1
|
Xu Y, Sun H, Chen J, Qin L, Wu M, Zhong Z, Zhang X. Loss of SIL1 Affects Actin Dynamics and Leads to Abnormal Neural Migration. Mol Neurobiol 2025; 62:335-350. [PMID: 38850350 DOI: 10.1007/s12035-024-04272-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/29/2024] [Indexed: 06/10/2024]
Abstract
SIL1 is a nucleotide exchange factor for the molecular chaperone protein Bip in the endoplasmic reticulum that plays a crucial role in protein folding. The Sil1 gene is currently the only known causative gene of Marinesco-Sjögren syndrome (MSS). Intellectual developmental disability is the main symptom of MSS, and its mechanism has not been fully elucidated. Studies have shown that mutations in the Sil1 gene can delay neuronal migration during cortical development, but the underlying molecular mechanisms remain unclear. To further identify potential molecules involved in the regulation of central nervous system development by SIL1, we established a cortical neuron model with SIL1 protein deficiency and used proteomic analysis to screen for differentially expressed proteins after Sil1 silencing, followed by GO functional enrichment and protein‒protein interaction (PPI) network analysis. We identified 68 upregulated and 137 downregulated proteins in total, and among them, 10 upregulated and 3 downregulated proteins were mainly related to actin cytoskeleton dynamics. We further validated the differential changes in actin-related molecules using qRT‒PCR and Western blotting of a Sil1 gene knockout (Sil1-/-) mouse model. The results showed that the protein levels of ACTN1 and VIM decreased, while their mRNA levels increased as a compensatory response to protein deficiency. The mRNA and protein levels of IQGAP1 both showed a secondary increase. In conclusion, we identified ACTN1 and VIM as the key molecules regulated by SIL1 that are involved in neuronal migration during cortical development.
Collapse
Affiliation(s)
- Yuanyuan Xu
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Hongji Sun
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Junyang Chen
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Liuting Qin
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Mengxue Wu
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Zhaoming Zhong
- Department of Medical Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Xiaomin Zhang
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
2
|
Falahati H, Wu Y, Feuerer V, Simon HG, De Camilli P. Proximity proteomics of synaptopodin provides insight into the molecular composition of the spine apparatus of dendritic spines. Proc Natl Acad Sci U S A 2022; 119:e2203750119. [PMID: 36215465 PMCID: PMC9586327 DOI: 10.1073/pnas.2203750119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/02/2022] [Indexed: 02/03/2023] Open
Abstract
The spine apparatus is a specialized compartment of the neuronal smooth endoplasmic reticulum (ER) located in a subset of dendritic spines. It consists of stacks of ER cisterns that are interconnected by an unknown dense matrix and are continuous with each other and with the ER of the dendritic shaft. While this organelle was first observed over 60 y ago, its molecular organization remains a mystery. Here, we performed in vivo proximity proteomics to gain some insight into its molecular components. To do so, we used the only known spine apparatus-specific protein, synaptopodin, to target a biotinylating enzyme to this organelle. We validated the specific localization in dendritic spines of a small subset of proteins identified by this approach, and we further showed their colocalization with synaptopodin when expressed in nonneuronal cells. One such protein is Pdlim7, an actin binding protein not previously identified in spines. Pdlim7, which we found to interact with synaptopodin through multiple domains, also colocalizes with synaptopodin on the cisternal organelle, a peculiar stack of ER cisterns resembling the spine apparatus and found at axon initial segments of a subset of neurons. Moreover, Pdlim7 has an expression pattern similar to that of synaptopodin in the brain, highlighting a functional partnership between the two proteins. The components of the spine apparatus identified in this work will help elucidate mechanisms in the biogenesis and maintenance of this enigmatic structure with implications for the function of dendritic spines in physiology and disease.
Collapse
Affiliation(s)
- Hanieh Falahati
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
- HHMI, Yale University School of Medicine, New Haven, CT 06510
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510
| | - Yumei Wu
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
- HHMI, Yale University School of Medicine, New Haven, CT 06510
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510
| | - Vanessa Feuerer
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
- HHMI, Yale University School of Medicine, New Haven, CT 06510
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510
| | - Hans-Georg Simon
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University and Stanley Manne Children’s Research Institute, Chicago, IL 60611
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
- HHMI, Yale University School of Medicine, New Haven, CT 06510
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
3
|
Phldb2 is essential for regulating hippocampal dendritic spine morphology through drebrin in an adult-type isoform-specific manner. Neurosci Res 2022; 185:1-10. [PMID: 36162735 DOI: 10.1016/j.neures.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022]
Abstract
Morphologically dynamic dendritic spines are the major sites of neuronal plasticity in the brain; however, the molecular mechanisms underlying their morphological dynamics have not been fully elucidated. Phldb2 is a protein that contains two predicted coiled-coil domains and the pleckstrin homology domain, whose binding is highly sensitive to PIP3. We have previously demonstrated that Phldb2 regulates synaptic plasticity, glutamate receptor trafficking, and PSD-95 turnover. Drebrin is one of the most abundant neuron-specific F-actin-binding proteins that are pivotal for synaptic morphology and plasticity. We observed that Phldb2 bound to drebrin A (adult-type drebrin), but not to drebrin E (embryonic-type drebrin). In the absence of Phldb2, the subcellular localization of drebrin A in the hippocampal spines and its distribution in the hippocampus were altered. Immature spines, such as the filopodium type, increased relatively in the CA1 regions of the hippocampus, whereas mushroom spines, a typical mature type, decreased in Phldb2-/- mice. Phldb2 suppressed the formation of an abnormal filopodium structure induced by drebrin A overexpression. Taken together, these findings demonstrate that Phldb2 is pivotal for dendritic spine morphology and possibly for synaptic plasticity in mature animals by regulating drebrin A localization.
Collapse
|
4
|
Stress vulnerability shapes disruption of motor cortical neuroplasticity. Transl Psychiatry 2022; 12:91. [PMID: 35246507 PMCID: PMC8897461 DOI: 10.1038/s41398-022-01855-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Chronic stress is a major cause of neuropsychiatric conditions such as depression. Stress vulnerability varies individually in mice and humans, measured by behavioral changes. In contrast to affective symptoms, motor retardation as a consequence of stress is not well understood. We repeatedly imaged dendritic spines of the motor cortex in Thy1-GFP M mice before and after chronic social defeat stress. Susceptible and resilient phenotypes were discriminated by symptom load and their motor learning abilities were assessed by a gross and fine motor task. Stress phenotypes presented individual short- and long-term changes in the hypothalamic-pituitary-adrenal axis as well as distinct patterns of altered motor learning. Importantly, stress was generally accompanied by a marked reduction of spine density in the motor cortex and spine dynamics depended on the stress phenotype. We found astrogliosis and altered microglia morphology along with increased microglia-neuron interaction in the motor cortex of susceptible mice. In cerebrospinal fluid, proteomic fingerprints link the behavioral changes and structural alterations in the brain to neurodegenerative disorders and dysregulated synaptic homeostasis. Our work emphasizes the importance of synaptic integrity and the risk of neurodegeneration within depression as a threat to brain health.
Collapse
|
5
|
Keith RE, Ogoe RH, Dumas TC. Behind the scenes: Are latent memories supported by calcium independent plasticity? Hippocampus 2022; 32:73-88. [PMID: 33905147 PMCID: PMC8548406 DOI: 10.1002/hipo.23332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 02/03/2023]
Abstract
N-methyl-D-aspartate receptors (NMDARs) can be considered to be the de facto "plasticity" receptors in the brain due to their central role in the activity-dependent modification of neuronal morphology and synaptic transmission. Since the 1980s, research on NMDARs has focused on the second messenger properties of calcium and the downstream signaling pathways that mediate alterations in neural form and function. Recently, NMDARs were shown to drive activity-dependent synaptic plasticity without calcium influx. How this "nonionotropic" plasticity occurs in vitro is becoming clearer, but research on its involvement in behavior and cognition is in its infancy. There is a partial overlap in the downstream signaling molecules that are involved in ionotropic and nonionotropic NMDAR-dependent plasticity. Given this, and prior studies of the cognitive impacts of ionotropic NMDAR plasticity, a preliminary model explaining how NMDAR nonionotropic plasticity affects learning and memory can be established. We hypothesize that nonionotropic NMDAR plasticity takes part in latent memory encoding in immature rodents through nonassociative depression of synaptic efficacy, and possibly shrinking of dendritic spines. Further, the late postnatal alteration in NMDAR composition in the hippocampus appears to reduce nonionotropic signaling and remove a restriction on memory retrieval. This framework substantially alters the canonical model of NMDAR involvement in spatial cognition and hippocampal maturation and provides novel and exciting inroads for future studies.
Collapse
Affiliation(s)
- Rachel E. Keith
- Interdisciplinary Program in Neuroscience, College of Science, George Mason University, Fairfax, Virginia
| | - Richard H. Ogoe
- Department of Psychology, College of Humanities and Social Sciences, George Mason University, Fairfax, Virginia
| | - Theodore C. Dumas
- Interdisciplinary Program in Neuroscience, College of Science, George Mason University, Fairfax, Virginia,Department of Psychology, College of Humanities and Social Sciences, George Mason University, Fairfax, Virginia
| |
Collapse
|
6
|
Cullinan MM, Klipp RC, Bankston JR. Regulation of acid-sensing ion channels by protein binding partners. Channels (Austin) 2021; 15:635-647. [PMID: 34704535 PMCID: PMC8555555 DOI: 10.1080/19336950.2021.1976946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are a family of proton-gated cation channels that contribute to a diverse array of functions including pain sensation, cell death during ischemia, and more broadly to neurotransmission in the central nervous system. There is an increasing interest in understanding the physiological regulatory mechanisms of this family of channels. ASICs have relatively short N- and C-termini, yet a number of proteins have been shown to interact with these domains both in vitro and in vivo. These proteins can impact ASIC gating, localization, cell-surface expression, and regulation. Like all ion channels, it is important to understand the cellular context under which ASICs function in neurons and other cells. Here we will review what is known about a number of these potentially important regulatory molecules.
