizing prepulses inactivated this component of the current, leaving a delayed, rectifier-like current. Micromolar concentrations of Cd2+ dramatically shifted the voltage dependence of the A current without significantly affecting the delayed rectifier. The A-channel antagonist 4-aminopyridine (4-AP) produced a voltage-dependent block (IC50, approximately 1 mM) with a prominent crossover at millimolar concentrations. On the other hand, TEA preferentially blocked the sustained current component at concentrations <10 mM. Single-cell mRNA profiling of subunits known to give rise to rapidly inactivating K+ currents revealed the coexpression of Kv4.1, Kv4.2, and Kv1.4 mRNAs but low or undetectable levels of Kv4.3 and Kv3.4 mRNAs. Kv1.1, beta1, and beta2 subunit mRNAs, but not beta3, were also commonly detected. The inactivation recovery kinetics of the A-type current were found to match those of Kv4.2 and 4.1 channels and not those of Kv1.4 or Kv1. 1 and beta1 channels. Immunocytochemical analysis confirmed the presence of Kv4.2 but not Kv1.4 subunits in the somatodendritic membrane of ChAT-immunoreactive neurons. These results argue that the depolarization-activated somatodendritic K+ currents in cholinergic interneurons are dominated by Kv4.2- and Kv4. 1-containing channels. The properties of these channels are consistent with their playing a prominent role in governing the slow, repetitive discharge of interneurons seen in vivo.


Subject(s)
Acetylcholine/physiology , Dendrites/physiology , Interneurons/physiology , Neostriatum/physiology , Peptide Fragments/physiology , Potassium Channels/physiology , 4-Aminopyridine/pharmacology , Animals , Cadmium/pharmacology , Dendrites/drug effects , Interneurons/drug effects , Interneurons/ultrastructure , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neostriatum/cytology , Neostriatum/drug effects , Patch-Clamp Techniques , Potassium Channels/drug effects , Rats , Tetraethylammonium/pharmacology
16.
Biochim Biophys Acta ; 1261(2): 243-8, 1995 Apr 04.
Article in English | MEDLINE | ID: mdl-7536040

ABSTRACT

The transcription of the mts1 gene putatively involved in the control of tumor metastasis was studied in three human lymphoma cell lines: MOLT-4, CEM and Jurkat. The level of the mts1 gene transcription is high in MOLT-4 cells, lower in CEM cells and hardly detectable in Jurkat cells. This correlates with the hypomethylation of DNA in the first exon and the first intron of the mts1 gene in the analyzed culture cells. This area was also found to be undermethylated in human peripheral blood cells--macrophages, neutrophils and lymphocytes where the mts 1 gene is highly expressed. 5-Azadeoxycytidine (AzadC)--an inhibitor of the eukaryotic DNA-methylase--significantly induces the expression of the mts1 gene in CEM and Jurkat cells and has little effect on mts1 gene transcription in MOLT-4 cells. The drug does not influence mts1 transcription in cultivated peripheral blood lymphocytes. These data indicate the possible involvement of the methylation of the first exon/first intron sequences in the transcriptional repression of the mts1 gene. The finding of two DNAaseI hypersensitivity sites (DHSs) mapped in the first intron of the mts1 gene supports this suggestion.


Subject(s)
DNA Modification Methylases/genetics , Lymphoma/genetics , Neoplasm Metastasis/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Decitabine , Gene Expression Regulation/drug effects , Humans , Methylation , Restriction Mapping , Transcription, Genetic/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...