Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Psychiatry ; 20(8): 959-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25266126

ABSTRACT

Neuregulin 1 (NRG1) is a trophic factor that has an essential role in the nervous system by modulating neurodevelopment, neurotransmission and synaptic plasticity. Despite the evidence that NRG1 and its receptors, ErbB tyrosine kinases, are expressed in mesencephalic dopaminergic nuclei and their functional alterations are reported in schizophrenia and Parkinson's disease, the role of NRG1/ErbB signalling in dopaminergic neurons remains unclear. Here we found that NRG1 selectively increases the metabotropic glutamate receptor 1 (mGluR1)-activated currents by inducing synthesis and trafficking to membrane of functional receptors and stimulates phosphatidylinositol 3-kinase-Akt-mammalian target of rapamycin (PI3K-Akt-mTOR) pathway, which is required for mGluR1 function. Notably, an endogenous NRG1/ErbB tone is necessary to maintain mGluR1 function, by preserving its surface membrane expression in dopaminergic neurons. Consequently, it enables striatal mGluR1-induced dopamine outflow in in vivo conditions. Our results identify a novel role of NRG1 in the dopaminergic neurons, whose functional alteration might contribute to devastating diseases, such as schizophrenia and Parkinson's disease.


Subject(s)
Dopaminergic Neurons/physiology , Mesencephalon/physiology , Neuregulin-1/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Dopamine/metabolism , Dopaminergic Neurons/drug effects , ErbB Receptors/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mesencephalon/drug effects , Microdialysis , Patch-Clamp Techniques , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tissue Culture Techniques
2.
Br J Pharmacol ; 153(5): 1022-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18223675

ABSTRACT

BACKGROUND AND PURPOSE: Reactive oxygen species (ROS) have been postulated to play a crucial role in the pathogenesis of ischaemia-reperfusion injury. Among these, hydrogen peroxide (H(2)O(2)) is known to be a toxic compound responsible for free-radical-dependent neuronal damage. In recent years, however, the 'bad reputation' of H(2)O(2) and other ROS molecules has changed. The aim of this study was to assess the protective role of H(2)O(2) and modification in its endogenous production on the electrophysiological and morphological changes induced by oxygen/glucose deprivation (OGD) on CA1 hippocampal neurons. EXPERIMENTAL APPROACH: Neuroprotective effects of exogenous and endogenous H(2)O(2) were determined using extracellular electrophysiological recordings of field excitatory post synaptic potentials (fEPSPs) and morphological studies in a hippocampal slice preparation. In vitro OGD was delivered by switching to an artificial cerebrospinal fluid solution with no glucose and with oxygen replaced by nitrogen. KEY RESULTS: Neuroprotection against in vitro OGD was observed in slices treated with H(2)O(2) (3 mM). The rescuing action of H(2)O(2) was mediated by catalase as pre-treatment with the catalase inhibitor 3-amino-1,2,4-triazole blocked this effect. More interestingly, we showed that an increase of the endogenous levels of H(2)O(2), due to a combination of an inhibitor of the glutathione peroxidase enzyme and addition of Cu,Zn-superoxide dismutase in the tissue bath, prevented the OGD-induced irreversible depression of fEPSPs. CONCLUSIONS AND IMPLICATIONS: Taken together, our results suggest new possible strategies to lessen the damage produced by a transient brain ischaemia by increasing the endogenous tissue level of H(2)O(2).


Subject(s)
Brain Ischemia/drug therapy , Hydrogen Peroxide/pharmacology , Neuroprotective Agents/pharmacology , Animals , Brain Ischemia/physiopathology , Catalase/drug effects , Catalase/metabolism , Disease Models, Animal , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Hydrogen Peroxide/metabolism , In Vitro Techniques , Male , Neuroprotective Agents/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Rats , Rats, Wistar
3.
Neuroscience ; 118(2): 399-408, 2003.
Article in English | MEDLINE | ID: mdl-12699776

