Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Sci Adv ; 9(2): eabo7421, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36638182

ABSTRACT

Synapse loss strongly correlates with cognitive decline in Alzheimer's disease (AD), but the underlying mechanisms are poorly understood. Deficient Wnt signaling contributes to synapse dysfunction and loss in AD. Consistently, a variant of the LRP6 receptor, (LRP6-Val), with reduced Wnt signaling, is linked to late-onset AD. However, the impact of LRP6-Val on the healthy and AD brain has not been examined. Knock-in mice, generated by gene editing, carrying this Lrp6 variant develop normally. However, neurons from Lrp6-val mice do not respond to Wnt7a, a ligand that promotes synaptic assembly through the Frizzled-5 receptor. Wnt7a stimulates the formation of the low-density lipoprotein receptor-related protein 6 (LRP6)-Frizzled-5 complex but not if LRP6-Val is present. Lrp6-val mice exhibit structural and functional synaptic defects that become pronounced with age. Lrp6-val mice present exacerbated synapse loss around plaques when crossed to the NL-G-F AD model. Our findings uncover a previously unidentified role for Lrp6-val in synapse vulnerability during aging and AD.


Subject(s)
Alzheimer Disease , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Animals , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Wnt Signaling Pathway , Synapses/metabolism , Aging/genetics
2.
Front Pharmacol ; 13: 970234, 2022.
Article in English | MEDLINE | ID: mdl-36160429

ABSTRACT

The expanding roles of macrophages in physiological and pathophysiological mechanisms now include normal tissue homeostasis, tissue repair and regeneration, including neuronal tissue; initiation, progression, and resolution of the inflammatory response and a diverse array of anti-microbial activities. Two hallmarks of macrophage activity which appear to be fundamental to their diverse cellular functionalities are cellular plasticity and phenotypic heterogeneity. Macrophage plasticity allows these cells to take on a broad spectrum of differing cellular phenotypes in response to local and possibly previous encountered environmental signals. Cellular plasticity also contributes to tissue- and stimulus-dependent macrophage heterogeneity, which manifests itself as different macrophage phenotypes being found at different tissue locations and/or after different cell stimuli. Together, plasticity and heterogeneity align macrophage phenotypes to their required local cellular functions and prevent inappropriate activation of the cell, which could lead to pathology. To execute the appropriate function, which must be regulated at the qualitative, quantitative, spatial and temporal levels, macrophages constantly monitor intracellular and extracellular parameters to initiate and control the appropriate cell signaling cascades. The sensors and signaling mechanisms which control macrophages are the focus of a considerable amount of research. Ion channels regulate the flow of ions between cellular membranes and are critical to cell signaling mechanisms in a variety of cellular functions. It is therefore surprising that the role of ion channels in the macrophage biology has been relatively overlooked. In this review we provide a summary of ion channel research in macrophages. We begin by giving a narrative-based explanation of the membrane potential and its importance in cell biology. We then report on research implicating different ion channel families in macrophage functions. Finally, we highlight some areas of ion channel research in macrophages which need to be addressed, future possible developments in this field and therapeutic potential.

3.
Br J Pharmacol ; 179(20): 4844-4856, 2022 10.
Article in English | MEDLINE | ID: mdl-35817954

ABSTRACT

BACKGROUND AND PURPOSE: Ischaemia is known to cause massive neuronal depolarization, termed anoxic depolarization (AD), due to energy failure and loss of membrane ion gradients. The neuromodulator adenosine accumulates extracellularly during ischaemia and activates four metabotropic receptors: A1 , A2A , A2B and A3 . Striatal medium spiny neurons (MSNs) express high levels of A2A receptors and are particularly vulnerable to ischaemic insults. A2A Receptor blockade reduces acute striatal post-ischaemic damage but the cellular mechanisms involved are still unknown. EXPERIMENTAL APPROACH: We performed patch-clamp recordings of MSNs in rat striatal slices subjected to oxygen and glucose deprivation (OGD) to investigate the effects of A2A receptor ligands or ion channel blockers on AD and OGD-induced ionic imbalance, measured as a positive shift in Erev of ramp currents. KEY RESULTS: Our data indicate that the A2A receptor antagonist SCH58261 (10 µM) significantly attenuated ionic imbalance and AD appearance in MSNs exposed to OGD. The K+ channel blocker Ba2+ (2 mM) or the Na+ channel blocker tetrodotoxin (1 µM) exacerbated and attenuated, respectively, OGD-induced changes. Spontaneous excitatory post-synaptic current (sEPSC) analysis in MSNs revealed that the A2A receptor agonist CGS21680 (1 µM) prevented OGD-induced decrease of sEPSCs within the first 5 min of the insult, an effect shared by the K+ channel blocker Ba2+ , indicating facilitated glutamate release. CONCLUSION AND IMPLICATIONS: Adenosine, released during striatal OGD, activates A2A receptors that may exacerbate OGD-induced damage through K+ channel inhibition. Our results could help to develop A2A receptor-selective therapeutic tools for the treatment of brain ischaemia.


