1
|
Griffin H, Hanson J, Phelan KD, Baldini G. MC4R Localizes at Excitatory Postsynaptic and Peri-Postsynaptic Sites of Hypothalamic Neurons in Primary Culture. Cells 2024; 13:1235. [PMID: 39120267 PMCID: PMC11311852 DOI: 10.3390/cells13151235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
The melanocortin-4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that is expressed in several brain locations encompassing the hypothalamus and the brainstem, where the receptor controls several body functions, including metabolism. In a well-defined pathway to decrease appetite, hypothalamic proopiomelanocortin (POMC) neurons localized in the arcuate nucleus (Arc) project to MC4R neurons in the paraventricular nuclei (PVN) to release the natural MC4R agonist α-melanocyte-stimulating hormone (α-MSH). Arc neurons also project excitatory glutamatergic fibers to the MC4R neurons in the PVN for a fast synaptic transmission to regulate a satiety pathway potentiated by α-MSH. By using super-resolution microscopy, we found that in hypothalamic neurons in a primary culture, postsynaptic density protein 95 (PSD95) colocalizes with GluN1, a subunit of the ionotropic N-methyl-D-aspartate receptor (NMDAR). Thus, hypothalamic neurons form excitatory postsynaptic specializations. To study the MC4R distribution at these sites, tagged HA-MC4R under the synapsin promoter was expressed in neurons by adeno-associated virus (AAV) gene transduction. HA-MC4R immunofluorescence peaked at the center and in proximity to the PSD95- and NMDAR-expressing sites. These data provide morphological evidence that MC4R localizes together with glutamate receptors at postsynaptic and peri-postsynaptic sites.
Collapse
Affiliation(s)
- Haven Griffin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Jude Hanson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Kevin D. Phelan
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| |
Collapse
|
2
|
Nakayama H, Miyazaki T, Abe M, Yamazaki M, Kawamura Y, Choo M, Konno K, Kawata S, Uesaka N, Hashimoto K, Miyata M, Sakimura K, Watanabe M, Kano M. Direct and indirect pathways for heterosynaptic interaction underlying developmental synapse elimination in the mouse cerebellum. Commun Biol 2024; 7:806. [PMID: 38961250 PMCID: PMC11222442 DOI: 10.1038/s42003-024-06447-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 06/12/2024] [Indexed: 07/05/2024] Open
Abstract
Developmental synapse elimination is crucial for shaping mature neural circuits. In the neonatal mouse cerebellum, Purkinje cells (PCs) receive excitatory synaptic inputs from multiple climbing fibers (CFs) and synapses from all but one CF are eliminated by around postnatal day 20. Heterosynaptic interaction between CFs and parallel fibers (PFs), the axons of cerebellar granule cells (GCs) forming excitatory synapses onto PCs and molecular layer interneurons (MLIs), is crucial for CF synapse elimination. However, mechanisms for this heterosynaptic interaction are largely unknown. Here we show that deletion of AMPA-type glutamate receptor functions in GCs impairs CF synapse elimination mediated by metabotropic glutamate receptor 1 (mGlu1) signaling in PCs. Furthermore, CF synapse elimination is impaired by deleting NMDA-type glutamate receptors from MLIs. We propose that PF activity is crucial for CF synapse elimination by directly activating mGlu1 in PCs and indirectly enhancing the inhibition of PCs through activating NMDA receptors in MLIs.
Collapse
Affiliation(s)
- Hisako Nakayama
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Taisuke Miyazaki
- Department of Functioning and Disability, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yoshinobu Kawamura
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Myeongjeong Choo
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kohtarou Konno
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shinya Kawata
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naofumi Uesaka
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mariko Miyata
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, Japan.
- Advanced Comprehensive Research Organization (ACRO), Teikyo University, Tokyo, Japan.
| |
Collapse
|
3
|
Pennock RL, Coddington LT, Yan X, Overstreet-Wadiche L, Wadiche JI. Afferent convergence to a shared population of interneuron AMPA receptors. Nat Commun 2023; 14:3113. [PMID: 37253743 PMCID: PMC10229553 DOI: 10.1038/s41467-023-38854-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/12/2023] [Indexed: 06/01/2023] Open
Abstract
Precise alignment of pre- and postsynaptic elements optimizes the activation of glutamate receptors at excitatory synapses. Nonetheless, glutamate that diffuses out of the synaptic cleft can have actions at distant receptors, a mode of transmission called spillover. To uncover the extrasynaptic actions of glutamate, we localized AMPA receptors (AMPARs) mediating spillover transmission between climbing fibers and molecular layer interneurons in the cerebellar cortex. We found that climbing fiber spillover generates calcium transients mediated by Ca2+-permeable AMPARs at parallel fiber synapses. Spillover occludes parallel fiber synaptic currents, indicating that separate, independently regulated afferent pathways converge onto a common pool of AMPARs. Together these findings demonstrate a circuit motif wherein glutamate 'spill-in' from an unconnected afferent pathway co-opts synaptic receptors, allowing activation of postsynaptic AMPARs even when canonical glutamate release is suppressed.
Collapse
Affiliation(s)
- Reagan L Pennock
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Luke T Coddington
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Xiaohui Yan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | | | - Jacques I Wadiche
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
4
|
Hindley N, Sanchez Avila A, Henstridge C. Bringing synapses into focus: Recent advances in synaptic imaging and mass-spectrometry for studying synaptopathy. Front Synaptic Neurosci 2023; 15:1130198. [PMID: 37008679 PMCID: PMC10050382 DOI: 10.3389/fnsyn.2023.1130198] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
Synapses are integral for healthy brain function and are becoming increasingly recognized as key structures in the early stages of brain disease. Understanding the pathological processes driving synaptic dysfunction will unlock new therapeutic opportunities for some of the most devastating diseases of our time. To achieve this we need a solid repertoire of imaging and molecular tools to interrogate synaptic biology at greater resolution. Synapses have historically been examined in small numbers, using highly technical imaging modalities, or in bulk, using crude molecular approaches. However, recent advances in imaging techniques are allowing us to analyze large numbers of synapses, at single-synapse resolution. Furthermore, multiplexing is now achievable with some of these approaches, meaning we can examine multiple proteins at individual synapses in intact tissue. New molecular techniques now allow accurate quantification of proteins from isolated synapses. The development of increasingly sensitive mass-spectrometry equipment means we can now scan the synaptic molecular landscape almost in totality and see how this changes in disease. As we embrace these new technical developments, synapses will be viewed with clearer focus, and the field of synaptopathy will become richer with insightful and high-quality data. Here, we will discuss some of the ways in which synaptic interrogation is being facilitated by methodological advances, focusing on imaging, and mass spectrometry.
Collapse
Affiliation(s)
- Nicole Hindley
- Division of Cellular and Systems Medicine, University of Dundee, Dundee, United Kingdom
| | - Anna Sanchez Avila
- Division of Cellular and Systems Medicine, University of Dundee, Dundee, United Kingdom
- Euan Macdonald Centre for Motor Neuron Disease, University of Edinburgh, Edinburgh, United Kingdom
| | - Christopher Henstridge
- Division of Cellular and Systems Medicine, University of Dundee, Dundee, United Kingdom
- Euan Macdonald Centre for Motor Neuron Disease, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
5
|
mGluR5 is transiently confined in perisynaptic nanodomains to shape synaptic function. Nat Commun 2023; 14:244. [PMID: 36646691 PMCID: PMC9842668 DOI: 10.1038/s41467-022-35680-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
The unique perisynaptic distribution of postsynaptic metabotropic glutamate receptors (mGluRs) at excitatory synapses is predicted to directly shape synaptic function, but mechanistic insight into how this distribution is regulated and impacts synaptic signaling is lacking. We used live-cell and super-resolution imaging approaches, and developed molecular tools to resolve and acutely manipulate the dynamic nanoscale distribution of mGluR5. Here we show that mGluR5 is dynamically organized in perisynaptic nanodomains that localize close to, but not in the synapse. The C-terminal domain of mGluR5 critically controlled perisynaptic confinement and prevented synaptic entry. We developed an inducible interaction system to overcome synaptic exclusion of mGluR5 and investigate the impact on synaptic function. We found that mGluR5 recruitment to the synapse acutely increased synaptic calcium responses. Altogether, we propose that transient confinement of mGluR5 in perisynaptic nanodomains allows flexible modulation of synaptic function.
Collapse
|
6
|
Surdin T, Preissing B, Rohr L, Grömmke M, Böke H, Barcik M, Azimi Z, Jancke D, Herlitze S, Mark MD, Siveke I. Optogenetic activation of mGluR1 signaling in the cerebellum induces synaptic plasticity. iScience 2022; 26:105828. [PMID: 36632066 PMCID: PMC9826949 DOI: 10.1016/j.isci.2022.105828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/21/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Neuronal plasticity underlying cerebellar learning behavior is strongly associated with type 1 metabotropic glutamate receptor (mGluR1) signaling. Activation of mGluR1 leads to activation of the Gq/11 pathway, which is involved in inducing synaptic plasticity at the parallel fiber-Purkinje cell synapse (PF-PC) in form of long-term depression (LTD). To optogenetically modulate mGluR1 signaling we fused mouse melanopsin (OPN4) that activates the Gq/11 pathway to the C-termini of mGluR1 splice variants (OPN4-mGluR1a and OPN4-mGluR1b). Activation of both OPN4-mGluR1 variants showed robust Ca2+ increase in HEK cells and PCs of cerebellar slices. We provide the prove-of-concept approach to modulate synaptic plasticity via optogenetic activation of OPN4-mGluR1a inducing LTD at the PF-PC synapse in vitro. Moreover, we demonstrate that light activation of mGluR1a signaling pathway by OPN4-mGluR1a in PCs leads to an increase in intrinsic activity of PCs in vivo and improved cerebellum driven learning behavior.
Collapse
Affiliation(s)
- Tatjana Surdin
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Bianca Preissing
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Lennard Rohr
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Michelle Grömmke
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Hanna Böke
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Maike Barcik
- Cardiovascular Research Institute Düsseldorf, Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Zohre Azimi
- Optical Imaging Group, Institut für Neuroinformatik, Ruhr-University Bochum, Bochum, Germany
| | - Dirk Jancke
- Optical Imaging Group, Institut für Neuroinformatik, Ruhr-University Bochum, Bochum, Germany
| | - Stefan Herlitze
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany,Corresponding author
| | - Melanie D. Mark
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Ida Siveke
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany,Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany,Corresponding author
| |
Collapse
|
7
|
Burghard AL, Lee CM, Fabrizio-Stover EM, Oliver DL. Long-Duration Sound-Induced Facilitation Changes Population Activity in the Inferior Colliculus. Front Syst Neurosci 2022; 16:920642. [PMID: 35873097 PMCID: PMC9301083 DOI: 10.3389/fnsys.2022.920642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
The inferior colliculus (IC) is at the midpoint of the auditory system and integrates virtually all information ascending from the auditory brainstem, organizes it, and transmits the results to the auditory forebrain. Its abundant, excitatory local connections are crucial for this task. This study describes a long duration sound (LDS)-induced potentiation in the IC that changes both subsequent tone-evoked responses and spontaneous activity. Afterdischarges, changes of spontaneous spiking following an LDS, were seen previously in single neurons. Here, we used multi-channel probes to record activity before and after a single, tetanic sound and describe the changes in a population of IC neurons. Following a 60 s narrowband-noise stimulation, a subset of recording channels (∼16%) showed afterdischarges. A facilitated response spike rate to tone pips following an LDS was also observed in ∼16% of channels. Both channels with an afterdischarge and channels with facilitated tone responses had higher firing rates in response to LDS, and the magnitude of the afterdischarges increased with increased responses to the LDS. This is the first study examining the effect of LDS stimulation on tone-evoked responses. This observed facilitation in vivo has similarities to post-tetanic potentiation in vitro as both manner of induction (strong stimulation for several seconds) as well as time-course of the facilitation (second to minute range) are comparable. Channels with and without facilitation appear to be intermixed and distributed widely in the central nucleus of IC, and this suggests a heretofore unknown property of some IC neurons or their circuits. Consequently, this sound-evoked facilitation may enhance the sound-evoked output of these neurons, while, simultaneously, most other IC neurons have reduced or unchanged output in response to the same stimulus.