ABSTRACT

Apart from the extensive loss of motor neurons, degeneration of midbrain dopaminergic cells has been described in both familial and sporadic forms of amyotrophic lateral sclerosis (ALS). Mice overexpressing the mutant human Cu/Zn superoxide dismutase (SOD1) show an ALS-like phenotype in that they show a progressive death of motor neurons accompanied by degeneration of dopaminergic cells. To describe the functional alterations specifically associated with this dopaminergic dysfunction, we have investigated the corticostriatal synaptic plasticity in mice overexpressing the human SOD1 (SOD1+) and the mutated (Gly(93)-->Ala) form (G93A+) of the same enzyme. We show that repetitive stimulation of the corticostriatal pathway generates long-term depression (LTD) in SOD1+ mice and in control (G93A-/SOD1-) animals, whereas in G93A+ mice the same stimulation generates an N-methyl-D-aspartic acid receptor-dependent long-term potentiation. No significant alterations were found in the intrinsic membrane properties of striatal medium spiny neurons and basal corticostriatal synaptic transmission of G93A+ mice. Bath perfusion of dopamine or the D(2) dopamine receptor agonist quinpirole restored LTD in G93A+ mice. Consistent with these in vitro results, habituation of locomotor activity and striatal-dependent active avoidance learning were impaired in G93A+ mice. Thus, degeneration of dopaminergic neurons in the substantia nigra of G93A+ mice causes substantial modifications in striatal synaptic plasticity and related behaviors, and may be a cellular substrate of the extrapyramidal motor and cognitive disorders observed in familial and sporadic ALS.


Subject(s)
Long-Term Potentiation/physiology , Mutation , Neuronal Plasticity/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Valine/analogs & derivatives , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Alanine/genetics , Animals , Avoidance Learning/physiology , Calcium/metabolism , Cell Membrane/physiology , Corpus Striatum/physiology , Disease Models, Animal , Dopamine Agonists/pharmacology , Electric Stimulation , Genotype , Glycine/genetics , Gyrus Cinguli/anatomy & histology , Gyrus Cinguli/physiology , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Mice, Transgenic , Motor Activity/drug effects , Neural Pathways , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Quinpirole/pharmacology , Valine/pharmacology
4.
Eur J Neurosci ; 14(9): 1492-504, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11722611

ABSTRACT

Transgenic Huntington's disease (HD) mice, expressing exon 1 of the human HD gene (lines R6/1 and R6/2), are totally resistant to striatal lesions caused by the NMDA receptor agonist quinolinic acid (QA). Here we show that this resistance develops gradually over time in both R6/1 and R6/2 mice, and that it occurred earlier in R6/2 (CAG-155) than in R6/1 (CAG-115) mice. The development of the resistance coincided with the appearance of nuclear inclusions and with the onset of motor deficits. In the HD mice, hippocampal neurons were also resistant to QA, especially in the CA1 region. Importantly, there was no change in susceptibility to QA in transgenic mice with a normal CAG repeat (CAG-18). R6/1 mice were also resistant to NMDA-, but not to AMPA-induced striatal damage. Interestingly, QA-induced current and calcium influx in striatal R6/2 neurons were not decreased. However, R6/2 neurons had a better capacity to handle cytoplasmic calcium ([Ca2+]c) overload following QA and could avoid [Ca2+]c deregulation and cell lysis. In addition, basal [Ca2+]c levels were increased five-fold in striatal R6/2 neurons. This might cause an adaptation of R6 neurons to excitotoxic stress resulting in an up-regulation of defense mechanisms, including an increased capacity to handle [Ca2+]c overload. However, the increased level of basal [Ca2+]c in the HD mice might also disturb intracellular signalling in striatal neurons and thereby cause neuronal dysfunction and behavioural deficits.


Subject(s)
Brain/drug effects , Calcium/metabolism , Drug Resistance/genetics , Excitatory Amino Acid Agonists/toxicity , Huntington Disease/genetics , Nerve Degeneration/genetics , Receptors, N-Methyl-D-Aspartate/drug effects , Aging/genetics , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Brain/physiopathology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Survival/drug effects , Cell Survival/physiology , Exons/genetics , Female , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Homeostasis/genetics , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/physiopathology , Immunohistochemistry , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Male , Mice , Mice, Transgenic , Neostriatum/drug effects , Neostriatum/metabolism , Neostriatum/physiopathology , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Trinucleotide Repeats/genetics
5.
J Neurophysiol ; 85(6): 2490-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11387395

