Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 4249, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31534164

ABSTRACT

The first wave of oligodendrocyte precursor cells (firstOPCs) and most GABAergic interneurons share common embryonic origins. Cortical firstOPCs are thought to be replaced by other OPC populations shortly after birth, maintaining a consistent OPC density and making postnatal interactions between firstOPCs and ontogenetically-related interneurons unlikely. Challenging these ideas, we show that a cortical firstOPC subpopulation survives and forms functional cell clusters with lineage-related interneurons. Favored by a common embryonic origin, these clusters display unexpected preferential synaptic connectivity and are anatomically maintained after firstOPCs differentiate into myelinating oligodendrocytes. While the concomitant rescue of interneurons and firstOPCs committed to die causes an exacerbated neuronal inhibition, it abolishes interneuron-firstOPC high synaptic connectivity. Further, the number of other oligodendroglia populations increases through a non-cell-autonomous mechanism, impacting myelination. These findings demonstrate unprecedented roles of interneuron and firstOPC apoptosis in regulating lineage-related cell interactions and the homeostatic oligodendroglia density.


Subject(s)
Apoptosis/physiology , Interneurons/metabolism , Neurogenesis/physiology , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Animals , Central Nervous System/cytology , Central Nervous System/embryology , Female , GABAergic Neurons/cytology , Homeodomain Proteins/metabolism , Interneurons/cytology , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Oligodendroglia/cytology
2.
Elife ; 82019 12 31.
Article in English | MEDLINE | ID: mdl-31891351

ABSTRACT

Programmed cell death and early activity contribute to the emergence of functional cortical circuits. While most neuronal populations are scaled-down by death, some subpopulations are entirely eliminated, raising the question of the importance of such demise for cortical wiring. Here, we addressed this issue by focusing on Cajal-Retzius neurons (CRs), key players in cortical development that are eliminated in postnatal mice in part via Bax-dependent apoptosis. Using Bax-conditional mutants and CR hyperpolarization, we show that the survival of electrically active subsets of CRs triggers an increase in both dendrite complexity and spine density of upper layer pyramidal neurons, leading to an excitation/inhibition imbalance. The survival of these CRs is induced by hyperpolarization, highlighting an interplay between early activity and neuronal elimination. Taken together, our study reveals a novel activity-dependent programmed cell death process required for the removal of transient immature neurons and the proper wiring of functional cortical circuits.


Subject(s)
Apoptosis/genetics , Neurogenesis/genetics , Pyramidal Cells/metabolism , bcl-2-Associated X Protein/genetics , Animals , Animals, Newborn , Cell Polarity/genetics , Cerebral Cortex/metabolism , Electric Stimulation , Interstitial Cells of Cajal/metabolism , Mice , Mutant Proteins/genetics , Pyramidal Cells/pathology
3.
Front Cell Neurosci ; 12: 477, 2018.
Article in English | MEDLINE | ID: mdl-30574070

ABSTRACT

Optogenetic and pharmacogenetic techniques have been effective to analyze the role of neuronal activity in controlling oligodendroglia lineage cells in behaving juvenile and adult mice. This kind of studies is also of high interest during early postnatal (PN) development since important changes in oligodendroglia dynamics occur during the first two PN weeks. Yet, neuronal manipulation is difficult to implement at an early age because high-level, specific protein expression is less reliable in neonatal mice. Here, we describe a protocol allowing for an optogenetic stimulation of neurons in awake mouse pups with the purpose of investigating the effect of neuronal activity on oligodendroglia dynamics during early PN stages. Since GABAergic interneurons contact oligodendrocyte precursor cells (OPCs) through bona fide synapses and maintain a close relationship with these progenitors during cortical development, we used this relevant example of neuron-oligodendroglia interaction to implement a proof-of-principle optogenetic approach. First, we tested Nkx2.1-Cre and Parvalbumin (PV)-Cre lines to drive the expression of the photosensitive ion channel channelrhodopsin-2 (ChR2) in subpopulations of interneurons at different developmental stages. By using patch-clamp recordings and photostimulation of ChR2-positive interneurons in acute somatosensory cortical slices, we analyzed the level of functional expression of ChR2 in these neurons. We found that ChR2 expression was insufficient in PV-Cre mouse at PN day 10 (PN10) and that this channel needs to be expressed from embryonic stages (as in the Nkx2.1-Cre line) to allow for a reliable photoactivation in mouse pups. Then, we implemented a stereotaxic surgery to place a mini-optic fiber at the cortical surface in order to photostimulate ChR2-positive interneurons at PN10. In vivo field potentials were recorded in Layer V to verify that photostimulation reaches deep cortical layers. Finally, we analyzed the effect of the photostimulation on the layer V oligodendroglia population by conventional immunostainings. Neither the total density nor a proliferative fraction of OPCs were affected by increasing interneuron activity in vivo, complementing previous findings showing the lack of effect of GABAergic synaptic activity on OPC proliferation. The methodology described here should provide a framework for future investigation of the role of early cellular interactions during PN brain maturation.