Collapse
Affiliation(s)
- Megan M Cullinan
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert C Klipp
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
7
|
Eliaz Y, Nedelec F, Morrison G, Levine H, Cheung MS. Insights from graph theory on the morphologies of actomyosin networks with multilinkers. Phys Rev E 2020; 102:062420. [PMID: 33466104 DOI: 10.1103/physreve.102.062420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/02/2020] [Indexed: 11/07/2022]
Abstract
Quantifying the influence of microscopic details on the dynamics of development of the overall structure of a filamentous network is important in a number of biologically relevant contexts, but it is not obvious what order parameters can be used to adequately describe this complex process. In this paper we investigated the role of multivalent actin-binding proteins (ABPs) in reorganizing actin filaments into higher-order complex networks via a computer model of semiflexible filaments. We characterize the importance of local connectivity among actin filaments, as well as the global features of actomyosin networks. We first map the networks into local graph representations and then, using principles from network-theory order parameters, combine properties from these representations to gain insight into the heterogeneous morphologies of actomyosin networks at a global level. We find that ABPs with a valency greater than 2 promote filament bundles and large filament clusters to a much greater extent than bivalent multilinkers. We also show that active myosinlike motor proteins promote the formation of dendritic branches from a stalk of actin bundles. Our work motivates future studies to embrace network theory as a tool to characterize complex morphologies of actomyosin detected by experiments, leading to a quantitative understanding of the role of ABPs in manipulating the self-assembly of actin filaments into unique architectures that underlie the structural scaffold of a cell relating to its mobility and shape.
Collapse
Affiliation(s)
- Yossi Eliaz
- Department of Physics, University of Houston, Houston, Texas 77204, USA
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
| | - Francois Nedelec
- Sainsbury Laboratory, Cambridge University, Bateman Street, CB2 1LR Cambridge, England, UK
| | - Greg Morrison
- Department of Physics, University of Houston, Houston, Texas 77204, USA
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
- Department of Physics, Northeastern University, Boston, Massachusetts 02115, USA
| | - Margaret S Cheung
- Department of Physics, University of Houston, Houston, Texas 77204, USA
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
- Department of Bioengineering, Rice University, Houston, Texas 77005, USA
| |
Collapse
|
8
|
Regulation of actin dynamics in dendritic spines: Nanostructure, molecular mobility, and signaling mechanisms. Mol Cell Neurosci 2020; 109:103564. [DOI: 10.1016/j.mcn.2020.103564] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/04/2020] [Indexed: 12/16/2022] Open
|
9
|
Malankhanova T, Suldina L, Grigor’eva E, Medvedev S, Minina J, Morozova K, Kiseleva E, Zakian S, Malakhova A. A Human Induced Pluripotent Stem Cell-Derived Isogenic Model of Huntington's Disease Based on Neuronal Cells Has Several Relevant Phenotypic Abnormalities. J Pers Med 2020; 10:jpm10040215. [PMID: 33182269 PMCID: PMC7712151 DOI: 10.3390/jpm10040215] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/30/2022] Open
Abstract
Huntington's disease (HD) is a severe neurodegenerative disorder caused by a CAG triplet expansion in the first exon of the HTT gene. Here we report the introduction of an HD mutation into the genome of healthy human embryonic fibroblasts through CRISPR/Cas9-mediated homologous recombination. We verified the specificity of the created HTT-editing system and confirmed the absence of undesirable genomic modifications at off-target sites. We showed that both mutant and control isogenic induced pluripotent stem cells (iPSCs) derived by reprogramming of the fibroblast clones can be differentiated into striatal medium spiny neurons. We next demonstrated phenotypic abnormalities in the mutant iPSC-derived neural cells, including impaired neural rosette formation and increased sensitivity to growth factor withdrawal. Moreover, using electron microscopic analysis, we detected a series of ultrastructural defects in the mutant neurons, which did not contain huntingtin aggregates, suggesting that these defects appear early in HD development. Thus, our study describes creation of a new isogenic iPSC-based cell system that models HD and recapitulates HD-specific disturbances in the mutant cells, including some ultrastructural features implemented for the first time.
Collapse
|
10
|
Sbai O, Soussi R, Bole A, Khrestchatisky M, Esclapez M, Ferhat L. The actin binding protein α-actinin-2 expression is associated with dendritic spine plasticity and migrating granule cells in the rat dentate gyrus following pilocarpine-induced seizures. Exp Neurol 2020; 335:113512. [PMID: 33098872 DOI: 10.1016/j.expneurol.2020.113512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
α-actinin-2 (α-actn-2) is an F-actin-crosslinking protein, localized in dendritic spines. In vitro studies suggested that it is involved in spinogenesis, morphogenesis, actin organization, cell migration and anchoring of the NR1 subunit of the N-methyl-D-aspartate (NMDA) receptors in dendritic spines. However, little is known regarding its function in vivo. We examined the levels of α-actn-2 expression within the dentate gyrus (DG) during the development of chronic limbic seizures (epileptogenesis) induced by pilocarpine in rats. In this model, plasticity of the DG glutamatergic granule cells including spine loss, spinogenesis, morphogenesis, neo-synaptogenesis, aberrant migration, and alterations of NMDA receptors have been well characterized. We showed that α-actn-2 immunolabeling was reduced in the inner molecular layer at 1-2 weeks post-status epilepticus (SE), when granule cell spinogenesis and morphogenesis occur. This low level persisted at the chronic stage when new functional synapses are established. This decreased of α-actn-2 protein is concomitant with the recovery of drebrin A (DA), another actin-binding protein, at the chronic stage. Indeed, we demonstrated in cultured cells that in contrast to DA, α-actn-2 did not protect F-actin destabilization and DA inhibited α-actn-2 binding to F-actin. Such alteration could affect the anchoring of NR1 in dendritic spines. Furthermore, we showed that the expression of α-actn-2 and NR1 are co-down-regulated in membrane fractions of pilocarpine animals at chronic stage. Last, we showed that α-actn-2 is expressed in migrating newly born granule cells observed within the hilus of pilocarpine-treated rats. Altogether, our results suggest that α-actn-2 is not critical for the structural integrity and stabilization of granule cell dendritic spines. Instead, its expression is regulated when spinogenesis and morphogenesis occur and within migrating granule cells. Our data also suggest that the balance between α-actn-2 and DA expression levels may modulate NR1 anchoring within dendritic spines.
Collapse
Affiliation(s)
- Oualid Sbai
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Rabia Soussi
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Angélique Bole
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | | | - Monique Esclapez
- Aix-Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Lotfi Ferhat
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.
| |
Collapse
|
11
|
Traceable stimulus-dependent rapid molecular changes in dendritic spines in the brain. Sci Rep 2020; 10:15266. [PMID: 32943708 PMCID: PMC7499203 DOI: 10.1038/s41598-020-72248-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 08/26/2020] [Indexed: 01/21/2023] Open
Abstract
Dendritic spines function as microcompartments that can modify the efficiency of their associated synapses. Here, we analyzed stimulus-dependent molecular changes in spines. The F-actin capping protein CapZ accumulates in parts of dendritic spines within regions where long-term potentiation has been induced. We produced a transgenic mouse line, AiCE-Tg, in which CapZ tagged with enhanced green fluorescence protein (EGFP-CapZ) is expressed. Twenty minutes after unilateral visual or somatosensory stimulation in AiCE-Tg mice, relative EGFP-CapZ signal intensification was seen in a subset of dendritic spines selectively in stimulated-side cortices; this right-left difference was abolished by NMDA receptor blockade. Immunolabeling of α-actinin, a PSD-95 binding protein that can recruit AMPA receptors, showed that the α-actinin signals colocalized more frequently in spines with the brightest EGFP-CapZ signals (top 100) than in spines with more typical EGFP-CapZ signal strength (top 1,000). This stimulus-dependent in vivo redistribution of EGFP-CapZ represents a novel molecular event with plasticity-like characteristics, and bright EGFP-CapZ in AiCE-Tg mice make high-CapZ spines traceable in vivo and ex vivo. This mouse line has the potential to be used to reveal sequential molecular events, including synaptic tagging, and to relate multiple types of plasticity in these spines, extending knowledge related to memory mechanisms.
Collapse
|
12
|
Konietzny A, González-Gallego J, Bär J, Perez-Alvarez A, Drakew A, Demmers JAA, Dekkers DHW, Hammer JA, Frotscher M, Oertner TG, Wagner W, Kneussel M, Mikhaylova M. Myosin V regulates synaptopodin clustering and localization in the dendrites of hippocampal neurons. J Cell Sci 2019; 132:jcs.230177. [PMID: 31371487 PMCID: PMC6737913 DOI: 10.1242/jcs.230177] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 07/19/2019] [Indexed: 01/15/2023] Open
Abstract
The spine apparatus (SA) is an endoplasmic reticulum-related organelle that is present in a subset of dendritic spines in cortical and pyramidal neurons, and plays an important role in Ca2+ homeostasis and dendritic spine plasticity. The protein synaptopodin is essential for the formation of the SA and is widely used as a maker for this organelle. However, it is still unclear which factors contribute to its localization at selected synapses, and how it triggers local SA formation. In this study, we characterized development, localization and mobility of synaptopodin clusters in hippocampal primary neurons, as well as the molecular dynamics within these clusters. Interestingly, synaptopodin at the shaft-associated clusters is less dynamic than at spinous clusters. We identify the actin-based motor proteins myosin V (herein referring to both the myosin Va and Vb forms) and VI as novel interaction partners of synaptopodin, and demonstrate that myosin V is important for the formation and/or maintenance of the SA. We found no evidence of active microtubule-based transport of synaptopodin. Instead, new clusters emerge inside spines, which we interpret as the SA being assembled on-site.