ABSTRACT

Metabotropic glutamate receptors (mGluRs) modulate neuronal function via different transduction mechanisms that are either dependent or independent on G-protein function. Here we investigated, using whole cell patch-clamp recordings in combination with fluorimetric measurements of intracellular calcium concentration ([Ca(2+)](i)), the metabolic pathways involved in the responses induced by group I mGluRs in dopamine neurons of the rat midbrain. The inward current and the [Ca(2+)](i) increase caused by the group I mGluR agonist (S)-3,5-dihydroxyphenylglycine (DHPG, 100 microM) were permanently activated and subsequently abolished in cells loaded with the nonhydrolizable GTP-analogue GTP-gamma-S (600 microM). In addition, when GDP-beta-S (600 microM) was dialyzed into the cells to produce the blockade of the G proteins, the DHPG-dependent responses were reduced. When the tissue was bathed with the phospholipase C inhibitor 1-[6[[(17 beta)-3-methoxyestra-1,3,5(10)-trien-17-yl]amino]exyl]-1H-pyrrole-2,5-dione (10 microM), the DHPG-induced calcium transients slightly diminished but the associated inward currents were not affected. Interestingly, a substantial depression of the DHPG-induced inward current and transient increase of [Ca(2+)](i) was caused by the protein tyrosine kinase inhibitors tyrphostin B52 (40 microM) and 4',5,7-trihydroxyisoflavone (genistein; 40 microM), whereas genistein's inactive analogue 4',5,7-trihydroxyisoflavone-7-glucoside (40 microM) was ineffective. The blockade of the Src family of tyrosine kinase by 4-amino-5-(4-methylphenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (20 microM), mitogen-activated protein kinase by 2'-amino-3' methoxyflavone (50 microM), and protein kinase C by staurosporine (1 microM) had no effect on the cellular responses caused by DHPG. The mGluR5-selective antagonist 2-methyl-6-(phenylethynyl)-pyridine (10--100 microM) did not affect the actions of DHPG. Thus our results indicate that the responses, mainly mediated by mGluRs1 in dopamine neurons, are activated by intracellular mechanisms coupled to G proteins and regulated by tyrosine kinases.


Subject(s)
Dopamine/physiology , GTP-Binding Proteins/metabolism , Guanosine Diphosphate/analogs & derivatives , Neurons/enzymology , Protein-Tyrosine Kinases/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Genistein/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Diphosphate/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mesencephalon/cytology , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Protein Kinase C/metabolism , Pyrrolidinones/pharmacology , Rats , Rats, Wistar , Resorcinols/pharmacology , Thionucleotides/pharmacology , Type C Phospholipases/metabolism , Tyrphostins/pharmacology
6.
J Neurosci ; 20(16): 6013-20, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10934250

ABSTRACT

In the present study, we characterized the intrinsic electrophysiological properties and the membrane currents activated by dopamine (DA) D(2) and GABA(B) receptors in midbrain dopaminergic neurons, maintained in vitro in a slice preparation, from wild-type and homozygous weaver (wv/wv) mice. By using patch-clamp techniques, we found that membrane potential, apparent input resistance, and spontaneous firing of wv/wv dopaminergic neurons were similar to those of dopamine-containing cells recorded from nonaffected (+/+) animals. More interestingly, the wv/wv neurons were excited rather than inhibited by dopamine and the GABA(B) agonist baclofen. This neurotransmitter-mediated excitation was attributable to the activation of a G-protein-gated inward current that reversed polarity at a membrane potential of approximately -30 mV. We suggest that the altered behavior of the receptor-operated wv G-protein-gated inwardly rectifying K(+) channel 2 (GIRK2) might be related to the selective degeneration of the dopaminergic neurons. In addition, the wv GIRK2 would not only suppress the autoreceptor-mediated feedback inhibition of DA release but could also establish a feedforward mechanism of DA release in the terminal fields.


Subject(s)
Dopamine/metabolism , Mesencephalon/metabolism , Mice, Neurologic Mutants/metabolism , Neurons/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Baclofen/pharmacology , Dopamine/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , In Vitro Techniques , Mesencephalon/cytology , Mice , Mice, Neurologic Mutants/anatomy & histology , Mice, Neurologic Mutants/genetics , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neurons/cytology , Potassium Channels/drug effects , Potassium Channels/genetics , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Receptors, GABA-B/drug effects , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/pharmacology
7.
J Neurophysiol ; 82(4): 1974-81, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10515987

