Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nature ; 622(7981): 120-129, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37674083

ABSTRACT

Multimodal astrocyte-neuron communications govern brain circuitry assembly and function1. For example, through rapid glutamate release, astrocytes can control excitability, plasticity and synchronous activity2,3 of synaptic networks, while also contributing to their dysregulation in neuropsychiatric conditions4-7. For astrocytes to communicate through fast focal glutamate release, they should possess an apparatus for Ca2+-dependent exocytosis similar to neurons8-10. However, the existence of this mechanism has been questioned11-13 owing to inconsistent data14-17 and a lack of direct supporting evidence. Here we revisited the astrocyte glutamate exocytosis hypothesis by considering the emerging molecular heterogeneity of astrocytes18-21 and using molecular, bioinformatic and imaging approaches, together with cell-specific genetic tools that interfere with glutamate exocytosis in vivo. By analysing existing single-cell RNA-sequencing databases and our patch-seq data, we identified nine molecularly distinct clusters of hippocampal astrocytes, among which we found a notable subpopulation that selectively expressed synaptic-like glutamate-release machinery and localized to discrete hippocampal sites. Using GluSnFR-based glutamate imaging22 in situ and in vivo, we identified a corresponding astrocyte subgroup that responds reliably to astrocyte-selective stimulations with subsecond glutamate release events at spatially precise hotspots, which were suppressed by astrocyte-targeted deletion of vesicular glutamate transporter 1 (VGLUT1). Furthermore, deletion of this transporter or its isoform VGLUT2 revealed specific contributions of glutamatergic astrocytes in cortico-hippocampal and nigrostriatal circuits during normal behaviour and pathological processes. By uncovering this atypical subpopulation of specialized astrocytes in the adult brain, we provide insights into the complex roles of astrocytes in central nervous system (CNS) physiology and diseases, and identify a potential therapeutic target.


Subject(s)
Astrocytes , Central Nervous System , Glutamic Acid , Signal Transduction , Adult , Humans , Astrocytes/classification , Astrocytes/cytology , Astrocytes/metabolism , Central Nervous System/cytology , Central Nervous System/metabolism , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Neurons/metabolism , Synaptic Transmission , Calcium/metabolism , Exocytosis , Single-Cell Gene Expression Analysis , Vesicular Glutamate Transport Protein 1/deficiency , Vesicular Glutamate Transport Protein 1/genetics , Gene Deletion , Cerebral Cortex/cytology , Cerebral Cortex/metabolism
2.
Neuron ; 111(7): 1094-1103.e8, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36731469

ABSTRACT

Parental behaviors secure the well-being of newborns and concomitantly limit negative affective states in adults, which emerge when coping with neonatal distress becomes challenging. Whether negative-affect-related neuronal circuits orchestrate parental actions is unknown. Here, we identify parental signatures in lateral habenula neurons receiving bed nucleus of stria terminalis innervation (BNSTLHb). We find that LHb neurons of virgin female mice increase their activity following pup distress vocalization and are necessary for pup-call-driven aversive behaviors. LHb activity rises during pup retrieval, a behavior worsened by LHb inactivation. Intersectional cell identification and transcriptional profiling associate BNSTLHb cells to parenting and outline a gene expression in female virgins similar to that in mothers but different from that in non-parental virgin male mice. Finally, tracking and manipulating BNSTLHb cell activity demonstrates their specificity for encoding negative affect and pup retrieval. Thus, a negative affect neural circuit processes newborn distress signals and may limit them by guiding female parenting.


