1
|
Bartol TM, Ordyan M, Sejnowski TJ, Rangamani P, Kennedy MB. A spatial model of autophosphorylation of CaMKII predicts that the lifetime of phospho-CaMKII after induction of synaptic plasticity is greatly prolonged by CaM-trapping. Front Synaptic Neurosci 2025; 17:1547948. [PMID: 40255983 PMCID: PMC12006173 DOI: 10.3389/fnsyn.2025.1547948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Long-term potentiation (LTP) is a biochemical process that underlies learning in excitatory glutamatergic synapses in the Central Nervous System (CNS). A critical early driver of LTP is autophosphorylation of the abundant postsynaptic enzyme, Ca2+/calmodulin-dependent protein kinase II (CaMKII). Autophosphorylation is initiated by Ca2+ flowing through NMDA receptors activated by strong synaptic activity. Its lifetime is ultimately determined by the balance of the rates of autophosphorylation and of dephosphorylation by protein phosphatase 1 (PP1). Here we have modeled the autophosphorylation and dephosphorylation of CaMKII during synaptic activity in a spine synapse using MCell4, an open source computer program for creating particle-based stochastic, and spatially realistic models of cellular microchemistry. The model integrates four earlier detailed models of separate aspects of regulation of spine Ca2+ and CaMKII activity, each of which incorporate experimentally measured biochemical parameters and have been validated against experimental data. We validate the composite model by showing that it accurately predicts previous experimental measurements of effects of NMDA receptor activation, including high sensitivity of induction of LTP to phosphatase activity in vivo, and persistence of autophosphorylation for a period of minutes after the end of synaptic stimulation. We then use the model to probe aspects of the mechanism of regulation of autophosphorylation of CaMKII that are difficult to measure in vivo. We examine the effects of "CaM-trapping," a process in which the affinity for Ca2+/CaM increases several hundred-fold after autophosphorylation. We find that CaM-trapping does not increase the proportion of autophosphorylated subunits in holoenzymes after a complex stimulus, as previously hypothesized. Instead, CaM-trapping may dramatically prolong the lifetime of autophosphorylated CaMKII through steric hindrance of dephosphorylation by protein phosphatase 1. The results provide motivation for experimental measurement of the extent of suppression of dephosphorylation of CaMKII by bound Ca2+/CaM. The composite MCell4 model of biochemical effects of complex stimuli in synaptic spines is a powerful new tool for realistic, detailed dissection of mechanisms of synaptic plasticity.
Collapse
Affiliation(s)
- Thomas M. Bartol
- The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Mariam Ordyan
- The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Terrence J. Sejnowski
- The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Neurobiology, University of California, San Diego, La Jolla, CA, United States
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Mary B. Kennedy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
2
|
Moldwin T, Azran LS, Segev I. A generalized mathematical framework for the calcium control hypothesis describes weight-dependent synaptic plasticity. J Comput Neurosci 2025:10.1007/s10827-025-00894-6. [PMID: 40100329 DOI: 10.1007/s10827-025-00894-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 03/20/2025]
Abstract
The brain modifies synaptic strengths to store new information via long-term potentiation (LTP) and long-term depression (LTD). Evidence has mounted that long-term synaptic plasticity is controlled via concentrations of calcium ([Ca2+]) in postsynaptic dendritic spines. Several mathematical models describe this phenomenon, including those of Shouval, Bear, and Cooper (SBC) (Shouval et al., 2002, 2010) and Graupner and Brunel (GB) (Graupner & Brunel, 2012). Here we suggest a generalized version of the SBC and GB models, the fixed point - learning rate (FPLR) framework, where the synaptic [Ca2+] specifies a fixed point toward which the synaptic weight approaches asymptotically at a [Ca2+]-dependent rate. The FPLR framework offers a straightforward phenomenological interpretation of calcium-based plasticity: the calcium concentration tells the synaptic weight where it is going and how quickly it goes there. The FPLR framework can flexibly incorporate various experimental findings, including the existence of multiple regions of [Ca2+] where no plasticity occurs, or plasticity observed experimentally in cerebellar Purkinje cells, where the directionality of calcium-based synaptic changes is reversed relative to cortical and hippocampal neurons. We also suggest a modeling approach that captures the dependency of late-phase plasticity stabilization on protein synthesis. We demonstrate that due to the asymptotic nature of synaptic changes in the FPLR rule, the plastic changes induced by frequency- and spike-timing-dependent plasticity protocols are weight-dependent. Finally, we show how the FPLR framework can explain the weight-dependence observed in behavioral time scale plasticity (BTSP).
Collapse
Affiliation(s)
- Toviah Moldwin
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Li Shay Azran
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Idan Segev
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
3
|
Good MA, Bannerman DM. Hippocampal Synaptic Plasticity: Integrating Memory and Anxiety Impairments in the Early Stages of Alzheimer's Disease. Curr Top Behav Neurosci 2025; 69:27-48. [PMID: 39747797 DOI: 10.1007/7854_2024_565] [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] [Indexed: 01/04/2025]
Abstract
A decline in hippocampal function has long been associated with the progression of cognitive impairments in patients with Alzheimer's disease (AD). The disruption of hippocampal synaptic plasticity [primarily the reduction of long-term potentiation LTP] by excess production of soluble beta-amyloid (Aβ) has long been accepted as the mechanism by which AD pathology impairs memory, at least during the early stages of AD pathogenesis. However, the premise that hippocampal LTP underpins the formation of associative, long-term memories has been challenged. Here, we consider evidence that this canonical view of LTP needs to be refined. Similarly, the view that the hippocampus simply supports memory ignores the wealth of data showing that the hippocampus is functionally heterogeneous along its septo-temporal axis. The ventral (but not the dorsal) hippocampus plays a major role in modulating emotional reactions to conflict. Here, we suggest that hippocampal LTP is not involved in forming long-term associative memories, but instead contributes to the disambiguation of overlapping memories in situations of conflict and associative interference. This conceptualisation of hippocampal synaptic plasticity may help explain how early-stage AD pathology may impact both memory and anxiety.
Collapse
Affiliation(s)
- Mark A Good
- School of Psychology, Cardiff University, Park Place, Cardiff, UK.
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Bartol TM, Ordyan M, Sejnowski TJ, Rangamani P, Kennedy MB. A spatial model of autophosphorylation of Ca 2+/calmodulin-dependent protein kinase II (CaMKII) predicts that the lifetime of phospho-CaMKII after induction of synaptic plasticity is greatly prolonged by CaM-trapping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578696. [PMID: 38352446 PMCID: PMC10862815 DOI: 10.1101/2024.02.02.578696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Long-term potentiation (LTP) is a biochemical process that underlies learning in excitatory glutamatergic synapses in the Central Nervous System (CNS). The critical early driver of LTP is autophosphorylation of the abundant postsynaptic enzyme, Ca2+/calmodulin-dependent protein kinase II (CaMKII). Autophosphorylation is initiated by Ca2+ flowing through NMDA receptors activated by strong synaptic activity. Its lifetime is ultimately determined by the balance of the rates of autophosphorylation and of dephosphorylation by protein phosphatase 1 (PP1). Here we have modeled the autophosphorylation and dephosphorylation of CaMKII during synaptic activity in a spine synapse using MCell4, an open source computer program for creating particle-based stochastic, and spatially realistic models of cellular microchemistry. The model integrates four earlier detailed models of separate aspects of regulation of spine Ca2+ and CaMKII activity, each of which incorporate experimentally measured biochemical parameters and have been validated against experimental data. We validate the composite model by showing that it accurately predicts previous experimental measurements of effects of NMDA receptor activation, including high sensitivity of induction of LTP to phosphatase activity in vivo, and persistence of autophosphorylation for a period of minutes after the end of synaptic stimulation. We then use the model to probe aspects of the mechanism of regulation of autophosphorylation of CaMKII that are difficult to measure in vivo. We examine the effects of "CaM-trapping," a process in which the affinity for Ca2+/CaM increases several hundred-fold after autophosphorylation. We find that CaM-trapping does not increase the proportion of autophosphorylated subunits in holoenzymes after a complex stimulus, as previously hypothesized. Instead, CaM-trapping may dramatically prolong the lifetime of autophosphorylated CaMKII through steric hindrance of dephosphorylation by protein phosphatase 1. The results provide motivation for experimental measurement of the extent of suppression of dephosphorylation of CaMKII by bound Ca2+/CaM. The composite MCell4 model of biochemical effects of complex stimuli in synaptic spines is a powerful new tool for realistic, detailed dissection of mechanisms of synaptic plasticity.
Collapse
Affiliation(s)
| | - Mariam Ordyan
- The Salk Institute for Biological Studies, La Jolla, CA
| | - Terrence J Sejnowski
- The Salk Institute for Biological Studies, La Jolla, CA
- Department of Neurobiology, University of California at San Diego, La Jolla, CA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA
| | - Mary B Kennedy
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| |
Collapse
|
5
|
Saccenti D, Lauro LJR, Crespi SA, Moro AS, Vergallito A, Grgič RG, Pretti N, Lamanna J, Ferro M. Boosting Psychotherapy With Noninvasive Brain Stimulation: The Whys and Wherefores of Modulating Neural Plasticity to Promote Therapeutic Change. Neural Plast 2024; 2024:7853199. [PMID: 39723244 PMCID: PMC11669434 DOI: 10.1155/np/7853199] [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: 05/03/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024] Open
Abstract
The phenomenon of neural plasticity pertains to the intrinsic capacity of neurons to undergo structural and functional reconfiguration through learning and experiential interaction with the environment. These changes could manifest themselves not only as a consequence of various life experiences but also following therapeutic interventions, including the application of noninvasive brain stimulation (NIBS) and psychotherapy. As standalone therapies, both NIBS and psychotherapy have demonstrated their efficacy in the amelioration of psychiatric disorders' symptoms, with a certain variability in terms of effect sizes and duration. Consequently, scholars suggested the convenience of integrating the two interventions into a multimodal treatment to boost and prolong the therapeutic outcomes. Such an approach is still in its infancy, and the physiological underpinnings substantiating the effectiveness and utility of combined interventions are still to be clarified. Therefore, this opinion paper aims to provide a theoretical framework consisting of compelling arguments as to why adding NIBS to psychotherapy can promote therapeutic change. Namely, we will discuss the physiological effects of the two interventions, thus providing a rationale to explain the potential advantages of a combined approach.