Subject(s)
Glucose , Oxygen , Adenosine/pharmacology , Animals , Glucose/pharmacology , Glutamic Acid/pharmacology , Ion Channels , Ischemia , Ligands , Neurons , Oxygen/metabolism , Rats , Rats, Wistar , Receptor, Adenosine A2A/metabolism , Tetrodotoxin
4.
Front Immunol ; 13: 840069, 2022.
Article in English | MEDLINE | ID: mdl-35757775

ABSTRACT

The traditional view of the nuclear envelope (NE) was that it represented a relatively inert physical barrier within the cell, whose main purpose was to separate the nucleoplasm from the cytoplasm. However, recent research suggests that this is far from the case, with new and important cellular functions being attributed to this organelle. In this review we describe research suggesting an important contribution of the NE and its constituents in regulating the functions of cells of the innate and adaptive immune system. One of the standout properties of immune cells is their ability to migrate around the body, allowing them to carry out their physiological/pathophysiology cellular role at the appropriate location. This together with the physiological role of the tissue, changes in tissue matrix composition due to disease and aging, and the activation status of the immune cell, all result in immune cells being subjected to different mechanical forces. We report research which suggests that the NE may be an important sensor/transducer of these mechanical signals and propose that the NE is an integrator of both mechanical and chemical signals, allowing the cells of the innate immune system to precisely regulate gene transcription and functionality. By presenting this overview we hope to stimulate the interests of researchers into this often-overlooked organelle and propose it should join the ranks of mitochondria and phagosome, which are important organelles contributing to immune cell function.


Subject(s)
Cell Nucleus , Nuclear Envelope , Cell Nucleus/genetics , Cytoplasm
5.
Neuropharmacology ; 202: 108861, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34736958

ABSTRACT

NMDA receptors are one subtype of glutamate receptor that play fundamental roles in synaptic physiology and synaptic plasticity in the nervous system, in addition to being implicated in several neurological disorders. It is now established that many NMDA receptors in the nervous system are triheteromeric, composed of two glycine-binding GluN1 subunits and two different glutamate binding GluN2 subunits. The pharmacology of NMDA receptor has become well established since the pioneering work of Watkins and Evans almost half a century ago and has seen a resurgence of interest in the past decade as new subtype-selective allosteric modulators have been discovered. In this article, features specific to allosteric antagonist action at triheteromeric NMDA receptors are reviewed with a focus on understanding the mechanism of action of drugs acting at triheteromeric GluN1/GluN2B/GluN2D receptors. These receptors are of importance in the basal ganglia and in interneurons of the hippocampus and implications for understanding the action of allosteric antagonists at synaptic triheteromeric receptors are considered.


Subject(s)
Receptors, N-Methyl-D-Aspartate/agonists , Allosteric Regulation , Animals , Basal Ganglia , Binding Sites , Glutamic Acid/metabolism , Glycine/metabolism , Hippocampus , Humans , Interneurons , Nervous System Diseases , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/classification , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Synaptic Transmission/physiology
6.
Neuropharmacology ; 198: 108743, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34363811

ABSTRACT

In 1981 Jeff Watkins and Dick Evans wrote what was to become a seminal review on excitatory amino acids (EAAs) and their receptors (Watkins and Evans, 1981). Bringing together various lines of evidence dating back over several decades on: the distribution in the nervous system of putative amino acid neurotransmitters; enzymes involved in their production and metabolism; the uptake and release of amino acids; binding of EAAs to membranes; the pharmacological action of endogenous excitatory amino acids and their synthetic analogues, and notably the actions of antagonists for the excitations caused by both nerve stimulation and exogenous agonists, often using pharmacological tools developed by Jeff and his colleagues, they provided a compelling account for EAAs, especially l-glutamate, as a bona fide neurotransmitter in the nervous system. The rest, as they say, is history, but far from being consigned to history, EAA research is in rude health well into the 21st Century as this series of Special Issues of Neuropharmacology exemplifies. With EAAs and their receptors flourishing across a wide range of disciplines and clinical conditions, we enter into a dialogue with two of the most prominent and influential figures in the early days of EAA research: Jeff Watkins and Dick Evans.