Subject(s)
Habenula , Neurons , Mice , Animals , Male , Female , Neurons/physiology , Avoidance Learning , Affect , Habenula/physiology
3.
Sci Adv ; 8(46): eabo4552, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36399562

ABSTRACT

During corticogenesis, dynamic regulation of apical adhesion is fundamental to generate correct numbers and cell identities. While radial glial cells (RGCs) maintain basal and apical anchors, basal progenitors and neurons detach and settle at distal positions from the apical border. Whether diffusible signals delivered from the cerebrospinal fluid (CSF) contribute to the regulation of apical adhesion dynamics remains fully unknown. Secreted class 3 Semaphorins (Semas) trigger cell responses via Plexin-Neuropilin (Nrp) membrane receptor complexes. Here, we report that unconventional Sema3-Nrp preformed complexes are delivered by the CSF from sources including the choroid plexus to Plexin-expressing RGCs via their apical endfeet. Through analysis of mutant mouse models and various ex vivo assays mimicking ventricular delivery to RGCs, we found that two different complexes, Sema3B/Nrp2 and Sema3F/Nrp1, exert dual effects on apical endfeet dynamics, nuclei positioning, and RGC progeny. This reveals unexpected balance of CSF-delivered guidance molecules during cortical development.

4.
Nat Commun ; 12(1): 7362, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934077

ABSTRACT

Neural stem/progenitor cells (NSPCs) generate new neurons throughout adulthood. However, the underlying regulatory processes are still not fully understood. Lipid metabolism plays an important role in regulating NSPC activity: build-up of lipids is crucial for NSPC proliferation, whereas break-down of lipids has been shown to regulate NSPC quiescence. Despite their central role for cellular lipid metabolism, the role of lipid droplets (LDs), the lipid storing organelles, in NSPCs remains underexplored. Here we show that LDs are highly abundant in adult mouse NSPCs, and that LD accumulation is significantly altered upon fate changes such as quiescence and differentiation. NSPC proliferation is influenced by the number of LDs, inhibition of LD build-up, breakdown or usage, and the asymmetric inheritance of LDs during mitosis. Furthermore, high LD-containing NSPCs have increased metabolic activity and capacity, but do not suffer from increased oxidative damage. Together, these data indicate an instructive role for LDs in driving NSPC behaviour.


Subject(s)
Lipid Droplets/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Inheritance Patterns/genetics , Lipid Peroxidation , Male , Mice, Inbred C57BL , Mitosis , Neurons/cytology , Neurons/metabolism , Perilipin-2/metabolism , Phospholipids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
5.
EMBO Rep ; 22(4): e51404, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33779029

ABSTRACT

Status epilepticus (SE) is a condition in which seizures are not self-terminating and thereby pose a serious threat to the patient's life. The molecular mechanisms underlying SE are likely heterogeneous and not well understood. Here, we reveal a role for the RNA-binding protein Fragile X-Related Protein 2 (FXR2P) in SE. Fxr2 KO mice display reduced sensitivity specifically to kainic acid-induced SE. Immunoprecipitation of FXR2P coupled to next-generation sequencing of associated mRNAs shows that FXR2P targets are enriched in genes that encode glutamatergic post-synaptic components. Of note, the FXR2P target transcriptome has a significant overlap with epilepsy and SE risk genes. In addition, Fxr2 KO mice fail to show sustained ERK1/2 phosphorylation induced by KA and present reduced burst activity in the hippocampus. Taken together, our findings show that the absence of FXR2P decreases the expression of glutamatergic proteins, and this decrease might prevent self-sustained seizures.


Subject(s)
Kainic Acid , Status Epilepticus , Animals , Hippocampus/metabolism , Kainic Acid/toxicity , Mice , Mice, Inbred C57BL , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Seizures/chemically induced , Seizures/genetics , Status Epilepticus/chemically induced , Status Epilepticus/genetics
6.
Curr Opin Neurobiol ; 66: 116-124, 2021 02.
Article in English | MEDLINE | ID: mdl-33171340

ABSTRACT

Our understanding of the central nervous system (CNS) development has been strongly enhanced by the recent progress of single-cell multiomics approaches. Certainly, the multiplex profiling of individual cell epigenomes and transcriptomes together with dynamic lineage tracing systems brings encouraging new perspectives and prompts a paradigm shift in neuroscience developmental research. In this review, we outline the latest multiomics -based findings in CNS development, from the early CNS patterning to the regional specification of the CNS along anterior-posterior axis (forebrain, midbrain, hindbrain and spinal cord). Overall, multiomics development has substantially impacted current knowledge and has challenged our classical models for embryonic CNS development. Integrating all these newly generated -omics databases represents the next step to overcome challenges in understanding developmental diseases.


