Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Brain Struct Funct ; 222(8): 3677-3690, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28466358

ABSTRACT

The perisomatic domain of cortical neurons is under the control of two major GABAergic inhibitory interneuron types: regular-spiking cholecystokinin (CCK) basket cells (BCs) and fast-spiking parvalbumin (PV) BCs. CCK and PV BCs are different not only in their intrinsic physiological, anatomical and molecular characteristics, but also in their presynaptic modulation of their synaptic output. Most GABAergic terminals are known to contain GABAB receptors (GABABR), but their role in presynaptic inhibition and surface expression have not been comparatively characterized in the two BC types. To address this, we performed whole-cell recordings from CCK and PV BCs and postsynaptic pyramidal cells (PCs), as well as freeze-fracture replica-based quantitative immunogold electron microscopy of their synapses in the rat hippocampal CA1 area. Our results demonstrate that while both CCK and PV BCs contain functional presynaptic GABABRs, their modulatory effects and relative abundance are markedly different at these two synapses: GABA release is dramatically inhibited by the agonist baclofen at CCK BC synapses, whereas a moderate reduction in inhibitory transmission is observed at PV BC synapses. Furthermore, GABABR activation has divergent effects on synaptic dynamics: paired-pulse depression (PPD) is enhanced at CCK BC synapses, but abolished at PV BC synapses. Consistent with the quantitative differences in presynaptic inhibition, virtually all CCK BC terminals were found to contain GABABRs at high densities, but only 40% of PV BC axon terminals contain GABABRs at detectable levels. These findings add to an increasing list of differences between these two interneuron types, with implications for their network functions.


Subject(s)
Cholecystokinin/physiology , Hippocampus/physiology , Parvalbumins/physiology , Pyramidal Cells/physiology , Receptors, GABA-B/physiology , Animals , Cholecystokinin/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Inhibitory Postsynaptic Potentials , Neural Inhibition , Parvalbumins/metabolism , Presynaptic Terminals/physiology , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Rats, Transgenic , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Receptor, Muscarinic M2/metabolism , Receptors, GABA-B/metabolism , Synapses/physiology
2.
Cereb Cortex ; 27(3): 2318-2334, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27073217

ABSTRACT

Cholecystokinin-expressing interneurons (CCK-INs) mediate behavior state-dependent inhibition in cortical circuits and themselves receive strong GABAergic input. However, it remains unclear to what extent GABAB receptors (GABABRs) contribute to their inhibitory control. Using immunoelectron microscopy, we found that CCK-INs in the rat hippocampus possessed high levels of dendritic GABABRs and KCTD12 auxiliary proteins, whereas postsynaptic effector Kir3 channels were present at lower levels. Consistently, whole-cell recordings revealed slow GABABR-mediated inhibitory postsynaptic currents (IPSCs) in most CCK-INs. In spite of the higher surface density of GABABRs in CCK-INs than in CA1 principal cells, the amplitudes of IPSCs were comparable, suggesting that the expression of Kir3 channels is the limiting factor for the GABABR currents in these INs. Morphological analysis showed that CCK-INs were diverse, comprising perisomatic-targeting basket cells (BCs), as well as dendrite-targeting (DT) interneurons, including a previously undescribed DT type. GABABR-mediated IPSCs in CCK-INs were large in BCs, but small in DT subtypes. In response to prolonged activation, GABABR-mediated currents displayed strong desensitization, which was absent in KCTD12-deficient mice. This study highlights that GABABRs differentially control CCK-IN subtypes, and the kinetics and desensitization of GABABR-mediated currents are modulated by KCTD12 proteins.