Collapse
Affiliation(s)
- Anja Konietzny
- DFG Emmy Noether Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Judit González-Gallego
- DFG Emmy Noether Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Julia Bär
- DFG Emmy Noether Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Alberto Perez-Alvarez
- Institute for Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Alexander Drakew
- Institute of Structural Neurobiology, Center for Molecular Neurobiology Hamburg, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Institute of Clinical Neuroanatomy, Faculty of Medicine, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | | - Dick H W Dekkers
- Center for Proteomics, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - John A Hammer
- Cell Biology and Physiology Center, National Heart, Lung Blood Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Michael Frotscher
- Institute of Structural Neurobiology, Center for Molecular Neurobiology Hamburg, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Wolfgang Wagner
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marina Mikhaylova
- DFG Emmy Noether Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
13
|
Tousley A, Iuliano M, Weisman E, Sapp E, Richardson H, Vodicka P, Alexander J, Aronin N, DiFiglia M, Kegel-Gleason KB. Huntingtin associates with the actin cytoskeleton and α-actinin isoforms to influence stimulus dependent morphology changes. PLoS One 2019; 14:e0212337. [PMID: 30768638 PMCID: PMC6377189 DOI: 10.1371/journal.pone.0212337] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
One response of cells to growth factor stimulus involves changes in morphology driven by the actin cytoskeleton and actin associated proteins which regulate functions such as cell adhesion, motility and in neurons, synaptic plasticity. Previous studies suggest that Huntingtin may be involved in regulating morphology however, there has been limited evidence linking endogenous Huntingtin localization or function with cytoplasmic actin in cells. We found that depletion of Huntingtin in human fibroblasts reduced adhesion and altered morphology and these phenotypes were made worse with growth factor stimulation, whereas the presence of the Huntington's Disease mutation inhibited growth factor induced changes in morphology and increased numbers of vinculin-positive focal adhesions. Huntingtin immunoreactivity localized to actin stress fibers, vinculin-positive adhesion contacts and membrane ruffles in fibroblasts. Interactome data from others has shown that Huntingtin can associate with α-actinin isoforms which bind actin filaments. Mapping studies using a cDNA encoding α-actinin-2 showed that it interacts within Huntingtin aa 399-969. Double-label immunofluorescence showed Huntingtin and α-actinin-1 co-localized to stress fibers, membrane ruffles and lamellar protrusions in fibroblasts. Proximity ligation assays confirmed a close molecular interaction between Huntingtin and α-actinin-1 in human fibroblasts and neurons. Huntingtin silencing with siRNA in fibroblasts blocked the recruitment of α-actinin-1 to membrane foci. These studies support the idea that Huntingtin is involved in regulating adhesion and actin dependent functions including those involving α-actinin.
Collapse
Affiliation(s)
- Adelaide Tousley
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Maria Iuliano
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Elizabeth Weisman
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Ellen Sapp
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Heather Richardson
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Petr Vodicka
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Jonathan Alexander
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Neil Aronin
- Department of Medicine and Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Marian DiFiglia
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Kimberly B. Kegel-Gleason
- Laboratory of Cellular Neurobiology, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
14
|
Matt L, Kim K, Hergarden AC, Patriarchi T, Malik ZA, Park DK, Chowdhury D, Buonarati OR, Henderson PB, Gökçek Saraç Ç, Zhang Y, Mohapatra D, Horne MC, Ames JB, Hell JW. α-Actinin Anchors PSD-95 at Postsynaptic Sites. Neuron 2018; 97:1094-1109.e9. [PMID: 29429936 PMCID: PMC5963734 DOI: 10.1016/j.neuron.2018.01.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/10/2017] [Accepted: 01/17/2018] [Indexed: 12/17/2022]
Abstract
Despite the central role PSD-95 plays in anchoring postsynaptic AMPARs, how PSD-95 itself is tethered to postsynaptic sites is not well understood. Here we show that the F-actin binding protein α-actinin binds to the very N terminus of PSD-95. Knockdown (KD) of α-actinin phenocopies KD of PSD-95. Mutating lysine at position 10 or lysine at position 11 of PSD-95 to glutamate, or glutamate at position 53 or glutamate and aspartate at positions 213 and 217 of α-actinin, respectively, to lysine impairs, in parallel, PSD-95 binding to α-actinin and postsynaptic localization of PSD-95 and AMPARs. These experiments identify α-actinin as a critical PSD-95 anchor tethering the AMPAR-PSD-95 complex to postsynaptic sites.
Collapse
Affiliation(s)
- Lucas Matt
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Karam Kim
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Anne C Hergarden
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Zulfiqar A Malik
- Department of Pharmacology, University of California, Davis, Davis, CA, USA; Department of Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Deborah K Park
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | | | - Olivia R Buonarati
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Peter B Henderson
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Çiğdem Gökçek Saraç
- Department of Pharmacology, University of California, Davis, Davis, CA, USA; Department of Biomedical Engineering, Faculty of Engineering, Akdeniz University, Antalya, Turkey
| | - Yonghong Zhang
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Durga Mohapatra
- Department of Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Mary C Horne
- Department of Pharmacology, University of California, Davis, Davis, CA, USA; Department of Pharmacology, University of Iowa, Iowa City, IA, USA
| | - James B Ames
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, Davis, CA, USA; Department of Pharmacology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
15
|
Chazeau A, Giannone G. Organization and dynamics of the actin cytoskeleton during dendritic spine morphological remodeling. Cell Mol Life Sci 2016; 73:3053-73. [PMID: 27105623 PMCID: PMC11108290 DOI: 10.1007/s00018-016-2214-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 12/18/2022]
Abstract
In the central nervous system, most excitatory post-synapses are small subcellular structures called dendritic spines. Their structure and morphological remodeling are tightly coupled to changes in synaptic transmission. The F-actin cytoskeleton is the main driving force of dendritic spine remodeling and sustains synaptic plasticity. It is therefore essential to understand how changes in synaptic transmission can regulate the organization and dynamics of actin binding proteins (ABPs). In this review, we will provide a detailed description of the organization and dynamics of F-actin and ABPs in dendritic spines and will discuss the current models explaining how the actin cytoskeleton sustains both structural and functional synaptic plasticity.
Collapse
Affiliation(s)
- Anaël Chazeau
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, UMR 5297, 33000, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, 33000, Bordeaux, France
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Grégory Giannone
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, UMR 5297, 33000, Bordeaux, France.
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, 33000, Bordeaux, France.
| |
Collapse
|
16
|
Swartzwelder HS, Risher ML, Miller KM, Colbran RJ, Winder DG, Wills TA. Changes in the Adult GluN2B Associated Proteome following Adolescent Intermittent Ethanol Exposure. PLoS One 2016; 11:e0155951. [PMID: 27213757 PMCID: PMC4877005 DOI: 10.1371/journal.pone.0155951] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/07/2016] [Indexed: 12/20/2022] Open
Abstract
Adolescent alcohol use is the strongest predictor for alcohol use disorders. In rodents, adolescents have distinct responses to acute ethanol, and prolonged alcohol exposure during adolescence can maintain these phenotypes into adulthood. One brain region that is particularly sensitive to the effects of both acute and chronic ethanol exposure is the hippocampus. Adolescent intermittent ethanol exposure (AIE) produces long lasting changes in hippocampal synaptic plasticity and dendritic morphology, as well as in the susceptibility to acute ethanol-induced spatial memory impairment. Given the pattern of changes in hippocampal structure and function, one potential target for these effects is the ethanol sensitive GluN2B subunit of the NMDA receptor, which is known to be involved in synaptic plasticity and dendritic morphology. Thus we sought to determine if there were persistent changes in hippocampal GluN2B signaling cascades following AIE. We employed a previously validated GluN2B-targeted proteomic strategy that was used to identify novel signaling mechanisms altered by chronic ethanol exposure in the adult hippocampus. We collected adult hippocampal tissue (P70) from rats that had been given 2 weeks of AIE from P30-45. Tissue extracts were fractionated into synaptic and non-synaptic pools, immuno-precipitated for GluN2B, and then analyzed using proteomic methods. We detected a large number of proteins associated with GluN2B. AIE produced significant changes in the association of many proteins with GluN2B in both synaptic and non-synaptic fractions. Intriguingly the number of proteins changed in the non-synaptic fraction was double that found in the synaptic fraction. Some of these proteins include those involved in glutamate signaling cytoskeleton rearrangement, calcium signaling, and plasticity. Disruptions in these pathways may contribute to the persistent cellular and behavioral changes found in the adult hippocampus following AIE. Further, the robust change in non-synaptic proteins suggests that AIE may prime this signaling pathway for future ethanol exposures in adulthood.
