1
|
Alexander CJ, Barzik M, Fujiwara I, Remmert K, Wang YX, Petralia RS, Friedman TB, Hammer JA. Myosin 18Aα targets the guanine nucleotide exchange factor β-Pix to the dendritic spines of cerebellar Purkinje neurons and promotes spine maturation. FASEB J 2021; 35:e21092. [PMID: 33378124 PMCID: PMC8357457 DOI: 10.1096/fj.202001449r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/24/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
Myosin 18Aα is a myosin 2-like protein containing unique N- and C-terminal protein interaction domains that co-assembles with myosin 2. One protein known to bind to myosin 18Aα is β-Pix, a guanine nucleotide exchange factor (GEF) for Rac1 and Cdc42 that has been shown to promote dendritic spine maturation by activating the assembly of actin and myosin filaments in spines. Here, we show that myosin 18A⍺ concentrates in the spines of cerebellar Purkinje neurons via co-assembly with myosin 2 and through an actin binding site in its N-terminal extension. miRNA-mediated knockdown of myosin 18A⍺ results in a significant defect in spine maturation that is rescued by an RNAi-immune version of myosin 18A⍺. Importantly, β-Pix co-localizes with myosin 18A⍺ in spines, and its spine localization is lost upon myosin 18A⍺ knockdown or when its myosin 18A⍺ binding site is deleted. Finally, we show that the spines of myosin 18A⍺ knockdown Purkinje neurons contain significantly less F-actin and myosin 2. Together, these data argue that mixed filaments of myosin 2 and myosin 18A⍺ form a complex with β-Pix in Purkinje neuron spines that promotes spine maturation by enhancing the assembly of actin and myosin filaments downstream of β-Pix's GEF activity.
Collapse
Affiliation(s)
- Christopher J Alexander
- Molecular Cell Biology Laboratory, Cell and Developmental Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Melanie Barzik
- Laboratory of Molecular Genetics, NIDCD, NIH, Bethesda, MD, USA
| | - Ikuko Fujiwara
- Graduate School of Science, Osaka City University, Osaka, Japan
| | | | - Ya-Xian Wang
- Advanced Imaging Core, NIDCD, NIH, Betheda, MD, USA
| | | | | | - John A Hammer
- Molecular Cell Biology Laboratory, Cell and Developmental Biology Center, NHLBI, NIH, Bethesda, MD, USA
| |
Collapse
|
2
|
Mikhaylova M, Rentsch J, Ewers H. Actomyosin Contractility in the Generation and Plasticity of Axons and Dendritic Spines. Cells 2020; 9:cells9092006. [PMID: 32882840 PMCID: PMC7565476 DOI: 10.3390/cells9092006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/19/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
Actin and non-muscle myosins have long been known to play important roles in growth cone steering and neurite outgrowth. More recently, novel functions for non-muscle myosin have been described in axons and dendritic spines. Consequently, possible roles of actomyosin contraction in organizing and maintaining structural properties of dendritic spines, the size and location of axon initial segment and axonal diameter are emerging research topics. In this review, we aim to summarize recent findings involving myosin localization and function in these compartments and to discuss possible roles for actomyosin in their function and the signaling pathways that control them.
Collapse
Affiliation(s)
- Marina Mikhaylova
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, 10115 Berlin, Germany
- DFG Emmy Noether Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Correspondence: (M.M.); (H.E.); Tel.: +49-4074-1055-815 (M.M.); +49-30-838-60644 (H.E.)
| | - Jakob Rentsch
- Institut für Chemie und Biochemie, Freie Universität Berlin, 14195 Berlin, Germany;
| | - Helge Ewers
- Institut für Chemie und Biochemie, Freie Universität Berlin, 14195 Berlin, Germany;
- Correspondence: (M.M.); (H.E.); Tel.: +49-4074-1055-815 (M.M.); +49-30-838-60644 (H.E.)
| |
Collapse
|
3
|
Conventional and Non-Conventional Roles of Non-Muscle Myosin II-Actin in Neuronal Development and Degeneration. Cells 2020; 9:cells9091926. [PMID: 32825197 PMCID: PMC7566000 DOI: 10.3390/cells9091926] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Myosins are motor proteins that use chemical energy to produce mechanical forces driving actin cytoskeletal dynamics. In the brain, the conventional non-muscle myosin II (NMII) regulates actin filament cytoskeletal assembly and contractile forces during structural remodeling of axons and dendrites, contributing to morphology, polarization, and migration of neurons during brain development. NMII isoforms also participate in neurotransmission and synaptic plasticity by driving actin cytoskeletal dynamics during synaptic vesicle release and retrieval, and formation, maturation, and remodeling of dendritic spines. NMIIs are expressed differentially in cerebral non-neuronal cells, such as microglia, astrocytes, and endothelial cells, wherein they play key functions in inflammation, myelination, and repair. Besides major efforts to understand the physiological functions and regulatory mechanisms of NMIIs in the nervous system, their contributions to brain pathologies are still largely unclear. Nonetheless, genetic mutations or deregulation of NMII and its regulatory effectors are linked to autism, schizophrenia, intellectual disability, and neurodegeneration, indicating non-conventional roles of NMIIs in cellular mechanisms underlying neurodevelopmental and neurodegenerative disorders. Here, we summarize the emerging biological roles of NMIIs in the brain, and discuss how actomyosin signaling contributes to dysfunction of neurons and glial cells in the context of neurological disorders. This knowledge is relevant for a deep understanding of NMIIs on the pathogenesis and therapeutics of neuropsychiatric and neurodegenerative diseases.
Collapse
|
4
|
Newell-Litwa KA, Horwitz R, Lamers ML. Non-muscle myosin II in disease: mechanisms and therapeutic opportunities. Dis Model Mech 2015; 8:1495-515. [PMID: 26542704 PMCID: PMC4728321 DOI: 10.1242/dmm.022103] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The actin motor protein non-muscle myosin II (NMII) acts as a master regulator of cell morphology, with a role in several essential cellular processes, including cell migration and post-synaptic dendritic spine plasticity in neurons. NMII also generates forces that alter biochemical signaling, by driving changes in interactions between actin-associated proteins that can ultimately regulate gene transcription. In addition to its roles in normal cellular physiology, NMII has recently emerged as a critical regulator of diverse, genetically complex diseases, including neuronal disorders, cancers and vascular disease. In the context of these disorders, NMII regulatory pathways can be directly mutated or indirectly altered by disease-causing mutations. NMII regulatory pathway genes are also increasingly found in disease-associated copy-number variants, particularly in neuronal disorders such as autism and schizophrenia. Furthermore, manipulation of NMII-mediated contractility regulates stem cell pluripotency and differentiation, thus highlighting the key role of NMII-based pharmaceuticals in the clinical success of stem cell therapies. In this Review, we discuss the emerging role of NMII activity and its regulation by kinases and microRNAs in the pathogenesis and prognosis of a diverse range of diseases, including neuronal disorders, cancer and vascular disease. We also address promising clinical applications and limitations of NMII-based inhibitors in the treatment of these diseases and the development of stem-cell-based therapies.
Collapse
Affiliation(s)
- Karen A Newell-Litwa
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rick Horwitz
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Marcelo L Lamers
- Department of Morphological Sciences, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-010, Brazil
| |
Collapse
|
5
|
Newell-Litwa KA, Badoual M, Asmussen H, Patel H, Whitmore L, Horwitz AR. ROCK1 and 2 differentially regulate actomyosin organization to drive cell and synaptic polarity. J Cell Biol 2015; 210:225-42. [PMID: 26169356 PMCID: PMC4508895 DOI: 10.1083/jcb.201504046] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/04/2015] [Indexed: 01/19/2023] Open
Abstract
RhoGTPases organize the actin cytoskeleton to generate diverse polarities, from front-back polarity in migrating cells to dendritic spine morphology in neurons. For example, RhoA through its effector kinase, RhoA kinase (ROCK), activates myosin II to form actomyosin filament bundles and large adhesions that locally inhibit and thereby polarize Rac1-driven actin polymerization to the protrusions of migratory fibroblasts and the head of dendritic spines. We have found that the two ROCK isoforms, ROCK1 and ROCK2, differentially regulate distinct molecular pathways downstream of RhoA, and their coordinated activities drive polarity in both cell migration and synapse formation. In particular, ROCK1 forms the stable actomyosin filament bundles that initiate front-back and dendritic spine polarity. In contrast, ROCK2 regulates contractile force and Rac1 activity at the leading edge of migratory cells and the spine head of neurons; it also specifically regulates cofilin-mediated actin remodeling that underlies the maturation of adhesions and the postsynaptic density of dendritic spines.
Collapse
Affiliation(s)
- Karen A Newell-Litwa
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Mathilde Badoual
- Laboratoire Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC), UMR 8165, Centre National de la Recherche Scientifique, University Paris-Sud and University Paris Diderot, 91405 Orsay, France
| | - Hannelore Asmussen
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Heather Patel
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Leanna Whitmore
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Alan Rick Horwitz
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
6
|
Harris KM, Weinberg RJ. Ultrastructure of synapses in the mammalian brain. Cold Spring Harb Perspect Biol 2012; 4:cshperspect.a005587. [PMID: 22357909 DOI: 10.1101/cshperspect.a005587] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The morphology and molecular composition of synapses provide the structural basis for synaptic function. This article reviews the electron microscopy of excitatory synapses on dendritic spines, using data from rodent hippocampus, cerebral cortex, and cerebellar cortex. Excitatory synapses have a prominent postsynaptic density, in contrast with inhibitory synapses, which have less dense presynaptic or postsynaptic specializations and are usually found on the cell body or proximal dendritic shaft. Immunogold labeling shows that the presynaptic active zone provides a scaffold for key molecules involved in the release of neurotransmitter, whereas the postsynaptic density contains ligand-gated ionic channels, other receptors, and a complex network of signaling molecules. Delineating the structure and molecular organization of these axospinous synapses represents a crucial step toward understanding the mechanisms that underlie synaptic transmission and the dynamic modulation of neurotransmission associated with short- and long-term synaptic plasticity.
