1
|
Kim YJ, Kim K, Lee Y, Min HW, Ko YH, Lee BR, Hur KH, Kim SK, Lee SY, Jang CG. The mutated cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2 S968F) regulates cocaine-induced reward behaviour and plasticity in the nucleus accumbens. Br J Pharmacol 2024; 181:3327-3345. [PMID: 38751203 DOI: 10.1111/bph.16427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2), as a component of the Wiskott-Aldrich syndrome protein family verprolin-homologous protein (WAVE) regulatory complex, is involved in actin polymerization, contributing to neuronal development and structural plasticity. Mutating serine-968 to phenylalanine (S968F) in CYFIP2 causes an altered cocaine response in mice. The neuronal mechanisms underlying this response remain unknown. EXPERIMENTAL APPROACH We performed cocaine reward-related behavioural tests and examined changes in synaptic protein phenotypes and neuronal morphology in the nucleus accumbens (NAc), using CYFIP2 S968F knock-in mice to investigate the role of CYFIP2 in regulating cocaine reward. KEY RESULTS CYFIP2 S968F mutation attenuated cocaine-induced behavioural sensitization and conditioned place preference. Cocaine-induced c-Fos was not observed in the NAc of CYFIP2 S968F knock-in mice. However, c-Fos induction was still evident in the medial prefrontal cortex (mPFC). CYFIP2 S968F mutation altered cocaine-associated CYFIP2 signalling, glutamatergic protein expression and synaptic density in the NAc following cocaine exposure. To further determine the role of CYFIP2 in NAc neuronal activity and the mPFC projecting to the NAc activity-mediating reward response, we used optogenetic tools to stimulate the NAc or mPFC-NAc pathway and observed that optogenetic activation of the NAc or mPFC-NAc pathway induced reward-related behaviours. This effect was not observed in the S968F mutation in CYFIP2. CONCLUSION AND IMPLICATIONS These results suggest that CYFIP2 plays a role in controlling cocaine-mediated neuronal function and structural plasticity in the NAc, and that CYFIP2 could serve as a target for regulating cocaine reward.
Collapse
Affiliation(s)
- Young-Jung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyungin Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hee-Won Min
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yong-Hyun Ko
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Bo-Ram Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kwang-Hyun Hur
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seon-Kyung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
2
|
Narayanan R, Levone BR, Winterer J, Nanda P, Müller A, Lobriglio T, Fiore R, Germain PL, Mihailovich M, Testa G, Schratt G. miRNA-mediated inhibition of an actomyosin network in hippocampal pyramidal neurons restricts sociability in adult male mice. Cell Rep 2024; 43:114429. [PMID: 38968074 DOI: 10.1016/j.celrep.2024.114429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/07/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024] Open
Abstract
Social deficits are frequently observed in patients suffering from neurodevelopmental disorders, but the molecular mechanisms regulating sociability are still poorly understood. We recently reported that the loss of the microRNA (miRNA) cluster miR-379-410 leads to hypersocial behavior and anxiety in mice. Here, we show that ablating miR-379-410 in excitatory neurons of the postnatal mouse hippocampus recapitulates hypersociability, but not anxiety. At the cellular level, miR-379-410 loss in excitatory neurons leads to larger dendritic spines, increased excitatory synaptic transmission, and upregulation of an actomyosin gene network. Re-expression of three cluster miRNAs, as well as pharmacological inhibition of the actomyosin activator ROCK, is sufficient to reinstate normal sociability in miR-379-410 knockout mice. Several actomyosin genes and miR-379-410 family members are reciprocally dysregulated in isogenic human induced pluripotent stem cell (iPSC)-derived neurons harboring a deletion present in patients with Williams-Beuren syndrome, characterized by hypersocial behavior. Together, our results show an miRNA-actomyosin pathway involved in social behavior regulation.
Collapse
Affiliation(s)
- Ramanathan Narayanan
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Brunno Rocha Levone
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Jochen Winterer
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Prakruti Nanda
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Alexander Müller
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Thomas Lobriglio
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Roberto Fiore
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland
| | - Pierre-Luc Germain
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland; Laboratory of Molecular and Behavioural Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland; Laboratory of Statistical Bioinformatics, IMLS, University of Zürich, Zürich, Switzerland
| | - Marija Mihailovich
- European Institute of Oncology (IEO) IRCCS, Milan, Italy; Human Technopole, Milan, Italy
| | - Giuseppe Testa
- European Institute of Oncology (IEO) IRCCS, Milan, Italy; Human Technopole, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Gerhard Schratt
- Laboratory of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, ETH-Zürich, Zürich, Switzerland.
| |
Collapse
|
3
|
McCormick LE, Evans EB, Barker NK, Herring LE, Diering GH, Gupton SL. The E3 ubiquitin ligase TRIM9 regulates synaptic function and actin dynamics in response to netrin-1. Mol Biol Cell 2024; 35:ar67. [PMID: 38507236 PMCID: PMC11151106 DOI: 10.1091/mbc.e23-12-0476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/04/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024] Open
Abstract
During neuronal development, dynamic filopodia emerge from dendrites and mature into functional dendritic spines during synaptogenesis. Dendritic filopodia and spines respond to extracellular cues, influencing dendritic spine shape and size as well as synaptic function. Previously, the E3 ubiquitin ligase TRIM9 was shown to regulate filopodia in early stages of neuronal development, including netrin-1-dependent axon guidance and branching. Here, we demonstrate that TRIM9 also localizes to dendritic filopodia and spines of murine cortical and hippocampal neurons during synaptogenesis and is required for synaptic responses to netrin. In particular, TRIM9 is enriched in the postsynaptic density (PSD) within dendritic spines and loss of Trim9 alters the PSD proteome, including the actin cytoskeleton landscape. While netrin exposure induces accumulation of the Arp2/3 complex and filamentous actin in dendritic spine heads, this response is disrupted by genetic deletion of Trim9. In addition, we document changes in the synaptic receptors associated with loss of Trim9. These defects converge on a loss of netrin-dependent increases in neuronal firing rates, indicating TRIM9 is required downstream of synaptic netrin-1 signaling. We propose that TRIM9 regulates cytoskeletal dynamics in dendritic spines and is required for the proper response to synaptic stimuli.
Collapse
Affiliation(s)
- Laura E. McCormick
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Elliot B. Evans
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Natalie K. Barker
- Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Laura E. Herring
- Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Graham H. Diering
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
4
|
Zeng Y, Wu T, Liang F, Long S, Guo W, Huang Y, Pei Z. Expression of human Ras-related protein Rab39B variant T168K in Caenorhabditis elegans leads to motor dysfunction and dopaminergic neuron degeneration. Heliyon 2024; 10:e26902. [PMID: 38444482 PMCID: PMC10912484 DOI: 10.1016/j.heliyon.2024.e26902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Human RAB39B gene is related to familial early-onset Parkinson disease. In early adulthood, men with the RAB39B c.503C > A (Thr168Lys, p. T168K) mutation develop typical tremor, bradykinesia, and alpha-synuclein accumulation. We investigated the pathological mechanism of RAB39B T168K in a Caenorhabditis elegans model. In early adult C. elegans, RAB39B T168K led to dopaminergic neuron degeneration that presented as disrupted dendrites and blunt neuronal cells. Abnormal dopamine secretion was inferred from a decline in motor function and a positive basal slowing phenotype. Dopamine-associated tests confirmed that synthesis and recycling of dopamine were normal. The RAB39B T168K mutation might impair dopamine vesicular transmission from the presynaptic membrane to the synaptic gap in dopaminergic neurons. The release-dependent feedback mechanism in neurotransmitters regulates the balance of receptor activities. Protein-protein interactions network analysis revealed that RAB39B may also function in lysosomal degradation and autophagy. Impaired disposal of misfolded α-synuclein eventually leads to protein aggregation. Thus, like other members of the Rab family, RAB39B may be involved in vesicular transport associated with dopamine secretion and α-synuclein clearance.
Collapse
Affiliation(s)
- Yixuan Zeng
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Tengteng Wu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fengyin Liang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Simei Long
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenyuan Guo
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Huang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Pei
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
McCormick LE, Evans EB, Barker NK, Herring LE, Diering GH, Gupton SL. The E3 ubiquitin ligase TRIM9 regulates synaptic function and actin dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.31.573790. [PMID: 38260647 PMCID: PMC10802335 DOI: 10.1101/2023.12.31.573790] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
During neuronal development, dynamic filopodia emerge from dendrites and mature into functional dendritic spines during synaptogenesis. Dendritic filopodia and spines respond to extracellular cues, influencing dendritic spine shape and size as well as synaptic function. Previously, the E3 ubiquitin ligase TRIM9 was shown to regulate filopodia in early stages of neuronal development, including netrin-1 dependent axon guidance and branching. Here we demonstrate TRIM9 also localizes to dendritic filopodia and spines of murine cortical and hippocampal neurons during synaptogenesis and is required for synaptic responses to netrin. In particular, TRIM9 is enriched in the post-synaptic density (PSD) within dendritic spines and loss of Trim9 alters the PSD proteome, including the actin cytoskeleton landscape. While netrin exposure induces accumulation of the Arp2/3 complex and filamentous actin in dendritic spine heads, this response is disrupted by genetic deletion of Trim9. In addition, we document changes in the synaptic receptors associated with loss of Trim9. These defects converge on a loss of netrin-dependent increases in neuronal firing rates, indicating TRIM9 is required downstream of synaptic netrin-1 signaling. We propose TRIM9 regulates cytoskeletal dynamics in dendritic spines and is required for the proper response to synaptic stimuli.