Collapse
Affiliation(s)
- H. Scott Swartzwelder
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Psychology and Neuroscience, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Mary-Louise Risher
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Kelsey M. Miller
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Roger J. Colbran
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States of America
- J. F. Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, United States of America
| | - Danny G. Winder
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States of America
- J. F. Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, United States of America
| | - Tiffany A. Wills
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, United States of America
- Department of Cell Biology and Anatomy, Louisiana State University Health Science Center, New Orleans, LA, United States of America
- * E-mail:
| |
Collapse
|
17
|
Sun X, Shi N, Li Y, Dong C, Zhang M, Guan Z, Duan M. Quantitative Proteome Profiling of Street Rabies Virus-Infected Mouse Hippocampal Synaptosomes. Curr Microbiol 2016; 73:301-311. [PMID: 27155843 DOI: 10.1007/s00284-016-1061-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/06/2016] [Indexed: 12/23/2022]
Abstract
It is well established now that neuronal dysfunction rather than structural damage may be responsible for the development of rabies. In order to explore the underlying mechanisms in rabies virus (RABV) and synaptic dysfunctions, a quantitative proteome profiling was carried out on synaptosome samples from mice hippocampus. Synaptosome samples from mice hippocampus were isolated and confirmed by Western blot and transmission electron microscopy. Synaptosome protein content changes were quantitatively detected by Nano-LC-MS/MS. Protein functions were classified by the Gene Ontology (GO) and KEGG pathway. PSICQUIC was used to create a network. MCODE algorithm was applied to obtain subnetworks. Of these protein changes, 45 were upregulated and 14 were downregulated following RABV infection relative to non-infected (mock) synaptosomes. 28 proteins were unique to mock treatment and 12 were unique to RABV treatment. Proteins related to metabolism and synaptic vesicle showed the most changes in expression levels. Furthermore, protein-protein interaction (PPI) networks revealed that several key biological processes related to synaptic functions potentially were modulated by RABV, including energy metabolism, cytoskeleton organization, and synaptic transmission. These data will be useful for better understanding of neuronal dysfunction of rabies and provide the foundation for future research.
Collapse
Affiliation(s)
- Xiaoning Sun
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, Jilin, China
| | - Ning Shi
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences CAAS, Changchun, 132109, China
| | - Ying Li
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Chunyan Dong
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, Jilin, China
| | - Maolin Zhang
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, Jilin, China
| | - Zhenhong Guan
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, Jilin, China
| | - Ming Duan
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, 130062, Jilin, China.
| |
Collapse
|
18
|
Regulation of the Postsynaptic Compartment of Excitatory Synapses by the Actin Cytoskeleton in Health and Its Disruption in Disease. Neural Plast 2016; 2016:2371970. [PMID: 27127658 PMCID: PMC4835652 DOI: 10.1155/2016/2371970] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 03/09/2016] [Indexed: 02/07/2023] Open
Abstract
Disruption of synaptic function at excitatory synapses is one of the earliest pathological changes seen in wide range of neurological diseases. The proper control of the segregation of neurotransmitter receptors at these synapses is directly correlated with the intact regulation of the postsynaptic cytoskeleton. In this review, we are discussing key factors that regulate the structure and dynamics of the actin cytoskeleton, the major cytoskeletal building block that supports the postsynaptic compartment. Special attention is given to the complex interplay of actin-associated proteins that are found in the synaptic specialization. We then discuss our current understanding of how disruption of these cytoskeletal elements may contribute to the pathological events observed in the nervous system under disease conditions with a particular focus on Alzheimer's disease pathology.
Collapse
|
19
|
Neurogranin regulates CaM dynamics at dendritic spines. Sci Rep 2015; 5:11135. [PMID: 26084473 PMCID: PMC4471661 DOI: 10.1038/srep11135] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 05/18/2015] [Indexed: 11/18/2022] Open
Abstract
Calmodulin (CaM) plays a key role in synaptic function and plasticity due to its ability to mediate Ca2+ signaling. Therefore, it is essential to understand the dynamics of CaM at dendritic spines. In this study we have explored CaM dynamics using live-cell confocal microscopy and fluorescence recovery after photobleaching (FRAP) to study CaM diffusion. We find that only a small fraction of CaM in dendritic spines is immobile. Furthermore, the diffusion rate of CaM was regulated by neurogranin (Ng), a CaM-binding protein enriched at dendritic spines. Interestingly, Ng did not influence the immobile fraction of CaM at recovery plateau. We have previously shown that Ng enhances synaptic strength in a CaM-dependent manner. Taken together, these data indicate that Ng-mediated enhancement of synaptic strength is due to its ability to target, rather than sequester, CaM within dendritic spines.
Collapse
|
20
|
Kalinowska M, Chávez AE, Lutzu S, Castillo PE, Bukauskas FF, Francesconi A. Actinin-4 Governs Dendritic Spine Dynamics and Promotes Their Remodeling by Metabotropic Glutamate Receptors. J Biol Chem 2015; 290:15909-20. [PMID: 25944910 DOI: 10.1074/jbc.m115.640136] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
Dendritic spines are dynamic, actin-rich protrusions in neurons that undergo remodeling during neuronal development and activity-dependent plasticity within the central nervous system. Although group 1 metabotropic glutamate receptors (mGluRs) are critical for spine remodeling under physiopathological conditions, the molecular components linking receptor activity to structural plasticity remain unknown. Here we identify a Ca(2+)-sensitive actin-binding protein, α-actinin-4, as a novel group 1 mGluR-interacting partner that orchestrates spine dynamics and morphogenesis in primary neurons. Functional silencing of α-actinin-4 abolished spine elongation and turnover stimulated by group 1 mGluRs despite intact surface receptor expression and downstream ERK1/2 signaling. This function of α-actinin-4 in spine dynamics was underscored by gain-of-function phenotypes in untreated neurons. Here α-actinin-4 induced spine head enlargement, a morphological change requiring the C-terminal domain of α-actinin-4 that binds to CaMKII, an interaction we showed to be regulated by group 1 mGluR activation. Our data provide mechanistic insights into spine remodeling by metabotropic signaling and identify α-actinin-4 as a critical effector of structural plasticity within neurons.
Collapse
Affiliation(s)
- Magdalena Kalinowska
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Andrés E Chávez
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Stefano Lutzu
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Pablo E Castillo
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Feliksas F Bukauskas
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Anna Francesconi
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
21
|
Ning L, Paetau S, Nyman-Huttunen H, Tian L, Gahmberg CG. ICAM-5 affects spine maturation by regulation of NMDA receptor binding to α-actinin. Biol Open 2015; 4:125-36. [PMID: 25572420 PMCID: PMC4365481 DOI: 10.1242/bio.201410439] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
ICAM-5 is a negative regulator of dendritic spine maturation and facilitates the formation of filopodia. Its absence results in improved memory functions, but the mechanisms have remained poorly understood. Activation of NMDA receptors induces ICAM-5 ectodomain cleavage through a matrix metalloproteinase (MMP)-dependent pathway, which promotes spine maturation and synapse formation. Here, we report a novel, ICAM-5-dependent mechanism underlying spine maturation by regulating the dynamics and synaptic distribution of α-actinin. We found that GluN1 and ICAM-5 partially compete for the binding to α-actinin; deletion of the cytoplasmic tail of ICAM-5 or ablation of the gene resulted in increased association of GluN1 with α-actinin, whereas internalization of ICAM-5 peptide perturbed the GluN1/α-actinin interaction. NMDA treatment decreased α-actinin binding to ICAM-5, and increased the binding to GluN1. Proper synaptic distribution of α-actinin requires the ICAM-5 cytoplasmic domain, without which α-actinin tended to accumulate in filopodia, leading to F-actin reorganization. The results indicate that ICAM-5 retards spine maturation by preventing reorganization of the actin cytoskeleton, but NMDA receptor activation is sufficient to relieve the brake and promote the maturation of spines.
Collapse
Affiliation(s)
- Lin Ning
- Division of Biochemistry and Biotechnology, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 5, FIN-00014, Helsinki, Finland
| | - Sonja Paetau
- Division of Biochemistry and Biotechnology, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 5, FIN-00014, Helsinki, Finland
| | - Henrietta Nyman-Huttunen
- Division of Biochemistry and Biotechnology, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 5, FIN-00014, Helsinki, Finland
| | - Li Tian
- Neuroscience Center, University of Helsinki, Viikinkaari 4, FIN-00014, Helsinki, Finland
| | - Carl G Gahmberg
- Division of Biochemistry and Biotechnology, Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 5, FIN-00014, Helsinki, Finland
| |
Collapse
|
22
|
Ammar MR, Kassas N, Bader MF, Vitale N. Phosphatidic acid in neuronal development: A node for membrane and cytoskeleton rearrangements. Biochimie 2014; 107 Pt A:51-7. [DOI: 10.1016/j.biochi.2014.07.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/30/2014] [Indexed: 12/22/2022]
|
23
|
Valencia A, Sapp E, Kimm JS, McClory H, Ansong KA, Yohrling G, Kwak S, Kegel KB, Green KM, Shaffer SA, Aronin N, DiFiglia M. Striatal synaptosomes from Hdh140Q/140Q knock-in mice have altered protein levels, novel sites of methionine oxidation, and excess glutamate release after stimulation. J Huntingtons Dis 2014; 2:459-75. [PMID: 24696705 DOI: 10.3233/jhd-130080] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Synaptic connections are disrupted in patients with Huntington's disease (HD). Synaptosomes from postmortem brain are ideal for synaptic function studies because they are enriched in pre- and post-synaptic proteins important in vesicle fusion, vesicle release, and neurotransmitter receptor activation. OBJECTIVE To examine striatal synaptosomes from 3, 6 and 12 month old WT and Hdh140Q/140Q knock-in mice for levels of synaptic proteins, methionine oxidation, and glutamate release. METHODS We used Western blot analysis, glutamate release assays, and liquid chromatography tandem mass spectrometry (LC-MS/MS). RESULTS Striatal synaptosomes of 6 month old Hdh140Q/140Q mice had less DARPP32, syntaxin 1 and calmodulin compared to WT. Striatal synaptosomes of 12 month old Hdh140Q/140Q mice had lower levels of DARPP32, alpha actinin, HAP40, Na+/K+-ATPase, PSD95, SNAP-25, TrkA and VAMP1, VGlut1 and VGlut2, increased levels of VAMP2, and modifications in actin and calmodulin compared to WT. More glutamate released from vesicles of depolarized striatal synaptosomes of 6 month old Hdh140Q/140Q than from age matched WT mice but there was no difference in glutamate release in synaptosomes of 3 and 12 month old WT and Hdh140Q/140Q mice. LC-MS/MS of 6 month old Hdh140Q/140Q mice striatal synaptosomes revealed that about 4% of total proteins detected (>600 detected) had novel sites of methionine oxidation including proteins involved with vesicle fusion, trafficking, and neurotransmitter function (synaptophysin, synapsin 2, syntaxin 1, calmodulin, cytoplasmic actin 2, neurofilament, and tubulin). Altered protein levels and novel methionine oxidations were also seen in cortical synaptosomes of 12 month old Hdh140Q/140Q mice. CONCLUSIONS Findings provide support for early synaptic dysfunction in Hdh140Q/140Q knock-in mice arising from altered protein levels, oxidative damage, and impaired glutamate neurotransmission and suggest that study of synaptosomes could be of value for evaluating HD therapies.
