Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-23459031

ABSTRACT

Juxtaglomerular neurons represent one of the largest cellular populations in the mammalian olfactory bulb yet their role for signal processing remains unclear. We used two-photon imaging and electrophysiological recordings to clarify the in vivo properties of these cells and their functional organization in the juxtaglomerular space. Juxtaglomerular neurons coded for many perceptual characteristics of the olfactory stimulus such as (1) identity of the odorant, (2) odorant concentration, (3) odorant onset, and (4) offset. The odor-responsive neurons clustered within a narrow area surrounding the glomerulus with the same odorant specificity, with ~80% of responding cells located ≤20 µm from the glomerular border. This stereotypic spatial pattern of activated cells persisted at different odorant concentrations and was found for neurons both activated and inhibited by the odorant. Our data identify a principal glomerulus with a narrow shell of juxtaglomerular neurons as a basic odor coding unit in the glomerular layer and underline the important role of intraglomerular circuitry.


Subject(s)
Nerve Net/cytology , Nerve Net/physiology , Odorants , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Olfactory Receptor Neurons/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neurons/physiology
2.
Neuroscience ; 162(3): 612-23, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19302832

ABSTRACT

Elimination of redundant synapses and strengthening of the surviving ones are crucial steps in the development of the nervous system. Both processes can be readily followed at the climbing fiber to Purkinje cell synapse in the cerebellum. Shortly after birth, around five equally strong climbing fiber synapses are established. Subsequently, one of these five synaptic connections starts to grow in size and synaptic strength, while the others degenerate and eventually disappear. Both the elimination of the redundant climbing fiber synapses and the strengthening of the surviving one depend on a combination of a genetically coded blueprint and synaptic activity. Recently, it has been shown that synaptic activity affects the synaptic strength of developing climbing fibers. Remarkably, the same pattern of paired activity of the presynaptic climbing fiber and the postsynaptic Purkinje cell resulted in strengthening of already "large" climbing fibers and weakening of already "weak" climbing fibers. In this review, we will integrate the current knowledge of synaptic plasticity of climbing fibers with that of other processes affecting climbing fiber development.


Subject(s)
Cerebellum/cytology , Cerebellum/growth & development , Nerve Fibers/physiology , Neuronal Plasticity/physiology , Purkinje Cells/physiology , Synapses/physiology , Animals
3.
J Neurosci ; 24(22): 5119-30, 2004 Jun 02.
Article in English | MEDLINE | ID: mdl-15175381

ABSTRACT

G-protein-coupled metabotropic glutamate group I receptors (mGluR1s) mediate synaptic transmission and plasticity in Purkinje cells and, therefore, critically determine cerebellar motor control and learning. Purkinje cells express two members of the G-protein G(q) family, namely G(q) and G11. Although in vitro coexpression of mGluR1 with either Galpha11 or Galpha(q) produces equally well functioning signaling cascades, Galpha(q)- and Galpha11-deficient mice exhibit distinct alterations in motor coordination. By using whole-cell recordings and Ca2+ imaging in Purkinje cells, we show that Galpha(q) is required for mGluR-dependent synaptic transmission and for long-term depression (LTD). Galpha11 has no detectable contribution for synaptic transmission but also contributes to LTD. Quantitative single-cell RT-PCR analyses in Purkinje cells demonstrate a more than 10-fold stronger expression of Galpha(q) versus Galpha11. Our findings suggest an expression level-dependent action of Galpha(q) and Galpha11 for Purkinje cell signaling and assign specific roles of these two G(q) isoforms for motor coordination.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Purkinje Cells/metabolism , Animals , Behavior, Animal/physiology , COS Cells , Calcium/metabolism , Calcium Signaling/genetics , Cerebellum/cytology , Cerebellum/metabolism , Chlorocebus aethiops , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Fluorescent Dyes , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Long-Term Synaptic Depression/genetics , Long-Term Synaptic Depression/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Subunits/physiology , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology
4.
Pflugers Arch ; 446(6): 766-73, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12883893