Collapse
Affiliation(s)
- Laura E McCormick
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Elliot B Evans
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Natalie K Barker
- Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Laura E Herring
- Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Graham H Diering
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
6
|
Cao YY, Wu LL, Li XN, Yuan YL, Zhao WW, Qi JX, Zhao XY, Ward N, Wang J. Molecular Mechanisms of AMPA Receptor Trafficking in the Nervous System. Int J Mol Sci 2023; 25:111. [PMID: 38203282 PMCID: PMC10779435 DOI: 10.3390/ijms25010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Synaptic plasticity enhances or reduces connections between neurons, affecting learning and memory. Postsynaptic AMPARs mediate greater than 90% of the rapid excitatory synaptic transmission in glutamatergic neurons. The number and subunit composition of AMPARs are fundamental to synaptic plasticity and the formation of entire neural networks. Accordingly, the insertion and functionalization of AMPARs at the postsynaptic membrane have become a core issue related to neural circuit formation and information processing in the central nervous system. In this review, we summarize current knowledge regarding the related mechanisms of AMPAR expression and trafficking. The proteins related to AMPAR trafficking are discussed in detail, including vesicle-related proteins, cytoskeletal proteins, synaptic proteins, and protein kinases. Furthermore, significant emphasis was placed on the pivotal role of the actin cytoskeleton, which spans throughout the entire transport process in AMPAR transport, indicating that the actin cytoskeleton may serve as a fundamental basis for AMPAR trafficking. Additionally, we summarize the proteases involved in AMPAR post-translational modifications. Moreover, we provide an overview of AMPAR transport and localization to the postsynaptic membrane. Understanding the assembly, trafficking, and dynamic synaptic expression mechanisms of AMPAR may provide valuable insights into the cognitive decline associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Yang Cao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Ling-Ling Wu
- School of Medicine, Shanghai University, Shanghai 200444, China;
| | - Xiao-Nan Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Yu-Lian Yuan
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Wan-Wei Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Jing-Xuan Qi
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Xu-Yu Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Natalie Ward
- Medical Laboratory, Exceptional Community Hospital, 19060 N John Wayne Pkwy, Maricopa, AZ 85139, USA;
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| |
Collapse
|
7
|
Raven F, Riemersma IW, Olthuis MF, Rybakovaite I, Meijer EL, Meerlo P, Van der Zee EA, Havekes R. Cofilin overactivation improves hippocampus-dependent short-term memory. Front Behav Neurosci 2023; 17:1243524. [PMID: 37638111 PMCID: PMC10448394 DOI: 10.3389/fnbeh.2023.1243524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Many living organisms of the animal kingdom have the fundamental ability to form and retrieve memories. Most information is initially stored as short-term memory, which is then converted to a more stable long-term memory through a process called memory consolidation. At the neuronal level, synaptic plasticity is crucial for memory storage. It includes the formation of new spines, as well as the modification of existing spines, thereby tuning and shaping synaptic efficacy. Cofilin critically contributes to memory processes as upon activation, it regulates the shape of dendritic spines by targeting actin filaments. We previously found that prolonged activation of cofilin in hippocampal neurons attenuated the formation of long-term object-location memories. Because the modification of spine shape and structure is also essential for short-term memory formation, we determined whether overactivation of hippocampal cofilin also influences the formation of short-term memories. To this end, mice were either injected with an adeno-associated virus expressing catalytically active cofilin, or an eGFP control, in the hippocampus. We show for the first time that cofilin overactivation improves short-term memory formation in the object-location memory task, without affecting anxiety-like behavior. Surprisingly, we found no effect of cofilin overactivation on AMPA receptor expression levels. Altogether, while cofilin overactivation might negatively impact the formation of long-lasting memories, it may benefit short-term plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
| |
Collapse
|
8
|
Gromova KV, Thies E, Janiesch PC, Lützenkirchen FP, Zhu Y, Stajano D, Dürst CD, Schweizer M, Konietzny A, Mikhaylova M, Gee CE, Kneussel M. The kinesin Kif21b binds myosin Va and mediates changes in actin dynamics underlying homeostatic synaptic downscaling. Cell Rep 2023; 42:112743. [PMID: 37418322 DOI: 10.1016/j.celrep.2023.112743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/15/2023] [Accepted: 06/19/2023] [Indexed: 07/09/2023] Open
Abstract
Homeostatic synaptic plasticity adjusts the strength of synapses to restrain neuronal activity within a physiological range. Postsynaptic guanylate kinase-associated protein (GKAP) controls the bidirectional synaptic scaling of AMPA receptors (AMPARs); however, mechanisms by which chronic activity triggers cytoskeletal remodeling to downscale synaptic transmission are barely understood. Here, we report that the microtubule-dependent kinesin motor Kif21b binds GKAP and likewise is located in dendritic spines in a myosin Va- and neuronal-activity-dependent manner. Kif21b depletion unexpectedly alters actin dynamics in spines, and adaptation of actin turnover following chronic activity is lost in Kif21b-knockout neurons. Consistent with a role of the kinesin in regulating actin dynamics, Kif21b overexpression promotes actin polymerization. Moreover, Kif21b controls GKAP removal from spines and the decrease of GluA2-containing AMPARs from the neuronal surface, thereby inducing homeostatic synaptic downscaling. Our data highlight a critical role of Kif21b at the synaptic actin cytoskeleton underlying homeostatic scaling of neuronal firing.
Collapse
Affiliation(s)
- Kira V Gromova
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| | - Edda Thies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Philipp C Janiesch
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Felix P Lützenkirchen
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Yipeng Zhu
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Daniele Stajano
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Céline D Dürst
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michaela Schweizer
- Core Facility Morphology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anja Konietzny
- RG Neuronal Protein Transport, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marina Mikhaylova
- RG Neuronal Protein Transport, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, 10099 Berlin, Germany
| | - Christine E Gee
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Hamburg Center of Neuroscience, HCNS, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| |
Collapse
|
9
|
Han KA, Ko J. Orchestration of synaptic functions by WAVE regulatory complex-mediated actin reorganization. Exp Mol Med 2023; 55:1065-1075. [PMID: 37258575 PMCID: PMC10318009 DOI: 10.1038/s12276-023-01004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 06/02/2023] Open
Abstract
The WAVE regulatory complex (WRC), composed of five components-Cyfip1/Sra1, WAVE/Scar, Abi, Nap1/Nckap1, and Brk1/HSPC300-is essential for proper actin cytoskeletal dynamics and remodeling in eukaryotic cells, likely by matching various patterned signals to Arp2/3-mediated actin nucleation. Accumulating evidence from recent studies has revealed diverse functions of the WRC in neurons, demonstrating its crucial role in dictating the assembly of molecular complexes for the patterning of various trans-synaptic signals. In this review, we discuss recent exciting findings on the physiological role of the WRC in regulating synaptic properties and highlight the involvement of WRC dysfunction in various brain disorders.
Collapse
Affiliation(s)
- Kyung Ah Han
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea.
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea.
| |
Collapse
|
10
|
Kinoshita M, Okamoto H. Acetylcholine potentiates glutamate transmission from the habenula to the interpeduncular nucleus in losers of social conflict. Curr Biol 2023:S0960-9822(23)00445-1. [PMID: 37105168 DOI: 10.1016/j.cub.2023.03.087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023]
Abstract
Switching behaviors from aggression to submission in losers at the end of conspecific social fighting is essential to avoid serious injury or death. We have previously shown that the experience of defeat induces a loser-specific potentiation in the habenula (Hb)-interpeduncular nucleus (IPN) and show here that this is induced by acetylcholine. Calcium imaging and electrophysiological recording using acute brain slices from winners and losers of fighting behavior in zebrafish revealed that the ventral IPN (vIPN) dominates over the dorsal IPN in the neural response to Hb stimulation in losers. We also show that GluA1 α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits on the postsynaptic membrane increased in the vIPN of losers. Furthermore, these loser-specific neural properties disappeared in the presence of an α7 nicotinic acetylcholine receptor (nAChR) antagonist and, conversely, were induced in brain slices of winners treated with α7 nAChR agonists. These data suggest that acetylcholine released from Hb terminals in the vIPN induces activation of α7 nAChR followed by an increase in postsynaptic membrane GluA1. This results in an increase in active synapses on postsynaptic neurons, resulting in the potentiation of neurotransmissions to the vIPN. This acetylcholine-induced neuromodulation could be the neural foundation for behavioral switching in losers. Our results could increase our understanding of the mechanisms of various mood disorders such as social anxiety disorder and social withdrawal.
Collapse
Affiliation(s)
- Masae Kinoshita
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Hitoshi Okamoto
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; RIKEN CBS-Kao Collaboration Center, Saitama 351-0198, Japan.
| |
Collapse
|
11
|
Filamin A organizes γ‑aminobutyric acid type B receptors at the plasma membrane. Nat Commun 2023; 14:34. [PMID: 36596803 PMCID: PMC9810740 DOI: 10.1038/s41467-022-35708-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/21/2022] [Indexed: 01/05/2023] Open
Abstract
The γ-aminobutyric acid type B (GABAB) receptor is a prototypical family C G protein-coupled receptor (GPCR) that plays a key role in the regulation of synaptic transmission. Although growing evidence suggests that GPCR signaling in neurons might be highly organized in time and space, limited information is available about the mechanisms controlling the nanoscale organization of GABAB receptors and other GPCRs on the neuronal plasma membrane. Using a combination of biochemical assays in vitro, single-particle tracking, and super-resolution microscopy, we provide evidence that the spatial organization and diffusion of GABAB receptors on the plasma membrane are governed by dynamic interactions with filamin A, which tethers the receptors to sub-cortical actin filaments. We further show that GABAB receptors are located together with filamin A in small nanodomains in hippocampal neurons. These interactions are mediated by the first intracellular loop of the GABAB1 subunit and modulate the kinetics of Gαi protein activation in response to GABA stimulation.
Collapse
|
12
|
Schirripa Spagnolo C, Luin S. Setting up multicolour TIRF microscopy down to the single molecule level. Biomol Concepts 2023; 14:bmc-2022-0032. [PMID: 37428621 DOI: 10.1515/bmc-2022-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/26/2023] [Indexed: 07/12/2023] Open
Abstract
Investigating biological mechanisms in ever greater detail requires continuous advances in microscopy techniques and setups. Total internal reflection fluorescence (TIRF) microscopy is a well-established technique for visualizing processes on the cell membrane. TIRF allows studies down to the single molecule level, mainly in single-colour applications. Instead, multicolour setups are still limited. Here, we describe our strategies for implementing a multi-channel TIRF microscopy system capable of simultaneous two-channel excitation and detection, starting from a single-colour commercial setup. First, we report some applications at high molecule density and then focus on the challenges we faced for achieving the single molecule level simultaneously in different channels, showing that rigorous optimizations on the setup are needed to increase its sensitivity up to this point, from camera setting to background minimization. We also discuss our strategies regarding crucial points of fluorescent labelling for this type of experiment: labelling strategy, kind of probe, efficiency, and orthogonality of the reaction, all of which are aspects that can influence the achievable results. This work may provide useful guidelines for setting up advanced single-molecule multi-channel TIRF experiments to obtain insights into interaction mechanisms on the cell membrane of living cells.