4.
Cell Rep ; 17(12): 3133-3141, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28009284

ABSTRACT

Cajal-Retzius cells (CRs), the first-born neurons in the developing cerebral cortex, coordinate crucial steps in the construction of functional circuits. CRs are thought to be transient, as they disappear during early postnatal life in both mice and humans, where their abnormal persistence is associated with pathological conditions. Embryonic CRs comprise at least three molecularly and functionally distinct subtypes: septum, ventral pallium/pallial-subpallial boundary (PSB), and hem. However, whether subtype-specific features exist postnatally and through which mechanisms they disappear remain unknown. We report that CR subtypes display unique distributions and dynamics of death in the postnatal mouse cortex. Surprisingly, although all CR subtypes undergo cell death, septum, but not hem, CRs die in a Bax-dependent manner. Bax-inactivated rescued septum-CRs maintain immature electrophysiological properties. These results underlie the existence of an exquisitely refined control of developmental cell death and provide a model to test the effect of maintaining immature circuits in the adult neocortex.


Subject(s)
Cell Death/genetics , Cerebral Cortex/metabolism , Neurons/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Lineage/genetics , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Embryo, Mammalian , Humans , Mice
6.
Elife ; 42015 Apr 22.
Article in English | MEDLINE | ID: mdl-25902404

ABSTRACT

NG2 cells, oligodendrocyte progenitors, receive a major synaptic input from interneurons in the developing neocortex. It is presumed that these precursors integrate cortical networks where they act as sensors of neuronal activity. We show that NG2 cells of the developing somatosensory cortex form a transient and structured synaptic network with interneurons that follows its own rules of connectivity. Fast-spiking interneurons, highly connected to NG2 cells, target proximal subcellular domains containing GABAA receptors with γ2 subunits. Conversely, non-fast-spiking interneurons, poorly connected with these progenitors, target distal sites lacking this subunit. In the network, interneuron-NG2 cell connectivity maps exhibit a local spatial arrangement reflecting innervation only by the nearest interneurons. This microcircuit architecture shows a connectivity peak at PN10, coinciding with a switch to massive oligodendrocyte differentiation. Hence, GABAergic innervation of NG2 cells is temporally and spatially regulated from the subcellular to the network level in coordination with the onset of oligodendrogenesis.


Subject(s)
Interneurons/ultrastructure , Neocortex/cytology , Neural Stem Cells/ultrastructure , Oligodendroglia/ultrastructure , Somatosensory Cortex/cytology , Action Potentials/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation , Gene Expression , Genes, Reporter , Interneurons/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Microtomy , Neocortex/growth & development , Neocortex/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Oligodendroglia/metabolism , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Somatosensory Cortex/growth & development , Somatosensory Cortex/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
7.
Front Cell Neurosci ; 8: 364, 2014.
Article in English | MEDLINE | ID: mdl-25414639