ABSTRACT

The zebrafish larva is a powerful model for the analysis of behaviour and the underlying neuronal network activity during early stages of development. Here we employ a new approach of "in vivo" Ca(2+) imaging in this preparation. We demonstrate that bolus injection of membrane-permeable Ca(2+) indicator dyes into the spinal cord of zebrafish larvae results in rapid staining of essentially the entire spinal cord. Using two-photon imaging, we could monitor Ca(2+) signals simultaneously from a large population of spinal neurons with single-cell resolution. To test the method, Ca(2+) transients were produced by iontophoretic application of glutamate and, as observed for the first time in a living preparation, of GABA or glycine. Glycine-evoked Ca(2+) transients were blocked by the application of strychnine. Sensory stimuli that trigger escape reflexes in mobile zebrafish evoked Ca(2+) transients in distinct neurons of the spinal network. Moreover, long-term recordings revealed spontaneous Ca(2+) transients in individual spinal neurons. Frequently, this activity occurred synchronously among many neurons in the network. In conclusion, the new approach permits a reliable analysis with single-cell resolution of the functional organisation of developing neuronal networks.


Subject(s)
Calcium/physiology , Diagnostic Imaging , Nerve Net/physiology , Zebrafish/physiology , Animals , Calcium/chemistry , Calcium Signaling/drug effects , Calcium Signaling/physiology , Coloring Agents , Excitatory Amino Acids/antagonists & inhibitors , Excitatory Amino Acids/pharmacology , Fluorescent Dyes , Glycine Agents/pharmacology , In Vitro Techniques , Larva/physiology , Nerve Net/drug effects , Nerve Net/growth & development , Neurons/physiology , Spinal Cord/cytology , Spinal Cord/growth & development , Spinal Cord/physiology , Strychnine/pharmacology
5.
J Neurosci ; 21(24): 9701-12, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739579

ABSTRACT

Climbing fiber (CF) synapse formation onto cerebellar Purkinje cells (PCs) is critically dependent on the synaptogenesis from parallel fibers (PFs), the other input to PCs. Previous studies revealed that deletion of the glutamate receptor delta2 subunit (GluRdelta2) gene results in persistent multiple CF innervation of PCs with impaired PF synaptogenesis, whereas mutation of the metabotropic glutamate receptor subtype 1 (mGluR1) gene causes multiple CF innervation with normal PF synaptogenesis. We demonstrate that atypical CF-mediated EPSCs (CF-EPSCs) with slow rise times and small amplitudes coexisted with typical CF-EPSCs with fast rise times and large amplitudes in PCs from GluRdelta2 mutant cerebellar slices. CF-EPSCs in mGluR1 mutant and wild-type PCs had fast rise times. Atypical slow CF responses of GluRdelta2 mutant PCs were associated with voltage-dependent Ca(2+) signals that were confined to PC distal dendrites. In the wild-type and mGluR1 mutant PCs, CF-induced Ca(2+) signals involved both proximal and distal dendrites. Morphologically, CFs of GluRdelta2 mutant mice extended to the superficial regions of the molecular layer, whereas those of wild-type and mGluR1 mutant mice did not innervate the superficial one-fifth of the molecular layer. It is therefore likely that surplus CFs of GluRdelta2 mutant mice form ectopic synapses onto distal dendrites, whereas those of wild-type and mGluR1 mutant mice innervate proximal dendrites. These findings suggest that GluRdelta2 is required for consolidating PF synapses and restricting CF synapses to the proximal dendrites, whereas the mGluR1-signaling pathway does not affect PF synaptogenesis but is involved in eliminating surplus CF synapses at the proximal dendrites.


Subject(s)
Cerebellum/metabolism , Nerve Fibers/physiology , Receptors, Glutamate/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synapses/metabolism , Aging/metabolism , Animals , Calcium Signaling/physiology , Cell Membrane/genetics , Cell Membrane/metabolism , Cerebellum/cytology , Cerebellum/growth & development , Crosses, Genetic , Dendrites/physiology , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Mutation , Olivary Nucleus/physiology , Patch-Clamp Techniques , Purkinje Cells/cytology , Purkinje Cells/physiology , Reaction Time/physiology , Receptors, Glutamate/genetics , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/physiology
6.
J Physiol ; 536(Pt 2): 429-37, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11600678