Collapse
Affiliation(s)
- Daniele Saccenti
- Department of Psychology, Sigmund Freud University, Milan, Italy
| | - Leonor J. Romero Lauro
- Department of Psychology and NeuroMi, University of Milano-Bicocca, Milan, Italy
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
| | - Sofia A. Crespi
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea S. Moro
- Department of Psychology, Sigmund Freud University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | | | | | - Novella Pretti
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
- Clinical Psychology Center, Division of Neurology, Galliera Hospital, Genoa, Italy
| | - Jacopo Lamanna
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | - Mattia Ferro
- Department of Psychology, Sigmund Freud University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
6
|
Dittmer PJ, Dell’Acqua ML. L-type Ca 2+ channel activation of STIM1-Orai1 signaling remodels the dendritic spine ER to maintain long-term structural plasticity. Proc Natl Acad Sci U S A 2024; 121:e2407324121. [PMID: 39178228 PMCID: PMC11363309 DOI: 10.1073/pnas.2407324121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/10/2024] [Indexed: 08/25/2024] Open
Abstract
Learning and memory require coordinated structural and functional plasticity at neuronal glutamatergic synapses located on dendritic spines. Here, we investigated how the endoplasmic reticulum (ER) controls postsynaptic Ca2+ signaling and long-term potentiation of dendritic spine size, i.e., sLTP that accompanies functional strengthening of glutamatergic synaptic transmission. In most ER-containing (ER+) spines, high-frequency optical glutamate uncaging (HFGU) induced long-lasting sLTP that was accompanied by a persistent increase in spine ER content downstream of a signaling cascade engaged by N-methyl-D-aspartate receptors (NMDARs), L-type Ca2+ channels (LTCCs), and Orai1 channels, the latter being activated by stromal interaction molecule 1 (STIM1) in response to ER Ca2+ release. In contrast, HFGU stimulation of ER-lacking (ER-) spines expressed only transient sLTP and exhibited weaker Ca2+ signals noticeably lacking Orai1 and ER contributions. Consistent with spine ER regulating structural metaplasticity, delivery of a second stimulus to ER- spines induced ER recruitment along with persistent sLTP, whereas ER+ spines showed no additional increases in size or ER content in response to sequential stimulation. Surprisingly, the physical interaction between STIM1 and Orai1 induced by ER Ca2+ release, but not the resulting Ca2+ entry through Orai1 channels, proved necessary for the persistent increases in both spine size and ER content required for expression of long-lasting late sLTP.
Collapse
Affiliation(s)
- Philip J. Dittmer
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Mark L. Dell’Acqua
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| |
Collapse
|
7
|
Tian C, Qi Y, Zheng Y, Xia P, Liu Q, Luan M, Zheng J, Song R, Wang M, Qi D, Xiong C, Dong L. Exploring the Effect of Arsenic-Containing Hydrocarbon on the Bidirectional Synaptic Plasticity of the Dorsal Hippocampus. Int J Mol Sci 2024; 25:7223. [PMID: 39000331 PMCID: PMC11241539 DOI: 10.3390/ijms25137223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Arsenic-containing hydrocarbons (AsHCs) are common in marine organisms. However, there is little research on their effects on the central nervous system's advanced activities, such as cognition. Bidirectional synaptic plasticity dynamically regulates cognition through the balance of long-term potentiation (LTP) and long-term depression (LTD). However, the effects of AsHCs on bidirectional synaptic plasticity and the underlying molecular mechanisms remain unexplored. This study provides the first evidence that 15 μg As L-1 AsHC 360 enhances bidirectional synaptic plasticity, occurring during the maintenance phase rather than the baseline phase. Further calcium gradient experiments hypothesize that AsHC 360 may enhance bidirectional synaptic plasticity by affecting calcium ion levels. The enhancement of bidirectional synaptic plasticity by 15 μg As L-1 AsHC 360 holds significant implications in improving cognitive function, treating neuro-psychiatric disorders, promoting neural recovery, and enhancing brain adaptability.
Collapse
Affiliation(s)
- Chunxiao Tian
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
- School of Biomedical Engineering, Tianjin Medical University, Tianjin 300070, China
| | - Yenan Qi
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
- School of Electronics & Information Engineering, Tiangong University, Tianjin 300387, China
| | - Yu Zheng
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
- School of Electronics & Information Engineering, Tiangong University, Tianjin 300387, China
- School of Control Science and Engineering, Tiangong University, Tianjin 300387, China;
| | - Pei Xia
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310012, China;
| | - Qiwen Liu
- School of Control Science and Engineering, Tiangong University, Tianjin 300387, China;
| | - Mengying Luan
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
| | - Junyao Zheng
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
| | - Rujuan Song
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
| | - Meng Wang
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
| | - Dejiao Qi
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
| | - Chan Xiong
- Analytical Chemistry, Institute of Chemistry, University of Graz, 8010 Graz, Austria
- BOKU Core Facility Mass Spectrometry, University of Natural Resources and Life Sciences (BOKU), 1190 Vienna, Austria
| | - Lei Dong
- School of Life Sciences, Tiangong University, Tianjin 300387, China; (C.T.); (Y.Q.); (Y.Z.); (M.L.); (J.Z.); (R.S.); (M.W.); (D.Q.)
- School of Electronics & Information Engineering, Tiangong University, Tianjin 300387, China
| |
Collapse
|
8
|
Prinkey K, Thompson E, Saikia J, Cid T, Dore K. Fluorescence lifetime imaging of AMPA receptor endocytosis in living neurons: effects of Aβ and PP1. Front Mol Neurosci 2024; 17:1409401. [PMID: 38915938 PMCID: PMC11194458 DOI: 10.3389/fnmol.2024.1409401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/22/2024] [Indexed: 06/26/2024] Open
Abstract
The relative amount of AMPA receptors expressed at the surface of neurons can be measured using superecliptic pHluorin (SEP) labeling at their N-terminus. However, the high signal variability resulting from protein overexpression in neurons and the low signal observed in intracellular vesicles make quantitative characterization of receptor trafficking difficult. Here, we establish a real-time live-cell assay of AMPAR trafficking based on fluorescence lifetime imaging (FLIM), which allows for simultaneous visualization of both surface and intracellular receptors. Using this assay, we found that elevating amyloid-beta (Aβ) levels leads to a strong increase in intracellular GluA1 and GluA2-containing receptors, indicating that Aβ triggers the endocytosis of these AMPARs. In APP/PS1 Alzheimer's disease model mouse neurons, FLIM revealed strikingly different AMPAR trafficking properties for GluA1- and GluA3-containing receptors, suggesting that chronic Aβ exposure triggered the loss of both surface and intracellular GluA3-containing receptors. Interestingly, overexpression of protein phosphatase 1 (PP1) also resulted in GluA1 endocytosis as well as depressed synaptic transmission, confirming the important role of phosphorylation in regulating AMPAR trafficking. This new approach allows for the quantitative measurement of extracellular pH, small changes in receptor trafficking, as well as simultaneous measurement of surface and internalized AMPARs in living neurons, and could therefore be applied to several different studies in the future.
Collapse
Affiliation(s)
| | | | | | | | - Kim Dore
- Center for Neural Circuits and Behavior, Department of Neuroscience, School of Medicine, University of California at San Diego, La Jolla, CA, United States
| |
Collapse
|
9
|
Carles A, Freyssin A, Perin-Dureau F, Rubinstenn G, Maurice T. Targeting N-Methyl-d-Aspartate Receptors in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:3733. [PMID: 38612544 PMCID: PMC11011887 DOI: 10.3390/ijms25073733] [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: 02/09/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
N-methyl-d-aspartate receptors (NMDARs) are the main class of ionotropic receptors for the excitatory neurotransmitter glutamate. They play a crucial role in the permeability of Ca2+ ions and excitatory neurotransmission in the brain. Being heteromeric receptors, they are composed of several subunits, including two obligatory GluN1 subunits (eight splice variants) and regulatory GluN2 (GluN2A~D) or GluN3 (GluN3A~B) subunits. Widely distributed in the brain, they regulate other neurotransmission systems and are therefore involved in essential functions such as synaptic transmission, learning and memory, plasticity, and excitotoxicity. The present review will detail the structure, composition, and localization of NMDARs, their role and regulation at the glutamatergic synapse, and their impact on cognitive processes and in neurodegenerative diseases (Alzheimer's, Huntington's, and Parkinson's disease). The pharmacology of different NMDAR antagonists and their therapeutic potentialities will be presented. In particular, a focus will be given on fluoroethylnormemantine (FENM), an investigational drug with very promising development as a neuroprotective agent in Alzheimer's disease, in complement to its reported efficacy as a tomography radiotracer for NMDARs and an anxiolytic drug in post-traumatic stress disorder.
Collapse
Affiliation(s)
- Allison Carles
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France; (A.C.); (A.F.)
| | - Aline Freyssin
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France; (A.C.); (A.F.)
- ReST Therapeutics, 34095 Montpellier, France; (F.P.-D.); (G.R.)
| | | | | | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France; (A.C.); (A.F.)
| |
Collapse
|
10
|
Kraner SD, Sompol P, Prateeptrang S, Promkan M, Hongthong S, Thongsopha N, Nelson PT, Norris CM. Development of a monoclonal antibody specific for a calpain-generated ∆48 kDa calcineurin fragment, a marker of distressed astrocytes. J Neurosci Methods 2024; 402:110012. [PMID: 37984591 PMCID: PMC10841921 DOI: 10.1016/j.jneumeth.2023.110012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/23/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Calcineurin (CN) is a Ca2+/calmodulin-dependent protein phosphatase. In healthy tissue, CN exists mainly as a full-length (∼60 kDa) highly-regulated protein phosphatase involved in essential cellular functions. However, in diseased or injured tissue, CN is proteolytically converted to a constitutively active fragment that has been causatively-linked to numerous pathophysiologic processes. These calpain-cleaved CN fragments (∆CN) appear at high levels in human brain at early stages of cognitive decline associated with Alzheimer's disease (AD). NEW METHOD We developed a monoclonal antibody to ∆CN, using an immunizing peptide corresponding to the C-terminal end of the ∆CN fragment. RESULTS We obtained a mouse monoclonal antibody, designated 26A6, that selectively detects ∆CN in Western analysis of calpain-cleaved recombinant human CN. Using this antibody, we screened both pathological and normal human brain sections provided by the University of Kentucky's Alzheimer's Disease Research Center. 26A6 showed low reactivity towards normal brain tissue, but detected astrocytes both surrounding AD amyloid plaques and throughout AD brain tissue. In brain tissue with infarcts, there was considerable concentration of 26A6-positive astrocytes within/around infarcts, suggesting a link with anoxic/ischemia pathways. COMPARISON WITH EXISTING METHOD The results obtained with the new monoclonal are similar to those obtained with a polyclonal we had previously developed. However, the monoclonal is an abundant tool available to the dementia research community. CONCLUSIONS The new monoclonal 26A6 antibody is highly selective for the ∆CN proteolytic fragment and labels a subset of astrocytes, and could be a useful tool for marking insidious brain pathology and identifying novel astrocyte phenotypes.
Collapse
Affiliation(s)
| | - Pradoldej Sompol
- Sanders Brown Center on Aging, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Siriyagon Prateeptrang
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Moltira Promkan
- Sanders Brown Center on Aging, USA; Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Suthida Hongthong
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Napasorn Thongsopha
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Peter T Nelson
- Sanders Brown Center on Aging, USA; Department of Pathology, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher M Norris
- Sanders Brown Center on Aging, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
11
|
Nicoll RA, Schulman H. Synaptic memory and CaMKII. Physiol Rev 2023; 103:2877-2925. [PMID: 37290118 PMCID: PMC10642921 DOI: 10.1152/physrev.00034.2022] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 06/10/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and long-term potentiation (LTP) were discovered within a decade of each other and have been inextricably intertwined ever since. However, like many marriages, it has had its up and downs. Based on the unique biochemical properties of CaMKII, it was proposed as a memory molecule before any physiological linkage was made to LTP. However, as reviewed here, the convincing linkage of CaMKII to synaptic physiology and behavior took many decades. New technologies were critical in this journey, including in vitro brain slices, mouse genetics, single-cell molecular genetics, pharmacological reagents, protein structure, and two-photon microscopy, as were new investigators attracted by the exciting challenge. This review tracks this journey and assesses the state of this marriage 40 years on. The collective literature impels us to propose a relatively simple model for synaptic memory involving the following steps that drive the process: 1) Ca2+ entry through N-methyl-d-aspartate (NMDA) receptors activates CaMKII. 2) CaMKII undergoes autophosphorylation resulting in constitutive, Ca2+-independent activity and exposure of a binding site for the NMDA receptor subunit GluN2B. 3) Active CaMKII translocates to the postsynaptic density (PSD) and binds to the cytoplasmic C-tail of GluN2B. 4) The CaMKII-GluN2B complex initiates a structural rearrangement of the PSD that may involve liquid-liquid phase separation. 5) This rearrangement involves the PSD-95 scaffolding protein, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), and their transmembrane AMPAR-regulatory protein (TARP) auxiliary subunits, resulting in an accumulation of AMPARs in the PSD that underlies synaptic potentiation. 6) The stability of the modified PSD is maintained by the stability of the CaMKII-GluN2B complex. 7) By a process of subunit exchange or interholoenzyme phosphorylation CaMKII maintains synaptic potentiation in the face of CaMKII protein turnover. There are many other important proteins that participate in enlargement of the synaptic spine or modulation of the steps that drive and maintain the potentiation. In this review we critically discuss the data underlying each of the steps. As will become clear, some of these steps are more firmly grounded than others, and we provide suggestions as to how the evidence supporting these steps can be strengthened or, based on the new data, be replaced. Although the journey has been a long one, the prospect of having a detailed cellular and molecular understanding of learning and memory is at hand.
Collapse
Affiliation(s)
- Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California, United States
| | - Howard Schulman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, United States
- Panorama Research Institute, Sunnyvale, California, United States
| |
Collapse
|
12
|
Moldwin T, Kalmenson M, Segev I. Asymmetric Voltage Attenuation in Dendrites Can Enable Hierarchical Heterosynaptic Plasticity. eNeuro 2023; 10:ENEURO.0014-23.2023. [PMID: 37414554 PMCID: PMC10354808 DOI: 10.1523/eneuro.0014-23.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 05/16/2023] [Accepted: 06/14/2023] [Indexed: 07/08/2023] Open
Abstract
Long-term synaptic plasticity is mediated via cytosolic calcium concentrations ([Ca2+]). Using a synaptic model that implements calcium-based long-term plasticity via two sources of Ca2+ - NMDA receptors and voltage-gated calcium channels (VGCCs) - we show in dendritic cable simulations that the interplay between these two calcium sources can result in a diverse array of heterosynaptic effects. When spatially clustered synaptic input produces a local NMDA spike, the resulting dendritic depolarization can activate VGCCs at nonactivated spines, resulting in heterosynaptic plasticity. NMDA spike activation at a given dendritic location will tend to depolarize dendritic regions that are located distally to the input site more than dendritic sites that are proximal to it. This asymmetry can produce a hierarchical effect in branching dendrites, where an NMDA spike at a proximal branch can induce heterosynaptic plasticity primarily at branches that are distal to it. We also explored how simultaneously activated synaptic clusters located at different dendritic locations synergistically affect the plasticity at the active synapses, as well as the heterosynaptic plasticity of an inactive synapse "sandwiched" between them. We conclude that the inherent electrical asymmetry of dendritic trees enables sophisticated schemes for spatially targeted supervision of heterosynaptic plasticity.
Collapse
Affiliation(s)
| | - Menachem Kalmenson
- Department of Neurobiology, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| | - Idan Segev
- Edmond and Lily Safra Center for Brain Sciences
- Department of Neurobiology, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| |
Collapse
|
13
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
14
|
Synaptic plasticity in Schizophrenia pathophysiology. IBRO Neurosci Rep 2023. [DOI: 10.1016/j.ibneur.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
15
|
Cornelius J, Haak S, Rothkegel M, Korte M, Michaelsen-Preusse K. Phosphorylation of the actin-binding protein profilin2a at S137 modulates bidirectional structural plasticity at dendritic spines. Front Cell Dev Biol 2023; 11:1107380. [PMID: 36875774 PMCID: PMC9975505 DOI: 10.3389/fcell.2023.1107380] [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: 11/24/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Background: Synaptic plasticity requires constant adaptation of functional and structural features at individual synaptic connections. Rapid re-modulation of the synaptic actin cytoskeleton provides the scaffold orchestrating both morphological and functional modifications. A major regulator of actin polymerization not only in neurons but also in various other cell types is the actin-binding protein profilin. While profilin is known to mediate the ADP to ATP exchange at actin monomers through its direct interaction with G-actin, it additionally is able to influence actin dynamics by binding to membrane-bound phospholipids as phosphatidylinositol (4,5)-bisphosphate (PIP2) as well as several other proteins containing poly-L-proline motifs including actin modulators like Ena/VASP, WAVE/WASP or formins. Notably, these interactions are proposed to be mediated by a fine-tuned regulation of post-translational phosphorylation of profilin. However, while phosphorylation sites of the ubiquitously expressed isoform profilin1 have been described and analyzed previously, there is still only little known about the phosphorylation of the profilin2a isoform predominantly expressed in neurons. Methods: Here, utilizing a knock-down/knock-in approach, we replaced endogenously expressed profilin2a by (de)phospho-mutants of S137 known to alter actin-, PIP2 and PLP-binding properties of profilin2a and analyzed their effect on general actin dynamics as well as activity-dependent structural plasticity. Results and Discussion: Our findings suggest that a precisely timed regulation of profilin2a phosphorylation at S137 is needed to mediate actin dynamics and structural plasticity bidirectionally during long-term potentiation and long-term depression, respectively.
Collapse
Affiliation(s)
- Jonas Cornelius
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Stefan Haak
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Martin Rothkegel
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany.,Helmholtz Centre for Infection Research, Research group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| | | |
Collapse
|
16
|
Hu Z, Ondrejcak T, Yu P, Zhang Y, Yang Y, Klyubin I, Kennelly SP, Rowan MJ, Hu NW. Do tau-synaptic long-term depression interactions in the hippocampus play a pivotal role in the progression of Alzheimer's disease? Neural Regen Res 2022; 18:1213-1219. [PMID: 36453396 PMCID: PMC9838152 DOI: 10.4103/1673-5374.360166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Cognitive decline in Alzheimer's disease correlates with the extent of tau pathology, in particular tau hyperphosphorylation that initially appears in the transentorhinal and related regions of the brain including the hippocampus. Recent evidence indicates that tau hyperphosphorylation caused by either amyloid-β or long-term depression, a form of synaptic weakening involved in learning and memory, share similar mechanisms. Studies from our group and others demonstrate that long-term depression-inducing low-frequency stimulation triggers tau phosphorylation at different residues in the hippocampus under different experimental conditions including aging. Conversely, certain forms of long-term depression at hippocampal glutamatergic synapses require endogenous tau, in particular, phosphorylation at residue Ser396. Elucidating the exact mechanisms of interaction between tau and long-term depression may help our understanding of the physiological and pathological functions of tau/tau (hyper)phosphorylation. We first summarize experimental evidence regarding tau-long-term depression interactions, followed by a discussion of possible mechanisms by which this interplay may influence the pathogenesis of Alzheimer's disease. Finally, we conclude with some thoughts and perspectives on future research about these interactions.
Collapse
Affiliation(s)
- Zhengtao Hu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China,Department of Gerontology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Tomas Ondrejcak
- Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Pengpeng Yu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yangyang Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yin Yang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China,Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Igor Klyubin
- Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Sean P. Kennelly
- Department of Age-Related Healthcare, Tallaght University Hospital, Dublin, Ireland,Department of Medical Gerontology, Trinity College, Dublin, Ireland
| | - Michael J. Rowan
- Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Neng-Wei Hu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China,Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland,Correspondence to: Neng-Wei Hu, .
| |
Collapse
|
17
|
Zhang K, Liao P, Wen J, Hu Z. Synaptic plasticity in schizophrenia pathophysiology. IBRO Neurosci Rep 2022; 13:478-487. [PMID: 36590092 PMCID: PMC9795311 DOI: 10.1016/j.ibneur.2022.10.008] [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: 05/22/2022] [Accepted: 10/21/2022] [Indexed: 11/05/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric syndrome with psychotic behavioral abnormalities and marked cognitive deficits. It is widely accepted that genetic and environmental factors contribute to the onset of schizophrenia. However, the etiology and pathology of the disease remain largely unexplored. Recently, the synaptopathology and the dysregulated synaptic plasticity and function have emerging as intriguing and prominent biological mechanisms of schizophrenia pathogenesis. Synaptic plasticity is the ability of neurons to change the strength of their connections in response to internal or external stimuli, which is essential for brain development and function, learning and memory, and vast majority of behavior responses relevant to psychiatric diseases including schizophrenia. Here, we reviewed molecular and cellular mechanisms of the multiple forms synaptic plasticity, and the functional regulations of schizophrenia-risk factors including disease susceptible genes and environmental alterations on synaptic plasticity and animal behavior. Recent genome-wide association studies have provided fruitful findings of hundreds of risk gene variances associated with schizophrenia, thus further clarifying the role of these disease-risk genes in synaptic transmission and plasticity will be beneficial to advance our understanding of schizophrenia pathology, as well as the molecular mechanism of synaptic plasticity.
Collapse
Affiliation(s)
- Kexuan Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China
| | - Panlin Liao
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jin Wen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhonghua Hu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha 410008, Hunan, PR China,Correspondence to: Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, PR China.
| |
Collapse
|
18
|
Sakimoto Y, Shintani A, Yoshiura D, Goshima M, Kida H, Mitsushima D. A critical period for learning and plastic changes at hippocampal CA1 synapses. Sci Rep 2022; 12:7199. [PMID: 35504922 PMCID: PMC9065057 DOI: 10.1038/s41598-022-10453-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/24/2022] [Indexed: 02/07/2023] Open
Abstract
Postnatal development of hippocampal function has been reported in many mammalian species, including humans. To obtain synaptic evidence, we analyzed developmental changes in plasticity after an inhibitory avoidance task in rats. Learning performance was low in infants (postnatal 2 weeks) but clearly improved from the juvenile period (3-4 weeks) to adulthood (8 weeks). One hour after the training, we prepared brain slices and sequentially recorded miniature excitatory postsynaptic currents (mEPSCs) and inhibitory postsynaptic currents (mIPSCs) from the same hippocampal CA1 neuron. Although the training failed to affect the amplitude of either mEPSCs or mIPSCs at 2 weeks, it increased mEPSC, but not mIPSC, amplitude at 3 weeks. At 4 weeks, the training had increased the amplitude of both mEPSCs and mIPSCs, whereas mIPSC, but not mEPSC, amplitude was increased at 8 weeks. Because early-life physiological functions can affect performance, we also evaluated sensory-motor functions together with emotional state and found adequate sensory/motor functions from infancy to adulthood. Moreover, by analyzing performance of rats in multiple hippocampal-dependent tasks, we found that the developmental changes in the performance are task dependent. Taken together, these findings delineate a critical period for learning and plastic changes at hippocampal CA1 synapses.
Collapse
Affiliation(s)
- Yuya Sakimoto
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan.
| | - Ako Shintani
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Daiki Yoshiura
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Makoto Goshima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Hiroyuki Kida
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Dai Mitsushima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan.
- The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, 753-8511, Japan.
| |
Collapse
|
19
|
Xolalpa-Cueva L, García-Carlos CA, Villaseñor-Zepeda R, Orta-Salazar E, Díaz-Cintra S, Peña-Ortega F, Perry G, Mondragón-Rodríguez S. Hyperphosphorylated Tau Relates to Improved Cognitive Performance and Reduced Hippocampal Excitability in the Young rTg4510 Mouse Model of Tauopathy. J Alzheimers Dis 2022; 87:529-543. [PMID: 35342085 DOI: 10.3233/jad-215186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Tau hyperphosphorylation at several sites, including those close to its microtubule domain (MD), is considered a key pathogenic event in the development of tauopathies. Nevertheless, we recently demonstrated that at the very early disease stage, tau phosphorylation (pTau) at MD sites promotes neuroprotection by preventing seizure-like activity. OBJECTIVE To further support the notion that very early pTau is not detrimental, the present work evaluated the young rTg4510 mouse model of tauopathy as a case study. Thus, in mice at one month of age (PN30-35), we studied the increase of pTau within the hippocampal area as well as hippocampal and locomotor function. METHODS We used immunohistochemistry, T-maze, nesting test, novel object recognition test, open field arena, and electrophysiology. RESULTS Our results showed that the very young rTg4510 mouse model has no detectable changes in hippocampal dependent tasks, such as spontaneous alternation and nesting, or in locomotor activity. However, at this very early stage the hippocampal neurons from PN30-35 rTg4510 mice accumulate pTau protein and exhibit changes in hippocampal oscillatory activity. Moreover, we found a significant reduction in the somatic area of pTau positive pyramidal and granule neurons in the young rTg4510 mice. Despite this, improved memory and increased number of dendrites per cell in granule neurons was found. CONCLUSION Altogether, this study provides new insights into the early pathogenesis of tauopathies and provides further evidence that pTau remodels hippocampal function and morphology.
Collapse
Affiliation(s)
- Lorena Xolalpa-Cueva
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, México
| | - Carlos Antonio García-Carlos
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, México
| | - Rocío Villaseñor-Zepeda
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, México
| | - Erika Orta-Salazar
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, México
| | - Sofia Díaz-Cintra
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, México
| | - Fernando Peña-Ortega
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, México
| | - George Perry
- UTSA Neuroscience Institute and Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Siddhartha Mondragón-Rodríguez
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, México.,CONACYT National Council for Science and Technology, México, México
| |
Collapse
|
20
|
Abstract
The last century was characterized by a significant scientific effort aimed at unveiling the neurobiological basis of learning and memory. Thanks to the characterization of the mechanisms regulating the long-term changes of neuronal synaptic connections, it was possible to understand how specific neural networks shape themselves during the acquisition of memory traces or complex motor tasks. In this chapter, we will summarize the mechanisms underlying the main forms of synaptic plasticity taking advantage of the studies performed in the hippocampus and in the nucleus striatum, key brain structures that play a crucial role in cognition. Moreover, we will discuss how the molecular pathways involved in the induction of physiologic synaptic long-term changes could be disrupted during neurodegenerative and neuroinflammatory disorders, highlighting the translational relevance of this intriguing research field.
Collapse
Affiliation(s)
- Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Antonio de Iure
- IRCCS San Raffaele Roma, Laboratory of Experimental Neurophysiology, Rome, Italy
| | - Barbara Picconi
- IRCCS San Raffaele Roma, Laboratory of Experimental Neurophysiology, Rome, Italy; University San Raffaele, Rome, Italy.
| |
Collapse
|
21
|
Niu X, Yu K, He B. Transcranial focused ultrasound induces sustained synaptic plasticity in rat hippocampus. Brain Stimul 2022; 15:352-359. [DOI: 10.1016/j.brs.2022.01.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 12/22/2022] Open
|
22
|
Tao W, Lee J, Chen X, Díaz-Alonso J, Zhou J, Pleasure S, Nicoll RA. Synaptic memory requires CaMKII. eLife 2021; 10:e60360. [PMID: 34908526 PMCID: PMC8798046 DOI: 10.7554/elife.60360] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/14/2021] [Indexed: 01/28/2023] Open
Abstract
Long-term potentiation (LTP) is arguably the most compelling cellular model for learning and memory. While the mechanisms underlying the induction of LTP ('learning') are well understood, the maintenance of LTP ('memory') has remained contentious over the last 20 years. Here, we find that Ca2+-calmodulin-dependent kinase II (CaMKII) contributes to synaptic transmission and is required LTP maintenance. Acute inhibition of CaMKII erases LTP and transient inhibition of CaMKII enhances subsequent LTP. These findings strongly support the role of CaMKII as a molecular storage device.
Collapse
Affiliation(s)
- Wucheng Tao
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical UniversityFuzhouChina
- Department of Cellular and Molecular Pharmacology, University of California, San FranciscoSan FranciscoUnited States
| | - Joel Lee
- Department of Cellular and Molecular Pharmacology, University of California, San FranciscoSan FranciscoUnited States
| | - Xiumin Chen
- Department of Cellular and Molecular Pharmacology, University of California, San FranciscoSan FranciscoUnited States
| | - Javier Díaz-Alonso
- Department of Cellular and Molecular Pharmacology, University of California, San FranciscoSan FranciscoUnited States
| | - Jing Zhou
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Samuel Pleasure
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California, San FranciscoSan FranciscoUnited States
- Physiology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
23
|
Coleman BC, Manz KM, Grueter BA. Kappa opioid receptor modulation of excitatory drive onto nucleus accumbens fast-spiking interneurons. Neuropsychopharmacology 2021; 46:2340-2349. [PMID: 34400782 PMCID: PMC8581025 DOI: 10.1038/s41386-021-01146-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023]
Abstract
The dynorphin/kappa opioid receptor (KOR) system within the nucleus accumbens (NAc) contributes to affective states. Parvalbumin fast-spiking interneurons (PV-FSIs), a key component of feedforward inhibition, participate in integration of excitatory inputs to the NAc by robustly inhibiting select populations of medium spiny output neurons, therefore greatly influencing NAc dependent behavior. How the dynorphin/KOR system regulates feedforward inhibition in the NAc remains unknown. Here, we elucidate the molecular mechanisms of KOR inhibition of excitatory transmission onto NAc PV-FSIs using a combination of whole-cell patch-clamp electrophysiology, optogenetics, pharmacology, and a parvalbumin reporter mouse. We find that postsynaptic KOR stimulation induces long-term depression (LTD) of excitatory synapses onto PV-FSI by stimulating the endocytosis of AMPARs via a PKA and calcineurin-dependent mechanism. Furthermore, KOR regulation of PV-FSI synapses are input specific, inhibiting thalamic but not cortical inputs. Finally, following acute stress, a protocol known to elevate dynorphin/KOR signaling in the NAc, KOR agonists no longer inhibit excitatory transmission onto PV-FSI. In conclusion, we delineate pathway-specific mechanisms mediating KOR control of feedforward inhibitory circuits in the NAc and provide evidence for the recruitment of this system in response to stress.
Collapse
Affiliation(s)
| | - Kevin M Manz
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Brad A Grueter
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
24
|
Yang YS, Choi JH, Rah JC. Hypoxia with inflammation and reperfusion alters membrane resistance by dynamically regulating voltage-gated potassium channels in hippocampal CA1 neurons. Mol Brain 2021; 14:147. [PMID: 34556177 PMCID: PMC8461870 DOI: 10.1186/s13041-021-00857-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/07/2021] [Indexed: 02/01/2023] Open
Abstract
Hypoxia typically accompanies acute inflammatory responses in patients and animal models. However, a limited number of studies have examined the effect of hypoxia in combination with inflammation (Hypo-Inf) on neural function. We previously reported that neuronal excitability in hippocampal CA1 neurons decreased during hypoxia and greatly rebounded upon reoxygenation. We attributed this altered excitability mainly to the dynamic regulation of hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels and input resistance. However, the molecular mechanisms underlying input resistance changes by Hypo-Inf and reperfusion remained unclear. In the present study, we found that a change in the density of the delayed rectifier potassium current (IDR) can explain the input resistance variability. Furthermore, voltage-dependent inactivation of A-type potassium (IA) channels shifted in the depolarizing direction during Hypo-Inf and reverted to normal upon reperfusion without a significant alteration in the maximum current density. Our results indicate that changes in the input resistance, and consequently excitability, caused by Hypo-Inf and reperfusion are at least partially regulated by the availability and voltage dependence of KV channels. Moreover, these results suggest that selective KV channel modulators can be used as potential neuroprotective drugs to minimize hypoxia- and reperfusion-induced neuronal damage.
Collapse
Affiliation(s)
- Yoon-Sil Yang
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
| | - Joon Ho Choi
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
| | - Jong-Cheol Rah
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333 Techno Jungang-daero, Dalseong-gun, Daegu, 42988 South Korea
| |
Collapse
|
25
|
Bin Ibrahim MZ, Benoy A, Sajikumar S. Long-term plasticity in the hippocampus: maintaining within and 'tagging' between synapses. FEBS J 2021; 289:2176-2201. [PMID: 34109726 DOI: 10.1111/febs.16065] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/15/2021] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
Synapses between neurons are malleable biochemical structures, strengthening and diminishing over time dependent on the type of information they receive. This phenomenon known as synaptic plasticity underlies learning and memory, and its different forms, long-term potentiation (LTP) and long-term depression (LTD), perform varied cognitive roles in reinforcement, relearning and associating memories. Moreover, both LTP and LTD can exist in an early transient form (early-LTP/LTD) or a late persistent form (late-LTP/LTD), which are triggered by different induction protocols, and also differ in their dependence on protein synthesis and the involvement of key molecular players. Beyond homosynaptic modifications, synapses can also interact with one another. This is encapsulated in the synaptic tagging and capture hypothesis (STC), where synapses expressing early-LTP/LTD present a 'tag' that can capture the protein synthesis products generated during a temporally proximal late-LTP/LTD induction. This 'tagging' phenomenon forms the framework of synaptic interactions in various conditions and accounts for the cellular basis of the time-dependent associativity of short-lasting and long-lasting memories. All these synaptic modifications take place under controlled neuronal conditions, regulated by subcellular elements such as epigenetic regulation, proteasomal degradation and neuromodulatory signals. Here, we review current understanding of the different forms of synaptic plasticity and its regulatory mechanisms in the hippocampus, a brain region critical for memory formation. We also discuss expression of plasticity in hippocampal CA2 area, a long-overlooked narrow hippocampal subfield and the behavioural correlate of STC. Lastly, we put forth perspectives for an integrated view of memory representation in synapses.
Collapse
Affiliation(s)
- Mohammad Zaki Bin Ibrahim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore
| | - Amrita Benoy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
26
|
Dore K, Carrico Z, Alfonso S, Marino M, Koymans K, Kessels HW, Malinow R. PSD-95 protects synapses from β-amyloid. Cell Rep 2021; 35:109194. [PMID: 34077732 PMCID: PMC8237704 DOI: 10.1016/j.celrep.2021.109194] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/16/2021] [Accepted: 05/10/2021] [Indexed: 01/22/2023] Open
Abstract
Beta-amyloid (Aβ) depresses excitatory synapses by a poorly understood mechanism requiring NMDA receptor (NMDAR) function. Here, we show that increased PSD-95, a major synaptic scaffolding molecule, blocks the effects of Aβ on synapses. The protective effect persists in tissue lacking the AMPA receptor subunit GluA1, which prevents the confounding synaptic potentiation by increased PSD-95. Aβ modifies the conformation of the NMDAR C-terminal domain (CTD) and its interaction with protein phosphatase 1 (PP1), producing synaptic weakening. Higher endogenous levels or overexpression of PSD-95 block Aβ-induced effects on the NMDAR CTD conformation, its interaction with PP1, and synaptic weakening. Our results indicate that increased PSD-95 protects synapses from Aβ toxicity, suggesting that low levels of synaptic PSD-95 may be a molecular sign indicating synapse vulnerability to Aβ. Importantly, pharmacological inhibition of its depalmitoylation increases PSD-95 at synapses and rescues deficits caused by Aβ, possibly opening a therapeutic avenue against Alzheimer's disease.
Collapse
Affiliation(s)
- Kim Dore
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA.
| | - Zachary Carrico
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA
| | - Stephanie Alfonso
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA
| | - Marc Marino
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA
| | - Karin Koymans
- Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Helmut W Kessels
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA; Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Roberto Malinow
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA
| |
Collapse
|
27
|
Fan Y, Gao Y, Therriault J, Luo J, Ba M, Zhang H, the Alzheimer’s Disease Neuroimaging Initiative. The Effects of CSF Neurogranin and APOE ε4 on Cognition and Neuropathology in Mild Cognitive Impairment and Alzheimer's Disease. Front Aging Neurosci 2021; 13:667899. [PMID: 33986657 PMCID: PMC8110906 DOI: 10.3389/fnagi.2021.667899] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/30/2021] [Indexed: 01/08/2023] Open
Abstract
Cerebrospinal fluid (CSF) measurements of neurogranin (Ng) have emerged as a promising biomarker for cognitive decline in mild cognitive impairment (MCI) and Alzheimer’s disease (AD). The apolipoprotein E ε4 (APOE ε4) allele is by far the most consistent genetic risk factor for AD. However, it is not known whether the pathophysiological roles of Ng in MCI or AD are related to APOEε4. We stratified 250 participants from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database into cognitively normal (CN) ε4 negative (CN ε4−), CN ε4 positive (CN ε4+), MCI ε4 negative (MCI ε4−), MCI ε4 positive (MCI ε4+), AD ε4 negative (AD ε4−), and AD ε4 positive (AD ε4+). CSF Ng levels were significantly increased in APOE ε4 carriers compared to APOE ε4 non-carriers with MCI. In addition, CSF Ng identified MCI ε4+ versus CN ε4−, but not MCI ε4− versus CN ε4−. Similarly, CSF Ng negatively correlated with Mini-Mental State Examination (MMSE) scores at baseline in the MCI ε4+ group. Our findings support the use of CSF Ng as a biomarker of synaptic pathology for AD. We propose that the roles of CSF Ng in the pathophysiology of MCI may be related to APOE ε4.
Collapse
|
28
|
Regulation of Synaptic Transmission and Plasticity by Protein Phosphatase 1. J Neurosci 2021; 41:3040-3050. [PMID: 33827970 DOI: 10.1523/jneurosci.2026-20.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 01/22/2023] Open
Abstract
Protein phosphatases, by counteracting protein kinases, regulate the reversible phosphorylation of many substrates involved in synaptic plasticity, a cellular model for learning and memory. A prominent phosphatase regulating synaptic plasticity and neurologic disorders is the serine/threonine protein phosphatase 1 (PP1). PP1 has three isoforms (α, β, and γ, encoded by three different genes), which are regulated by a vast number of interacting subunits that define their enzymatic substrate specificity. In this review, we discuss evidence showing that PP1 regulates synaptic transmission and plasticity, as well as presenting novel models of PP1 regulation suggested by recent experimental evidence. We also outline the required targeting of PP1 by neurabin and spinophilin to achieve substrate specificity at the synapse to regulate AMPAR and NMDAR function. We then highlight the role of inhibitor-2 in regulating PP1 function in plasticity, including its positive regulation of PP1 function in vivo in memory formation. We also discuss the distinct function of the three PP1 isoforms in synaptic plasticity and brain function, as well as briefly discuss the role of inhibitory phosphorylation of PP1, which has received recent emphasis in the regulation of PP1 activity in neurons.
Collapse
|
29
|
Sleep Deprivation and Neurological Disorders. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5764017. [PMID: 33381558 PMCID: PMC7755475 DOI: 10.1155/2020/5764017] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022]
Abstract
Sleep plays an important role in maintaining neuronal circuitry, signalling and helps maintain overall health and wellbeing. Sleep deprivation (SD) disturbs the circadian physiology and exerts a negative impact on brain and behavioural functions. SD impairs the cellular clearance of misfolded neurotoxin proteins like α-synuclein, amyloid-β, and tau which are involved in major neurodegenerative diseases like Alzheimer's disease and Parkinson's disease. In addition, SD is also shown to affect the glymphatic system, a glial-dependent metabolic waste clearance pathway, causing accumulation of misfolded faulty proteins in synaptic compartments resulting in cognitive decline. Also, SD affects the immunological and redox system resulting in neuroinflammation and oxidative stress. Hence, it is important to understand the molecular and biochemical alterations that are the causative factors leading to these pathophysiological effects on the neuronal system. This review is an attempt in this direction. It provides up-to-date information on the alterations in the key processes, pathways, and proteins that are negatively affected by SD and become reasons for neurological disorders over a prolonged period of time, if left unattended.
Collapse
|
30
|
Platholi J, Hemmings HC. Modulation of dendritic spines by protein phosphatase-1. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:117-144. [PMID: 33706930 DOI: 10.1016/bs.apha.2020.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Protein phosphatase-1 (PP-1), a highly conserved multifunctional serine/threonine phosphatase, is enriched in dendritic spines where it plays a major role in modulating excitatory synaptic activity. In addition to established functions in spine maturation and development, multi-subunit holoenzyme forms of PP-1 modulate higher-order cognitive functions such learning and memory. Mechanisms involved in regulating PP-1 activity and localization in spines include interactions with neurabin and spinophilin, structurally related synaptic scaffolding proteins associated with the actin cytoskeleton. Since PP-1 is a critical element in synaptic development, signaling, and plasticity, alterations in PP-1 signaling in dendritic spines are implicated in various neurological and psychiatric disorders. The effects of PP-1 depend on its isoform-specific association with regulatory proteins and activation of downstream signaling pathways. Here we review the role of PP-1 and its binding proteins neurabin and spinophilin in both developing and established dendritic spines, as well as some of the disorders that result from its dysregulation.
Collapse
Affiliation(s)
- Jimcy Platholi
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
31
|
Mondragón-Rodríguez S, Salgado-Burgos H, Peña-Ortega F. Circuitry and Synaptic Dysfunction in Alzheimer's Disease: A New Tau Hypothesis. Neural Plast 2020; 2020:2960343. [PMID: 32952546 PMCID: PMC7481966 DOI: 10.1155/2020/2960343] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/20/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
For more than five decades, the field of Alzheimer's disease (AD) has focused on two main hypotheses positing amyloid-beta (Aβ) and Tau phosphorylation (pTau) as key pathogenic mediators. In line with these canonical hypotheses, several groups around the world have shown that the synaptotoxicity in AD depends mainly on the increase in pTau levels. Confronting this leading hypothesis, a few years ago, we reported that the increase in phosphorylation levels of dendritic Tau, at its microtubule domain (MD), acts as a neuroprotective mechanism that prevents N-methyl-D-aspartate receptor (NMDAr) overexcitation, which allowed us to propose that Tau protein phosphorylated near MD sites is involved in neuroprotection, rather than in neurodegeneration. Further supporting this alternative role of pTau, we have recently shown that early increases in pTau close to MD sites prevent hippocampal circuit overexcitation in a transgenic AD mouse model. Here, we will synthesize this new evidence that confronts the leading Tau-based AD hypothesis and discuss the role of pTau modulating neural circuits and network connectivity. Additionally, we will briefly address the role of brain circuit alterations as a potential biomarker for detecting the prodromal AD stage.
Collapse
Affiliation(s)
- Siddhartha Mondragón-Rodríguez
- CONACYT National Council for Science and Technology, México, Mexico
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, Mexico
| | - Humberto Salgado-Burgos
- UADY Neurosciences Department, Autonomous University of Yucatán, 97000 Mérida, Yucatán, Mexico
| | - Fernando Peña-Ortega
- UNAM Developmental Neurobiology and Neurophysiology, Institute of Neurobiology, National Autonomous University of México, Querétaro, Mexico
| |
Collapse
|
32
|
Fontaine CJ, Gräfe EL, Pinar C, Bonilla-Del Río I, Grandes P, Christie BR. Endocannabinoid receptors contribute significantly to multiple forms of long-term depression in the rat dentate gyrus. LEARNING & MEMORY (COLD SPRING HARBOR, N.Y.) 2020; 27:380-389. [PMID: 32817304 PMCID: PMC7433656 DOI: 10.1101/lm.050666.119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
Abstract
Cannabinoid receptors are widely expressed throughout the hippocampal formation, but are particularly dense in the dentate gyrus (DG) subregion. We, and others, have shown in mice that cannabinoid type 1 receptors (CB1Rs) are involved in a long-term depression (LTD) that can be induced by prolonged 10 Hz stimulation of the medial perforant path (MPP)-granule cell synaptic input to the DG. Here, we extend this work to examine the involvement of CB1Rs in other common forms of LTD in the hippocampus of juvenile male and female Sprague–Dawley rats (Rattus norvegicus). We found, as in mice, that prolonged 10 Hz stimulation (6000 pulses) could reliably induce a form of LTD that was dependent upon CB1R activation. In addition, we also discovered a role for both CB1R and mGluR proteins in LTD induced with 1 Hz low-frequency stimulation (1 Hz-LTD; 900 pulses) and in LTD induced by bath application of the group I mGluR agonist (RS)-3,5-Dihydroxyphenylglycine (DHPG; DHPG-LTD). This study elucidates an essential role for endocannabinoid receptors in a number of forms of LTD in the rat DG, and identifies a novel role for CB1Rs as potential therapeutic targets for conditions that involve impaired LTD in the DG.
Collapse
Affiliation(s)
- Christine J Fontaine
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Erin L Gräfe
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Cristina Pinar
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Itziar Bonilla-Del Río
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain.,Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940 Leioa, Spain
| | - Pedro Grandes
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada.,Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain.,Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, E-48940 Leioa, Spain
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada.,Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, USA
| |
Collapse
|
33
|
Moreno A. Molecular mechanisms of forgetting. Eur J Neurosci 2020; 54:6912-6932. [DOI: 10.1111/ejn.14839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/23/2020] [Accepted: 05/18/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Andrea Moreno
- Danish Institute of Translational Neuroscience (DANDRITE) Aarhus University Aarhus C Denmark
| |
Collapse
|
34
|
Purkey AM, Dell’Acqua ML. Phosphorylation-Dependent Regulation of Ca 2+-Permeable AMPA Receptors During Hippocampal Synaptic Plasticity. Front Synaptic Neurosci 2020; 12:8. [PMID: 32292336 PMCID: PMC7119613 DOI: 10.3389/fnsyn.2020.00008] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/18/2020] [Indexed: 01/28/2023] Open
Abstract
Experience-dependent learning and memory require multiple forms of plasticity at hippocampal and cortical synapses that are regulated by N-methyl-D-aspartate receptors (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type ionotropic glutamate receptors (NMDAR, AMPAR). These plasticity mechanisms include long-term potentiation (LTP) and depression (LTD), which are Hebbian input-specific mechanisms that rapidly increase or decrease AMPAR synaptic strength at specific inputs, and homeostatic plasticity that globally scales-up or -down AMPAR synaptic strength across many or even all inputs. Frequently, these changes in synaptic strength are also accompanied by a change in the subunit composition of AMPARs at the synapse due to the trafficking to and from the synapse of receptors lacking GluA2 subunits. These GluA2-lacking receptors are most often GluA1 homomeric receptors that exhibit higher single-channel conductance and are Ca2+-permeable (CP-AMPAR). This review article will focus on the role of protein phosphorylation in regulation of GluA1 CP-AMPAR recruitment and removal from hippocampal synapses during synaptic plasticity with an emphasis on the crucial role of local signaling by the cAMP-dependent protein kinase (PKA) and the Ca2+calmodulin-dependent protein phosphatase 2B/calcineurin (CaN) that is coordinated by the postsynaptic scaffold protein A-kinase anchoring protein 79/150 (AKAP79/150).
Collapse
Affiliation(s)
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
35
|
Ryu HH, Kim SY, Lee YS. Connecting the dots between SHP2 and glutamate receptors. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:129-135. [PMID: 32140036 PMCID: PMC7043995 DOI: 10.4196/kjpp.2020.24.2.129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 11/18/2022]
Abstract
SHP2 is an unusual protein phosphatase that functions as an activator for several signaling pathways, including the RAS pathway, while most other phosphatases suppress their downstream signaling cascades. The physiological and pathophysiological roles of SHP2 have been extensively studied in the field of cancer research. Mutations in the PTPN11 gene which encodes SHP2 are also highly associated with developmental disorders, such as Noonan syndrome (NS), and cognitive deficits including learning disabilities are common among NS patients. However, the molecular and cellular mechanism by which SHP2 is involved in cognitive functions is not well understood. Recent studies using SHP2 mutant mice or pharmacological inhibitors have shown that SHP2 plays critical role in learning and memory and synaptic plasticity. Here, we review the recent studies demonstrating that SHP2 is involved in synaptic plasticity, and learning and memory, by the regulation of the expression and/or function of glutamate receptors. We suggest that each cell type may have distinct paths connecting the dots between SHP2 and glutamate receptors, and these paths may also change with aging.
Collapse
Affiliation(s)
- Hyun-Hee Ryu
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sun Yong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
36
|
Pan Y, Monje M. Activity Shapes Neural Circuit Form and Function: A Historical Perspective. J Neurosci 2020; 40:944-954. [PMID: 31996470 PMCID: PMC6988998 DOI: 10.1523/jneurosci.0740-19.2019] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/30/2022] Open
Abstract
The brilliant and often prescient hypotheses of Ramon y Cajal have proven foundational for modern neuroscience, but his statement that "In adult centers the nerve paths are something fixed, ended, immutable … " is an exception that did not stand the test of empirical study. Mechanisms of cellular and circuit-level plasticity continue to shape and reshape many regions of the adult nervous system long after the neurodevelopmental period. Initially focused on neurons alone, the field has followed a meteoric trajectory in understanding of activity-regulated neurodevelopment and ongoing neuroplasticity with an arc toward appreciating neuron-glial interactions and the role that each neural cell type plays in shaping adaptable neural circuity. In this review, as part of a celebration of the 50th anniversary of Society for Neuroscience, we provide a historical perspective, following this arc of inquiry from neuronal to neuron-glial mechanisms by which activity and experience modulate circuit structure and function. The scope of this consideration is broad, and it will not be possible to cover the wealth of knowledge about all aspects of activity-dependent circuit development and plasticity in depth.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305
| |
Collapse
|
37
|
Elliott T. Dynamic Integrative Synaptic Plasticity Explains the Spacing Effect in the Transition from Short- to Long-Term Memory. Neural Comput 2019; 31:2212-2251. [PMID: 31525308 DOI: 10.1162/neco_a_01227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Repeated stimuli that are spaced apart in time promote the transition from short- to long-term memory, while massing repetitions together does not. Previously, we showed that a model of integrative synaptic plasticity, in which plasticity induction signals are integrated by a low-pass filter before plasticity is expressed, gives rise to a natural timescale at which to repeat stimuli, hinting at a partial account of this spacing effect. The account was only partial because the important role of neuromodulation was not considered. We now show that by extending the model to allow dynamic integrative synaptic plasticity, the model permits synapses to robustly discriminate between spaced and massed repetition protocols, suppressing the response to massed stimuli while maintaining that to spaced stimuli. This is achieved by dynamically coupling the filter decay rate to neuromodulatory signaling in a very simple model of the signaling cascades downstream from cAMP production. In particular, the model's parameters may be interpreted as corresponding to the duration and amplitude of the waves of activity in the MAPK pathway. We identify choices of parameters and repetition times for stimuli in this model that optimize the ability of synapses to discriminate between spaced and massed repetition protocols. The model is very robust to reasonable changes around these optimal parameters and times, but for large changes in parameters, the model predicts that massed and spaced stimuli cannot be distinguished or that the responses to both patterns are suppressed. A model of dynamic integrative synaptic plasticity therefore explains the spacing effect under normal conditions and also predicts its breakdown under abnormal conditions.
Collapse
Affiliation(s)
- Terry Elliott
- Department of Electronics and Computer Science, University of Southampton, Highfield, Southampton, SO17 1BJ, U.K.
| |
Collapse
|
38
|
Reiner A, Levitz J. Glutamatergic Signaling in the Central Nervous System: Ionotropic and Metabotropic Receptors in Concert. Neuron 2019; 98:1080-1098. [PMID: 29953871 DOI: 10.1016/j.neuron.2018.05.018] [Citation(s) in RCA: 407] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/19/2018] [Accepted: 05/10/2018] [Indexed: 12/28/2022]
Abstract
Glutamate serves as both the mammalian brain's primary excitatory neurotransmitter and as a key neuromodulator to control synapse and circuit function over a wide range of spatial and temporal scales. This functional diversity is decoded by two receptor families: ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs). The challenges posed by the complexity and physiological importance of each of these subtypes has limited our appreciation and understanding of how these receptors work in concert. In this review, by comparing both receptor families with a focus on their crosstalk, we argue for a more holistic understanding of neural glutamate signaling.
Collapse
Affiliation(s)
- Andreas Reiner
- Department of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany.
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
39
|
Wang J, Xie R, Kou X, Liu Y, Qi C, Liu R, You W, Gao J, Gao X. A protein phosphatase 2A deficit in the hippocampal CA1 area impairs memory extinction. Mol Brain 2019; 12:51. [PMID: 31113458 PMCID: PMC6528246 DOI: 10.1186/s13041-019-0469-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/30/2019] [Indexed: 01/17/2023] Open
Abstract
Protein phosphorylation plays an important role in learning and memory. Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase involved in the regulation of neural synaptic plasticity. Here, to determine if PP2A is necessary for successful learning and memory, we have utilized a Tg (Camk2a-cre) T29–2Stl mice to specific knock down the expression of hippocampal PP2A in mice. By analysing behavioural, we observed that loss of PP2A in the hippocampal CA1 area did not affect the formation of memory but impaired contextual fear memory extinction. We use the electrophysiological recording to find the synaptic mechanisms. The results showed that the basic synapse transmission and synaptic plasticity of PP2A conditional knockout (CKO) mice were impaired. Moreover, PP2A CKO mice exhibited a saturating long-term potentiation inducted by strong theta burst stimulation but no depotentiation after low-frequency stimulation. Taken together, our results provide the evidence that PP2A is involved in synaptic transmission and hippocampus-dependent memory extinction.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ran Xie
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xiaolin Kou
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yu Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Cui Qi
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Rui Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Weiyan You
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jun Gao
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Xiang Gao
- Model Animal Research Center and MOE Key Laboratory of Model Animals for Disease Study, Nanjing University, Nanjing, 210093, Jiangsu, China.
| |
Collapse
|
40
|
Van Dyke AM, Francis TC, Chen H, Bailey AM, Thompson SM. RETRACTED: Chronic fluoxetine treatment in vivo enhances excitatory synaptic transmission in the hippocampus. Neuropharmacology 2019; 150:38-45. [PMID: 30851310 PMCID: PMC6475886 DOI: 10.1016/j.neuropharm.2019.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/07/2019] [Accepted: 03/02/2019] [Indexed: 12/17/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Authors. After publication, Scott M. Thompson found significant concerns about the data and duly notified The University of Maryland. The University of Maryland conducted an internal investigation which confirmed that the article was compromised. Namely in Figure 2B, the Investigation Committee determined that the western blots used to create the figure were not the ones used for the quantification and concluded that the figure was falsified to fit the hypothesis. In Figure 2C and 2D, the Investigation Committee determined that the densitometry data (pCaMKII, pS831, CamKII and GluA1) used to create the histogram were falsified to fit the hypothesis.
Collapse
Affiliation(s)
- Adam M Van Dyke
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Training Programs in Neuroscience, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Membrane Biology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - T Chase Francis
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Training Programs in Neuroscience, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Haiwen Chen
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Training Programs in Neuroscience, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Membrane Biology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA; Medical Scientist Training Program, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Aileen M Bailey
- Department of Psychology, Saint Mary's College of Maryland, St. Mary's City, MD, USA
| | - Scott M Thompson
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
41
|
Chirillo MA, Waters MS, Lindsey LF, Bourne JN, Harris KM. Local resources of polyribosomes and SER promote synapse enlargement and spine clustering after long-term potentiation in adult rat hippocampus. Sci Rep 2019; 9:3861. [PMID: 30846859 PMCID: PMC6405867 DOI: 10.1038/s41598-019-40520-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 02/07/2019] [Indexed: 12/11/2022] Open
Abstract
Synapse clustering facilitates circuit integration, learning, and memory. Long-term potentiation (LTP) of mature neurons produces synapse enlargement balanced by fewer spines, raising the question of how clusters form despite this homeostatic regulation of total synaptic weight. Three-dimensional reconstruction from serial section electron microscopy (3DEM) revealed the shapes and distributions of smooth endoplasmic reticulum (SER) and polyribosomes, subcellular resources important for synapse enlargement and spine outgrowth. Compared to control stimulation, synapses were enlarged two hours after LTP on resource-rich spines containing polyribosomes (4% larger than control) or SER (15% larger). SER in spines shifted from a single tubule to complex spine apparatus after LTP. Negligible synapse enlargement (0.6%) occurred on resource-poor spines lacking SER and polyribosomes. Dendrites were divided into discrete synaptic clusters surrounded by asynaptic segments. Spine density was lowest in clusters having only resource-poor spines, especially following LTP. In contrast, resource-rich spines preserved neighboring resource-poor spines and formed larger clusters with elevated total synaptic weight following LTP. These clusters also had more shaft SER branches, which could sequester cargo locally to support synapse growth and spinogenesis. Thus, resources appear to be redistributed to synaptic clusters with LTP-related synapse enlargement while homeostatic regulation suppressed spine outgrowth in resource-poor synaptic clusters.
Collapse
Affiliation(s)
- Michael A Chirillo
- Center for Learning and Memory, Department of Neuroscience, The University of Texas at Austin, Austin, Texas, 78712, USA.,Fulbright U.S. Scholar Program, University of Belgrade, Studentski trg 1, Belgrade, 11000, Serbia
| | - Mikayla S Waters
- Center for Learning and Memory, Department of Neuroscience, The University of Texas at Austin, Austin, Texas, 78712, USA.,McGovern Medical School in Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Laurence F Lindsey
- Center for Learning and Memory, Department of Neuroscience, The University of Texas at Austin, Austin, Texas, 78712, USA.,Google Seattle, Seattle, Washington, 98103, USA
| | - Jennifer N Bourne
- Center for Learning and Memory, Department of Neuroscience, The University of Texas at Austin, Austin, Texas, 78712, USA.,Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Kristen M Harris
- Center for Learning and Memory, Department of Neuroscience, The University of Texas at Austin, Austin, Texas, 78712, USA.
| |
Collapse
|
42
|
Khoo GH, Lin YT, Tsai TC, Hsu KS. Perineuronal Nets Restrict the Induction of Long-Term Depression in the Mouse Hippocampal CA1 Region. Mol Neurobiol 2019; 56:6436-6450. [PMID: 30826967 DOI: 10.1007/s12035-019-1526-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/08/2019] [Indexed: 10/27/2022]
Abstract
Long-term depression (LTD) of synaptic efficacy is widely regarded as a cellular basis of learning and memory. The magnitude of hippocampal CA1 LTD induced by low-frequency stimulation (LFS) declines with age, but the mechanisms involved remain poorly understood. Perineuronal nets (PNNs) are specialized extracellular matrix structures that function in dampening synaptic plasticity during postnatal development, suggesting that PNN formation may restrict LTD induction in the adult hippocampus. Here, we show that PNNs tightly enwrap a subpopulation of parvalbumin (PV) interneurons in the hippocampal CA1 region and enzymatic removal of PNNs with the chondroitinase ABC alters the excitatory/inhibitory synaptic balance toward more excitation and restores the ability of LFS to induce an N-methyl-D-aspartate receptor-dependent LTD at Schaffer collateral-CA1 synapses in slices from male adult mice. Early interference with depolarizing GABA with Na+-K+-2Cl- cotransporter inhibitor bumetanide impairs the maturation of PNNs and enhances LTD induction. These results provide novel insights into a previously unrecognized role for PNNs around PV interneurons in restricting long-term synaptic plasticity at excitatory synapses on hippocampal CA1 neurons in adulthood.
Collapse
Affiliation(s)
- Guan Hock Khoo
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan City, 70101, Taiwan
| | - Yu-Ting Lin
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan City, 70101, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Tsung-Chih Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Kuei-Sen Hsu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan City, 70101, Taiwan. .,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
43
|
Bear MF, Cooke SF, Giese KP, Kaang BK, Kennedy MB, Kim JI, Morris RGM, Park P. In memoriam: John Lisman - commentaries on CaMKII as a memory molecule. Mol Brain 2018; 11:76. [PMID: 30593282 PMCID: PMC6309094 DOI: 10.1186/s13041-018-0419-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/24/2018] [Indexed: 11/10/2022] Open
Abstract
Shortly before he died in October 2017, John Lisman submitted an invited review to Molecular Brain on 'Criteria for identifying the molecular basis of the engram (CaMKII, PKMζ)'. John had no opportunity to read the referees' comments, and as a mark of the regard in which he was held by the neuroscience community the Editors decided to publish his review as submitted. This obituary takes the form of a series of commentaries on Lisman's review. At the same time we are publishing as a separate article a longer response by Todd Sacktor and André Fenton entitled 'What does LTP tell us about the roles of CaMKII and PKMζ in memory?' which presents the case for a rival memory molecule, PKMζ.
Collapse
Affiliation(s)
- Mark F. Bear
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Sam F. Cooke
- King’s College London, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, London, SE5 8AF UK
| | - Karl Peter Giese
- King’s College London, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, De Crespigny Park, London, SE5 8AF UK
| | - Bong-Kiun Kaang
- Department of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | - Mary B. Kennedy
- The Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
| | - Ji-il Kim
- Department of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | - Richard G. M. Morris
- Laboratory for Cognitive Neuroscience, Centre for Discovery Brain Sciences, Edinburgh Neuroscience, Edinburgh, EH8 9JZ UK
| | - Pojeong Park
- Department of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| |
Collapse
|
44
|
Distinct Roles of Protein Phosphatase 1 Bound on Neurabin and Spinophilin and Its Regulation in AMPA Receptor Trafficking and LTD Induction. Mol Neurobiol 2018; 55:7179-7186. [PMID: 29383693 DOI: 10.1007/s12035-018-0886-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/08/2018] [Indexed: 01/30/2023]
Abstract
Protein phosphatase-1 (PP1) constrains learning and memory formation in part through its effects on the induction threshold of long-term potentiation (LTP) and depression (LTD). LTD induction requires both the enzymatic activity of PP1 and its proper anchoring to synaptic spines. We have shown previously that neurabin, a major synaptic scaffolding protein, targets PP1 to synapses for LTD induction. Here, we show that PP1 bound on spinophilin, a close homolog of neurabin and another major synaptic PP1 anchoring protein, does not play a role in LTD induction, which suggests that neurabin plays a privileged role in nanodomain targeting of PP1 in LTD induction. We found that protein kinase A can significantly weaken the neurabin-PP1 interaction in neurons via phosphorylation of neurabin at serine 461, a phosphorylation site adjacent to the PP1-binding motif that is not conserved in spinophilin. Finally, we found that a neurabin mutation (S461E), which mimics phosphorylation, blocked AMPA receptor endocytosis and LTD induction. The results indicate the critical importance of nanodomain targeting of PP1 within synaptic spines and its regulation in LTD induction.
Collapse
|
45
|
Woolfrey KM, O'Leary H, Goodell DJ, Robertson HR, Horne EA, Coultrap SJ, Dell'Acqua ML, Bayer KU. CaMKII regulates the depalmitoylation and synaptic removal of the scaffold protein AKAP79/150 to mediate structural long-term depression. J Biol Chem 2017; 293:1551-1567. [PMID: 29196604 DOI: 10.1074/jbc.m117.813808] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/21/2017] [Indexed: 11/06/2022] Open
Abstract
Both long-term potentiation (LTP) and depression (LTD) of excitatory synapse strength require the Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) and its autonomous activity generated by Thr-286 autophosphorylation. Additionally, LTP and LTD are correlated with dendritic spine enlargement and shrinkage that are accompanied by the synaptic accumulation or removal, respectively, of the AMPA-receptor regulatory scaffold protein A-kinase anchoring protein (AKAP) 79/150. We show here that the spine shrinkage associated with LTD indeed requires synaptic AKAP79/150 removal, which in turn requires CaMKII activity. In contrast to normal CaMKII substrates, the substrate sites within the AKAP79/150 N-terminal polybasic membrane-cytoskeletal targeting domain were phosphorylated more efficiently by autonomous compared with Ca2+/CaM-stimulated CaMKII activity. This unusual regulation was mediated by Ca2+/CaM binding to the substrate sites resulting in protection from phosphorylation in the presence of Ca2+/CaM, a mechanism that favors phosphorylation by prolonged, weak LTD stimuli versus brief, strong LTP stimuli. Phosphorylation by CaMKII inhibited AKAP79/150 association with F-actin; it also facilitated AKAP79/150 removal from spines but was not required for it. By contrast, LTD-induced spine removal of AKAP79/150 required its depalmitoylation on two Cys residues within the N-terminal targeting domain. Notably, such LTD-induced depalmitoylation was also blocked by CaMKII inhibition. These results provide a mechanism how CaMKII can indeed mediate not only LTP but also LTD through regulated substrate selection; however, in the case of AKAP79/150, indirect CaMKII effects on palmitoylation are more important than the effects of direct phosphorylation. Additionally, our results provide the first direct evidence for a function of the well-described AKAP79/150 trafficking in regulating LTD-induced spine shrinkage.
Collapse
Affiliation(s)
- Kevin M Woolfrey
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Heather O'Leary
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Dayton J Goodell
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Holly R Robertson
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Eric A Horne
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Steven J Coultrap
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Mark L Dell'Acqua
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - K Ulrich Bayer
- From the Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| |
Collapse
|
46
|
Unconventional NMDA Receptor Signaling. J Neurosci 2017; 37:10800-10807. [PMID: 29118208 DOI: 10.1523/jneurosci.1825-17.2017] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/28/2017] [Accepted: 09/28/2017] [Indexed: 11/21/2022] Open
Abstract
In the classical view, NMDA receptors (NMDARs) are stably expressed at the postsynaptic membrane, where they act via Ca2+ to signal coincidence detection in Hebbian plasticity. More recently, it has been established that NMDAR-mediated transmission can be dynamically regulated by neural activity. In addition, NMDARs have been found presynaptically, where they cannot act as conventional coincidence detectors. Unexpectedly, NMDARs have also been shown to signal metabotropically, without the need for Ca2+ This review highlights novel findings concerning these unconventional modes of NMDAR action.
Collapse
|
47
|
McGregor G, Irving AJ, Harvey J. Canonical JAK‐STAT signaling is pivotal for long‐term depression at adult hippocampal temporoammonic‐CA1 synapses. FASEB J 2017; 31:3449-3466. [DOI: 10.1096/fj.201601293rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/11/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Gemma McGregor
- Division of NeuroscienceSchool of MedicineNinewells Hospital and Medical SchoolUniversity of Dundee Dundee United Kingdom
| | - Andrew J. Irving
- School of Biomolecular and Biomedical ScienceThe Conway InstituteUniversity College Dublin Dublin Ireland
| | - Jenni Harvey
- Division of NeuroscienceSchool of MedicineNinewells Hospital and Medical SchoolUniversity of Dundee Dundee United Kingdom
| |
Collapse
|
48
|
Pinar C, Fontaine CJ, Triviño-Paredes J, Lottenberg CP, Gil-Mohapel J, Christie BR. Revisiting the flip side: Long-term depression of synaptic efficacy in the hippocampus. Neurosci Biobehav Rev 2017. [PMID: 28624435 DOI: 10.1016/j.neubiorev.2017.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Synaptic plasticity is widely regarded as a putative biological substrate for learning and memory processes. While both decreases and increases in synaptic strength are seen as playing a role in learning and memory, long-term depression (LTD) of synaptic efficacy has received far less attention than its counterpart long-term potentiation (LTP). Never-the-less, LTD at synapses can play an important role in increasing computational flexibility in neural networks. In addition, like learning and memory processes, the magnitude of LTD can be modulated by factors that include stress and sex hormones, neurotrophic support, learning environments, and age. Examining how these factors modulate hippocampal LTD can provide the means to better elucidate the molecular underpinnings of learning and memory processes. This is in turn will enhance our appreciation of how both increases and decreases in synaptic plasticity can play a role in different neurodevelopmental and neurodegenerative conditions.
Collapse
Affiliation(s)
- Cristina Pinar
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Christine J Fontaine
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Juan Triviño-Paredes
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Carina P Lottenberg
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada; Faculty of Medical Sciences of Santa Casa de São Paulo, Sao Paulo, SP, Brazil
| | - Joana Gil-Mohapel
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences and UBC Island Medical Program, University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
49
|
Di Mauro M, Tozzi A, Calabresi P, Pettorossi VE, Grassi S. Different synaptic stimulation patterns influence the local androgenic and estrogenic neurosteroid availability triggering hippocampal synaptic plasticity in the male rat. Eur J Neurosci 2017; 45:499-509. [DOI: 10.1111/ejn.13455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/21/2016] [Accepted: 10/27/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Michela Di Mauro
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| | - Alessandro Tozzi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
- Fondazione Santa Lucia – I.R.C.C.S. Rome Italy
| | - Paolo Calabresi
- Fondazione Santa Lucia – I.R.C.C.S. Rome Italy
- Dipartimento di Medicina Clinica Neurologica Università di Perugia Perugia Italy
| | - Vito Enrico Pettorossi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| | - Silvarosa Grassi
- Dipartimento di Medicina Sperimentale Sezione di Fisiologia e Biochimica Università di Perugia Via Gambuli 06156 Perugia Italy
| |
Collapse
|
50
|
Wang Z, Joshi S, Savel'ev SE, Jiang H, Midya R, Lin P, Hu M, Ge N, Strachan JP, Li Z, Wu Q, Barnell M, Li GL, Xin HL, Williams RS, Xia Q, Yang JJ. Memristors with diffusive dynamics as synaptic emulators for neuromorphic computing. NATURE MATERIALS 2017; 16:101-108. [PMID: 27669052 DOI: 10.1038/nmat4756] [Citation(s) in RCA: 680] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/17/2016] [Indexed: 05/20/2023]
Abstract
The accumulation and extrusion of Ca2+ in the pre- and postsynaptic compartments play a critical role in initiating plastic changes in biological synapses. To emulate this fundamental process in electronic devices, we developed diffusive Ag-in-oxide memristors with a temporal response during and after stimulation similar to that of the synaptic Ca2+ dynamics. In situ high-resolution transmission electron microscopy and nanoparticle dynamics simulations both demonstrate that Ag atoms disperse under electrical bias and regroup spontaneously under zero bias because of interfacial energy minimization, closely resembling synaptic influx and extrusion of Ca2+, respectively. The diffusive memristor and its dynamics enable a direct emulation of both short- and long-term plasticity of biological synapses, representing an advance in hardware implementation of neuromorphic functionalities.
Collapse
Affiliation(s)
- Zhongrui Wang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Saumil Joshi
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Sergey E Savel'ev
- Department of Physics, Loughborough University, Loughborough LE11 3TU, UK
| | - Hao Jiang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Rivu Midya
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Peng Lin
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Miao Hu
- Hewlett Packard Labs, Palo Alto, California 94304, USA
| | - Ning Ge
- Hewlett Packard Labs, Palo Alto, California 94304, USA
| | | | - Zhiyong Li
- Hewlett Packard Labs, Palo Alto, California 94304, USA
| | - Qing Wu
- Air Force Research Lab, Information Directorate, Rome, New York 13441, USA
| | - Mark Barnell
- Air Force Research Lab, Information Directorate, Rome, New York 13441, USA
| | - Geng-Lin Li
- Biology Department, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Huolin L Xin
- Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, New York 11973, USA
| | | | - Qiangfei Xia
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - J Joshua Yang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|