Collapse
Affiliation(s)
- Kristen M Harris
- Center for Learning and Memory, Neurobiology Section, University of Texas, Austin, 78712, USA.
| | | |
Collapse
|
7
|
Johnson HW, Schell MJ. Neuronal IP3 3-kinase is an F-actin-bundling protein: role in dendritic targeting and regulation of spine morphology. Mol Biol Cell 2010; 20:5166-80. [PMID: 19846664 DOI: 10.1091/mbc.e09-01-0083] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The actin microstructure in dendritic spines is involved in synaptic plasticity. Inositol trisphosphate 3-kinase A (ITPKA) terminates Ins(1,4,5)P(3) signals emanating from spines and also binds filamentous actin (F-actin) through its amino terminal region (amino acids 1-66, N66). Here we investigated how ITPKA, independent of its kinase activity, regulates dendritic spine F-actin microstructure. We show that the N66 region of the protein mediates F-actin bundling. An N66 fusion protein bundled F-actin in vitro, and the bundling involved N66 dimerization. By mutagenesis we identified a point mutation in a predicted helical region that eliminated both F-actin binding and bundling, rendering the enzyme cytosolic. A fusion protein containing a minimal helical region (amino acids 9-52, N9-52) bound F-actin in vitro and in cells, but had lower affinity. In hippocampal neurons, GFP-tagged N66 expression was highly polarized, with targeting of the enzyme predominantly to spines. By contrast, N9-52-GFP expression occurred in actin-rich structures in dendrites and growth cones. Expression of N66-GFP tripled the length of dendritic protrusions, induced longer dendritic spine necks, and induced polarized actin motility in time-lapse assays. These results suggest that, in addition to its ability to regulate intracellular Ca(2+) via Ins(1,4,5)P(3) metabolism, ITPKA regulates structural plasticity.
Collapse
Affiliation(s)
- Hong W Johnson
- Department of Pharmacology, Uniformed Services University, Bethesda, MD 20814, USA
| | | |
Collapse
|
8
|
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
|
9
|
Korobova F, Svitkina T. Molecular architecture of synaptic actin cytoskeleton in hippocampal neurons reveals a mechanism of dendritic spine morphogenesis. Mol Biol Cell 2009; 21:165-76. [PMID: 19889835 PMCID: PMC2801710 DOI: 10.1091/mbc.e09-07-0596] [Citation(s) in RCA: 286] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Excitatory synapses in the brain play key roles in learning and memory. The formation and functions of postsynaptic mushroom-shaped structures, dendritic spines, and possibly of presynaptic terminals, rely on actin cytoskeleton remodeling. However, the cytoskeletal architecture of synapses remains unknown hindering the understanding of synapse morphogenesis. Using platinum replica electron microscopy, we characterized the cytoskeletal organization and molecular composition of dendritic spines, their precursors, dendritic filopodia, and presynaptic boutons. A branched actin filament network containing Arp2/3 complex and capping protein was a dominant feature of spine heads and presynaptic boutons. Surprisingly, the spine necks and bases, as well as dendritic filopodia, also contained a network, rather than a bundle, of branched and linear actin filaments that was immunopositive for Arp2/3 complex, capping protein, and myosin II, but not fascin. Thus, a tight actin filament bundle is not necessary for structural support of elongated filopodia-like protrusions. Dynamically, dendritic filopodia emerged from densities in the dendritic shaft, which by electron microscopy contained branched actin network associated with dendritic microtubules. We propose that dendritic spine morphogenesis begins from an actin patch elongating into a dendritic filopodium, which tip subsequently expands via Arp2/3 complex-dependent nucleation and which length is modulated by myosin II-dependent contractility.
Collapse
Affiliation(s)
- Farida Korobova
- Department of Biology, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
10
|
Kioussi C, Appu M, Löhr CV, Fischer KA, Bajaj G, Leid M, Ishmael JE. Co-expression of myosin II regulatory light chain and the NMDAR1 subunit in neonatal and adult mouse brain. Brain Res Bull 2007; 74:439-51. [DOI: 10.1016/j.brainresbull.2007.07.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Accepted: 07/12/2007] [Indexed: 01/26/2023]
|
11
|
Sekino Y, Kojima N, Shirao T. Role of actin cytoskeleton in dendritic spine morphogenesis. Neurochem Int 2007; 51:92-104. [PMID: 17590478 DOI: 10.1016/j.neuint.2007.04.029] [Citation(s) in RCA: 227] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Revised: 04/25/2007] [Accepted: 04/27/2007] [Indexed: 11/20/2022]
Abstract
Dendritic spines are the postsynaptic receptive regions of most excitatory synapses, and their morphological plasticity play a pivotal role in higher brain functions, such as learning and memory. The dynamics of spine morphology is due to the actin cytoskeleton concentrated highly in spines. Filopodia, which are thin and headless protrusions, are thought to be precursors of dendritic spines. Drebrin, a spine-resident side-binding protein of filamentous actin (F-actin), is responsible for recruiting F-actin and PSD-95 into filopodia, and is suggested to govern spine morphogenesis. Interestingly, some recent studies on neurological disorders accompanied by cognitive deficits suggested that the loss of drebrin from dendritic spines is a common pathognomonic feature of synaptic dysfunction. In this review, to understand the importance of actin-binding proteins in spine morphogenesis, we first outline the well-established knowledge pertaining to the actin cytoskeleton in non-neuronal cells, such as the mechanism of regulation by small GTPases, the equilibrium between globular actin (G-actin) and F-actin, and the distinct roles of various actin-binding proteins. Then, we review the dynamic changes in the localization of drebrin during synaptogenesis and in response to glutamate receptor activation. Because side-binding proteins are located upstream of the regulatory pathway for actin organization via other actin-binding proteins, we discuss the significance of drebrin in the regulatory mechanism of spine morphology through the reorganization of the actin cytoskeleton. In addition, we discuss the possible involvement of an actin-myosin interaction in the morphological plasticity of spines.
Collapse
Affiliation(s)
- Yuko Sekino
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | | | | |
Collapse
|
12
|
Deller T, Bas Orth C, Del Turco D, Vlachos A, Burbach GJ, Drakew A, Chabanis S, Korte M, Schwegler H, Haas CA, Frotscher M. A role for synaptopodin and the spine apparatus in hippocampal synaptic plasticity. Ann Anat 2007; 189:5-16. [PMID: 17319604 DOI: 10.1016/j.aanat.2006.06.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Spines are considered sites of synaptic plasticity in the brain and are capable of remodeling their shape and size. A molecule thathas been implicated in spine plasticity is the actin-associated protein synaptopodin. This article will review a series of studies aimed at elucidating the role of synaptopodin in the rodent brain. First, the developmental expression of synaptopodin mRNA and protein were studied; secondly, the subcellular localization of synaptopodin in hippocampal principal neurons was analyzed using confocal microscopy as well as electron microscopy and immunogold labelling; and, finally, the functional role of synaptopodin was investigated using a synaptopodin-deficient mouse. The results of these studies are: (1) synaptopodin expression byhippocampal principal neurons develops during the first postnatal weeks and increases in parallel with the maturation of spines in the hippocampus. (2) Synaptopodin is sorted to the spine compartment, where it is tightly associated with the spine apparatus, an enigmatic organelle believed to be involved in calcium storage or local protein synthesis. (3) Synaptopodin-deficient mice generated by gene targeting are viable but lack the spine apparatus organelle. These mice show deficitsin synaptic plasticity as well as impaired learning and memory. Taken together, these data implicate synaptopodin and the spine apparatus in the regulation of synaptic plasticity in the hippocampus. Future studies will be aimed at finding the molecular link between synaptopodin, the spine apparatus organelle, and synaptic plasticity.
Collapse
Affiliation(s)
- Thomas Deller
- Institute of Clinical Neuroanatomy, J.W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Ishmael JE, Safic M, Amparan D, Vogel WK, Pham T, Marley K, Filtz TM, Maier CS. Nonmuscle myosins II-B and Va are components of detergent-resistant membrane skeletons derived from mouse forebrain. Brain Res 2007; 1143:46-59. [PMID: 17321505 DOI: 10.1016/j.brainres.2007.01.061] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 01/14/2007] [Accepted: 01/16/2007] [Indexed: 10/23/2022]
Abstract
Myosins are actin-based molecular motors that may have specialized trafficking and contractile functions in cytoskeletal compartments that lack microtubules. The postsynaptic excitatory synapse is one such specialization, yet little is known about the spatial organization of myosin motor proteins in the mature brain. We used a proteomics approach to determine if class II and class V myosin isoforms are associated with Triton X-100-resistant membranes isolated from mouse forebrain. Two nonmuscle myosin isoforms (II-B and Va), were identified as components of lipid raft fractions that also contained typical membrane skeletal proteins such as non-erythrocyte spectrins, actin, alpha-actinin-2 and tubulin subunits. Other raft-associated proteins included lipid raft markers, proteins involved in cell adhesion and membrane dynamics, receptors and channels including glutamate receptor subunits, scaffolding and regulatory proteins. Myosin II-B and Va were also present in standard postsynaptic density (PSD) fractions, however retention of myosin II-B was strongly influenced by ATP status. If homogenates were supplemented with ATP, myosin II-B could be extracted from PSD I whereas myosin Va and other postsynaptic proteins were resistant to extraction. In summary, both myosin isoforms are components of a raft-associated membrane skeleton and are likely detected in standard PSD fractions as a result of their intrinsic ability to form actomyosin. Myosin II-B, however, is more loosely associated with PSD fractions than myosin Va, which appears to be a core PSD protein.