Subject(s)
Excitatory Amino Acids/physiology , Neurotransmitter Agents/physiology , Receptors, Glutamate/physiology , Animals , Excitatory Amino Acids/pharmacology , Humans , Receptors, Glutamate/drug effects , Synapses/physiology
7.
Front Synaptic Neurosci ; 13: 670467, 2021.
Article in English | MEDLINE | ID: mdl-34149390

ABSTRACT

Synapse degeneration in the striatum has been associated with the early stages of Parkinson's and Huntington's diseases (PD and HD). However, the molecular mechanisms that trigger synaptic dysfunction and loss are not fully understood. Increasing evidence suggests that deficiency in Wnt signaling triggers synapse degeneration in the adult brain and that this pathway is affected in neurodegenerative diseases. Here, we demonstrate that endogenous Wnt signaling is essential for the integrity of a subset of inhibitory synapses on striatal medium spiny neurons (MSNs). We found that inducible expression of the specific Wnt antagonist Dickkopf-1 (Dkk1) in the adult striatum leads to the loss of inhibitory synapses on MSNs and affects the synaptic transmission of D2-MSNs. We also discovered that re-activation of the Wnt pathway by turning off Dkk1 expression after substantial loss of synapses resulted in the complete recovery of GABAergic and dopamine synapse number. Our results also show that re-activation of the Wnt pathway leads to a recovery of amphetamine response and motor function. Our studies identify the Wnt signaling pathway as a potential therapeutic target for restoring neuronal circuits after synapse degeneration.

8.
Pharmacol Rep ; 73(3): 881-890, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33713314

ABSTRACT

BACKGROUND: Macrophages are important cells of the innate immune system and contribute to a variety of physiological and pathophysiological responses. Monovalent and divalent ion channels have been studied in macrophage function, and while much research is still required, a role for these channels is beginning to emerge in macrophages. In addition to the plasma membrane, ion channels are also found in intracellular membranes including mitochondrial, lysosomal and nuclear membranes. While studying the function of plasma membrane located large conductance voltage- and calcium-activated potassium channels (BK channels) in a macrophage cell line RAW264.7, we became aware of the expression of these ion channels in other cellular locations. METHODS: Immunofluorescence and Western blot analysis were used to identify the expression of BK channels. To demonstrate a functional role for the nuclear located channel, we investigated the effect of the lipid soluble BK channel inhibitor paxilline on CREB phosphorylation. RESULTS: Treatment of resting macrophages with paxilline resulted in increased CREB phosphorylation. To confirm a role for nuclear BK channels, these experiments were repeated in isolated nuclei and similar results were found. Ca2+ and calmodulin-dependent kinases (CaMK) have been demonstrated to regulate CREB phosphorylation. Inhibition of CaMKII and CaMKIV resulted in the reversal of paxilline-induced CREB phosphorylation. CONCLUSIONS: These results suggest that nuclear BK channels regulate CREB phosphorylation in macrophages. Nuclear located ion channels may therefore be part of novel signalling pathways in macrophages and should be taken into account when studying the role of ion channels in these and other cells.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Macrophages/metabolism , Phosphorylation/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Indoles/pharmacology , Macrophages/drug effects , Mice , Nuclear Envelope/metabolism , Phosphorylation/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects
9.
Front Synaptic Neurosci ; 12: 575863, 2020.
Article in English | MEDLINE | ID: mdl-33013349

ABSTRACT

Structural plasticity of synapses correlates with changes in synaptic strength. Dynamic modifications in dendritic spine number and size are crucial for long-term potentiation (LTP), the cellular correlate of learning and memory. Recent studies have suggested the generation of multi-innervated spines (MIS), in the form of several excitatory presynaptic inputs onto one spine, are crucial for hippocampal memory storage. However, little is known about the molecular mechanisms underlying MIS formation and their contribution to LTP. Using 3D enhanced resolution confocal images, we examined the contribution of Wnt synaptic modulators in MIS formation in the context of LTP. We show that blockage of endogenous Wnts with specific Wnt antagonists supresses the formation of MIS upon chemical LTP induction in cultured hippocampal neurons. Gain- and loss-of-function studies demonstrate that Wnt7a signaling promotes MIS formation through the postsynaptic Wnt scaffold protein Disheveled 1 (Dvl1) by stimulating neuronal nitric oxide (NO) synthase (nNOS). Subsequently, NO activates soluble guanylyl cyclase (sGC) to increase MIS formation. Consistently, we observed an enhanced frequency and amplitude of excitatory postsynaptic currents. Collectively, our findings identify a unique role for Wnt secreted proteins through nNOS/NO/sGC signaling to modulate MIS formation during LTP.