Collapse
|
24
|
McVicker DP, Millette MM, Dent EW. Signaling to the microtubule cytoskeleton: an unconventional role for CaMKII. Dev Neurobiol 2014; 75:423-34. [PMID: 25156276 DOI: 10.1002/dneu.22227] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/20/2014] [Indexed: 12/29/2022]
Abstract
Synaptic plasticity is a hallmark of the nervous system and is thought to be integral to higher brain functions such as learning and memory. Calcium, acting as a second messenger, and the calcium/calmodulin dependent kinase CaMKII are key regulators of neuronal plasticity. Given the importance of the actin and microtubule (MT) cytoskeleton in dendritic spine morphology, composition and plasticity, it is not surprising that many regulators of these cytoskeletal elements are downstream of the CaMKII pathway. In this review, we discuss the emerging role of calcium and CaMKII in the regulation of MTs and cargo unloading during synaptic plasticity.
Collapse
Affiliation(s)
- Derrick P McVicker
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, 53705
| | | | | |
Collapse
|
25
|
Hodges JL, Vilchez SM, Asmussen H, Whitmore LA, Horwitz AR. α-Actinin-2 mediates spine morphology and assembly of the post-synaptic density in hippocampal neurons. PLoS One 2014; 9:e101770. [PMID: 25007055 PMCID: PMC4090192 DOI: 10.1371/journal.pone.0101770] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/10/2014] [Indexed: 11/18/2022] Open
Abstract
Dendritic spines are micron-sized protrusions that constitute the primary post-synaptic sites of excitatory neurotransmission in the brain. Spines mature from a filopodia-like protrusion into a mushroom-shaped morphology with a post-synaptic density (PSD) at its tip. Modulation of the actin cytoskeleton drives these morphological changes as well as the spine dynamics that underlie learning and memory. Several PSD molecules respond to glutamate receptor activation and relay signals to the underlying actin cytoskeleton to regulate the structural changes in spine and PSD morphology. α-Actinin-2 is an actin filament cross-linker, which localizes to dendritic spines, enriched within the post-synaptic density, and implicated in actin organization. We show that loss of α-actinin-2 in rat hippocampal neurons creates an increased density of immature, filopodia-like protrusions that fail to mature into a mushroom-shaped spine during development. α-Actinin-2 knockdown also prevents the recruitment and stabilization of the PSD in the spine, resulting in failure of synapse formation, and an inability to structurally respond to chemical stimulation of the N-methyl-D-aspartate (NMDA)-type glutamate receptor. The Ca2+-insensitive EF-hand motif in α-actinin-2 is necessary for the molecule's function in regulating spine morphology and PSD assembly, since exchanging it for the similar but Ca2+-sensitive domain from α-actinin-4, another α-actinin isoform, inhibits its function. Furthermore, when the Ca2+-insensitive domain from α-actinin-2 is inserted into α-actinin-4 and expressed in neurons, it creates mature spines. These observations support a model whereby α-actinin-2, partially through its Ca2+-insensitive EF-hand motif, nucleates PSD formation via F-actin organization and modulates spine maturation to mediate synaptogenesis.
Collapse
Affiliation(s)
- Jennifer L. Hodges
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail: (ARH); (JLH)
| | - Samuel Martin Vilchez
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Hannelore Asmussen
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Leanna A. Whitmore
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Alan Rick Horwitz
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail: (ARH); (JLH)
| |
Collapse
|
26
|
Abstract
α-Actinins are a major class of actin filament cross-linking proteins expressed in virtually all cells. In muscle, actinins cross-link thin filaments from adjacent sarcomeres. In non-muscle cells, different actinin isoforms play analogous roles in cross-linking actin filaments and anchoring them to structures such as cell-cell and cell-matrix junctions. Although actinins have long been known to play roles in cytokinesis, cell adhesion and cell migration, recent studies have provided further mechanistic insights into these functions. Roles for actinins in synaptic plasticity and membrane trafficking events have emerged more recently, as has a 'non-canonical' function for actinins in transcriptional regulation in the nucleus. In the present paper we review recent advances in our understanding of these diverse cell biological functions of actinins in non-muscle cells, as well as their roles in cancer and in genetic disorders affecting platelet and kidney physiology. We also make two proposals with regard to the actinin nomenclature. First, we argue that naming actinin isoforms according to their expression patterns is problematic and we suggest a more precise nomenclature system. Secondly, we suggest that the α in α-actinin is superfluous and can be omitted.
Collapse
|
27
|
Hell JW. CaMKII: claiming center stage in postsynaptic function and organization. Neuron 2014; 81:249-65. [PMID: 24462093 DOI: 10.1016/j.neuron.2013.12.024] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2013] [Indexed: 11/16/2022]
Abstract
While CaMKII has long been known to be essential for synaptic plasticity and learning, recent work points to new dimensions of CaMKII function in the nervous system, revealing that CaMKII also plays an important role in synaptic organization. Ca(2+)-triggered autophosphorylation of CaMKII not only provides molecular memory by prolonging CaMKII activity during long-term plasticity (LTP) and learning but also represents a mechanism for autoactivation of CaMKII's multifaceted protein-docking functions. New details are also emerging about the distinct roles of CaMKIIα and CaMKIIβ in synaptic homeostasis, further illustrating the multilayered and complex nature of CaMKII's involvement in synaptic regulation. Here, I review novel molecular and functional insight into how CaMKII supports synaptic function.
Collapse
Affiliation(s)
- Johannes W Hell
- Department of Pharmacology, University of California, Davis, Davis, CA 95615, USA.
| |
Collapse
|
28
|
Abstract
The development of methods to follow the dynamics of synaptic molecules in living neurons has radically altered our view of the synapse, from that of a generally static structure to that of a dynamic molecular assembly at steady state. This view holds not only for relatively labile synaptic components, such as synaptic vesicles, cytoskeletal elements, and neurotransmitter receptors, but also for the numerous synaptic molecules known as scaffolding molecules, a generic name for a diverse class of molecules that organize synaptic function in time and space. Recent studies reveal that these molecules, which confer a degree of stability to synaptic assemblies over time scales of hours and days, are themselves subject to significant dynamics. Furthermore, these dynamics are probably not without effect; wherever studied, these seem to be associated with spontaneous changes in scaffold molecule content, synaptic size, and possibly synaptic function. This review describes the dynamics exhibited by synaptic scaffold molecules, their typical time scales, and the potential implications to our understanding of synaptic function.
Collapse
Affiliation(s)
- Noam E. Ziv
- Technion–Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
29
|
Ma XM, Miller MB, Vishwanatha KS, Gross MJ, Wang Y, Abbott T, Lam TT, Mains RE, Eipper BA. Nonenzymatic domains of Kalirin7 contribute to spine morphogenesis through interactions with phosphoinositides and Abl. Mol Biol Cell 2014; 25:1458-71. [PMID: 24600045 PMCID: PMC4004595 DOI: 10.1091/mbc.e13-04-0215] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Like several Rho GDP/GTP exchange factors (GEFs), Kalirin7 (Kal7) contains an N-terminal Sec14 domain and multiple spectrin repeats. A natural splice variant of Kalrn lacking the Sec14 domain and four spectrin repeats is unable to increase spine formation; our goal was to understand the function of the Sec14 and spectrin repeat domains. Kal7 lacking its Sec14 domain still increased spine formation, but the spines were short. Strikingly, Kal7 truncation mutants containing only the Sec14 domain and several spectrin repeats increased spine formation. The Sec14 domain bound phosphoinositides, a minor but crucial component of cellular membranes, and binding was increased by a phosphomimetic mutation. Expression of KalSec14-GFP in nonneuronal cells impaired receptor-mediated endocytosis, linking Kal7 to membrane trafficking. Consistent with genetic studies placing Abl, a non-receptor tyrosine kinase, and the Drosophila orthologue of Kalrn into the same signaling pathway, Abl1 phosphorylated two sites in the fourth spectrin repeat of Kalirin, increasing its sensitivity to calpain-mediated degradation. Treating cortical neurons of the wild-type mouse, but not the Kal7(KO) mouse, with an Abl inhibitor caused an increase in linear spine density. Phosphorylation of multiple sites in the N-terminal Sec14/spectrin region of Kal7 may allow coordination of the many signaling pathways contributing to spine morphogenesis.
Collapse
Affiliation(s)
- Xin-Ming Ma
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030 WM Keck Foundation Biotechnology Resource Laboratory, Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hazai D, Szudoczki R, Ding J, Soderling SH, Weinberg RJ, Sótonyi P, Rácz B. Ultrastructural abnormalities in CA1 hippocampus caused by deletion of the actin regulator WAVE-1. PLoS One 2013; 8:e75248. [PMID: 24086480 PMCID: PMC3783472 DOI: 10.1371/journal.pone.0075248] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 08/12/2013] [Indexed: 11/18/2022] Open
Abstract
By conveying signals from the small GTPase family of proteins to the Arp2/3 complex, proteins of the WAVE family facilitate actin remodeling. The WAVE-1 isoform is expressed at high levels in brain, where it plays a role in normal synaptic processing, and is implicated in hippocampus-dependent memory retention. We used electron microscopy to determine whether synaptic structure is modified in the hippocampus of WAVE-1 knockout mice, focusing on the neuropil of CA1 stratum radiatum. Mice lacking WAVE-1 exhibited alterations in the morphology of both axon terminals and dendritic spines; the relationship between the synaptic partners was also modified. The abnormal synaptic morphology we observed suggests that signaling through WAVE-1 plays a critical role in establishing normal synaptic architecture in the rodent hippocampus.