ABSTRACT

Purkinje cells (PC) control spike timing of neighboring PC by their recurrent axon collaterals. These synapses underlie fast cerebellar oscillations and are characterized by a strong facilitation within a time window of <20 ms during paired-pulse protocols. PC express high levels of the fast Ca(2+) buffer protein calbindin D-28k (CB). As expected from the absence of a fast Ca(2+) buffer, presynaptic action potential-evoked [Ca(2+)]i transients were previously shown to be bigger in PC boutons of young (second postnatal week) CB-/- mice, yet IPSC mean amplitudes remained unaltered in connected CB-/- PC. Since PC spine morphology is altered in adult CB-/- mice (longer necks, larger spine head volume), we summoned that morphological compensation/adaptation mechanisms might also be induced in CB-/- PC axon collaterals including boutons. In these mice, biocytin-filled PC reconstructions revealed that the number of axonal varicosities per PC axon collateral was augmented, mostly confined to the granule cell layer. Additionally, the volume of individual boutons was increased, evidenced from z-stacks of confocal images. EM analysis of PC-PC synapses revealed an enhancement in active zone (AZ) length by approximately 23%, paralleled by a higher number of docked vesicles per AZ in CB-/- boutons. Moreover, synaptic cleft width was larger in CB-/- (23.8 ± 0.43 nm) compared to wild type (21.17 ± 0.39 nm) synapses. We propose that the morphological changes, i.e., the larger bouton volume, the enhanced AZ length and the higher number of docked vesicles, in combination with the increase in synaptic cleft width likely modifies the GABA release properties at this synapse in CB-/- mice. We view these changes as adaptation/homeostatic mechanisms to likely maintain characteristics of synaptic transmission in the absence of the fast Ca(2+) buffer CB. Our study provides further evidence on the functioning of the Ca(2+) homeostasome.

8.
J Physiol ; 591(13): 3215-32, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23551945

ABSTRACT

Striatal fast spiking interneurons (FSIs) modulate output of the striatum by synchronizing medium-sized spiny neurons (MSNs). Recent studies have broadened our understanding of FSIs, showing that they are implicated in severe motor disorders such as parkinsonism, dystonia and Tourette syndrome. FSIs are the only striatal neurons to express the calcium-binding protein parvalbumin (PV). This selective expression of PV raises questions about the functional role of this Ca(2+) buffer in controlling FSI Ca(2+) dynamics and, consequently, FSI spiking mode and neurotransmission. To study the functional involvement of FSIs in striatal microcircuit activity and the role of PV in FSI function, we performed perforated patch recordings on enhanced green fluorescent protein-expressing FSIs in brain slices from control and PV-/- mice. Our results revealed that PV-/- FSIs fired more regularly and were more excitable than control FSIs by a mechanism in which Ca(2+) buffering is linked to spiking activity as a result of the activation of small conductance Ca(2+)-dependent K(+) channels. A modelling approach of striatal FSIs supports our experimental results. Furthermore, PV deletion modified frequency-specific short-term plasticity at inhibitory FSI to MSN synapses. Our results therefore reinforce the hypothesis that in FSIs, PV is crucial for fine-tuning of the temporal responses of the FSI network and for the orchestration of MSN populations. This, in turn, may play a direct role in the generation and pathology-related worsening of motor rhythms.


Subject(s)
Corpus Striatum/physiology , Interneurons/physiology , Parvalbumins/physiology , Animals , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neuronal Plasticity
9.
Neuron ; 77(3): 440-56, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23395372

ABSTRACT

The study of human cortical development has major implications for brain evolution and diseases but has remained elusive due to paucity of experimental models. Here we found that human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), cultured without added morphogens, recapitulate corticogenesis leading to the sequential generation of functional pyramidal neurons of all six layer identities. After transplantation into mouse neonatal brain, human ESC-derived cortical neurons integrated robustly and established specific axonal projections and dendritic patterns corresponding to native cortical neurons. The differentiation and connectivity of the transplanted human cortical neurons complexified progressively over several months in vivo, culminating in the establishment of functional synapses with the host circuitry. Our data demonstrate that human cortical neurons generated in vitro from ESC/iPSC can develop complex hodological properties characteristic of the cerebral cortex in vivo, thereby offering unprecedented opportunities for the modeling of human cortex diseases and brain repair.