ABSTRACT

1. Cellular responses to GABA(A) receptor activation were studied in developing cerebellar Purkinje neurones (PNs) in brain slices obtained from 2- to 22-day-old rats. Two-photon fluorescence imaging of fura-2-loaded cells and perforated-patch recordings were used to monitor intracellular Ca2+ transients and to estimate the reversal potential of GABA-induced currents, respectively. 2. During the 1st postnatal week, focal application of GABA or the GABA(A) receptor agonist muscimol evoked transient increases in [Ca2+]i in immature PNs. These Ca2+ transients were reversibly abolished by the GABA(A) receptor antagonist bicuculline and by Ni2+, a blocker of voltage-activated Ca2+ channels. 3. Perforated-patch recordings were used to measure the reversal potential of GABA-evoked currents (E(GABA)) at different stages of development. It was found that E(GABA) was about -44 mV at postnatal day 3 (P3), it shifted to gradually more negative values during the 1st week and finally equilibrated at -87 mV at around the end of the 2nd postnatal week. This transition was well described by a sigmoidal function. The largest change in E(GABA) was -7 mV x day(-1), which occurred at around P6. 4. The transition in GABA-mediated signalling occurs during a period in which striking changes in PN morphology and synaptic connectivity are known to take place. Since such changes were shown to be Ca2+ dependent, we propose that GABA-evoked Ca2+ signalling is one of the critical determinants for the normal development of cerebellar PNs.


Subject(s)
Calcium Signaling/physiology , Purkinje Cells/metabolism , gamma-Aminobutyric Acid/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bicuculline/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Cerebellum/cytology , Cerebellum/growth & development , GABA Antagonists/pharmacology , Gramicidin/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nickel/pharmacology , Patch-Clamp Techniques , Purkinje Cells/drug effects , Rats , Rats, Wistar , Receptors, GABA-A/metabolism
7.
Neuron ; 31(4): 519-22, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11545711

ABSTRACT

Activation of most excitatory synapses of central neurons produces calcium release signals from intracellular stores. Synaptically evoked calcium release from stores is frequently triggered by the binding of glutamate to metabotropic receptors and the subsequent activation of IP(3) receptors in spines and dendrites. There is increasing evidence for the presence of local calcium signals caused by calcium-induced calcium release (CICR) through activation of ryanodine or IP(3) receptors. Recent work on mutant mice indicates that store signaling determines activity-dependent synaptic plasticity.


Subject(s)
Calcium/metabolism , Neuronal Plasticity/physiology , Purkinje Cells/physiology , Pyramidal Cells/physiology , Synapses/metabolism , Animals
9.
J Neurosci ; 21(15): 5520-7, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11466423

ABSTRACT

The pituitary adenylate cyclase activating polypeptide (PACAP) type I receptor (PAC1) is a G-protein-coupled receptor binding the strongly conserved neuropeptide PACAP with 1000-fold higher affinity than the related peptide vasoactive intestinal peptide. PAC1-mediated signaling has been implicated in neuronal differentiation and synaptic plasticity. To gain further insight into the biological significance of PAC1-mediated signaling in vivo, we generated two different mutant mouse strains, harboring either a complete or a forebrain-specific inactivation of PAC1. Mutants from both strains show a deficit in contextual fear conditioning, a hippocampus-dependent associative learning paradigm. In sharp contrast, amygdala-dependent cued fear conditioning remains intact. Interestingly, no deficits in other hippocampus-dependent tasks modeling declarative learning such as the Morris water maze or the social transmission of food preference are observed. At the cellular level, the deficit in hippocampus-dependent associative learning is accompanied by an impairment of mossy fiber long-term potentiation (LTP). Because the hippocampal expression of PAC1 is restricted to mossy fiber terminals, we conclude that presynaptic PAC1-mediated signaling at the mossy fiber synapse is involved in both LTP and hippocampus-dependent associative learning.


