Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Neurophysiol ; 131(4): 768-777, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38380828

ABSTRACT

NMDA-type glutamate receptors (NMDARs) play a crucial role in synaptogenesis, circuit development, and synaptic plasticity, serving as fundamental components in cellular models of learning and memory. Their dysregulation has been implicated in several neurological disorders and synaptopathies. NMDARs are heterotetrameric complexes composed of two GluN1 and two GluN2 subunits. The composition of GluN2 subunits determines the main biophysical properties of the channel, such as calcium permeability and gating kinetics, and influences the ability of the receptor to interact with postsynaptic proteins involved in normal synaptic physiology and plasticity, including scaffolding proteins and signaling molecules. During early development, NMDARs in the forebrain contain solely the GluN2B subunit, a necessary subunit for proper synaptogenesis and synaptic plasticity. As the animal matures, the expression of the GluN2A subunit increases, leading to a partial replacement of GluN2B-containing synaptic NMDARs with GluN2A-containing receptors. The switch in the synaptic GluN2A-to-GluN2B ratio has a significant impact on the kinetics of excitatory postsynaptic currents and diminishes the synaptic plasticity capacity. In this study, we present findings indicating that GluN2A expression occurs earlier in a mouse model of fragile X syndrome (FXS). This altered timing of GluN2A expression affects various important parameters of NMDAR-mediated excitatory postsynaptic currents, including maximal current amplitude, decay time, and response to consecutive stimuli delivered in close temporal proximity. These observations suggest that the early expression of GluN2A during a critical period when synapses and circuits are developing could be an underlying factor contributing to the formation of pathological circuits in the FXS mouse model.NEW & NOTEWORTHY NMDA receptors (NMDARs) play important roles in synaptic transmission and are involved in multiple neurological disorders. During development, GluN2A in the forebrain becomes incorporated into previously GluN2B-dominated NMDARs, leading to the "GluN2A/GluN2B ratio switch." This is a crucial step for normal brain development. Here we present findings indicating that GluN2A expression occurs earlier in the fragile X mouse and this could be an underlying factor contributing to the pathology found in the fragile X model.


Subject(s)
Fragile X Syndrome , Receptors, N-Methyl-D-Aspartate , Mice , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Synaptic Transmission , Neuronal Plasticity
2.
J Biol Chem ; 299(11): 105350, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37832874

ABSTRACT

Wnt signaling plays a key role in the mature CNS by regulating trafficking of NMDA-type glutamate receptors and intrinsic properties of neurons. The Wnt receptor ROR2 has been identified as a necessary component of the neuronal Wnt5a/Ca2+ signaling pathway that regulates synaptic and neuronal function. Since ROR2 is considered a pseudokinase, its mechanism for downstream signaling upon ligand binding has been controversial. It has been suggested that its role is to function as a coreceptor of a G-protein-coupled Wnt receptor of the Frizzled family. We show that chemically induced homodimerization of ROR2 is sufficient to recapitulate key signaling events downstream of receptor activation in neurons, including PKC and JNK kinases activation, elevation of somatic and dendritic Ca2+ levels, and increased trafficking of NMDARs to synapses. In addition, we show that homodimerization of ROR2 induces phosphorylation of the receptor on Tyr residues. Point mutations in the conserved but presumed nonfunctional ATP-binding site of the receptor prevent its phosphorylation, as well as downstream signaling. This suggests an active kinase domain. Our results indicate that ROR2 can signal independently of Frizzled receptors to regulate the trafficking of a key synaptic component. Additionally, they suggest that homodimerization can overcome structural conformations that render the tyrosine kinase inactive. A better understanding of ROR2 signaling is crucial for comprehending the regulation of synaptic and neuronal function in normal brain processes in mature animals.