Subject(s)
Brain/cytology , Embryonic Stem Cells/cytology , Nerve Net/physiology , Pluripotent Stem Cells/physiology , Pyramidal Cells/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Age Factors , Animals , Axons/physiology , Bromodeoxyuridine , Calcium/metabolism , Cell Differentiation , Cell Transplantation , Cells, Cultured , Dendrites/physiology , Evoked Potentials/physiology , Excitatory Amino Acid Antagonists/pharmacology , Female , Fetus , Fluorescent Dyes/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Humans , In Vitro Techniques , Mice , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Nerve Net/ultrastructure , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Patch-Clamp Techniques , Pregnancy , Pyramidal Cells/cytology , RNA, Messenger/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptic Potentials/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic , Tyrosine 3-Monooxygenase/metabolism , Valine/analogs & derivatives , Valine/pharmacology , Vesicular Glutamate Transport Protein 1/metabolism
10.
J Neurosci ; 31(33): 11795-807, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849540

ABSTRACT

The contribution of neuronal dysfunction to neurodegeneration is studied in a mouse model of spinocerebellar ataxia type 1 (SCA1) displaying impaired motor performance ahead of loss or atrophy of cerebellar Purkinje cells. Presymptomatic SCA1 mice show a reduction in the firing rate of Purkinje cells (both in vivo and in slices) associated with a reduction in the efficiency of the main glutamatergic synapse onto Purkinje cells and with increased A-type potassium current. The A-type potassium channel Kv4.3 appears to be internalized in response to glutamatergic stimulation in Purkinje cells and accumulates in presymptomatic SCA1 mice. SCA1 mice are treated with aminopyridines, acting as potassium channel blockers to test whether the treatment could improve neuronal dysfunction, motor behavior, and neurodegeneration. In acutely treated young SCA1 mice, aminopyridines normalize the firing rate of Purkinje cells and the motor behavior of the animals. In chronically treated old SCA1 mice, 3,4-diaminopyridine improves the firing rate of Purkinje cells, the motor behavior of the animals, and partially protects against cell atrophy. Chronic treatment with 3,4-diaminopyridine is associated with increased cerebellar levels of BDNF, suggesting that partial protection against atrophy of Purkinje cells is possibly provided by an increased production of growth factors secondary to the reincrease in electrical activity. Our data suggest that aminopyridines might have symptomatic and/or neuroprotective beneficial effects in SCA1, that reduction in the firing rate of Purkinje cells can cause cerebellar ataxia, and that treatment of early neuronal dysfunction is relevant in neurodegenerative disorders such as SCA1.


Subject(s)
Aminopyridines/therapeutic use , Motor Skills Disorders/prevention & control , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Neuroprotective Agents/therapeutic use , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/physiopathology , Action Potentials/drug effects , Action Potentials/physiology , Aminopyridines/pharmacology , Animals , Conditioning, Eyelid/drug effects , Conditioning, Eyelid/physiology , Disease Models, Animal , Mice , Mice, Transgenic , Motor Skills Disorders/physiopathology , Neuroprotective Agents/pharmacology , Time Factors
11.
Mol Neurobiol ; 44(1): 111-21, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21710140

ABSTRACT

The potassium channel tetramerization domain-containing protein 7 (KCTD7) was named after the structural homology of its predicted N-terminal broad complex, tramtrack and bric à brac/poxvirus and zinc finger domain with the T1 domain of the Kv potassium channel, but its expression profile and cellular function are still largely unknown. We have recently reported a homozygous nonsense mutation of KCTD7 in patients with a novel form of autosomal recessive progressive myoclonic epilepsy. Here, we show that KCTD7 expression hyperpolarizes the cell membrane and reduces the excitability of transfected neurons in patch clamp experiments. We found the expression of KCTD7 in the hippocampal and Purkinje cells of the murine brain, an expression profile consistent with our patients' phenotype. The effect on the plasma membrane resting potential is possibly mediated by Cullin-3, as we demonstrated direct molecular interaction of KCTD7 with Cullin-3 in co-immunoprecipitation assays. Our data link progressive myoclonic epilepsy to an inherited defect of the neuron plasma membrane's resting potential in the brain.