Collapse
Affiliation(s)
- Jane E Ishmael
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Fujisawa S, Shirao T, Aoki C. In vivo, competitive blockade of N-methyl-D-aspartate receptors induces rapid changes in filamentous actin and drebrin A distributions within dendritic spines of adult rat cortex. Neuroscience 2006; 140:1177-87. [PMID: 16650941 PMCID: PMC2844451 DOI: 10.1016/j.neuroscience.2006.03.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Accepted: 03/06/2006] [Indexed: 10/24/2022]
Abstract
In vitro studies have demonstrated that prolonged N-methyl-D-aspartate receptor (NMDAR) blockade triggers a homeostatic up-regulation of NMDARs at synapses. Such upregulation can also be seen within 30 min in vivo in adult rats, implicating trafficking of reserve pools of NMDARs. Here, we evaluated the involvement of filamentous actin (F-actin), the major cytoskeletal component in spines, in this rapid in vivo homeostatic response, using biotinylated phalloidin as its probe. We also immuno-labeled spines for drebrin A, an F-actin-binding protein found at excitatory synapses and with a proposed role of modulating F-actin's cross-linking with one another and interactions with NMDARs. Quantitative 2-D analysis of ultrastructural images revealed that NMDAR blockade increased filamentous actin labeling per spine by 62.5% (P<0.005). The proportion of dendritic spines immuno-labeled for drebrin A also increased significantly, from 67.5% to 85% following NMDAR blockade (P<0.001), especially among larger spines. The frequency distributions of spine widths and postsynaptic density lengths were not affected by the D-(+)-2-amino-5-phosphonopentanoic acid (D-APV) treatment. However, the average postsynaptic density length was reduced by 25 nm among the fewer, drebrin A immuno-negative spines, indicating that drebrin A confers stability to synapse size. We propose that, in a homeostatic in vivo response, increases of drebrin A and F-actin within spines can enhance NMDAR trafficking by reducing cytoskeletal rigidity within the spine cytoplasm without changing the overt morphology of axo-spinous synapses. Alternatively or in addition, the cytoskeletal redistribution within spine cytoplasm may be triggered by the D-APV-induced, homeostatic up-regulation of NMDAR.
Collapse
Affiliation(s)
- S Fujisawa
- Center for Neural Science, New York University, 4 Washington Place #809, New York, NY 10003, USA.
| | | | | |
Collapse
|
15
|
Rami G, Caillard O, Medina I, Pellegrino C, Fattoum A, Ben-Ari Y, Ferhat L. Change in the shape and density of dendritic spines caused by overexpression of acidic calponin in cultured hippocampal neurons. Hippocampus 2006; 16:183-97. [PMID: 16358313 DOI: 10.1002/hipo.20145] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dendritic spines are morphing structures believed to provide a cellular substrate for synaptic plasticity. It has been suggested that the actin cytoskeleton is the target of molecular mechanisms regulating spine morphology. Here we hypothesized that acidic calponin, an actin-binding protein, is one of the key regulators of actin filaments during spine plasticity. Our data showed that the overexpression of acidic calponin-GFP (green fluorescent protein) in primary cultures of rat hippocampal neurons causes an elongation of spines and an increase of their density as compared with those of GFP-expressing neurons. These effects required the actin-binding domains of acidic calponin. The close apposition of the presynatic marker synaptophysin to these long spines and the presence of specific postsynaptic markers actin, PSD-95, NR1, and GluR1 suggested the existence of functional excitatory synaptic contacts. Indeed, electrophysiological data showed that the postsynaptic overexpression of acidic calponin enhanced the frequency of miniature excitatory postsynaptic currents as compared with that of GFP-expressing neurons, but did not affect their properties such as amplitude, rise time, and half width. Studies in heterologous cells revealed that acidic calponin reorganized the actin filaments and stabilized them. Taken together, these findings show that acidic calponin regulates dendritic spine morphology and density, likely via regulation of the actin cytoskeleton reorganization and dynamic. Furthermore, the acidic calponin-induced spines are able to establish functional glutamatergic synapses. Such data suggest that acidic calponin is a key factor in the regulation of spine plasticity and synaptic activity.
Collapse
Affiliation(s)
- Guillaume Rami
- INMED/INSERM U29, 163 rue de Luminy, BP 13, 13273, Marseille Cedex 09, France
| | | | | | | | | | | | | |
Collapse
|
16
|
Amparan D, Avram D, Thomas CG, Lindahl MG, Yang J, Bajaj G, Ishmael JE. Direct interaction of myosin regulatory light chain with the NMDA receptor. J Neurochem 2005; 92:349-61. [PMID: 15663482 DOI: 10.1111/j.1471-4159.2004.02869.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
NMDA receptors interact with a variety of intracellular proteins at excitatory synapses. In this paper we show that myosin regulatory light chain (RLC) isolated from mouse brain is a NMDA receptor-interacting protein. Myosin RLC bound directly to the C-termini of both NMDA receptor 1 (NR1) and NMDA receptor 2 (NR2) subunits, rendering the interaction of myosin RLC with NMDA receptors distinct from that of calmodulin which is considered a NR1-interacting protein. Myosin RLC co-localized with NR1 in the dendritic spines of isolated hippocampal neurons, and was co-immunoprecipitated from brain extracts in a complex with NR1, NR2A, NR2B, PSD-95, Adaptor protein-2 and myosin II heavy chain. The C0 region of NR1 was necessary and sufficient for binding myosin RLC. Ca2+/calmodulin, but not calmodulin alone, displaced recombinant myosin RLC from the carboxy tail of NR1 indicating that myosin RLC and Ca2+/calmodulin can compete for a common binding site on NR1 in vitro. Myosin RLC is the only known substrate for myosin regulatory light chain kinase, which has recently been shown to modulate NMDA receptor function in isolated hippocampal neurons. Our results suggest that an additional level of NMDA receptor regulation may be mediated via a direct interaction with a light chain of myosin II. Thus, myosin RLC-NMDA receptor interactions may contribute to the contractile and motile forces that are placed upon NMDA receptor subunits during changes associated with synaptic plasticity and neural morphogenesis.
Collapse
Affiliation(s)
- David Amparan
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon 97331, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Ethell IM, Pasquale EB. Molecular mechanisms of dendritic spine development and remodeling. Prog Neurobiol 2005; 75:161-205. [PMID: 15882774 DOI: 10.1016/j.pneurobio.2005.02.003] [Citation(s) in RCA: 264] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Revised: 01/28/2005] [Accepted: 02/22/2005] [Indexed: 12/19/2022]
Abstract
Dendritic spines are small protrusions that cover the surface of dendrites and bear the postsynaptic component of excitatory synapses. Having an enlarged head connected to the dendrite by a narrow neck, dendritic spines provide a postsynaptic biochemical compartment that separates the synaptic space from the dendritic shaft and allows each spine to function as a partially independent unit. Spines develop around the time of synaptogenesis and are dynamic structures that continue to undergo remodeling over time. Changes in spine morphology and density influence the properties of neural circuits. Our knowledge of the structure and function of dendritic spines has progressed significantly since their discovery over a century ago, but many uncertainties still remain. For example, several different models have been put forth outlining the sequence of events that lead to the genesis of a spine. Although spines are small and apparently simple organelles with a cytoskeleton mainly composed of actin filaments, regulation of their morphology and physiology appears to be quite sophisticated. A multitude of molecules have been implicated in dendritic spine development and remodeling, suggesting that intricate networks of interconnected signaling pathways converge to regulate actin dynamics in spines. This complexity is not surprising, given the likely importance of dendritic spines in higher brain functions. In this review, we discuss the molecules that are currently known to mediate the exquisite sensitivity of spines to perturbations in their environment and we outline how these molecules interface with each other to mediate cascades of signals flowing from the spine surface to the actin cytoskeleton.
Collapse
Affiliation(s)
- Iryna M Ethell
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92521, USA
| | | |
Collapse
|
18
|
Hayashi ML, Choi SY, Rao BSS, Jung HY, Lee HK, Zhang D, Chattarji S, Kirkwood A, Tonegawa S. Altered cortical synaptic morphology and impaired memory consolidation in forebrain- specific dominant-negative PAK transgenic mice. Neuron 2004; 42:773-87. [PMID: 15182717 DOI: 10.1016/j.neuron.2004.05.003] [Citation(s) in RCA: 231] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2004] [Revised: 04/23/2004] [Accepted: 05/06/2004] [Indexed: 11/18/2022]
Abstract
Molecular and cellular mechanisms for memory consolidation in the cortex are poorly known. To study the relationships between synaptic structure and function in the cortex and consolidation of long-term memory, we have generated transgenic mice in which catalytic activity of PAK, a critical regulator of actin remodeling, is inhibited in the postnatal forebrain. Cortical neurons in these mice displayed fewer dendritic spines and an increased proportion of larger synapses compared to wild-type controls. These alterations in basal synaptic morphology correlated with enhanced mean synaptic strength and impaired bidirectional synaptic modifiability (enhanced LTP and reduced LTD) in the cortex. By contrast, spine morphology and synaptic plasticity were normal in the hippocampus of these mice. Importantly, these mice exhibited specific deficits in the consolidation phase of hippocampus-dependent memory. Thus, our results provide evidence for critical relationships between synaptic morphology and bidirectional modifiability of synaptic strength in the cortex and consolidation of long-term memory.