Collapse
Affiliation(s)
| | - Stefano Luin
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, I-56127, Pisa, Italy
- NEST Laboratory, Istituto Nanoscienze-Consiglio Nazionale delle ricerche (CNR), Piazza San Silvestro 12, I-56127, Pisa, Italy
| |
Collapse
|
13
|
Álvarez A, Gutiérrez D, Chandía-Cristi A, Yáñez M, Zanlungo S. c-Abl kinase at the crossroads of healthy synaptic remodeling and synaptic dysfunction in neurodegenerative diseases. Neural Regen Res 2023; 18:237-243. [PMID: 35900397 PMCID: PMC9396477 DOI: 10.4103/1673-5374.346540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Our ability to learn and remember depends on the active formation, remodeling, and elimination of synapses. Thus, the development and growth of synapses as well as their weakening and elimination are essential for neuronal rewiring. The structural reorganization of synaptic complexes, changes in actin cytoskeleton and organelle dynamics, as well as modulation of gene expression, determine synaptic plasticity. It has been proposed that dysregulation of these key synaptic homeostatic processes underlies the synaptic dysfunction observed in many neurodegenerative diseases. Much is known about downstream signaling of activated N-methyl-D-aspartate and α-amino-3-hydroxy-5-methyl-4-isoazolepropionate receptors; however, other signaling pathways can also contribute to synaptic plasticity and long-lasting changes in learning and memory. The non-receptor tyrosine kinase c-Abl (ABL1) is a key signal transducer of intra and extracellular signals, and it shuttles between the cytoplasm and the nucleus. This review focuses on c-Abl and its synaptic and neuronal functions. Here, we discuss the evidence showing that the activation of c-Abl can be detrimental to neurons, promoting the development of neurodegenerative diseases. Nevertheless, c-Abl activity seems to be in a pivotal balance between healthy synaptic plasticity, regulating dendritic spines remodeling and gene expression after cognitive training, and synaptic dysfunction and loss in neurodegenerative diseases. Thus, c-Abl genetic ablation not only improves learning and memory and modulates the brain genetic program of trained mice, but its absence provides dendritic spines resiliency against damage. Therefore, the present review has been designed to elucidate the common links between c-Abl regulation of structural changes that involve the actin cytoskeleton and organelles dynamics, and the transcriptional program activated during synaptic plasticity. By summarizing the recent discoveries on c-Abl functions, we aim to provide an overview of how its inhibition could be a potentially fruitful treatment to improve degenerative outcomes and delay memory loss.
Collapse
|
14
|
Post-Synapses in the Brain: Role of Dendritic and Spine Structures. Biomedicines 2022; 10:biomedicines10081859. [PMID: 36009405 PMCID: PMC9405724 DOI: 10.3390/biomedicines10081859] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/26/2022] [Accepted: 07/22/2022] [Indexed: 02/07/2023] Open
Abstract
Brain synapses are neuronal structures of the greatest interest. For a long time, however, the knowledge about them was variable, and interest was mostly focused on their pre-synaptic portions, especially neurotransmitter release from axon terminals. In the present review interest is focused on post-synapses, the structures receiving and converting pre-synaptic messages. Upon further modulation, such messages are transferred to dendritic fibers. Dendrites are profoundly different from axons; they are shorter and of variable thickness. Their post-synapses are of two types. Those called flat/intended/aspines, integrated into dendritic fibers, are very frequent in inhibitory neurons. The spines, small and stemming protrusions, connected to dendritic fibers by their necks, are present in almost all excitatory neurons. Several structures and functions including the post-synaptic densities and associated proteins, the nanoscale mechanisms of compartmentalization, the cytoskeletons of actin and microtubules, are analogous in the two post-synaptic forms. However other properties, such as plasticity and its functions of learning and memory, are largely distinct. Several properties of spines, including emersion from dendritic fibers, growth, change in shape and decreases in size up to disappearance, are specific. Spinal heads correspond to largely independent signaling compartments. They are motile, their local signaling is fast, however transport through their thin necks is slow. When single spines are activated separately, their dendritic effects are often lacking; when multiple spines are activated concomitantly, their effects take place. Defects of post-synaptic responses, especially those of spines, take place in various brain diseases. Here alterations affecting symptoms and future therapy are shown to occur in neurodegenerative diseases and autism spectrum disorders.
Collapse
|
15
|
Royo M, Escolano BA, Madrigal MP, Jurado S. AMPA Receptor Function in Hypothalamic Synapses. Front Synaptic Neurosci 2022; 14:833449. [PMID: 35173598 PMCID: PMC8842481 DOI: 10.3389/fnsyn.2022.833449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
AMPA receptors (AMPARs) are critical for mediating glutamatergic synaptic transmission and plasticity, thus playing a major role in the molecular machinery underlying cellular substrates of memory and learning. Their expression pattern, transport and regulatory mechanisms have been extensively studied in the hippocampus, but their functional properties in other brain regions remain poorly understood. Interestingly, electrophysiological and molecular evidence has confirmed a prominent role of AMPARs in the regulation of hypothalamic function. This review summarizes the existing evidence on AMPAR-mediated transmission in the hypothalamus, where they are believed to orchestrate the role of glutamatergic transmission in autonomous, neuroendocrine function, body homeostasis, and social behavior.
Collapse
|
16
|
Longatti A, Ponzoni L, Moretto E, Giansante G, Lattuada N, Colombo MN, Francolini M, Sala M, Murru L, Passafaro M. Arhgap22 Disruption Leads to RAC1 Hyperactivity Affecting Hippocampal Glutamatergic Synapses and Cognition in Mice. Mol Neurobiol 2021; 58:6092-6110. [PMID: 34455539 PMCID: PMC8639580 DOI: 10.1007/s12035-021-02502-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 07/15/2021] [Indexed: 11/03/2022]
Abstract
Rho GTPases are a class of G-proteins involved in several aspects of cellular biology, including the regulation of actin cytoskeleton. The most studied members of this family are RHOA and RAC1 that act in concert to regulate actin dynamics. Recently, Rho GTPases gained much attention as synaptic regulators in the mammalian central nervous system (CNS). In this context, ARHGAP22 protein has been previously shown to specifically inhibit RAC1 activity thus standing as critical cytoskeleton regulator in cancer cell models; however, whether this function is maintained in neurons in the CNS is unknown. Here, we generated a knockout animal model for arhgap22 and provided evidence of its role in the hippocampus. Specifically, we found that ARHGAP22 absence leads to RAC1 hyperactivity and to an increase in dendritic spine density with defects in synaptic structure, molecular composition, and plasticity. Furthermore, arhgap22 silencing causes impairment in cognition and a reduction in anxiety-like behavior in mice. We also found that inhibiting RAC1 restored synaptic plasticity in ARHGAP22 KO mice. All together, these results shed light on the specific role of ARHGAP22 in hippocampal excitatory synapse formation and function as well as in learning and memory behaviors.
Collapse
Affiliation(s)
- Anna Longatti
- Institute of Neuroscience, CNR, Milan, 20129, Italy
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, 20133, Milan, Italy
| | | | - Edoardo Moretto
- Institute of Neuroscience, CNR, Milan, 20129, Italy
- NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, 20126, Milan, Italy
| | - Giorgia Giansante
- Institute of Neuroscience, CNR, Milan, 20129, Italy
- NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, 20126, Milan, Italy
| | - Norma Lattuada
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi Di Milano, 20129, Milan, Italy
| | - Maria Nicol Colombo
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi Di Milano, 20129, Milan, Italy
| | - Maura Francolini
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi Di Milano, 20129, Milan, Italy
| | - Mariaelvina Sala
- Institute of Neuroscience, CNR, Milan, 20129, Italy
- NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, 20126, Milan, Italy
| | - Luca Murru
- Institute of Neuroscience, CNR, Milan, 20129, Italy.
- NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, 20126, Milan, Italy.
| | - Maria Passafaro
- Institute of Neuroscience, CNR, Milan, 20129, Italy.
- NeuroMI Milan Center for Neuroscience, Università Milano-Bicocca, 20126, Milan, Italy.
| |
Collapse
|
17
|
Dutta P, Bharti P, Kumar J, Maiti S. Role of actin cytoskeleton in the organization and function of ionotropic glutamate receptors. Curr Res Struct Biol 2021; 3:277-289. [PMID: 34766008 PMCID: PMC8569634 DOI: 10.1016/j.crstbi.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/04/2021] [Accepted: 10/09/2021] [Indexed: 12/22/2022] Open
Abstract
Neural networks with precise connection are compulsory for learning and memory. Various cellular events occur during the genesis of dendritic spines to their maturation, synapse formation, stabilization of the synapse, and proper signal transmission. The cortical actin cytoskeleton and its multiple regulatory proteins are crucial for the above cellular events. The different types of ionotropic glutamate receptors (iGluRs) present on the postsynaptic density (PSD) are also essential for learning and memory. Interaction of the iGluRs in association of their auxiliary proteins with actin cytoskeleton regulated by actin-binding proteins (ABPs) are required for precise long-term potentiation (LTP) and long-term depression (LTD). There has been a quest to understand the mechanistic detail of synapse function involving these receptors with dynamic actin cytoskeleton. A major, emerging area of investigation is the relationship between ABPs and iGluRs in synapse development. In this review we have summarized the current understanding of iGluRs functioning with respect to the actin cytoskeleton, scaffolding proteins, and their regulators. The AMPA, NMDA, Delta and Kainate receptors need the stable underlying actin cytoskeleton to anchor through synaptic proteins for precise synapse formation. The different types of ABPs present in neurons play a critical role in dynamizing/stabilizing the actin cytoskeleton needed for iGluRs function.
Collapse
Affiliation(s)
- Priyanka Dutta
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Pratibha Bharti
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Janesh Kumar
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Sankar Maiti
- Indian Institute of Science Education and Research, Kolkata, 741246, India
| |
Collapse
|
18
|
Godó S, Barabás K, Lengyel F, Ernszt D, Kovács T, Kecskés M, Varga C, Jánosi TZ, Makkai G, Kovács G, Orsolits B, Fujiwara T, Kusumi A, Ábrahám IM. Single-Molecule Imaging Reveals Rapid Estradiol Action on the Surface Movement of AMPA Receptors in Live Neurons. Front Cell Dev Biol 2021; 9:708715. [PMID: 34631701 PMCID: PMC8495425 DOI: 10.3389/fcell.2021.708715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
Gonadal steroid 17β-estradiol (E2) exerts rapid, non-genomic effects on neurons and strictly regulates learning and memory through altering glutamatergic neurotransmission and synaptic plasticity. However, its non-genomic effects on AMPARs are not well understood. Here, we analyzed the rapid effect of E2 on AMPARs using single-molecule tracking and super-resolution imaging techniques. We found that E2 rapidly decreased the surface movement of AMPAR via membrane G protein-coupled estrogen receptor 1 (GPER1) in neurites in a dose-dependent manner. The cortical actin network played a pivotal role in the GPER1 mediated effects of E2 on the surface mobility of AMPAR. E2 also decreased the surface movement of AMPAR both in synaptic and extrasynaptic regions on neurites and increased the synaptic dwell time of AMPARs. Our results provide evidence for understanding E2 action on neuronal plasticity and glutamatergic neurotransmission at the molecular level.
