Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cereb Cortex ; 29(12): 5166-5179, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31050701

ABSTRACT

GABA signaling sustains fundamental brain functions, from nervous system development to the synchronization of population activity and synaptic plasticity. Despite these pivotal features, molecular determinants underscoring the rapid and cell-autonomous replenishment of the vesicular neurotransmitter GABA and its impact on synaptic plasticity remain elusive. Here, we show that genetic disruption of the glutamine transporter Slc38a1 in mice hampers GABA synthesis, modifies synaptic vesicle morphology in GABAergic presynapses and impairs critical period plasticity. We demonstrate that Slc38a1-mediated glutamine transport regulates vesicular GABA content, induces high-frequency membrane oscillations and shapes cortical processing and plasticity. Taken together, this work shows that Slc38a1 is not merely a transporter accumulating glutamine for metabolic purposes, but a key component regulating several neuronal functions.


Subject(s)
Amino Acid Transport System A/metabolism , Brain/physiology , GABAergic Neurons/physiology , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Animals , Mice
2.
Cereb Cortex ; 24(12): 3277-88, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23897649

ABSTRACT

The distribution and (patho-)physiological role of neuropeptides in the adult and aging brain have been extensively studied. Galanin is an inhibitory neuropeptide that can coexist with γ-aminobutyric acid (GABA) in the adult forebrain. However, galanin's expression sites, mode of signaling, impact on neuronal morphology, and colocalization with amino acid neurotransmitters during brain development are less well understood. Here, we show that galaninergic innervation of cholinergic projection neurons, which preferentially express galanin receptor 2 (GalR2) in the neonatal mouse basal forebrain, develops by birth. Nerve growth factor (NGF), known to modulate cholinergic morphogenesis, increases GalR2 expression. GalR2 antagonism (M871) in neonates reduces the in vivo expression and axonal targeting of the vesicular acetylcholine transporter (VAChT), indispensable for cholinergic neurotransmission. During cholinergic neuritogenesis in vitro, GalR2 can recruit Rho-family GTPases to induce the extension of a VAChT-containing primary neurite, the prospective axon. In doing so, GalR2 signaling dose-dependently modulates directional filopodial growth and antagonizes NGF-induced growth cone differentiation. Galanin accumulates in GABA-containing nerve terminals in the neonatal basal forebrain, suggesting its contribution to activity-driven cholinergic development during the perinatal period. Overall, our data define the cellular specificity and molecular complexity of galanin action in the developing basal forebrain.


Subject(s)
Cholinergic Neurons/cytology , Galanin/metabolism , Gene Expression Regulation, Developmental/physiology , Presynaptic Terminals/metabolism , Prosencephalon , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Cholinergic Neurons/drug effects , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Galanin/pharmacology , Glutamate Decarboxylase/genetics , Mice , Mice, Transgenic , Nerve Growth Factor/pharmacology , Presynaptic Terminals/drug effects , Prosencephalon/cytology , Prosencephalon/embryology , Prosencephalon/growth & development , Rats , Rats, Wistar , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism
3.
J Neurosci ; 29(11): 3453-62, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19295151

ABSTRACT

Alzheimer's disease is associated with an increased risk of unprovoked seizures. However, the underlying mechanisms of seizure induction remain elusive. Here, we performed video-EEG recordings in mice carrying mutant human APPswe and PS1dE9 genes (APdE9 mice) and their wild-type littermates to determine the prevalence of unprovoked seizures. In two recording episodes at the onset of amyloid beta (Abeta) pathogenesis (3 and 4.5 months of age), at least one unprovoked seizure was detected in 65% of APdE9 mice, of which 46% had multiple seizures and 38% had a generalized seizure. None of the wild-type mice had seizures. In a subset of APdE9 mice, seizure phenotype was associated with a loss of calbindin-D28k immunoreactivity in dentate granular cells and ectopic expression of neuropeptide Y in mossy fibers. In APdE9 mice, persistently decreased resting membrane potential in neocortical layer 2/3 pyramidal cells and dentate granule cells underpinned increased network excitability as identified by patch-clamp electrophysiology. At stimulus strengths evoking single-component EPSPs in wild-type littermates, APdE9 mice exhibited decreased action potential threshold and burst firing of pyramidal cells. Bath application (1 h) of Abeta1-42 or Abeta25-35 (proto-)fibrils but not oligomers induced significant membrane depolarization of pyramidal cells and increased the activity of excitatory cell populations as measured by extracellular field recordings in the juvenile rodent brain, confirming the pathogenic significance of bath-applied Abeta (proto-)fibrils. Overall, these data identify fibrillar Abeta as a pathogenic entity powerfully altering neuronal membrane properties such that hyperexcitability of pyramidal cells culminates in epileptiform activity.