Collapse
Affiliation(s)
- Mansuo L Hayashi
- The Picower Center for Learning and Memory, Howard Hughes Medical Institute, RIKEN-MIT Neuroscience Research Center, Center for Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Muly EC, Smith Y, Allen P, Greengard P. Subcellular distribution of spinophilin immunolabeling in primate prefrontal cortex: localization to and within dendritic spines. J Comp Neurol 2004; 469:185-97. [PMID: 14694533 DOI: 10.1002/cne.11001] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Signal transduction in the nervous system depends on kinases and phosphatases, whose localization is regulated by a large group of scaffolding proteins. In particular, protein phosphatase-1 mediates dopamine's actions on a variety of substrates, including glutamate receptors, and this, in turn, depends on the binding of protein phosphatase-1 to its binding protein spinophilin. To better understand spinophilin's role in targeting protein phosphatase-1 within neurons, we used a combination of preembedding immunoperoxidase and postembedding immunogold labeling and electron microscopy to determine the localization of this scaffolding protein in macaque prefrontal cortex. Consistent with previous reports, spinophilin was found predominantly in dendritic spines, but a significant number of labeled dendritic shafts and, less frequently, glia and preterminal axons were also identified. By using the postembedding immunogold method, we further examined the distribution of spinophilin within dendritic spines. Spinophilin immunoreactivity was present throughout the spine, but the density of label was heterogeneous and defined two domains. The highest density of label was associated with the postsynaptic density and the 100 nm immediately subjacent to it. The deeper region of the spine, further than 100 nm from the postsynaptic density, had a lower density of spinophilin label. The distribution of spinophilin reported in this study supports its role in modulating glutamatergic neurotransmission but also suggests the possibility that spinophilin may target protein phosphatase-1 to other sites within the spine or to other neuronal or glial compartments.
Collapse
Affiliation(s)
- E Chris Muly
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia 30329, USA.
| | | | | | | |
Collapse
|
20
|
Krieger C, Hu JH, Pelech S. Aberrant protein kinases and phosphoproteins in amyotrophic lateral sclerosis. Trends Pharmacol Sci 2004; 24:535-41. [PMID: 14559406 DOI: 10.1016/j.tips.2003.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Charles Krieger
- School of Kinesiology, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| | | | | |
Collapse
|
21
|
Hu JH, Zhang H, Wagey R, Krieger C, Pelech SL. Protein kinase and protein phosphatase expression in amyotrophic lateral sclerosis spinal cord. J Neurochem 2003; 85:432-42. [PMID: 12675919 DOI: 10.1046/j.1471-4159.2003.01670.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The Kinetworks trade mark multi-immunoblotting technique was used to evaluate the expressions of 78 protein kinases, 24 protein phosphatases and phosphorylation states of 31 phosphoproteins in thoracic spinal cord tissue from control subjects and patients having the sporadic form of amyotrophic lateral sclerosis (ALS). In both the cytosolic (C) and particulate (P) fractions of spinal cord from ALS patients as compared with controls, there were increased levels of calcium/calmodulin-dependent protein kinase kinase (CaMKK; C = 120% increase/P = 580% increase;% change, compared with control), extracellular regulated kinase 2 (ERK2; C = 120% increase/P = 170% increase), G protein-coupled receptor kinase 2 (GRK2; C = 140% increase/P = 140% increase), phospho-Y279/216 glycogen synthase kinase 3 alpha/beta (GSK3alpha/beta; C = 90% increase/P = 220% increase), protein kinase B alpha (PKBalpha; C = 360% increase/P = 200% increase), phospho-T638 PKCalpha/beta (C = 630% increase/P = 170% increase), cGMP-dependent protein kinase (PKG; C = 100% increase/P = 75% increase), phospho-T451 dsRNA-dependent protein kinase (PKR; C = 2600% increase/P = 3330% increase), ribosomal S6 kinase 1 (RSK1; C = 750% increase/P = 630% increase), phospho-T389 p70 S6 kinase (S6K; C = 1000% increase/P = 460% increase), and protein-tyrosine phosphatase 1 delta (PTP1delta; C = 43% increase/P = 70% increase). Cytosolic increases in phospho-alpha-S724/gamma-S662 adducin (C = 15650% increase), PKCalpha (C = 100% increase) and PKCzeta (C = 190% increase) were found in ALS patients as compared with controls, while particulate increases in cAMP-dependent protein kinase (PKA; 43% increase), protein kinase C beta (PKCbeta; 330% increase), and stress-activated protein kinase beta (SAPKbeta; 34% increase) were also observed. Cyclin-dependent kinase-associated phosphatase (KAP) was apparently translocated, as it was reduced (31% decrease) in cytosolic fractions but elevated (100% increase) in particulate fractions of ALS spinal cord tissue. Our observations indicate that ALS is associated with the elevated expression and/or activation of many protein kinases, including PKCalpha, PKCbeta, PKCzeta and GSK3alpha/beta, which may augment neural death in ALS, and CaMKK, PKBalpha, Rsk1, S6K, and SAPK, which may be a response to neuronal injury that potentially can mitigate cell death.
Collapse
Affiliation(s)
- J-H Hu
- School of Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | | | | | | |
Collapse
|
22
|
Ferhat L, Esclapez M, Represa A, Fattoum A, Shirao T, Ben-Ari Y. Increased levels of acidic calponin during dendritic spine plasticity after pilocarpine-induced seizures. Hippocampus 2003; 13:845-58. [PMID: 14620880 DOI: 10.1002/hipo.10136] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We have previously shown that, in HEK 293 cells, overexpression of acidic calponin, an actin-binding protein, induces remodeling of actin filaments, leading to a change in cell morphology. In addition, this protein is found in dendritic spines of adult hippocampal neurons. We hypothesized that this protein plays a role in regulating actin-based filaments during dendritic spine plasticity. To assess this hypothesis, the pilocarpine model of temporal lobe epilepsy was selected because an important reorganization of the glutamatergic network, which includes an aberrant sprouting of granule cell axons, neo-synaptogenesis, and dendritic spine remodeling, is well established in the dentate gyrus. This reorganization begins after the initial period of status epilepticus after pilocarpine injection, during the silent period when animals display a normal behavior, and reaches a plateau at the chronic stage when the animals have developed spontaneous recurrent seizures. Our data show that the intensity of immunolabeling for acidic calponin was clearly increased in the inner one-third of the molecular layer of the dentate gyrus, the site of mossy fiber sprouting, and neo-synaptogenesis, at 1 and 2 weeks after pilocarpine injection (silent period) when the reorganization was taking place. In contrast, in chronic pilocarpine-treated animals, when the reorganization was established, the levels of labeling for acidic calponin in the inner molecular layer were similar to those observed in control rats. In addition, double immunostaining studies suggested that the increase in acidic calponin levels occurred within the dendritic spines. Altogether, these results are consistent with an involvement of acidic calponin in dendritic spine plasticity.
Collapse
|
23
|
Wright JW, Reichert JR, Davis CJ, Harding JW. Neural plasticity and the brain renin-angiotensin system. Neurosci Biobehav Rev 2002; 26:529-52. [PMID: 12367589 DOI: 10.1016/s0149-7634(02)00019-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The brain renin-angiotensin system mediates several classic physiologies including body water balance, maintenance of blood pressure, cyclicity of reproductive hormones and sexual behaviors, and regulation of pituitary gland hormones. In addition, angiotensin peptides have been implicated in neural plasticity and memory. The present review initially describes the extracellular matrix (ECM) and the roles of cell adhesion molecules (CAMs), matrix metalloproteinases, and tissue inhibitors of metalloproteinases in the maintenance and degradation of the ECM. It is the ECM that appears to permit synaptic remodeling and thus is critical to the plasticity that is presumed to underlie mechanisms of memory consolidation and retrieval. The interrelationship among long-term potentiation (LTP), CAMs, and synaptic strengthening is described, followed by the influence of angiotensins on LTP. There is strong support for an inhibitory influence by angiotensin II (AngII) and a facilitory role by angiotensin IV (AngIV), on LTP. Next, the influences of AngII and IV on associative and spatial memories are summarized. Finally, the impact of sleep deprivation on matrix metalloproteinases and memory function is described. Recent findings indicate that sleep deprivation-induced memory impairment is accompanied by a lack of appropriate changes in matrix metalloproteinases within the hippocampus and neocortex as compared with non-sleep deprived animals. These findings generally support an important contribution by angiotensin peptides to neural plasticity and memory consolidation.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology and Program in Neuroscience, Washington State University, PO Box 644820, Pullman, WA 99164-4820, USA.
| | | | | | | |
Collapse
|
24
|
Wright JW, Kramár EA, Meighan SE, Harding JW. Extracellular matrix molecules, long-term potentiation, memory consolidation and the brain angiotensin system. Peptides 2002; 23:221-46. [PMID: 11814638 DOI: 10.1016/s0196-9781(01)00599-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Considerable evidence now suggests an interrelationship among long-term potentiation (LTP), extracellular matrix (ECM) reconfiguration, synaptogenesis, and memory consolidation within the mammalian central nervous system. Extracellular matrix molecules provide the scaffolding necessary to permit synaptic remodeling and contribute to the regulation of ionic and nutritional homeostasis of surrounding cells. These molecules also facilitate cellular proliferation, movement, differentiation, and apoptosis. The present review initially focuses on characterizing the ECM and the roles of cell adhesion molecules (CAMs), matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs), in the maintenance and degradation of the ECM. The induction and maintenance of LTP is described. Debate continues over whether LTP results in some form of synaptic strengthening and in turn promotes memory consolidation. Next, the contribution of CAMs and TIMPs to the facilitation of LTP and memory consolidation is discussed. Finally, possible roles for angiotensins, MMPs, and tissue plasminogen activators in the facilitation of LTP and memory consolidation are described. These enzymatic pathways appear to be very important to an understanding of dysfunctional memory diseases such as Alzheimer's disease, multiple sclerosis, brain tumors, and infections.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, PO Box 644820, Pullman, WA 99164-4820, USA.