Collapse
Affiliation(s)
- Soma Godó
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Klaudia Barabás
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Ferenc Lengyel
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Dávid Ernszt
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Tamás Kovács
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Miklós Kecskés
- PTE-NAP Cortical Microcircuits Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, Pécs, Hungary
| | - Csaba Varga
- PTE-NAP Cortical Microcircuits Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Institute, Pécs, Hungary
| | - Tibor Z Jánosi
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Géza Makkai
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Gergely Kovács
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| | - Barbara Orsolits
- Laboratory of Neuroimmunology, Institute of Experimental Medicine of the Hungarian Academy of Sciences, Budapest, Hungary
| | - Takahiro Fujiwara
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
| | - Akihiro Kusumi
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Onna, Japan
| | - István M Ábrahám
- PTE-NAP Molecular Neuroendocrinology Research Group, Centre for Neuroscience, Szentágothai Research Center, Medical School, Institute of Physiology, University of Pécs, Pécs, Hungary
| |
Collapse
|
19
|
Oueslati Morales CO, Ignácz A, Bencsik N, Sziber Z, Rátkai AE, Lieb WS, Eisler SA, Szűcs A, Schlett K, Hausser A. Protein kinase D promotes activity-dependent AMPA receptor endocytosis in hippocampal neurons. Traffic 2021; 22:454-470. [PMID: 34564930 DOI: 10.1111/tra.12819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/12/2021] [Accepted: 09/14/2021] [Indexed: 12/18/2022]
Abstract
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) type glutamate receptors (AMPARs) mediate the majority of fast excitatory neurotransmission in the brain. The continuous trafficking of AMPARs into and out of synapses is a core feature of synaptic plasticity, which is considered as the cellular basis of learning and memory. The molecular mechanisms underlying the postsynaptic AMPAR trafficking, however, are still not fully understood. In this work, we demonstrate that the protein kinase D (PKD) family promotes basal and activity-induced AMPAR endocytosis in primary hippocampal neurons. Pharmacological inhibition of PKD increased synaptic levels of GluA1-containing AMPARs, slowed down their endocytic trafficking and increased neuronal network activity. By contrast, ectopic expression of constitutive active PKD decreased the synaptic level of AMPARs, while increasing their colocalization with early endosomes. Our results thus establish an important role for PKD in the regulation of postsynaptic AMPAR trafficking during synaptic plasticity.
Collapse
Affiliation(s)
- Carlos O Oueslati Morales
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Attila Ignácz
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Norbert Bencsik
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Zsofia Sziber
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Anikó Erika Rátkai
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Wolfgang S Lieb
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Stephan A Eisler
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Attila Szűcs
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Katalin Schlett
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Angelika Hausser
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
20
|
Wu QL, Gao Y, Li JT, Ma WY, Chen NH. The Role of AMPARs Composition and Trafficking in Synaptic Plasticity and Diseases. Cell Mol Neurobiol 2021; 42:2489-2504. [PMID: 34436728 DOI: 10.1007/s10571-021-01141-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/11/2021] [Indexed: 11/28/2022]
Abstract
AMPA receptors are tetrameric ionic glutamate receptors, which mediate 90% fast excitatory synaptic transmission induced by excitatory glutamate in the mammalian central nervous system through the activation or inactivation of ion channels. The alternation of synaptic AMPA receptor number and subtype is thought to be one of the primary mechanisms that involve in synaptic plasticity regulation and affect the functions in learning, memory, and cognition. The increasing of surface AMPARs enhances synaptic strength during long-term potentiation, whereas the decreasing of AMPARs weakens synaptic strength during the long-term depression. It is closely related to the AMPA receptor as well as its subunits assembly, trafficking, and degradation. The dysfunction of any step in these precise regulatory processes is likely to induce the disorder of synaptic transmission and loss of neurons, or even cause neuropsychiatric diseases ultimately. Therefore, it is useful to understand how AMPARs regulate synaptic plasticity and its role in related neuropsychiatric diseases via comprehending architecture and trafficking of the receptors. Here, we reviewed the progress in structure, expression, trafficking, and relationship with synaptic plasticity of AMPA receptor, especially in anxiety, depression, neurodegenerative disorders, and cerebral ischemia.
Collapse
Affiliation(s)
- Qing-Lin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yan Gao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jun-Tong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Yu Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China. .,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
21
|
Li ZZ, Han WJ, Sun ZC, Chen Y, Sun JY, Cai GH, Liu WN, Wang TZ, Xie YD, Mao HH, Wang F, Ma SB, Wang FD, Xie RG, Wu SX, Luo C. Extracellular matrix protein laminin β1 regulates pain sensitivity and anxiodepression-like behaviors in mice. J Clin Invest 2021; 131:e146323. [PMID: 34156983 DOI: 10.1172/jci146323] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/16/2021] [Indexed: 01/11/2023] Open
Abstract
Patients with neuropathic pain often experience comorbid psychiatric disorders. Cellular plasticity in the anterior cingulate cortex (ACC) is assumed to be a critical interface for pain perception and emotion. However, substantial efforts have thus far been focused on the intracellular mechanisms of plasticity rather than the extracellular alterations that might trigger and facilitate intracellular changes. Laminin, a key element of the extracellular matrix (ECM), consists of one α-, one β-, and one γ-chain and is implicated in several pathophysiological processes. Here, we showed in mice that laminin β1 (LAMB1) in the ACC was significantly downregulated upon peripheral neuropathy. Knockdown of LAMB1 in the ACC exacerbated pain sensitivity and induced anxiety and depression. Mechanistic analysis revealed that loss of LAMB1 caused actin dysregulation via interaction with integrin β1 and the subsequent Src-dependent RhoA/LIMK/cofilin pathway, leading to increased presynaptic transmitter release probability and abnormal postsynaptic spine remodeling, which in turn orchestrated the structural and functional plasticity of pyramidal neurons and eventually resulted in pain hypersensitivity and anxiodepression. This study sheds new light on the functional capability of ECM LAMB1 in modulating pain plasticity and identifies a mechanism that conveys extracellular alterations to intracellular plasticity. Moreover, we identified cingulate LAMB1/integrin β1 signaling as a promising therapeutic target for the treatment of neuropathic pain and associated anxiodepression.
Collapse
Affiliation(s)
- Zhen-Zhen Li
- Department of Neurobiology, School of Basic Medicine.,Department of Neurosurgery, Xijing Hospital, and
| | - Wen-Juan Han
- Department of Neurobiology, School of Basic Medicine
| | - Zhi-Chuan Sun
- Department of Neurobiology, School of Basic Medicine
| | - Yun Chen
- The Second Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jun-Yi Sun
- The Second Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Guo-Hong Cai
- Department of Neurobiology, School of Basic Medicine
| | - Wan-Neng Liu
- Department of Neurobiology, School of Basic Medicine.,College of Life Sciences, Northwest University, Xi'an, China
| | - Tao-Zhi Wang
- Department of Neurobiology, School of Basic Medicine.,Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Yang-Dan Xie
- The Second Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hong-Hui Mao
- Department of Neurobiology, School of Basic Medicine
| | - Fei Wang
- Department of Neurobiology, School of Basic Medicine.,Medical Experiment Center, Shaanxi University of Chinese Medicine, China
| | - Sui-Bin Ma
- Department of Neurobiology, School of Basic Medicine
| | - Fu-Dong Wang
- Department of Neurobiology, School of Basic Medicine
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine
| |
Collapse
|
22
|
Junyent S, Reeves JC, Szczerkowski JLA, Garcin CL, Trieu TJ, Wilson M, Lundie-Brown J, Habib SJ. Wnt- and glutamate-receptors orchestrate stem cell dynamics and asymmetric cell division. eLife 2021; 10:59791. [PMID: 34028355 PMCID: PMC8177892 DOI: 10.7554/elife.59791] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 05/21/2021] [Indexed: 12/16/2022] Open
Abstract
The Wnt-pathway is part of a signalling network that regulates many aspects of cell biology. Recently, we discovered crosstalk between AMPA/Kainate-type ionotropic glutamate receptors (iGluRs) and the Wnt-pathway during the initial Wnt3a-interaction at the cytonemes of mouse embryonic stem cells (ESCs). Here, we demonstrate that this crosstalk persists throughout the Wnt3a-response in ESCs. Both AMPA and Kainate receptors regulate early Wnt3a-recruitment, dynamics on the cell membrane, and orientation of the spindle towards a Wnt3a-source at mitosis. AMPA receptors specifically are required for segregating cell fate components during Wnt3a-mediated asymmetric cell division (ACD). Using Wnt-pathway component knockout lines, we determine that Wnt co-receptor Lrp6 has particular functionality over Lrp5 in cytoneme formation, and in facilitating ACD. Both Lrp5 and 6, alongside pathway effector β-catenin act in concert to mediate the positioning of the dynamic interaction with, and spindle orientation to, a localised Wnt3a-source. Wnt-iGluR crosstalk may prove pervasive throughout embryonic and adult stem cell signalling.
Collapse
Affiliation(s)
- Sergi Junyent
- Centre for Stem Cells and Regenerative Medicine, King's College LondonLondonUnited Kingdom
| | - Joshua C Reeves
- Centre for Stem Cells and Regenerative Medicine, King's College LondonLondonUnited Kingdom
| | - James LA Szczerkowski
- Centre for Stem Cells and Regenerative Medicine, King's College LondonLondonUnited Kingdom
| | - Clare L Garcin
- Centre for Stem Cells and Regenerative Medicine, King's College LondonLondonUnited Kingdom
| | - Tung-Jui Trieu
- Centre for Stem Cells and Regenerative Medicine, King's College LondonLondonUnited Kingdom
| | - Matthew Wilson
- Centre for Stem Cells and Regenerative Medicine, King's College LondonLondonUnited Kingdom
| | - Jethro Lundie-Brown
- Centre for Stem Cells and Regenerative Medicine, King's College LondonLondonUnited Kingdom
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College LondonLondonUnited Kingdom
| |
Collapse
|
23
|
Chemical Stimulation of Rodent and Human Cortical Synaptosomes: Implications in Neurodegeneration. Cells 2021; 10:cells10051174. [PMID: 34065927 PMCID: PMC8151714 DOI: 10.3390/cells10051174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/29/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022] Open
Abstract
Synaptic plasticity events, including long-term potentiation (LTP), are often regarded as correlates of brain functions of memory and cognition. One of the central players in these plasticity-related phenomena is the α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor (AMPAR). Increased levels of AMPARs on postsynaptic membranes thus constitute a biochemical measure of LTP. Isolated synaptic terminals (synaptosomes) are an excellent ex vivo tool to monitor synaptic physiology in healthy and diseased brains, particularly in human research. We herein describe three protocols for chemically-induced LTP (cLTP) in synaptosomes from both rodent and human brain tissues. Two of these chemical stimulation protocols are described for the first time in synaptosomes. A pharmacological block of synaptosomal actin dynamics confirmed the efficiency of the cLTP protocols. Furthermore, the study prototypically evaluated the deficiency of cLTP in cortical synaptosomes obtained from human cases of early-onset Alzheimer’s disease (EOAD) and frontotemporal lobar degeneration (FLTD), as well as an animal model that mimics FLTD.