Subject(s)
Ion Channel Gating/genetics , Myoclonic Epilepsies, Progressive/genetics , Neurons/metabolism , Potassium Channels/genetics , Action Potentials/physiology , Animals , Antibody Specificity , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cullin Proteins/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunoprecipitation , Mice , Neurons/pathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Potassium Channels/metabolism , Protein Binding , Protein Multimerization , Protein Transport , Purkinje Cells/metabolism , Purkinje Cells/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
J Neurosci ; 29(42): 13126-35, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-19846700

ABSTRACT

The B05 transgenic SCA1 mice, expressing human ataxin-1 with an expanded polyglutamine tract in cerebellar Purkinje cells (PCs), recapitulate many pathological and behavioral characteristics of the neurodegenerative disease spinocerebellar ataxia type 1 (SCA1), including progressive ataxia and PC loss. We transplanted neural precursor cells (NPCs) derived from the subventricular zone of GFP-expressing adult mice into the cerebellar white matter of SCA1 mice when they showed absent (5 weeks), initial (13 weeks), and significant (24 weeks) PC loss. Only in mice with significant cell loss, grafted NPCs migrated into the cerebellar cortex. These animals showed improved motor skills compared with sham-treated controls. No grafted cell adopted the morphological and immunohistochemical characteristics of PCs, but the cerebellar cortex in NPC-grafted SCA1 mice had a significantly thicker molecular layer and more surviving PCs. Perforated patch-clamp recordings revealed a normalization of the PC basal membrane potential, which was abnormally depolarized in sham-treated animals. No significant increase in levels of several neurotrophic factors was observed, suggesting, along with morphological observation, that the neuroprotective effect of grafted NPCs was mediated by direct contact with the host PCs. We postulate that a similar neuroprotective effect of NPCs may be applicable to other cerebellar degenerative diseases.


Subject(s)
Adult Stem Cells/physiology , Neurons/physiology , Recovery of Function/physiology , Spinocerebellar Ataxias/surgery , Stem Cell Transplantation/methods , Adult Stem Cells/transplantation , Analysis of Variance , Animals , Ataxin-1 , Ataxins , Cell Movement/physiology , Cerebral Ventricles/cytology , Dendrites/pathology , Dendrites/physiology , Disease Models, Animal , Green Fluorescent Proteins/genetics , Hand Strength/physiology , Humans , Membrane Potentials/genetics , Membrane Potentials/physiology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Motor Activity/genetics , Motor Activity/physiology , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/pathology , Nuclear Proteins/genetics , Patch-Clamp Techniques , Peptides/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Spinocerebellar Ataxias/physiopathology , Time Factors
13.
Proc Natl Acad Sci U S A ; 104(45): 17831-6, 2007 Nov 06.
Article in English | MEDLINE | ID: mdl-17965230

ABSTRACT

Morphological studies have provided ample evidence for synaptic connections between cerebellar Purkinje cells (PCs), but the functional properties of these synapses remain elusive. We report on direct recordings of synaptically connected PCs in mice cerebellar slices. In PCs filled with a fluorescent dye to aid axon visualization and postsynaptic target identification, presynaptic action potentials elicited unitary inhibitory postsynaptic currents in neighboring PCs in 10% of potential connections tested. In 11 pairs, postsynaptic currents had a delay onset of 1.62 +/- 0.16 ms with respect to the presynaptic spike, a 10-90% rise time of 2.20 +/- 0.33 ms, and a monoexponential decay with a time constant of 13.3 +/- 1.7 ms. Average values for peak current and variance-to-mean ratio were 55 +/- 14 and 30 +/- 3 pA, respectively. In contrast to the depressing nature of the synapse between PCs and deep cerebellar nuclei neurons, PC-PC synapses exhibited strong facilitation operating within a time window of a few milliseconds; paired-pulse ratios for 3- and 20-ms intervals were 1.79 +/- 0.18 and 1.01 +/- 0.14, respectively (n = 6). The facilitation is of presynaptic nature because it is accompanied by a decrease in failure rate. Trains of action potentials evoked in presynaptic varicosities volume-averaged calcium transients whose peak increased 1.7-fold as the frequency increased from 50 to 166 Hz. We suggest that PC-PC synapses are tuned for high fidelity of transmission during bursts of PC activity and that their operation in the cerebellar circuit modulates synchronized PC firing.


Subject(s)
Axons/physiology , Cell Communication/physiology , Cerebellum/physiology , Purkinje Cells/physiology , Synapses/physiology , Action Potentials , Animals , Cerebellar Nuclei/physiology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/physiology , Mice , Reaction Time
SELECTION OF CITATIONS
SEARCH DETAIL
...