| | | | | | | |
Collapse
|
25
|
Petralia RS, Wang YX, Sans N, Worley PF, Hammer JA, Wenthold RJ. Glutamate receptor targeting in the postsynaptic spine involves mechanisms that are independent of myosin Va. Eur J Neurosci 2001; 13:1722-32. [PMID: 11359524 DOI: 10.1046/j.0953-816x.2001.01553.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Targeting of glutamate receptors (GluRs) to synapses involves rapid movement of intracellular receptors. This occurs in forms of synaptic upregulation of receptors, such as long-term potentiation. Thus, many GluRs are retained in a cytoplasmic pool in dendrites, and are transported to synapses for upregulation, presumably via motor proteins such as myosins travelling along cytoskeletal elements that extend up into the spine. In this ultrastructural immunogold study of the cerebellar cortex, we compared synapses between normal rats/mice and dilute lethal mutant mice. These mutant mice lack myosin Va, which has been implicated in protein trafficking at synapses. The postsynaptic spine in the cerebellum lacks the inositol trisphosphate receptor (IP3R) -laden reticular tubules that are found in normal mice and rats (Takagishi et al., Neurosci. Lett., 1996, 215, 169). Thus, we tested the hypothesis that myosin Va is necessary for transport of GluRs and associated proteins to spine synapses. We found that these spines retain a normal distribution of (i) GluRs (delta 1/2, GluR2/3 and mGluR1alpha), (ii) at least one associated MAGUK (membrane-associated guanylate kinase) protein, (iii) Homer (which interacts with mGluR1alpha and IP3Rs), (iv) the actin cytoskeleton, (v) the reticulum-associated protein BiP, and (vi) the motor-associated protein, dynein light chain. Thus, while myosin Va may maintain the IP3R-laden reticulum in the spine for proper calcium regulation, other mechanisms must be involved in the delivery of GluRs and associated proteins to synapses. Other possible mechanisms include diffusion along the extrasynaptic membrane and delivery via other motors running along the spine's actin cytoskeleton.
Collapse
Affiliation(s)
- R S Petralia
- Laboratory of Neurochemistry, NIDCD/NIH, 36/5D08, 36 Convent Dr MSC 4162, Bethesda MD 20892-4162, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Capani F, Martone ME, Deerinck TJ, Ellisman MH. Selective localization of high concentrations of F-actin in subpopulations of dendritic spines in rat central nervous system: a three-dimensional electron microscopic study. J Comp Neurol 2001; 435:156-70. [PMID: 11391638 DOI: 10.1002/cne.1199] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Dendritic spines differ considerably in their size, shape, and internal organization between brain regions. We examined the actin cytoskeleton in dendritic spines in hippocampus (areas CA1, CA3, and dentate gyrus), neostriatum, and cerebellum at both light and electron microscopic levels by using a novel high-resolution photoconversion method based in the high affinity of phalloidin for filamentous (F)-actin. In all brain regions, labeling was strongest in the heads of dendritic spines, diminishing in the spine neck. The number of labeled spines varied by region. Compared with the cerebellar molecular layer and area CA3, where nearly every dendritic spine was labeled, less than half the spines were labeled in CA1, dentate gyrus, and neostriatum. Serial section reconstructions of spines in these areas indicated that phalloidin labeling was restricted to the largest and most morphologically diverse dendritic spines. The resolution of the photoconversion technique allowed us to examine the localization and organization of actin filaments in the spine. The most intense staining for actin was found in the postsynaptic density and associated with the spines internal membrane system. In mushroom-shaped spines, F-actin staining was particularly strong between the lamellae of the spine apparatus. Three-dimensional reconstruction of labeled spines by using electron tomography showed that the labeled dense material was in continuity with the postsynaptic density. These results highlight differences in the actin cytoskeleton between different spine populations and provide novel information on the organization of the actin cytoskeleton in vivo.
Collapse
Affiliation(s)
- F Capani
- Department of Neurosciences, National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, California 92093-0608, USA
| | | | | | | |
Collapse
|
27
|
Roth SU, Sommer C, Mundel P, Kiessling M. Expression of synaptopodin, an actin-associated protein, in the rat hippocampus after limbic epilepsy. Brain Pathol 2001; 11:169-81. [PMID: 11303792 PMCID: PMC8098178 DOI: 10.1111/j.1750-3639.2001.tb00389.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Synaptopodin, a 100 kD protein, associated with the actin cytoskeleton of the postsynaptic density and dendritic spines, is thought to play a role in modulating actin-based shape and motility of dendritic spines during formation or elimination of synaptic contacts. Temporal lobe epilepsy in humans and in rats shows neuronal damage, aberrant sprouting of hippocampal mossy fibers and subsequent synaptic remodeling processes. Using kainic acid (KA) induced epilepsy in rats, the postictal hippocampal expression of synaptopodin was analyzed by in situ hybridization (ISH) and immunohistochemistry. Sprouting of mossy fibers was visualized by a modified Timm's staining. ISH showed elevated levels of Synaptopodin mRNA in perikarya of CA3 principal neurons, dentate granule cells and in surviving hilar neurons these levels persisted up to 8 weeks after seizure induction. Synaptopodin immunoreactivity in the dendritic layers of CA3, in the hilus and in the inner molecular layer of the dentate gyrus (DG) was initially reduced. Eight weeks after KA treatment Synaptopodin protein expression returned to control levels in dendritic layers of CA3 and in the entire molecular layer of the DG. The recovery of protein expression was accompanied by simultaneous supra- and infragranular mossy fiber sprouting. Postictal upregulation of Synaptopodin mRNA levels in target cell populations of limbic epilepsy-elicited damage and subsequent Synaptopodin protein expression largely co-localized with remodeling processes as demonstrated by mossy fiber sprouting. It may thus represent a novel postsynaptic molecular correlate of hippocampal neuroplasticity.
Collapse
Affiliation(s)
- S U Roth
- Department of Neuropathology, University of Heidelberg, Germany.
| | | | | | | |
Collapse
|
28
|
Abstract
Dendritic spines are dynamic structures that rapidly remodel their shape and size. These morphological adaptations are regulated by changes in synaptic activity, and result from rearrangements of the postsynaptic cytoskeleton. A cytoskeletal molecule preferentially found in mature spines is the actin-associated protein synaptopodin. It is strongly expressed by spine-bearing neurons in the olfactory bulb, striatum, cerebral cortex, and hippocampus. In the hippocampus, principal cells express synaptopodin mRNA and sort the protein to the spine compartment. Within the spine microdomain, synaptopodin is preferentially located in the spine neck and is closely associated with the spine apparatus. On the basis of these data we hypothesize that synaptopodin could affect spine motility by bundling actin filaments in the spine neck. In addition, it could link the actin cytoskeleton of spines to intracellular calcium stores, i.e., the spine apparatus and the smooth endoplasmic reticulum.
Collapse
Affiliation(s)
- T Deller
- Institute of Anatomy, University of Freiburg, Germany
| | | | | |
Collapse
|
29
|
Abstract
The dendritic spine may be considered a fusion of a specialized actin-based structure akin to filopodia and lamellopodia, with an excitatory postsynaptic density containing glutamate receptors and signal-transducing machinery. This specialized neuronal microdomain is the site of the majority of excitatory synaptic contacts in the mammalian brain. Regulation of spine morphology, composition, and stability are likely to contribute to long-lasting changes in synaptic efficacy. Thus, understanding the function and regulation of dendritic spines is a fundamental problem ranging from molecular through behavioral neurobiology. A complete understanding of dendritic spines will require a knowledge of all the molecular components and how these components interact. Here we wish to accomplish two goals: to catalog many of the known components of hippocampal dendritic spines and suggest how these may contribute to spine function; and to compare dendritic spines with other actin-based structures, namely lamellopodia, filopodia, microvilli, and stereocilia, to gain some insight into possible common vs. specialized mechanisms of regulation of the shape, motility, and longevity of these actin-based structures.
Collapse
Affiliation(s)
- A Rao
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
30
|
Abstract
Nearly all excitatory input in the hippocampus impinges on dendritic spines which serve as multifunctional compartments that can, at the very least, selectively isolate and amplify incoming signals. Their importance to normal brain function is highlighted by the severe mental impairment observed in most individuals having poorly developed spines (Purpura, Science 1974;186:1126-1128). Distinct groups of membrane proteins, cytoskeletal elements, scaffolding proteins, and second messenger-related proteins are concentrated particularly in dendritic spines, but their ability to generate, maintain, and coordinately regulate spine structure or function is poorly understood. Here we review the unique molecular composition of dendritic spines along with the factors known to influence dendritic spine development in order to construct a model of dendritic spine development in relation to synaptogenesis.
Collapse
Affiliation(s)
- W Zhang
- Fishberg Research Center for Neurobiology and Program in Cell Adhesion, Mount Sinai School of Medicine, New York, New York 10029,USA
| | | |
Collapse
|
31
|
Agassandian C, Plantier M, Fattoum A, Represa A, der Terrossian E. Subcellular distribution of calponin and caldesmon in rat hippocampus. Brain Res 2000; 887:444-9. [PMID: 11134639 DOI: 10.1016/s0006-8993(00)03030-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Caldesmon and calponin are two actin- and calmodulin-binding proteins involved in the 'actin-linked' regulation of smooth muscle and non-muscle Mg(2+)-actin-activated myosin II ATPase activity. In the present report we show that caldesmon and calponin are present in the post-synaptic side of symmetric synapses and accumulate in the post-synaptic densities of asymmetric synapses. Caldesmon- and calponin-immunoreactivities are also observed at the plasma membrane of the hippocampal neurones. Finally, while caldesmon seems strictly distributed to neurones, acidic calponin is present in both neurones and astrocytes.