Subject(s)
Association Learning/physiology , Long-Term Potentiation/physiology , Mossy Fibers, Hippocampal/metabolism , Receptors, Pituitary Hormone/deficiency , Receptors, Pituitary Hormone/metabolism , Animals , Avoidance Learning/physiology , Cues , Electroshock , In Vitro Techniques , Maze Learning/physiology , Mice , Mice, Knockout , Mice, Mutant Strains , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Presynaptic Terminals/metabolism , Prosencephalon/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/genetics , Signal Transduction/physiology , Social Behavior
10.
J Neurosci ; 21(12): 4207-14, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11404406

ABSTRACT

Spines and dendrites of central neurons represent an important site of synaptic signaling and integration. Here we identify a new, synaptically mediated spine signal with unique properties. Using two-photon Na(+) imaging, we show that suprathreshold synaptic stimulation leads to transient increases in Na(+) concentration in postsynaptic spines and their adjacent dendrites. This local signal is restricted to a dendritic domain near the site of synaptic input. In presumed active spines within this domain, the Na(+) level increases by 30-40 mm even during short bursts of synaptic stimulation. During a long-term potentiation induction protocol (100 Hz, 1 sec), the Na(+) level in the active spines reaches peak amplitudes of approximately 100 mm. We find that the Na(+) transients are mainly mediated by Na(+) entry through NMDA receptor channels and are detected during the coincident occurrence of synaptic potentials and backpropagating action potentials. The large amplitudes of the Na(+) transients and their location on dendritic spines suggest that this signal is an important determinant of electrical and biochemical spine characteristics.


Subject(s)
Cell Surface Extensions/metabolism , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology , Sodium/metabolism , Action Potentials/physiology , Animals , Dendrites/metabolism , Electric Stimulation/methods , Hippocampus/cytology , Hippocampus/metabolism , In Vitro Techniques , Long-Term Potentiation/physiology , Membrane Potentials/physiology , Mice , Microscopy, Confocal , Patch-Clamp Techniques , Sensory Thresholds/physiology , Synaptic Transmission/physiology
13.
Nat Neurosci ; 3(5): 452-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10769384

ABSTRACT

Two-photon imaging of large neuronal networks in cortical slices of newborn rats revealed synchronized oscillations in intracellular Ca2+ concentration. These spontaneous Ca2+ waves usually started in the posterior cortex and propagated slowly (2.1 mm per second) toward its anterior end. Ca2+ waves were associated with field-potential changes and required activation of AMPA and NMDA receptors. Although GABAA receptors were not involved in wave initiation, the developmental transition of GABAergic transmission from depolarizing to hyperpolarizing (around postnatal day 7) stopped the oscillatory activity. Thus we identified a type of large-scale Ca2+ wave that may regulate long-distance wiring in the immature cortex.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Bicuculline/pharmacology , Calcium Signaling/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Electric Conductivity , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , In Vitro Techniques , Membrane Potentials/drug effects , Nerve Net/drug effects , Nerve Net/metabolism , Neurons/drug effects , Neurons/metabolism , Photons , Rats , Rats, Wistar , Receptors, AMPA/physiology , Receptors, GABA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Tetrodotoxin/pharmacology , gamma-Aminobutyric Acid/pharmacology
14.
J Neurosci ; 20(5): 1791-9, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10684880

ABSTRACT

We have used rapid confocal microscopy to investigate the mechanism of Ca(2+) signals in individual dendritic spines of hippocampal CA1 pyramidal cells. The experiments focused on the signals that occur during single weak synaptic responses that were subthreshold for triggering postsynaptic action potentials. These Ca(2+) signals were not strongly affected by blocking the EPSPs with the AMPA receptor antagonist CNQX. The signals were also not strongly reduced by blocking T-type voltage-gated Ca(2+) channels (VGCCs) with Ni(2+) or by blocking a broad range of VGCCs with intracellular D890. The spine Ca(2+) signals were blocked by NMDA receptor channel (NMDAR) antagonist and had the voltage dependence characteristic of these channels. Neither ryanodine nor cyclopiazonic acid (CPA), substances known to deplete intracellular Ca(2+) stores, substantially reduced the amplitude of synaptically evoked Ca(2+) signals. CPA slowed the recovery phase of Ca(2+) signals in spines produced by synaptic stimulation or by backpropagating action potentials, suggesting a role of intracellular stores in Ca(2+) reuptake. Thus, we find that Ca(2+) release from intracellular stores is not required to produce spine Ca(2+) signals. We conclude that synaptic Ca(2+) signals in spines are primarily caused by Ca(2+) entry through NMDARs. Although these channels are largely blocked by Mg(2+) at voltages near the resting potential, they can nevertheless produce significant Ca(2+) elevation. The resulting Ca(2+) signals are an integral component of individual evoked or spontaneous synaptic events and may be important in the maintenance of synaptic function.


Subject(s)
Calcium Signaling/physiology , Dendrites/physiology , Hippocampus/cytology , Receptors, N-Methyl-D-Aspartate/physiology , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Calcium/metabolism , Calcium Channels, T-Type/physiology , Calcium Signaling/drug effects , Cells, Cultured , Dendrites/chemistry , Dinucleoside Phosphates/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microscopy, Confocal , Pyramidal Cells/physiology , Pyramidal Cells/ultrastructure , Rats , Rats, Wistar , Receptors, AMPA/physiology , Ryanodine/pharmacology
15.
J Physiol ; 521 Pt 1: 99-111, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10562337

ABSTRACT

1. Whole-cell patch-clamp recordings of iontophoretically induced N-methyl-D-aspartate (NMDA) receptor-mediated currents (INMDA) in CA1 pyramidal cells in hippocampal slices from 1- to 40-day-old rats were used to characterize developmental changes in the Mg2+ sensitivity of NMDA receptors. 2. The dose-response relations for extracellular Mg2+ blockade of INMDA indicated a high affinity binding of Mg2+ to NMDA receptors at membrane potentials more negative than -60 mV, independent of postnatal age. 3. Depolarizing the cells unblocked NMDA receptors by decreasing their affinity for Mg2+. The efficacy of depolarization in unblocking NMDA receptors markedly increased after postnatal day 4 (P4), endowing the receptors with a greater voltage dependence. 4. The NR2B subunit-specific NMDA antagonist ifenprodil reduced INMDA in pyramidal cells of all ages. The sensitivity of INMDA to ifenprodil was greatest during the first postnatal week and decreased thereafter, indicating an enhanced contribution of NR2B subunit-containing NMDA receptors to INMDA in the first week after birth. 5. In the first postnatal week, the ifenprodil-insensitive INMDA component had a lower voltage dependence than the total INMDA. In older pyramidal cells, the voltage dependence of the ifenprodil-insensitive component and the total INMDA were similar. 6. In another set of CA1 pyramidal cells, single-cell reverse transcription and polymerase chain reaction (RT-PCR) were used to characterize concomitant developmental changes in NMDA subunit mRNA expression. The mRNA for the NR2D subunit was detected during the first postnatal week in 50 % of the cells and disappeared thereafter. The proportion of cells expressing the NR2A and NR2B subunits remained relatively constant throughout the first five postnatal weeks. 7. We conclude that NMDA receptors in hippocampal CA1 pyramidal cells are effectively blocked by Mg2+ at all ages. After 4 days they become much less sensitive to Mg2+ at depolarized membrane potentials. This postnatal switch in voltage control of Mg2+ binding to NMDA receptors may be due to the downregulation of NR2D subunit expression in developing CA1 pyramidal cells.


Subject(s)
Magnesium/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Animals, Newborn , Gene Expression Regulation, Developmental , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , In Vitro Techniques , Membrane Potentials , Patch-Clamp Techniques , Piperidines/pharmacology , Protein Structure, Quaternary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Nature ; 401(6756): 918-21, 1999 Oct 28.
Article in English | MEDLINE | ID: mdl-10553907

ABSTRACT

Neurotrophins are a family of structurally related proteins that regulate the survival, differentiation and maintenance of function of different populations of peripheral and central neurons. They are also essential for modulating activity-dependent neuronal plasticity. Here we show that neurotrophins elicit action potentials in central neurons. Even at low concentrations, brain-derived neurotrophic factor (BDNF) excited neurons in the hippocampus, cortex and cerebellum. We found that BDNF and neurotrophin-4/5 depolarized neurons just as rapidly as the neurotransmitter glutamate, even at a more than thousand-fold lower concentration. Neurotrophin-3 produced much smaller responses, and nerve growth factor was ineffective. The neurotrophin-induced depolarization resulted from the activation of a sodium ion conductance which was reversibly blocked by K-252a, a protein kinase blocker which prefers tyrosine kinase Trk receptors. Our results demonstrate a very rapid excitatory action of neurotrophins, placing them among the most potent endogenous neuro-excitants in the mammalian central nervous system described so far.


Subject(s)
Nerve Growth Factors/physiology , Receptor, trkB/physiology , Synaptic Transmission/physiology , Action Potentials , Animals , Brain-Derived Neurotrophic Factor/physiology , Calcium/metabolism , Carbazoles/pharmacology , Glutamic Acid/physiology , In Vitro Techniques , Indole Alkaloids , Membrane Potentials , Neurotransmitter Agents/physiology , Patch-Clamp Techniques , Pyramidal Cells/physiology , Rats , Rats, Wistar , Sodium Channels/physiology
17.
J Physiol ; 514 ( Pt 2): 327-41, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9852317

ABSTRACT

1. The distribution of ATP-sensitive K+ channels (KATP channels) was investigated in four cell types in hippocampal slices prepared from 10- to 13-day-old rats: CA1 pyramidal cells, interneurones of stratum radiatum in CA1, complex glial cells of the same area and granule cells of the dentate gyrus. The neuronal cell types were identified visually and characterized by the shapes and patterns of their action potentials and by neurobiotin labelling. 2. The patch-clamp technique was used to study the sensitivity of whole-cell currents to diazoxide (0.3 mM), a KATP channel opener, and to tolbutamide (0.5 mM) or glibenclamide (20 microM), two KATP channel inhibitors. The fraction of cells in which whole-cell currents were activated by diazoxide and inhibited by tolbutamide was 26% of pyramidal cells, 89 % of interneurones, 100% of glial cells and 89% of granule cells. The reversal potential of the diazoxide-induced current was at the K+ equilibrium potential and a similar current activated spontaneously when cells were dialysed with an ATP-free pipette solution. 3. Using the single-cell RT-PCR method, the presence of mRNA encoding KATP channel subunits (Kir6.1, Kir6.2, SUR1 and SUR2) was examined in CA1 pyramidal cells and interneurones. Subunit mRNA combinations that can result in functional KATP channels (Kir6.1 together with SUR1, Kir6.2 together with SUR1 or SUR2) were detected in only 17% of the pyramidal cells. On the other hand, KATP channels may be formed in 75% of the interneurones, mainly by the combination of Kir6.2 with SUR1 (58% of all interneurones). 4. The results of these combined analyses indicate that functional KATP channels are present in principal neurones, interneurones and glial cells of the rat hippocampus, but at highly different densities in the four cell types studied.


Subject(s)
ATP-Binding Cassette Transporters , Hippocampus/physiology , Neurons/physiology , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Transcription, Genetic , Adenosine Triphosphate/physiology , Animals , Diazoxide/pharmacology , In Vitro Techniques , Interneurons/drug effects , Interneurons/physiology , Neuroglia/drug effects , Neuroglia/physiology , Neurons/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Drug/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sulfonylurea Compounds/pharmacology , Sulfonylurea Receptors , Tolbutamide/pharmacology
18.
Pflugers Arch ; 439(1-2): 201-7, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10651018

ABSTRACT

Dendritic spines are assumed to be the smallest units of neuronal integration. Because of their miniature size, however, many of their functional properties are still unclear. New insights in spine physiology have been provided by two-photon laser-scanning microscopy which allows fluorescence imaging with high spatial resolution and minimal photodamage. For example, two-photon imaging has been employed successfully for the measurement of activity-induced calcium transients in individual spines. Here, we describe the first application of two-photon imaging to measure Na+ transients in spines and dendrites of CA1 pyramidal neurons in hippocampal slices. Whole-cell patch-clamped neurons were loaded with the Na(+)-indicator dye SBFI (sodium-binding benzofuran-isophthalate). In situ calibration of SBFI fluorescence with ionophores enabled the determination of the actual magnitude of the [Na+]i changes. We found that back-propagating action potentials (APs) evoked Na+ transients throughout the proximal part of the dendritic tree and adjacent spines. The action-potential-induced [Na+]i transients reached values of 4 mM for a train of 20 APs and monotonically decayed with a time constant of several seconds. These results represent the first demonstration of activity-induced Na+ accumulation in spines. Our results demonstrate that two-photon Na+ imaging represents a powerful tool for extending our knowledge on Na+ signaling in fine cellular subcompartments.