ABSTRACT

Metabotropic glutamate receptors modulate neuronal excitability via a multitude of mechanisms, and they have been implicated in the pathogenesis of neurodegenerative processes. Here we investigated the responses mediated by group I metabotropic glutamate receptors (mGluRs) in dopamine neurons of the rat substantia nigra pars compacta, using whole cell patch-clamp recordings in combination with microfluorometric measurements of [Ca(2+)](i) and [Na(+)](i). The selective group I mGluR agonist (S)-3,5-dihydroxyphenylglycine (3,5-DHPG) was bath-applied (20 microM, 30 s to 2 min) or applied locally by means of short-lasting (2-4 s) pressure pulses, delivered through an agonist-containing pipette positioned close to the cell body of the neuron. 3,5-DHPG evoked an inward current characterized by a transient and a sustained component, the latter of which was uncovered only with long-lasting agonist applications. The fast component coincided with a transient elevation of [Ca(2+)](i), whereas the total current was associated with a rise in [Na(+)](i). These responses were not affected either by the superfusion of ionotropic excitatory amino acid antagonists 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) and D-2-amino-5-phosphono-pentanoic acid (D-APV), nor by the sodium channel blocker tetrodotoxin (TTX). (S)-alpha-methyl-4-carboxyphenylglycine (S-MCPG) and the more selective mGluR1 antagonist 7(hydroxyimino)cyclopropa[b]chromen-1a-carboxylate (CPCCOEt) depressed both 3,5-DHPG-induced inward current components and, although less effectively, the associated [Ca(2+)](i) elevations. On repeated agonist applications the inward current and the calcium transients both desensitized. The time constant of recovery from desensitization differed significantly between these two responses, being 67.4+/-4.4 s for the inward current and 28.6+/-2.7 s for the calcium response. Bathing the tissue in a calcium-free/EGTA medium or adding thapsigargin (1 microM) to the extracellular medium prevented the generation of the [Ca(2+)](i) transient, but did not prevent the activation of the inward current. These electrophysiological and fluorometric results show that the 3, 5-DHPG-induced inward current and the [Ca(2+)](i) elevations are mediated by independent pathways downstream the activation of mGluR1.


Subject(s)
Calcium/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Neurons/physiology , Receptors, Metabotropic Glutamate/physiology , Substantia Nigra/physiology , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Calcium/pharmacology , Chromones/pharmacology , Cycloleucine/analogs & derivatives , Cycloleucine/pharmacology , Egtazic Acid/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , In Vitro Techniques , Microscopy, Fluorescence , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Rats , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Resorcinols/pharmacology , Tetrodotoxin/pharmacology , Thapsigargin/pharmacology
8.
J Neurophysiol ; 80(5): 2237-43, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9819239

ABSTRACT

We investigated the hypoxia-induced disturbance of cytosolic sodium concentration ([Na+]i) and of cytosolic calcium concentration ([Ca2+]i) in dopamine neurons of the substantia nigra pars compacta in rat midbrain slices, by combining whole cell patch-clamp recordings and microfluorometry. Transient hypoxia (3-5 min) induced an outward current (118.7 +/- 15.1 pA, mean +/- SE; VH = -60 mV). The development of this outward current was associated with an elevation in [Na+]i and in [Ca2+]i. The hypoxia-induced outward current as well as the elevations in [Na+]i and [Ca2+]i were not affected by the ionotropic and metabotropic glutamate receptor antagonists -amino-phosphonovalerate (50 microM), 6nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione (10 microM) and S-(alpha)-methyl-4-carboxyphenylglycine (500 microM). Tolbutamide, a blocker of ATP-dependent K+ channels, depressed the hypoxia-induced outward current but did not affect the increases in [Na+]i or [Ca2+]i. Increasing the concentration of ATP in the internal solution from 2 to 10 mM strongly reduced the hypoxia-induced outward current but did not reduce the rise in [Na+]i. Decreasing the concentration of extracellular Na+ to 19.2 mM depressed the hypoxia-induced outward current and resulted in a decrease in resting [Na+]i. Under this condition hypoxia still increased [Na+]i, albeit to levels not exceeding those of resting [Na+]i observed under control conditions. We conclude that 1) a major component of the hypoxia-induced outward current of these cells is caused by a depletion of intracellular ATP in combination with an increase in [Na+]i, 2) that the [Na+]i and [Ca2+]i responses are not mediated by glutamate receptors, 3) that the [Na+]i and [Ca2+]i responses are not depressed by activation of sulfonylurea receptors, and 4) that the rise in [Na+]i induced by short-lasting hypoxia is not due to a ATP depletion-induced failure of Na+ extrusion.