Subject(s)
Action Potentials/physiology , Amyloid beta-Peptides/toxicity , Epilepsy/etiology , Epilepsy/metabolism , Neurons/physiology , Peptide Fragments/toxicity , Animals , Epilepsy/genetics , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Rats, Sprague-Dawley
4.
Endocrinology ; 147(2): 683-6, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16293666

ABSTRACT

Dehydroepiandrosterone and its sulfate (DHEAS) are sex hormone precursors that exert marked neurotrophic and/or neuroprotective activity in the central nervous system. The present study evaluated the effects of DHEAS and 17beta-estradiol (E2) in a focal cortical cold lesion model, in which DHEAS (50 mg/kg, sc) and E2 (35 mg/kg, sc) were administered either as pretreatment (two subsequent injections 1 d and 1 h before lesion induction) or posttreatment (immediately after lesion induction). The focal cortical cold lesion was induced in the primary motor cortex by means of a cooled copper cylinder placed directly onto the cortical surface. One hour later, the animals were killed, the brains cut into 0.4-mm-thick slices, and the sections stained with 1% triphenyltetrazolium chloride. The volume of the hemispheric lesion was calculated for each animal. The results demonstrated that the lesion area was significantly attenuated in both the DHEAS- and E2- pre- and posttreated groups and that in the presence of letrozole, a nonsteroidal aromatase inhibitor, no neuroprotection was observed, suggesting that the beneficial effect of DHEAS on the cold injury might depend on the conversion of DHEAS to E2 within the brain. It is concluded that even a single posttraumatic administration of DHEAS may be of substantial therapeutic benefit in the treatment of focal brain injury with vasogenic edema.


Subject(s)
Brain Injuries/prevention & control , Cold Temperature , Dehydroepiandrosterone/administration & dosage , Estradiol/physiology , Motor Cortex/injuries , Neuroprotective Agents/administration & dosage , Animals , Brain Injuries/etiology , Dehydroepiandrosterone/metabolism , Drug Administration Schedule , Estradiol/administration & dosage , Male , Models, Animal , Motor Cortex/drug effects , Motor Cortex/metabolism , Neuroprotective Agents/metabolism , Rats , Rats, Wistar
5.
J Neurosci ; 24(21): 4978-88, 2004 May 26.
Article in English | MEDLINE | ID: mdl-15163690

