首页 | 本学科首页   官方微博 | 高级检索  
检索        


NMDA receptor–BK channel coupling regulates synaptic plasticity in the barrel cortex
Authors:Ricardo Gmez  Laura E Maglio  Alberto J Gonzalez-Hernandez  Belinda Rivero-Prez  David Bartolom-Martín  Teresa Giraldez
Institution:aDepartamento de Ciencias Médicas Básicas-Fisiología, Facultad de Medicina, Universidad de La Laguna, 38200 Tenerife, Spain;bInstituto de Tecnologías Biomédicas, Universidad de La Laguna, 38200 Tenerife, Spain
Abstract:Postsynaptic N-methyl-D-aspartate receptors (NMDARs) are crucial mediators of synaptic plasticity due to their ability to act as coincidence detectors of presynaptic and postsynaptic neuronal activity. However, NMDARs exist within the molecular context of a variety of postsynaptic signaling proteins, which can fine-tune their function. Here, we describe a form of NMDAR suppression by large-conductance Ca2+- and voltage-gated K+ (BK) channels in the basal dendrites of a subset of barrel cortex layer 5 pyramidal neurons. We show that NMDAR activation increases intracellular Ca2+ in the vicinity of BK channels, thus activating K+ efflux and strong negative feedback inhibition. We further show that neurons exhibiting such NMDAR–BK coupling serve as high-pass filters for incoming synaptic inputs, precluding the induction of spike timing–dependent plasticity. Together, these data suggest that NMDAR-localized BK channels regulate synaptic integration and provide input-specific synaptic diversity to a thalamocortical circuit.

Glutamate is the primary excitatory chemical transmitter in the mammalian central nervous system (CNS), where it is essential for neuronal viability, network function, and behavioral responses (1). Glutamate activates a variety of pre- and postsynaptic receptors, including ionotropic receptors (iGluRs) that form ligand-gated cation-permeable ion channels. The iGluR superfamily includes α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), kainate receptors, and N-methyl-D-aspartate receptors (NMDARs), all of which form tetrameric assemblies that are expressed throughout the CNS (2).NMDARs exhibit high sensitivity to glutamate (apparent half maximal effective concentration in the micromolar range) and a voltage-dependent block by Mg2+ (3, 4), slow gating kinetics (5), and high permeability to Ca2+ (6, 7) (for a review, see ref. 8). Together, these characteristics confer postsynaptic NMDARs with the ability to detect and decode coincidental activity of pre- and postsynaptic neurons: presynaptic glutamate release brings about the occupation of the agonist-binding site and AMPAR-driven postsynaptic depolarization, removing the voltage-dependent Mg2+ block. The coincidence of these two events leads to NMDAR activation and a Ca2+ influx through the channel (8, 9), which initiates several forms of synaptic plasticity (10, 11).Large-conductance Ca2+- and voltage-gated K+ (BK) channels are opened by a combination of membrane depolarization and relatively high levels of intracellular Ca2+ (12, 13). In CNS neurons, such micromolar Ca2+ increases are usually restricted to the immediate vicinity of Ca2+ sources, including voltage-gated Ca2+ channels (VGCCs) (1416) and ryanodine receptors (RyRs) (17, 18). In addition, Ca2+ influx through nonselective cation-permeable channels, including NMDARs, has also been shown to activate BK channels in granule cells from the olfactory bulb and dentate gyrus (1921). In these neurons, Ca2+ entry through NMDARs opens BK channels in somatic and perisomatic regions, causing the repolarization of the surrounding plasma membrane and subsequent closure of NMDARs. Because BK channel activation blunts NMDAR-mediated excitatory responses, it provides a negative feedback mechanism that modulates the excitability of these neurons (19, 20). Thus, the same characteristics that make NMDARs key components in excitatory synaptic transmission and plasticity can paradoxically give rise to an inhibitory response when NMDARs are located in the proximity of BK channels. However, it is unclear whether functional NMDAR–BK coupling is relevant at dendrites and dendritic spines.The barrel field area in the primary somatosensory cortex, also known as the barrel cortex (BC), processes information from peripheral sensory receptors for onward transmission to cortical and subcortical brain regions (22, 23). Sensory information is received in the BC from different nuclei of the thalamus. Among these nuclei, the ventral posterior medial nucleus, ventrobasal nucleus, and posterior medial nucleus are known to directly innervate layer 5 pyramidal neurons (BC-L5PNs) (2427). In basal dendrites of BC-L5PN, the coactivation of neighboring dendritic inputs can initiate NMDAR-mediated dendritically restricted spikes characterized by large Ca2+ transients and long-lasting depolarizations (2830), providing the appropriate environment for BK activation.To determine whether functional NMDAR–BK coupling plays a role in synaptic transmission, and potentially synaptic plasticity, we investigated the thalamocortical synapses at basal dendrites of BC-L5PNs. We found that the suppression of NMDAR activity by BK channels occurs in the basal dendrites of about 40% of BC-L5PNs, where NMDAR activation triggers strong negative feedback inhibition by delivering Ca2+ to nearby BK channels. This inhibition regulates the amplitude of postsynaptic responses and increases the threshold for the induction of synaptic plasticity. Our findings thus unveil a calibration mechanism that can decode the amount and frequency of afferent synaptic inputs by selectively attenuating synaptic plasticity and providing input-specific synaptic diversity to a thalamocortical circuit.
Keywords:large-conductance calcium- and voltage-activated potassium channels  functional coupling  ion channel macromolecular complexes  synaptic plasticity
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号