Subject(s)
Calcium/metabolism , Dopamine/metabolism , Homeostasis/physiology , Neurons/metabolism , Sodium/metabolism , Substantia Nigra/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Excitatory Amino Acid Antagonists/pharmacology , Fluorometry , Homeostasis/drug effects , In Vitro Techniques , Patch-Clamp Techniques , Potassium Channel Blockers , Rats , Substantia Nigra/cytology , Substantia Nigra/drug effects , Tolbutamide/pharmacology
9.
Synapse ; 30(4): 371-9, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9826229

ABSTRACT

We used field potential recording techniques to examine whether felbamate (FBM), lamotrigine (LTG), and lidocaine (LID) protect against the irreversible functional damage induced by transient ischemia. Five minutes of ischemia caused a depression of the field potential in rat cortical slices, which did not recover even after more than 1 h of washout. The N-methyl-D-aspartate (NMDA) antagonist ketamine (50 microM) protected against depression of the field caused by ischemia. On the other hand, the non-NMDA antagonist 6-cyano-7-nitroquinoxaline-2.3-dione (CNQX) (10 microM) had protective effects only if co-applied with ketamine. We found that either FBM (30-300 microM), which did not modify the amplitude of the field EPSP, or LTG (10-300 microM), which reversibly depressed the excitatory synaptic transmission, had a marked protective effect when superfused before and during the ischemic insult. After FBM (100 microM) and LTG (100 microM), the field EPSP recovered by 84 +/- 1% and 73 +/- 2.7% of control, respectively. Furthermore, LID (30-300 microM) was less effective than FBM and LTG in inducing a functional recovery from the damage caused by ischemia (58 +/- 1.8%). The rank order of potency, based on the maximal protection caused by the three drugs, was FBM > LTG > LID. Our results suggest that a noticeable neuroprotection can be obtained during glucose and O2 deprivation by preventive therapeutic regimens which use the two recently marketed anticonvulsant drugs, FBM and LTG.


Subject(s)
Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Ischemia/physiopathology , Lidocaine/pharmacology , Neuroprotective Agents/pharmacology , Propylene Glycols/pharmacology , Triazines/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , Felbamate , In Vitro Techniques , Lamotrigine , Male , Phenylcarbamates , Rats , Rats, Wistar
10.
Eur J Neurosci ; 10(5): 1926-9, 1998 May.
Article in English | MEDLINE | ID: mdl-9751162

ABSTRACT

We investigated the effect of changes in membrane-voltage on intracellular sodium concentration ([Na+]i) of dopamine-sensitive neurons of the substantia nigra pars compacta in a slice preparation of rat mesencephalon. Whole-cell patch-clamp techniques were combined with microfluorometric measurements of [Na+]i using the Na+-sensitive probe, sodium-binding benzofuran isophthalate (SBFI). Hyperpolarization of spontaneously active dopamine neurons (recorded in current-clamp mode) caused the cessation of action potential firing accompanied by an elevation in [Na+]i. In dopamine neurons voltage-clamped at a holding potential of -60 mV elevations of [Na+]i were induced by long-lasting (45-60 s) voltage jumps to more negative membrane potentials (-90 to -120 mV) but not by corresponding voltage jumps to -30 mV. These hyperpolarization-induced elevations of [Na+]i were depressed during inhibition of I(h), a hyperpolarization-activated inward current, by Cs+. Hyperpolarization-induced elevations in [Na+]i might occur also in other cell types which express a powerful I(h) and might signal lack of postsynaptic activity.


Subject(s)
Dopamine/physiology , Sodium/metabolism , Substantia Nigra/metabolism , Animals , In Vitro Techniques , Membrane Potentials/physiology , Patch-Clamp Techniques , Rats , Substantia Nigra/cytology
11.
J Neurophysiol ; 79(3): 1239-45, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9497405