Subject(s)
Body Patterning , Spinal Cord , Gene Expression Regulation, Developmental , Prosencephalon
7.
Int J Mol Sci ; 21(20)2020 Oct 11.
Article in English | MEDLINE | ID: mdl-33050604

ABSTRACT

The complexity of brain structure and function is rooted in the precise spatial and temporal regulation of selective developmental events. During neurogenesis, both vertebrates and invertebrates generate a wide variety of specialized cell types through the expansion and specification of a restricted set of neuronal progenitors. Temporal patterning of neural progenitors rests on fine regulation between cell-intrinsic and cell-extrinsic mechanisms. The rapid emergence of high-throughput single-cell technologies combined with elaborate computational analysis has started to provide us with unprecedented biological insights related to temporal patterning in the developing central nervous system (CNS). Here, we present an overview of recent advances in Drosophila and vertebrates, focusing both on cell-intrinsic mechanisms and environmental influences. We then describe the various multi-omics approaches that have strongly contributed to our current understanding and discuss perspectives on the various -omics approaches that hold great potential for the future of temporal patterning research.


Subject(s)
Body Patterning/genetics , Genomics , Metabolomics , Neurogenesis/genetics , Proteomics , Single-Cell Analysis , Temporal Lobe/embryology , Temporal Lobe/metabolism , Animals , Gene Expression Regulation, Developmental , Genomics/methods , Proteomics/methods , Single-Cell Analysis/methods
8.
Cell ; 174(5): 1264-1276.e15, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30057116

ABSTRACT

During corticogenesis, ventricular zone progenitors sequentially generate distinct subtypes of neurons, accounting for the diversity of neocortical cells and the circuits they form. While activity-dependent processes are critical for the differentiation and circuit assembly of postmitotic neurons, how bioelectrical processes affect nonexcitable cells, such as progenitors, remains largely unknown. Here, we reveal that, in the developing mouse neocortex, ventricular zone progenitors become more hyperpolarized as they generate successive subtypes of neurons. Experimental in vivo hyperpolarization shifted the transcriptional programs and division modes of these progenitors to a later developmental status, with precocious generation of intermediate progenitors and a forward shift in the laminar, molecular, morphological, and circuit features of their neuronal progeny. These effects occurred through inhibition of the Wnt-beta-catenin signaling pathway by hyperpolarization. Thus, during corticogenesis, bioelectric membrane properties are permissive for specific molecular pathways to coordinate the temporal progression of progenitor developmental programs and thus neocortical neuron diversity.


Subject(s)
Membrane Potentials , Neocortex/embryology , Neurons/metabolism , Stem Cells/cytology , Animals , Brain/cytology , Brain/embryology , Cell Differentiation , Disease Progression , Electroporation , Female , Gene Expression Regulation, Developmental , Male , Mice , Neocortex/cytology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neurogenesis , Potassium Channels, Inwardly Rectifying/metabolism , Sequence Analysis, RNA , Signal Transduction , Time Factors , Wnt Proteins/metabolism , beta Catenin/metabolism
9.
Nature ; 555(7697): 452-454, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29565398

Subject(s)
Neurons
10.
Nature ; 555(7697): 452-454, 2018 Mar.
Article in English | MEDLINE | ID: mdl-32034370
11.
Nat Commun ; 8(1): 2015, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29222517

ABSTRACT

Input from the sensory organs is required to pattern neurons into topographical maps during development. Dendritic complexity critically determines this patterning process; yet, how signals from the periphery act to control dendritic maturation is unclear. Here, using genetic and surgical manipulations of sensory input in mouse somatosensory thalamocortical neurons, we show that membrane excitability is a critical component of dendritic development. Using a combination of genetic approaches, we find that ablation of N-methyl-D-aspartate (NMDA) receptors during postnatal development leads to epigenetic repression of Kv1.1-type potassium channels, increased excitability, and impaired dendritic maturation. Lesions to whisker input pathways had similar effects. Overexpression of Kv1.1 was sufficient to enable dendritic maturation in the absence of sensory input. Thus, Kv1.1 acts to tune neuronal excitability and maintain it within a physiological range, allowing dendritic maturation to proceed. Together, these results reveal an input-dependent control over neuronal excitability and dendritic complexity in the development and plasticity of sensory pathways.