ABSTRACT

Recent studies implicate dendritic endocannabinoid release from subsynaptic dendrites and subsequent inhibition of neurotransmitter release from nerve terminals as a means of retrograde signaling in multiple brain regions. Here we show that type 1 cannabinoid receptor-mediated endocannabinoid signaling is not involved in the retrograde control of synaptic efficacy at inhibitory synapses between fast-spiking interneurons and pyramidal cells in layer 2/3 of the neocortex. Vesicular neurotransmitter transporters, such as vesicular glutamate transporters (VGLUTs) 1 and 2, are localized to presynaptic terminals and accumulate neurotransmitters into synaptic vesicles. A third subtype of VGLUTs (VGLUT3) was recently identified and found localized to dendrites of various cell types. We demonstrate, using multiple immunofluorescence labeling and confocal laser-scanning microscopy, that VGLUT3-like immunoreactivity is present in dendrites of layer 2/3 pyramidal neurons in the rat neocortex. Electron microscopy analysis confirmed that VGLUT3-like labeling is localized to vesicular structures, which show a tendency to accumulate in close proximity to postsynaptic specializations in dendritic shafts of pyramidal cells. Dual whole-cell recordings revealed that retrograde signaling between fast-spiking interneurons and pyramidal cells was enhanced under conditions of maximal efficacy of VGLUT3-mediated glutamate uptake, whereas it was reduced when glutamate uptake was inhibited by incrementing concentrations of the nonselective VGLUT inhibitor Evans blue (0.5-5.0 microm) or intracellular Cl- concentrations (4-145 mm). Our results present further evidence that dendritic vesicular glutamate release, controlled by novel VGLUT isoforms, provides fast negative feedback at inhibitory neocortical synapses, and demonstrate that glutamate can act as a retrograde messenger in the CNS.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Neocortex/physiology , Receptors, Cannabinoid/metabolism , Synapses/physiology , Animals , Dendrites/physiology , Dendrites/ultrastructure , Interneurons/metabolism , Neocortex/ultrastructure , Patch-Clamp Techniques , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/ultrastructure , Vesicular Glutamate Transport Proteins
6.
Int J Dev Neurosci ; 22(7): 533-43, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15465283

ABSTRACT

The amazing morphological and electrophysiological diversity of cortical GABAergic interneurons subserves the broad diversity of processes these cells modulate in neuronal networks. Until recently, interneuron development and functions have been extensively studied in heterogeneous in vitro and in vivo systems containing both excitatory and inhibitory components. However, mechanisms of interneuron specification during development, key signaling mechanisms controlling the establishment of particular inhibitory neuron subsets, and the spatial and temporal regulation of their integration in neuronal microcircuits remain poorly understood. Selective isolation of particular interneuron subsets may significantly extend our knowledge on the scenario of neurochemical and electrophysiological specification of developing interneurons, identification of signaling cues directing their axon growth, and principles of their anterograde and retrograde synaptic communication with other cell types. Here, we show that selective isolation of perisomatic inhibitory cells containing either parvalbumin or cholecystokinin reveals major differences in the temporal dynamics of their functional differentiation, and their dependence on target-derived signals like brain-derived neurotrophic factor and endocannabinoids. In addition, we discuss therapeutic prospects of modulating increased excitatory output in the hippocampus and subthalamic nucleus by re-adjusting the inhibitory control of principal cells.


Subject(s)
Brain/metabolism , Huntington Disease/metabolism , Interneurons/metabolism , Nerve Net/metabolism , Parkinson Disease/metabolism , Receptors, GABA/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Biomarkers/metabolism , Brain/embryology , Brain/pathology , Humans , Huntington Disease/embryology , Huntington Disease/pathology , Interneurons/pathology , Nerve Net/pathology , Neuronal Plasticity , Parkinson Disease/embryology , Parkinson Disease/pathology , Synaptic Transmission
7.
J Neurobiol ; 66(13): 1437-51, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17013928