ABSTRACT

The effects of brief (2-4 min) hypoxia on presumed dopaminergic "principal" neurons of the rat ventral mesencephalon were investigated by using either intracellular or whole cell patch-clamp recordings in in vitro conditions. Under single-electrode voltage clamp, with sharp microelectrode (Vh -60 mV), a brief hypoxia caused an outward current (hypoOUT) of 110.2 +/- 15.2 (SE) pA (n = 18), which was followed by a posthypoxic outward current (posthypoOUT) of 149.6 +/- 10.6 pA (n = 18). Although the hypoOUT reversed at -83.7 +/- 3.8 mV (n = 18), the posthypoOUT did not reverse. The K+ATP-blocking sulphonylureas tolbutamide (100 microM) and glibenclamide (30 microM), significantly reduced the peak of the hypoOUT by 47.6 +/- 7.7% (n = 16) and 54.18 +/- 7.5% (n = 3), respectively. In contrast, they did not affect the posthypoOUT. Extracellular barium (300 microM to 1 mM) almost abolished the hypoOUT, leaving the posthypoOUT unchanged. The large K+ channel blocker charybdotoxin (10-50 nM), depressed the hypoOUT after tolbutamide treatment. To investigate whether or not cytosolic factors might control the development of the hypoOUT, we dialyzed the principal neurons by patch-clamp recordings (Vh -60 mV). Under whole cell recordings hypoxia evoked an hypoOUT of 70.2 +/- 14.5 pA that reversed polarity at -87.9 +/- 5.1 mV (n = 8). A small posthypoxic response was detected upon reoxygenation in a few neurons (4 out of 14). Three different sulphonylureas, tolbutamide (100 microM), glibenclamide (10-30 microM), and glipizide (100 nM) completely blocked the hypoOUT in patch-clamped neurons. The hypoOUT was also abolished by extracellular BaCl2 (300 microM). When the content of ATP in the dialyzate was raised from 2 to 10 mM no outward current/hyperpolarization was evoked by hypoxia. These data suggest that the hypoOUT, in principal neurons, is a complex response sustained by at least two barium-sensitive components: 1) an ATP-dependent, sulphonylurea-sensitive K+ conductance which could be isolated by the patch-clamp techniques and 2) a K+ conductance remaining after tolbutamide in intracellularly recorded neurons, which is sensitive to charybdotoxin and dependent on dialyzable cytosolic factors.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Hypoxia , Dopamine/metabolism , Mesencephalon/physiology , Neurons/physiology , Potassium Channels/physiology , Sulfonylurea Compounds/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Barium Compounds/pharmacology , Charybdotoxin/pharmacology , Chlorides/pharmacology , Glipizide/pharmacology , Glyburide/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microelectrodes , Neurons/drug effects , Patch-Clamp Techniques , Potassium Channels/drug effects , Rats , Rats, Wistar , Reaction Time , Tolbutamide/pharmacology
12.
Trends Neurosci ; 19(12): 530-4, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8961481

ABSTRACT

A growing number of experimental studies have used patch-clamp amplifiers (PCAs) in the current-clamp (CC) mode to investigate classical excitability. In this paper we show that the measurements obtained in this way are affected by errors due to the electronic design of the PCA input section. We present experimental evidence of such errors, and demonstrate that they derive from PCA current absorption. Moreover, we propose a new PCA input-circuit configuration for the CC mode, which is suitable for accurately recording physiological voltage signals and is perfectly compatible with the standard voltage-clamp mode.


Subject(s)
Patch-Clamp Techniques/standards , Action Potentials/physiology , Artifacts , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods
13.
Neuroreport ; 7(15-17): 2575-9, 1996 Nov 04.
Article in English | MEDLINE | ID: mdl-8981426

ABSTRACT

Combretastatin B1, a polyhydroxybibenzyl compound extracted from the fruit of Combretum kraussii, known to contain 'hiccup nut' toxin, reversibly increased the duration, but not the peak or the rate of rise, of the action potential in rat sensory neurones by approximately 300%. This effect was only seen when it was applied to the extracellular side of the membrane. No effects on the resting potential were observed. K+ delayed rectifier currents were inhibited in neurones and in human myotubes with an IC50 of about 300 microM; the HERG-type inward rectifier channels in tumour cells were inhibited to a greater degree. Due to its selective action and the similarity of its blockade to that produced by class III antiarrhythmic drugs, the toxin could be the origin of compounds of potentially significant pharmacological interest.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Bibenzyls/pharmacology , Ganglia, Spinal/drug effects , Potassium Channels/drug effects , Stilbenes , Animals , Dose-Response Relationship, Drug , Humans , Rats
14.
Brain Res ; 741(1-2): 1-12, 1996 Nov 25.
Article in English | MEDLINE | ID: mdl-9001698

ABSTRACT

Pyramidal neurons were acutely isolated from neocortex slices of 14- to 20-day-old rats and patch-clamped under physiological conditions. Current-clamp recordings revealed firing patterns corresponding to those previously reported in slices as regular spiking (RS) and intrinsically bursting (IB), i.e., single action potentials (AP), trains of regular spikes and bursts with depolarizing after-potentials (DAP). In IB neurons, intracellular perfusion with KF blocked the high-voltage-activated Ca2+ and the Ca(2+)-dependent K+ currents, revealing APs with a 10-30 ms shoulder at -35 mV (shoulder AP), which was the supporting plateau of the intraburst spikes. The use of the A channel blocker, 4-aminopyridine, caused a three-fold reduction in the AP repolarizing rate. A study of the de- and repolarizing rates modulating the spike shape (shoulder AP, burst or single APs) suggested that the percentage of available A channels could play a crucial role in burst formation. Blockade of the residual T-type Ca2+ current by Ni2+ did not inhibit the AP shoulder, whereas it was completely and reversibly inhibited by 30 nM TTX, which did not affect AP amplitude. The AP rising rate was only halved by 100 nM TTX. The data concerning the A channel-mediated burst formation and the role of the TTX-sensitive conductance have been successfully simulated in a model cell. We suggest that bursting is an intrinsic property of the membrane of neocortex neurons, and is sustained by TTX-sensitive slowly inactivating and/or persistent Na+ conductances.