Subject(s)
Dendrites/ultrastructure , Neurons/ultrastructure , Sodium/metabolism , Action Potentials/physiology , Animals , Benzofurans , Calibration , Dendrites/metabolism , Dendrites/physiology , Ethers, Cyclic , Fluorescent Dyes , Kinetics , Mice , Microscopy, Confocal , Neurons/metabolism , Neurons/physiology , Photons , Sodium Channels/metabolism
19.
J Neurosci ; 18(23): 9573-84, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9822718

ABSTRACT

Voltage-dependent Ca2+ channels are a major pathway for Ca2+ entry in neurons. We have studied the electrophysiological, pharmacological, and molecular properties of voltage-gated Ca2+ channels in motoneurons of the rat facial nucleus in slices of the brainstem. Most facial motoneurons express both low voltage-activated (LVA) and high voltage-activated (HVA) Ca2+ channel currents. The HVA current is composed of a number of pharmacologically separable components, including 30% of N-type and approximately 5% of L-type. Despite the dominating role of P-type Ca2+ channels in transmitter release at facial motoneuron terminals described in previous studies, these channels were not present in the cell body. Remarkably, most of the HVA current was carried through a new type of Ca2+ channel that is resistant to toxin and dihydropyridine block but distinct from the R-type currents described in other neurons. Using reverse transcription followed by PCR amplification (RT-PCR) with a powerful set of primers designed to amplify all HVA subtypes of the alpha1-subunit, we identified a highly heterogeneous expression pattern of Ca2+ channel alpha1-subunit mRNA in individual neurons consistent with the Ca2+ current components found in the cell bodies and axon terminals. We detected mRNA for alpha1A in 86% of neurons, alpha1B in 59%, alpha1C in 18%, alpha1D in 18%, and alpha1E in 59%. Either alpha1A or alpha1B mRNAs (or both) were present in all neurons, together with various other alpha1-subunit mRNAs. The most frequently occurring combination was alpha1A with alpha1B and alpha1E. Taken together, these results demonstrate that the Ca2+ channel pattern found in facial motoneurons is highly distinct from that found in other brainstem motoneurons.


Subject(s)
Calcium Channels/genetics , Facial Nerve/cytology , Motor Neurons/chemistry , Receptors, Cholinergic/genetics , omega-Conotoxins , Animals , Animals, Newborn , Brain Stem/cytology , Cadmium/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type , DNA, Complementary , Dihydropyridines/pharmacology , Facial Nerve/chemistry , Ion Channel Gating/drug effects , Nickel/pharmacology , Nitrendipine/pharmacology , Patch-Clamp Techniques , Peptides/pharmacology , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Spider Venoms/pharmacology , omega-Agatoxin IVA , omega-Conotoxin GVIA
20.
Prog Neurobiol ; 55(6): 563-75, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9670218

ABSTRACT

Neurons are extraordinarily complicated devices, in which physical and chemical processes are intercoupled, in spatially non-uniform manner, over distances of millimeters or more, and over time scales of < 1 msec up to the lifetime of the animal. The fact that neuronal populations generating most brain activities of interest are very large-perhaps many millions of cells-makes the task of analysis seem hopeless. Yet, during at least some population activities, neuronal networks oscillate synchronously. The emergence of such oscillations generates precise temporal relationship between neuronal inputs and outputs, thus rendering tractable the analysis of network function at a cellular level. We illustrate this idea with a review of recent data and a network model of synchronized gamma frequency (> 20 Hz) oscillations in vitro, and discuss how these and other oscillations may relate to recent data on back-propagating, action potentials, dendritic Ca2+ transients, long-term potentiation and GABAA receptor-mediated synaptic potentials.


Subject(s)
Electroencephalography , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/physiology , Animals , Hippocampus/physiology , Nerve Net/physiology , Oscillometry , Receptors, AMPA/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...