10.
J Physiol ; 598(15): 3071-3083, 2020 08.
Article in English | MEDLINE | ID: mdl-32468591

ABSTRACT

Glutamate receptors are essential ligand-gated ion channels in the central nervous system that mediate excitatory synaptic transmission in response to the release of glutamate from presynaptic terminals. The structural and biophysical basis underlying the function of these receptors has been studied for decades by a wide range of approaches. However recent structural, pharmacological and genetic studies have provided new insight into the regions of this protein that are critical determinants of receptor function. Lack of variation in specific areas of the protein amino acid sequences in the human population has defined three regions in each receptor subunit that are under selective pressure, which has focused research efforts and driven new hypotheses. In addition, these three closely positioned elements reside near a cavity that is shown by multiple studies to be a likely site of action for allosteric modulators, one of which is currently in use as an FDA-approved anticonvulsant. These structural elements are capable of controlling gating of the pore, and appear to permit some modulators bound within the cavity to also alter permeation properties. This creates a new precedent whereby features of the channel pore can be modulated by exogenous drugs that bind outside the pore. The convergence of structural, genetic, biophysical and pharmacological approaches is a powerful means to gain insight into the complex biological processes defined by neurotransmitter receptor function.


Subject(s)
Awards and Prizes , Ligand-Gated Ion Channels , Biophysical Phenomena , Glutamic Acid , Humans , Receptors, Glutamate
11.
Sci Rep ; 8(1): 15068, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30305717

ABSTRACT

Proteasome inhibitors (PIs) are now standard of care for several cancers, and noninvasive biomarkers of treatment response are critically required for early patient stratification and treatment personalization. The present study evaluated whether chemical exchange (CEST) magnetic resonance imaging (MRI) can provide measurements that can be used as the noninvasive biomarkers of proteasome inhibition, alongside diffusion MRI and relaxometry. The sensitivity of human colorectal carcinoma cells to the PI Ixazomib was assessed via in vitro and in vivo dose-response experiments. Acute in vivo response to Ixazomib was assessed at three dosing concentrations, using CEST MRI (amide, amine, hydroxyl signals), diffusion MRI (ADC) and relaxometry (T1, T2). These responses were further evaluated with the known histological markers for Ixazomib and Bradford assay ex vivo. The CEST signal from amides and amines increased in proportion to Ixazomib dose in colorectal cancer xenografts. The cell lines differed in their sensitivity to Ixazomib, which was reflected in the MRI measurements. A mild stimulation in tumor growth was observed at low Ixazomib doses. Our results identify CEST MRI as a promising method for safely and noninvasively monitoring disrupted tumor protein homeostasis induced by proteasome inhibitor treatment, and for stratifying sensitivity between tumor types.


Subject(s)
Magnetic Resonance Imaging , Proteasome Inhibitors/pharmacology , Proteostasis/drug effects , Amides/analysis , Amines/analysis , Animals , Apoptosis/drug effects , Boron Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/pathology , Diffusion Magnetic Resonance Imaging , Dose-Response Relationship, Drug , Female , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Image Interpretation, Computer-Assisted , Mice, Nude , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
J Gen Physiol ; 150(8): 1081-1105, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30037851

ABSTRACT

NMDA-type glutamate receptors are ligand-gated ion channels that mediate a Ca2+-permeable component of excitatory neurotransmission in the central nervous system (CNS). They are expressed throughout the CNS and play key physiological roles in synaptic function, such as synaptic plasticity, learning, and memory. NMDA receptors are also implicated in the pathophysiology of several CNS disorders and more recently have been identified as a locus for disease-associated genomic variation. NMDA receptors exist as a diverse array of subtypes formed by variation in assembly of seven subunits (GluN1, GluN2A-D, and GluN3A-B) into tetrameric receptor complexes. These NMDA receptor subtypes show unique structural features that account for their distinct functional and pharmacological properties allowing precise tuning of their physiological roles. Here, we review the relationship between NMDA receptor structure and function with an emphasis on emerging atomic resolution structures, which begin to explain unique features of this receptor.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Humans , Ions/metabolism , Protein Structure, Quaternary , Receptors, N-Methyl-D-Aspartate/chemistry
13.
J Physiol ; 596(17): 4057-4089, 2018 09.
Article in English | MEDLINE | ID: mdl-29917241