Subject(s)
Dendrites/physiology , Neurons/physiology , Somatosensory Cortex/physiology , Thalamus/physiology , Animals , Female , Gene Expression Profiling , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Somatosensory Cortex/cytology , Synaptic Transmission/physiology , Thalamus/cytology , Vibrissae/innervation , Vibrissae/physiology
12.
Nat Commun ; 8: 14219, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28134272

ABSTRACT

Cortical GABAergic interneurons constitute a highly diverse population of inhibitory neurons that are key regulators of cortical microcircuit function. An important and heterogeneous group of cortical interneurons specifically expresses the serotonin receptor 3A (5-HT3AR) but how this diversity emerges during development is poorly understood. Here we use single-cell transcriptomics to identify gene expression patterns operating in Htr3a-GFP+ interneurons during early steps of cortical circuit assembly. We identify three main molecular types of Htr3a-GFP+ interneurons, each displaying distinct developmental dynamics of gene expression. The transcription factor Meis2 is specifically enriched in a type of Htr3a-GFP+ interneurons largely confined to the cortical white matter. These MEIS2-expressing interneurons appear to originate from a restricted region located at the embryonic pallial-subpallial boundary. Overall, this study identifies MEIS2 as a subclass-specific marker for 5-HT3AR-containing interstitial interneurons and demonstrates that the transcriptional and anatomical parcellation of cortical interneurons is developmentally coupled.


Subject(s)
Cerebral Cortex/growth & development , GABAergic Neurons/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Interneurons/physiology , Animals , Biomarkers , COUP Transcription Factor II/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/anatomy & histology , Cerebral Cortex/cytology , Embryo, Mammalian , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Profiling/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfluidics/methods , Nerve Net/growth & development , Nerve Tissue Proteins/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Reelin Protein , Sequence Analysis, RNA/methods , Serine Endopeptidases/metabolism , Single-Cell Analysis/methods
13.
Nature ; 538(7623): 96-98, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27669022

ABSTRACT

Modality-specific sensory inputs from individual sense organs are processed in parallel in distinct areas of the neocortex. For each sensory modality, input follows a cortico-thalamo-cortical loop in which a 'first-order' exteroceptive thalamic nucleus sends peripheral input to the primary sensory cortex, which projects back to a 'higher order' thalamic nucleus that targets a secondary sensory cortex. This conserved circuit motif raises the possibility that shared genetic programs exist across sensory modalities. Here we report that, despite their association with distinct sensory modalities, first-order nuclei in mice are genetically homologous across somatosensory, visual, and auditory pathways, as are higher order nuclei. We further reveal peripheral input-dependent control over the transcriptional identity and connectivity of first-order nuclei by showing that input ablation leads to induction of higher-order-type transcriptional programs and rewiring of higher-order-directed descending cortical input to deprived first-order nuclei. These findings uncover an input-dependent genetic logic for the design and plasticity of sensory pathways, in which conserved developmental programs lead to conserved circuit motifs across sensory modalities.