Subject(s)
Receptor Tyrosine Kinase-like Orphan Receptors , Wnt Signaling Pathway , Animals , Calcium/metabolism , Calcium Signaling , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Neurons/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein/metabolism , Dimerization
3.
Neurochem Res ; 48(10): 3212-3227, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37402036

ABSTRACT

Astrocytes release numerous factors known to contribute to the process of synaptogenesis, yet knowledge about the signals that control their release is limited. We hypothesized that neuron-derived signals stimulate astrocytes, which respond to neurons through the modulation of astrocyte-released synaptogenic factors. Here we investigate the effect of cholinergic stimulation of astrocytes on synaptogenesis in co-cultured neurons. Using a culture system where primary rat astrocytes and primary rat neurons are first grown separately allowed us to independently manipulate astrocyte cholinergic signaling. Subsequent co-culture of pre-stimulated astrocytes with naïve neurons enabled us to assess how prior stimulation of astrocyte acetylcholine receptors uniquely modulates neuronal synapse formation. Pre-treatment of astrocytes with the acetylcholine receptor agonist carbachol increased the expression of synaptic proteins, the number of pre- and postsynaptic puncta, and the number of functional synapses in hippocampal neurons after 24 h in co-culture. Astrocyte secretion of the synaptogenic protein thrombospondin-1 increased after cholinergic stimulation and inhibition of the receptor for thrombospondins prevented the increase in neuronal synaptic structures. Thus, we identified a novel mechanism of neuron-astrocyte-neuron communication, where neuronal release of acetylcholine stimulates astrocytes to release synaptogenic proteins leading to increased synaptogenesis in neurons. This study provides new insights into the role of neurotransmitter receptors in developing astrocytes and into our understanding of the modulation of astrocyte-induced synaptogenesis.


Subject(s)
Astrocytes , Synapses , Rats , Animals , Astrocytes/metabolism , Synapses/metabolism , Neurons/metabolism , Coculture Techniques , Cholinergic Agents/pharmacology , Cholinergic Agents/metabolism
4.
Res Sq ; 2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36824819

ABSTRACT

Astrocytes release numerous factors known to contribute to the process of synaptogenesis, yet knowledge about the signals that control their release is limited. We hypothesized that neuron-derived signals stimulate astrocytes, which respond by signaling back to neurons through the modulation of astrocyte-released synaptogenic factors. Here we investigate the effect of cholinergic stimulation of astrocytes on synaptogenesis in co-cultured neurons. Using a culture system where primary rat astrocytes and primary rat neurons are first grown separately allowed us to independently manipulate astrocyte cholinergic signaling. Subsequent co-culture of pre-stimulated astrocytes with naïve neurons enabled us to assess how prior stimulation of astrocyte acetylcholine receptors uniquely modulates neuronal synapse formation. Pre-treatment of astrocytes with the acetylcholine receptor agonist carbachol increased the expression of synaptic proteins, the number of pre- and postsynaptic puncta, and the number of functional synapses in hippocampal neurons after 24 hours in co-culture. Astrocyte secretion of the synaptogenic protein thrombospondin-1 increased after cholinergic stimulation and the inhibition of the target receptor for thrombospondins prevented the observed increase in neuronal synaptic structures. Thus, we identified a novel mechanism of neuron-astrocyte-neuron communication, i.e. , neuronal release of acetylcholine stimulates astrocytes to release synaptogenic proteins leading to increased synaptogenesis in neurons. This study provides new insights into the role of neurotransmitter receptors in developing astrocytes and into our understanding of the modulation of astrocyte-induced synaptogenesis.

5.
Nutr Hosp ; 39(6): 1389-1396, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36327126

ABSTRACT

Introduction: Irisin is an adipomyokine involved in white adipose tissue browning, therefore, could be a key protein in metabolic health. However, exercise effects on irisin in subjects with overweight and/or obesity are conflicting. Therefore, this systematic review aims to search and analyse the literature available on this topic. From three databases: PubMed, ScienceDirect, and Medline, clinical studies published between 2010 and 2021 were considered. From 134 found, 14 studies were included. Only six reported plasma increases after exercise (~1.2 to 3-fold from pre-exercise levels). In addition, only 1 reported significant increases in skeletal muscle irisin mRNA levels (~2-fold). Also, irisin was measured from subcutaneous adipose tissue and saliva, where a ~2-fold increase in its protein levels was found in the latter. Exercise seems to increase the circulatory concentrations of irisin in subjects with overweight or obesity. However, this response is highly variable, therefore, a more integrative approach is urgently needed.


Introducción: La irisina es una adipomioquina relacionada a la transformación del tejido adiposo blanco a marrón, por tanto, podría ser una proteína clave para la salud metabólica. Sin embargo, los efectos del ejercicio sobre la irisina en personas con sobrepeso u obesidad son poco claros. Por lo anterior, esta revisión sistemática apunta a buscar y analizar la literatura disponible en este tema. Desde tres bases de datos: PubMed, ScienceDirect y Medline se buscaron estudios clínicos publicados entre el 2010 y 2021. De 134 estudios encontrados, 14 fueron incluidos. Solo 6 reportaron incrementos plasmáticos de irisina después del ejercicio (~1.2 a 3-veces respecto a niveles preejercicio). Además, solo 1 estudio describió incrementos significativos en el ARNm de irisina en el músculo esquelético (~2 veces sobre niveles preejercicio). La irisina también se medió desde tejido adiposo subcutáneo y saliva, encontrándose una elevación de (~2 veces sobre niveles preejercicio) en esta última. El ejercicio físico incrementaría las concentraciones circulatorias de irisina en personas con sobrepeso u obesidad. Sin embargo, esta respuesta es muy variable, por lo que se requiere una mirada más integrativa a la hora de estudiar este fenómeno.


Subject(s)
Exercise Therapy , Exercise , Fibronectins , Overweight , Humans , Exercise/physiology , Muscle, Skeletal/metabolism , Obesity/therapy , Overweight/metabolism
6.
Nutr. hosp ; 39(6): 1389-1396, nov.-dic. 2022. ilus, tab
Article in English | IBECS | ID: ibc-214848

ABSTRACT

Irisin is an adipomyokine involved in white adipose tissue browning, therefore, could be a key protein in metabolic health. However, exercise effects on irisin in subjects with overweight and/or obesity are conflicting. Therefore, this systematic review aims to search and analyse the literature available on this topic. From three databases: PubMed, ScienceDirect, and Medline, clinical studies published between 2010 and 2021 were considered. From 134 found, 14 studies were included. Only six reported plasma increases after exercise (~1.2 to 3-fold from pre-exercise levels). In addition, only 1 reported significant increases in skeletal muscle irisin mRNA levels (~2-fold). Also, irisin was measured from subcutaneous adipose tissue and saliva, where a ~2-fold increase in its protein levels was found in the latter. Exercise seems to increase the circulatory concentrations of irisin in subjects with overweight or obesity. However, this response is highly variable, therefore, a more integrative approach is urgently needed. (AU)


La irisina es una adipomioquina relacionada a la transformación del tejido adiposo blanco a marrón, por tanto, podría ser una proteína clave para la salud metabólica. Sin embargo, los efectos del ejercicio sobre la irisina en personas con sobrepeso u obesidad son poco claros. Por lo anterior, esta revisión sistemática apunta a buscar y analizar la literatura disponible en este tema. Desde tres bases de datos: PubMed, ScienceDirect y Medline se buscaron estudios clínicos publicados entre el 2010 y 2021. De 134 estudios encontrados, 14 fueron incluidos. Solo 6 reportaron incrementos plasmáticos de irisina después del ejercicio (~1.2 a 3-veces respecto a niveles preejercicio). Además, solo 1 estudio describió incrementos significativos en el ARNm de irisina en el músculo esquelético (~2 veces sobre niveles preejercicio). La irisina también se medió desde tejido adiposo subcutáneo y saliva, encontrándose una elevación de (~2 veces sobre niveles preejercicio) en esta última. El ejercicio físico incrementaría las concentraciones circulatorias de irisina en personas con sobrepeso u obesidad. Sin embargo, esta respuesta es muy variable, por lo que se requiere una mirada más integrativa a la hora de estudiar este fenómeno. (AU)


Subject(s)
Humans , Exercise Therapy , Overweight/metabolism , Obesity/therapy , Fibronectins , Muscle, Skeletal/metabolism
7.
Int J Mol Sci ; 23(12)2022 Jun 07.
Article in English | MEDLINE | ID: mdl-35742824

ABSTRACT

Both hypothalamic microglial inflammation and melanocortin pathway dysfunction contribute to diet-induced obesity (DIO) pathogenesis. Previous studies involving models of altered microglial signaling demonstrate altered DIO susceptibility with corresponding POMC neuron cytological changes, suggesting a link between microglia and the melanocortin system. We addressed this hypothesis using the specific microglial silencing molecule, CX3CL1 (fractalkine), to determine whether reducing hypothalamic microglial activation can restore POMC/melanocortin signaling to protect against DIO. We performed metabolic analyses in high fat diet (HFD)-fed mice with targeted viral overexpression of CX3CL1 in the hypothalamus. Electrophysiologic recording in hypothalamic slices from POMC-MAPT-GFP mice was used to determine the effects of HFD feeding and microglial silencing via minocycline or CX3CL1 on GFP-labeled POMC neurons. Finally, mice with hypothalamic overexpression of CX3CL1 received central treatment with the melanocortin receptor antagonist SHU9119 to determine whether melanocortin signaling is required for the metabolic benefits of CX3CL1. Hypothalamic overexpression of CX3CL1 increased leptin sensitivity and POMC gene expression, while reducing weight gain in animals fed an HFD. In electrophysiological recordings from hypothalamic slice preparations, HFD feeding was associated with reduced POMC neuron excitability and increased amplitude of inhibitory postsynaptic currents. Microglial silencing using minocycline or CX3CL1 treatment reversed these HFD-induced changes in POMC neuron electrophysiologic properties. Correspondingly, blockade of melanocortin receptor signaling in vivo prevented both the acute and chronic reduction in food intake and body weight mediated by CX3CL1. Our results show that suppressing microglial activation during HFD feeding reduces DIO susceptibility via a mechanism involving increased POMC neuron excitability and melanocortin signaling.


Subject(s)
Diet, High-Fat , Melanocortins , Animals , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Melanocortins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Minocycline/pharmacology , Neurons/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
8.
J Cell Sci ; 135(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-34982154

ABSTRACT

Phosphatidylinositol(4,5)-bisphosphate (PtdInsP2) is an important modulator of many cellular processes, and its abundance in the plasma membrane is closely regulated. We examined the hypothesis that members of the Dishevelled scaffolding protein family can bind the lipid kinases phosphatidylinositol 4-kinase (PI4K) and phosphatidylinositol 4-phosphate 5-kinase (PIP5K), facilitating synthesis of PtdInsP2 directly from phosphatidylinositol. We used several assays for PtdInsP2 to examine the cooperative function of phosphoinositide kinases and the Dishevelled protein Dvl3 in the context of two receptor signaling cascades. Simultaneous overexpression of PI4KIIIα (also known as PI4KA) and PIP5KIγ (also known as PIP5K1C) had a synergistic effect on PtdInsP2 synthesis that was recapitulated by overexpression of Dvl3. Increasing the activity of Dvl3 by overexpression increased resting plasma membrane PtdInsP2. Knockdown of Dvl3 reduced resting plasma membrane PtdInsP2 and slowed PtdInsP2 resynthesis following receptor activation. We confirm that Dvl3 promotes coupling of PI4KIIIα and PIP5KIγ and show that this interaction is essential for efficient resynthesis of PtdInsP2 following receptor activation.