Collapse
Affiliation(s)
- Diána Hazai
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, Budapest, Hungary
| | - Róbert Szudoczki
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, Budapest, Hungary
| | - Jindong Ding
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, North Carolina, United States of America
| | - Scott H. Soderling
- Departments of Cell Biology and Neurobiology, Duke University, Durham, North Carolina, United States of America
| | - Richard J. Weinberg
- Department of Cell Biology & Physiology and Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Péter Sótonyi
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, Budapest, Hungary
| | - Bence Rácz
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
31
|
Abstract
Like all cells, neurons are made of proteins that have characteristic synthesis and degradation profiles. Unlike other cells, however, neurons have a unique multipolar architecture that makes ∼10,000 synaptic contacts with other neurons. Both the stability and modifiability of the neuronal proteome are crucial for its information-processing, storage and plastic properties. The cell biological mechanisms that synthesize, modify, deliver and degrade dendritic and synaptic proteins are not well understood but appear to reflect unique solutions adapted to the particular morphology of neurons.
Collapse
|
32
|
Zhang F, Chen L, Liu C, Qiu P, Wang A, Li L, Wang H. Up-regulation of protein tyrosine nitration in methamphetamine-induced neurotoxicity through DDAH/ADMA/NOS pathway. Neurochem Int 2013; 62:1055-64. [PMID: 23583342 DOI: 10.1016/j.neuint.2013.03.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 03/24/2013] [Accepted: 03/29/2013] [Indexed: 12/22/2022]
Abstract
Protein tyrosine nitration is an important post-translational modification mediated by nitric oxide (NO) associated oxidative stress, occurring in a variety of neurodegenerative diseases. In our previous study, an elevated level of dimethylarginine dimethylaminohydrolase 1 (DDAH1) protein was observed in different brain regions of acute methamphetamine (METH) treated rats, indicating the possibility of an enhanced expression of protein nitration that is mediated by excess NO through the DDAH1/ADMA (Asymmetric Dimethylated l-arginine)/NOS (Nitric Oxide Synthase) pathway. In the present study, proteomic methods, including stable isotope labeling with amino acids in cell culture (SILAC) and two dimensional electrophoresis, were used to determine the relationship between protein nitration and METH induced neurotoxicity in acute METH treated rats and PC12 cells. We found that acute METH administration evokes a positive activation of DDAH1/ADMA/NOS pathway and results in an over-production of NO in different brain regions of rat and PC12 cells, whereas the whole signaling could be repressed by DDAH1 inhibitor N(ω)-(2-methoxyethyl)-arginine (l-257). In addition, enhanced expressions of 3 nitroproteins were identified in rat striatum and increased levels of 27 nitroproteins were observed in PC12 cells. These nitrated proteins are key factors for Cdk5 activation, cytoskeletal structure, ribosomes function, etc. l-257 also displayed significant protective effects against METH-induced protein nitration, apoptosis and cell death. The overall results illustrate that protein nitration plays a significant role in the acute METH induced neurotoxicity via the activation of DDAH1/ADMA/NOS pathway.
Collapse
Affiliation(s)
- Fu Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | | | | | | | | | | | | |
Collapse
|
33
|
Rácz B, Weinberg RJ. Microdomains in forebrain spines: an ultrastructural perspective. Mol Neurobiol 2013; 47:77-89. [PMID: 22983912 PMCID: PMC3538892 DOI: 10.1007/s12035-012-8345-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 08/27/2012] [Indexed: 12/21/2022]
Abstract
Glutamatergic axons in the mammalian forebrain terminate predominantly onto dendritic spines. Long-term changes in the efficacy of these excitatory synapses are tightly coupled to changes in spine morphology. The reorganization of the actin cytoskeleton underlying this spine "morphing" involves numerous proteins that provide the machinery needed for adaptive cytoskeletal remodeling. Here, we review recent literature addressing the chemical architecture of the spine, focusing mainly on actin-binding proteins (ABPs). Accumulating evidence suggests that ABPs are organized into functionally distinct microdomains within the spine cytoplasm. This functional compartmentalization provides a structural basis for regulation of the spinoskeleton, offering a novel window into mechanisms underlying synaptic plasticity.
Collapse
Affiliation(s)
- Bence Rácz
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, 1078, Budapest, Hungary.
| | | |
Collapse
|
34
|
Mandela P, Yankova M, Conti LH, Ma XM, Grady J, Eipper BA, Mains RE. Kalrn plays key roles within and outside of the nervous system. BMC Neurosci 2012; 13:136. [PMID: 23116210 PMCID: PMC3541206 DOI: 10.1186/1471-2202-13-136] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 10/22/2012] [Indexed: 11/23/2022] Open
Abstract
Background The human KALRN gene, which encodes a complex, multifunctional Rho GDP/GTP exchange factor, has been linked to cardiovascular disease, psychiatric disorders and neurodegeneration. Examination of existing Kalrn knockout mouse models has focused only on neuronal phenotypes. However, Kalirin was first identified through its interaction with an enzyme involved in the synthesis and secretion of multiple bioactive peptides, and studies in C.elegans revealed roles for its orthologue in neurosecretion. Results We used a broad array of tests to evaluate the effects of ablating a single exon in the spectrin repeat region of Kalrn (KalSRKO/KO); transcripts encoding Kalrn isoforms containing only the second GEF domain can still be produced from the single remaining functional Kalrn promoter. As expected, KalSRKO/KO mice showed a decrease in anxiety-like behavior and a passive avoidance deficit. No changes were observed in prepulse inhibition of acoustic startle or tests of depression-like behavior. Growth rate, parturition and pituitary secretion of growth hormone and prolactin were deficient in the KalSRKO/KO mice. Based on the fact that a subset of Kalrn isoforms is expressed in mouse skeletal muscle and the observation that muscle function in C.elegans requires its Kalrn orthologue, KalSRKO/KO mice were evaluated in the rotarod and wire hang tests. KalSRKO/KO mice showed a profound decrease in neuromuscular function, with deficits apparent in KalSR+/KO mice; these deficits were not as marked when loss of Kalrn expression was restricted to the nervous system. Pre- and postsynaptic deficits in the neuromuscular junction were observed, along with alterations in sarcomere length. Conclusions Many of the widespread and diverse deficits observed both within and outside of the nervous system when expression of Kalrn is eliminated may reflect its role in secretory granule function and its expression outside of the nervous system.
Collapse
Affiliation(s)
- Prashant Mandela
- Department of Neuroscience, University of Connecticut Health Science Center, Farmington, CT 06030-3401, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Frick KM. Building a better hormone therapy? How understanding the rapid effects of sex steroid hormones could lead to new therapeutics for age-related memory decline. Behav Neurosci 2012; 126:29-53. [PMID: 22289043 DOI: 10.1037/a0026660] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A wealth of data collected in recent decades has demonstrated that ovarian sex-steroid hormones, particularly 17β-estradiol (E2), are important trophic factors that regulate the function of cognitive regions of the brain such as the hippocampus. The loss of hormone cycling at menopause is associated with cognitive decline and dementia in women, and the onset of memory decline in animal models. However, hormone therapy is not currently recommended to prevent or treat cognitive decline, in part because of its detrimental side effects. In this article, it is proposed that investigations of the rapid effects of E2 on hippocampal function be used to further the design of new drugs that mimic the beneficial effects of E2 on memory without the side effects of current therapies. A conceptual model is presented for elucidating the molecular and biochemical mechanisms through which sex-steroid hormones modulate memory, and a specific hypothesis is proposed to account for the rapid memory-enhancing effects of E2. Empirical support for this hypothesis is discussed as a means of stimulating the consideration of new directions for the development of hormone-based therapies to preserve memory function in menopausal women.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 East Hartford Avenue, Milwaukee, WI 53211, USA.
| |
Collapse
|
36
|
Brunelli L, Llansola M, Felipo V, Campagna R, Airoldi L, De Paola M, Fanelli R, Mariani A, Mazzoletti M, Pastorelli R. Insight into the neuroproteomics effects of the food-contaminant non-dioxin like polychlorinated biphenyls. J Proteomics 2012; 75:2417-30. [PMID: 22387315 DOI: 10.1016/j.jprot.2012.02.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 02/14/2012] [Accepted: 02/16/2012] [Indexed: 01/20/2023]
Abstract
Recent studies showed that food-contaminant non-dioxin-like polychlorinated biphenyls (NDL-PCBs) congeners (PCB52, PCB138, PCB180) have neurotoxic potential, but the cellular and molecular mechanisms underlying neuronal damage are not entirely known. The aim of this study was to assess whether in-vitro exposure to NDL-PCBs may alter the proteome profile of primary cerebellar neurons in order to expand our knowledge on NDL-PCBs neurotoxicity. Comparison of proteome from unexposed and exposed rat cerebellar neurons was performed using state-of-the-art label-free semi-quantitative mass-spectrometry method. We observed significant changes in the abundance of several proteins, that fall into two main classes: (i) novel targets for both PCB138 and 180, mediating the dysregulation of CREB pathways and ubiquitin-proteasome system; (ii) different congeners-specific targets (alpha-actinin-1 for PCB138; microtubule-associated-protein-2 for PCB180) that might lead to similar deleterious consequences on neurons cytoskeleton organization. Interference of the PCB congeners with synaptic formation was supported by the increased expression of pre- and post-synaptic proteins quantified by western blot and immunocytochemistry. Expression alteration of synaptic markers was confirmed in the cerebellum of rats developmentally exposed to these congeners, suggesting an adaptive response to neurodevelopmental toxicity on brain structures. As such, our work is expected to lead to new insights into the mechanisms of NDL-PCBs neurotoxicity.