Subject(s)
Afferent Pathways/physiology , Models, Genetic , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Afferent Pathways/cytology , Animals , Auditory Pathways/cytology , Auditory Pathways/physiology , Female , Gene Expression Regulation, Developmental , Geniculate Bodies/cytology , Geniculate Bodies/physiology , Male , Mice , Mice, Inbred C57BL , Somatosensory Cortex/physiology , Thalamic Nuclei/cytology , Thalamic Nuclei/physiology , Transcription, Genetic , Visual Pathways/cytology , Visual Pathways/physiology
14.
Neuron ; 91(6): 1276-1291, 2016 Sep 21.
Article in English | MEDLINE | ID: mdl-27618676

ABSTRACT

Subcellular target recognition in the CNS is the culmination of a multiple-step program including axon guidance, target recognition, and synaptogenesis. In cerebellum, basket cells (BCs) innervate the soma and axon initial segment (AIS) of Purkinje cells (PCs) to form the pinceau synapse, but the underlying mechanisms remain incompletely understood. Here, we demonstrate that neuropilin-1 (NRP1), a Semaphorin receptor expressed in BCs, controls both axonal guidance and subcellular target recognition. We show that loss of Semaphorin 3A function or specific deletion of NRP1 in BCs alters the stereotyped organization of BC axon and impairs pinceau synapse formation. Further, we identified NRP1 as a trans-synaptic binding partner of the cell adhesion molecule neurofascin-186 (NF186) expressed in the PC AIS during pinceau synapse formation. These findings identify a dual function of NRP1 in both axon guidance and subcellular target recognition in the construction of GABAergic circuitry.


Subject(s)
Axon Guidance/physiology , Cerebellum/cytology , Cerebellum/growth & development , GABAergic Neurons/physiology , Neuropilin-1/physiology , Animals , CHO Cells , Cell Adhesion Molecules/metabolism , Coculture Techniques , Cricetulus , Humans , Nerve Growth Factors/metabolism , Neurogenesis/physiology , Purkinje Cells/physiology , Semaphorin-3A/physiology , Synapses/physiology
15.
Science ; 351(6280): 1443-6, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26940868

ABSTRACT

During corticogenesis, excitatory neurons are born from progenitors located in the ventricular zone (VZ), from where they migrate to assemble into circuits. How neuronal identity is dynamically specified upon progenitor division is unknown. Here, we study this process using a high-temporal-resolution technology allowing fluorescent tagging of isochronic cohorts of newborn VZ cells. By combining this in vivo approach with single-cell transcriptomics in mice, we identify and functionally characterize neuron-specific primordial transcriptional programs as they dynamically unfold. Our results reveal early transcriptional waves that instruct the sequence and pace of neuronal differentiation events, guiding newborn neurons toward their final fate, and contribute to a road map for the reverse engineering of specific classes of cortical neurons from undifferentiated cells.


Subject(s)
Neocortex/embryology , Neurogenesis/genetics , Neurons/cytology , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cerebral Ventricles/cytology , Cerebral Ventricles/embryology , DNA-Binding Proteins/genetics , Female , GPI-Linked Proteins/genetics , Green Fluorescent Proteins/genetics , Male , Mice , Neocortex/cytology , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neuropeptides/genetics , SOXB1 Transcription Factors/genetics , T-Box Domain Proteins , Transcriptome
16.
Nature ; 511(7510): 471-4, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24828045

ABSTRACT

During development, thalamocortical (TC) input has a critical role in the spatial delineation and patterning of cortical areas, yet the underlying cellular and molecular mechanisms that drive cortical neuron differentiation are poorly understood. In the primary (S1) and secondary (S2) somatosensory cortex, layer 4 (L4) neurons receive mutually exclusive input originating from two thalamic nuclei: the ventrobasalis (VB), which conveys tactile input, and the posterior nucleus (Po), which conveys modulatory and nociceptive input. Recently, we have shown that L4 neuron identity is not fully committed postnatally, implying a capacity for TC input to influence differentiation during cortical circuit assembly. Here we investigate whether the cell-type-specific molecular and functional identity of L4 neurons is instructed by the origin of their TC input. Genetic ablation of the VB at birth resulted in an anatomical and functional rewiring of Po projections onto L4 neurons in S1. This induced acquisition of Po input led to a respecification of postsynaptic L4 neurons, which developed functional molecular features of Po-target neurons while repressing VB-target traits. Respecified L4 neurons were able to respond both to touch and to noxious stimuli, in sharp contrast to the normal segregation of these sensory modalities in distinct cortical circuits. These findings reveal a behaviourally relevant TC-input-type-specific control over the molecular and functional differentiation of postsynaptic L4 neurons and cognate intracortical circuits, which instructs the development of modality-specific neuronal and circuit properties during corticogenesis.