Collapse
Affiliation(s)
- C Agassandian
- INMED/INSERM U29, Parc Scientifique de Luminy, 13009 Marseille Cedex, France
| | | | | | | | | |
Collapse
|
32
|
Miyazaki T, Watanabe M, Yamagishi A, Takahashi M. B2 exon splicing of nonmuscle myosin heavy chain IIB is differently regulated in developing and adult rat brain. Neurosci Res 2000; 37:299-306. [PMID: 10958978 DOI: 10.1016/s0168-0102(00)00130-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Two isoforms of nonmuscle myosin heavy chain IIB (MHC-IIB) are generated by alternative splicing; MHC-IIB(B2) differs from MHC-IIB(DeltaB2) by the insertion of B2 exon cassette near the actin binding region. Here we examined expressions of the two splice variants in developing and adult rat brains by in situ hybridization with isoform-specific oligonucleotide probes. In adult, MHC-IIB(DeltaB2) mRNA was highly expressed in neurons of the cerebral cortex, hippocampus, and cerebellum, whereas MHC-IIB(B2) mRNA was mainly distributed in the brainstem and cerebellum, with the highest level in Purkinje cells. During development, MHC-IIB(DeltaB2) mRNA was predominantly expressed in various regions of embryonic and neonatal brains, whereas MHC-IIB(B2) mRNA was low during embryonic stages. Up-regulation of MHC-IIB(B2) started in the cerebellum during early postnatal stages when dendritogenesis and synaptogenesis occur actively in Purkinje cells. We further employed immunofluorescence using two antibodies (one recognizing both splicing variants and another specific to MHC-IIB(B2)), and found similar and dense localization in cell bodies and dendrites of Purkinje cells. Therefore, splicing of the B2 exon cassette undergoes distinct temporal and spatial regulations in the brain in vivo, and the different exon usage seems unlikely to affect the somato-dendritic localization of MHC-IIB.
Collapse
Affiliation(s)
- T Miyazaki
- Division of Biological Sciences, Graduate School of Science, Hokkaido University, Sapporo, Japan
| | | | | | | |
Collapse
|
33
|
Postsynaptic scaffolds of excitatory and inhibitory synapses in hippocampal neurons: maintenance of core components independent of actin filaments and microtubules. J Neurosci 2000. [PMID: 10844024 DOI: 10.1523/jneurosci.20-12-04545.2000] [Citation(s) in RCA: 143] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The mechanisms responsible for anchoring molecular components of postsynaptic specializations in the mammalian brain are not well understood but are presumed to involve associations with cytoskeletal elements. Here we build on previous studies of neurotransmitter receptors (Allison et al., 1998) to analyze the modes of attachment of scaffolding and signal transducing proteins of both glutamate and GABA postsynaptic sites to either the microtubule or microfilament cytoskeleton. Hippocampal pyramidal neurons in culture were treated with latrunculin A to depolymerize actin, with vincristine to depolymerize microtubules, or with Triton X-100 to extract soluble proteins. The synaptic clustering of PSD-95, a putative NMDA receptor anchoring protein and a core component of the postsynaptic density (PSD), was unaffected by actin depolymerization, microtubule depolymerization, or detergent extraction. The same was largely true for GKAP, a PSD-95-interacting protein. In contrast, the synaptic clustering of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII)alpha, another core component of the PSD, was completely dependent on an intact actin cytoskeleton and was partially disrupted by detergent. Drebrin and alpha-actinin-2, actin-binding proteins concentrated in spines, were also dependent on F-actin for synaptic localization but were unaffected by detergent extraction. Surprisingly, the subcellular distributions of the inhibitory synaptic proteins GABA(A)R and gephyrin, which has a tubulin-binding motif, were unaffected by depolymerization of microtubules or actin or by detergent extraction. These studies reveal an unsuspected heterogeneity in the modes of attachment of postsynaptic proteins to the cytoskeleton and support the idea that PSD-95 and gephyrin may be core scaffolding components independent of the actin or tubulin cytoskeleton.
Collapse
|
34
|
Abstract
Our understanding of the organization of postsynaptic signaling systems at excitatory synapses has been aided by the identification of proteins in the postsynaptic density (PSD) fraction, a subcellular fraction enriched in structures with the morphology of PSDs. In this study, we have completed the identification of most major proteins in the PSD fraction with the use of an analytical method based on mass spectrometry coupled with searching of the protein sequence databases. At least one protein in each of 26 prominent protein bands from the PSD fraction has now been identified. We found 7 proteins not previously known to be constituents of the PSD fraction and 24 that had previously been associated with the PSD by other methods. The newly identified proteins include the heavy chain of myosin-Va (dilute myosin), a motor protein thought to be involved in vesicle trafficking, and the mammalian homolog of the yeast septin protein cdc10, which is important for bud formation in yeast. Both myosin-Va and cdc10 are threefold to fivefold enriched in the PSD fraction over brain homogenates. Immunocytochemical localization of myosin-Va in cultured hippocampal neurons shows that it partially colocalizes with PSD-95 at synapses and is also diffusely localized in cell bodies, dendrites, and axons. Cdc10 has a punctate distribution in cell bodies and dendrites, with some of the puncta colocalizing with PSD-95. The results support a role for myosin-Va in transport of materials into spines and for septins in the formation or maintenance of spines.
Collapse
|
35
|
Deller T, Merten T, Roth SU, Mundel P, Frotscher M. Actin-associated protein synaptopodin in the rat hippocampal formation: localization in the spine neck and close association with the spine apparatus of principal neurons. J Comp Neurol 2000; 418:164-81. [PMID: 10701442 DOI: 10.1002/(sici)1096-9861(20000306)418:2<164::aid-cne4>3.0.co;2-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Dendritic spines are sites of synaptic plasticity in the brain and are capable of remodeling their shape and size. However, little is known about the cellular mechanisms that regulate spine morphology and motility. Synaptopodin is a recently described actin-associated protein found in renal podocytes and dendritic spines (Mundel et al. J Cell Biol. [1997] 139:193-204), which is believed to play a role in spine plasticity. The present study analyzed the distribution of synaptopodin in the hippocampal formation. In situ hybridization histochemistry revealed a high constitutive expression of synaptopodin mRNA in the principal cell layers. Light microscopic immunohistochemistry showed that the protein is distributed throughout the hippocampal formation in a region- and lamina-specific manner. Postembedding immunogold histochemistry demonstrated that synaptopodin is exclusively present in dendrites and spines, specifically in the spine neck in close association with the spine apparatus. Spines lacking a spine apparatus are not immunoreactive for synaptopodin. These data suggest that synaptopodin links the spine apparatus to actin and may thus be involved in the actin-based plasticity of spines.
Collapse
Affiliation(s)
- T Deller
- Institute of Anatomy, University of Freiburg, Germany.
| | | | | | | | | |
Collapse
|
36
|
Cheng XT, Hayashi K, Shirao T. Non-muscle myosin IIB-like immunoreactivity is present at the drebrin-binding cytoskeleton in neurons. Neurosci Res 2000; 36:167-73. [PMID: 10711814 DOI: 10.1016/s0168-0102(99)00123-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Dendritic spines are extremely motile, providing a structural mechanism for synaptic plasticity. Actin-myosin interaction is thought to be responsible for the change in the shape of spine. We have already reported that drebrin, an actin-binding protein, inhibits actin-myosin interaction and is enriched in the dendritic spine of mature neurons. In this study, we prepared the actin cytoskeleton of dendritic spines as an immunoprecipitate with anti-drebrin antibody from adult guinea-pig brain, immunized mice with the cytoskeleton, and obtained a monoclonal antibody (MAb) called MAb G650. MAb G650 reacted with non-muscle myosin IIB, but it did not react with muscle myosin II or non-muscle myosin IIA. Immunoblotting with this antibody revealed that drebrin-binding cytoskeleton contains this myosin IIB-like immunreactivity. Immunohistochemistry using MAb G650 demonstrated that this myosin IIB-like immunreactivity can be detected in the neuronal cell bodies and their apical dendrites, where drebrin is hardly detected. These data demonstrate that a myosin subtype associated with drebrin-binding actin filaments in the dendritic spines is myosin IIB, although this myosin is widely distributed in somato-dendritic subdomains of neurons. Furthermore, it is indicated that the cytoskeletons in dendritic spine were uniquely characterized with actin-binding proteins such as drebrin, but not with myosins.
Collapse
Affiliation(s)
- X T Cheng
- Department of Neurobiology and Behavior, Gunma University School of Medicine, Japan
| | | | | |
Collapse
|
37
|
van Rossum D, Hanisch UK. Cytoskeletal dynamics in dendritic spines: direct modulation by glutamate receptors? Trends Neurosci 1999; 22:290-5. [PMID: 10370249 DOI: 10.1016/s0166-2236(99)01404-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A wide heterogeneity in dendritic-spine morphology is observed and ultrastructural changes can be induced following experimental stimulation of neurons. Morphological adaptation of a given spine might, thus, reflect its history or the current state of synaptic activity. These changes could conceivably result from rearrangements of the cytoskeleton that is subjacent to excitatory synapses. This article dicusses the direct and indirect interactions, between glutamate receptors and the cytoskeletal proteins, which include PDZ-containing proteins, actin and tubulin, as well as associated proteins. In fact, the synaptic-activity-controlled balancing of monomeric, dimeric and polymeric forms of actin and tubulin might underlie the changes in spine shape. These continuous adaptations could be relevant for physiological events, such as learning and the formation of memory.