Subject(s)
Cerebral Cortex/physiology , Neural Conduction/drug effects , Pyramidal Cells/physiology , Tetrodotoxin/pharmacology , Animals , Axons/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Computer Simulation , Electrophysiology , Fluorides/metabolism , Membrane Potentials/physiology , Nickel/pharmacology , Patch-Clamp Techniques , Rats , Rats, Wistar
15.
Glia ; 16(3): 196-209, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8833190

ABSTRACT

Hyperpolarization-activated currents were recorded from rat brain cortical and spinal cord astrocytes maintained in culture. Spinal cord astrocytes expressed primarily an inward rectifier potassium current characterized by time-dependent inactivation, a strong dependence on extracellular Na+ and insensitivity to intracellular GTP-gamma-S (0.2 mM). In cortical astrocytes voltage clamp protocols aimed to elicit currents activated at, or negative to cell membrane potentials led to the development of two distinct ion currents. The most prominent current resembled the inward rectifier potassium current. This component was sensitive to blockade by extracellular cesium and was greatly reduced during recordings performed with GTP-gamma-S (0.2 Mm) added to the pipette solutions. The remaining current component was similar to the endothelial I ha current. I ha conductance was enhanced by extracellular potassium and the current reversal potential behaved as expected for a mixed cation, Na+/K+ current. I ha was nearly abolished after removal of extracellular Na. These results are consistent with the expression of a novel mixed cation conductance in glial cells, possibly involved in extracellular potassium buffering.


Subject(s)
Astrocytes/physiology , Cerebral Cortex/physiology , Spinal Cord/physiology , Animals , Cations/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Electrophysiology , Ions , Rats , Spinal Cord/cytology
16.
Neurotoxicology ; 17(2): 481-96, 1996.
Article in English | MEDLINE | ID: mdl-8856743

ABSTRACT

Cell culture models have been widely used for screening of neurotoxicants and represent a viable alternative to direct in vivo experiments. We have developed a dynamic in vitro blood-brain barrier model designed to allow for extensive toxicological, pharmacological and physiological testing. Induction of blood-brain barrier properties in a tri-dimensional hollow fiber culturing apparatus was investigated by co-culturing a bovine aortic endothelial cell line (or rat brain endothelial cells) with rat brain astrocytes (or C6 rat glioma cells) under pulsatile flow conditions to mimic intraluminal blood flow. Cell growth was monitored over time by measuring glucose consumption and lactate production: these experiments confirmed that the hollow fiber cell culturing systems can maintain viable cells in culture for extended (> 1 month) periods of time. Cells were visually inspected after culturing and dissociation from the hollow fiber cartridge and identified as endothelial (by fluorescent Dil-Ac-LDL uptake) or glial (by GFAP immunoreactivity). Blood-brain barrier properties were tested by intraluminal injection of horse-radish peroxidase (HRP, mol. weight 44,000), glucose (m.w. 180) or potassium. Either procedure demonstrated that aortic cells co-cultured with astrocytes (or C6 cells) developed a selective barrier with an estimated electrical resistance of 2,900 omega/cm2. The electrophysiological and morphological properties of BAEC were also affected by the co-culturing process, suggesting that astrocytes induced CNS properties in these cells. These results demonstrate that the hollow fiber cell co-culturing system may be used as a dynamic model of the mammalian blood-brain barrier.


Subject(s)
Blood-Brain Barrier/drug effects , Toxicity Tests/methods , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/physiology , Cattle , Cells, Cultured , Coculture Techniques , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Horseradish Peroxidase , Lactates/analysis , Patch-Clamp Techniques , Rats
17.
Brain Res ; 696(1-2): 127-39, 1995 Oct 23.
Article in English | MEDLINE | ID: mdl-8574660