Subject(s)
Cell Differentiation , Neural Pathways/physiology , Neurons/cytology , Neurons/physiology , Post-Synaptic Density/physiology , Somatosensory Cortex/physiology , Thalamic Nuclei/physiology , Animals , Axons/drug effects , Axons/physiology , Capsaicin/pharmacology , Cell Differentiation/drug effects , Female , Male , Mice, Inbred C57BL , Neural Pathways/drug effects , Neurons/drug effects , Noxae/pharmacology , Optogenetics , Post-Synaptic Density/drug effects , Somatosensory Cortex/cytology , Somatosensory Cortex/drug effects , Synaptic Potentials/drug effects , Thalamic Nuclei/cytology , Thalamic Nuclei/drug effects , Touch/physiology , Vibrissae/drug effects , Vibrissae/physiology
17.
Curr Biol ; 23(10): 850-61, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23602477

ABSTRACT

BACKGROUND: GABAergic interneurons regulate the balance and dynamics of neural circuits, in part, by elaborating their strategically placed axon branches that innervate specific cellular and subcellular targets. However, the molecular mechanisms that regulate target-directed GABAergic axon branching are not well understood. RESULTS: Here we show that the secreted axon guidance molecule, SEMA3A, expressed locally by Purkinje cells, regulates cerebellar basket cell axon branching through its cognate receptor Neuropilin-1 (NRP1). SEMA3A was specifically localized and enriched in the Purkinje cell layer (PCL). In sema3A(-/-) and nrp1(sema-/sema-) mice lacking SEMA3A-binding domains, basket axon branching in PCL was reduced. We demonstrate that SEMA3A-induced axon branching was dependent on local recruitment of soluble guanylyl cyclase (sGC) to the plasma membrane of basket cells, and sGC subcellular trafficking was regulated by the Src kinase FYN. In fyn-deficient mice, basket axon terminal branching was reduced in PCL, but not in the molecular layer. CONCLUSIONS: These results demonstrate a critical role of local SEMA3A signaling in layer-specific axonal branching, which contributes to target innervation.


Subject(s)
Cerebellum/cytology , Interneurons/cytology , Semaphorin-3A/metabolism , Signal Transduction , Animals , Axons , Cerebellum/metabolism , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Mice , Mice, Knockout , Protein Transport , gamma-Aminobutyric Acid/metabolism
18.
J Biol Chem ; 283(11): 6799-805, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18182392

ABSTRACT

Functional interplay between ionotropic and metabotropic receptors frequently involves complex intracellular signaling cascades. The group I metabotropic glutamate receptor mGlu5a co-clusters with the ionotropic N-methyl-d-aspartate (NMDA) receptor in hippocampal neurons. In this study, we report that a more direct cross-talk can exist between these types of receptors. Using bioluminescence resonance energy transfer in living HEK293 cells, we demonstrate that mGlu5a and NMDA receptor clustering reflects the existence of direct physical interactions. Consequently, the mGlu5a receptor decreased NMDA receptor current, and reciprocally, the NMDA receptor strongly reduced the ability of the mGlu5a receptor to release intracellular calcium. We show that deletion of the C terminus of the mGlu5a receptor abolished both its interaction with the NMDA receptor and reciprocal inhibition of the receptors. This direct functional interaction implies a higher degree of target-effector specificity, timing, and subcellular localization of signaling than could ever be predicted with complex signaling pathways.


Subject(s)
Gene Expression Regulation , Receptors, Kainic Acid/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Calcium/metabolism , Cell Line , Energy Transfer , GTP-Binding Proteins/metabolism , Hippocampus/metabolism , Humans , Luminescent Proteins/chemistry , Models, Biological , Protein Structure, Tertiary , Receptor, Metabotropic Glutamate 5 , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...