Subject(s)
1-Phosphatidylinositol 4-Kinase , Phosphatidylinositol 4,5-Diphosphate , 1-Phosphatidylinositol 4-Kinase/metabolism , Cell Membrane/metabolism , Dishevelled Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositols/metabolism , Phosphotransferases (Alcohol Group Acceptor)
9.
Front Synaptic Neurosci ; 12: 588295, 2020.
Article in English | MEDLINE | ID: mdl-33343326

ABSTRACT

AMPA-type glutamate receptors (AMPARs) are tetrameric ligand-gated channels made up of combinations of GluA1-4 subunits and play important roles in synaptic transmission and plasticity. Here, we have investigated the development of AMPAR-mediated synaptic transmission in the hippocampus of the Fmr1 knock-out (KO) mouse, a widely used model of Fragile X syndrome (FXS). FXS is the leading monogenic cause of intellectual disability and autism spectrum disorders (ASD) and it is considered a neurodevelopmental disorder. For that reason, we investigated synaptic properties and dendritic development in animals from an early stage when synapses are starting to form up to adulthood. We found that hippocampal CA1 pyramidal neurons in the Fmr1-KO mouse exhibit a higher AMPAR-NMDAR ratio early in development but reverses to normal values after P13. This increase was accompanied by a larger presence of the GluA2-subunit in synaptic AMPARs that will lead to altered Ca2+ permeability of AMPARs that could have a profound impact upon neural circuits, learning, and diseases. Following this, we found that young KO animals lack Long-term potentiation (LTP), a well-understood model of synaptic plasticity necessary for proper development of circuits, and exhibit an increased frequency of spontaneous miniature excitatory postsynaptic currents, a measure of synaptic density. Furthermore, post hoc morphological analysis of recorded neurons revealed altered dendritic branching in the KO group. Interestingly, all these anomalies are transitory and revert to normal values in older animals. Our data suggest that loss of FMRP during early development leads to temporary upregulation of the GluA2 subunit and this impacts synaptic plasticity and altering morphological dendritic branching.

10.
J Neurophysiol ; 123(3): 1004-1014, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31995440

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) are fundamental coincidence detectors of synaptic activity necessary for the induction of synaptic plasticity and synapse stability. Adjusting NMDAR synaptic content, whether by receptor insertion or lateral diffusion between extrasynaptic and synaptic compartments, could play a substantial role defining the characteristics of the NMDAR-mediated excitatory postsynaptic current (EPSC), which in turn would mediate the ability of the synapse to undergo plasticity. Lateral NMDAR movement has been observed in dissociated neurons; however, it is currently unclear whether NMDARs are capable of lateral surface diffusion in hippocampal slices, a more physiologically relevant environment. To test for lateral mobility in rat hippocampal slices, we rapidly blocked synaptic NMDARs using MK-801, a use-dependent and irreversible NMDAR blocker. Following a 5-min washout period, we observed a strong recovery of NMDAR-mediated responses. The degree of the observed recovery was proportional to the amount of induced blockade, independent of levels of intracellular calcium, and mediated primarily by GluN2B-containing NMDA receptors. These results indicate that lateral diffusion of NMDARs could be a mechanism by which synapses rapidly adjust parameters to fine-tune synaptic plasticity.NEW & NOTEWORTHYN-methyl-d-aspartate-type glutamate receptors (NMDARs) have always been considered stable components of synapses. We show that in rat hippocampal slices synaptic NMDARs are in constant exchange with extrasynaptic receptors. This exchange of receptors is mediated primarily by NMDA receptors containing GluN2B, a subunit necessary to undergo synaptic plasticity. Thus this lateral movement of synaptic receptors allows synapses to rapidly regulate the total number of synaptic NMDARs with potential consequences for synaptic plasticity.


Subject(s)
CA1 Region, Hippocampal/metabolism , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , Animals , CA1 Region, Hippocampal/drug effects , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptic Transmission/drug effects
11.
Nature ; 573(7775): 499-501, 2019 09.
Article in English | MEDLINE | ID: mdl-31551543
12.
Methods Mol Biol ; 1941: 57-64, 2019.
Article in English | MEDLINE | ID: mdl-30707427

ABSTRACT

Organotypic slice cultures enable the study of glutamate receptors in an environment closely related to the in vivo situation but with easy access to genetic manipulation of the receptors and regulatory signaling cascades as well as a more precise pharmacological intervention. We describe a method to prepare organotypic hippocampal slices that can be easily adapted to other brain regions. Brain slices are laid on porous membranes, and culture medium is allowed to form an interface. This method preserves the functionality and gross architecture of the hippocampus for up to 2 weeks in culture.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Organ Culture Techniques/methods , Receptors, Glutamate/metabolism , Animals
13.
Cell Rep ; 21(1): 60-69, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978484

ABSTRACT

Wnt signaling controls multiple biological process, particularly the embryonic development of metazoans. Sustained expression of Wnt signaling components in the mature mammalian CNS and their apparent deregulation in certain neuropathologies suggest that it also plays a part beyond embryonic development to regulate normal brain function. We describe a noncanonical Wnt/Ca2+ signaling cascade that regulates the electrophysiological intrinsic properties of rat neurons, resulting in sustained membrane depolarization and the mobilization of Ca2+ from internal stores. These effects require tyrosine kinase-like orphan receptor 2 (RoR2), activation of PLC, and voltage-gated Ca2+ channels. Activation of this signaling cascade then promotes surface expression of N-methyl-D-aspartate receptors (NMDARs) through a SNARE-dependent mechanism. This neuronal Wnt/Ca2+ signaling pathway represents a mechanism for Wnt ligands to regulate normal brain processes in the mature animal and provides a framework for understanding how alterations in this pathway may contribute to the etiology of psychiatric disorders where NMDARs are compromised.


Subject(s)
Calcium Signaling , Hippocampus/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Wnt Signaling Pathway , Action Potentials/physiology , Animals , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Female , Gene Expression Regulation , HEK293 Cells , Hippocampus/cytology , Humans , Male , Microtomy , Neurons/cytology , Patch-Clamp Techniques , Protein Transport , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptor Tyrosine Kinase-like Orphan Receptors/deficiency , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Tissue Culture Techniques , Type C Phospholipases/genetics , Type C Phospholipases/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism
14.
Proc Natl Acad Sci U S A ; 112(15): 4797-802, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25825749

ABSTRACT

Wnt signaling has a well-established role as a regulator of nervous system development, but its role in the maintenance and regulation of established synapses in the mature brain remains poorly understood. At excitatory glutamatergic synapses, NMDA receptors (NMDARs) have a fundamental role in synaptogenesis, synaptic plasticity, and learning and memory; however, it is not known what controls their number and subunit composition. Here we show that the receptor tyrosine kinase-like orphan receptor 2 (RoR2) functions as a Wnt receptor required to maintain basal NMDAR-mediated synaptic transmission. In addition, RoR2 activation by a noncanonical Wnt ligand activates PKC and JNK and acutely enhances NMDAR synaptic responses. Regulation of a key component of glutamatergic synapses through RoR2 provides a mechanism for Wnt signaling to modulate synaptic transmission, synaptic plasticity, and brain function acutely beyond embryonic development.


Subject(s)
Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission , Wnt Proteins/metabolism , Action Potentials/physiology , Animals , Cells, Cultured , Gene Expression , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , In Situ Hybridization , JNK Mitogen-Activated Protein Kinases/metabolism , Microscopy, Confocal , Patch-Clamp Techniques , Protein Kinase C/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , RNA Interference , Rats , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Wnt Proteins/genetics , Wnt-5a Protein
15.
J Neurosci ; 34(14): 4914-9, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24695710

ABSTRACT

Information processing in the brain relies on precise timing of signal propagation. The highly conserved neuronal network for computing spatial representations of acoustic signals resolves microsecond timing of sounds processed by the two ears. As such, it provides an excellent model for understanding how precise temporal regulation of neuronal signals is achieved and maintained. The well described avian and mammalian brainstem circuit for computation of interaural time differences is composed of monaural cells in the cochlear nucleus (CN; nucleus magnocellularis in birds) projecting to binaurally innervated coincidence detection neurons in the medial superior olivary nucleus (MSO) in mammals or nucleus laminaris (NL) in birds. Individual axons from CN neurons issue a single axon that bifurcates into an ipsilateral branch and a contralateral branch that innervate segregated dendritic regions of the MSO/NL coincidence detector neurons. We measured conduction velocities of the ipsilateral and contralateral branches of these bifurcating axon collaterals in the chicken by antidromic stimulation of two sites along each branch and whole-cell recordings in the parent neurons. At the end of each experiment, the individual CN neuron and its axon collaterals were filled with dye. We show that the two collaterals of a single axon adjust the conduction velocities individually to achieve the specific conduction velocities essential for precise temporal integration of information from the two ears, as required for sound localization. More generally, these results suggest that individual axonal segments in the CNS interact locally with surrounding neural structures to determine conduction velocity.


Subject(s)
Brain Stem/cytology , Functional Laterality/physiology , Neural Conduction/physiology , Neural Pathways/physiology , Neurons/physiology , Animals , Axons/physiology , Chick Embryo , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Antagonists/pharmacology , Imaging, Three-Dimensional , In Vitro Techniques , Male , Models, Neurological , Neural Conduction/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Picrotoxin/pharmacology , Quinoxalines/pharmacology , Valine/analogs & derivatives , Valine/pharmacology
16.
PLoS One ; 7(11): e49089, 2012.
Article in English | MEDLINE | ID: mdl-23166606

ABSTRACT

Palmitoylation of NMDARs occurs at two distinct cysteine clusters in the carboxyl-terminus of GluN2A and GluN2B subunits that differentially regulates retention in the Golgi apparatus and surface expression of NMDARs. Mutations of palmitoylatable cysteine residues in the membrane-proximal cluster to non-palmitoylatable serines leads to a reduction in the surface expression of recombinant NMDARs via enhanced internalization of the receptors. Mutations in a cluster of cysteines in the middle of the carboxyl-terminus of GluN2A and GluN2B, leads to an increase in the surface expression of NMDARs via an increase in post-Golgi trafficking. Using a quantitative electrophysiological assay, we investigated whether palmitoylation of GluN2 subunits and the differential regulation of surface expression affect functional synaptic incorporation of NMDARs. We show that a reduction in surface expression due to mutations in the membrane-proximal cluster translates to a reduction in synaptic expression of NMDARs. However, increased surface expression induced by mutations in the cluster of cysteines that regulates post-Golgi trafficking of NMDARs does not increase the synaptic pool of NMDA receptors, indicating that the number of synaptic receptors is tightly regulated.


Subject(s)
Cell Membrane/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Cysteine/metabolism , Electric Stimulation , Golgi Apparatus/metabolism , Hippocampus/metabolism , Lipoylation , Mutation/genetics , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics
17.
J Physiol ; 590(19): 4801-18, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22826130

ABSTRACT

Precise control of neuronal excitability in the auditory brainstem is fundamental for processing timing cues used for sound localization and signal discrimination in complex acoustic environments. In mature nucleus laminaris (NL), the first nucleus responsible for binaural processing in chickens, neuronal excitability is governed primarily by voltage-activated potassium conductances (K(VA)). High levels of K(VA) expression in NL neurons result in one or two initial action potentials (APs) in response to high-frequency synaptic activity or sustained depolarization. Here we show that during a period of synaptogenesis and circuit refinement, before hearing onset, K(VA) conductances are relatively small, in particular low-voltage-activated K(+) conductances (K(LVA)). In spite of this, neuronal output is filtered and repetitive synaptic activity generates only one or two initial APs during a train of stimuli. During this early developmental time period, synaptic NMDA-type glutamate receptors (NMDA-Rs) contain primarily the GluN2B subunit. We show that the slow decay kinetics of GluN2B-containing NMDA-Rs allows synaptic responses to summate, filtering the output of NL neurons before intrinsic properties are fully developed. Weaker Mg(2+) blockade of NMDA-Rs and ambient glutamate early in development generate a tonic NMDA-R-mediated current that sets the membrane potential at more depolarized values. Small KLVA conductances, localized in dendrites, prevent excessive depolarization caused by tonic activation of NMDA-Rs. Thus, before intrinsic properties are fully developed, NMDA-Rs control the output of NL neurons during evoked synaptic transmission.


Subject(s)
Brain Stem/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Action Potentials , Animals , Chick Embryo , Excitatory Postsynaptic Potentials , Glutamic Acid/physiology , In Vitro Techniques , Potassium Channels, Voltage-Gated/physiology
18.
J Neurosci ; 31(26): 9466-71, 2011 Jun 29.
Article in English | MEDLINE | ID: mdl-21715611

ABSTRACT

Wnt ligands are secreted glycoproteins controlling gene expression and cytoskeleton reorganization involved in embryonic development of the nervous system. However, their role in later stages of brain development, particularly in the regulation of established synaptic connections, is not known. We found that Wnt-5a acutely and specifically upregulates synaptic NMDAR currents in rat hippocampal slices, facilitating induction of long-term potentiation, a cellular model of learning and memory. This effect requires an increase in postsynaptic Ca(2+) and activation of noncanonical downstream effectors of the Wnt signaling pathway. In contrast, Wnt-7a, an activator of the canonical Wnt signaling pathway, has no effect on NMDAR-mediated synaptic transmission. Moreover, endogenous Wnt ligands are necessary to maintain basal NMDAR synaptic transmission, adjusting the threshold for synaptic potentiation. This novel role for Wnt ligands provides a mechanism for Wnt signaling to acutely modulate synaptic plasticity and brain function in later stages of development and in the mature organism.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology , Synaptic Transmission/physiology , Wnt Proteins/metabolism , Animals , Culture Media, Conditioned , Electric Stimulation , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/drug effects , Hippocampus/physiology , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/drug effects , Wnt Proteins/administration & dosage
19.
J Neurosci ; 31(17): 6311-6, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21525271

ABSTRACT

Synaptic incorporation of NMDA receptors (NMDARs) is regulated by GluN2 subunits with different rules controlling GluN2A- and GluN2B-containing receptors; whereas GluN2B-containing receptors are constitutively incorporated into synapses, GluN2A incorporation is activity-dependent. We expressed electrophysiologically tagged NMDARs in rat hippocampal slices to identify the molecular determinants controlling the mode of synaptic incorporation of NMDARs. Expressing chimeric GluN2 subunits, we identified a putative N-glycosylation site present in GluN2B, but not in GluN2A, as necessary and sufficient to drive NMDARs into synapses in an activity-independent manner. This suggests a novel mechanism for regulating activity-driven changes and trafficking of NMDARs to the synapse.


Subject(s)
Hippocampus/cytology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Animals , Animals, Newborn , Biophysics , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Green Fluorescent Proteins/genetics , Macromolecular Substances/chemistry , Male , Mutation/genetics , Organ Culture Techniques , Patch-Clamp Techniques/methods , Protein Subunits , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Time Factors , Transfection/methods , Valine/analogs & derivatives , Valine/pharmacology
20.
Proc Natl Acad Sci U S A ; 108(14): 5855-60, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21427228

ABSTRACT

During early postnatal development in the rat hippocampus, synaptogenesis occurs in parallel with a developmental switch in the subunit composition of NMDA receptors from NR2B to NR2A. It is unclear how this switch affects the process of synaptogenesis, synapse maturation, and synapse stabilization. We investigated the role of NR2 subunits in synaptogenesis during the period in which expression and synaptic incorporation of the NR2A protein begins through the time when it reaches adult levels. We found that early expression of NR2A in organotypic hippocampal slices reduces the number of synapses and the volume and dynamics of spines. In contrast, overexpression of NR2B does not affect the normal number and growth of synapses; however, it does increase spine motility, adding and retracting spines at a higher rate. The C terminus of NR2B, and specifically its ability to bind CaMKII, is sufficient to allow proper synapse formation and maturation. Conversely, the C terminus of NR2A was sufficient to stop the development of synapse number and spine growth. Our results indicate that the ratio of synaptic NR2B over NR2A controls spine motility and synaptogenesis, and suggest a structural role for the intracellular C terminus of NR2 in recruiting the signaling and scaffolding molecules necessary for proper synaptogenesis.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hippocampus/growth & development , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Microscopy, Fluorescence , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...