Collapse
Affiliation(s)
- Laura Brunelli
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Seabold GK, Wang PY, Petralia RS, Chang K, Zhou A, McDermott MI, Wang YX, Milgram SL, Wenthold RJ. Dileucine and PDZ-binding motifs mediate synaptic adhesion-like molecule 1 (SALM1) trafficking in hippocampal neurons. J Biol Chem 2012; 287:4470-84. [PMID: 22174418 PMCID: PMC3281672 DOI: 10.1074/jbc.m111.279661] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 11/22/2011] [Indexed: 12/18/2022] Open
Abstract
Synaptic adhesion-like molecules (SALMs) are a family of cell adhesion molecules involved in neurite outgrowth and synapse formation. Of the five family members, only SALM1, -2, and -3 contain a cytoplasmic C-terminal PDZ-binding motif. We have found that SALM1 is unique among the SALMs because deletion of its PDZ-binding motif (SALM1ΔPDZ) blocks its surface expression in heterologous cells. When expressed in hippocampal neurons, SALM1ΔPDZ had decreased surface expression in dendrites and the cell soma but not in axons, suggesting that the PDZ-binding domain may influence cellular trafficking of SALMs to specific neuronal locations. Endoglycosidase H digestion assays indicated that SALM1ΔPDZ is retained in the endoplasmic reticulum (ER) in heterologous cells. However, when the entire C-terminal tail of SALM1 was deleted, SALM1 was detected on the cell surface. Using serial deletions, we identified a region of SALM1 that contains a putative dileucine ER retention motif, which is not present in the other SALMs. Mutation of this DXXXLL motif allowed SALM1 to leave the ER and enhanced its surface expression in heterologous cells and neurons. An increase in the number of protrusions at the dendrites and cell body was observed when this SALM1 mutant was expressed in hippocampal neurons. With electron microscopy, these protrusions appeared to be irregular, enlarged spines and filopodia. Thus, enrichment of SALM1 on the cell surface affects dendritic arborization, and intracellular motifs regulate its dendritic versus axonal localization.
Collapse
Affiliation(s)
- Gail K Seabold
- Laboratory of Neurochemistry, NIDCD/National Institutes of Health, 50 South Dr., Bldg. 50, Rm. 4144, Bethesda, MD20892-8027, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cheyne JE, Grant L, Butler-Munro C, Foote JW, Connor B, Montgomery JM. Synaptic integration of newly generated neurons in rat dissociated hippocampal cultures. Mol Cell Neurosci 2011; 47:203-14. [DOI: 10.1016/j.mcn.2011.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 04/20/2011] [Accepted: 04/26/2011] [Indexed: 10/18/2022] Open
|
39
|
Nestor MW, Cai X, Stone MR, Bloch RJ, Thompson SM. The actin binding domain of βI-spectrin regulates the morphological and functional dynamics of dendritic spines. PLoS One 2011; 6:e16197. [PMID: 21297961 PMCID: PMC3031527 DOI: 10.1371/journal.pone.0016197] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 12/07/2010] [Indexed: 01/30/2023] Open
Abstract
Actin microfilaments regulate the size, shape and mobility of dendritic spines and are in turn regulated by actin binding proteins and small GTPases. The βI isoform of spectrin, a protein that links the actin cytoskeleton to membrane proteins, is present in spines. To understand its function, we expressed its actin-binding domain (ABD) in CA1 pyramidal neurons in hippocampal slice cultures. The ABD of βI-spectrin bundled actin in principal dendrites and was concentrated in dendritic spines, where it significantly increased the size of the spine head. These effects were not observed after expression of homologous ABDs of utrophin, dystrophin, and α-actinin. Treatment of slice cultures with latrunculin-B significantly decreased spine head size and decreased actin-GFP fluorescence in cells expressing the ABD of α-actinin, but not the ABD of βI-spectrin, suggesting that its presence inhibits actin depolymerization. We also observed an increase in the area of GFP-tagged PSD-95 in the spine head and an increase in the amplitude of mEPSCs at spines expressing the ABD of βI-spectrin. The effects of the βI-spectrin ABD on spine size and mEPSC amplitude were mimicked by expressing wild-type Rac3, a small GTPase that co-immunoprecipitates specifically with βI-spectrin in extracts of cultured cortical neurons. Spine size was normal in cells co-expressing a dominant negative Rac3 construct with the βI-spectrin ABD. We suggest that βI-spectrin is a synaptic protein that can modulate both the morphological and functional dynamics of dendritic spines, perhaps via interaction with actin and Rac3.
Collapse
Affiliation(s)
- Michael W. Nestor
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Xiang Cai
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Michele R. Stone
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Robert J. Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Scott M. Thompson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
40
|
Bennett MR, Farnell L, Gibson WG. A Model of NMDA Receptor Control of F-actin Treadmilling in Synaptic Spines and Their Growth. Bull Math Biol 2010; 73:2109-31. [DOI: 10.1007/s11538-010-9614-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 11/26/2010] [Indexed: 12/16/2022]
|
41
|
Lek M, Quinlan KGR, North KN. The evolution of skeletal muscle performance: gene duplication and divergence of human sarcomeric alpha-actinins. Bioessays 2010; 32:17-25. [PMID: 19967710 DOI: 10.1002/bies.200900110] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In humans, there are two skeletal muscle alpha-actinins, encoded by ACTN2 and ACTN3, and the ACTN3 genotype is associated with human athletic performance. Remarkably, approximately 1 billion people worldwide are deficient in alpha-actinin-3 due to the common ACTN3 R577X polymorphism. The alpha-actinins are an ancient family of actin-binding proteins with structural, signalling and metabolic functions. The skeletal muscle alpha-actinins diverged approximately 250-300 million years ago, and ACTN3 has since developed restricted expression in fast muscle fibres. Despite ACTN2 and ACTN3 retaining considerable sequence similarity, it is likely that following duplication there was a divergence in function explaining why alpha-actinin-2 cannot completely compensate for the absence of alpha-actinin-3. This paper focuses on the role of skeletal muscle alpha-actinins, and how possible changes in functions between these duplicates fit in the context of gene duplication paradigms.
Collapse
Affiliation(s)
- Monkol Lek
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | | | | |
Collapse
|
42
|
Baucum AJ, Jalan-Sakrikar N, Jiao Y, Gustin RM, Carmody LC, Tabb DL, Ham AJL, Colbran RJ. Identification and validation of novel spinophilin-associated proteins in rodent striatum using an enhanced ex vivo shotgun proteomics approach. Mol Cell Proteomics 2010; 9:1243-59. [PMID: 20124353 DOI: 10.1074/mcp.m900387-mcp200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Spinophilin regulates excitatory postsynaptic function and morphology during development by virtue of its interactions with filamentous actin, protein phosphatase 1, and a plethora of additional signaling proteins. To provide insight into the roles of spinophilin in mature brain, we characterized the spinophilin interactome in subcellular fractions solubilized from adult rodent striatum by using a shotgun proteomics approach to identify proteins in spinophilin immune complexes. Initial analyses of samples generated using a mouse spinophilin antibody detected 23 proteins that were not present in an IgG control sample; however, 12 of these proteins were detected in complexes isolated from spinophilin knock-out tissue. A second screen using two different spinophilin antibodies and either knock-out or IgG controls identified a total of 125 proteins. The probability of each protein being specifically associated with spinophilin in each sample was calculated, and proteins were ranked according to a chi(2) analysis of the probabilities from analyses of multiple samples. Spinophilin and the known associated proteins neurabin and multiple isoforms of protein phosphatase 1 were specifically detected. Multiple, novel, spinophilin-associated proteins (myosin Va, calcium/calmodulin-dependent protein kinase II, neurofilament light polypeptide, postsynaptic density 95, alpha-actinin, and densin) were then shown to interact with GST fusion proteins containing fragments of spinophilin. Additional biochemical and transfected cell imaging studies showed that alpha-actinin and densin directly interact with residues 151-300 and 446-817, respectively, of spinophilin. Taken together, we have developed a multi-antibody, shotgun proteomics approach to characterize protein interactomes in native tissues, delineating the importance of knock-out tissue controls and providing novel insights into the nature and function of the spinophilin interactome in mature striatum.
Collapse
Affiliation(s)
- Anthony J Baucum
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232-0615, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Pontrello CG, Ethell IM. Accelerators, Brakes, and Gears of Actin Dynamics in Dendritic Spines. ACTA ACUST UNITED AC 2009; 3:67-86. [PMID: 20463852 DOI: 10.2174/1874082000903020067] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic spines are actin-rich structures that accommodate the postsynaptic sites of most excitatory synapses in the brain. Although dendritic spines form and mature as synaptic connections develop, they remain plastic even in the adult brain, where they can rapidly grow, change, or collapse in response to normal physiological changes in synaptic activity that underlie learning and memory. Pathological stimuli can adversely affect dendritic spine shape and number, and this is seen in neurodegenerative disorders and some forms of mental retardation and autism as well. Many of the molecular signals that control these changes in dendritic spines act through the regulation of filamentous actin (F-actin), some through direct interaction with actin, and others via downstream effectors. For example, cortactin, cofilin, and gelsolin are actin-binding proteins that directly regulate actin dynamics in dendritic spines. Activities of these proteins are precisely regulated by intracellular signaling events that control their phosphorylation state and localization. In this review, we discuss how actin-regulating proteins maintain the balance between F-actin assembly and disassembly that is needed to stabilize mature dendritic spines, and how changes in their activities may lead to rapid remodeling of dendritic spines.