ABSTRACT

KEY POINTS: The kinetics of NMDA receptor (NMDAR) signalling are a critical aspect of the physiology of excitatory synaptic transmission in the brain. Here we develop a mechanistic description of NMDAR function based on the receptor tetrameric structure and the principle that each agonist-bound subunit must undergo some rate-limiting conformational change after agonist binding, prior to channel opening. By fitting this mechanism to single channel data using a new MATLAB-based software implementation of maximum likelihood fitting with correction for limited time resolution, rate constants were derived for this mechanism that reflect distinct structural changes and predict the properties of macroscopic and synaptic NMDAR currents. The principles applied here to develop a mechanistic description of the heterotetrameric NMDAR, and the software used in this analysis, can be equally applied to other heterotetrameric glutamate receptors, providing a unifying mechanistic framework to understanding the physiology of glutamate receptor signalling in the brain. ABSTRACT: NMDA receptors (NMDARs) are tetrameric complexes comprising two glycine-binding GluN1 and two glutamate-binding GluN2 subunits. Four GluN2 subunits encoded by different genes can produce up to 10 different di- and triheteromeric receptors. In addition, some neurological patients contain a de novo mutation or inherited rare variant in only one subunit. There is currently no mechanistic framework to describe tetrameric receptor function that can be extended to receptors with two different GluN1 or GluN2 subunits. Here we use the structural features of glutamate receptors to develop a mechanism describing both single channel and macroscopic NMDAR currents. We propose that each agonist-bound subunit undergoes some rate-limiting conformational change after agonist binding, prior to channel opening. We hypothesize that this conformational change occurs within a triad of interactions between a short helix preceding the M1 transmembrane helix, the highly conserved M3 motif encoded by the residues SYTANLAAF, and the linker preceding the M4 transmembrane helix of the adjacent subunit. Molecular dynamics simulations suggest that pre-M1 helix motion is uncorrelated between subunits, which we interpret to suggest independent subunit-specific conformational changes may influence these pre-gating steps. According to this interpretation, these conformational changes are the main determinants of the key kinetic properties of NMDA receptor activation following agonist binding, and so these steps sculpt their physiological role. We show that this structurally derived tetrameric model describes both single channel and macroscopic data, giving a new approach to interpreting functional properties of synaptic NMDARs that provides a logical framework to understanding receptors with non-identical subunits.


Subject(s)
Glutamic Acid/metabolism , Ion Channel Gating , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission , HEK293 Cells , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Multimerization , Protein Subunits
14.
Cell Rep ; 23(4): 1060-1071, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29694885

ABSTRACT

The structural and functional plasticity of synapses is critical for learning and memory. Long-term potentiation (LTP) induction promotes spine growth and AMPAR accumulation at excitatory synapses, leading to increased synaptic strength. Glutamate initiates these processes, but the contribution from extracellular modulators is not fully established. Wnts are required for spine formation; however, their impact on activity-mediated spine plasticity and AMPAR localization is unknown. We found that LTP induction rapidly increased synaptic Wnt7a/b protein levels. Acute blockade of endogenous Wnts or loss of postsynaptic Frizzled-7 (Fz7) receptors impaired LTP-mediated synaptic strength, spine growth, and AMPAR localization at synapses. Live imaging of SEP-GluA1 and single-particle tracking revealed that Wnt7a rapidly promoted synaptic AMPAR recruitment and trapping. Wnt7a, through Fz7, induced CaMKII-dependent loss of SynGAP from spines and increased extrasynaptic AMPARs by PKA phosphorylation. We identify a critical role for Wnt-Fz7 signaling in LTP-mediated synaptic accumulation of AMPARs and spine plasticity.