Subject(s)
Cholecystokinin/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Inhibitory Postsynaptic Potentials/physiology , Interneurons/metabolism , Potassium Channels/metabolism , Receptors, GABA-A/metabolism , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Immunohistochemistry , Interneurons/ultrastructure , Male , Microscopy, Immunoelectron , Patch-Clamp Techniques , Rats, Wistar , Tissue Culture Techniques
3.
Front Cell Neurosci ; 9: 315, 2015.
Article in English | MEDLINE | ID: mdl-26321916

ABSTRACT

Tuning of the time course and strength of inhibitory and excitatory neurotransmitter release is fundamental for the precise operation of cortical network activity and is controlled by Ca(2+) influx into presynaptic terminals through the high voltage-activated P/Q-type Ca(2+) (Cav2.1) channels. Proper channel-mediated Ca(2+)-signaling critically depends on the topographical arrangement of the channels in the presynaptic membrane. Here, we used high-resolution SDS-digested freeze-fracture replica immunoelectron microscopy together with automatized computational analysis of Cav2.1 immunogold labeling to determine the precise subcellular organization of Cav2.1 channels in both inhibitory and excitatory terminals. Immunoparticles labeling the pore-forming α1 subunit of Cav2.1 channels were enriched over the active zone of the boutons with the number of channels (3-62) correlated with the area of the synaptic membrane. Detailed analysis showed that Cav2.1 channels are non-uniformly distributed over the presynaptic membrane specialization where they are arranged in clusters of an average five channels per cluster covering a mean area with a diameter of about 70 nm. Importantly, clustered arrangement and cluster properties did not show any significant difference between GABAergic and glutamatergic terminals. Our data demonstrate a common nano-architecture of Cav2.1 channels in inhibitory and excitatory boutons in stratum radiatum of the hippocampal CA1 area suggesting that the cluster arrangement is crucial for the precise release of transmitters from the axonal boutons.

4.
PLoS One ; 10(3): e0119643, 2015.
Article in English | MEDLINE | ID: mdl-25760459

ABSTRACT

Adult hippocampal neurogenesis has been implicated in hippocampus-dependent learning and memory. Furthermore, the decline of neurogenesis accompanying aging could be involved in age-related cognitive deficits. It is believed that the neural stem cell niche comprises a specialized microenvironment regulating stem cell activation and maintenance. However, little is known about the significance of the extracellular matrix in controlling adult stem cells. Reelin is a large glycoprotein of the extracelluar matrix known to be of crucial importance for neuronal migration. Here, we examined the local interrelation between Reelin expressing interneurons and putative hippocampal stem cells and investigated the effects of Reelin deficiency on stem cell and progenitor cell proliferation. Reelin-positive cells are found in close vicinity to putative stem cell processes, which would allow for stem cell regulation by Reelin. We investigated the proliferation of stem cells in the Reelin-deficient reeler hippocampus by Ki67 labeling and found a strong reduction of mitotic cells. A detailed analysis of dividing Type 1, type 2 and type 3 cells indicated that once a stem cell is recruited for proliferation, the progression to the next progenitor stage as well as the number of mitotic cycles is not altered in reeler. Our data point to a role for Reelin in either regulating stem cell quiescence or maintenance.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Dentate Gyrus/cytology , Extracellular Matrix Proteins/metabolism , Interneurons/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Stem Cells/physiology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Differentiation , Cell Proliferation , Dentate Gyrus/physiology , Extracellular Matrix Proteins/genetics , Male , Mice , Mitosis , Nerve Tissue Proteins/genetics , Reelin Protein , Serine Endopeptidases/genetics
5.
J Neurosci ; 35(5): 1858-71, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25653347

ABSTRACT

Tight coupling between Ca(2+) channels and the sensor for vesicular transmitter release at the presynaptic active zone (AZ) is crucial for high-fidelity synaptic transmission. It has been hypothesized that a switch from a loosely coupled to a tightly coupled transmission mode is a common step in the maturation of CNS synapses. However, this hypothesis has never been tested at cortical synapses. We addressed this hypothesis at a representative small cortical synapse: the synapse connecting mouse cerebellar cortical parallel fibers to Purkinje neurons. We found that the slow Ca(2+) chelator EGTA affected release significantly stronger at immature than at mature synapses, while the fast chelator BAPTA was similarly effective in both groups. Analysis of paired-pulse ratios and quantification of release probability (pr) with multiple-probability fluctuation analysis revealed increased facilitation at immature synapses accompanied by reduced pr. Cav2.1 Ca(2+) channel immunoreactivity, assessed by quantitative high-resolution immuno-electron microscopy, was scattered over immature boutons but confined to putative AZs at mature boutons. Presynaptic Ca(2+) signals were quantified with two-photon microscopy and found to be similar between maturation stages. Models adjusted to fit EGTA dose-response curves as well as differential effects of the Ca(2+) channel blocker Cd(2+) indicate looser and less homogenous coupling at immature terminals compared with mature ones. These results demonstrate functionally relevant developmental tightening of influx-release coupling at a single AZ cortical synapse and corroborate developmental tightening of coupling as a prevalent phenomenon in the mammalian brain.