Collapse
|
8
|
Westra M, MacGillavry HD. Precise Detection and Visualization of Nanoscale Temporal Confinement in Single-Molecule Tracking Analysis. MEMBRANES 2022; 12:membranes12070650. [PMID: 35877853 PMCID: PMC9320997 DOI: 10.3390/membranes12070650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022]
Abstract
The plasma membrane consists of a diverse mixture of molecules that dynamically assemble into a highly non-random organization. The formation of nanoscale domains in the membrane is of particular interest as these domains underlie critical cellular functions. Single-molecule tracking is a powerful method to detect and quantify molecular motion at high temporal and spatial resolution and has therefore been instrumental in understanding mechanisms that underlie membrane organization. In single-molecule trajectories, regions of temporal confinement can be determined that might reveal interesting biophysical interactions important for domain formation. However, analytical methods for the detection of temporal confinement in single-molecule trajectories depend on a variety of parameters that heavily depend on experimental factors and the influence of these factors on the performance of confinement detection are not well understood. Here, we present elaborate confinement analyses on simulated random walks and trajectories that display transient confined behavior to optimize the parameters for different experimental conditions. Furthermore, we demonstrate a heatmap visualization tool that allows spatial mapping of confinement hotspots relative to subcellular markers. Using these optimized tools, we reliably detected subdiffusive behavior of different membrane components and observed differences in the confinement behavior of two types of glutamate receptors in neurons. This study will help in further understanding the dynamic behavior of the complex membrane and its role in cellular functioning.
Collapse
|
9
|
Wichmann C, Kuner T. Heterogeneity of glutamatergic synapses: cellular mechanisms and network consequences. Physiol Rev 2022; 102:269-318. [PMID: 34727002 DOI: 10.1152/physrev.00039.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chemical synapses are commonly known as a structurally and functionally highly diverse class of cell-cell contacts specialized to mediate communication between neurons. They represent the smallest "computational" unit of the brain and are typically divided into excitatory and inhibitory as well as modulatory categories. These categories are subdivided into diverse types, each representing a different structure-function repertoire that in turn are thought to endow neuronal networks with distinct computational properties. The diversity of structure and function found among a given category of synapses is referred to as heterogeneity. The main building blocks for this heterogeneity are synaptic vesicles, the active zone, the synaptic cleft, the postsynaptic density, and glial processes associated with the synapse. Each of these five structural modules entails a distinct repertoire of functions, and their combination specifies the range of functional heterogeneity at mammalian excitatory synapses, which are the focus of this review. We describe synapse heterogeneity that is manifested on different levels of complexity ranging from the cellular morphology of the pre- and postsynaptic cells toward the expression of different protein isoforms at individual release sites. We attempt to define the range of structural building blocks that are used to vary the basic functional repertoire of excitatory synaptic contacts and discuss sources and general mechanisms of synapse heterogeneity. Finally, we explore the possible impact of synapse heterogeneity on neuronal network function.
Collapse
Affiliation(s)
- Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg, Germany
| |
Collapse
|
10
|
Membrane trafficking and positioning of mGluRs at presynaptic and postsynaptic sites of excitatory synapses. Neuropharmacology 2021; 200:108799. [PMID: 34592242 DOI: 10.1016/j.neuropharm.2021.108799] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 01/21/2023]
Abstract
The plethora of functions of glutamate in the brain are mediated by the complementary actions of ionotropic and metabotropic glutamate receptors (mGluRs). The ionotropic glutamate receptors carry most of the fast excitatory transmission, while mGluRs modulate transmission on longer timescales by triggering multiple intracellular signaling pathways. As such, mGluRs mediate critical aspects of synaptic transmission and plasticity. Interestingly, at synapses, mGluRs operate at both sides of the cleft, and thus bidirectionally exert the effects of glutamate. At postsynaptic sites, group I mGluRs act to modulate excitability and plasticity. At presynaptic sites, group II and III mGluRs act as auto-receptors, modulating release properties in an activity-dependent manner. Thus, synaptic mGluRs are essential signal integrators that functionally couple presynaptic and postsynaptic mechanisms of transmission and plasticity. Understanding how these receptors reach the membrane and are positioned relative to the presynaptic glutamate release site are therefore important aspects of synapse biology. In this review, we will discuss the currently known mechanisms underlying the trafficking and positioning of mGluRs at and around synapses, and how these mechanisms contribute to synaptic functioning. We will highlight outstanding questions and present an outlook on how recent technological developments will move this exciting research field forward.
Collapse
|
11
|
Petralia RS, Wang YX. Review of Post-embedding Immunogold Methods for the Study of Neuronal Structures. Front Neuroanat 2021; 15:763427. [PMID: 34720893 PMCID: PMC8551803 DOI: 10.3389/fnana.2021.763427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/28/2021] [Indexed: 01/03/2023] Open
Abstract
The post-embedding immunogold (PI) technique for immunolabeling of neuronal tissues utilizing standard thin-section transmission electron microscopy (TEM) continues to be a prime method for understanding the functional localization of key proteins in neuronal function. Its main advantages over other immunolabeling methods for thin-section TEM are (1) fairly accurate and quantifiable localization of proteins in cells; (2) double-labeling of sections using two gold particle sizes; and (3) the ability to perform multiple labeling for different proteins by using adjacent sections. Here we first review in detail a common method for PI of neuronal tissues. This method has two major parts. First, we describe the freeze-substitution embedding method: cryoprotected tissue is frozen in liquid propane via plunge-freezing, and is placed in a freeze-substitution instrument in which the tissue is embedded in Lowicryl at low temperatures. We highlight important aspects of freeze-substitution embedding. Then we outline how thin sections of embedded tissue on grids are labeled with a primary antibody and a secondary gold particle-conjugated antibody, and the particular problems encountered in TEM of PI-labeled sections. In the Discussion, we compare our method both to earlier PI methods and to more recent PI methods used by other laboratories. We also compare TEM immunolabeling using PI vs. various pre-embedding immunolabeling methods, especially relating to neuronal tissue.
Collapse
Affiliation(s)
- Ronald S. Petralia
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | | |
Collapse
|
12
|
Zieger HL, Choquet D. Nanoscale synapse organization and dysfunction in neurodevelopmental disorders. Neurobiol Dis 2021; 158:105453. [PMID: 34314857 DOI: 10.1016/j.nbd.2021.105453] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022] Open
Abstract
Neurodevelopmental disorders such as those linked to intellectual disabilities or autism spectrum disorder are thought to originate in part from genetic defects in synaptic proteins. Single gene mutations linked to synapse dysfunction can broadly be separated in three categories: disorders of transcriptional regulation, disorders of synaptic signaling and disorders of synaptic scaffolding and structures. The recent developments in super-resolution imaging technologies and their application to synapses have unraveled a complex nanoscale organization of synaptic components. On the one hand, part of receptors, adhesion proteins, ion channels, scaffold elements and the pre-synaptic release machinery are partitioned in subsynaptic nanodomains, and the respective organization of these nanodomains has tremendous impact on synaptic function. For example, pre-synaptic neurotransmitter release sites are partly aligned with nanometer precision to postsynaptic receptor clusters. On the other hand, a large fraction of synaptic components is extremely dynamic and constantly exchanges between synaptic domains and extrasynaptic or intracellular compartments. It is largely the combination of the exquisitely precise nanoscale synaptic organization of synaptic components and their high dynamic that allows the rapid and profound regulation of synaptic function during synaptic plasticity processes that underlie adaptability of brain function, learning and memory. It is very tempting to speculate that genetic defects that lead to neurodevelopmental disorders and target synaptic scaffolds and structures mediate their deleterious impact on brain function through perturbing synapse nanoscale dynamic organization. We discuss here how applying super-resolution imaging methods in models of neurodevelopmental disorders could help in addressing this question.
Collapse
Affiliation(s)
- Hanna L Zieger
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Daniel Choquet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France; Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, F-33000 Bordeaux, France.
| |
Collapse
|
13
|
Nagaraja RY, Sherry DM, Fessler JL, Stiles MA, Li F, Multani K, Orock A, Ahmad M, Brush RS, Anderson RE, Agbaga MP, Deák F. W246G Mutant ELOVL4 Impairs Synaptic Plasticity in Parallel and Climbing Fibers and Causes Motor Defects in a Rat Model of SCA34. Mol Neurobiol 2021; 58:4921-4943. [PMID: 34227061 PMCID: PMC8497303 DOI: 10.1007/s12035-021-02439-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022]
Abstract
Spinocerebellar ataxia (SCA) is a neurodegenerative disorder characterized by ataxia and cerebellar atrophy. A number of different mutations gives rise to different types of SCA with characteristic ages of onset, symptomatology, and rates of progression. SCA type 34 (SCA34) is caused by mutations in ELOVL4 (ELOngation of Very Long-chain fatty acids 4), a fatty acid elongase essential for biosynthesis of Very Long Chain Saturated and Polyunsaturated Fatty Acids (VLC-SFA and VLC-PUFA, resp., ≥28 carbons), which have important functions in the brain, skin, retina, Meibomian glands, testes, and sperm. We generated a rat model of SCA34 by knock-in of the SCA34-causing 736T>G (p.W246G) ELOVL4 mutation. Rats carrying the mutation developed impaired motor deficits by 2 months of age. To understand the mechanism of these motor deficits, we performed electrophysiological studies using cerebellar slices from rats homozygous for W246G mutant ELOVL4 and found marked reduction of long-term potentiation at parallel fiber synapses and long-term depression at climbing fiber synapses onto Purkinje cells. Neuroanatomical analysis of the cerebellum showed normal cytoarchitectural organization with no evidence of degeneration out to 6 months of age. These results point to ELOVL4 as essential for motor function and cerebellar synaptic plasticity. The results further suggest that ataxia in SCA34 patients may arise from a primary impairment of synaptic plasticity and cerebellar network desynchronization before onset of neurodegeneration and progression of the disease at a later age.