Subject(s)
Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Glutamate/metabolism , Spine/metabolism , Wnt Signaling Pathway/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Frizzled Receptors , Mice , Proto-Oncogene Proteins/metabolism , Spine/cytology , Wnt Proteins/metabolism
15.
Nat Commun ; 9(1): 957, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29511171

ABSTRACT

Genetic and bioinformatic analyses have identified missense mutations in GRIN2B encoding the NMDA receptor GluN2B subunit in autism, intellectual disability, Lennox Gastaut and West Syndromes. Here, we investigated several such mutations using a near-complete, hybrid 3D model of the human NMDAR and studied their consequences with kinetic modelling and electrophysiology. The mutants revealed reductions in glutamate potency; increased receptor desensitisation; and ablation of voltage-dependent Mg2+ block. In addition, we provide new views on Mg2+ and NMDA channel blocker binding sites. We demonstrate that these mutants have significant impact on excitatory transmission in developing neurons, revealing profound changes that could underlie their associated neurological disorders. Of note, the NMDAR channel mutant GluN2BV618G unusually allowed Mg2+ permeation, whereas nearby N615I reduced Ca2+ permeability. By identifying the binding site for an NMDAR antagonist that is used in the clinic to rescue gain-of-function phenotypes, we show that drug binding may be modified by some GluN2B disease-causing mutations.


Subject(s)
Disease/genetics , Ion Channels/metabolism , Mutation, Missense/genetics , Protein Subunits/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Amino Acid Sequence , Animals , Cell Membrane Permeability/drug effects , Computational Biology , Excitatory Postsynaptic Potentials/drug effects , Glutamates/metabolism , HEK293 Cells , Humans , Ligands , Magnesium/pharmacology , Memantine/pharmacology , Models, Molecular , Neurons/metabolism , Protein Subunits/chemistry , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/chemistry
16.
17.
Curr Biol ; 26(19): 2551-2561, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27593374

ABSTRACT

Synapse degeneration occurs early in neurodegenerative diseases and correlates strongly with cognitive decline in Alzheimer's disease (AD). The molecular mechanisms that trigger synapse vulnerability and those that promote synapse regeneration after substantial synaptic failure remain poorly understood. Increasing evidence suggests a link between a deficiency in Wnt signaling and AD. The secreted Wnt antagonist Dickkopf-1 (Dkk1), which is elevated in AD, contributes to amyloid-ß-mediated synaptic failure. However, the impact of Dkk1 at the circuit level and the mechanism by which synapses disassemble have not yet been explored. Using a transgenic mouse model that inducibly expresses Dkk1 in the hippocampus, we demonstrate that Dkk1 triggers synapse loss, impairs long-term potentiation, enhances long-term depression, and induces learning and memory deficits. We decipher the mechanism involved in synapse loss induced by Dkk1 as it can be prevented by combined inhibition of the Gsk3 and RhoA-Rock pathways. Notably, after loss of synaptic connectivity, reactivation of the Wnt pathway by cessation of Dkk1 expression completely restores synapse number, synaptic plasticity, and long-term memory. These findings demonstrate the remarkable capacity of adult neurons to regenerate functional circuits and highlight Wnt signaling as a targetable pathway for neuronal circuit recovery after synapse degeneration.


Subject(s)
Hippocampus/physiopathology , Intercellular Signaling Peptides and Proteins/genetics , Memory, Long-Term , Neuronal Plasticity , Synapses/physiology , Wnt Signaling Pathway , Animals , Female , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic
18.
Nat Commun ; 6: 8302, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26400647

ABSTRACT

The functional assembly of the synaptic release machinery is well understood; however, how signalling factors modulate this process remains unknown. Recent studies suggest that Wnts play a role in presynaptic function. To examine the mechanisms involved, we investigated the interaction of release machinery proteins with Dishevelled-1 (Dvl1), a scaffold protein that determines the cellular locale of Wnt action. Here we show that Dvl1 directly interacts with Synaptotagmin-1 (Syt-1) and indirectly with the SNARE proteins SNAP25 and Syntaxin (Stx-1). Importantly, the interaction of Dvl1 with Syt-1, which is regulated by Wnts, modulates neurotransmitter release. Moreover, presynaptic terminals from Wnt signalling-deficient mice exhibit reduced release probability and are unable to sustain high-frequency release. Consistently, the readily releasable pool size and formation of SNARE complexes are reduced. Our studies demonstrate that Wnt signalling tunes neurotransmitter release and identify Syt-1 as a target for modulation by secreted signalling proteins.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Neurons/metabolism , Neurotransmitter Agents/metabolism , Phosphoproteins/genetics , Synaptic Vesicles/metabolism , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmin I/metabolism , Syntaxin 1/metabolism , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dishevelled Proteins , Fluorescent Antibody Technique , Hippocampus/cytology , Hippocampus/metabolism , Immunoprecipitation , Mice , Mice, Knockout , Microscopy, Electron , Patch-Clamp Techniques , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Transmission , Wnt Proteins/genetics
19.
Nat Commun ; 5: 4992, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25318560