Collapse
Affiliation(s)
- D van Rossum
- Max Delbrück Centre for Molecular Medicine, 13092 Berlin-Buch, Germany
| | | |
Collapse
|
38
|
Andersen P, Soleng AF. A thorny question: how does activity maintain dendritic spines? Nat Neurosci 1999; 2:5-7. [PMID: 10195170 DOI: 10.1038/4506] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
39
|
Matsuoka Y, Li X, Bennett V. Adducin is an in vivo substrate for protein kinase C: phosphorylation in the MARCKS-related domain inhibits activity in promoting spectrin-actin complexes and occurs in many cells, including dendritic spines of neurons. J Biophys Biochem Cytol 1998; 142:485-97. [PMID: 9679146 PMCID: PMC2133059 DOI: 10.1083/jcb.142.2.485] [Citation(s) in RCA: 170] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Adducin is a heteromeric protein with subunits containing a COOH-terminal myristoylated alanine-rich C kinase substrate (MARCKS)-related domain that caps and preferentially recruits spectrin to the fast-growing ends of actin filaments. The basic MARCKS-related domain, present in alpha, beta, and gamma adducin subunits, binds calmodulin and contains the major phosphorylation site for protein kinase C (PKC). This report presents the first evidence that phosphorylation of the MARCKS-related domain modifies in vitro and in vivo activities of adducin involving actin and spectrin, and we demonstrate that adducin is a prominent in vivo substrate for PKC or other phorbol 12-myristate 13-acetate (PMA)-activated kinases in multiple cell types, including neurons. PKC phosphorylation of native and recombinant adducin inhibited actin capping measured using pyrene-actin polymerization and abolished activity of adducin in recruiting spectrin to ends and sides of actin filaments. A polyclonal antibody specific to the phosphorylated state of the RTPS-serine, which is the major PKC phosphorylation site in the MARCKS-related domain, was used to evaluate phosphorylation of adducin in cells. Reactivity with phosphoadducin antibody in immunoblots increased twofold in rat hippocampal slices, eight- to ninefold in human embryonal kidney (HEK 293) cells, threefold in MDCK cells, and greater than 10-fold in human erythrocytes after treatments with PMA, but not with forskolin. Thus, the RTPS-serine of adducin is an in vivo phosphorylation site for PKC or other PMA-activated kinases but not for cAMP-dependent protein kinase in a variety of cell types. Physiological consequences of the two PKC phosphorylation sites in the MARCKS-related domain were investigated by stably transfecting MDCK cells with either wild-type or PKC-unphosphorylatable S716A/S726A mutant alpha adducin. The mutant alpha adducin was no longer concentrated at the cell membrane at sites of cell-cell contact, and instead it was distributed as a cytoplasmic punctate pattern. Moreover, the cells expressing the mutant alpha adducin exhibited increased levels of cytoplasmic spectrin, which was colocalized with the mutant alpha adducin in a punctate pattern. Immunofluorescence with the phosphoadducin-specific antibody revealed the RTPS-serine phosphorylation of adducin in postsynaptic areas in the developing rat hippocampus. High levels of the phosphoadducin were detected in the dendritic spines of cultured hippocampal neurons. Spectrin also was a component of dendritic spines, although at distinct sites from the ones containing phosphoadducin. These data demonstrate that adducin is a significant in vivo substrate for PKC or other PMA-activated kinases in a variety of cells, and that phosphorylation of adducin occurs in dendritic spines that are believed to respond to external signals by changes in morphology and reorganization of cytoskeletal structures.
Collapse
Affiliation(s)
- Y Matsuoka
- Howard Hughes Medical Institute and Departments of Cell Biology and Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
40
|
Changes in the ultrastructure of axospike contacts in human brain during normal aging and in vascular disorders. Bull Exp Biol Med 1998. [DOI: 10.1007/bf02446050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
41
|
Role of actin in anchoring postsynaptic receptors in cultured hippocampal neurons: differential attachment of NMDA versus AMPA receptors. J Neurosci 1998. [PMID: 9502803 DOI: 10.1523/jneurosci.18-07-02423.1998] [Citation(s) in RCA: 393] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We used actin-perturbing agents and detergent extraction of primary hippocampal cultures to test directly the role of the actin cytoskeleton in localizing GABAA receptors, AMPA- and NMDA-type glutamate receptors, and potential anchoring proteins at postsynaptic sites. Excitatory postsynaptic sites on dendritic spines contained a high concentration of F-actin that was resistant to cytochalasin D but could be depolymerized using the novel compound latrunculin A. Depolymerization of F-actin led to a 40% decrease in both the number of synaptic NMDA receptor (NMDAR1) clusters and the number of AMPA receptor (GluR1)-labeled spines. The nonsynaptic NMDA receptors appeared to remain clustered and to coalesce in cell bodies. alpha-Actinin-2, which binds both actin and NMDA receptors, dissociated from the receptor clusters, but PSD-95 remained associated with both the synaptic and nonsynaptic receptor clusters, consistent with a proposed cross-linking function. AMPA receptors behaved differently; on GABAergic neurons, the clusters redistributed to nonsynaptic sites, whereas on pyramidal neurons, many of the clusters appeared to disperse. Furthermore, in control neurons, AMPA receptors were detergent extractable from pyramidal cell spines, whereas AMPA receptors on GABAergic neurons and NMDA receptors were unextractable. GABAA receptors were not dependent on F-actin for the maintenance or synaptic localization of clusters. These results indicate fundamental differences in the mechanisms of receptor anchoring at postsynaptic sites, both regarding the anchoring of a single receptor (the AMPA receptor) in pyramidal cells versus GABAergic interneurons and regarding the anchoring of different receptors (AMPA vs NMDA receptors) at a single class of postsynaptic sites on pyramidal cell dendritic spines.
Collapse
|
42
|
Abstract
Dendritic spines contain high concentrations of actin, but neither the isoforms involved nor the mechanism of accumulation is known. In situ hybridization with specific probes established that beta- and gamma-cytoplasmic actins are selectively expressed at high levels by spine-bearing neurons. Transfecting cultured hippocampal neurons with epitope-tagged actin isoforms showed that cytoplasmic beta- and gamma-cytoplasmic actins are correctly targeted to spines, whereas alpha-cardiac muscle actin, which is normally absent from neurons, formed aggregates in dendrites. The transfected actin cDNAs contained only coding domains, suggesting that spine targeting involves amino acid sequences in the proteins, an interpretation supported by experiments with chimeric cDNAs in which C-terminal actin sequences were found to be determinative in spine targeting. By contrast to actin, microtubule components, including tubulin and MAP2, were restricted to the dendritic shaft domain. The close association of cytoplasmic actins with spines together with their general involvement in cell surface motility further supports the idea that actin motility-based changes in spine shape may contribute to synaptic plasticity.
Collapse
|
43
|
Abstract
Morphological changes in the dendritic spines have been postulated to participate in the expression of synaptic plasticity. The cytoskeleton is likely to play a key role in regulating spine structure. Here we examine the molecular mechanisms responsible for the changes in spine morphology, focusing on drebrin, an actin-binding protein that is known to change the properties of actin filaments. We found that adult-type drebrin is localized in the dendritic spines of rat forebrain neurons, where it binds to the cytoskeleton. To identify the cytoskeletal proteins that associated with drebrin, we isolated drebrin-containing cytoskeletons using immunoprecipitation with a drebrin antibody. Drebrin, actin, myosin, and gelsolin were co-precipitated. We next examined the effect of drebrin on actomyosin interaction. In vitro, drebrin reduced the sliding velocity of actin filaments on immobilized myosin and inhibited the actin-activated ATPase activity of myosin. These results suggest that drebrin may modulate the actomyosin interaction within spines and may play a role in the structure-based plasticity of synapses.