Collapse
Affiliation(s)
- Raghavendra Y Nagaraja
- Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Neuroscience Program, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Cell Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - David M Sherry
- Neuroscience Program, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Cell Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Jennifer L Fessler
- Cell Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Megan A Stiles
- Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Feng Li
- Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Karanpreet Multani
- Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Albert Orock
- Neuroscience Program, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Reynolds Center on Aging, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Mohiuddin Ahmad
- Neuroscience Program, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Cell Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Richard S Brush
- Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Robert E Anderson
- Neuroscience Program, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Cell Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA
| | - Martin-Paul Agbaga
- Neuroscience Program, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA. .,Cell Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA. .,Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA. .,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA. .,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA.
| | - Ferenc Deák
- Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA. .,Neuroscience Program, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA. .,Reynolds Center on Aging, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, DMEI 428PP, Oklahoma City, OK, 73104, USA. .,Dept. of Neuroscience & Regenerative Medicine, Medical College of Georgia, 1120 15th Str, CA4010, Augusta, GA, 30912, USA.
| |
Collapse
|
14
|
Bockaert J, Perroy J, Ango F. The Complex Formed by Group I Metabotropic Glutamate Receptor (mGluR) and Homer1a Plays a Central Role in Metaplasticity and Homeostatic Synaptic Scaling. J Neurosci 2021; 41:5567-5578. [PMID: 34193623 PMCID: PMC8244974 DOI: 10.1523/jneurosci.0026-21.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/28/2022] Open
Abstract
G-protein-coupled receptors can be constitutively activated following physical interaction with intracellular proteins. The first example described was the constitutive activation of Group I metabotropic glutamate receptors (mGluR: mGluR1,5) following their interaction with Homer1a, an activity-inducible early-termination variant of the scaffolding protein Homer that lacks dimerization capacity (Ango et al., 2001). Homer1a disrupts the links, maintained by the long form of Homer (cross-linking Homers), between mGluR1,5 and the Shank-GKAP-PSD-95-ionotropic glutamate receptor network. Two characteristics of the constitutive activation of the Group I mGluR-Homer1a complex are particularly interesting: (1) it affects a large number of synapses in which Homer1a is upregulated following enhanced, long-lasting neuronal activity; and (2) it mainly depends on Homer1a protein turnover. The constitutively active Group I mGluR-Homer1a complex is involved in the two main forms of non-Hebbian neuronal plasticity: "metaplasticity" and "homeostatic synaptic scaling," which are implicated in a large series of physiological and pathologic processes. Those include non-Hebbian plasticity observed in visual system, synapses modulated by addictive drugs (rewarded synapses), chronically overactivated synaptic networks, normal sleep, and sleep deprivation.
Collapse
Affiliation(s)
- Joël Bockaert
- Institut de Génomique Fonctionnelle, Université Montpellier, Center National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, 34094 Montpellier, France
| | - Julie Perroy
- Institut de Génomique Fonctionnelle, Université Montpellier, Center National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, 34094 Montpellier, France
| | - Fabrice Ango
- Institut des Neurosciences de Montpellier, Université Montpellier, Center National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, 34295 Montpellier, France
| |
Collapse
|
15
|
Burada AP, Vinnakota R, Lambolez B, Tricoire L, Kumar J. Structural biology of ionotropic glutamate delta receptors and their crosstalk with metabotropic glutamate receptors. Neuropharmacology 2021; 196:108683. [PMID: 34181979 DOI: 10.1016/j.neuropharm.2021.108683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/31/2021] [Accepted: 06/17/2021] [Indexed: 10/21/2022]
Abstract
Enigmatic orphan glutamate delta receptors (GluD) are one of the four classes of the ionotropic glutamate receptors (iGluRs) that play key roles in synaptic transmission and plasticity. While members of other iGluR families viz AMPA, NMDA, and kainate receptors are gated by glutamate, the GluD receptors neither bind glutamate nor evoke ligand-induced currents upon binding of glycine and D-serine. Thus, the GluD receptors were considered to function as structural proteins that facilitate the formation, maturation, and maintenance of synapses in the hippocampus and cerebellum. Recent work has revealed that GluD receptors have extensive crosstalk with metabotropic glutamate receptors (mGlus) and are also gated by their activation. The latest development of a novel optopharamcological tool and the cryoEM structures of GluD receptors would help define the molecular and chemical basis of the GluD receptor's role in synaptic physiology. This article is part of the special Issue on "Glutamate Receptors - Orphan iGluRs".
Collapse
Affiliation(s)
- Ananth Prasad Burada
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, 411007, India
| | - Rajesh Vinnakota
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, 411007, India
| | - Bertrand Lambolez
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Ludovic Tricoire
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France.
| | - Janesh Kumar
- Laboratory of Membrane Protein Biology, National Centre for Cell Science, NCCS Complex, S. P. Pune University, Pune, Maharashtra, 411007, India.
| |
Collapse
|
16
|
Feng X, Yang F, Rabenstein M, Wang Z, Frech MJ, Wree A, Bräuer AU, Witt M, Gläser A, Hermann A, Rolfs A, Luo J. Stimulation of mGluR1/5 Improves Defective Internalization of AMPA Receptors in NPC1 Mutant Mouse. Cereb Cortex 2021; 30:1465-1480. [PMID: 31599924 DOI: 10.1093/cercor/bhz179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Niemann-Pick type C1 (NPC1) disease is characterized by neurodegeneration caused by cholesterol accumulation in the late endosome/lysosome. In this study, a defective basal and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-stimulated internalization of GluR2-containing AMPA receptors in NPC1-/- cortical neurons was detected. Our results show that the amount of cholesterol and group I metabotropic glutamate receptors (mGluR1/5) in lipid rafts of NPC1-/- cortical tissue and neurons are decreased and their downstream signals of p-ERK are defective, which are restored by a rebalance of cholesterol homeostasis through β-cyclodextrin (β-CD) treatment. Application of 3,5-dihydroxyphenylglycine (DHPG)-a mGluR1/5 agonist-and β-CD markedly increases the internalization of AMPA receptors and decreases over-influx of calcium in NPC1-/- neurons, respectively. Furthermore, the defective phosphorylated GluR2 and protein kinase C signals are ameliorated by the treatment with DHPG and β-CD, respectively, suggesting an involvement of them in internalization dysfunction. Taken together, our data imply that abnormal internalization of AMPA receptors is a critical mechanism for neuronal dysfunction and the correction of dysfunctional mGluR1/5 is a potential therapeutic strategy for NPC1 disease.
Collapse
Affiliation(s)
- Xiao Feng
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany
| | - Fan Yang
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany
| | - Michael Rabenstein
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany
| | - Zhen Wang
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Moritz J Frech
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany.,Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
| | - Andreas Wree
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock 18147, Germany.,Institute of Anatomy, University Medical Center Rostock, Rostock 18055, Germany
| | - Anja U Bräuer
- Institute of Anatomy, University Medical Center Rostock, Rostock 18055, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University, Oldenburg 26129, Germany.,Research Center for Neurosensory Science, Carl von Ossietzky University, Oldenburg 26129, Germany
| | - Martin Witt
- Institute of Anatomy, University Medical Center Rostock, Rostock 18055, Germany
| | - Anne Gläser
- Institute of Anatomy, University Medical Center Rostock, Rostock 18055, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany.,Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock 18147, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Rostock 18147, Germany
| | | | - Jiankai Luo
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany.,Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
| |
Collapse
|
17
|
mGluR1 signaling in cerebellar Purkinje cells: Subcellular organization and involvement in cerebellar function and disease. Neuropharmacology 2021; 194:108629. [PMID: 34089728 DOI: 10.1016/j.neuropharm.2021.108629] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/20/2022]
Abstract
The cerebellum is essential for the control, coordination, and learning of movements, and for certain aspects of cognitive function. Purkinje cells are the sole output neurons in the cerebellar cortex and therefore play crucial roles in the diverse functions of the cerebellum. The type 1 metabotropic glutamate receptor (mGluR1) is prominently enriched in Purkinje cells and triggers downstream signaling pathways that are required for functional and structural plasticity, and for synaptic responses. To understand how mGluR1 contributes to cerebellar functions, it is important to consider not only the operational properties of this receptor, but also its spatial organization and the molecular interactions that enable its proper functioning. In this review, we highlight how mGluR1 and its related signaling molecules are organized into tightly coupled microdomains to fulfill physiological functions. We also describe emerging evidence that altered mGluR1 signaling in Purkinje cells underlies cerebellar dysfunction in ataxias of human patients and mouse models.
Collapse
|
18
|
Zhang X, Alshakhshir N, Zhao L. Glycolytic Metabolism, Brain Resilience, and Alzheimer's Disease. Front Neurosci 2021; 15:662242. [PMID: 33994936 PMCID: PMC8113697 DOI: 10.3389/fnins.2021.662242] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of age-related dementia. Despite decades of research, the etiology and pathogenesis of AD are not well understood. Brain glucose hypometabolism has long been recognized as a prominent anomaly that occurs in the preclinical stage of AD. Recent studies suggest that glycolytic metabolism, the cytoplasmic pathway of the breakdown of glucose, may play a critical role in the development of AD. Glycolysis is essential for a variety of neural activities in the brain, including energy production, synaptic transmission, and redox homeostasis. Decreased glycolytic flux has been shown to correlate with the severity of amyloid and tau pathology in both preclinical and clinical AD patients. Moreover, increased glucose accumulation found in the brains of AD patients supports the hypothesis that glycolytic deficit may be a contributor to the development of this phenotype. Brain hyperglycemia also provides a plausible explanation for the well-documented link between AD and diabetes. Humans possess three primary variants of the apolipoprotein E (ApoE) gene - ApoE∗ϵ2, ApoE∗ϵ3, and ApoE∗ϵ4 - that confer differential susceptibility to AD. Recent findings indicate that neuronal glycolysis is significantly affected by human ApoE isoforms and glycolytic robustness may serve as a major mechanism that renders an ApoE2-bearing brain more resistant against the neurodegenerative risks for AD. In addition to AD, glycolytic dysfunction has been observed in other neurodegenerative diseases, including Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, strengthening the concept of glycolytic dysfunction as a common pathway leading to neurodegeneration. Taken together, these advances highlight a promising translational opportunity that involves targeting glycolysis to bolster brain metabolic resilience and by such to alter the course of brain aging or disease development to prevent or reduce the risks for not only AD but also other neurodegenerative diseases.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Nadine Alshakhshir
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
19
|
Alcoreza OB, Patel DC, Tewari BP, Sontheimer H. Dysregulation of Ambient Glutamate and Glutamate Receptors in Epilepsy: An Astrocytic Perspective. Front Neurol 2021; 12:652159. [PMID: 33828523 PMCID: PMC8019783 DOI: 10.3389/fneur.2021.652159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
Given the important functions that glutamate serves in excitatory neurotransmission, understanding the regulation of glutamate in physiological and pathological states is critical to devising novel therapies to treat epilepsy. Exclusive expression of pyruvate carboxylase and glutamine synthetase in astrocytes positions astrocytes as essential regulators of glutamate in the central nervous system (CNS). Additionally, astrocytes can significantly alter the volume of the extracellular space (ECS) in the CNS due to their expression of the bi-directional water channel, aquaporin-4, which are enriched at perivascular endfeet. Rapid ECS shrinkage has been observed following epileptiform activity and can inherently concentrate ions and neurotransmitters including glutamate. This review highlights our emerging knowledge on the various potential contributions of astrocytes to epilepsy, particularly supporting the notion that astrocytes may be involved in seizure initiation via failure of homeostatic responses that lead to increased ambient glutamate. We also review the mechanisms whereby ambient glutamate can influence neuronal excitability, including via generation of the glutamate receptor subunit GluN2B-mediated slow inward currents, as well as indirectly affect neuronal excitability via actions on metabotropic glutamate receptors that can potentiate GluN2B currents and influence neuronal glutamate release probabilities. Additionally, we discuss evidence for upregulation of System x c - , a cystine/glutamate antiporter expressed on astrocytes, in epileptic tissue and changes in expression patterns of glutamate receptors.