ABSTRACT

Molecular mechanisms of neurotrophin signaling on dendrite development and dynamics are only partly understood. To address the role of brain-derived neurotrophic factor (BDNF) in the morphogenesis of GABAergic neurons of the main olfactory bulb, we analyzed mice lacking BDNF, mice carrying neurotrophin-3 (NT3) in the place of BDNF, and TrkB signaling mutant mice with a receptor that can activate phospholipase Cgamma (PLCgamma) but is unable to recruit the adaptors Shc/Frs2. BDNF deletion yielded a compressed olfactory bulb with a significant loss of parvalbumin (PV) immunoreactivity in GABAergic interneurons of the external plexiform layer. Dendrite development of PV-positive interneurons was selectively attenuated by BDNF since other Ca2+ -binding protein-containing neuron populations appeared unaffected. The deficit in PV-positive neurons could be rescued by the NT3/NT3 alleles. The degree of PV immunoreactivity was dependent on BDNF and TrkB recruitment of the adaptor proteins Shc/Frs2. In contrast, PLCgamma signaling from the TrkB receptor was sufficient for dendrite growth in vivo and consistently, blocking PLCgamma prevented BDNF-dependent dendrite development in vitro. Collectively, our results provide genetic evidence that BDNF and TrkB signaling selectively regulate PV expression and dendrite growth in a subset of neurochemically-defined GABAergic interneurons via activation of the PLCgamma pathway.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Dendrites/drug effects , Interneurons , Olfactory Bulb/cytology , Parvalbumins/metabolism , Phospholipase C gamma/metabolism , Animals , Animals, Newborn , Blotting, Western , Brain-Derived Neurotrophic Factor/deficiency , Cells, Cultured , Dendrites/ultrastructure , Drug Interactions , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry/methods , Interneurons/cytology , Interneurons/drug effects , Interneurons/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques/methods , Phospholipase C gamma/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, trkB/genetics , Signal Transduction/physiology , Silver Staining/methods
8.
Proc Natl Acad Sci U S A ; 102(52): 19115-20, 2005 Dec 27.
Article in English | MEDLINE | ID: mdl-16357196

ABSTRACT

In utero exposure to Delta(9)-tetrahydrocannabinol (Delta(9)-THC), the active component from marijuana, induces cognitive deficits enduring into adulthood. Although changes in synaptic structure and plasticity may underlie Delta(9)-THC-induced cognitive impairments, the neuronal basis of Delta(9)-THC-related developmental deficits remains unknown. Using a Boyden chamber assay, we show that agonist stimulation of the CB(1) cannabinoid receptor (CB(1)R) on cholecystokinin-expressing interneurons induces chemotaxis that is additive with brain-derived neurotrophic factor (BDNF)-induced interneuron migration. We find that Src kinase-dependent TrkB receptor transactivation mediates endocannabinoid (eCB)-induced chemotaxis in the absence of BDNF. Simultaneously, eCBs suppress the BDNF-dependent morphogenesis of interneurons, and this suppression is abolished by Src kinase inhibition in vitro. Because sustained prenatal Delta(9)-THC stimulation of CB(1)Rs selectively increases the density of cholecystokinin-expressing interneurons in the hippocampus in vivo, we conclude that prenatal CB(1)R activity governs proper interneuron placement and integration during corticogenesis. Moreover, eCBs use TrkB receptor-dependent signaling pathways to regulate subtype-selective interneuron migration and specification.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Cerebral Cortex/drug effects , Endocannabinoids , Interneurons/metabolism , Neurons/metabolism , Receptor, trkB/metabolism , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/physiology , Cell Differentiation , Cell Movement , Cerebral Cortex/embryology , Chemotaxis , Dose-Response Relationship, Drug , Electrophysiology , Hippocampus/metabolism , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Morphogenesis , Neuronal Plasticity , Rabbits , Rats , Receptors, Cannabinoid/metabolism , Receptors, Drug/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Substance-Related Disorders , Time Factors , Transcriptional Activation , Transfection , gamma-Aminobutyric Acid/metabolism , src-Family Kinases/metabolism
9.
Eur J Neurosci ; 18(7): 1979-92, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14622230