Subject(s)
Calcium Signaling , Neurogenesis , Purkinje Cells/metabolism , Synapses/metabolism , Animals , Calcium/metabolism , Calcium Channels, N-Type/metabolism , Calcium Chelating Agents/pharmacology , Excitatory Postsynaptic Potentials , Female , Male , Mice , Mice, Inbred C57BL , Purkinje Cells/drug effects , Purkinje Cells/physiology , Synapses/physiology
6.
J Neurosci ; 34(41): 13586-99, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25297088

ABSTRACT

Parkinson disease (PD) is an α-synucleinopathy resulting in the preferential loss of highly vulnerable dopamine (DA) substantia nigra (SN) neurons. Mutations (e.g., A53T) in the α-synuclein gene (SNCA) are sufficient to cause PD, but the mechanism of their selective action on vulnerable DA SN neurons is unknown. In a mouse model overexpressing mutant α-synuclein (A53T-SNCA), we identified a SN-selective increase of in vivo firing frequencies in DA midbrain neurons, which was not observed in DA neurons in the ventral tegmental area. The selective and age-dependent gain-of-function phenotype of A53T-SCNA overexpressing DA SN neurons was in part mediated by an increase of their intrinsic pacemaker frequency caused by a redox-dependent impairment of A-type Kv4.3 potassium channels. This selective enhancement of "stressful pacemaking" of DA SN neurons in vivo defines a functional response to mutant α-synuclein that might be useful as a novel biomarker for the "DA system at risk" before the onset of neurodegeneration in PD.


Subject(s)
Dopaminergic Neurons/physiology , Mutation/physiology , Oxidative Stress/physiology , Shal Potassium Channels/physiology , Substantia Nigra/physiology , alpha-Synuclein/genetics , Aging/physiology , Animals , Electrophysiological Phenomena , Glutathione/metabolism , Glutathione/physiology , Ion Channel Gating/physiology , Male , Mice , Mutation/genetics , Substantia Nigra/cytology , Substantia Nigra/growth & development , Ventral Tegmental Area/growth & development , Ventral Tegmental Area/physiology
7.
J Neurosci ; 33(18): 7961-74, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23637187

ABSTRACT

Inhibitory parvalbumin-containing interneurons (PVIs) control neuronal discharge and support the generation of theta- and gamma-frequency oscillations in cortical networks. Fast GABAergic input onto PVIs is crucial for their synchronization and oscillatory entrainment, but the role of metabotropic GABA(B) receptors (GABA(B)Rs) in mediating slow presynaptic and postsynaptic inhibition remains unknown. In this study, we have combined high-resolution immunoelectron microscopy, whole-cell patch-clamp recording, and computational modeling to investigate the subcellular distribution and effects of GABA(B)Rs and their postsynaptic effector Kir3 channels in rat hippocampal PVIs. Pre-embedding immunogold labeling revealed that the receptors and channels localize at high levels to the extrasynaptic membrane of parvalbumin-immunoreactive dendrites. Immunoreactivity for GABA(B)Rs was also present at lower levels on PVI axon terminals. Whole-cell recordings further showed that synaptically released GABA in response to extracellular stimulation evokes large GABA(B)R-mediated slow IPSCs in perisomatic-targeting (PT) PVIs, but only small or no currents in dendrite-targeting (DT) PVIs. In contrast, paired recordings demonstrated that GABA(B)R activation results in presynaptic inhibition at the output synapses of both PT and DT PVIs, but more strongly in the latter. Finally, computational analysis indicated that GABA(B) IPSCs can phasically modulate the discharge of PT interneurons at theta frequencies. In summary, our results show that GABA(B)Rs differentially mediate slow presynaptic and postsynaptic inhibition in PVIs and can contribute to the dynamic modulation of their activity during oscillations. Furthermore, these data provide evidence for a compartment-specific molecular divergence of hippocampal PVI subtypes, suggesting that activation of GABA(B)Rs may shift the balance between perisomatic and dendritic inhibition.