Collapse
Affiliation(s)
- Oscar B Alcoreza
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States.,School of Medicine, Virginia Tech Carilion, Roanoke, VA, United States.,Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA, United States
| | - Dipan C Patel
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| | - Bhanu P Tewari
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| | - Harald Sontheimer
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| |
Collapse
|
20
|
Li S, Raychaudhuri S, Lee SA, Brockmann MM, Wang J, Kusick G, Prater C, Syed S, Falahati H, Ramos R, Bartol TM, Hosy E, Watanabe S. Asynchronous release sites align with NMDA receptors in mouse hippocampal synapses. Nat Commun 2021; 12:677. [PMID: 33514725 PMCID: PMC7846561 DOI: 10.1038/s41467-021-21004-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 01/08/2021] [Indexed: 01/30/2023] Open
Abstract
Neurotransmitter is released synchronously and asynchronously following an action potential. Our recent study indicates that the release sites of these two phases are segregated within an active zone, with asynchronous release sites enriched near the center in mouse hippocampal synapses. Here we demonstrate that synchronous and asynchronous release sites are aligned with AMPA receptor and NMDA receptor clusters, respectively. Computational simulations indicate that this spatial and temporal arrangement of release can lead to maximal membrane depolarization through AMPA receptors, alleviating the pore-blocking magnesium leading to greater activation of NMDA receptors. Together, these results suggest that release sites are likely organized to activate NMDA receptors efficiently.
Collapse
Affiliation(s)
- Shuo Li
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Biological and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sumana Raychaudhuri
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Stephen Alexander Lee
- Neurobiology Course, The Marine Biological Laboratory, Woods Hole, MA, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Marisa M Brockmann
- Institute of Neurophysiology, Charité Universitätsmedizin, Berlin, Germany
| | - Jing Wang
- ThermoFisher Scientific, Hillsboro, OR, USA
| | - Grant Kusick
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Christine Prater
- Neurobiology Course, The Marine Biological Laboratory, Woods Hole, MA, USA
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sarah Syed
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Hanieh Falahati
- Neurobiology Course, The Marine Biological Laboratory, Woods Hole, MA, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Raul Ramos
- Neurobiology Course, The Marine Biological Laboratory, Woods Hole, MA, USA
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Tomas M Bartol
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Eric Hosy
- Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Nanoscale co-organization and coactivation of AMPAR, NMDAR, and mGluR at excitatory synapses. Proc Natl Acad Sci U S A 2020; 117:14503-14511. [PMID: 32513712 PMCID: PMC7321977 DOI: 10.1073/pnas.1922563117] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The nanoscale co-organization of neurotransmitter receptors facing presynaptic release sites is a fundamental determinant of their coactivation and of synaptic physiology. At excitatory synapses, how endogenous AMPARs, NMDARs, and mGluRs are co-organized inside the synapse and their respective activation during glutamate release are still unclear. Combining single-molecule superresolution microscopy, electrophysiology, and modeling, we determined the average quantity of each glutamate receptor type, their nanoscale organization, and their respective activation. We observed that NMDARs form a unique cluster mainly at the center of the PSD, while AMPARs segregate in clusters surrounding the NMDARs. mGluR5 presents a different organization and is homogenously dispersed at the synaptic surface. From these results, we build a model predicting the synaptic transmission properties of a unitary synapse, allowing better understanding of synaptic physiology.
Collapse
|
22
|
Yeung JHY, Palpagama TH, Tate WP, Peppercorn K, Waldvogel HJ, Faull RLM, Kwakowsky A. The Acute Effects of Amyloid-Beta 1-42 on Glutamatergic Receptor and Transporter Expression in the Mouse Hippocampus. Front Neurosci 2020; 13:1427. [PMID: 32009891 PMCID: PMC6978747 DOI: 10.3389/fnins.2019.01427] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/17/2019] [Indexed: 01/24/2023] Open
Abstract
Alzheimer’s disease (AD) is the leading type of dementia worldwide. Despite an increasing burden of disease due to a rapidly aging population, there is still a lack of complete understanding of the precise pathological mechanisms which drive its progression. Glutamate is the main excitatory neurotransmitter in the brain and plays an essential role in the normal function and excitability of neuronal networks. While previous studies have shown alterations in the function of the glutamatergic system in AD, the underlying etiology of beta amyloid (Aβ1–42) induced changes has not been explored. Here we have investigated the acute effects of stereotaxic hippocampal Aβ1–42 injection on specific glutamatergic receptors and transporters in the mouse hippocampus, using immunohistochemistry and confocal microscopy 3 days after Aβ1–42 injection in aged male C57BL/6 mice, before the onset of neuronal cell death. We show that acute injection of Aβ1–42 is sufficient to induce cognitive deficits 3 days post-injection. We also report no significant changes in glutamate receptor subunits GluA1, GluA2, VGluT1, and VGluT2 in response to acute injection of Aβ1–42 when compared with the ACSF-vehicle injected mice. However, we observed increased expression in the DG hilus and ventral stratum (str.) granulosum, CA3 str. radiatum and str. oriens, and CA1 str. radiatum of the GluN1 subunit, and increased expression within the CA3 str. radiatum and decreased expression within the DG str. granulosum of the GluN2A subunit in Aβ1–42 injected mice compared to NC, and a similar trend observed when compared to ACSF-injected mice. We also observed alterations in expression patterns of glutamatergic receptor subunits and transporters within specific layers of hippocampal subregions in response to a microinjection stimulus. These findings indicate that the pathological alterations in the glutamatergic system observed in AD are likely to be partially a result of both acute and chronic exposure to Aβ1–42 and implies a much more complex circuit mechanism associated with glutamatergic dysfunction than simply glutamate-mediated excitotoxic neuronal death.
Collapse
Affiliation(s)
- Jason H Y Yeung
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Warren P Tate
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Katie Peppercorn
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
23
|
Masilamoni GJ, Smith Y. Group I metabotropic glutamate receptors in the primate motor thalamus: subsynaptic association with cortical and sub-cortical glutamatergic afferents. Brain Struct Funct 2019; 224:2787-2804. [PMID: 31422483 DOI: 10.1007/s00429-019-01937-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022]
Abstract
Preclinical evidence indicates that mGluR5 is a potential therapeutic target for Parkinson's disease and L-DOPA-induced dyskinesia. However, the mechanisms through which these therapeutic benefits are mediated remain poorly understood. Although the regulatory role of mGluR5 on glutamatergic transmission has been examined in various basal ganglia nuclei, very little is known about the localization and function of mGluR5 in the ventral motor and intralaminar thalamic nuclei, the main targets of basal ganglia output in mammals. Thus, we used immuno-electron microscopy to map the cellular and subcellular localization of group I mGluRs (mGluR1a and mGluR5) in the ventral motor and caudal intralaminar thalamic nuclei in rhesus monkeys. Furthermore, using double immuno-electron microscopy, we examined the subsynaptic localization of mGluR5 in relation to cortical and sub-cortical glutamatergic afferents. Four major conclusions can be drawn from these data. First, mGluR1a and mGluR5 are expressed postsynaptically on the plasma membrane of dendrites of projection neurons and GABAergic interneurons in the basal ganglia- and cerebellar-receiving regions of the ventral motor thalamus and in CM. Second, the plasma membrane-bound mGluR5 immunoreactivity is preferentially expressed perisynaptically at the edges of cortical and sub-cortical glutamatergic afferents. Third, the mGluR5 immunoreactivity is more strongly expressed in the lateral than the medial tiers of CM, suggesting a preferential association with thalamocortical over thalamostriatal neurons in the primate CM. Overall, mGluR5 is located to subserve powerful modulatory role of cortical and subcortical glutamatergic transmission in the primate ventral motor thalamus and CM.
Collapse
Affiliation(s)
- Gunasingh Jeyaraj Masilamoni
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30329, USA. .,Udall Center of Excellence for Parkinson's Disease, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Udall Center of Excellence for Parkinson's Disease, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
24
|
Reiner A, Levitz J. Glutamatergic Signaling in the Central Nervous System: Ionotropic and Metabotropic Receptors in Concert. Neuron 2019; 98:1080-1098. [PMID: 29953871 DOI: 10.1016/j.neuron.2018.05.018] [Citation(s) in RCA: 399] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/19/2018] [Accepted: 05/10/2018] [Indexed: 12/28/2022]
Abstract
Glutamate serves as both the mammalian brain's primary excitatory neurotransmitter and as a key neuromodulator to control synapse and circuit function over a wide range of spatial and temporal scales. This functional diversity is decoded by two receptor families: ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs). The challenges posed by the complexity and physiological importance of each of these subtypes has limited our appreciation and understanding of how these receptors work in concert. In this review, by comparing both receptor families with a focus on their crosstalk, we argue for a more holistic understanding of neural glutamate signaling.
Collapse
Affiliation(s)
- Andreas Reiner
- Department of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany.
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Chen JH, Blanpied TA, Tang AH. Quantification of trans-synaptic protein alignment: A data analysis case for single-molecule localization microscopy. Methods 2019; 174:72-80. [PMID: 31325491 DOI: 10.1016/j.ymeth.2019.07.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/14/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
Nanoscale distribution of proteins and their relative positioning within a defined subcellular region are key to their physiological functions. Thanks to the super-resolution imaging methods, especially single-molecule localization microscopy (SMLM), mapping the three-dimensional distribution of multiple proteins has been easier and more efficient than ever. Nevertheless, in spite of the many tools available for efficient localization detection and image rendering, it has been a challenge to quantitatively analyze the 3D distribution and relative positioning of proteins in these SMLM data. Here, using heterogeneously distributed synaptic proteins as examples, we describe in detail a series of analytical methods including detection of nanoscale density clusters, quantification of the trans-synaptic alignment between these protein densities, and automatic en face projection and averaging. These analyses were performed within customized Matlab routines and we make the full scripts available. The concepts behind these analytical methods and the scripts can be adapted for quantitative analysis of spatial organization of other macromolecular complexes.
Collapse
Affiliation(s)
- Jia-Hui Chen
- CAS Key Laboratory of Brain Function and Disease and Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Thomas A Blanpied
- Program in Neuroscience and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ai-Hui Tang
- CAS Key Laboratory of Brain Function and Disease and Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
26
|
Galvan A, Raper J, Hu X, Paré JF, Bonaventura J, Richie CT, Michaelides M, Mueller SAL, Roseboom PH, Oler JA, Kalin NH, Hall RA, Smith Y. Ultrastructural localization of DREADDs in monkeys. Eur J Neurosci 2019; 50:2801-2813. [PMID: 31063250 DOI: 10.1111/ejn.14429] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/15/2019] [Accepted: 04/23/2019] [Indexed: 01/01/2023]
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) are extensively used to modulate neuronal activity in rodents, but their use in primates remains limited. An essential need that remains is the demonstration that DREADDs are efficiently expressed on the plasma membrane of primate neurons. To address this issue, electron microscopy immunogold was used to determine the subcellular localization of the AAV vector-induced DREADDs hM4Di and hM3Dq fused to different tags in various brain areas of rhesus monkeys and mice. When hM4Di was fused to mCherry, the immunogold labelling was mostly confined to the intracellular space, and poorly expressed at the plasma membrane in monkey dendrites. In contrast, the hM4Di-mCherry labelling was mostly localized to the dendritic plasma membrane in mouse neurons, suggesting species differences in the plasma membrane expression of these exogenous proteins. The lack of hM4Di plasma membrane expression may limit the functional effects of systemic administration of DREADD-actuators in monkey neurons. Removing the mCherry and fusing of hM4Di with the haemagglutinin (HA) tag resulted in strong neuronal plasma membrane immunogold labelling in both monkeys and mice neurons. Finally, hM3Dq-mCherry was expressed mostly at the plasma membrane in monkey neurons, indicating that the fusion of mCherry with hM3Dq does not hamper membrane incorporation of this specific DREADD. Our results suggest that the pattern of ultrastructural expression of DREADDs in monkey neurons depends on the DREADD/tag combination. Therefore, a preliminary characterization of plasma membrane expression of specific DREADD/tag combinations is recommended when using chemogenetic approaches in primates.