ABSTRACT

Basal forebrain cholinergic neurons project to diverse cortical and hippocampal areas and receive reciprocal projections therefrom. Maintenance of a fine-tuned synaptic communication between pre- and postsynaptic cells in neuronal circuitries also requires feedback mechanisms to control the probability of neurotransmitter release from the presynaptic terminal. Release of endocannabinoids or glutamate from a postsynaptic neuron has been identified as a means of retrograde synaptic signalling. Presynaptic action of endocannabinoids is largely mediated by type 1 cannabinoid (CB1) receptors, while fatty-acid amide hydrolase (FAAH) is involved in inactivating some endocannabinoids postsynaptically. Alternatively, vesicular glutamate transporter 3 (VGLUT3) controls release of glutamate from postsynaptic cells. Here, we studied the distribution of CB1 receptors, FAAH and VGLUT3 in cholinergic basal forebrain nuclei of mouse and rat. Cholinergic neurons were devoid of CB1 receptor immunoreactivity. A fine CB1 receptor-immunoreactive (ir) fibre meshwork was present in medial septum, diagonal bands and nucleus basalis. In contrast, the ventral pallidum and substantia innominata received dense CB1 receptor-ir innervation and cholinergic neurons received CB1 receptor-ir presumed synaptic contacts. Consistent with CB1 receptor distribution, FAAH-ir somata were abundant in basal forebrain and appeared in contact with CB1 receptor-containing terminals. Virtually all cholinergic neurons were immunoreactive for FAAH. A significant proportion of cholinergic cells exhibited VGLUT3 immunoreactivity in medial septum, diagonal bands and nucleus basalis, and were in close apposition to VGLUT3-ir terminals. VGLUT3 immunoreactivity was largely absent in ventral pallidum and substantia innominata. We propose that specific subsets of cholinergic neurons may utilize endocannabinoids or glutamate for retrograde control of the efficacy of input synapses, and the mutually exclusive complementary distribution pattern of CB1 receptor-ir and VGLUT3-ir fibres in basal forebrain suggests segregated input-specific signalling mechanisms by cholinergic neurons.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Choline O-Acetyltransferase/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Receptor, Cannabinoid, CB1/metabolism , Amidohydrolases/metabolism , Animals , Fluorescent Antibody Technique/methods , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Parvalbumins/metabolism , Phosphopyruvate Hydratase/metabolism , Prosencephalon/cytology , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism , Signal Transduction , Sincalide/metabolism , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism , Vesicular Glutamate Transport Proteins
10.
Eur J Neurosci ; 20(5): 1290-306, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15341601

ABSTRACT

GABAergic interneurons with high-frequency firing, fast-spiking (FS) cells, form synapses on perisomatic regions of principal cells in the neocortex and hippocampus to control the excitability of cortical networks. Brain-derived neurotrophic factor (BDNF) is essential for the differentiation of multiple interneuron subtypes and the formation of their synaptic contacts. Here, we examined whether BDNF, alone or in conjunction with sustained KCl-induced depolarization, drives functional FS cell differentiation and the formation of inhibitory microcircuits. Homogeneous FS cell cultures were established by target-specific isolation using the voltage-gated potassium channel 3.1b subunit as the selection marker. Isolated FS cells expressed parvalbumin, were surrounded by perineuronal nets, formed immature inhibitory connections and generated slow action potentials at 12 days in vitro. Brain-derived neurotrophic factor (BDNF) promoted FS cell differentiation by increasing the somatic diameter, dendritic branching and the frequency of action potential firing. In addition, BDNF treatment led to a significant up-regulation of synaptophysin and vesicular GABA transporter expression, components of the synaptic machinery critical for GABA release, which was paralleled by an increase in synaptic strength. Long-term membrane depolarization alone was detrimental to dendritic branching. However, we observed that BDNF and KCl exerted additive effects, as reflected by the significantly accelerated maturation of synaptic contacts and high discharge frequencies, and was required for the formation of reciprocal connections between FS cells. Our results show that BDNF, along with membrane depolarization, is critical for FS cells to establish inhibitory circuitries during corticogenesis.


Subject(s)
Action Potentials/physiology , Brain-Derived Neurotrophic Factor/physiology , Cell Differentiation/physiology , Nerve Net/physiology , Potassium Channels, Voltage-Gated , gamma-Aminobutyric Acid/physiology , Animals , Cells, Cultured , Female , Interneurons/cytology , Interneurons/physiology , Nerve Net/cytology , Nerve Tissue Proteins/physiology , Potassium Channels/physiology , Pregnancy , Rats , Rats, Sprague-Dawley , Shaw Potassium Channels
SELECTION OF CITATIONS
SEARCH DETAIL