Subject(s)
Dendrites/metabolism , Hippocampus/cytology , Interneurons/metabolism , Interneurons/ultrastructure , Parvalbumins/metabolism , Receptors, GABA-B/metabolism , Animals , Animals, Newborn , Axons/metabolism , Axons/ultrastructure , Cholecystokinin/metabolism , Computer Simulation , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GABA Agents/pharmacology , Green Fluorescent Proteins/genetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Male , Models, Neurological , Neural Inhibition , Neuropeptide Y/metabolism , Nipecotic Acids/pharmacology , Rats , Rats, Transgenic , Rats, Wistar , Tiagabine , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
8.
Eur J Neurosci ; 37(2): 190-202, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23134516

ABSTRACT

Intracortical axons originating from pyramidal cells in layer 3 of the rat somatosensory cortex are shared between adjacent columns, and receive the presynaptic inhibition that is mediated by the GABA(B) receptor. Synaptic actions by intracortical axons of single layer 3 pyramidal cells covary between the two adjacent columns in response to stimulation of layer 3 of either column. We examined whether GABA(B) receptor-mediated presynaptic inhibition affects the covariability of synaptic actions by intracortical axons between adjacent columns in slice preparations of the rat barrel cortex. Paired stimulations of superficial layer 3 evoked first and second excitatory postsynaptic currents (EPSCs) of varying amplitudes, yielding varying paired-pulse depression of EPSCs in layer 3 pyramidal cells that were located in the stimulated column, but not in its adjacent column. The amplitude of the second EPSC was inversely proportional to that of the first EPSC in layer 3 pyramidal cells in the stimulated column, yielding a negative correlation coefficient between the first and second EPSCs. Baclofen and CGP55845 attenuated paired-pulse depression and abolished the inverse relationship. Simultaneous recordings from two layer 3 pyramidal cells in the stimulated and adjacent columns revealed a positive correlation between the paired first EPSC amplitudes and a negative correlation between the paired second EPSC amplitudes, which, respectively, indicate the positive and negative covariability of synaptic actions by intracortical axons between the two adjacent columns. These results suggest that GABA(B) receptor-mediated presynaptic inhibition can reverse the positive covariability of inter-columnar synaptic actions, which may serve as a basis for inter-columnar desynchronisation.


Subject(s)
Neural Inhibition , Pyramidal Cells/metabolism , Receptors, GABA-B/metabolism , Somatosensory Cortex/metabolism , Synapses/metabolism , Animals , Axons/metabolism , Axons/physiology , Baclofen/pharmacology , Excitatory Postsynaptic Potentials , GABA-B Receptor Agonists/pharmacology , GABA-B Receptor Antagonists/pharmacology , Pyramidal Cells/physiology , Rats , Rats, Wistar , Somatosensory Cortex/cytology , Somatosensory Cortex/physiology , Synapses/physiology
9.
Eur J Neurosci ; 36(12): 3643-52, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22978624

ABSTRACT

Temporal lobe epilepsy (TLE) is the most frequent form of epilepsy in adults. In addition to recurrent focal seizures, patients suffer from memory loss and depression. The factors contributing to these symptoms are unknown. In recent years, adult hippocampal neurogenesis has been implicated in certain aspects of learning and memory, as well as in depression and anhedonia. Here we investigated whether the adult hippocampal stem cell niche is affected by status epilepticus in a mouse model of TLE using unilateral intrahippocampal kainic acid injection. Eight days after status epilepticus, we found a strong diminution in Notch signalling, a key pathway involved in stem cell maintenance, as assayed by hes5 reporter gene activity. In particular, hes5-GFP expression in the subgranular zone of the dentate gyrus was diminished. Furthermore, Sox2-positive cells as well as stem cell proliferation were reduced, thus pointing to a disruption of the stem cell niche in epilepsy under the present experimental conditions.


Subject(s)
Adult Stem Cells/metabolism , Dentate Gyrus/pathology , Epilepsy, Temporal Lobe/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Gene Expression , Genes, Reporter , Kainic Acid , Male , Mice , Mice, Inbred C57BL , Repressor Proteins/genetics , Repressor Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Status Epilepticus/pathology , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL
...