ABSTRACT

In in vitro slices prepared from rat sensorimotor cortex, intracellular recordings were obtained from 107 layer V pyramidal neurons, subsequently injected with biocytin for morphological reconstruction. Of the 107 neurons, 59 (55.1%) were identified as adapting (45) or non-adapting (13) regular spiking neurons (RS), and 48 (44.9%) as intrinsically bursting (IB) neurons discharging with an initial cluster of action potentials, which tended to recur rhythmically in a subset of 19 cells. The block of IAR by extracellular Cs+ did not affect burst generation, but enhanced the tendency to reburst in IB neurons. A similar effect was induced by other procedures affecting K(+)-dependent post-burst hyperpolarization. In IB neurons Ca2+ spikes had a longer decay time than in RS neurons, however selective blockers of both low and high threshold Ca2+ conductances failed to impair bursting activity. On the contrary, the perfusion of the slices with 0.5-1 microM TTX suppressed bursting behaviour in a critical time interval preceding the complete block of Na(+)-dependent action potentials. It is concluded that the persistent Na+ current INAP is the most important intrinsic factor for the typical firing properties of IB neurons, while Ca2+ and K+ conductances appear to contribute towards shaping bursts and controlling their recurrence rate. The morphology, connectivity and physiological properties of adapting and non-adapting RS neurons are particularly suited to the processing of respectively phasic and tonic inputs, whereas the properties of IB neurons are consistent with their suggested role in cortical rhythmogenesis and in the pathophysiological synchronized activities underlying epileptogenesis.


Subject(s)
Motor Cortex/physiology , Pyramidal Cells/physiology , Somatosensory Cortex/physiology , Action Potentials/drug effects , Animals , Calcium Channel Blockers/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Motor Cortex/cytology , Motor Cortex/metabolism , Patch-Clamp Techniques , Potassium Channels/drug effects , Potassium Channels/metabolism , Rats , Rats, Wistar , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism , Tetraethylammonium Compounds/pharmacology , Tetrodotoxin/pharmacology
18.
Neurosci Lett ; 175(1-2): 117-20, 1994 Jul 04.
Article in English | MEDLINE | ID: mdl-7970191

ABSTRACT

Neurons acutely dissociated from neocortex slices of 14-16-day-old rats were patch-clamped in physiological conditions. Different pyramidal cells, spontaneously or in response to current steps, generate regular spiking and intrinsically bursting behaviour during long periods of time. We show that typical firing properties recorded in somatosensory neocortex slices are preserved in dissociated pyramidal neurons originating from the slices themselves, thus, providing a way for the related characterization of biophysical properties of currents in identified subtypes of pyramidal neurons.


Subject(s)
Cerebral Cortex/physiology , Neurons/physiology , Action Potentials , Animals , Electric Stimulation , Evoked Potentials , In Vitro Techniques , Pyramidal Tracts/physiology , Rats , Rats, Wistar , Time Factors
19.
Eur J Neurosci ; 6(3): 381-91, 1994 Mar 01.
Article in English | MEDLINE | ID: mdl-8019675

ABSTRACT

We studied, in rat sensory neurons, the modulation of high voltage-activated Ca2+ currents (ICa) mediated by the pertussis toxin-sensitive activation of muscarinic receptors, which were found to be of subtypes M2 or M4. Muscarine reversibly blocked somatic Ca2+ spikes but strong predepolarizations only partially relieved the inhibited Ca2+ current. On the other hand, the putative coupling messenger could not rapidly diffuse towards channels whose activity was recorded from a macro-patch. The perforated patch technique virtually prevented the response rundown present during whole-cell experiments. Both omega-conotoxin GVIA (omega-CgTx)-sensitive channels and omega-CgTx- and dihydropyridine-resistant channels are coupled to the muscarinic receptor, but not the L-channel. When measured in the same neuron, dose-response relationships for the first and subsequent agonist applications differed; maximal inhibition, the reciprocal of half-maximal concentration and the Hill coefficient were always highest in the first trial. Muscarine and oxotremorine exhibited monotone dose-response curves, but oxotremorine-M showed non-linear relationships which became monotonic when cells were intracellularly perfused with inhibitors of protein kinase A (PKA) and C (PKC), suggesting that either PKA or receptor-induced PKC could phosphorylate and thus inactive G-proteins or other unknown proteins involved in inhibitory muscarinic actions on ICa. In summary, these data provide a preliminary pharmacological characterization of the muscarinic inhibition of the Ca2+ channels in sensory neurons, with implications about agonist specificity and the interplay between signalling pathways.


Subject(s)
Calcium Channels/drug effects , Neurons, Afferent/drug effects , Receptors, Muscarinic/drug effects , Signal Transduction/drug effects , omega-Conotoxins , Action Potentials/drug effects , Animals , Calcium Channel Blockers/pharmacology , Electric Conductivity , In Vitro Techniques , Kinetics , Membrane Potentials/drug effects , Mollusk Venoms/pharmacology , Muscarine/pharmacology , Peptides/pharmacology , Protein Kinase Inhibitors , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...