Collapse
|
24
|
Kloc M, Uosef A, Villagran M, Zdanowski R, Kubiak JZ, Wosik J, Ghobrial RM. RhoA- and Actin-Dependent Functions of Macrophages from the Rodent Cardiac Transplantation Model Perspective -Timing Is the Essence. BIOLOGY 2021; 10:biology10020070. [PMID: 33498417 PMCID: PMC7909416 DOI: 10.3390/biology10020070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary The functions of animal and human cells depend on the actin cytoskeleton and its regulating protein called the RhoA. The actin cytoskeleton and RhoA also regulate the response of the immune cells such as macrophages to the microbial invasion and/or the presence of a non-self, such as a transplanted organ. The immune response against transplant occurs in several steps. The early step occurring within days post-transplantation is called the acute rejection and the late step, occurring months to years post-transplantation, is called the chronic rejection. In clinical transplantation, acute rejection is easily manageable by the anti-rejection drugs. However, there is no cure for chronic rejection, which is caused by the macrophages entering the transplant and promoting blockage of its blood vessels and destruction of tissue. We discuss here how the inhibition of the RhoA and actin cytoskeleton polymerization in the macrophages, either by genetic interference or pharmacologically, prevents macrophage entry into the transplanted organ and prevents chronic rejection, and also how it affects the anti-microbial function of the macrophages. We also focus on the importance of timing of the macrophage functions in chronic rejection and how the circadian rhythm may affect the anti-chronic rejection and anti-microbial therapies. Abstract The small GTPase RhoA, and its down-stream effector ROCK kinase, and the interacting Rac1 and mTORC2 pathways, are the principal regulators of the actin cytoskeleton and actin-related functions in all eukaryotic cells, including the immune cells. As such, they also regulate the phenotypes and functions of macrophages in the immune response and beyond. Here, we review the results of our and other’s studies on the role of the actin and RhoA pathway in shaping the macrophage functions in general and macrophage immune response during the development of chronic (long term) rejection of allografts in the rodent cardiac transplantation model. We focus on the importance of timing of the macrophage functions in chronic rejection and how the circadian rhythm may affect the anti-chronic rejection therapies.
Collapse
Affiliation(s)
- Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- M.D. Anderson Cancer Center, Department of Genetics, The University of Texas, Houston, TX 77030, USA
- Correspondence:
| | - Ahmed Uosef
- The Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
| | - Martha Villagran
- Electrical and Computer Engineering Department, University of Houston, Houston, TX 77204, USA; (M.V.); (J.W.)
- Texas Center for Superconductivity, University of Houston, Houston, TX 77204, USA
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine (WIM), 04-141 Warsaw, Poland;
| | - Jacek Z. Kubiak
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), 01-163 Warsaw, Poland;
- Cell Cycle Group, CNRS, Faculty of Medicine, Institute of Genetics and Development of Rennes, University of Rennes, UMR, 6290 Rennes, France
| | - Jarek Wosik
- Electrical and Computer Engineering Department, University of Houston, Houston, TX 77204, USA; (M.V.); (J.W.)
- Texas Center for Superconductivity, University of Houston, Houston, TX 77204, USA
| | - Rafik M. Ghobrial
- The Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
25
|
Ji B, Skup M. Roles of palmitoylation in structural long-term synaptic plasticity. Mol Brain 2021; 14:8. [PMID: 33430908 PMCID: PMC7802216 DOI: 10.1186/s13041-020-00717-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/15/2020] [Indexed: 11/30/2022] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are important cellular mechanisms underlying learning and memory processes. N-Methyl-d-aspartate receptor (NMDAR)-dependent LTP and LTD play especially crucial roles in these functions, and their expression depends on changes in the number and single channel conductance of the major ionotropic glutamate receptor α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) located on the postsynaptic membrane. Structural changes in dendritic spines comprise the morphological platform and support for molecular changes in the execution of synaptic plasticity and memory storage. At the molecular level, spine morphology is directly determined by actin cytoskeleton organization within the spine and indirectly stabilized and consolidated by scaffold proteins at the spine head. Palmitoylation, as a uniquely reversible lipid modification with the ability to regulate protein membrane localization and trafficking, plays significant roles in the structural and functional regulation of LTP and LTD. Altered structural plasticity of dendritic spines is also considered a hallmark of neurodevelopmental disorders, while genetic evidence strongly links abnormal brain function to impaired palmitoylation. Numerous studies have indicated that palmitoylation contributes to morphological spine modifications. In this review, we have gathered data showing that the regulatory proteins that modulate the actin network and scaffold proteins related to AMPAR-mediated neurotransmission also undergo palmitoylation and play roles in modifying spine architecture during structural plasticity.
Collapse
Affiliation(s)
- Benjun Ji
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| | - Małgorzata Skup
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| |
Collapse
|
26
|
Kloc M, Uosef A, Kubiak JZ, Ghobrial RM. Role of Macrophages and RhoA Pathway in Atherosclerosis. Int J Mol Sci 2020; 22:ijms22010216. [PMID: 33379334 PMCID: PMC7796231 DOI: 10.3390/ijms22010216] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/20/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
The development, progression, or stabilization of the atherosclerotic plaque depends on the pro-inflammatory and anti-inflammatory macrophages. The influx of the macrophages and the regulation of macrophage phenotype, inflammatory or anti-inflammatory, are controlled by the small GTPase RhoA and its downstream effectors. Therefore, macrophages and the components of the RhoA pathway are attractive targets for anti-atherosclerotic therapies, which would inhibit macrophage influx and inflammatory phenotype, maintain an anti-inflammatory environment, and promote tissue remodeling and repair. Here, we discuss the recent findings on the role of macrophages and RhoA pathway in the atherosclerotic plaque formation and resolution and the novel therapeutic approaches.
Collapse
Affiliation(s)
- Malgorzata Kloc
- Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, USA
- M.D. Anderson Cancer Center, Department of Genetics, University of Texas, Houston, TX 77030, USA
- Correspondence:
| | - Ahmed Uosef
- Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Jacek Z. Kubiak
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), 01-001 Warsaw, Poland;
- Cell Cycle Group, Institute of Genetics and Development of Rennes (IGDR), Faculty of Medicine, Univ Rennes, CNRS, UMR 6290, 35000 Rennes, France
| | - Rafik Mark Ghobrial
- Houston Methodist Research Institute, Houston, TX 77030, USA; (A.U.); (R.M.G.)
- Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
27
|
Arriagada-Diaz J, Prado-Vega L, Cárdenas Díaz AM, Ardiles AO, Gonzalez-Jamett AM. Dynamin Superfamily at Pre- and Postsynapses: Master Regulators of Synaptic Transmission and Plasticity in Health and Disease. Neuroscientist 2020; 28:41-58. [PMID: 33300419 DOI: 10.1177/1073858420974313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Dynamin superfamily proteins (DSPs) comprise a large group of GTP-ases that orchestrate membrane fusion and fission, and cytoskeleton remodeling in different cell-types. At the central nervous system, they regulate synaptic vesicle recycling and signaling-receptor turnover, allowing the maintenance of synaptic transmission. In the presynapses, these GTP-ases control the recycling of synaptic vesicles influencing the size of the ready-releasable pool and the release of neurotransmitters from nerve terminals, whereas in the postsynapses, they are involved in AMPA-receptor trafficking to and from postsynaptic densities, supporting excitatory synaptic plasticity, and consequently learning and memory formation. In agreement with these relevant roles, an important number of neurological disorders are associated with mutations and/or dysfunction of these GTP-ases. Along the present review we discuss the importance of DSPs at synapses and their implication in different neuropathological contexts.
Collapse
Affiliation(s)
- Jorge Arriagada-Diaz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Magister en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Prado-Vega
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Magister en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Ana M Cárdenas Díaz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Alvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| | - Arlek M Gonzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
28
|
Regulation of actin dynamics in dendritic spines: Nanostructure, molecular mobility, and signaling mechanisms. Mol Cell Neurosci 2020; 109:103564. [DOI: 10.1016/j.mcn.2020.103564] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/04/2020] [Indexed: 12/16/2022] Open
|
29
|
Ben Zablah Y, Merovitch N, Jia Z. The Role of ADF/Cofilin in Synaptic Physiology and Alzheimer's Disease. Front Cell Dev Biol 2020; 8:594998. [PMID: 33282872 PMCID: PMC7688896 DOI: 10.3389/fcell.2020.594998] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/23/2020] [Indexed: 12/21/2022] Open
Abstract
Actin-depolymerization factor (ADF)/cofilin, a family of actin-binding proteins, are critical for the regulation of actin reorganization in response to various signals. Accumulating evidence indicates that ADF/cofilin also play important roles in neuronal structure and function, including long-term potentiation and depression. These are the most extensively studied forms of long-lasting synaptic plasticity and are widely regarded as cellular mechanisms underlying learning and memory. ADF/cofilin regulate synaptic function through their effects on dendritic spines and the trafficking of glutamate receptors, the principal mediator of excitatory synaptic transmission in vertebrates. Regulation of ADF/cofilin involves various signaling pathways converging on LIM domain kinases and slingshot phosphatases, which phosphorylate/inactivate and dephosphorylate/activate ADF/cofilin, respectively. Actin-depolymerization factor/cofilin activity is also regulated by other actin-binding proteins, activity-dependent subcellular distribution and protein translation. Abnormalities in ADF/cofilin have been associated with several neurodegenerative disorders such as Alzheimer’s disease. Therefore, investigating the roles of ADF/cofilin in the brain is not only important for understanding the fundamental processes governing neuronal structure and function, but also may provide potential therapeutic strategies to treat brain disorders.
Collapse
Affiliation(s)
- Youssif Ben Zablah
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Neil Merovitch
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Zhengping Jia
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Gobbo F, Cattaneo A. Neuronal Activity at Synapse Resolution: Reporters and Effectors for Synaptic Neuroscience. Front Mol Neurosci 2020; 13:572312. [PMID: 33192296 PMCID: PMC7609880 DOI: 10.3389/fnmol.2020.572312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022] Open
Abstract
The development of methods for the activity-dependent tagging of neurons enabled a new way to tackle the problem of engram identification at the cellular level, giving rise to groundbreaking findings in the field of memory studies. However, the resolution of activity-dependent tagging remains limited to the whole-cell level. Notably, events taking place at the synapse level play a critical role in the establishment of new memories, and strong experimental evidence shows that learning and synaptic plasticity are tightly linked. Here, we provide a comprehensive review of the currently available techniques that enable to identify and track the neuronal activity with synaptic spatial resolution. We also present recent technologies that allow to selectively interfere with specific subsets of synapses. Lastly, we discuss how these technologies can be applied to the study of learning and memory.
Collapse
Affiliation(s)
- Francesco Gobbo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
31
|
Zhang H, Bramham CR. Arc/Arg3.1 function in long-term synaptic plasticity: Emerging mechanisms and unresolved issues. Eur J Neurosci 2020; 54:6696-6712. [PMID: 32888346 DOI: 10.1111/ejn.14958] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/18/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Arc (activity-regulated cytoskeleton-associated protein) is posited as a critical regulator of long-term synaptic plasticity at excitatory synapses, including long-term potentiation, long-term depression, inverse synaptic tagging and homoeostatic scaling, with pivotal roles in memory and postnatal cortical development. However, the mechanisms underlying the bidirectional regulation of synaptic strength are poorly understood. Here we review evidence from different plasticity paradigms, highlight outstanding issues and discuss stimulus-specific mechanisms that dictate Arc function. We propose a model in which Arc bidirectionally controls synaptic strength by coordinate regulation of AMPA-type glutamate receptor (AMPAR) trafficking and actin cytoskeletal dynamics in dendritic spines. Key to this model, Arc is proposed to function as an activity-dependent regulator of AMPAR lateral membrane diffusion and trapping at synapses.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
32
|
Hong Y, Liang X, Gilhus NE. AChR antibodies show a complex interaction with human skeletal muscle cells in a transcriptomic study. Sci Rep 2020; 10:11230. [PMID: 32641696 PMCID: PMC7343820 DOI: 10.1038/s41598-020-68185-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 06/17/2020] [Indexed: 01/26/2023] Open
Abstract
Acetylcholine receptor (AChR) antibodies are the most important pathogenic marker in patients with myasthenia gravis (MG). The antibodies bind to AChRs on the postsynaptic membrane, and this leads to receptor degradation, destruction, or functional blocking with impaired signal at the neuromuscular junction. In this study, we have explored the effects of AChR antibodies binding to mature human myotubes with agrin-induced AChR clusters and pathways relevant for AChR degradation using bulk RNA sequencing. Protein-coding RNAs and lncRNAs were examined by RNA sequencing analysis. AChR antibodies induced marked changes of the transcriptomic profiles, with over 400 genes differentially expressed. Cholesterol metabolic processes and extracellular matrix organization gene sets were influenced and represent AChR-trafficking related pathways. Muscle contraction and cellular homeostasis gene sets were also affected, and independently of AChR trafficking. Furthermore, we found changes in a protein-coding RNA and lncRNA network, where expression of lncRNA MEG3 correlated closely with protein-coding genes for cellular homeostasis. We conclude that AChR antibodies induce an active response in human skeletal muscle cells which affects key intra- and extracellular pathways.
Collapse
Affiliation(s)
- Yu Hong
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Xiao Liang
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Nils Erik Gilhus
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
33
|
Gavrilovici C, Jiang Y, Kiroski I, Teskey GC, Rho JM, Nguyen MD. Postnatal Role of the Cytoskeleton in Adult Epileptogenesis. Cereb Cortex Commun 2020; 1:tgaa024. [PMID: 32864616 PMCID: PMC7446231 DOI: 10.1093/texcom/tgaa024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Mutations in cytoskeletal proteins can cause early infantile and childhood epilepsies by misplacing newly born neurons and altering neuronal connectivity. In the adult epileptic brain, cytoskeletal disruption is often viewed as being secondary to aberrant neuronal activity and/or death, and hence simply represents an epiphenomenon. Here, we review the emerging evidence collected in animal models and human studies implicating the cytoskeleton as a potential causative factor in adult epileptogenesis. Based on the emerging evidence, we propose that cytoskeletal disruption may be an important pathogenic mechanism in the mature epileptic brain.
Collapse
Affiliation(s)
- Cezar Gavrilovici
- Departments of Neurosciences & Pediatrics, University of California San Diego, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
| | - Yulan Jiang
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - Ivana Kiroski
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - G Campbell Teskey
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| | - Jong M Rho
- Departments of Neurosciences & Pediatrics, University of California San Diego, Rady Children’s Hospital San Diego, San Diego, CA 92123, USA
| | - Minh Dang Nguyen
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children Hospital Research Institute, University of Calgary, Calgary T2N 4N1, Canada
| |
Collapse
|
34
|
Abstract
Regulation of neurotransmitter receptor content at synapses is achieved through a dynamic equilibrium between biogenesis and degradation pathways, receptor stabilization at synaptic sites, and receptor trafficking in and out synapses. In the past 20 years, the movements of receptors to and from synapses have emerged as a series of highly regulated processes that mediate postsynaptic plasticity. Our understanding of the properties and roles of receptor movements has benefited from technological advances in receptor labeling and tracking capacities, as well as from new methods to interfere with their movements. Focusing on two key glutamatergic receptors, we review here our latest understanding of the characteristics of receptor movements and their role in tuning the efficacy of synaptic transmission in health and brain disease.
Collapse
Affiliation(s)
- Laurent Groc
- Interdisciplinary Institute for NeuroScience, CNRS, UMR 5297, Centre Broca Nouvelle-Aquitaine, 146, rue Léo-Saignat, 33076 Bordeaux, France
- IINS, University of Bordeaux, UMR5297, Bordeaux, France
| | - Daniel Choquet
- Interdisciplinary Institute for NeuroScience, CNRS, UMR 5297, Centre Broca Nouvelle-Aquitaine, 146, rue Léo-Saignat, 33076 Bordeaux, France
- IINS, University of Bordeaux, UMR5297, Bordeaux, France
- Bordeaux Imaging Center, UMS 3420 CNRS, US4 INSERM, University of Bordeaux, Bordeaux, France
| |
Collapse
|
35
|
Eguchi K, Velicky P, Hollergschwandtner E, Itakura M, Fukazawa Y, Danzl JG, Shigemoto R. Advantages of Acute Brain Slices Prepared at Physiological Temperature in the Characterization of Synaptic Functions. Front Cell Neurosci 2020; 14:63. [PMID: 32265664 PMCID: PMC7096554 DOI: 10.3389/fncel.2020.00063] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/02/2020] [Indexed: 11/27/2022] Open
Abstract
Acute brain slice preparation is a powerful experimental model for investigating the characteristics of synaptic function in the brain. Although brain tissue is usually cut at ice-cold temperature (CT) to facilitate slicing and avoid neuronal damage, exposure to CT causes molecular and architectural changes of synapses. To address these issues, we investigated ultrastructural and electrophysiological features of synapses in mouse acute cerebellar slices prepared at ice-cold and physiological temperature (PT). In the slices prepared at CT, we found significant spine loss and reconstruction, synaptic vesicle rearrangement and decrease in synaptic proteins, all of which were not detected in slices prepared at PT. Consistent with these structural findings, slices prepared at PT showed higher release probability. Furthermore, preparation at PT allows electrophysiological recording immediately after slicing resulting in higher detectability of long-term depression (LTD) after motor learning compared with that at CT. These results indicate substantial advantages of the slice preparation at PT for investigating synaptic functions in different physiological conditions.
Collapse
Affiliation(s)
- Kohgaku Eguchi
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Philipp Velicky
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | | | - Makoto Itakura
- Department of Biochemistry, Kitasato University School of Medicine, Sagamihara-shi, Japan
| | - Yugo Fukazawa
- Department of Brain Structure and Function, Research Center for Child Mental Development, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Johann Georg Danzl
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| |
Collapse
|
36
|
Optogenetic Control of Spine-Head JNK Reveals a Role in Dendritic Spine Regression. eNeuro 2020; 7:ENEURO.0303-19.2019. [PMID: 31937523 PMCID: PMC7053173 DOI: 10.1523/eneuro.0303-19.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/27/2022] Open
Abstract
In this study, we use an optogenetic inhibitor of c-Jun NH2-terminal kinase (JNK) in dendritic spine sub-compartments of rat hippocampal neurons. We show that JNK inhibition exerts rapid (within seconds) reorganization of actin in the spine-head. Using real-time Förster resonance energy transfer (FRET) to measure JNK activity, we find that either excitotoxic insult (NMDA) or endocrine stress (corticosterone), activate spine-head JNK causing internalization of AMPARs and spine retraction. Both events are prevented upon optogenetic inhibition of JNK, and rescued by JNK inhibition even 2 h after insult. Moreover, we identify that the fast-acting anti-depressant ketamine reduces JNK activity in hippocampal neurons suggesting that JNK inhibition may be a downstream mediator of its anti-depressant effect. In conclusion, we show that JNK activation plays a role in triggering spine elimination by NMDA or corticosterone stress, whereas inhibition of JNK facilitates regrowth of spines even in the continued presence of glucocorticoid. This identifies that JNK acts locally in the spine-head to promote AMPAR internalization and spine shrinkage following stress, and reveals a protective function for JNK inhibition in preventing spine regression.
Collapse
|
37
|
Pinho J, Marcut C, Fonseca R. Actin remodeling, the synaptic tag and the maintenance of synaptic plasticity. IUBMB Life 2020; 72:577-589. [DOI: 10.1002/iub.2261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/06/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Júlia Pinho
- Cellular and Systems Neurobiology, Chronic Disease Research CenterNOVA Medical School Lisbon Portugal
| | - Cristina Marcut
- Cellular and Systems Neurobiology, Chronic Disease Research CenterNOVA Medical School Lisbon Portugal
| | - Rosalina Fonseca
- Cellular and Systems Neurobiology, Chronic Disease Research CenterNOVA Medical School Lisbon Portugal
| |
Collapse
|
38
|
Dendritic Spines in Alzheimer's Disease: How the Actin Cytoskeleton Contributes to Synaptic Failure. Int J Mol Sci 2020; 21:ijms21030908. [PMID: 32019166 PMCID: PMC7036943 DOI: 10.3390/ijms21030908] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/24/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by Aβ-driven synaptic dysfunction in the early phases of pathogenesis. In the synaptic context, the actin cytoskeleton is a crucial element to maintain the dendritic spine architecture and to orchestrate the spine’s morphology remodeling driven by synaptic activity. Indeed, spine shape and synaptic strength are strictly correlated and precisely governed during plasticity phenomena in order to convert short-term alterations of synaptic strength into long-lasting changes that are embedded in stable structural modification. These functional and structural modifications are considered the biological basis of learning and memory processes. In this review we discussed the existing evidence regarding the role of the spine actin cytoskeleton in AD synaptic failure. We revised the physiological function of the actin cytoskeleton in the spine shaping and the contribution of actin dynamics in the endocytosis mechanism. The internalization process is implicated in different aspects of AD since it controls both glutamate receptor membrane levels and amyloid generation. The detailed understanding of the mechanisms controlling the actin cytoskeleton in a unique biological context as the dendritic spine could pave the way to the development of innovative synapse-tailored therapeutic interventions and to the identification of novel biomarkers to monitor synaptic loss in AD.
Collapse
|
39
|
Fukuda T, Nagashima S, Inatome R, Yanagi S. CAMDI interacts with the human memory-associated protein KIBRA and regulates AMPAR cell surface expression and cognition. PLoS One 2019; 14:e0224967. [PMID: 31730661 PMCID: PMC6857912 DOI: 10.1371/journal.pone.0224967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 10/25/2019] [Indexed: 01/06/2023] Open
Abstract
Little is known about the molecular mechanisms of cognitive deficits in psychiatric disorders. CAMDI is a psychiatric disorder-related factor, the deficiency of which in mice results in delayed neuronal migration and psychiatrically abnormal behaviors. Here, we found that CAMDI-deficient mice exhibited impaired recognition memory and spatial reference memory. Knockdown of CAMDI in hippocampal neurons increased the amount of internalized alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor (AMPAR) and attenuated the chemical long-term potentiation (LTP)-dependent cell surface expression of AMPAR. KIBRA was identified as a novel CAMDI-binding protein that retains AMPAR in the cytosol after internalization. KIBRA inhibited CAMDI-dependent Rab11 activation, thereby attenuating AMPAR cell surface expression. These results suggest that CAMDI regulates AMPAR cell surface expression during LTP. CAMDI dysfunction may partly explain the mechanism underlying cognitive deficits in psychiatric diseases.
Collapse
Affiliation(s)
- Toshifumi Fukuda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- * E-mail: (SY); (TF)
| | - Shun Nagashima
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Ryoko Inatome
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- * E-mail: (SY); (TF)
| |
Collapse
|
40
|
Guerra MJ, González‐Jamett AM, Báez‐Matus X, Navarro‐Quezada N, Martínez AD, Neely A, Cárdenas AM. The Ca2+channel subunit CaVβ2a‐subunit down‐regulates voltage‐activated ion current densities by disrupting actin‐dependent traffic in chromaffin cells. J Neurochem 2019; 151:703-715. [DOI: 10.1111/jnc.14851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/01/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022]
Affiliation(s)
- María J. Guerra
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Arlek M. González‐Jamett
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Ximena Báez‐Matus
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Nieves Navarro‐Quezada
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Agustín D. Martínez
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Alan Neely
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| | - Ana M. Cárdenas
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia Universidad de Valparaíso Valparaíso Chile
| |
Collapse
|
41
|
Morini R, Ferrara S, Perrucci F, Zambetti S, Pelucchi S, Marcello E, Gardoni F, Antonucci F, Matteoli M, Menna E. Lack of the Actin Capping Protein, Eps8, Affects NMDA-Type Glutamate Receptor Function and Composition. Front Mol Neurosci 2018; 11:313. [PMID: 30233314 PMCID: PMC6133960 DOI: 10.3389/fnmol.2018.00313] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 08/15/2018] [Indexed: 11/26/2022] Open
Abstract
Actin-based remodeling underlines spine morphogenesis and plasticity and is crucially involved in the processes that constantly reshape the circuitry of the adult brain in response to external stimuli, leading to learning and memory formation and supporting cognitive functions. Hence spine morphology and synaptic strength are tightly linked and indeed abnormalities in spine number and morphology have been described in a number of neurological disorders such as autism spectrum disorders (ASDs), schizophrenia and intellectual disabilities. We have recently demonstrated that the actin regulating protein, Epidermal growth factor receptor pathway substrate 8 (Eps8), is essential for spine growth and long term potentiation. Indeed, mice lacking Eps8 display immature filopodia-like spines, which are unable to undergo potentiation, and are impaired in cognitive functions. Furthermore, reduced levels of Eps8 have been found in the brain of a cohort of patients affected by ASD compared to controls. Here we investigated whether the lack of Eps8, which is also part of the N-methyl-d-aspartate (NMDA) receptor complex, affects the functional maturation of the postsynaptic compartment. Our results demonstrate that Eps8 knock out mice (Eps8 KO) neurons display altered synaptic expression and subunit composition of NMDA receptors (i.e., increased GluN2B-, decreased GluN2A-containing receptors) and impaired GluN2B to GluN2A subunit shift. Indeed Eps8 KO neurons display increased content of GluN2B containing NMDA receptors both at the synaptic and extrasynaptic level. Furthermore, Eps8 KO neurons display an increased content of extra-synaptic GluN2B-containing receptors, suggesting that also the synaptic targeting of NMDA receptors is affected by the lack of Eps8. These data demonstrate that, besides regulation of spine morphogenesis, Eps8 also regulates the synaptic balance of NMDA receptors subunits GluN2A and GluN2B.
Collapse
Affiliation(s)
- Raffaella Morini
- Laboratory of Pharmacology and Brain Pathology, Neurocenter IRCCS Humanitas, Milan, Italy
| | - Silvia Ferrara
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università di Milano, Milan, Italy
| | - Fabio Perrucci
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Stefania Zambetti
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università di Milano, Milan, Italy
| | - Silvia Pelucchi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università di Milano, Milan, Italy.,NEUROFARBA, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Università degli Studi di Firenze, Florence, Italy
| | - Elena Marcello
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università di Milano, Milan, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università di Milano, Milan, Italy
| | - Flavia Antonucci
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università di Milano, Milan, Italy
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Neurocenter IRCCS Humanitas, Milan, Italy.,CNR-Istituto di Neuroscienze (IN), Milan, Italy
| | - Elisabetta Menna
- Laboratory of Pharmacology and Brain Pathology, Neurocenter IRCCS Humanitas, Milan, Italy.,CNR-Istituto di Neuroscienze (IN), Milan, Italy
| |
Collapse
|
42
|
Zhang B, Li Y, Liu JW, Liu XW, Wen W, Cui Y, Huang SM. Postsynaptic GluR2 Involved in Amelioration of Aβ-Induced Memory Dysfunction by KAIXIN-San Through Rescuing Hippocampal LTP in Mice. Rejuvenation Res 2018; 22:131-137. [PMID: 30009679 DOI: 10.1089/rej.2018.2080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Kaixin-San (KXS), a Chinese formula, was used to treat "amnesia," a senile dementia in the modern world. This formula was reported to improve behavioral performances in many animal models. This study was designed to explore how KXS has improved amyloid-β (Aβ)-induced memory dysfunction in mice. The mouse models were achieved through unilateral ventricle injection with Aβ42. The effects of KXS on memory improvement were evaluated by the step-down test. The electrophysiological changes induced by KXS were measured by long-term potentiation (LTP) analysis in the hippocampus in vivo. The expression of glutamate receptor 2 (GluR2) was observed through immunohistochemical staining. Behavioral experiment outcome demonstrated reduced avoidance time and increased error time during the step-down test in the mice of Aβ group. This memory impairment, however, was reversed by KXS. Electrophysiological experiment showed no significant difference between Aβ group and KXS group either in the size or the shape of field excitatory postsynaptic potentiation recorded from perforant path to dentate gyrus pathway. However, LTP in this region was reduced by Aβ and recovered by KXS administration. Moreover, immunohistochemical staining showed increased postsynaptic GluR2 expression in DG area in KXS group. These findings suggest that Aβ results in impairment to memory function of the animals, and KXS protects the animal from memory loss by rescuing LTP through postsynaptic mechanism which refers to increasing GluR2 expression.
Collapse
Affiliation(s)
- Bo Zhang
- 1 Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan Li
- 1 Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia-Wei Liu
- 2 Department of Organic Chemistry, School of Pharmacy, Mudanjiang Medical University, Mudanjiang, China
| | - Xue-Wei Liu
- 1 Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
- 3 Department of Neuropharmacology, College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Wei Wen
- 1 Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Cui
- 4 Department of Veterinary Medicine, Institute of tropical agriculture and forestry, Hainan University, Haikou, China
| | - Shu-Ming Huang
- 1 Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
43
|
Zhou ZW, Yang QD, Tang QP, Yang J, Guo RJ, Jiang W. Effect of willed movement training on neurorehabilitation after focal cerebral ischemia and on the neural plasticity-associated signaling pathway. Mol Med Rep 2017; 17:1173-1181. [PMID: 29115485 DOI: 10.3892/mmr.2017.7964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 09/12/2017] [Indexed: 11/06/2022] Open
Abstract
Neurorehabilitation training is a therapeutic intervention for the loss of neural function induced by focal cerebral ischemia, however, the effect varies depending on the neurorehabilitation exercises. Willed movement (WM) training is defined as task‑oriented training, which increases enthusiasm of patients to accomplish a specific task. The current study was performed to the evaluate effect of WM training on neurorehabilitation following focal cerebral ischemia, and further investigate the influence on neural plasticity‑associated signaling pathway. Sprague‑Dawley rats following temporary middle cerebral artery occlusion (tMCAO) were randomly divided into four groups: tMCAO (no rehabilitation training), CR (control rehabilitation), EM (environmental modification) and WM groups. Rats in the CR group were forced to exercise (running) in a rotating wheel. In the WM group, food was used to entice rats to climb on a herringbone ladder. Herringbone ladders were also put into the cages of the rats in the CR and EM groups, however without the food attraction. WM group exhibited an improvement in neurobehavioral performance compared with other groups. TTC staining indicated an evident reduction in brain damage in the WM group. There were increased synaptic junctions following WM training, based on the observations of transmission election microscopy. Investigation of the molecular mechanism suggested that WM training conferred the greatest effect on stimulating the extracellular signal‑related kinase (ERK)/cyclic adenosine monophosphate response element‑binding protein 1 (CREB) pathway and glutamate receptor 2 (GluR2)/glutamate receptor interacting protein 1‑associated protein 1 (GRASP‑1)/protein interacting with C‑kinase 1 (PICK1) cascades among groups. Collectively, the improvement of neurobehavioral performance by WM training following tMCAO is suggested to involve the ERK/CREB pathway and GluR2/GRASP‑1/PICK1 cascades. The present study provided a preliminary foundation for future research on the therapeutic effect of WM training against stroke‑induced neuron damage.
Collapse
Affiliation(s)
- Zhi-Wen Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qi-Dong Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qing-Ping Tang
- Department of Physiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Rong-Jing Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Wen Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
44
|
Mittal N, Minasyan A, Romaneschi N, Hakimian JK, Gonzalez-Fernandez G, Albert R, Desai N, Mendez IA, Schallert T, Ostlund SB, Walwyn W. Beta-arrestin 1 regulation of reward-motivated behaviors and glutamatergic function. PLoS One 2017; 12:e0185796. [PMID: 28973019 PMCID: PMC5626489 DOI: 10.1371/journal.pone.0185796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/19/2017] [Indexed: 01/10/2023] Open
Abstract
The two highly homologous non-visual arrestins, beta-arrestin 1 and 2, are ubiquitously expressed in the central nervous system, yet knowledge of their disparate roles is limited. While beta-arrestin 2 (βarr2) has been implicated in several aspects of reward-related learning and behavior, very little is known about the behavioral function of beta-arrestin 1 (βarr1). Using mice lacking βarr1, we focused on the role of this scaffolding and signal transduction protein in reward-motivated behaviors and in striatal glutamatergic function. We found that βarr1 KO mice were both slower in acquiring cocaine self-administration and in extinguishing this behavior. They also showed deficits in learning tasks supported by a natural food reward, suggesting a general alteration in reward processing. We then examined glutamatergic synaptic strength in WT and KO medium spiny neurons (MSNs) of the Nucleus Accumbens (NAc) shell in naïve animals, and from those that underwent cocaine self-administration. An increase in the AMPA/NMDA (A/N) ratio and a relative lack of GluN2B-enriched NMDARs was found in naïve KO vs WT MSNs. Applying Lim Domain Kinase (LIMK1), the kinase that phosphorylates and inactivates cofilin, to these cells, showed that both βarr1 and LIMK regulate the A/N ratio and GluN2B-NMDARs. Cocaine self-administration increased the A/N ratio and GluN2B-NMDARs in WT MSNs and, although the A/N ratio also increased in KO MSNs, this was accompanied by fewer GluN2B-NMDARs and an appearance of calcium-permeable AMPARs. Finally, to examine the consequences of reduced basal GluN2B-NMDARs in reward-processing seen in KO mice, we chronically infused ifenprodil, a GluN2B antagonist, into the NAc shell of WT mice. This intervention substantially reduced food-motivated behavior. Together these findings identify a previously unknown role of βarr1 in regulating specific reward-motivated behaviors and glutamatergic function.
Collapse
Affiliation(s)
- Nitish Mittal
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States of America
| | - Ani Minasyan
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Nicole Romaneschi
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Joshua K. Hakimian
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Gabriel Gonzalez-Fernandez
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Ralph Albert
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Nina Desai
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Ian A. Mendez
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Timothy Schallert
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States of America
| | - Sean B. Ostlund
- Department of Anesthesiology and Perioperative Care, School of Medicine, University of California, Irvine, UCI Center for Addiction Neuroscience, School of Biological Sciences, University of California Irvine, Irvine, United States of America
| | - Wendy Walwyn
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, CA, United States of America
- * E-mail:
| |
Collapse
|
45
|
Laguesse S, Morisot N, Shin JH, Liu F, Adrover MF, Sakhai SA, Lopez MF, Phamluong K, Griffin WC, Becker HC, Bender KJ, Alvarez VA, Ron D. Prosapip1-Dependent Synaptic Adaptations in the Nucleus Accumbens Drive Alcohol Intake, Seeking, and Reward. Neuron 2017; 96:145-159.e8. [PMID: 28890345 PMCID: PMC6014831 DOI: 10.1016/j.neuron.2017.08.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/13/2017] [Accepted: 08/24/2017] [Indexed: 12/15/2022]
Abstract
The mammalian target of rapamycin complex 1 (mTORC1), a transducer of local dendritic translation, participates in learning and memory processes as well as in mechanisms underlying alcohol-drinking behaviors. Using an unbiased RNA-seq approach, we identified Prosapip1 as a novel downstream target of mTORC1 whose translation and consequent synaptic protein expression are increased in the nucleus accumbens (NAc) of mice excessively consuming alcohol. We demonstrate that alcohol-dependent increases in Prosapip1 levels promote the formation of actin filaments, leading to changes in dendritic spine morphology of NAc medium spiny neurons (MSNs). We further demonstrate that Prosapip1 is required for alcohol-dependent synaptic localization of GluA2 lacking AMPA receptors in NAc shell MSNs. Finally, we present data implicating Prosapip1 in mechanisms underlying alcohol self-administration and reward. Together, these data suggest that Prosapip1 in the NAc is a molecular transducer of structural and synaptic alterations that drive and/or maintain excessive alcohol use.
Collapse
Affiliation(s)
- Sophie Laguesse
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Nadege Morisot
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Jung Hoon Shin
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute of Alcohol Abuse and Alcoholism, US National Institutes of Health, Bethesda, MD, USA
| | - Feng Liu
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Martin F Adrover
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute of Alcohol Abuse and Alcoholism, US National Institutes of Health, Bethesda, MD, USA
| | - Samuel A Sakhai
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Marcelo F Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California, San Francisco, CA, USA
| | - William C Griffin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA; Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA; RHJ Department of Veterans Affairs Medical Center, Charleston, SC, USA
| | - Kevin J Bender
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute of Alcohol Abuse and Alcoholism, US National Institutes of Health, Bethesda, MD, USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, CA, USA.
| |
Collapse
|
46
|
The non-coding RNA BC1 regulates experience-dependent structural plasticity and learning. Nat Commun 2017; 8:293. [PMID: 28819097 PMCID: PMC5561022 DOI: 10.1038/s41467-017-00311-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/19/2017] [Indexed: 11/09/2022] Open
Abstract
The brain cytoplasmic (BC1) RNA is a non-coding RNA (ncRNA) involved in neuronal translational control. Absence of BC1 is associated with altered glutamatergic transmission and maladaptive behavior. Here, we show that pyramidal neurons in the barrel cortex of BC1 knock out (KO) mice display larger excitatory postsynaptic currents and increased spontaneous activity in vivo. Furthermore, BC1 KO mice have enlarged spine heads and postsynaptic densities and increased synaptic levels of glutamate receptors and PSD-95. Of note, BC1 KO mice show aberrant structural plasticity in response to whisker deprivation, impaired texture novel object recognition and altered social behavior. Thus, our study highlights a role for BC1 RNA in experience-dependent plasticity and learning in the mammalian adult neocortex, and provides insight into the function of brain ncRNAs regulating synaptic transmission, plasticity and behavior, with potential relevance in the context of intellectual disabilities and psychiatric disorders. Brain cytoplasmic (BC1) RNA is a non-coding RNA that has been implicated in translational regulation, seizure, and anxiety. Here, the authors show that in the cortex, BC1 RNA is required for sensory deprivation-induced structural plasticity of dendritic spines, as well as for correct sensory learning and social behaviors.
Collapse
|
47
|
Huntingtin-associated protein-1 (HAP1) regulates endocytosis and interacts with multiple trafficking-related proteins. Cell Signal 2017; 35:176-187. [DOI: 10.1016/j.cellsig.2017.02.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/16/2017] [Accepted: 02/28/2017] [Indexed: 12/24/2022]
|
48
|
Cyclin Y-mediated transcript profiling reveals several important functional pathways regulated by Cyclin Y in hippocampal neurons. PLoS One 2017; 12:e0172547. [PMID: 28241067 PMCID: PMC5328252 DOI: 10.1371/journal.pone.0172547] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/05/2017] [Indexed: 12/20/2022] Open
Abstract
Cyclin Y (CCNY), which is a cyclin protein known to play a role in cell division, is unexpectedly and thus interestingly expressed in non-proliferating neuronal cells. There have been only a few studies reporting the neuronal functions of CCNY in synapse remodeling and hippocampal long-term potentiation. Therefore, we here provide global and comprehensive information on the putative functions of CCNY in biological and functional pathways in neuronal systems. We adopted high-throughput RNA-sequencing technology for analyzing transcriptomes regulated by CCNY and utilized bioinformatics for identifying putative molecules, biological processes, and functional pathways that are possibly connected to CCNY functions in hippocampal neuronal cells of rats. We revealed that several enriched annotation terms and pathways associated with CCNY expression within neurons, including apoptosis, learning or memory, synaptic plasticity, actin cytoskeleton, focal adhesion, extracellular matrix-receptor interaction and chemokine signaling pathway are targeted by CCNY. In addition, the mRNA levels of some genes enriched for those annotation terms and pathways or genes reported to be altered in Alzheimer's disease mouse model were further validated by quantitative real-time PCR in hippocampal neuronal cells. The present study provides an excellent resource for future investigations of CCNY functions in neuronal systems.
Collapse
|
49
|
Willmes CG, Mack TGA, Ledderose J, Schmitz D, Wozny C, Eickholt BJ. Investigation of hippocampal synaptic transmission and plasticity in mice deficient in the actin-binding protein Drebrin. Sci Rep 2017; 7:42652. [PMID: 28198431 PMCID: PMC5309812 DOI: 10.1038/srep42652] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/11/2017] [Indexed: 12/22/2022] Open
Abstract
The dynamic regulation of the actin cytoskeleton plays a key role in controlling the structure and function of synapses. It is vital for activity-dependent modulation of synaptic transmission and long-term changes in synaptic morphology associated with memory consolidation. Several regulators of actin dynamics at the synapse have been identified, of which a salient one is the postsynaptic actin stabilising protein Drebrin (DBN). It has been suggested that DBN modulates neurotransmission and changes in dendritic spine morphology associated with synaptic plasticity. Given that a decrease in DBN levels is correlated with cognitive deficits associated with ageing and dementia, it was hypothesised that DBN protein abundance instructs the integrity and function of synapses. We created a novel DBN deficient mouse line. Analysis of gross brain and neuronal morphology revealed no phenotype in the absence of DBN. Electrophysiological recordings in acute hippocampal slices and primary hippocampal neuronal cultures showed that basal synaptic transmission, and both long-term and homeostatic synaptic plasticity were unchanged, suggesting that loss of DBN is not sufficient in inducing synapse dysfunction. We propose that the overall lack of changes in synaptic function and plasticity in DBN deficient mice may indicate robust compensatory mechanisms that safeguard cytoskeleton dynamics at the synapse.
Collapse
Affiliation(s)
- Claudia G Willmes
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,NeuroCure - Cluster of Excellence, Charité - Universitätsmedizin Berlin, Germany
| | - Till G A Mack
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Julia Ledderose
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Dietmar Schmitz
- NeuroCure - Cluster of Excellence, Charité - Universitätsmedizin Berlin, Germany.,Neuroscience Research Center (NWFZ), Charité Universitätsmedizin, 10117 Berlin, Germany
| | - Christian Wozny
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Britta J Eickholt
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,NeuroCure - Cluster of Excellence, Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
50
|
Lunardi P, Sachser RM, Sierra RO, Pedraza LK, Medina C, de la Fuente V, Romano A, Quillfeldt JA, de Oliveira Alvares L. Effects of Hippocampal LIMK Inhibition on Memory Acquisition, Consolidation, Retrieval, Reconsolidation, and Extinction. Mol Neurobiol 2017; 55:958-967. [DOI: 10.1007/s12035-016-0361-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/28/2016] [Indexed: 02/06/2023]
|