101
|
Catanese A, Garrido D, Walther P, Roselli F, Boeckers TM. Nutrient limitation affects presynaptic structures through dissociable Bassoon autophagic degradation and impaired vesicle release. J Cereb Blood Flow Metab 2018; 38:1924-1939. [PMID: 29972341 PMCID: PMC6259322 DOI: 10.1177/0271678x18786356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acute mismatch between metabolic requirements of neurons and nutrients/growth factors availability characterizes several neurological conditions such as traumatic brain injury, stroke and hypoglycemia. Although the effects of this mismatch have been investigated at cell biological level, the effects on synaptic structure and function are less clear. Since synaptic activity is the most energy-demanding neuronal function and it is directly linked to neuronal networks functionality, we have explored whether nutrient limitation (NL) affects the ultrastructure, function and composition of pre and postsynaptic terminals. We show that upon NL, presynaptic terminals show disorganized vesicle pools and reduced levels of the active zone protein Bassoon (but not of Piccolo). Moreover, NL triggers an impaired vesicle release, which is reversed by re-administration of glucose but not by the blockade of autophagic or proteasomal protein degradation. This reveals a dissociable correlation between presynaptic architecture and vesicle release, since restoring vesicle fusion does not necessarily depend from the rescue of Bassoon levels. Thus, our data show that the presynaptic compartment is highly sensitive to NL and the rescue of presynaptic function requires re-establishment of the metabolic supply rather than preventing local protein degradation.
Collapse
Affiliation(s)
- Alberto Catanese
- 1 Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.,2 International Graduate School in Molecular Medicine Ulm (IGradU), Ulm University, Ulm, Germany
| | - Débora Garrido
- 1 Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.,2 International Graduate School in Molecular Medicine Ulm (IGradU), Ulm University, Ulm, Germany
| | - Paul Walther
- 3 Electron Microscopy Institute, Ulm University, Ulm, Germany
| | - Francesco Roselli
- 1 Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.,4 Department of Neurology, Ulm University, Ulm, Germany
| | - Tobias M Boeckers
- 1 Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| |
Collapse
|
102
|
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: 49] [Impact Index Per Article: 7.0] [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
|
103
|
Yang Y, Tian X, Xu D, Zheng F, Lu X, Zhang Y, Ma Y, Li Y, Xu X, Zhu B, Wang X. GPR40 modulates epileptic seizure and NMDA receptor function. SCIENCE ADVANCES 2018; 4:eaau2357. [PMID: 30345361 PMCID: PMC6192686 DOI: 10.1126/sciadv.aau2357] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 09/12/2018] [Indexed: 05/21/2023]
Abstract
Epilepsy is a common neurological disease, and approximately 30% of patients do not respond adequately to antiepileptic drug treatment. Recent studies suggest that G protein-coupled receptor 40 (GPR40) is expressed in the central nervous system and is involved in the regulation of neurological function. However, the impact of GPR40 on epileptic seizures remains unclear. In this study, we first reported that GPR40 expression was increased in epileptic brains. In the kainic acid-induced epilepsy model, GPR40 activation after status epilepticus alleviated epileptic activity, whereas GPR40 inhibition showed the opposite effect. In the pentylenetetrazole-induced kindling model, susceptibility to epilepsy was reduced with GPR40 activation and increased with GPR40 inhibition. Whole-cell patch-clamp recordings demonstrated that GPR40 affected N-methyl-d-aspartate (NMDA) receptor-mediated synaptic transmission. Moreover, GPR40 regulated NR2A and NR2B expression on the surface of neurons. In addition, endocytosis of NMDA receptors and binding of GPR40 with NR2A and NR2B can be regulated by GPR40. Together, our findings indicate that GPR40 modulates epileptic seizures, providing a novel antiepileptic target.
Collapse
Affiliation(s)
- Yong Yang
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Neurology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266003 Shandong, China
| | - Xin Tian
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Demei Xu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fangshuo Zheng
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xi Lu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yanke Zhang
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuanlin Ma
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yun Li
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xin Xu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Binglin Zhu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Corresponding author. (B.Z.); (X.W.)
| | - Xuefeng Wang
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing 100101, China
- Corresponding author. (B.Z.); (X.W.)
| |
Collapse
|
104
|
Sachana M, Rolaki A, Bal-Price A. Development of the Adverse Outcome Pathway (AOP): Chronic binding of antagonist to N-methyl-d-aspartate receptors (NMDARs) during brain development induces impairment of learning and memory abilities of children. Toxicol Appl Pharmacol 2018; 354:153-175. [PMID: 29524501 PMCID: PMC6095943 DOI: 10.1016/j.taap.2018.02.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 01/06/2023]
Abstract
The Adverse Outcome Pathways (AOPs) are designed to provide mechanistic understanding of complex biological systems and pathways of toxicity that result in adverse outcomes (AOs) relevant to regulatory endpoints. AOP concept captures in a structured way the causal relationships resulting from initial chemical interaction with biological target(s) (molecular initiating event) to an AO manifested in individual organisms and/or populations through a sequential series of key events (KEs), which are cellular, anatomical and/or functional changes in biological processes. An AOP provides the mechanistic detail required to support chemical safety assessment, the development of alternative methods and the implementation of an integrated testing strategy. An example of the AOP relevant to developmental neurotoxicity (DNT) is described here following the requirements of information defined by the OECD Users' Handbook Supplement to the Guidance Document for developing and assessing AOPs. In this AOP, the binding of an antagonist to glutamate receptor N-methyl-d-aspartate (NMDAR) receptor is defined as MIE. This MIE triggers a cascade of cellular KEs including reduction of intracellular calcium levels, reduction of brain derived neurotrophic factor release, neuronal cell death, decreased glutamate presynaptic release and aberrant dendritic morphology. At organ level, the above mentioned KEs lead to decreased synaptogenesis and decreased neuronal network formation and function causing learning and memory deficit at organism level, which is defined as the AO. There are in vitro, in vivo and epidemiological data that support the described KEs and their causative relationships rendering this AOP relevant to DNT evaluation in the context of regulatory purposes.
Collapse
Affiliation(s)
| | | | - Anna Bal-Price
- European Commission, Joint Research Centre, Ispra, Italy.
| |
Collapse
|
105
|
Ishida H, Skorobogatov A, Yamniuk AP, Vogel HJ. Solution structures of the
SH
3 domains from Shank scaffold proteins and their interactions with Cav1.3 calcium channels. FEBS Lett 2018; 592:2786-2797. [DOI: 10.1002/1873-3468.13209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/05/2018] [Accepted: 07/12/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Hiroaki Ishida
- Department of Biological Sciences University of Calgary Canada
| | | | | | - Hans J. Vogel
- Department of Biological Sciences University of Calgary Canada
| |
Collapse
|
106
|
Collins SM, Galvez R. Neocortical SHANK1 regulation of forebrain dependent associative learning. Neurobiol Learn Mem 2018; 155:173-179. [PMID: 30053575 DOI: 10.1016/j.nlm.2018.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 11/18/2022]
Abstract
Learning-induced neocortical synaptic plasticity is a well-established mechanism mediating memory consolidation. Classic learning paradigms elicit synaptic changes in various brain regions including the neocortex. Work from our laboratory has further suggested synaptic remodeling in primary somatosensory cortex (S1) during forebrain-dependent associative learning. While this process of synaptic remodeling is largely believed to contribute to memory consolidation, the underlying processes mediating this plasticity are poorly understood. Interestingly, abnormal expression of the synaptic scaffolding protein SHANK1 has been linked with aberrant synaptic plasticity and learning impairments, suggesting that it plays a critical role in these processes. However, a direct analysis of the role for SHANK1 during learning in the neocortex, the most likely site for memory storage, has never been adequately explored. To directly examine SHANK1's potential role during learning and memory, the following study set out to both examine neocortical SHANK1 expression during a learning event and determine the consequences of reducing neocortical SHANK1 expression on learning. The current study found that SHANK1 expression is transiently increased during periods of learning-induced dendritic spine plasticity in the neocortex. Furthermore, shRNA-mediated neocortical SHANK1 knockdown significantly impairs acquisition for the forebrain-dependent associative learning task (whisker-trace-eyeblink conditioning). Consistent with these findings, SHANK1 has been implicated in various neurological disorders. Collectively, these findings suggest a role for SHANK1 in neocortical learning-induced dendritic spine plasticity underlying learning and normal cognition; thus, providing potential insight into neurological mechanisms mediating abnormalities of impaired cognition.
Collapse
Affiliation(s)
- Sean M Collins
- Psychology Department, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States
| | - Roberto Galvez
- Psychology Department, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States; Neuroscience Program, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States; Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States.
| |
Collapse
|
107
|
Eltokhi A, Rappold G, Sprengel R. Distinct Phenotypes of Shank2 Mouse Models Reflect Neuropsychiatric Spectrum Disorders of Human Patients With SHANK2 Variants. Front Mol Neurosci 2018; 11:240. [PMID: 30072871 PMCID: PMC6060255 DOI: 10.3389/fnmol.2018.00240] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/21/2018] [Indexed: 12/26/2022] Open
Abstract
The SHANK scaffolding proteins are important organizers for signaling proteins in the postsynapse of excitatory neurons. The functional significance of SHANK proteins becomes apparent by the wide spectrum of neurodevelopmental and neuropsychiatric disorders associated with SHANK variants in human patients. A similar diversity of neuropsychiatric-like phenotypes is described for numerous Shank2 and Shank3 knockout (KO) mouse lines. In this review, we will focus on and discuss the experimental results obtained from different, but genetically related and therefore comparable, Shank2 mouse models. First, we will describe the distinct SHANK2 variant-mediated neurodevelopmental and neuropsychiatric disorders in human patients. Then we will discuss the current knowledge of the expressed SHANK2 isoforms in the mouse, and we will describe the genetic strategies used for generating three conventional and seven conditional Shank2 mouse lines. The distinct impairments i.e., autistic-like and mania-like behavior and the alterations on the molecular, electrophysiological and behavioral levels will be compared between the different Shank2 mouse models. We will present our view as to why in these mouse models a spectrum of phenotypes can arise from similar Shank2 gene manipulations and how Shank2 mutant mice can be used and should be analyzed on the behavioral level in future research.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Max Planck Research Group "Molecular Neurobiology", Max Planck Institute for Medical Research, Heidelberg, Germany.,Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany.,Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Gudrun Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Rolf Sprengel
- Max Planck Research Group "Molecular Neurobiology", Max Planck Institute for Medical Research, Heidelberg, Germany.,Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
108
|
Defective Synapse Maturation and Enhanced Synaptic Plasticity in Shank2 Δex7 -/- Mice. eNeuro 2018; 5:eN-NWR-0398-17. [PMID: 30023428 PMCID: PMC6049608 DOI: 10.1523/eneuro.0398-17.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 04/19/2018] [Accepted: 05/07/2018] [Indexed: 11/21/2022] Open
Abstract
Autism spectrum disorders (ASDs) are neurodevelopmental disorders with a strong genetic etiology. Since mutations in human SHANK genes have been found in patients with autism, genetic mouse models are used for a mechanistic understanding of ASDs and the development of therapeutic strategies. SHANKs are scaffold proteins in the postsynaptic density of mammalian excitatory synapses with proposed functions in synaptogenesis, regulation of dendritic spine morphology, and instruction of structural synaptic plasticity. In contrast to all studies so far on the function of SHANK proteins, we have previously observed enhanced synaptic plasticity in Shank2 Δex7−/− mice. In a series of experiments, we now reproduce these results, further explore the synaptic phenotype, and directly compare our model to the independently generated Shank2 Δex6-7−/− mice. Minimal stimulation experiments reveal that Shank2 Δex7−/− mice possess an excessive fraction of silent (i.e., α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, short, AMPA receptor lacking) synapses. The synaptic maturation deficit emerges during the third postnatal week and constitutes a plausible mechanistic explanation for the mutants’ increased capacity for long-term potentiation, both in vivo and in vitro. A direct comparison with Shank2 Δex6-7−/− mice adds weight to the hypothesis that both mouse models show a different set of synaptic phenotypes, possibly due to differences in their genetic background. These findings add to the diversity of synaptic phenotypes in neurodevelopmental disorders and further support the supposed existence of “modifier genes” in the expression and inheritance of ASDs.
Collapse
|
109
|
Heise C, Preuss JM, Schroeder JC, Battaglia CR, Kolibius J, Schmid R, Kreutz MR, Kas MJH, Burbach JPH, Boeckers TM. Heterogeneity of Cell Surface Glutamate and GABA Receptor Expression in Shank and CNTN4 Autism Mouse Models. Front Mol Neurosci 2018; 11:212. [PMID: 29970989 PMCID: PMC6018460 DOI: 10.3389/fnmol.2018.00212] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/30/2018] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorder (ASD) refers to a large set of neurodevelopmental disorders, which have in common both repetitive behavior and abnormalities in social interactions and communication. Interestingly, most forms of ASD have a strong genetic contribution. However, the molecular underpinnings of this disorder remain elusive. The SHANK3 gene (and to a lesser degree SHANK2) which encode for the postsynaptic density (PSD) proteins SHANK3/SHANK2 and the CONTACTIN 4 gene which encodes for the neuronal glycoprotein CONTACTIN4 (CNTN4) exhibit mutated variants which are associated with ASD. Like many of the other genes associated with ASD, both SHANKs and CNTN4 affect synapse formation and function and are therefore related to the proper development and signaling capability of excitatory and inhibitory neuronal networks in the adult mammal brain. In this study, we used mutant/knock-out mice of Shank2 (Shank2−/−), Shank3 (Shank3αβ−/−), and Cntn4 (Cntn4−/−) as ASD-models to explore whether these mice share a molecular signature in glutamatergic and GABAergic synaptic transmission in ASD-related brain regions. Using a biotinylation assay and subsequent western blotting we focused our analysis on cell surface expression of several ionotropic glutamate and GABA receptor subunits: GluA1, GluA2, and GluN1 were analyzed for excitatory synaptic transmission, and the α1 subunit of the GABAA receptor was analyzed for inhibitory synaptic transmission. We found that both Shank2−/− and Shank3αβ−/− mice exhibit reduced levels of several cell surface glutamate receptors in the analyzed brain regions—especially in the striatum and thalamus—when compared to wildtype controls. Interestingly, even though Cntn4−/− mice also show reduced levels of some cell surface glutamate receptors in the cortex and hippocampus, increased levels of cell surface glutamate receptors were found in the striatum. Moreover, Cntn4−/− mice do not only show brain region-specific alterations in cell surface glutamate receptors but also a downregulation of cell surface GABA receptors in several of the analyzed brain regions. The results of this study suggest that even though mutations in defined genes can be associated with ASD this does not necessarily result in a common molecular phenotype in surface expression of glutamatergic and GABAergic receptor subunits in defined brain regions.
Collapse
Affiliation(s)
- Christopher Heise
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Jonathan M Preuss
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Jan C Schroeder
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Jonas Kolibius
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Rebecca Schmid
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands.,Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| |
Collapse
|
110
|
Yamazaki H, Sasagawa Y, Yamamoto H, Bito H, Shirao T. CaMKIIβ is localized in dendritic spines as both drebrin-dependent and drebrin-independent pools. J Neurochem 2018; 146:145-159. [PMID: 29675826 PMCID: PMC6099455 DOI: 10.1111/jnc.14449] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 03/14/2018] [Accepted: 04/04/2018] [Indexed: 12/21/2022]
Abstract
Drebrin is a major F-actin binding protein in dendritic spines that is critically involved in the regulation of dendritic spine morphogenesis, pathology, and plasticity. In this study, we aimed to identify a novel drebrin-binding protein involved in spine morphogenesis and synaptic plasticity. We confirmed the beta subunit of Ca2+ /calmodulin-dependent protein kinase II (CaMKIIβ) as a drebrin-binding protein using a yeast two-hybrid system, and investigated the drebrin-CaMKIIβ relationship in dendritic spines using rat hippocampal neurons. Drebrin knockdown resulted in diffuse localization of CaMKIIβ in dendrites during the resting state, suggesting that drebrin is involved in the accumulation of CaMKIIβ in dendritic spines. Fluorescence recovery after photobleaching analysis showed that drebrin knockdown increased the stable fraction of CaMKIIβ, indicating the presence of drebrin-independent, more stable CaMKIIβ. NMDA receptor activation also increased the stable fraction in parallel with drebrin exodus from dendritic spines. These findings suggest that CaMKIIβ can be classified into distinct pools: CaMKIIβ associated with drebrin, CaMKIIβ associated with post-synaptic density (PSD), and CaMKIIβ free from PSD and drebrin. CaMKIIβ appears to be anchored to a protein complex composed of drebrin-binding F-actin during the resting state. NMDA receptor activation releases CaMKIIβ from drebrin resulting in CaMKIIβ association with PSD.
Collapse
Affiliation(s)
- Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yoshio Sasagawa
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hideyuki Yamamoto
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
111
|
Joensuu M, Lanoue V, Hotulainen P. Dendritic spine actin cytoskeleton in autism spectrum disorder. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:362-381. [PMID: 28870634 DOI: 10.1016/j.pnpbp.2017.08.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/21/2017] [Accepted: 08/30/2017] [Indexed: 01/01/2023]
Abstract
Dendritic spines are small actin-rich protrusions from neuronal dendrites that form the postsynaptic part of most excitatory synapses. Changes in the shape and size of dendritic spines correlate with the functional changes in excitatory synapses and are heavily dependent on the remodeling of the underlying actin cytoskeleton. Recent evidence implicates synapses at dendritic spines as important substrates of pathogenesis in neuropsychiatric disorders, including autism spectrum disorder (ASD). Although synaptic perturbations are not the only alterations relevant for these diseases, understanding the molecular underpinnings of the spine and synapse pathology may provide insight into their etiologies and could reveal new drug targets. In this review, we will discuss recent findings of defective actin regulation in dendritic spines associated with ASD.
Collapse
Affiliation(s)
- Merja Joensuu
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland; Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Vanessa Lanoue
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland.
| |
Collapse
|
112
|
Moretto E, Murru L, Martano G, Sassone J, Passafaro M. Glutamatergic synapses in neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:328-342. [PMID: 28935587 DOI: 10.1016/j.pnpbp.2017.09.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/28/2017] [Accepted: 09/16/2017] [Indexed: 12/22/2022]
Abstract
Neurodevelopmental disorders (NDDs) are a group of diseases whose symptoms arise during childhood or adolescence and that impact several higher cognitive functions such as learning, sociability and mood. Accruing evidence suggests that a shared pathogenic mechanism underlying these diseases is the dysfunction of glutamatergic synapses. We summarize present knowledge on autism spectrum disorders (ASD), intellectual disability (ID), Down syndrome (DS), Rett syndrome (RS) and attention-deficit hyperactivity disorder (ADHD), highlighting the involvement of glutamatergic synapses and receptors in these disorders. The most commonly shared defects involve α-amino-3-hydroxy-5-methyl- 4-isoxazole propionic acid receptors (AMPARs), N-methyl-d-aspartate receptors (NMDARs) and metabotropic glutamate receptors (mGluRs), whose functions are strongly linked to synaptic plasticity, affecting both cell-autonomous features as well as circuit formation. Moreover, the major scaffolding proteins and, thus, the general structure of the synapse are often deregulated in neurodevelopmental disorders, which is not surprising considering their crucial role in the regulation of glutamate receptor positioning and functioning. This convergence of defects supports the definition of neurodevelopmental disorders as a continuum of pathological manifestations, suggesting that glutamatergic synapses could be a therapeutic target to ameliorate patient symptomatology.
Collapse
Affiliation(s)
- Edoardo Moretto
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Luca Murru
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Giuseppe Martano
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Jenny Sassone
- San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - Maria Passafaro
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy.
| |
Collapse
|
113
|
Mossa A, Giona F, Pagano J, Sala C, Verpelli C. SHANK genes in autism: Defining therapeutic targets. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:416-423. [PMID: 29175319 DOI: 10.1016/j.pnpbp.2017.11.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Adele Mossa
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Federica Giona
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Jessica Pagano
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Carlo Sala
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
114
|
Lrfn2-Mutant Mice Display Suppressed Synaptic Plasticity and Inhibitory Synapse Development and Abnormal Social Communication and Startle Response. J Neurosci 2018; 38:5872-5887. [PMID: 29798891 DOI: 10.1523/jneurosci.3321-17.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 05/03/2018] [Accepted: 05/16/2018] [Indexed: 11/21/2022] Open
Abstract
SALM1 (SALM (synaptic adhesion-like molecule), also known as LRFN2 (leucine rich repeat and fibronectin type III domain containing), is a postsynaptic density (PSD)-95-interacting synaptic adhesion molecule implicated in the regulation of NMDA receptor (NMDAR) clustering largely based on in vitro data, although its in vivo functions remain unclear. Here, we found that mice lacking SALM1/LRFN2 (Lrfn2-/- mice) show a normal density of excitatory synapses but altered excitatory synaptic function, including enhanced NMDAR-dependent synaptic transmission but suppressed NMDAR-dependent synaptic plasticity in the hippocampal CA1 region. Unexpectedly, SALM1 expression was detected in both glutamatergic and GABAergic neurons and Lrfn2-/- CA1 pyramidal neurons showed decreases in the density of inhibitory synapses and the frequency of spontaneous inhibitory synaptic transmission. Behaviorally, ultrasonic vocalization was suppressed in Lrfn2-/- pups separated from their mothers and acoustic startle was enhanced, but locomotion, anxiety-like behavior, social interaction, repetitive behaviors, and learning and memory were largely normal in adult male Lrfn2-/- mice. These results suggest that SALM1/LRFN2 regulates excitatory synapse function, inhibitory synapse development, and social communication and startle behaviors in mice.SIGNIFICANCE STATEMENT Synaptic adhesion molecules regulate synapse development and function, which govern neural circuit and brain functions. The SALM/LRFN (synaptic adhesion-like molecule/leucine rich repeat and fibronectin type III domain containing) family of synaptic adhesion proteins consists of five known members for which the in vivo functions are largely unknown. Here, we characterized mice lacking SALM1/LRFN2 (SALM1 KO) known to associate with NMDA receptors (NMDARs) and found that these mice showed altered NMDAR-dependent synaptic transmission and plasticity, as expected, but unexpectedly also exhibited suppressed inhibitory synapse development and synaptic transmission. Behaviorally, SALM1 KO pups showed suppressed ultrasonic vocalization upon separation from their mothers and SALM1 KO adults showed enhanced responses to loud acoustic stimuli. These results suggest that SALM1/LRFN2 regulates excitatory synapse function, inhibitory synapse development, social communication, and acoustic startle behavior.
Collapse
|
115
|
USP8 Deubiquitinates SHANK3 to Control Synapse Density and SHANK3 Activity-Dependent Protein Levels. J Neurosci 2018; 38:5289-5301. [PMID: 29735556 DOI: 10.1523/jneurosci.3305-17.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/28/2018] [Accepted: 03/29/2018] [Indexed: 11/21/2022] Open
Abstract
Mutations or altered protein levels of SHANK3 are implicated in neurodevelopmental disorders such as Phelan-McDermid syndrome, autism spectrum disorders, and schizophrenia (Guilmatre et al., 2014). Loss of SHANK3 in mouse models results in decreased synapse density and reduction in the levels of multiple synaptic proteins (Jiang and Ehlers, 2013). The family of SHANK scaffolding molecules are among the most heavily ubiquitinated proteins at the postsynaptic density. The ubiquitin-dependent proteasome degradation of SHANK is regulated by synaptic activity and may contribute to activity-dependent synaptic remodeling (Ehlers, 2003; Shin et al., 2012). However, the identity of the specific deubiquitinating enzymes and E3 ligases that regulate SHANK ubiquitination at synapses are unknown. Here we identify USP8/UBPY as a deubiquitinating enzyme that regulates SHANK3 and SHANK1 ubiquitination and protein levels. In primary rat neurons, USP8 enhances SHANK3 and SHANK1 protein levels via deubiquitination and increases dendritic spine density. Additionally, USP8 is essential for changes in SHANK3 protein levels following synaptic activity modulation. These data identify USP8 as a key modulator of SHANK3 downstream of synaptic activity.SIGNIFICANCE STATEMENT Precise regulation of the protein levels of the postsynaptic scaffolding protein SHANK3 is essential for proper neurodevelopment. Mutations of SHANK3 have been identified in Phelan-McDermid syndrome, autism spectrum disorders, and schizophrenia (Guilmatre et al., 2014). In this research, we identify USP8 as a key enzyme that regulates SHANK3 protein levels in neurons. USP8 acts to deubiquitinate SHANK3, which prevents its proteasomal-mediated degradation and enhances overall dendritic spine stability. In the future, the modulation of USP8 deubiquitinating activity could potentially be used to titrate the protein levels of SHANK3 to ameliorate disease.
Collapse
|
116
|
Ali Rodriguez R, Joya C, Hines RM. Common Ribs of Inhibitory Synaptic Dysfunction in the Umbrella of Neurodevelopmental Disorders. Front Mol Neurosci 2018; 11:132. [PMID: 29740280 PMCID: PMC5928253 DOI: 10.3389/fnmol.2018.00132] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/03/2018] [Indexed: 01/06/2023] Open
Abstract
The term neurodevelopmental disorder (NDD) is an umbrella term used to group together a heterogeneous class of disorders characterized by disruption in cognition, emotion, and behavior, early in the developmental timescale. These disorders are heterogeneous, yet they share common behavioral symptomatology as well as overlapping genetic contributors, including proteins involved in the formation, specialization, and function of synaptic connections. Advances may arise from bridging the current knowledge on synapse related factors indicated from both human studies in NDD populations, and in animal models. Mounting evidence has shown a link to inhibitory synapse formation, specialization, and function among Autism, Angelman, Rett and Dravet syndromes. Inhibitory signaling is diverse, with numerous subtypes of inhibitory interneurons, phasic and tonic modes of inhibition, and the molecular and subcellular diversity of GABAA receptors. We discuss common ribs of inhibitory synapse dysfunction in the umbrella of NDD, highlighting alterations in the developmental switch to inhibitory GABA, dysregulation of neuronal activity patterns by parvalbumin-positive interneurons, and impaired tonic inhibition. Increasing our basic understanding of inhibitory synapses, and their role in NDDs is likely to produce significant therapeutic advances in behavioral symptom alleviation for interrelated NDDs.
Collapse
Affiliation(s)
- Rachel Ali Rodriguez
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Christina Joya
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Rochelle M Hines
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| |
Collapse
|
117
|
Li L, Su S, Perry CJ, Elphick MR, Chittka L, Søvik E. Large-scale transcriptome changes in the process of long-term visual memory formation in the bumblebee, Bombus terrestris. Sci Rep 2018; 8:534. [PMID: 29323174 PMCID: PMC5765018 DOI: 10.1038/s41598-017-18836-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/14/2017] [Indexed: 01/05/2023] Open
Abstract
Many genes have been implicated in mechanisms of long-term memory formation, but there is still much to be learnt about how the genome dynamically responds, transcriptionally, during memory formation. In this study, we used high-throughput sequencing to examine how transcriptome profiles change during visual memory formation in the bumblebee (Bombus terrestris). Expression of fifty-five genes changed immediately after bees were trained to associate reward with a single coloured chip, and the upregulated genes were predominantly genes known to be involved in signal transduction. Changes in the expression of eighty-one genes were observed four hours after learning a new colour, and the majority of these were upregulated and related to transcription and translation, which suggests that the building of new proteins may be the predominant activity four hours after training. Several of the genes identified in this study (e.g. Rab10, Shank1 and Arhgap44) are interesting candidates for further investigation of the molecular mechanisms of long-term memory formation. Our data demonstrate the dynamic gene expression changes after associative colour learning and identify genes involved in each transcriptional wave, which will be useful for future studies of gene regulation in learning and long-term memory formation.
Collapse
Affiliation(s)
- Li Li
- School of Biological and Chemical Sciences, Queen Mary University of London, London, E1 4NS, UK.
| | - Songkun Su
- College of Bee Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| | - Clint J Perry
- School of Biological and Chemical Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Maurice R Elphick
- School of Biological and Chemical Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Lars Chittka
- School of Biological and Chemical Sciences, Queen Mary University of London, London, E1 4NS, UK
- Institute for Advanced Study, Wallotstrasse 19, D-14193, Berlin, Germany
| | - Eirik Søvik
- Department of Science and Mathematics, Volda University College, 6100, Volda, Norway
| |
Collapse
|
118
|
Suzuki T, Kametani K, Guo W, Li W. Protein components of post-synaptic density lattice, a backbone structure for type I excitatory synapses. J Neurochem 2017; 144:390-407. [PMID: 29134655 DOI: 10.1111/jnc.14254] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/17/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022]
Abstract
It is essential to study the molecular architecture of post-synaptic density (PSD) to understand the molecular mechanism underlying the dynamic nature of PSD, one of the bases of synaptic plasticity. A well-known model for the architecture of PSD of type I excitatory synapses basically comprises of several scaffolding proteins (scaffold protein model). On the contrary, 'PSD lattice' observed through electron microscopy has been considered a basic backbone of type I PSDs. However, major constituents of the PSD lattice and the relationship between the PSD lattice and the scaffold protein model, remain unknown. We purified a PSD lattice fraction from the synaptic plasma membrane of rat forebrain. Protein components of the PSD lattice were examined through immuno-gold negative staining electron microscopy. The results indicated that tubulin, actin, α-internexin, and Ca2+ /calmodulin-dependent kinase II are major constituents of the PSD lattice, whereas scaffold proteins such as PSD-95, SAP102, GKAP, Shank1, and Homer, were rather minor components. A similar structure was also purified from the synaptic plasma membrane of forebrains from 7-day-old rats. On the basis of this study, we propose a 'PSD lattice-based dynamic nanocolumn' model for PSD molecular architecture, in which the scaffold protein model and the PSD lattice model are combined and an idea of dynamic nanocolumn PSD subdomain is also included. In the model, cytoskeletal proteins, in particular, tubulin, actin, and α-internexin, may play major roles in the construction of the PSD backbone and provide linker sites for various PSD scaffold protein complexes/subdomains.
Collapse
Affiliation(s)
- Tatsuo Suzuki
- Department of Neuroplasticity, Institute of Pathogenesis and Disease Prevention, Graduate School of Medicine, Shinshu University Academic Assembly, Matsumoto, Japan.,Department of Biological Sciences for Intractable Neurological Diseases, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research Shinshu University, Matsumoto, Japan.,Department of Molecular and Cellular physiology, Shinshu University Academic Assembly, Institute of Medicine, Matsumoto, Japan
| | - Kiyokazu Kametani
- Department of Instrumental Analysis, Research Center for Human and Environmental Science, Shinshu University, Matsumoto, Nagano, Japan
| | - Weiheng Guo
- Department of Neuroplasticity, Institute of Pathogenesis and Disease Prevention, Graduate School of Medicine, Shinshu University Academic Assembly, Matsumoto, Japan
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China.,Distinguished Visiting Professor, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research Shinshu University, Matsumoto, Japan
| |
Collapse
|
119
|
Collins SM, Belagodu AP, Reed SL, Galvez R. SHANK1 is differentially expressed during development in CA1 hippocampal neurons and astrocytes. Dev Neurobiol 2017; 78:363-373. [PMID: 29218848 DOI: 10.1002/dneu.22564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/19/2017] [Accepted: 11/30/2017] [Indexed: 01/25/2023]
Abstract
Recent studies have strongly suggested a role for the synaptic scaffolding protein SHANK1 in normal synaptic structure and signaling. Global SHANK1 knockout (SHANK1-/-) mice demonstrate reduced dendritic spine density, an immature dendritic spine phenotype and impairments in various cognitive tasks. SHANK1 overexpression is associated with increased dendritic spine size and impairments in fear conditioning. These studies suggest proper regulation of SHANK1 is crucial for appropriate synaptic structure and cognition. However, little is known regarding SHANK1's developmental expression in brain regions critical for learning. The current study quantified cell specific developmental expression of SHANK1 in the hippocampus, a brain region critically involved in various learning paradigms shown to be disrupted by SHANK1 dysregulation. Consistent with prior studies, SHANK1 was found to be strongly co-expressed with dendritic markers, with significant increased co-expression at postnatal day (P) 15, an age associated with increased synaptogenesis in the hippocampus. Interestingly, SHANK1 was also found to be expressed in astrocytes and microglia. To our knowledge, this is the first demonstration of glial SHANK1 localization; therefore, these findings were further examined via a glial purified primary cell culture fraction using magnetic cell sorting. This additional analysis further demonstrated that SHANK1 was expressed in glial cells, supporting our immunofluorescence co-expression findings. Developmentally, astroglial SHANK1 co-expression was found to be significantly elevated at P5 with a reduction into adulthood, while SHANK1 microglial co-expression did not significantly change across development. These data collectively implicate a more global role for SHANK1 in mediating normal cellular signaling in the brain. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 363-373, 2018.
Collapse
Affiliation(s)
- Sean M Collins
- Psychology Department, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801
| | - Amogh P Belagodu
- Neuroscience Program, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801
| | - Samantha L Reed
- Psychology Department, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801
| | - Roberto Galvez
- Psychology Department, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801.,Neuroscience Program, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801
| |
Collapse
|
120
|
Zhu J, Zhou Q, Shang Y, Li H, Peng M, Ke X, Weng Z, Zhang R, Huang X, Li SS, Feng G, Lu Y, Zhang M. Synaptic Targeting and Function of SAPAPs Mediated by Phosphorylation-Dependent Binding to PSD-95 MAGUKs. Cell Rep 2017; 21:3781-3793. [DOI: 10.1016/j.celrep.2017.11.107] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/13/2017] [Accepted: 11/29/2017] [Indexed: 10/18/2022] Open
|
121
|
Queenan BN, Dunn RL, Santos VR, Feng Y, Huizenga MN, Hammack RJ, Vicini S, Forcelli PA, Pak DTS. Kappa opioid receptors regulate hippocampal synaptic homeostasis and epileptogenesis. Epilepsia 2017; 59:106-122. [PMID: 29114861 DOI: 10.1111/epi.13941] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Homeostatic synaptic plasticity (HSP) serves as a gain control mechanism at central nervous system (CNS) synapses, including those between the dentate gyrus (DG) and CA3. Improper circuit control of DG-CA3 synapses is hypothesized to underlie epileptogenesis. Here, we sought to (1) identify compounds that preferentially modulate DG-CA3 synapses in primary neuronal culture and (2) determine if these compounds would delay or prevent epileptogenesis in vivo. METHODS We previously developed and validated an in vitro assay to visualize the behavior of DG-CA3 synapses and predict functional changes. We used this "synapse-on-chip" assay (quantification of synapse size, number, and type using immunocytochemical markers) to dissect the mechanisms of HSP at DG-CA3 synapses. Using chemogenetic constructs and pharmacological agents we determined the signaling cascades necessary for gain control at DG-CA3 synapses. Finally, we tested the implicated cascades (using kappa opioid receptor (OR) agonists and antagonists) in two models of epileptogenesis: electrical amygdala kindling in the mouse and chemical (pentylenetetrazole) kindling in the rat. RESULTS In vitro, synapses between DG mossy fibers (MFs) and CA3 neurons are the primary homeostatic responders during sustained periods of activity change. Kappa OR signaling is both necessary and sufficient for the homeostatic elaboration of DG-CA3 synapses, induced by presynaptic DG activity levels. Blocking kappa OR signaling in vivo attenuates the development of seizures in both mouse and rat models of epilepsy. SIGNIFICANCE This study elucidates mechanisms by which synapses between DG granule cells and CA3 pyramidal neurons undergo activity-dependent homeostatic compensation, via OR signaling in vitro. Modulation of kappa OR signaling in vivo alters seizure progression, suggesting that breakdown of homeostatic closed-loop control at DG-CA3 synapses contributes to seizures, and that targeting endogenous homeostatic mechanisms at DG-CA3 synapses may prove useful in combating epileptogenesis.
Collapse
Affiliation(s)
- Bridget N Queenan
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA.,Department of Mechanical Engineering, Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Raymond L Dunn
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA.,Georgetown Hughes Scholars Program, Department of Biology, Georgetown University, Washington, DC, USA
| | - Victor R Santos
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Yang Feng
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Megan N Huizenga
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Robert J Hammack
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Stefano Vicini
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Patrick A Forcelli
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
122
|
Lu WH, Yeh NH, Huang YS. CPEB2 Activates GRASP1 mRNA Translation and Promotes AMPA Receptor Surface Expression, Long-Term Potentiation, and Memory. Cell Rep 2017; 21:1783-1794. [DOI: 10.1016/j.celrep.2017.10.073] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/15/2017] [Accepted: 10/19/2017] [Indexed: 01/09/2023] Open
|
123
|
Datko MC, Hu JH, Williams M, Reyes CM, Lominac KD, von Jonquieres G, Klugmann M, Worley PF, Szumlinski KK. Behavioral and Neurochemical Phenotyping of Mice Incapable of Homer1a Induction. Front Behav Neurosci 2017; 11:208. [PMID: 29163080 PMCID: PMC5672496 DOI: 10.3389/fnbeh.2017.00208] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/12/2017] [Indexed: 11/18/2022] Open
Abstract
Immediate early and constitutively expressed products of the Homer1 gene regulate the functional assembly of post-synaptic density proteins at glutamatergic synapses to influence excitatory neurotransmission and synaptic plasticity. Earlier studies of Homer1 gene knock-out (KO) mice indicated active, but distinct, roles for IEG and constitutively expressed Homer1 gene products in regulating cognitive, emotional, motivational and sensorimotor processing, as well as behavioral and neurochemical sensitivity to cocaine. More recent characterization of transgenic mice engineered to prevent generation of the IEG form (a.k.a Homer1a KO) pose a critical role for Homer1a in cocaine-induced behavioral and neurochemical sensitization of relevance to drug addiction and related neuropsychiatric disorders. Here, we extend our characterization of the Homer1a KO mouse and report a modest pro-depressant phenotype, but no deleterious effects of the KO upon spatial learning/memory, prepulse inhibition, or cocaine-induced place-conditioning. As we reported previously, Homer1a KO mice did not develop cocaine-induced behavioral or neurochemical sensitization within the nucleus accumbens; however, virus-mediated Homer1a over-expression within the nucleus accumbens reversed the sensitization phenotype of KO mice. We also report several neurochemical abnormalities within the nucleus accumbens of Homer1a KO mice that include: elevated basal dopamine and reduced basal glutamate content, Group1 mGluR agonist-induced glutamate release and high K+-stimulated release of dopamine and glutamate within this region. Many of the neurochemical anomalies exhibited by Homer1a KO mice are recapitulated upon deletion of the entire Homer1 gene; however, Homer1 deletion did not affect NAC dopamine or alter K+-stimulated neurotransmitter release within this region. These data show that the selective deletion of Homer1a produces a behavioral and neurochemical phenotype that is distinguishable from that produced by deletion of the entire Homer1 gene. Moreover, the data indicate a specific role for Homer1a in regulating cocaine-induced behavioral and neurochemical sensitization of potential relevance to the psychotogenic properties of this drug.
Collapse
Affiliation(s)
- Michael C Datko
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Jia-Hua Hu
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Melanie Williams
- Department of Molecular, Developmental and Cell Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Cindy M Reyes
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Kevin D Lominac
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Georg von Jonquieres
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matthias Klugmann
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Paul F Worley
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States.,Department of Molecular, Developmental and Cell Biology, University of California, Santa Barbara, Santa Barbara, CA, United States.,Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
124
|
Shi R, Redman P, Ghose D, Hwang H, Liu Y, Ren X, Ding LJ, Liu M, Jones KJ, Xu W. Shank Proteins Differentially Regulate Synaptic Transmission. eNeuro 2017; 4:ENEURO.0163-15.2017. [PMID: 29250591 PMCID: PMC5731535 DOI: 10.1523/eneuro.0163-15.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 11/23/2017] [Accepted: 11/28/2017] [Indexed: 11/21/2022] Open
Abstract
Shank proteins, one of the principal scaffolds in the postsynaptic density (PSD) of the glutamatergic synapses, have been associated with autism spectrum disorders and neuropsychiatric diseases. However, it is not known whether different Shank family proteins have distinct functions in regulating synaptic transmission, and how they differ from other scaffold proteins in this aspect. Here, we investigate the role of Shanks in regulating glutamatergic synaptic transmission at rat hippocampal SC-CA1 synapses, using lentivirus-mediated knockdown and molecular replacement combined with dual whole-cell patch clamp in hippocampal slice culture. In line with previous findings regarding PSD-MAGUK scaffold manipulation, we found that loss of scaffold proteins via knockdown of Shank1 or Shank2, but not Shank3, led to a reduction of the number but not the unitary response of AMPAR-containing synapses. Only when both Shank1 and Shank2 were knocked down, were both the number and the unitary response of active synapses reduced. This reduction was accompanied by a decrease in NMDAR-mediated synaptic response, indicating more profound deficits in synaptic transmission. Molecular replacement with Shank2 and Shank3c rescued the synaptic transmission to the basal level, and the intact sterile α-motif (SAM) of Shank proteins is required for maintaining glutamatergic synaptic transmission. We also found that altered neural activity did not influence the effect of Shank1 or Shank2 knockdown on AMPAR synaptic transmission, in direct contrast to the activity dependence of the effect of PSD-95 knockdown, revealing differential interaction between activity-dependent signaling and scaffold protein families in regulating synaptic AMPAR function.
Collapse
Affiliation(s)
- Rebecca Shi
- Picower Institute for Learning and Memory
- Department of Brain and Cognitive Sciences
- Department of Biology
| | | | | | - Hongik Hwang
- Picower Institute for Learning and Memory
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Yan Liu
- Picower Institute for Learning and Memory
| | | | - Lei J. Ding
- Picower Institute for Learning and Memory
- Department of Biology
| | - Mingna Liu
- Picower Institute for Learning and Memory
| | | | - Weifeng Xu
- Picower Institute for Learning and Memory
- Department of Brain and Cognitive Sciences
| |
Collapse
|
125
|
Zhu L, Wang L, Ju F, Ran Y, Wang C, Zhang S. Transient global cerebral ischemia induces rapid and sustained reorganization of synaptic structures. J Cereb Blood Flow Metab 2017; 37:2756-2767. [PMID: 27798269 PMCID: PMC5536786 DOI: 10.1177/0271678x16674736] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ischemia can cause rapid neuronal damage. Previous studies have suggested that synaptic structures and cortical functions can be rescued if therapeutic interventions are applied in time, but the structural basis for this resilience remains incompletely understood. Here, we investigated the restoration of synaptic structures and postischemic plasticity of dendritic spines in the somatosensory cortices of mice by taking advantage of a reversible global cerebral ischemia model. Intravital two-photon imaging revealed that although dendritic structures were rapidly distorted after global ischemia, only a small percentage of spines were actually lost after transient ischemia. Electron microscopy indicated that most presynaptic electron-dense structures were still apposed to postsynaptic densities, and that the majority of disrupted synaptic structures were rapidly reinstated following reperfusion after transient ischemia. Repeated imaging suggested that restored dendrites survived the initial ischemia -reperfusion challenge. Importantly, spines on the restored dendrites underwent a rapid and sustained structural reorganization following transient ischemia. These findings suggested that disrupted synapses during transient ischemia could be rapidly restored after ischemia/reperfusion, and that restored dendritic structures remained plastic to rebuild the cortical network.
Collapse
Affiliation(s)
- Lirui Zhu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lei Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Furong Ju
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yanli Ran
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Cong Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
126
|
Cabrera-Pedraza VR, de Jesús Gómez-Villalobos M, de la Cruz F, Aguilar-Alonso P, Zamudio S, Flores G. Pregnancy improves cognitive deficit and neuronal morphology atrophy in the prefrontal cortex and hippocampus of aging spontaneously hypertensive rats. Synapse 2017; 71:e21991. [DOI: 10.1002/syn.21991] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/24/2017] [Accepted: 07/02/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Verónica R. Cabrera-Pedraza
- Instituto de Fisiología; Benemérita Universidad Autónoma de Puebla; Puebla Pue México
- Depto. de Fisiología; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional; Cdmx México
| | | | - Fidel de la Cruz
- Depto. de Fisiología; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional; Cdmx México
| | | | - Sergio Zamudio
- Depto. de Fisiología; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional; Cdmx México
| | - Gonzalo Flores
- Instituto de Fisiología; Benemérita Universidad Autónoma de Puebla; Puebla Pue México
| |
Collapse
|
127
|
Goulding SP, Szumlinski KK, Contet C, MacCoss MJ, Wu CC. A mass spectrometry-based proteomic analysis of Homer2-interacting proteins in the mouse brain. J Proteomics 2017; 166:127-137. [PMID: 28728878 DOI: 10.1016/j.jprot.2017.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 07/08/2017] [Accepted: 07/12/2017] [Indexed: 11/26/2022]
Abstract
In the brain, the Homer protein family modulates excitatory signal transduction and receptor plasticity through interactions with other proteins in dendritic spines. Homer proteins are implicated in a variety of psychiatric disorders such as schizophrenia and addiction. Since long Homers serve as scaffolding proteins, identifying their interacting partners is an important first step in understanding their biological function and could help to guide the design of new therapeutic strategies. The present study set out to document Homer2-interacting proteins in the mouse brain using a co-immunoprecipitation-based mass spectrometry approach where Homer2 knockout samples were used to filter out non-specific interactors. We found that in the mouse brain, Homer2 interacts with a limited subset of its previously reported interacting partners (3 out of 31). Importantly, we detected an additional 15 novel Homer2-interacting proteins, most of which are part of the N-methyl-D-aspartate receptor signaling pathway. These results corroborate the central role Homer2 plays in glutamatergic transmission and expand the network of proteins potentially contributing to the behavioral abnormalities associated with altered Homer2 expression. SIGNIFICANCE Long Homer proteins are scaffolding proteins that regulate signal transduction in neurons. Identifying their interacting partners is key to understanding their function. We used co-immunoprecipitation in combination with mass spectrometry to establish the first comprehensive list of Homer2-interacting partners in the mouse brain. The specificity of interactions was evaluated using Homer2 knockout brain tissue as a negative control. The set of proteins that we identified minimally overlaps with previously reported interacting partners of Homer2; however, we identified novel interactors that are part of a signaling cascade activated by glutamatergic transmission, which improves our mechanistic understanding of the role of Homer2 in behavior.
Collapse
Affiliation(s)
- Scott P Goulding
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, United States; Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, United States.
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, Molecular, Cellular and Developmental Biology, The Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Candice Contet
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, United States
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, United States
| | - Christine C Wu
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
128
|
Neuronal activity-regulated alternative mRNA splicing. Int J Biochem Cell Biol 2017; 91:184-193. [PMID: 28591617 DOI: 10.1016/j.biocel.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 11/20/2022]
Abstract
Activity-regulated gene transcription underlies plasticity-dependent changes in the molecular composition and structure of neurons. Numerous genes whose expression is induced by different neuronal plasticity inducing pathways have been identified, but the alteration of gene expression levels represents only part of the complexity of the activity-regulated transcriptional program. Alternative splicing of precursor mRNA is an additional mechanism that modulates the activity-dependent transcriptional signature. Recently developed splicing sensitive transcriptome wide analyses improve our understanding of the underlying mechanisms and demonstrate to what extend the activity regulated transcriptome is alternatively spliced. So far, only for a small group of differentially spliced mRNAs of synaptic proteins, the functional implications have been studied in detail. These include examples in which differential exon usage can result in the expression of alternative proteins which interfere with or alter the function of preexisting proteins and cause a dominant negative functional block of constitutively expressed variants. Such altered proteins contribute to the structural and functional reorganization of pre- and postsynaptic terminals and to the maintenance and formation of synapses. In addition, activity-induced alternative splicing can affect the untranslated regions (UTRs) and generates mRNAs harboring different cis-regulatory elements. Such differential UTRs can influence mRNA stability, translation, and can change the targeting of mRNAs to subcellular compartments. Here, we summarize different categories of alternative splicing which are thought to contribute to synaptic remodeling, give an overview of activity-regulated alternatively spliced mRNAs of synaptic proteins that impact synaptic functions, and discuss splicing factors and epigenetic modifications as regulatory determinants.
Collapse
|
129
|
Sungur AÖ, Jochner MCE, Harb H, Kılıç A, Garn H, Schwarting RKW, Wöhr M. Aberrant cognitive phenotypes and altered hippocampal BDNF expression related to epigenetic modifications in mice lacking the post-synaptic scaffolding protein SHANK1: Implications for autism spectrum disorder. Hippocampus 2017; 27:906-919. [PMID: 28500650 DOI: 10.1002/hipo.22741] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/05/2017] [Accepted: 05/03/2017] [Indexed: 12/29/2022]
Abstract
Autism spectrum disorder (ASD) is a class of neurodevelopmental disorders characterized by persistent deficits in social communication/interaction, together with restricted/repetitive patterns of behavior. ASD is among the most heritable neuropsychiatric conditions, and while available evidence points to a complex set of genetic factors, the SHANK gene family has emerged as one of the most promising candidates. Here, we assessed ASD-related phenotypes with particular emphasis on social behavior and cognition in Shank1 mouse mutants in comparison to heterozygous and wildtype littermate controls across development in both sexes. While social approach behavior was evident in all experimental conditions and social recognition was only mildly affected by genotype, Shank1-/- null mutant mice were severely impaired in object recognition memory. This effect was particularly prominent in juveniles, not due to impairments in object discrimination, and replicated in independent mouse cohorts. At the neurobiological level, object recognition deficits were paralleled by increased brain-derived neurotrophic factor (BDNF) protein expression in the hippocampus of Shank1-/- mice; yet BDNF levels did not differ under baseline conditions. We therefore investigated changes in the epigenetic regulation of hippocampal BDNF expression and detected an enrichment of histone H3 acetylation at the Bdnf promoter1 in Shank1-/- mice, consistent with increased learning-associated BDNF. Together, our findings indicate that Shank1 deletions lead to an aberrant cognitive phenotype characterized by severe impairments in object recognition memory and increased hippocampal BDNF levels, possibly due to epigenetic modifications. This result supports the link between ASD and intellectual disability, and suggests epigenetic regulation as a potential therapeutic target.
Collapse
Affiliation(s)
- A Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - Magdalena C E Jochner
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - Hani Harb
- Institute of Laboratory Medicine and Pathobiochemistry-Molecular Diagnostics, Philipps-University of Marburg, Marburg, Germany
| | - Ayşe Kılıç
- Institute of Laboratory Medicine and Pathobiochemistry-Molecular Diagnostics, Philipps-University of Marburg, Marburg, Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry-Molecular Diagnostics, Philipps-University of Marburg, Marburg, Germany
| | - Rainer K W Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| |
Collapse
|
130
|
Vicidomini C, Ponzoni L, Lim D, Schmeisser M, Reim D, Morello N, Orelanna D, Tozzi A, Durante V, Scalmani P, Mantegazza M, Genazzani AA, Giustetto M, Sala M, Calabresi P, Boeckers TM, Sala C, Verpelli C. Pharmacological enhancement of mGlu5 receptors rescues behavioral deficits in SHANK3 knock-out mice. Mol Psychiatry 2017; 22:689-702. [PMID: 27021819 PMCID: PMC5014121 DOI: 10.1038/mp.2016.30] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 12/23/2015] [Accepted: 01/25/2016] [Indexed: 02/08/2023]
Abstract
SHANK3 (also called PROSAP2) genetic haploinsufficiency is thought to be the major cause of neuropsychiatric symptoms in Phelan-McDermid syndrome (PMS). PMS is a rare genetic disorder that causes a severe form of intellectual disability (ID), expressive language delays and other autistic features. Furthermore, a significant number of SHANK3 mutations have been identified in patients with autism spectrum disorders (ASD), and SHANK3 truncating mutations are associated with moderate to profound ID. The Shank3 protein is a scaffold protein that is located in the postsynaptic density (PSD) of excitatory synapses and is crucial for synapse development and plasticity. In this study, we investigated the molecular mechanisms associated with the ASD-like behaviors observed in Shank3Δ11-/- mice, in which exon 11 has been deleted. Our results indicate that Shank3 is essential to mediating metabotropic glutamate receptor 5 (mGlu5)-receptor signaling by recruiting Homer1b/c to the PSD, specifically in the striatum and cortex. Moreover, augmenting mGlu5-receptor activity by administering 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide ameliorated the functional and behavioral defects that were observed in Shank3Δ11-/- mice, suggesting that pharmaceutical treatments that increase mGlu5 activity may represent a new approach for treating patients that are affected by PMS and SHANK3 mutations.
Collapse
Affiliation(s)
| | | | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università degli Studi
del Piemonte Orientale “Amedeo Avogadro”, Novara
| | | | | | - Noemi Morello
- Department of Neuroscience, University of Turin, Torino
| | | | - Alessandro Tozzi
- University of Perugia, Department of Experimental Medicine,
Perugia
| | - Valentina Durante
- Department of Medicine, University of Perugia and Clinica
Neurologica, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Paolo Scalmani
- U.O. of Neurophysiopathology and Diagnostic Epileptology, Foundation
Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Neurological Institute
Carlo Besta, Milan
| | - Massimo Mantegazza
- Institute of Molecular and Cellular Pharmacology (IPMC), Laboratory
of Excellence Ion Channel Science and Therapeutics (LabEx ICST), CNRS UMR7275 and
University of Nice-Sophia Antipolis, Valbonne
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università degli Studi
del Piemonte Orientale “Amedeo Avogadro”, Novara
| | | | - Mariaelvina Sala
- CNR Neuroscience Institute, Milan, Milano,Institute for Anatomy and Cell Biology, Ulm University, Ulm
| | - Paolo Calabresi
- Department of Medicine, University of Perugia and Clinica
Neurologica, Santa Maria della Misericordia Hospital, Perugia, Italy
| | | | - Carlo Sala
- CNR Neuroscience Institute, Milan, Milano
| | | |
Collapse
|
131
|
Hong F, Ze Y, Zhou Y, Hong J, Yu X, Sheng L, Wang L. Nanoparticulate TiO 2 -mediated inhibition of the Wnt signaling pathway causes dendritic development disorder in cultured rat hippocampal neurons. J Biomed Mater Res A 2017; 105:2139-2149. [PMID: 28371053 DOI: 10.1002/jbm.a.36073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 02/26/2017] [Accepted: 03/24/2017] [Indexed: 11/11/2022]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) are increasingly used in daily life, in industry, and in environmental clearing, but their potential neurodevelopmental toxicity has been highly debated. In this study, we explored whether TiO2 NPs inhibited development of dendritic morphology and identified possible molecular mechanisms associated with this inhibition in primary cultured rat hippocampal neurons. Results showed that TiO2 NPs decreased neurite length, the number of branches and the spine density, and impaired mitochondrial function in the developing neurons. Furthermore, TiO2 NPs significantly reduced the expression of several proteins involved in canonical Wnt3a/β-catenin signaling including Wnt3a, β-catenin, p-GSK-3β, and CyclinD1 and conversely, elevated GSK-3β expression. In addition to altering expression of proteins involved in canonical Wnt3a/β-catenin signaling, TiO2 NPs decreased expression of proteins invovled in non-canonical Wnt signaling, including, MKLP1, CRMP3, ErbB4, and KIF17. Taken together, these results indicate that suppression of dendritic development caused by TiO2 NPs is associated with inhibition of activation of the Wnt/β-catenin pathway or non-canonical Wnt pathway-induced expression of microtubule cytoskeletal components in the developing neurons. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2139-2149, 2017.
Collapse
Affiliation(s)
- Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, 223300, China.,Jiangsu Key Laboratory for Food Safety and Nutritional Function, Huaiyin Normal University, Huaian, 223300, China.,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Yuguan Ze
- Medical College of Soochow University, Suzhou, 215123, China
| | - Yaoming Zhou
- Food Department, Jiangsu Food and Pharmaceutical Science College, Huaian, 223303, China
| | - Jie Hong
- Medical College of Soochow University, Suzhou, 215123, China
| | - Xiaohon Yu
- Medical College of Soochow University, Suzhou, 215123, China
| | - Lei Sheng
- Medical College of Soochow University, Suzhou, 215123, China
| | - Ling Wang
- Library of Soochow University, Suzhou, China, Suzhou, 215123, China
| |
Collapse
|
132
|
Wang W, Li C, Chen Q, van der Goes MS, Hawrot J, Yao AY, Gao X, Lu C, Zang Y, Zhang Q, Lyman K, Wang D, Guo B, Wu S, Gerfen CR, Fu Z, Feng G. Striatopallidal dysfunction underlies repetitive behavior in Shank3-deficient model of autism. J Clin Invest 2017; 127:1978-1990. [PMID: 28414301 DOI: 10.1172/jci87997] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 02/16/2017] [Indexed: 11/17/2022] Open
Abstract
The postsynaptic scaffolding protein SH3 and multiple ankyrin repeat domains 3 (SHANK3) is critical for the development and function of glutamatergic synapses. Disruption of the SHANK3-encoding gene has been strongly implicated as a monogenic cause of autism, and Shank3 mutant mice show repetitive grooming and social interaction deficits. Although basal ganglia dysfunction has been proposed to underlie repetitive behaviors, few studies have provided direct evidence to support this notion and the exact cellular mechanisms remain largely unknown. Here, we utilized the Shank3B mutant mouse model of autism to investigate how Shank3 mutation may differentially affect striatonigral (direct pathway) and striatopallidal (indirect pathway) medium spiny neurons (MSNs) and its relevance to repetitive grooming behavior in Shank3B mutant mice. We found that Shank3 deletion preferentially affects synapses onto striatopallidal MSNs. Striatopallidal MSNs showed profound defects, including alterations in synaptic transmission, synaptic plasticity, and spine density. Importantly, the repetitive grooming behavior was rescued by selectively enhancing the striatopallidal MSN activity via a Gq-coupled human M3 muscarinic receptor (hM3Dq), a type of designer receptors exclusively activated by designer drugs (DREADD). Our findings directly demonstrate the existence of distinct changes between 2 striatal pathways in a mouse model of autism and indicate that the indirect striatal pathway disruption might play a causative role in repetitive behavior of Shank3B mutant mice.
Collapse
|
133
|
Martínez-Cerdeño V. Dendrite and spine modifications in autism and related neurodevelopmental disorders in patients and animal models. Dev Neurobiol 2017; 77:393-404. [PMID: 27390186 PMCID: PMC5219951 DOI: 10.1002/dneu.22417] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Dendrites and spines are the main neuronal structures receiving input from other neurons and glial cells. Dendritic and spine number, size, and morphology are some of the crucial factors determining how signals coming from individual synapses are integrated. Much remains to be understood about the characteristics of neuronal dendrites and dendritic spines in autism and related disorders. Although there have been many studies conducted using autism mouse models, few have been carried out using postmortem human tissue from patients. Available animal models of autism include those generated through genetic modifications and those non-genetic models of the disease. Here, we review how dendrite and spine morphology and number is affected in autism and related neurodevelopmental diseases, both in human, and genetic and non-genetic animal models of autism. Overall, data obtained from human and animal models point to a generalized reduction in the size and number, as well as an alteration of the morphology of dendrites; and an increase in spine densities with immature morphology, indicating a general spine immaturity state in autism. Additional human studies on dendrite and spine number and morphology in postmortem tissue are needed to understand the properties of these structures in the cerebral cortex of patients with autism. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.
Collapse
Affiliation(s)
- Verónica Martínez-Cerdeño
- Department of Pathology and Laboratory Medicine, UC Davis, Sacramento, California
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, North California, Sacramento, California
- MIND Institute, UC Davis School of Medicine, Sacramento, California
| |
Collapse
|
134
|
Buscemi L, Ginet V, Lopatar J, Montana V, Pucci L, Spagnuolo P, Zehnder T, Grubišić V, Truttman A, Sala C, Hirt L, Parpura V, Puyal J, Bezzi P. Homer1 Scaffold Proteins Govern Ca2+ Dynamics in Normal and Reactive Astrocytes. Cereb Cortex 2017; 27:2365-2384. [PMID: 27075036 PMCID: PMC5963825 DOI: 10.1093/cercor/bhw078] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In astrocytes, the intracellular calcium (Ca2+) signaling mediated by activation of metabotropic glutamate receptor 5 (mGlu5) is crucially involved in the modulation of many aspects of brain physiology, including gliotransmission. Here, we find that the mGlu5-mediated Ca2+ signaling leading to release of glutamate is governed by mGlu5 interaction with Homer1 scaffolding proteins. We show that the long splice variants Homer1b/c are expressed in astrocytic processes, where they cluster with mGlu5 at sites displaying intense local Ca2+ activity. We show that the structural and functional significance of the Homer1b/c-mGlu5 interaction is to relocate endoplasmic reticulum (ER) to the proximity of the plasma membrane and to optimize Ca2+ signaling and glutamate release. We also show that in reactive astrocytes the short dominant-negative splice variant Homer1a is upregulated. Homer1a, by precluding the mGlu5-ER interaction decreases the intensity of Ca2+ signaling thus limiting the intensity and the duration of glutamate release by astrocytes. Hindering upregulation of Homer1a with a local injection of short interfering RNA in vivo restores mGlu5-mediated Ca2+ signaling and glutamate release and sensitizes astrocytes to apoptosis. We propose that Homer1a may represent one of the cellular mechanisms by which inflammatory astrocytic reactions are beneficial for limiting brain injury.
Collapse
Affiliation(s)
- Lara Buscemi
- Department of Fundamental Neurosciences, University of Lausanne, CH1005Lausanne, Switzerland
- Stroke Laboratory, Neurology Service, Department of Clinical Neurosciences, University Hospital Centre and University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Vanessa Ginet
- Department of Fundamental Neurosciences, University of Lausanne, CH1005Lausanne, Switzerland
- Division of Neonatology, Department of Paediatrics and Paediatric Surgery, University Hospital Centre and University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Jan Lopatar
- Department of Fundamental Neurosciences, University of Lausanne, CH1005Lausanne, Switzerland
| | - Vedrana Montana
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
- Department of Neurobiology, Center for Glial Biology in Medicine, Civitan International Research Center, Atomic Force Microscopy and Nanotechnology Laboratories, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Luca Pucci
- Department of Fundamental Neurosciences, University of Lausanne, CH1005Lausanne, Switzerland
| | - Paola Spagnuolo
- Department of Fundamental Neurosciences, University of Lausanne, CH1005Lausanne, Switzerland
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Tamara Zehnder
- Department of Fundamental Neurosciences, University of Lausanne, CH1005Lausanne, Switzerland
| | - Vladimir Grubišić
- Department of Neurobiology, Center for Glial Biology in Medicine, Civitan International Research Center, Atomic Force Microscopy and Nanotechnology Laboratories, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anita Truttman
- Division of Neonatology, Department of Paediatrics and Paediatric Surgery, University Hospital Centre and University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Carlo Sala
- CNR Institute of Neuroscience and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Lorenz Hirt
- Stroke Laboratory, Neurology Service, Department of Clinical Neurosciences, University Hospital Centre and University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine, Civitan International Research Center, Atomic Force Microscopy and Nanotechnology Laboratories, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, CH1005Lausanne, Switzerland
- Division of Neonatology, Department of Paediatrics and Paediatric Surgery, University Hospital Centre and University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Paola Bezzi
- Department of Fundamental Neurosciences, University of Lausanne, CH1005Lausanne, Switzerland
| |
Collapse
|
135
|
Emerging Synaptic Molecules as Candidates in the Etiology of Neurological Disorders. Neural Plast 2017; 2017:8081758. [PMID: 28331639 PMCID: PMC5346360 DOI: 10.1155/2017/8081758] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023] Open
Abstract
Synapses are complex structures that allow communication between neurons in the central nervous system. Studies conducted in vertebrate and invertebrate models have contributed to the knowledge of the function of synaptic proteins. The functional synapse requires numerous protein complexes with specialized functions that are regulated in space and time to allow synaptic plasticity. However, their interplay during neuronal development, learning, and memory is poorly understood. Accumulating evidence links synapse proteins to neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. In this review, we describe the way in which several proteins that participate in cell adhesion, scaffolding, exocytosis, and neurotransmitter reception from presynaptic and postsynaptic compartments, mainly from excitatory synapses, have been associated with several synaptopathies, and we relate their functions to the disease phenotype.
Collapse
|
136
|
Reim D, Distler U, Halbedl S, Verpelli C, Sala C, Bockmann J, Tenzer S, Boeckers TM, Schmeisser MJ. Proteomic Analysis of Post-synaptic Density Fractions from Shank3 Mutant Mice Reveals Brain Region Specific Changes Relevant to Autism Spectrum Disorder. Front Mol Neurosci 2017; 10:26. [PMID: 28261056 PMCID: PMC5306440 DOI: 10.3389/fnmol.2017.00026] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/23/2017] [Indexed: 12/03/2022] Open
Abstract
Disruption of the human SHANK3 gene can cause several neuropsychiatric disease entities including Phelan-McDermid syndrome, autism spectrum disorder (ASD), and intellectual disability. Although, a wide array of neurobiological studies strongly supports a major role for SHANK3 in organizing the post-synaptic protein scaffold, the molecular processes at synapses of individuals harboring SHANK3 mutations are still far from being understood. In this study, we biochemically isolated the post-synaptic density (PSD) fraction from striatum and hippocampus of adult Shank3Δ11-/- mutant mice and performed ion-mobility enhanced data-independent label-free LC-MS/MS to obtain the corresponding PSD proteomes (Data are available via ProteomeXchange with identifier PXD005192). This unbiased approach to identify molecular disturbances at Shank3 mutant PSDs revealed hitherto unknown brain region specific alterations including a striatal decrease of several molecules encoded by ASD susceptibility genes such as the serine/threonine kinase Cdkl5 and the potassium channel KCa1.1. Being the first comprehensive analysis of brain region specific PSD proteomes from a Shank3 mutant line, our study provides crucial information on molecular alterations that could foster translational treatment studies for SHANK3 mutation-associated synaptopathies and possibly also ASD in general.
Collapse
Affiliation(s)
- Dominik Reim
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany
- International Graduate School in Molecular Medicine, Ulm UniversityUlm, Germany
| | - Ute Distler
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University MainzMainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of the Johannes-Gutenberg University MainzMainz, Germany
| | - Sonja Halbedl
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany
- International Graduate School in Molecular Medicine, Ulm UniversityUlm, Germany
| | - Chiara Verpelli
- CNR Neuroscience InstituteMilan, Italy
- BIOMETRA, University of MilanMilan, Italy
| | - Carlo Sala
- CNR Neuroscience InstituteMilan, Italy
- BIOMETRA, University of MilanMilan, Italy
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany
| | - Stefan Tenzer
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University MainzMainz, Germany
| | | | - Michael J. Schmeisser
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany
- Division of Neuroanatomy, Institute of Anatomy, Otto-von-Guericke UniversityMagdeburg, Germany
- Leibniz Institute for NeurobiologyMagdeburg, Germany
| |
Collapse
|
137
|
Abstract
Several large-scale genomic studies have supported an association between cases of autism spectrum disorder and mutations in the genes SH3 and multiple ankyrin repeat domains protein 1 (SHANK1), SHANK2 and SHANK3, which encode a family of postsynaptic scaffolding proteins that are present at glutamatergic synapses in the CNS. An evaluation of human genetic data, as well as of in vitro and in vivo animal model data, may allow us to understand how disruption of SHANK scaffolding proteins affects the structure and function of neural circuits and alters behaviour.
Collapse
|
138
|
Niu Y, Dai Z, Liu W, Zhang C, Yang Y, Guo Z, Li X, Xu C, Huang X, Wang Y, Shi YS, Liu JJ. Ablation of SNX6 leads to defects in synaptic function of CA1 pyramidal neurons and spatial memory. eLife 2017; 6. [PMID: 28134614 PMCID: PMC5323044 DOI: 10.7554/elife.20991] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 01/28/2017] [Indexed: 11/14/2022] Open
Abstract
SNX6 is a ubiquitously expressed PX-BAR protein that plays important roles in retromer-mediated retrograde vesicular transport from endosomes. Here we report that CNS-specific Snx6 knockout mice exhibit deficits in spatial learning and memory, accompanied with loss of spines from distal dendrites of hippocampal CA1 pyramidal cells. SNX6 interacts with Homer1b/c, a postsynaptic scaffold protein crucial for the synaptic distribution of other postsynaptic density (PSD) proteins and structural integrity of dendritic spines. We show that SNX6 functions independently of retromer to regulate distribution of Homer1b/c in the dendritic shaft. We also find that Homer1b/c translocates from shaft to spines by protein diffusion, which does not require SNX6. Ablation of SNX6 causes reduced distribution of Homer1b/c in distal dendrites, decrease in surface levels of AMPAR and impaired AMPAR-mediated synaptic transmission. These findings reveal a physiological role of SNX6 in CNS excitatory neurons. DOI:http://dx.doi.org/10.7554/eLife.20991.001 Neurons are the building blocks of the nervous system. These cells generally consist of a round portion called the cell body and a long cable-like axon. The cell body bears numerous branches called dendrites, which are in turn covered in spines. Neurons communicate with one another at junctions – or synapses – that typically form between the end of the axon of one cell and a dendritic spine on another. Specialized proteins stabilize the dendritic spines and enable the cells to exchange messages across the synapse. However, it is the cell body – rather than the dendrites – that produces most of these proteins. Structures called molecular motors transport proteins to their destinations within the cell along fixed tracks, similar to how a freight train carries cargo over the rail network. One of the key molecular motors within neurons is called dynein‒dynactin. This in turn interacts with other proteins called adaptors, enabling it to transport specific types of cargo. Niu, Dai, Liu et al. have now examined the role of SNX6, an adaptor protein for the dynein‒dynactin motor. Mice that have been genetically modified to lack SNX6 in their brains have fewer spines on their dendrites compared with normal mice. This was particularly true for dendrites that contain AMPAR, a protein that receives signals sent across synapses. Niu, Dai, Liu et al. showed that SNX6 interacts with another protein called Homer1b/c and is responsible for distributing this protein in dendrites far from the cell body. The Homer1b/c protein helps to stabilize dendritic spines and to regulate the number of AMPAR proteins within them. Mice that lack SNX6 therefore have less Homer1b/c in the dendrites furthest from the cell body, and fewer spines on these dendrites too. These mice also have fewer AMPAR proteins at their synapses than control mice. Mice that lack SNX6 show impaired learning and memory compared to control mice. This is consistent with the fact that changes in the strength of synapses that possess AMPAR proteins are thought to underlie learning and memory. Additional experiments are required to explore these relationships further, and to determine whether SNX6 helps to localize any other proteins that also contribute to changes in the strength of synapses. DOI:http://dx.doi.org/10.7554/eLife.20991.002
Collapse
Affiliation(s)
- Yang Niu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Zhonghua Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wenxue Liu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China.,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China.,Model Animal Research Center, Nanjing University, Nanjing, China
| | - Cheng Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yanrui Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhenzhen Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyu Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Chenchang Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yun S Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China.,Model Animal Research Center, Nanjing University, Nanjing, China
| | - Jia-Jia Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
139
|
Homer, Spikar, and Other Drebrin-Binding Proteins in the Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:249-268. [PMID: 28865024 DOI: 10.1007/978-4-431-56550-5_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drebrin is a major F-actin-binding protein in the brain. In the past two decades, many drebrin-binding proteins in addition to F-actin have been identified in several research fields including neuroscience, oncology, and immunology. Among the drebrin-binding proteins, there are various kinds of proteins including scaffold proteins, nuclear proteins, phosphatases, microtubule-binding proteins, G-actin-binding proteins, gap junction proteins, chemokine receptors, and cell-adhesion-related proteins. The interaction between drebrin and its binding partners seems to play important roles in higher brain functions, because drebrin is involved in the pathogenesis of some neurological diseases with cognitive defects. In this chapter, we will first review the interaction of Homer and spikar with drebrin, particularly focusing on spine morphogenesis and synaptic function. Homer contributes to spine morphogenesis by cooperating with shank and activated Cdc42 small GTPase, suggesting a novel signaling pathway comprising Homer, drebrin, shank, and Cdc42 for spine morphogenesis. Drebrin sequesters spikar in the cytoplasm and stabilizes it in dendritic spines, leading to spine formation. Finally, we will introduce some other drebrin-binding proteins including end-binding protein 3 (EB3), profilin, progranulin, and phosphatase and tensin homologue (PTEN). These proteins are involved in Alzheimer's disease and cancer. Therefore, further studies on drebrin and its binding proteins will be of great importance to elucidate the pathologies of various diseases and may contribute to their medical treatment and diagnostics development.
Collapse
|
140
|
|
141
|
Nie J, Yang X. Modulation of Synaptic Plasticity by Exercise Training as a Basis for Ischemic Stroke Rehabilitation. Cell Mol Neurobiol 2017; 37:5-16. [PMID: 26910247 PMCID: PMC11482112 DOI: 10.1007/s10571-016-0348-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/11/2016] [Indexed: 12/23/2022]
Abstract
In recent years, rehabilitation of ischemic stroke draws more and more attention in the world, and has been linked to changes of synaptic plasticity. Exercise training improves motor function of ischemia as well as cognition which is associated with formation of learning and memory. The molecular basis of learning and memory might be synaptic plasticity. Research has therefore been conducted in an attempt to relate effects of exercise training to neuroprotection and neurogenesis adjacent to the ischemic injury brain. The present paper reviews the current literature addressing this question and discusses the possible mechanisms involved in modulation of synaptic plasticity by exercise training. This review shows the pathological process of synaptic dysfunction in ischemic roughly and then discusses the effects of exercise training on scaffold proteins and regulatory protein expression. The expression of scaffold proteins generally increased after training, but the effects on regulatory proteins were mixed. Moreover, the compositions of postsynaptic receptors were changed and the strength of synaptic transmission was enhanced after training. Finally, the recovery of cognition is critically associated with synaptic remodeling in an injured brain, and the remodeling occurs through a number of local regulations including mRNA translation, remodeling of cytoskeleton, and receptor trafficking into and out of the synapse. We do provide a comprehensive knowledge of synaptic plasticity enhancement obtained by exercise training in this review.
Collapse
Affiliation(s)
- Jingjing Nie
- Department of Neurology, Xiang Ya Hospital, Central South University, Xiang Ya Road 87, Changsha, 410008, Hunan, China
| | - Xiaosu Yang
- Department of Neurology, Xiang Ya Hospital, Central South University, Xiang Ya Road 87, Changsha, 410008, Hunan, China.
| |
Collapse
|
142
|
Lin YC, Frei JA, Kilander MBC, Shen W, Blatt GJ. A Subset of Autism-Associated Genes Regulate the Structural Stability of Neurons. Front Cell Neurosci 2016; 10:263. [PMID: 27909399 PMCID: PMC5112273 DOI: 10.3389/fncel.2016.00263] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/28/2016] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) comprises a range of neurological conditions that affect individuals’ ability to communicate and interact with others. People with ASD often exhibit marked qualitative difficulties in social interaction, communication, and behavior. Alterations in neurite arborization and dendritic spine morphology, including size, shape, and number, are hallmarks of almost all neurological conditions, including ASD. As experimental evidence emerges in recent years, it becomes clear that although there is broad heterogeneity of identified autism risk genes, many of them converge into similar cellular pathways, including those regulating neurite outgrowth, synapse formation and spine stability, and synaptic plasticity. These mechanisms together regulate the structural stability of neurons and are vulnerable targets in ASD. In this review, we discuss the current understanding of those autism risk genes that affect the structural connectivity of neurons. We sub-categorize them into (1) cytoskeletal regulators, e.g., motors and small RhoGTPase regulators; (2) adhesion molecules, e.g., cadherins, NCAM, and neurexin superfamily; (3) cell surface receptors, e.g., glutamatergic receptors and receptor tyrosine kinases; (4) signaling molecules, e.g., protein kinases and phosphatases; and (5) synaptic proteins, e.g., vesicle and scaffolding proteins. Although the roles of some of these genes in maintaining neuronal structural stability are well studied, how mutations contribute to the autism phenotype is still largely unknown. Investigating whether and how the neuronal structure and function are affected when these genes are mutated will provide insights toward developing effective interventions aimed at improving the lives of people with autism and their families.
Collapse
Affiliation(s)
- Yu-Chih Lin
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Jeannine A Frei
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Michaela B C Kilander
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Wenjuan Shen
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Gene J Blatt
- Laboratory of Autism Neurocircuitry, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| |
Collapse
|
143
|
Bennett MR, Arnold J, Hatton SN, Lagopoulos J. Regulation of fear extinction by long-term depression: The roles of endocannabinoids and brain derived neurotrophic factor. Behav Brain Res 2016; 319:148-164. [PMID: 27867101 DOI: 10.1016/j.bbr.2016.11.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 11/19/2022]
Abstract
The extinction of a conditioned fear response is of great interest in the search for a means of ameliorating adverse neurobiological changes resulting from stress. The discovery that endocannibinoid (EC) levels are inversely related to the extent of such stress, and that the amygdala is a primary site mediating stress, suggests that ECs in this brain region might play a major role in extinction. Supporting this are the observations that the basolateral complex of the amygdala shows an increase in ECs only during extinction and that early clinical trials indicate that cannabinoid-like agents, when taken orally by patients suffering from post traumatic stress disorder (PTSD), reduce insomnia and nightmares. In order to optimize the potential of these agents to ameliorate symptoms of PTSD four important questions need to be answered: first, what is the identity of the cells that release ECs in the amygdala during extinction; second, what are their sites of action; third, what roles do the ECs play in the alleviation of long- depression (LTD), a process central to extinction; and finally, to what extent does brain derived neurotrophic factor (BDNF) facilitate the release of ECs? A review of the relevant literature is presented in an attempt to answer these questions. It is suggested that the principal cell involved in EC synthesis and release during extinction is the so-called excitatory extinction neuron in the basal nucleus of the amygdala. Furthermore that the main site of action of the ECs is the adjacent calcitonin gene-related peptide inhibitory interneurons, whose normal role of blocking the excitatory neurons is greatly diminished. The molecular pathways leading (during extinction trials) to the synthesis and release of ECs from synaptic spines of extinction neurons, that is potentiated by BDNF, are also delineated in this review. Finally, consideration is given to how the autocrine action of BDNF, linked to the release of ECs, can lead to the sustained release of these, so maintaining extinction over long times.
Collapse
Affiliation(s)
- Maxwell R Bennett
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.
| | - Jonathon Arnold
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Sean N Hatton
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Jim Lagopoulos
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia; The Sunshine Coast Mind and Neuroscience, Thompson Institute, The University of the Sunshine Coast, QLD, Australia
| |
Collapse
|
144
|
Determining the Roles of Inositol Trisphosphate Receptors in Neurodegeneration: Interdisciplinary Perspectives on a Complex Topic. Mol Neurobiol 2016; 54:6870-6884. [PMID: 27771899 DOI: 10.1007/s12035-016-0205-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023]
Abstract
It is well known that calcium (Ca2+) is involved in the triggering of neuronal death. Ca2+ cytosolic levels are regulated by Ca2+ release from internal stores located in organelles, such as the endoplasmic reticulum. Indeed, Ca2+ transit from distinct cell compartments follows complex dynamics that are mediated by specific receptors, notably inositol trisphosphate receptors (IP3Rs). Ca2+ release by IP3Rs plays essential roles in several neurological disorders; however, details of these processes are poorly understood. Moreover, recent studies have shown that subcellular location, molecular identity, and density of IP3Rs profoundly affect Ca2+ transit in neurons. Therefore, regulation of IP3R gene products in specific cellular vicinities seems to be crucial in a wide range of cellular processes from neuroprotection to neurodegeneration. In this regard, microRNAs seem to govern not only IP3Rs translation levels but also subcellular accumulation. Combining new data from molecular cell biology with mathematical modelling, we were able to summarize the state of the art on this topic. In addition to presenting how Ca2+ dynamics mediated by IP3R activation follow a stochastic regimen, we integrated a theoretical approach in an easy-to-apply, cell biology-coherent fashion. Following the presented premises and in contrast to previously tested hypotheses, Ca2+ released by IP3Rs may play different roles in specific neurological diseases, including Alzheimer's disease and Parkinson's disease.
Collapse
|
145
|
Stanika R, Campiglio M, Pinggera A, Lee A, Striessnig J, Flucher BE, Obermair GJ. Splice variants of the Ca V1.3 L-type calcium channel regulate dendritic spine morphology. Sci Rep 2016; 6:34528. [PMID: 27708393 PMCID: PMC5052568 DOI: 10.1038/srep34528] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 09/15/2016] [Indexed: 01/13/2023] Open
Abstract
Dendritic spines are the postsynaptic compartments of glutamatergic synapses in the brain. Their number and shape are subject to change in synaptic plasticity and neurological disorders including autism spectrum disorders and Parkinson’s disease. The L-type calcium channel CaV1.3 constitutes an important calcium entry pathway implicated in the regulation of spine morphology. Here we investigated the importance of full-length CaV1.3L and two C-terminally truncated splice variants (CaV1.342A and CaV1.343S) and their modulation by densin-180 and shank1b for the morphology of dendritic spines of cultured hippocampal neurons. Live-cell immunofluorescence and super-resolution microscopy of epitope-tagged CaV1.3L revealed its localization at the base-, neck-, and head-region of dendritic spines. Expression of the short splice variants or deletion of the C-terminal PDZ-binding motif in CaV1.3L induced aberrant dendritic spine elongation. Similar morphological alterations were induced by co-expression of densin-180 or shank1b with CaV1.3L and correlated with increased CaV1.3 currents and dendritic calcium signals in transfected neurons. Together, our findings suggest a key role of CaV1.3 in regulating dendritic spine structure. Under physiological conditions it may contribute to the structural plasticity of glutamatergic synapses. Conversely, altered regulation of CaV1.3 channels may provide an important mechanism in the development of postsynaptic aberrations associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Ruslan Stanika
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Marta Campiglio
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Alexandra Pinggera
- Department of Pharmacology and Toxicology, University of Innsbruck, 6020 Innsbruck, Austria
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, Otolaryngology Head-Neck Surgery, and Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, University of Innsbruck, 6020 Innsbruck, Austria
| | - Bernhard E Flucher
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Gerald J Obermair
- Division of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
146
|
Actin-Dependent Alterations of Dendritic Spine Morphology in Shankopathies. Neural Plast 2016; 2016:8051861. [PMID: 27795858 PMCID: PMC5067329 DOI: 10.1155/2016/8051861] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/30/2016] [Indexed: 12/11/2022] Open
Abstract
Shank proteins (Shank1, Shank2, and Shank3) act as scaffolding molecules in the postsynaptic density of many excitatory neurons. Mutations in SHANK genes, in particular SHANK2 and SHANK3, lead to autism spectrum disorders (ASD) in both human and mouse models. Shank3 proteins are made of several domains-the Shank/ProSAP N-terminal (SPN) domain, ankyrin repeats, SH3 domain, PDZ domain, a proline-rich region, and the sterile alpha motif (SAM) domain. Via various binding partners of these domains, Shank3 is able to bind and interact with a wide range of proteins including modulators of small GTPases such as RICH2, a RhoGAP protein, and βPIX, a RhoGEF protein for Rac1 and Cdc42, actin binding proteins and actin modulators. Dysregulation of all isoforms of Shank proteins, but especially Shank3, leads to alterations in spine morphogenesis, shape, and activity of the synapse via altering actin dynamics. Therefore, here, we highlight the role of Shank proteins as modulators of small GTPases and, ultimately, actin dynamics, as found in multiple in vitro and in vivo models. The failure to mediate this regulatory role might present a shared mechanism in the pathophysiology of autism-associated mutations, which leads to dysregulation of spine morphogenesis and synaptic signaling.
Collapse
|
147
|
de Bartolomeis A, Marmo F, Buonaguro EF, Latte G, Tomasetti C, Iasevoli F. Switching antipsychotics: Imaging the differential effect on the topography of postsynaptic density transcripts in antipsychotic-naïve vs. antipsychotic-exposed rats. Prog Neuropsychopharmacol Biol Psychiatry 2016; 70:24-38. [PMID: 27177972 DOI: 10.1016/j.pnpbp.2016.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/17/2016] [Accepted: 04/27/2016] [Indexed: 10/24/2022]
Abstract
The postsynaptic density (PSD) has been regarded as a functional switchboard at the crossroads of a dopamine-glutamate interaction, and it is putatively involved in the pathophysiology of psychosis. Indeed, it has been demonstrated that antipsychotics may modulate several PSD transcripts, such as PSD-95, Shank, and Homer. Despite switching antipsychotics is a frequent strategy to counteract lack of efficacy and/or side effect onset in clinical practice, no information is available on the effects of sequential treatments with different antipsychotics on PSD molecules. The aim of this study was to evaluate whether a previous exposure to a typical antipsychotic and a switch to an atypical one may affect the expression of PSD transcripts, in order to evaluate potential neurobiological correlates of this common clinical practice, with specific regards to putative synaptic plasticity processes. We treated male Sprague-Dawley rats intraperitoneally for 15days with haloperidol or vehicle, then from the sixteenth day we switched the animals to amisulpride or continued to treat them with vehicle or haloperidol for 15 additional days. In this way we got six first treatment/second treatment groups: vehicle/vehicle, vehicle/haloperidol, vehicle/amisulpride, haloperidol/vehicle, haloperidol/haloperidol, haloperidol/amisulpride. In this paradigm, we evaluated the expression of brain transcripts belonging to relevant and interacting PSD proteins, both of the Immediate-Early Gene (Homer1a, Arc) and the constitutive classes (Homer1b/c and PSD-95). The major finding was the differential effect of amisulpride on gene transcripts when administered in naïve vs. antipsychotic-pretreated rats, with modifications of the ratio between Homer1a/Homer1b transcripts and differential effects in cortex and striatum. These results suggest that the neurobiological effects on PSD transcripts of amisulpride, and possibly of other antipsychotics, may be greatly affected by prior antipsychotic treatments and may impact significantly on the switching procedure.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy.
| | - Federica Marmo
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Gianmarco Latte
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| |
Collapse
|
148
|
Putative Cell Adhesion Membrane Protein Vstm5 Regulates Neuronal Morphology and Migration in the Central Nervous System. J Neurosci 2016; 36:10181-97. [PMID: 27683913 DOI: 10.1523/jneurosci.0541-16.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/16/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED During brain development, dynamic changes in neuronal membranes perform critical roles in neuronal morphogenesis and migration to create functional neural circuits. Among the proteins that induce membrane dynamics, cell adhesion molecules are important in neuronal membrane plasticity. Here, we report that V-set and transmembrane domain-containing protein 5 (Vstm5), a cell-adhesion-like molecule belonging to the Ig superfamily, was found in mouse brain. Knock-down of Vstm5 in cultured hippocampal neurons markedly reduced the complexity of dendritic structures, as well as the number of dendritic filopodia. Vstm5 also regulates neuronal morphology by promoting dendritic protrusions that later develop into dendritic spines. Using electroporation in utero, we found that Vstm5 overexpression delayed neuronal migration and induced multiple branches in leading processes during corticogenesis. These results indicate that Vstm5 is a new cell-adhesion-like molecule and is critically involved in synaptogenesis and corticogenesis by promoting neuronal membrane dynamics. SIGNIFICANCE STATEMENT Neuronal migration and morphogenesis play critical roles in brain development and function. In this study, we demonstrate for the first time that V-set and transmembrane domain-containing protein 5 (Vstm5), a putative cell adhesion membrane protein, modulates both the position and complexity of central neurons by altering their membrane morphology and dynamics. Vstm5 is also one of the target genes responsible for variations in patient responses to treatments for major depressive disorder. Our results provide the first evidence that Vstm5 is a novel factor involved in the modulation of the neuronal membrane and a critical element in normal neural circuit formation during mammalian brain development.
Collapse
|
149
|
Elevated ERK/p90 ribosomal S6 kinase activity underlies audiogenic seizure susceptibility in fragile X mice. Proc Natl Acad Sci U S A 2016; 113:E6290-E6297. [PMID: 27663742 DOI: 10.1073/pnas.1610812113] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common heritable cause of intellectual disability and a leading genetic form of autism. The Fmr1 KO mouse, a model of FXS, exhibits elevated translation in the hippocampus and the cortex. ERK (extracellular signal-regulated kinase) and mTOR (mechanistic target of rapamycin) signaling regulate protein synthesis by activating downstream targets critical to translation initiation and elongation and are known to contribute to hippocampal defects in fragile X. Here we show that the effect of loss of fragile X mental retardation protein (FMRP) on these pathways is brain region specific. In contrast to the hippocampus, ERK (but not mTOR) signaling is elevated in the neocortex of fragile X mice. Phosphorylation of ribosomal protein S6, typically a downstream target of mTOR, is elevated in the neocortex, despite normal mTOR activity. This is significant in that S6 phosphorylation facilitates translation, correlates with neuronal activation, and is altered in neurodevelopmental disorders. We show that in fragile X mice, S6 is regulated by ERK via the "alternative" S6 kinase p90-ribosomal S6 kinase (RSK), as evidenced by the site of elevated phosphorylation and the finding that ERK inhibition corrects elevated RSK and S6 activity. These findings indicate that signaling networks are altered in the neocortex of fragile X mice such that S6 phosphorylation receives aberrant input from ERK/RSK. Importantly, an RSK inhibitor reduces susceptibility to audiogenic seizures in fragile X mice. Our findings identify RSK as a therapeutic target for fragile X and suggest the therapeutic potential of drugs for the treatment of FXS may vary in a brain-region-specific manner.
Collapse
|
150
|
Tong BCK, Lee CSK, Cheng WH, Lai KO, Foskett JK, Cheung KH. Familial Alzheimer's disease-associated presenilin 1 mutants promote γ-secretase cleavage of STIM1 to impair store-operated Ca2+ entry. Sci Signal 2016; 9:ra89. [PMID: 27601731 DOI: 10.1126/scisignal.aaf1371] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Some forms of familial Alzheimer's disease (FAD) are caused by mutations in presenilins (PSs), catalytic components of a γ-secretase complex that cleaves target proteins, including amyloid precursor protein (APP). Calcium (Ca(2+)) dysregulation in cells with these FAD-causing PS mutants has been attributed to attenuated store-operated Ca(2+) entry [SOCE; also called capacitative Ca(2+) entry (CCE)]. CCE occurs when STIM1 detects decreases in Ca(2+) in the endoplasmic reticulum (ER) and activates ORAI channels to replenish Ca(2+) stores in the ER. We showed that CCE was attenuated by PS1-associated γ-secretase activity. Endogenous PS1 and STIM1 interacted in human neuroblastoma SH-SY5Y cells, patient fibroblasts, and mouse primary cortical neurons. Forms of PS1 with FAD-associated mutations enhanced γ-secretase cleavage of the STIM1 transmembrane domain at a sequence that was similar to the γ-secretase cleavage sequence of APP. Cultured hippocampal neurons expressing mutant PS1 had attenuated CCE that was associated with destabilized dendritic spines, which were rescued by either γ-secretase inhibition or overexpression of STIM1. Our results indicate that γ-secretase activity may physiologically regulate CCE by targeting STIM1 and that restoring STIM1 may be a therapeutic approach in AD.
Collapse
Affiliation(s)
- Benjamin Chun-Kit Tong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Claire Shuk-Kwan Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Wing-Hei Cheng
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Kwok-On Lai
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China. State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - J Kevin Foskett
- Departments of Physiology and Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - King-Ho Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China. State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China. HKU-Shenzhen Institute of Research and Innovation, Shenzhen, Guangdong, China.
| |
Collapse
|