Collapse
Affiliation(s)
- Crystal G Pontrello
- Biomedical Sciences Division and Neuroscience program, University of California Riverside, USA
| | | |
Collapse
|
45
|
Abstract
Dendritic spines are the postsynaptic components of most excitatory synapses in the mammalian brain. Spines accumulate rapidly during early postnatal development and undergo a substantial loss as animals mature into adulthood. In past decades, studies have revealed that the number and size of dendritic spines are regulated by a variety of gene products and environmental factors, underscoring the dynamic nature of spines and their importance to brain plasticity. Recently, in vivo time-lapse imaging of dendritic spines in the cerebral cortex suggests that, although spines are highly plastic during development, they are remarkably stable in adulthood, and most of them last throughout life. Therefore, dendritic spines may provide a structural basis for lifelong information storage, in addition to their well-established role in brain plasticity. Because dendritic spines are the key elements for information acquisition and retention, understanding how spines are formed and maintained, particularly in the intact brain, will likely provide fundamental insights into how the brain possesses the extraordinary capacity to learn and to remember.
Collapse
Affiliation(s)
- D Harshad Bhatt
- Molecular Neurobiology Program, The Helen and Martin Kimmel Center for Biology and Medicine at Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | |
Collapse
|
46
|
NMDA receptor GluN2B (GluR epsilon 2/NR2B) subunit is crucial for channel function, postsynaptic macromolecular organization, and actin cytoskeleton at hippocampal CA3 synapses. J Neurosci 2009; 29:10869-82. [PMID: 19726645 DOI: 10.1523/jneurosci.5531-08.2009] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
GluN2B (GluRepsilon2/NR2B) subunit is involved in synapse development, synaptic plasticity, and cognitive function. However, its roles in synaptic expression and function of NMDA receptors (NMDARs) in the brain remain mostly unknown because of the neonatal lethality of global knock-out mice. To address this, we generated conditional knock-out mice, in which GluN2B was ablated exclusively in hippocampal CA3 pyramidal cells. By immunohistochemistry, GluN2B disappeared and GluN1 (GluRzeta1/NR1) was moderately reduced, whereas GluN2A (GluRepsilon1/NR2A) and postsynaptic density-95 (PSD-95) were unaltered in the mutant CA3. This was consistent with protein contents in the CA3 crude fraction: 9.6% of control level for GluN2B, 47.7% for GluN1, 90.6% for GluN2A, and 98.0% for PSD-95. Despite the remaining NMDARs, NMDAR-mediated currents and long-term potentiation were virtually lost at various CA3 synapses. Then, we compared synaptic NMDARs by postembedding immunogold electron microscopy and immunoblot using the PSD fraction. In the mutant CA3, GluN1 was severely reduced in both immunogold (20.6-23.6%) and immunoblot (24.6%), whereas GluN2A and PSD-95 were unchanged in immunogold but markedly reduced in the PSD fraction (51.4 and 36.5%, respectively), indicating increased detergent solubility of PSD molecules. No such increased solubility was observed for GluN2B in the CA3 of GluN2A-knock-out mice. Furthermore, significant decreases were found in the ratio of filamentous to globular actin (49.5%) and in the density of dendritic spines (76.2%). These findings suggest that GluN2B is critically involved in NMDAR channel function, organization of postsynaptic macromolecular complexes, formation or maintenance of dendritic spines, and regulation of the actin cytoskeleton.
Collapse
|
47
|
Robertson HR, Gibson ES, Benke TA, Dell'Acqua ML. Regulation of postsynaptic structure and function by an A-kinase anchoring protein-membrane-associated guanylate kinase scaffolding complex. J Neurosci 2009; 29:7929-43. [PMID: 19535604 PMCID: PMC2716089 DOI: 10.1523/jneurosci.6093-08.2009] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 04/29/2009] [Accepted: 05/19/2009] [Indexed: 01/08/2023] Open
Abstract
A-kinase anchoring protein (AKAP) 79/150 is a scaffold protein found in dendritic spines that recruits the cAMP-dependent protein kinase (PKA) and protein phosphatase 2B-calcineurin (CaN) to membrane-associated guanylate kinase (MAGUK)-linked AMPA receptors (AMPARs) to control receptor phosphorylation and synaptic plasticity. However, AKAP79/150 may also coordinate regulation of AMPAR activity with spine structure directly through MAGUK binding and membrane-cytoskeletal interactions of its N-terminal targeting domain. In cultured hippocampal neurons, we observed that rat AKAP150 expression was low early in development but then increased coincident with spine formation and maturation. Overexpression of human AKAP79 in immature or mature neurons increased the number of dendritic filopodia and spines and enlarged spine area. However, RNA interference knockdown of AKAP150 decreased dendritic spine area only in mature neurons. Importantly, AKAP79 overexpression in immature neurons increased AMPAR postsynaptic localization and activity. Neither the AKAP79 PKA nor CaN anchoring domain was required for increasing dendritic protrusion numbers, spine area, or AMPAR synaptic localization; however, an internal region identified as the MAGUK binding domain was found to be essential as shown by expression of a MAGUK binding mutant that formed mainly filopodia and decreased AMPAR synaptic localization and activity. Expression of the AKAP79 N-terminal targeting domain alone also increased filopodia numbers but not spine area. Overall, these results demonstrate a novel structural role for AKAP79/150 in which the N-terminal targeting domain induces dendritic filopodia and binding to MAGUKs promotes spine enlargement and AMPAR recruitment.
Collapse
Affiliation(s)
| | | | - Timothy A. Benke
- Departments of Pharmacology
- Pediatrics, and
- Neurology and
- Program in Neuroscience, School of Medicine, University of Colorado Denver, Aurora, Colorado 80045
| | - Mark L. Dell'Acqua
- Departments of Pharmacology
- Program in Neuroscience, School of Medicine, University of Colorado Denver, Aurora, Colorado 80045
| |
Collapse
|
48
|
Hoe HS, Lee JY, Pak DTS. Combinatorial morphogenesis of dendritic spines and filopodia by SPAR and alpha-actinin2. Biochem Biophys Res Commun 2009; 384:55-60. [PMID: 19393616 DOI: 10.1016/j.bbrc.2009.04.069] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Accepted: 04/11/2009] [Indexed: 12/20/2022]
Abstract
Rap small GTPases regulate excitatory synaptic strength and morphological plasticity of dendritic spines. Changes in spine structure are mediated by the F-actin cytoskeleton, but the link between Rap activity and actin dynamics is unclear. Here, we report a novel interaction between SPAR, a postsynaptic inhibitor of Rap, and alpha-actinin, a family of actin-cross-linking proteins. SPAR and alpha-actinin engage in bidirectional structural plasticity of dendritic spines: SPAR promotes spine head enlargement, whereas increased alpha-actinin2 expression favors dendritic spine elongation and thinning. Surprisingly, SPAR and alpha-actinin2 can function in an additive rather than antagonistic fashion at the same dendritic spine, generating combination spine/filopodia hybrids. These data identify a molecular pathway bridging the actin cytoskeleton and Rap at synapses, and suggest that formation of spines and filopodia are not necessarily opposing forms of structural plasticity.
Collapse
Affiliation(s)
- Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | | | | |
Collapse
|
49
|
Preso, a novel PSD-95-interacting FERM and PDZ domain protein that regulates dendritic spine morphogenesis. J Neurosci 2009; 28:14546-56. [PMID: 19118189 DOI: 10.1523/jneurosci.3112-08.2008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PSD-95 is an abundant postsynaptic density (PSD) protein involved in the formation and regulation of excitatory synapses and dendritic spines, but the underlying mechanisms are not comprehensively understood. Here we report a novel PSD-95-interacting protein Preso that regulates spine morphogenesis. Preso is mainly expressed in the brain and contains WW (domain with two conserved Trp residues), PDZ (PSD-95/Dlg/ZO-1), FERM (4.1, ezrin, radixin, and moesin), and C-terminal PDZ-binding domains. These domains associate with actin filaments, the Rac1/Cdc42 guanine nucleotide exchange factor betaPix, phosphatidylinositol-4,5-bisphosphate, and the postsynaptic scaffolding protein PSD-95, respectively. Preso overexpression increases the density of dendritic spines in a manner requiring WW, PDZ, FERM, and PDZ-binding domains. Conversely, knockdown or dominant-negative inhibition of Preso decreases spine density, excitatory synaptic transmission, and the spine level of filamentous actin. These results suggest that Preso positively regulates spine density through its interaction with the synaptic plasma membrane, actin filaments, PSD-95, and the betaPix-based Rac1 signaling pathway.
Collapse
|
50
|
Sekimoto K, Triller A. Compatibility between itinerant synaptic receptors and stable postsynaptic structure. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2009; 79:031905. [PMID: 19391969 DOI: 10.1103/physreve.79.031905] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 01/13/2009] [Indexed: 05/27/2023]
Abstract
The density of synaptic receptors in front of presynaptic release sites is stabilized in the presence of scaffold proteins, but the receptors and scaffold molecules have local exchanges with characteristic times shorter than that of the receptor-scaffold assembly. We propose a mesoscopic model to account for the regulation of the local density of receptors as quasiequilibrium. It is based on two zones (synaptic and extrasynaptic) and multilayer (membrane, submembrane, and cytoplasmic) topological organization. The model includes the balance of chemical potentials associated with the receptor and scaffold protein concentrations in the various compartments. The model shows highly cooperative behavior including a "phase change" resulting in the formation of well-defined postsynaptic domains. This study provides theoretical tools to approach the complex issue of synaptic stability at the synapse, where receptors are transiently trapped yet rapidly diffuse laterally on the plasma membrane.
Collapse
Affiliation(s)
- Ken Sekimoto
- Laboratoire Matières et Systèmes Complexes, Université Paris Diderot and CNRS-UMR 7057, 10 rue Alice Domont et Léonie Duquet, 75013 Paris, France
| | | |
Collapse
|