Collapse
|
44
|
Pinault D. Backpropagation of action potentials generated at ectopic axonal loci: hypothesis that axon terminals integrate local environmental signals. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 1995; 21:42-92. [PMID: 8547954 DOI: 10.1016/0165-0173(95)00004-m] [Citation(s) in RCA: 84] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This review deals with the fascinating complexity of presynaptic axon terminals that are characterized by a high degree of functional distinctiveness. In vertebrate and invertebrate neurons, all-or-none APs can take off not only from the axon hillock, but also from ectopic axonal loci including terminals. Invertebrate neurons display EAPs, for instance alternating with somatic APs, during survival functions. In vertebrate, EAPs have been recorded in the peripheral and central nervous systems in time relationship with physiological or pathological neuronal activities. In motor or sensory axon, EAP generation may be the cause of motor dysfunctioning or sensory perceptions and pain respectively. Locomotion is associated with rhythmic depolarizations of the presynaptic axonal membrane of primary afferents, which are ridden by robust EAP bursts. In central axons lying within an epileptic tissue EAP discharges, coinciding with paroxysmal ECoG waves, get longer as somatic discharges get shorter during seizure progression. Once invaded by an orthodromic burst, an ectopic axonal locus can display an EAP after discharge. Such loci can also fire during hyperpolarization or the postinhibitory excitatory period of the parent somata, but not during their tonic excitation. Neurons are thus endowed with electrophysiological intrinsic properties making possible the alternate discharges of somatic APs and EAPs. In invertebrate and vertebrate neurons, ectopic axonal loci fire while the parent somata stop firing, further suggesting that axon terminal networks are unique and individual functional entities. The functional importance of EAPs in the nervous systems is, however, not yet well understood. Ectopically generated axonal APs propagate backwards and forwards along the axon, thus acting as a retrograde and anterograde signal. In invertebrate neurons, somatically and ectopically generated APs cannot have the same effect on the postsynaptic membrane. As suggested by studies related to the dorsal root reflex, EAPs may not only be implied in the presynaptic modulation of transmitter release but also contribute significantly during their backpropagation to a powerful control (collision process) of incoming volleys. From experimental data related to epileptiform activities it is proposed that EAPs, once orthodromically conducted, might potentiate synapses, initiate, spread or maintain epileptic cellular processes. For instance, paroxysmal discharges of EAPs would exert, like a booster-driver, a powerful synchronizing synaptic drive upon a large number of excitatory and inhibitory postsynaptic neurons. We have proposed that, once backpropagated, EAPs are likewise capable of initiating (and anticipating) threshold and low-threshold somatodendritic depolarizations. Interestingly, an antidromic EAP can modulate the excitability of the parent soma.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- D Pinault
- Université Laval, Centre de Recherches en Neurobiologie, Hôpital de l'Enfant-Jésus, Québec, Canada
| |
Collapse
|
45
|
Mochida S. Role of myosin in neurotransmitter release: functional studies at synapses formed in culture. JOURNAL OF PHYSIOLOGY, PARIS 1995; 89:83-94. [PMID: 8520575 DOI: 10.1016/0928-4257(96)80555-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
To determine the functional role of presynaptic proteins in the neurotransmitter release, I have employed cholinergic synapses formed between superior cervical ganglion neurons in culture. These synapses expressed proteins characteristic of mature synapses: immunofluorescence staining showed the presence of synaptophysin, synaptotagmin, VAMP/synaptobrevin-2, syntaxin and neurexin. The function of these proteins seems to be similar to that of mature synapses because botulinum neurotoxins A, E and C1 inhibited neurotransmitter release evoked by presynaptic action potentials. With this preparation, I have obtained evidence supporting roles for myosin II and myosin light chain kinase in neurotransmitter secretion. Acetylcholine release was inhibited by introduction of antibody against myosin II or inhibitors of myosin light chain kinase. This evidence suggests a model in which myosin light chain kinase phosphorylates myosin, and the resultant change in actin-myosin interactions is involved in some steps of transmitter release.
Collapse
Affiliation(s)
- S Mochida
- Department of Physiology, Tokyo Medical College, Japan
| |
Collapse
|
46
|
Mochida S, Kobayashi H, Matsuda Y, Yuda Y, Muramoto K, Nonomura Y. Myosin II is involved in transmitter release at synapses formed between rat sympathetic neurons in culture. Neuron 1994; 13:1131-42. [PMID: 7946350 DOI: 10.1016/0896-6273(94)90051-5] [Citation(s) in RCA: 130] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The presynaptic function of myosin II was studied at cholinergic synapses formed between rat superior cervical ganglion neurons in culture. Immunofluorescent staining showed that myosin II was colocalized with synaptophysin at the presynaptic nerve terminals. Antimyosin II antibody introduced into presynaptic neurons inhibited synaptic transmission. Transmission was also inhibited in a dose-dependent manner by two inhibitors of myosin light chain kinase: a peptide, SM-1, and an organic inhibitor, wortmannin. The inhibition produced by these agents was dependent on presynaptic activity. Extracellularly applied wortmannin also blocked synaptic transmission, but its effects were slower in onset. Wortmannin also decreased postsynaptic potentials and post-tetanic potentiation in intact superior cervical ganglia. These results suggest a model in which myosin light chain kinase phosphorylates myosin, and the resultant change in actin-myosin interactions is involved in neurotransmitter release.
Collapse
Affiliation(s)
- S Mochida
- Department of Physiology, Tokyo Medical College, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Mochida S, Nonomura Y, Kobayashi H. Analysis of the mechanism for acetylcholine release at the synapse formed between rat sympathetic neurons in culture. Microsc Res Tech 1994; 29:94-102. [PMID: 7812040 DOI: 10.1002/jemt.1070290206] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Superior cervical ganglion neurons (SCGNs) were isolated from 7-day-old rat SCG and cultured in MEM containing horse serum, fetal calf serum, and nerve growth factor. In this culture condition, it is well known that the SCGNs form cholinergic synapse. In 3-4 weeks cultured neurons, immunofluorescent staining for synaptophysin, a small synaptic vesicle associated protein, showed the presence of synaptophysin as small dots on the surface of the soma. Postsynaptic potentials could be recorded in 50-80% of the neurons responding to evoked action potentials elicited in neighboring neurons. Because of its relatively large cell size and the short distance to the terminal, this synapse is a useful model for studying the mechanisms of acetylcholine (ACh) release by introducing substances such as antibodies or selective inhibitors into the presynaptic neuron by means of the whole-cell clamp technique. In this model synapse we tested the possible role of myosin in ACh release. The distribution of myosin was studied by the immunofluorescent staining technique. Myosin was recognized by the anti-myosin II IgG at the same synaptic terminals that showed the presence of synaptophysin with its antibody. The functional blockade of myosin by the antibody itself, and that of myosin light chain kinase (MLCK) by a pseudosubstrate inhibitor of MLCK, SM-1, or by a selective inhibitor of MLCK, wortmannin, induced depression of synaptic transmission in a dose-dependent manner. These indicate that phosphorylation of myosin by MLCK may be necessary for ACh release mechanisms.
Collapse
Affiliation(s)
- S Mochida
- Department of Physiology, Tokyo Medical College, Japan
| | | | | |
Collapse
|
48
|
García-Segura LM, Chowen JA, Párducz A, Naftolin F. Gonadal hormones as promoters of structural synaptic plasticity: cellular mechanisms. Prog Neurobiol 1994; 44:279-307. [PMID: 7886228 DOI: 10.1016/0301-0082(94)90042-6] [Citation(s) in RCA: 202] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
It is now obvious that the CNS is capable of undergoing a variety of plastic changes at all stages of development. Although the magnitude and distribution of these changes may be more dramatic in the immature animal, the adult brain retains a remarkable capacity for undergoing morphological and functional modifications. Throughout development, as well as in the postpubertal animal, gonadal steroids exert an important influence over the architecture of specific sex steroid-responsive areas, resulting in sexual dimorphisms at both morphological and physiological levels. We are only now beginning to gain insight into the mechanisms involved in gonadal steroid-induced synaptic changes. The number of synaptic inputs to specific neuronal populations is sexually dimorphic and this can be modulated by changes in the sex steroid environment. These modifications can be correlated with other morphological changes, such as glial cell activation, that are occurring simultaneously in the same anatomical area. Indeed, the close physical relationship between glial cells and neuronal synaptic contacts makes them an ideal candidate for participating in this process. Interestingly, not only can the morphology and immunoreactivity of glial cells be modulated by gonadal steroids, but a close negative correlation between the number of synapses and the amount of glial ensheathing of a neuron has been demonstrated, suggesting an active participation of these cells in this process. Glia have sex steroid receptors, are capable of producing and metabolizing steroids, and can produce other neuronal trophic factors in response to sex steroids. Hence, their role in gonadal steroid-induced synaptic plasticity is becoming more apparent. In addition, there is recent evidence that this process may involve certain cell surface molecules, such as the N-CAMs, since a specific isoform of this molecule, previously referred to as the embryonic form, is found in those areas of the brain which maintain the capacity to undergo synaptic remodelling. However, there is much work to be done in order to fully understand this phenomenon and before bringing it into a clinical setting in hopes of treating neurodegenerative diseases or injuries to the nervous system.
Collapse
|
49
|
Abstract
Changes in osmotic and hydrostatic pressure were found to modulate NMDA responses of cultured embryonic mouse neurons recorded in various patch-clamp configurations. In nucleated patches, NMDA currents were potentiated by reductions in external osmolarity and were reduced in hyper-osmotic solutions. These changes, which were greater for low concentrations of NMDA, were not observed for responses to kainate, glycine, or GABA. They could be mimicked by directly changing the pipette pressure in nucleated, outside-out, inside-out, and cell-attached patches. Osmosensitivity of NMDA responses was also observed in the whole-cell mode, but only after prolonged dialysis. Mechanosensitivity of NMDA receptors could have an important role in neuronal regions experiencing changes in membrane tension, such as spines or growth cones.
Collapse
Affiliation(s)
- P Paoletti
- Laboratoire de Neurobiologie, Ecole Normale Supérieure, Paris, France
| | | |
Collapse
|
50
|
Fifková E, Eason H, Bueltmann K, Lanman J. Changes in GABAergic and non-GABAergic synapses during chronic ethanol exposure and withdrawal in the dentate fascia of LS and SS mice. Alcohol Clin Exp Res 1994; 18:989-97. [PMID: 7978114 DOI: 10.1111/j.1530-0277.1994.tb00071.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Ethanol-sensitive LSIBG and ethanol-insensitive SSIBG mice were exposed to ethanol (23.5% ethanol-derived calories) for 4 months. Half of the animals was sacrificed at this time and the other half was withdrawn from the ethanol diet for 1 month. GABA immunoelectron microscopy was used to study the impact of the treatments on synaptic contacts in the dentate molecular layer. In the LS mice a significant loss of non-GABAergic axospinous synapses (26.7%; p < 0.05) was observed during ethanol exposure which was followed by a loss of GABAergic synapses on dendritic shafts (54.7%; p < 0.01) during withdrawal. In the SS mice there was a significant decrease in the non-GABAergic axospinous synapses (23.5%; p < 0.05) and a significant increase in axodendritic synapses (63.3%; p < 0.05) during ethanol exposure. The observed changes in the GABAergic and non-GABAergic innervation of the dentate fascia induced by ethanol were observed in the projection zone of the perforant path. They could adversely affect the hippocampal physiology with a consequent impairment of mnemonic functions.
Collapse
Affiliation(s)
- E Fifková
- Department of Psychology, University of Colorado, Boulder 80309
| | | | | | | |
Collapse
|