Collapse
Affiliation(s)
- Adriana Galvan
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Jessica Raper
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Xing Hu
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Jean-François Paré
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Jordi Bonaventura
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse (NIDA), Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Christopher T Richie
- Genetic Engineering and Viral Vector Core, National Institute on Drug Abuse (NIDA), Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse (NIDA), Intramural Research Program, National Institutes of Health, Baltimore, Maryland.,Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sascha A L Mueller
- Department of Psychiatry, University of Wisconsin, Madison, Wisconsin.,Molecular and Cellular Pharmacology Training Program, University of Wisconsin, Madison, Wisconsin
| | | | - Jonathan A Oler
- Department of Psychiatry, University of Wisconsin, Madison, Wisconsin
| | - Ned H Kalin
- Department of Psychiatry, University of Wisconsin, Madison, Wisconsin.,Molecular and Cellular Pharmacology Training Program, University of Wisconsin, Madison, Wisconsin.,Wisconsin National Primate Research Center, Madison, Wisconsin
| | - Randy A Hall
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
27
|
Naito R, Kassai H, Sakai Y, Schönherr S, Fukaya M, Schwarzer C, Sakagami H, Nakao K, Aiba A, Ferraguti F. New Features on the Expression and Trafficking of mGluR1 Splice Variants Exposed by Two Novel Mutant Mouse Lines. Front Mol Neurosci 2018; 11:439. [PMID: 30559646 PMCID: PMC6287019 DOI: 10.3389/fnmol.2018.00439] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/13/2018] [Indexed: 01/08/2023] Open
Abstract
Metabotropic glutamate receptors (mGluRs) couple to G-proteins to modulate slow synaptic transmission via intracellular second messengers. The first cloned mGluR, mGluR1, regulates motor coordination, synaptic plasticity and synapse elimination. mGluR1 undergoes alternative splicing giving rise to four translated variants that differ in their intracellular C-terminal domains. Our current knowledge about mGluR1 relates almost entirely to the long mGluR1α isoform, whereas little is known about the other shorter variants. To study the expression of mGluR1γ, we have generated by means of the CRISPR/Cas9 system a new knock-in (KI) mouse line in which the C-terminus of this variant carries two short tags. Using this mouse line, we could establish that mGluR1γ is either untranslated or in amounts that are undetectable in the mouse cerebellum, indicating that only mGluR1α and mGluR1β are present and active at cerebellar synapses. The trafficking and function of mGluR1 appear strongly influenced by adaptor proteins such as long Homers that bind to the C-terminus of mGluR1α. We generated a second transgenic (Tg) mouse line in which mGluR1α carries a point mutation in its Homer binding domain and studied whether disruption of this interaction influenced mGluR1 subcellular localization at cerebellar parallel fiber (PF)-Purkinje cell (PC) synapses by means of the freeze-fracture replica immunolabeling technique. These Tg animals did not show any overt behavioral phenotype, and despite the typical mGluR1 perisynaptic distribution was not significantly changed, we observed a higher probability of intrasynaptic diffusion suggesting that long Homers regulate the lateral mobility of mGluR1. We extended our ultrastructural analysis to other mouse lines in which only one mGluR1 variant was reintroduced in PC of mGluR1-knock out (KO) mice. This work revealed that mGluR1α preferentially accumulates closer to the edge of the postsynaptic density (PSD), whereas mGluR1β has a less pronounced perijunctional distribution and, in the absence of mGluR1α, its trafficking to the plasma membrane is impaired with an accumulation in intracellular organelles. In conclusion, our study sets several firm points on largely disputed matters, namely expression of mGluR1γ and role of the C-terminal domain of mGluR1 splice variants on their perisynaptic clustering.
Collapse
Affiliation(s)
- Rika Naito
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hidetoshi Kassai
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Genetics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yusuke Sakai
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sabine Schönherr
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Masahiro Fukaya
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazuki Nakao
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Atsu Aiba
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Genetics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Dillon J, Holden-Dye L, O'Connor V. Yeast two-hybrid screening identifies MPZ-1 and PTP-1 as candidate scaffolding proteins of metabotropic glutamate receptors in Caenorhabditis elegans. INVERTEBRATE NEUROSCIENCE 2018; 18:16. [PMID: 30417267 DOI: 10.1007/s10158-018-0218-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/19/2018] [Indexed: 10/27/2022]
Abstract
The metabotropic glutamate receptors (mGluRs) are a class of G-protein-coupled receptor that undergo extensive interactions with scaffolding proteins, and this is intrinsic to their function as an important group of neuromodulators at glutamatergic synapses. The Caenorhabditis elegans nervous system expresses three metabotropic glutamate receptors, MGL-1, MGL-2 and MGL-3. Relatively little is known about how the function and signalling of these receptors is organised in C. elegans. To identify proteins that scaffold the MGL-1 receptor, we have conducted a yeast two-hybrid screen. Three of the interacting proteins, MPZ-1, NRFL-1 and PTP-1, displayed motifs characteristic of mammalian mGluR scaffolding proteins. Using cellular co-expression criterion, we show mpz-1 and ptp-1 exhibited overlapping expression patterns with subsets of mgl-1 neurons. This included neurones in the pharyngeal nervous system that control the feeding organ of the worm. The mGluR agonist L-CCG-I inhibits the activity of this network in wild-type worms, in an MGL-1 and dose-dependent manner. We utilised L-CCG-I to identify if MGL-1 function was disrupted in mutants with deletions in the mpz-1 gene. The mpz-1 mutants displayed a largely wild-type response to L-CCG-I, suggesting MGL-1 signalling is not overtly disrupted consistent with a non-obligatory modulatory function in receptor scaffolding. The selectivity of the protein interactions and overlapping expression identified here warrant further investigation of the functional significance of scaffolding of metabotropic glutamate receptor function.
Collapse
Affiliation(s)
- James Dillon
- Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| | - Lindy Holden-Dye
- Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Vincent O'Connor
- Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
29
|
Patriarchi T, Buonarati OR, Hell JW. Postsynaptic localization and regulation of AMPA receptors and Cav1.2 by β2 adrenergic receptor/PKA and Ca 2+/CaMKII signaling. EMBO J 2018; 37:e99771. [PMID: 30249603 PMCID: PMC6187224 DOI: 10.15252/embj.201899771] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/25/2018] [Accepted: 08/17/2018] [Indexed: 11/09/2022] Open
Abstract
The synapse transmits, processes, and stores data within its tiny space. Effective and specific signaling requires precise alignment of the relevant components. This review examines current insights into mechanisms of AMPAR and NMDAR localization by PSD-95 and their spatial distribution at postsynaptic sites to illuminate the structural and functional framework of postsynaptic signaling. It subsequently delineates how β2 adrenergic receptor (β2 AR) signaling via adenylyl cyclase and the cAMP-dependent protein kinase PKA is organized within nanodomains. Here, we discuss targeting of β2 AR, adenylyl cyclase, and PKA to defined signaling complexes at postsynaptic sites, i.e., AMPARs and the L-type Ca2+ channel Cav1.2, and other subcellular surface localizations, the role of A kinase anchor proteins, the physiological relevance of the spatial restriction of corresponding signaling, and their interplay with signal transduction by the Ca2+- and calmodulin-dependent kinase CaMKII How localized and specific signaling by cAMP occurs is a central cellular question. The dendritic spine constitutes an ideal paradigm for elucidating the dimensions of spatially restricted signaling because of their small size and defined protein composition.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Signaling/physiology
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cyclic AMP-Dependent Protein Kinases/genetics
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Humans
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Synapses/genetics
- Synapses/metabolism
Collapse
Affiliation(s)
- Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | | | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
30
|
Scheefhals N, MacGillavry HD. Functional organization of postsynaptic glutamate receptors. Mol Cell Neurosci 2018; 91:82-94. [PMID: 29777761 PMCID: PMC6276983 DOI: 10.1016/j.mcn.2018.05.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/16/2018] [Accepted: 05/07/2018] [Indexed: 01/28/2023] Open
Abstract
Glutamate receptors are the most abundant excitatory neurotransmitter receptors in the brain, responsible for mediating the vast majority of excitatory transmission in neuronal networks. The AMPA- and NMDA-type ionotropic glutamate receptors (iGluRs) are ligand-gated ion channels that mediate the fast synaptic responses, while metabotropic glutamate receptors (mGluRs) are coupled to downstream signaling cascades that act on much slower timescales. These functionally distinct receptor sub-types are co-expressed at individual synapses, allowing for the precise temporal modulation of postsynaptic excitability and plasticity. Intriguingly, these receptors are differentially distributed with respect to the presynaptic release site. While iGluRs are enriched in the core of the synapse directly opposing the release site, mGluRs reside preferentially at the border of the synapse. As such, to understand the differential contribution of these receptors to synaptic transmission, it is important to not only consider their signaling properties, but also the mechanisms that control the spatial segregation of these receptor types within synapses. In this review, we will focus on the mechanisms that control the organization of glutamate receptors at the postsynaptic membrane with respect to the release site, and discuss how this organization could regulate synapse physiology.
Collapse
Affiliation(s)
- Nicky Scheefhals
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Harold D MacGillavry
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands.
| |
Collapse
|
31
|
Pál B. Involvement of extrasynaptic glutamate in physiological and pathophysiological changes of neuronal excitability. Cell Mol Life Sci 2018; 75:2917-2949. [PMID: 29766217 PMCID: PMC11105518 DOI: 10.1007/s00018-018-2837-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Glutamate is the most abundant neurotransmitter of the central nervous system, as the majority of neurons use glutamate as neurotransmitter. It is also well known that this neurotransmitter is not restricted to synaptic clefts, but found in the extrasynaptic regions as ambient glutamate. Extrasynaptic glutamate originates from spillover of synaptic release, as well as from astrocytes and microglia. Its concentration is magnitudes lower than in the synaptic cleft, but receptors responding to it have higher affinity for it. Extrasynaptic glutamate receptors can be found in neuronal somatodendritic location, on astroglia, oligodendrocytes or microglia. Activation of them leads to changes of neuronal excitability with different amplitude and kinetics. Extrasynaptic glutamate is taken up by neurons and astrocytes mostly via EAAT transporters, and astrocytes, in turn metabolize it to glutamine. Extrasynaptic glutamate is involved in several physiological phenomena of the central nervous system. It regulates neuronal excitability and synaptic strength by involving astroglia; contributing to learning and memory formation, neurosecretory and neuromodulatory mechanisms, as well as sleep homeostasis.The extrasynaptic glutamatergic system is affected in several brain pathologies related to excitotoxicity, neurodegeneration or neuroinflammation. Being present in dementias, neurodegenerative and neuropsychiatric diseases or tumor invasion in a seemingly uniform way, the system possibly provides a common component of their pathogenesis. Although parts of the system are extensively discussed by several recent reviews, in this review I attempt to summarize physiological actions of the extrasynaptic glutamate on neuronal excitability and provide a brief insight to its pathology for basic understanding of the topic.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, Debrecen, 4012, Hungary.
| |
Collapse
|
32
|
Feng X, Bader BM, Yang F, Segura M, Schultz L, Schröder OHU, Rolfs A, Luo J. Improvement of impaired electrical activity in NPC1 mutant cortical neurons upon DHPG stimulation detected by micro-electrode array. Brain Res 2018; 1694:87-93. [PMID: 29753706 DOI: 10.1016/j.brainres.2018.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/20/2018] [Accepted: 05/09/2018] [Indexed: 12/20/2022]
Abstract
Niemann-Pick Type C1 (NPC1) disease is an autosomal recessive neurodegenerative disease characterized by an excessive accumulation of unesterified cholesterol in late endosomes/lysosomes. Patients with NPC1 disease show a series of symptoms in neuropathology, including a gradually increased loss of motor control and seizures. However, mechanism of the neurological manifestations in NPC1 disease is not fully understood yet. In this study, we utilized the micro-electrode array (MEA) to analyze the spontaneous extracellular electrical activity in cultivated cortical neurons of the NPC1 mutant (NPC1-/-) mouse. Our results show a decrease of the spontaneous electrical activity in NPC1-/- neuronal network when compared to wild type neurons, as indicated by the decreased spike rate, burst rate, event rate, and the increased burst period and event period. Application of 3,5-dihydroxyphenylglycine (DHPG), a specific agonist of group I metabotropic glutamate receptors, improved the electrical activity of the NPC1-/- neuronal network, suggesting that DHPG can be used as a potential therapeutic strategy for recovery of the electrical activity in NPC1 disease.
Collapse
Affiliation(s)
- Xiao Feng
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Benjamin M Bader
- NeuroProof GmbH, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany
| | - Fan Yang
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Monica Segura
- NeuroProof GmbH, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany
| | - Luise Schultz
- NeuroProof GmbH, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany
| | - Olaf H-U Schröder
- NeuroProof GmbH, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany
| | - Arndt Rolfs
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Jiankai Luo
- Albrecht-Kossel-Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Straße 20, 18147 Rostock, Germany; Centre for Transdisciplinary Neuroscience Rostock, Rostock University Medical Center, Gehlsheimer Straße 20, 18147 Rostock, Germany.
| |
Collapse
|
33
|
Yoshikura N, Kimura A, Fukata M, Fukata Y, Yokoi N, Harada N, Hayashi Y, Inuzuka T, Shimohata T. Long-term clinical follow-up of a patient with non-paraneoplastic cerebellar ataxia associated with anti-mGluR1 autoantibodies. J Neuroimmunol 2018; 319:63-67. [PMID: 29685291 DOI: 10.1016/j.jneuroim.2018.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/26/2018] [Accepted: 04/02/2018] [Indexed: 02/03/2023]
Abstract
The clinical features of cerebellar ataxia associated with anti-metabotropic glutamate receptor 1 (mGluR1) autoantibodies, a rare autoimmune-mediated cerebellar ataxia, remain to be elucidated. Here, we describe a patient with non-paraneoplastic cerebellar ataxia associated with anti-mGluR1 autoantibodies, who was followed up over 5 years. She presented with relapses and remissions of subacute progressive cerebellar ataxia that were responsive to immunotherapy. Although serum anti-mGluR1 autoantibodies were continuously detected and cerebellar atrophy gradually progressed, repeated intravenous immunoglobulin therapy and oral immunosuppressants ensured cerebellar ataxia remained at almost the same level during the observation period.
Collapse
Affiliation(s)
- Nobuaki Yoshikura
- Department of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akio Kimura
- Department of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan.
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Norihiko Yokoi
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Naoko Harada
- Department of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yuichi Hayashi
- Department of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takashi Inuzuka
- Department of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takayoshi Shimohata
- Department of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
34
|
Narushima M. Comparison of the role of metabotropic glutamate receptor subtype 1 in developmental refinement of neuronal connectivity between the cerebellum and the sensory thalamus. Neurosci Res 2017; 129:24-31. [PMID: 28711710 DOI: 10.1016/j.neures.2017.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/15/2017] [Accepted: 06/15/2017] [Indexed: 11/30/2022]
Abstract
Developmental refinement of neuronal connectivity is crucial for proper brain function. In the early phase of development, input fibers arrive at their target areas guided by specific molecular cues and form abundant immature synapses. Then, functionally important synapses are preserved and strengthened by neural activity while unnecessary synapses are eliminated. Afferent synapses in the sensory thalamus, such as from retina to lateral geniculate nucleus, and climbing fiber (CF)-Purkinje cell (PC) synapses in the cerebellum are valuable models for studying this developmental refinement of synaptic connectivity because only a limited number of input fibers innervate a given postsynaptic thalamocortical (TC) neuron or PC. The metabotropic glutamate receptor subtype 1 (mGluR1) is required for the refinement of both afferent-TC neuron and CF-PC synapses. However, mGluR1 functions differently at these synapses. While mGluR1 is critical for elimination of surplus CF-PC synapses in the cerebellum, retinogeniculate synapses require mGluR1 for maintenance of mature connectivity.
Collapse
Affiliation(s)
- Madoka Narushima
- Department of Physiology (I), School of Medicine, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan.
| |
Collapse
|
35
|
Meera P, Pulst S, Otis T. A positive feedback loop linking enhanced mGluR function and basal calcium in spinocerebellar ataxia type 2. eLife 2017; 6. [PMID: 28518055 PMCID: PMC5444899 DOI: 10.7554/elife.26377] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/16/2017] [Indexed: 12/11/2022] Open
Abstract
Metabotropic glutamate receptor 1 (mGluR1) function in Purkinje neurons (PNs) is essential for cerebellar development and for motor learning and altered mGluR1 signaling causes ataxia. Downstream of mGluR1, dysregulation of calcium homeostasis has been hypothesized as a key pathological event in genetic forms of ataxia but the underlying mechanisms remain unclear. We find in a spinocerebellar ataxia type 2 (SCA2) mouse model that calcium homeostasis in PNs is disturbed across a broad range of physiological conditions. At parallel fiber synapses, mGluR1-mediated excitatory postsynaptic currents (EPSCs) and associated calcium transients are increased and prolonged in SCA2 PNs. In SCA2 PNs, enhanced mGluR1 function is prevented by buffering [Ca2+] at normal resting levels while in wildtype PNs mGluR1 EPSCs are enhanced by elevated [Ca2+]. These findings demonstrate a deleterious positive feedback loop involving elevated intracellular calcium and enhanced mGluR1 function, a mechanism likely to contribute to PN dysfunction and loss in SCA2. DOI:http://dx.doi.org/10.7554/eLife.26377.001
Collapse
Affiliation(s)
- Pratap Meera
- Department of Neurobiology, Geffen School of Medicine, University of California, Los Angeles, United States
| | - Stefan Pulst
- Department of Neurology, University of Utah, Salt Lake, United States
| | - Thomas Otis
- Department of Neurobiology, Geffen School of Medicine, University of California, Los Angeles, United States.,Neuroscience, Ophthalmology, and Rare Diseases, Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| |
Collapse
|
36
|
Hoxha E, Tempia F, Lippiello P, Miniaci MC. Modulation, Plasticity and Pathophysiology of the Parallel Fiber-Purkinje Cell Synapse. Front Synaptic Neurosci 2016; 8:35. [PMID: 27857688 PMCID: PMC5093118 DOI: 10.3389/fnsyn.2016.00035] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 10/19/2016] [Indexed: 12/24/2022] Open
Abstract
The parallel fiber-Purkinje cell (PF-PC) synapse represents the point of maximal signal divergence in the cerebellar cortex with an estimated number of about 60 billion synaptic contacts in the rat and 100,000 billions in humans. At the same time, the Purkinje cell dendritic tree is a site of remarkable convergence of more than 100,000 parallel fiber synapses. Parallel fiber activity generates fast postsynaptic currents via α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, and slower signals, mediated by mGlu1 receptors, resulting in Purkinje cell depolarization accompanied by sharp calcium elevation within dendritic regions. Long-term depression (LTD) and long-term potentiation (LTP) have been widely described for the PF-PC synapse and have been proposed as mechanisms for motor learning. The mechanisms of induction for LTP and LTD involve different signaling mechanisms within the presynaptic terminal and/or at the postsynaptic site, promoting enduring modification in the neurotransmitter release and change in responsiveness to the neurotransmitter. The PF-PC synapse is finely modulated by several neurotransmitters, including serotonin, noradrenaline and acetylcholine. The ability of these neuromodulators to gate LTP and LTD at the PF-PC synapse could, at least in part, explain their effect on cerebellar-dependent learning and memory paradigms. Overall, these findings have important implications for understanding the cerebellar involvement in a series of pathological conditions, ranging from ataxia to autism. For example, PF-PC synapse dysfunctions have been identified in several murine models of spino-cerebellar ataxia (SCA) types 1, 3, 5 and 27. In some cases, the defect is specific for the AMPA receptor signaling (SCA27), while in others the mGlu1 pathway is affected (SCA1, 3, 5). Interestingly, the PF-PC synapse has been shown to be hyper-functional in a mutant mouse model of autism spectrum disorder, with a selective deletion of Pten in Purkinje cells. However, the full range of methodological approaches, that allowed the discovery of the physiological principles of PF-PC synapse function, has not yet been completely exploited to investigate the pathophysiological mechanisms of diseases involving the cerebellum. We, therefore, propose to extend the spectrum of experimental investigations to tackle this problem.
Collapse
Affiliation(s)
- Eriola Hoxha
- Neuroscience Institute Cavalieri Ottolenghi (NICO) and Department of Neuroscience, University of TorinoTorino, Italy
| | - Filippo Tempia
- Neuroscience Institute Cavalieri Ottolenghi (NICO) and Department of Neuroscience, University of TorinoTorino, Italy
| | | | | |
Collapse
|
37
|
Olmo IG, Ferreira-Vieira TH, Ribeiro FM. Dissecting the Signaling Pathways Involved in the Crosstalk between Metabotropic Glutamate 5 and Cannabinoid Type 1 Receptors. Mol Pharmacol 2016; 90:609-619. [PMID: 27338080 DOI: 10.1124/mol.116.104372] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/16/2016] [Indexed: 02/06/2023] Open
Abstract
The metabotropic glutamate 5 receptor and the cannabinoid type 1 receptor are G protein-coupled receptors that are widely expressed in the central nervous system. Metabotropic glutamate 5 receptors, present at the postsynaptic site, are coupled to Gαq/11 proteins and display an excitatory response upon activation, whereas the cannabinoid type 1 receptor, mainly present at presynaptic terminals, is coupled to the Gi/o protein and triggers an inhibitory response. Recent studies suggest that the glutamatergic and endocannabinoid systems exhibit a functional interaction to modulate several neural processes. In this review, we discuss possible mechanisms involved in this crosstalk and its relationship with physiologic and pathologic conditions, including nociception, addiction, and fragile X syndrome.
Collapse
Affiliation(s)
- Isabella G Olmo
- Department of Biochemistry and Immunology, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Talita H Ferreira-Vieira
- Department of Biochemistry and Immunology, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabiola M Ribeiro
- Department of Biochemistry and Immunology, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
38
|
Pittaluga A. Presynaptic Release-Regulating mGlu1 Receptors in Central Nervous System. Front Pharmacol 2016; 7:295. [PMID: 27630571 PMCID: PMC5006178 DOI: 10.3389/fphar.2016.00295] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/22/2016] [Indexed: 01/14/2023] Open
Abstract
Group I metabotropic glutamate (mGlu) receptors consists of mGlu1 and mGlu5 receptor subtypes. These receptors are widely distributed in the central nervous system (CNS), where they preferentially mediate facilitatory signaling in neurones and glial cells, mainly by favoring phospholipase (PLC) translocation. Based on the literature so far available, group I Metabotropic glutamate receptors (mGluRs) are preferentially expressed at the postsynaptic side of chemical synapsis, where they participate in the progression of the chemical stimulus. Studies, however, have shown the presence of these receptors also at the presynaptic level, where they exert several functions, including the modulation of transmitter exocytosis. Presynaptic Group I mGluRs can be both autoreceptors regulating release of glutamate and heteroreceptors regulating the release of various transmitters, including GABA, dopamine, noradrenaline, and acetylcholine. While the existence of presynaptic release-regulating mGlu5 receptors is largely recognized, the possibility that mGlu1 receptors also are present at this level has been a matter of discussion for a long time. A large body of evidence published in the last decade, however, supports this notion. This review aims at revisiting the data from in vitro studies concerning the existence and the role of release-regulating mGlu1 receptors presynaptically located in nerve terminals isolated from selected regions of the CNS. The functional interaction linking mGlu5 and mGlu1 receptor subtypes at nerve terminals and their relative contributions as modulators of central transmission will also be discussed. We apologize in advance for omission in our coverage of the existing literature.
Collapse
Affiliation(s)
- Anna Pittaluga
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of GenoaGenoa, Italy
- Center of Excellence for Biomedical Research, University of GenoaGenoa, Italy
| |
Collapse
|
39
|
Li Q, Cui P, Miao Y, Gao F, Li XY, Qian WJ, Jiang SX, Wu N, Sun XH, Wang Z. Activation of group I metabotropic glutamate receptors regulates the excitability of rat retinal ganglion cells by suppressing Kir and I h. Brain Struct Funct 2016; 222:813-830. [PMID: 27306787 DOI: 10.1007/s00429-016-1248-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/05/2016] [Indexed: 10/21/2022]
Abstract
Group I metabotropic glutamate receptor (mGluR I) activation exerts a slow postsynaptic excitatory effect in the CNS. Here, the issues of whether and how this receptor is involved in regulating retinal ganglion cell (RGC) excitability were investigated in retinal slices using patch-clamp techniques. Under physiological conditions, RGCs displayed spontaneous firing. Extracellular application of LY367385 (10 µM)/MPEP (10 µM), selective mGluR1 and mGluR5 antagonists, respectively, significantly reduced the firing frequency, suggesting that glutamate endogenously released from bipolar cells constantly modulates RGC firing. DHPG (10 µM), an mGluR I agonist, significantly increased the firing and caused depolarization of the cells, which were reversed by LY367385, but not by MPEP, suggesting the involvement of the mGluR1 subtype. Intracellular Ca2+-dependent PI-PLC/PKC and calcium/calmodulin-dependent protein kinase II (CaMKII) signaling pathways mediated the DHPG-induced effects. In the presence of cocktail synaptic blockers (CNQX, D-AP5, bicuculline, and strychnine), which terminated the spontaneous firing in both ON and OFF RGCs, DHPG still induced depolarization and triggered the cells to fire. The DHPG-induced depolarization could not be blocked by TTX. In contrast, Ba2+, an inwardly rectifying potassium channel (Kir) blocker, and Cs+ and ZD7288, hyperpolarization-activated cation channel (I h) blockers, mimicked the effect of DHPG. Furthermore, in the presence of Ba2+/ZD7288, DHPG did not show further effects. Moreover, Kir and I h currents could be recorded in RGCs, and extracellular application of DHPG indeed suppressed these currents. Our results suggest that activation of mGluR I regulates the excitability of rat RGCs by inhibiting Kir and I h.
Collapse
Affiliation(s)
- Qian Li
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China.,Institute of Neurobiology, Fudan University, Shanghai, 200032, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Peng Cui
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China.,Institute of Neurobiology, Fudan University, Shanghai, 200032, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yanying Miao
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China.,Institute of Neurobiology, Fudan University, Shanghai, 200032, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Feng Gao
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China.,Eye & ENT Hospital, Fudan University, Shanghai, 200031, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xue-Yan Li
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China.,Institute of Neurobiology, Fudan University, Shanghai, 200032, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Wen-Jing Qian
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China.,Institute of Neurobiology, Fudan University, Shanghai, 200032, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Shu-Xia Jiang
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China.,Institute of Neurobiology, Fudan University, Shanghai, 200032, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Na Wu
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China.,Eye & ENT Hospital, Fudan University, Shanghai, 200031, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xing-Huai Sun
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China. .,Eye & ENT Hospital, Fudan University, Shanghai, 200031, China. .,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, China. .,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| | - Zhongfeng Wang
- Institutes of Brain Science, Fudan University, 138 Yixueyuan Rd, Shanghai, 200032, China. .,Eye & ENT Hospital, Fudan University, Shanghai, 200031, China. .,Institute of Neurobiology, Fudan University, Shanghai, 200032, China. .,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, 200031, China. .,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
40
|
Metabotropic glutamate receptors are involved in the detection of IMP and L-amino acids by mouse taste sensory cells. Neuroscience 2015; 316:94-108. [PMID: 26701297 DOI: 10.1016/j.neuroscience.2015.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/03/2015] [Accepted: 12/04/2015] [Indexed: 11/22/2022]
Abstract
G-protein-coupled receptors are thought to be involved in the detection of umami and L-amino acid taste. These include the heterodimer taste receptor type 1 member 1 (T1r1)+taste receptor type 1 member 3 (T1r3), taste and brain variants of mGluR4 and mGluR1, and calcium sensors. While several studies suggest T1r1+T1r3 is a broadly tuned lLamino acid receptor, little is known about the function of metabotropic glutamate receptors (mGluRs) in L-amino acid taste transduction. Calcium imaging of isolated taste sensory cells (TSCs) of T1r3-GFP and T1r3 knock-out (T1r3 KO) mice was performed using the ratiometric dye Fura 2 AM to investigate the role of different mGluRs in detecting various L-amino acids and inosine 5' monophosphate (IMP). Using agonists selective for various mGluRs such as (RS)-3,5-dihydroxyphenylglycine (DHPG) (an mGluR1 agonist) and L-(+)-2-amino-4-phosphonobutyric acid (l-AP4) (an mGluR4 agonist), we evaluated TSCs to determine if they might respond to these agonists, IMP, and three L-amino acids (monopotassium L-glutamate, L-serine and L-arginine). Additionally, we used selective antagonists against different mGluRs such as (RS)-L-aminoindan-1,5-dicarboxylic acid (AIDA) (an mGluR1 antagonist), and (RS)-α-methylserine-O-phosphate (MSOP) (an mGluR4 antagonist) to determine if they can block responses elicited by these L-amino acids and IMP. We found that L-amino acid- and IMP-responsive cells also responded to each agonist. Antagonists for mGluR4 and mGluR1 significantly blocked the responses elicited by IMP and each of the L-amino acids. Collectively, these data provide evidence for the involvement of taste and brain variants of mGluR1 and mGluR4 in L-amino acid and IMP taste responses in mice, and support the concept that multiple receptors contribute to IMP and L-amino acid taste.
Collapse
|
41
|
Allam SL, Bouteiller JMC, Hu EY, Ambert N, Greget R, Bischoff S, Baudry M, Berger TW. Synaptic Efficacy as a Function of Ionotropic Receptor Distribution: A Computational Study. PLoS One 2015; 10:e0140333. [PMID: 26480028 PMCID: PMC4610697 DOI: 10.1371/journal.pone.0140333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 09/24/2015] [Indexed: 11/22/2022] Open
Abstract
Glutamatergic synapses are the most prevalent functional elements of information processing in the brain. Changes in pre-synaptic activity and in the function of various post-synaptic elements contribute to generate a large variety of synaptic responses. Previous studies have explored postsynaptic factors responsible for regulating synaptic strength variations, but have given far less importance to synaptic geometry, and more specifically to the subcellular distribution of ionotropic receptors. We analyzed the functional effects resulting from changing the subsynaptic localization of ionotropic receptors by using a hippocampal synaptic computational framework. The present study was performed using the EONS (Elementary Objects of the Nervous System) synaptic modeling platform, which was specifically developed to explore the roles of subsynaptic elements as well as their interactions, and that of synaptic geometry. More specifically, we determined the effects of changing the localization of ionotropic receptors relative to the presynaptic glutamate release site, on synaptic efficacy and its variations following single pulse and paired-pulse stimulation protocols. The results indicate that changes in synaptic geometry do have consequences on synaptic efficacy and its dynamics.
Collapse
Affiliation(s)
- Sushmita L. Allam
- Center for Neural Engineering, Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States of America
| | - Jean-Marie C. Bouteiller
- Center for Neural Engineering, Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States of America
- Rhenovia Pharma, Mulhouse, France
- * E-mail:
| | - Eric Y. Hu
- Center for Neural Engineering, Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States of America
| | | | | | | | - Michel Baudry
- Rhenovia Pharma, Mulhouse, France
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States of America
| | - Theodore W. Berger
- Center for Neural Engineering, Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States of America
| |
Collapse
|
42
|
Shifting towards a model of mGluR5 dysregulation in schizophrenia: Consequences for future schizophrenia treatment. Neuropharmacology 2015; 115:73-91. [PMID: 26349010 DOI: 10.1016/j.neuropharm.2015.08.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 08/02/2015] [Accepted: 08/03/2015] [Indexed: 12/22/2022]
Abstract
Metabotropic glutamate receptor subtype 5 (mGluR5), encoded by the GRM5 gene, represents a compelling novel drug target for the treatment of schizophrenia. mGluR5 is a postsynaptic G-protein coupled glutamate receptor strongly linked with several critical cellular processes that are reported to be disrupted in schizophrenia. Accordingly, mGluR5 positive allosteric modulators show encouraging therapeutic potential in preclinical schizophrenia models, particularly for the treatment of cognitive dysfunctions against which currently available therapeutics are largely ineffective. More work is required to support the progression of mGluR5-targeting drugs into the clinic for schizophrenia treatment, although some obstacles may be overcome by comprehensively understanding how mGluR5 itself is involved in the neurobiology of the disorder. Several processes that are necessary for the regulation of mGluR5 activity have been identified, but not examined, in the context of schizophrenia. These processes include protein-protein interactions, dimerisation, subcellular trafficking, the impact of genetic variability or mutations on protein function, as well as epigenetic, post-transcriptional and post-translational processes. It is essential to understand these aspects of mGluR5 to determine whether they are affected in schizophrenia pathology, and to assess the consequences of mGluR5 dysfunction for the future use of mGluR5-based drugs. Here, we summarise the known processes that regulate mGluR5 and those that have already been studied in schizophrenia, and discuss the consequences of this dysregulation for current mGluR5 pharmacological strategies. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
|
43
|
Tashiro A, Nishida Y, Bereiter DA. Local group I mGluR antagonists reduce TMJ-evoked activity of trigeminal subnucleus caudalis neurons in female rats. Neuroscience 2015; 299:125-33. [PMID: 25934040 DOI: 10.1016/j.neuroscience.2015.04.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/17/2015] [Accepted: 04/21/2015] [Indexed: 10/23/2022]
Abstract
Group I metabotropic glutamate receptors (mGluR1 and mGluR5) are functionally linked to estrogen receptors and play a key role in the plasticity of central neurons. Estrogen status strongly influences sensory input from the temporomandibular joint (TMJ) to neurons at the spinomedullary (Vc/C1-2) region. This study tested the hypothesis that TMJ input to trigeminal subnucleus caudalis/upper cervical cord (Vc/C1-2) neurons involved group I mGluR activation and depended on estrogen status. TMJ-responsive neurons were recorded in superficial laminae at the Vc/C1-2 region in ovariectomized (OvX) female rats treated with low-dose estradiol (2 μg/day, LE) or high-dose estradiol (20 μg/day, HE) for 2 days. TMJ-responsive units were activated by adenosine triphosphate (ATP, 1mM) injected into the joint space. Receptor antagonists selective for mGluR1 (CPCCOEt) or mGluR5 (MPEP) were applied topically to the Vc/C1-2 surface at the site of recording 10 min prior to the intra-TMJ ATP stimulus. In HE rats, CPCCOEt (50 and 500 μM) markedly reduced ATP-evoked unit activity. By contrast, in LE rats, a small but significant increase in neural activity was seen after 50 μM CPCCOEt, while 500 μM caused a large reduction in activity that was similar in magnitude as that seen in HE rats. Local application of MPEP produced a significant inhibition of TMJ-evoked unit activity independent of estrogen status. Neither mGluR1 nor mGluR5 antagonism altered the spontaneous activity of TMJ units in HE or LE rats. High-dose MPEP caused a small reduction in the size of the convergent cutaneous receptive field in HE rats, while CPCCOEt had no effect. These data suggest that group I mGluRs play a key role in sensory integration of TMJ nociceptive input to the Vc/C1-2 region and are largely independent of estrogen status.
Collapse
Affiliation(s)
- A Tashiro
- Department of Physiology, National Defense Medical College, Namiki 3-2, Tokorozawa City, Saitama 359-8513, Japan.
| | - Y Nishida
- Department of Physiology, National Defense Medical College, Namiki 3-2, Tokorozawa City, Saitama 359-8513, Japan
| | - D A Bereiter
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Moos Tower 18-214, 515 Delaware Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
44
|
Metabotropic glutamate receptor 5 as drug target for Fragile X syndrome. Curr Opin Pharmacol 2015; 20:124-34. [DOI: 10.1016/j.coph.2014.11.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/10/2014] [Accepted: 11/10/2014] [Indexed: 11/17/2022]
|
45
|
Mansouri M, Kasugai Y, Fukazawa Y, Bertaso F, Raynaud F, Perroy J, Fagni L, Kaufmann WA, Watanabe M, Shigemoto R, Ferraguti F. Distinct subsynaptic localization of type 1 metabotropic glutamate receptors at glutamatergic and
GABA
ergic synapses in the rodent cerebellar cortex. Eur J Neurosci 2014; 41:157-67. [DOI: 10.1111/ejn.12779] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 09/26/2014] [Accepted: 10/05/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Mahnaz Mansouri
- Department of Pharmacology Innsbruck Medical University Innsbruck Austria
| | - Yu Kasugai
- Department of Pharmacology Innsbruck Medical University Innsbruck Austria
| | - Yugo Fukazawa
- Division of Cerebral Structure National Institute for Physiological Sciences Okazaki Japan
| | - Federica Bertaso
- CNRS UMR‐5203 Institut de Génomique Fonctionnelle Montpellier France
- INSERM U661 Montpellier France
- Universités de Montpellier 1 & 2 UMR‐5203 Montpellier France
| | - Fabrice Raynaud
- CNRS UMR‐5203 Institut de Génomique Fonctionnelle Montpellier France
- INSERM U661 Montpellier France
- Universités de Montpellier 1 & 2 UMR‐5203 Montpellier France
| | - Julie Perroy
- CNRS UMR‐5203 Institut de Génomique Fonctionnelle Montpellier France
- INSERM U661 Montpellier France
- Universités de Montpellier 1 & 2 UMR‐5203 Montpellier France
| | - Laurent Fagni
- CNRS UMR‐5203 Institut de Génomique Fonctionnelle Montpellier France
- INSERM U661 Montpellier France
- Universités de Montpellier 1 & 2 UMR‐5203 Montpellier France
| | - Walter A. Kaufmann
- Department of Pharmacology Innsbruck Medical University Innsbruck Austria
| | | | - Ryuichi Shigemoto
- Division of Cerebral Structure National Institute for Physiological Sciences Okazaki Japan
| | | |
Collapse
|
46
|
Auditory neuroplasticity, hearing loss and cochlear implants. Cell Tissue Res 2014; 361:251-69. [DOI: 10.1007/s00441-014-2004-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/04/2014] [Indexed: 10/24/2022]
|
47
|
Hu SSJ, Ho YC, Chiou LC. No more pain upon Gq-protein-coupled receptor activation: role of endocannabinoids. Eur J Neurosci 2014; 39:467-84. [PMID: 24494686 DOI: 10.1111/ejn.12475] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 12/02/2013] [Accepted: 12/05/2013] [Indexed: 01/24/2023]
Abstract
Marijuana has been used to relieve pain for centuries. The analgesic mechanism of its constituents, the cannabinoids, was only revealed after the discovery of cannabinoid receptors (CB1 and CB2) two decades ago. The subsequent identification of the endocannabinoids, anandamide and 2-arachidonoylglycerol (2-AG), and their biosynthetic and degradation enzymes discloses the therapeutic potential of compounds targeting the endocannabinoid system for pain control. Inhibitors of the anandamide and 2-AG degradation enzymes, fatty acid amide hydrolase and monoacylglycerol lipase, respectively, may be superior to direct cannabinoid receptor ligands as endocannabinoids are synthesized on demand and rapidly degraded, focusing action at generating sites. Recently, a promising strategy for pain relief was revealed in the periaqueductal gray (PAG). It is initiated by Gq-protein-coupled receptor (Gq PCR) activation of the phospholipase C-diacylglycerol lipase enzymatic cascade, generating 2-AG that produces inhibition of GABAergic transmission (disinhibition) in the PAG, thereby leading to analgesia. Here, we introduce the antinociceptive properties of exogenous cannabinoids and endocannabinoids, involving their biosynthesis and degradation processes, particularly in the PAG. We also review recent studies disclosing the Gq PCR-phospholipase C-diacylglycerol lipase-2-AG retrograde disinhibition mechanism in the PAG, induced by activating several Gq PCRs, including metabotropic glutamatergic (type 5 metabotropic glutamate receptor), muscarinic acetylcholine (M1/M3), and orexin 1 receptors. Disinhibition mediated by type 5 metabotropic glutamate receptor can be initiated by glutamate transporter inhibitors or indirectly by substance P, neurotensin, cholecystokinin and capsaicin. Finally, the putative role of 2-AG generated after activating the above neurotransmitter receptors in stress-induced analgesia is discussed.
Collapse
Affiliation(s)
- Sherry Shu-Jung Hu
- Department of Psychology, National Cheng Kung University, Tainan, Taiwan
| | | | | |
Collapse
|
48
|
Lippiello P, Hoxha E, Volpicelli F, Lo Duca G, Tempia F, Miniaci MC. Noradrenergic modulation of the parallel fiber-Purkinje cell synapse in mouse cerebellum. Neuropharmacology 2014; 89:33-42. [PMID: 25218865 DOI: 10.1016/j.neuropharm.2014.08.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 08/05/2014] [Accepted: 08/19/2014] [Indexed: 10/24/2022]
Abstract
The signals arriving to Purkinje cells via parallel fibers are essential for all tasks in which the cerebellum is involved, including motor control, learning new motor skills and calibration of reflexes. Since learning also requires the activation of adrenergic receptors, we investigated the effects of adrenergic receptor agonists on the main plastic site of the cerebellar cortex, the parallel fiber-Purkinje cell synapse. Here we show that noradrenaline serves as an endogenous ligand for both α1-and α2-adrenergic receptors to produce synaptic depression between parallel fibers and Purkinje cells. On the contrary, PF-EPSCs were potentiated by the β-adrenergic receptor agonist isoproterenol. This short-term potentiation was postsynaptically expressed, required protein kinase A, and was mimicked by the β2-adrenoceptor agonist clenbuterol, suggesting that the β2-adrenoceptors mediate the noradrenergic facilitation of synaptic transmission between parallel fibers and Purkinje cells. Moreover, β-adrenoceptor activation lowered the threshold for cerebellar long-term potentiation induced by 1 Hz parallel fiber stimulation. The presence of both α and β adrenergic receptors on Purkinje cells suggests the existence of bidirectional mechanisms of regulation allowing the noradrenergic afferents to refine the signals arriving to Purkinje cells at particular arousal states or during learning.
Collapse
Affiliation(s)
| | - Eriola Hoxha
- Dept. of Neuroscience, University of Torino, Torino, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Italy
| | - Floriana Volpicelli
- Dept. of Pharmacy, University of Naples Federico II, Naples, Italy; Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR, Naples, Italy
| | | | - Filippo Tempia
- Dept. of Neuroscience, University of Torino, Torino, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Italy.
| | | |
Collapse
|
49
|
Abstract
Metabotropic glutamate receptors (mGluRs) are found throughout thalamus and cortex and are clearly important to circuit behavior in both structures, and so considering only participation of ionotropic glutamate receptors (e.g., [R,S]-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid [AMPA] and N-methyl-d-aspartate receptors [NMDA] receptors) in glutamatergic processing would be an unfortunate oversimplification. These mGluRs are found both postsynaptically, on target cells of glutamatergic afferents, and presynaptically, on various synaptic terminals themselves, and when activated, they produce prolonged effects lasting at least hundreds of msec to several sec and perhaps longer. Two main types exist: activation of group I mGluRs causes postsynaptic depolarization, and group II, hyperpolarization. Both types are implicated in synaptic plasticity, both short term and long term. Their evident importance in functioning of thalamus and cortex makes it critical to develop a better understanding of how these receptors are normally activated, especially because they also seem implicated in a wide range of neurological and cognitive pathologies.
Collapse
|
50
|
van Coevorden-Hameete MH, de Graaff E, Titulaer MJ, Hoogenraad CC, Sillevis Smitt PAE. Molecular and cellular mechanisms underlying anti-neuronal antibody mediated disorders of the central nervous system. Autoimmun Rev 2014; 13:299-312. [PMID: 24225076 DOI: 10.1016/j.autrev.2013.10.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 10/30/2013] [Indexed: 12/31/2022]
Abstract
Over the last decade multiple autoantigens located on the plasma membrane of neurons have been identified. Neuronal surface antigens include molecules directly involved in neurotransmission and excitability. Binding of the antibody to the antigen may directly alter the target protein's function, resulting in neurological disorders. The often striking reversibility of symptoms following early aggressive immunotherapy supports a pathogenic role for autoantibodies to neuronal surface antigens. In order to better understand and treat these neurologic disorders it is important to gain insight in the underlying mechanisms of antibody pathogenicity. In this review we discuss the clinical, circumstantial, in vitro and in vivo evidence for neuronal surface antibody pathogenicity and the possible underlying cellular and molecular mechanisms. This review shows that antibodies to neuronal surface antigens are often directed at conformational epitopes located in the extracellular domain of the antigen. The conformation of the epitope can be affected by specific posttranslational modifications. This may explain the distinct clinical phenotypes that are seen in patients with antibodies to antigens that are expressed throughout the brain. Furthermore, it is likely that there is a heterogeneous antibody population, consisting of different IgG subtypes and directed at multiple epitopes located in an immunogenic region. Binding of these antibodies may result in different pathophysiological mechanisms occurring in the same patient, together contributing to the clinical syndrome. Unraveling the predominant mechanism in each distinct antigen could provide clues for therapeutic interventions.
Collapse
Affiliation(s)
- M H van Coevorden-Hameete
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - E de Graaff
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - M J Titulaer
- Department of Neurology, Erasmus MC, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - C C Hoogenraad
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - P A E Sillevis Smitt
- Department of Neurology, Erasmus MC, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| |
Collapse
|