ABSTRACT

Synapse degeneration is an early and invariant feature of neurodegenerative diseases. Indeed, synapse loss occurs prior to neuronal degeneration and correlates with the symptom severity of these diseases. However, the molecular mechanisms that trigger synaptic loss remain poorly understood. Here we demonstrate that deficient Wnt signalling elicits synaptic degeneration in the adult striatum. Inducible expression of the secreted Wnt antagonist Dickkopf1 (Dkk1) in adult mice (iDkk1) decreases the number of cortico-striatal glutamatergic synapses and of D1 and D2 dopamine receptor clusters. Synapse loss occurs in the absence of axon retraction or cell death. The remaining excitatory terminals contain fewer synaptic vesicles and have a reduced probability of evoked transmitter release. IDkk1 mice show impaired motor coordination and are irresponsive to amphetamine. These studies identify Wnts as key endogenous regulators of synaptic maintenance and suggest that dysfunction in Wnt signalling contributes to synaptic degeneration at early stages in neurodegenerative diseases.


Subject(s)
Motor Skills , Neurodegenerative Diseases/physiopathology , Synapses/pathology , Wnt Proteins/metabolism , Amphetamines/chemistry , Animals , Axons/metabolism , Cell Death , Corpus Striatum/pathology , Dopamine/metabolism , Doxycycline/chemistry , Female , Heterozygote , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction
20.
J Physiol ; 592(10): 2059-78, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24614743

ABSTRACT

Native NMDA receptors (NMDARs) are tetrameric channels formed by two GluN1 and two GluN2 subunits. So far, seven NMDARs subunits have been identified and they can form diheteromeric or triheteromeric NMDARs (more than one type of GluN2 subunit). Extracellular Mg(2+) is an important regulator of NMDARs, and particularly the voltage dependence of Mg(2+) block is crucial to the roles of NMDARs in synaptic plasticity and the integration of synaptic activity with neuronal activity. Although the Mg(2+) block properties of diheteromeric NMDARs are fully investigated, properties of triheteromeric NMDARs are still not clear. Our previous data suggested that dopaminergic neurones expressed triheteromeric GluN1-GluN2B-GluN2D NMDARs. Here, using NMDARs in dopaminergic neurones from postnatal day 7 (P7) rats as a model system, we characterize the voltage-dependent Mg(2+) block properties of triheteromeric NMDARs. In control conditions, external Mg(2+) significantly inhibits the whole cell NMDA-evoked current in a voltage-dependent manner with IC50 values of 20.9 µm, 53.3 µm and 173 µm at -90 mV, -70 mV and -50 mV, respectively. When the GluN2B-selective antagonist ifenprodil was applied, the Mg(2+) sensitivity of the residual NMDA-mediated currents (which is mainly carried by GluN1-GluN2B-GluN2D NMDARs) is reduced to IC50 values of 45.9 µm (-90 mV), 104 µm (-70 mV) and 276 µm (-50 mV), suggesting that triheteromeric GluN1-GluN2B-GluN2D NMDARs have less affinity for external Mg(2+) than GluN1-GluN2B receptors. In addition, fitting INMDA-V curves with a trapping Mg(2+) block model shows the triheteromeric GluN1-GluN2B-GluN2D NMDARs have weaker voltage-dependent Mg(2+) block (δ = 0.56) than GluN1-GluN2B NMDARs. Finally, our concentration jump and single channel recordings suggest that GluN1-GluN2B-GluN2D rather than GluN1-GluN2D NMDARs are present. These data provide information relevant to Mg(2+) block characteristics of triheteromeric NMDARs and may help to better understand synaptic plasticity, which is dependent on these triheteromeric NMDARs.


Subject(s)
Dopaminergic Neurons/metabolism , Ion Channel Gating/physiology , Magnesium/pharmacology , Membrane Potentials/physiology , Pars Compacta/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Animals, Newborn , Cells, Cultured , Dopaminergic Neurons/drug effects , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Pars Compacta/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL