1
|
Chen SF, Cheng WJ, Chao CC, Kuo CH, Liao RM. Baseline-dependent enhancement of working memory by memantine in male rats: Involvement of NMDA receptor subunits and CaMKII signaling. Pharmacol Biochem Behav 2024; 245:173904. [PMID: 39522650 DOI: 10.1016/j.pbb.2024.173904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/26/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
N-methyl-d-aspartate (NMDA) receptors, activated by glutamate, play a crucial role in learning and memory. Memantine (MEM), a non-competitive NMDA receptor antagonist, is currently prescribed for the treatment of Alzheimer's disease or dementia, which meanwhile simultaneously promotes a need to clarify its potential pro-cognitive effects that exist in normal healthy individuals. However, the neurobehavioral mechanisms underlying the cognitive improvement by MEM in normal individuals remain to be elucidated. This study aimed to assess the effects of MEM on working memory, measured by a discrete paired-trial delay alternation task in a T-maze in normal male rats. The impacts of MEM were hypothesized to vary depending on different baseline levels of working memory performance. Neurochemical examination of the levels of calcium/calmodulin-dependent kinase 2 (CaMKII) and NMDA receptor subunits within five targeted brain regions was conducted after behavioral tests. The results showed that acute administration of MEM enhanced working memory performance, with 2.5, 5.0, and 10 mg/kg doses increasing task accuracy compared to the vehicle, particularly in low performers. Neurochemically, the protein expression of CaMKII in the amygdala and that of the glutamate (Glu) N2A subunit in the dorsal striatum were greater in the low-performance group than in the high-performance group. Additionally, the protein expression of the GluN2A subunit in the dorsal striatum was negatively associated with task performance at baseline. The expression of GluN1 and GluN2B in the nucleus accumbens was negatively associated with task performance in the retest three weeks after drug treatment. These findings underscore the baseline-dependent improvement of working memory resulting from MEM administration, with observed drug effects associated with alterations in the levels of NMDA receptor subunits in striatal subareas and CaMKII in the amygdala.
Collapse
Affiliation(s)
- Shuo-Fu Chen
- Department of Psychology, Asia University, Taichung, Taiwan; Institute of Neuroscience, National Cheng-Chi University, Taipei, Taiwan
| | - Wan-Ju Cheng
- Department of Psychiatry, China Medical University Hospital, China Medical University, Taichung, Taiwan; Department of Public Health, China Medical University, Taichung, Taiwan; National Center for Geriatrics and Welfare Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chih-Chang Chao
- Institute of Neuroscience, National Cheng-Chi University, Taipei, Taiwan
| | - Chun-Hsien Kuo
- Department of Psychology, Asia University, Taichung, Taiwan; Center for Prevention and Treatment of Internet Addiction, Asia University, Taichung, Taiwan
| | - Ruey-Ming Liao
- Department of Psychology, Asia University, Taichung, Taiwan; Institute of Neuroscience, National Cheng-Chi University, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
2
|
Sreelatha I, Choi GY, Lee IS, Inturu O, Lee HS, Park YN, Lee CW, Yang I, Maeng S, Park JH. Neuroprotective Properties of Rutin Hydrate against Scopolamine-Induced Deficits in BDNF/TrkB/ERK/CREB/Bcl2 Pathways. Neurol Int 2024; 16:1094-1111. [PMID: 39452684 PMCID: PMC11510686 DOI: 10.3390/neurolint16050082] [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: 08/16/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Alzheimer's disease (AD) is an age-related degenerative brain disorder characterized by a progressive decline in cognitive function and memory. This study aimed to evaluate whether rutin hydrate (RH) has neuroprotective effects in an AD-like learning and memory impairment rat model induced by scopolamine (SCO). Methods: The rats were administered with RH (100 mg/kg) and SCO (1.5 mg/kg) and underwent behavioral tests, including the Morris water maze test, Y-maze test, and passive avoidance test, to evaluate their learning and memory abilities. Additionally, long-term potentiation (LTP) was induced to observe changes in the field excitatory postsynaptic potential (fEPSP) activity. Results: RH treatment attenuated the SCO-induced shortening of step-through latency in the passive avoidance (PA) test, increased the percentage of alternation in the Y-maze, and increased the time spent in the target zone in the Morris water maze (MWM). Moreover, RH increased the total activity of fEPSP following theta burst stimulation and attenuated the SCO-induced blockade of fEPSP. RH also ameliorated the SCO-induced decrease in the expression levels of the BDNF, TrkB, ERK, CREB, and Bcl-2 proteins and the increase in the Bax protein level in the rat hippocampus. This demonstrates that RH has beneficial neuroprotective effects in the brain, improving learning, memory, and synaptic plasticity in rats. Conclusions: Our results highlight the molecular and cellular mechanisms through which RH exerts its neuroprotective effects in the prevention and treatment of learning and memory deficit disorders. RH could potentially be used as a therapeutic strategy for the restoration of learning and memory function and the prevention of the progression of AD.
Collapse
Affiliation(s)
- Inturu Sreelatha
- Department of Gerontology (AgeTech-Service Convergence Major), Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea; (I.S.); (I.-S.L.); (S.M.)
| | - Ga-Young Choi
- Center for Research Equipment, Korea Basic Science Institute, Cheongju 28119, Republic of Korea;
| | - In-Seo Lee
- Department of Gerontology (AgeTech-Service Convergence Major), Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea; (I.S.); (I.-S.L.); (S.M.)
| | - Omkaram Inturu
- Department of Applied Physics and Institute of Natural Sciences, Kyung Hee University, Yongin 17104, Republic of Korea;
| | - Hyun-Sook Lee
- Department of East-West Medicine, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea; (H.-S.L.); (Y.-N.P.); (I.Y.)
| | - Yea-Na Park
- Department of East-West Medicine, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea; (H.-S.L.); (Y.-N.P.); (I.Y.)
| | - Cheol-Won Lee
- Convergence Healthcare Research Institute, Myong Ji University, Yongin 17058, Republic of Korea;
| | - Inkyou Yang
- Department of East-West Medicine, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea; (H.-S.L.); (Y.-N.P.); (I.Y.)
| | - Sungho Maeng
- Department of Gerontology (AgeTech-Service Convergence Major), Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea; (I.S.); (I.-S.L.); (S.M.)
| | - Ji-Ho Park
- Department of Gerontology (AgeTech-Service Convergence Major), Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea; (I.S.); (I.-S.L.); (S.M.)
| |
Collapse
|
3
|
Isensee J, van Cann M, Despang P, Araldi D, Moeller K, Petersen J, Schmidtko A, Matthes J, Levine JD, Hucho T. Depolarization induces nociceptor sensitization by CaV1.2-mediated PKA-II activation. J Cell Biol 2021; 220:212600. [PMID: 34431981 PMCID: PMC8404467 DOI: 10.1083/jcb.202002083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/14/2021] [Accepted: 08/05/2021] [Indexed: 01/20/2023] Open
Abstract
Depolarization drives neuronal plasticity. However, whether depolarization drives sensitization of peripheral nociceptive neurons remains elusive. By high-content screening (HCS) microscopy, we revealed that depolarization of cultured sensory neurons rapidly activates protein kinase A type II (PKA-II) in nociceptors by calcium influx through CaV1.2 channels. This effect was modulated by calpains but insensitive to inhibitors of cAMP formation, including opioids. In turn, PKA-II phosphorylated Ser1928 in the distal C terminus of CaV1.2, thereby increasing channel gating, whereas dephosphorylation of Ser1928 involved the phosphatase calcineurin. Patch-clamp and behavioral experiments confirmed that depolarization leads to calcium- and PKA-dependent sensitization of calcium currents ex vivo and local peripheral hyperalgesia in the skin in vivo. Our data suggest a local activity-driven feed-forward mechanism that selectively translates strong depolarization into further activity and thereby facilitates hypersensitivity of nociceptor terminals by a mechanism inaccessible to opioids.
Collapse
Affiliation(s)
- Jörg Isensee
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Marianne van Cann
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Patrick Despang
- Department of Pharmacology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Dioneia Araldi
- Division of Neuroscience, Departments of Medicine and Oral & Maxillofacial Surgery, University of California, San Francisco, San Francisco, CA
| | - Katharina Moeller
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Jonas Petersen
- Institute for Pharmacology and Clinical Pharmacy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Achim Schmidtko
- Institute for Pharmacology and Clinical Pharmacy, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jan Matthes
- Department of Pharmacology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Jon D Levine
- Division of Neuroscience, Departments of Medicine and Oral & Maxillofacial Surgery, University of California, San Francisco, San Francisco, CA
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
Signalling pathways contributing to learning and memory deficits in the Ts65Dn mouse model of Down syndrome. Neuronal Signal 2021; 5:NS20200011. [PMID: 33763235 PMCID: PMC7955101 DOI: 10.1042/ns20200011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 01/30/2023] Open
Abstract
Down syndrome (DS) is a genetic trisomic disorder that produces life-long changes in physiology and cognition. Many of the changes in learning and memory seen in DS are reminiscent of disorders involving the hippocampal/entorhinal circuit. Mouse models of DS typically involve trisomy of murine chromosome 16 is homologous for many of the genes triplicated in human trisomy 21, and provide us with good models of changes in, and potential pharmacotherapy for, human DS. Recent careful dissection of the Ts65Dn mouse model of DS has revealed differences in key signalling pathways from the basal forebrain to the hippocampus and associated rhinal cortices, as well as changes in the microstructure of the hippocampus itself. In vivo behavioural and electrophysiological studies have shown that Ts65Dn animals have difficulties in spatial memory that mirror hippocampal deficits, and have changes in hippocampal electrophysiological phenomenology that may explain these differences, and align with expectations generated from in vitro exploration of this model. Finally, given the existing data, we will examine the possibility for pharmacotherapy for DS, and outline the work that remains to be done to fully understand this system.
Collapse
|
5
|
Chen W, Jiang L, Hu Y, Tang N, Liang N, Li XF, Chen YW, Qin H, Wu L. Ferritin reduction is essential for cerebral ischemia-induced hippocampal neuronal death through p53/SLC7A11-mediated ferroptosis. Brain Res 2021; 1752:147216. [PMID: 33333054 DOI: 10.1016/j.brainres.2020.147216] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/02/2020] [Accepted: 11/25/2020] [Indexed: 12/23/2022]
Abstract
Cerebral ischemia is the most common cause of hippocampal neuronal death and the most prevalent cause of stroke with high mortality rate. Ferroptosis has been suggested to affect the role of hippocampal neurons. This study explores the influence of lentivirus infection-induced ferritin overexpression in hippocampal neuronal injury and death through simulations in August Copenhagen Irish rat models. Twenty-four-hour cerebral ischemia-reperfusion injury was induced in the rats after 90-min middle cerebral artery occlusion (MCAO). Ferritin overexpression was induced through lentivirus infection. The Morris Water Maze (MWM) test and tau hyperphosphorylation test were performed on hippocampal neurons to establish a MCAO model. The effect of ferritin overexpression on hippocampal neuronal death was evaluated using hematoxylin-eosin staining and annexin V/propidium iodide flow cytometry. The MWM test revealed that MCAO modeling decreased the cognitive and locomotor capacity of the rats, whereas ferritin overexpression partially reversed the effect of MCAO. In addition, the hyperphosphorylation of tau caused by MCAO was reduced by ferritin. Pathogenic changes, impaired viability, increased apoptosis, and elevated caspase-9 cleavage in hippocampal neurons were clearly recovered by ferritin. Moreover, robust reactive oxygen species production and glutathione consumption, which was induced by MCAO modeling, were ameliorated by ferritin. Furthermore, two key modulators of ferroptosis, p53 and SLC7A11, were demonstrated to be upregulated by MCAO modeling and downregulated by ferritin. Ferritin reduction is essential for cerebral ischemia-induced hippocampal neuronal ferroptosis mediated via p53 and SLC7A11.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Lingfei Jiang
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China; Graduate College of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Yueqiang Hu
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Nong Tang
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| | - Ni Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xing-Feng Li
- Graduate College of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Ye-Wen Chen
- Graduate College of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Hongling Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Lin Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi, China; Scientific Laboratorial Centre Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| |
Collapse
|
6
|
Wang Y, Liu Y, Bi X, Baudry M. Calpain-1 and Calpain-2 in the Brain: New Evidence for a Critical Role of Calpain-2 in Neuronal Death. Cells 2020; 9:E2698. [PMID: 33339205 PMCID: PMC7765587 DOI: 10.3390/cells9122698] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 01/24/2023] Open
Abstract
Calpains are a family of soluble calcium-dependent proteases that are involved in multiple regulatory pathways. Our laboratory has focused on the understanding of the functions of two ubiquitous calpain isoforms, calpain-1 and calpain-2, in the brain. Results obtained over the last 30 years led to the remarkable conclusion that these two calpain isoforms exhibit opposite functions in the brain. Calpain-1 activation is required for certain forms of synaptic plasticity and corresponding types of learning and memory, while calpain-2 activation limits the extent of plasticity and learning. Calpain-1 is neuroprotective both during postnatal development and in adulthood, while calpain-2 is neurodegenerative. Several key protein targets participating in these opposite functions have been identified and linked to known pathways involved in synaptic plasticity and neuroprotection/neurodegeneration. We have proposed the hypothesis that the existence of different PDZ (PSD-95, DLG and ZO-1) binding domains in the C-terminal of calpain-1 and calpain-2 is responsible for their association with different signaling pathways and thereby their different functions. Results with calpain-2 knock-out mice or with mice treated with a selective calpain-2 inhibitor indicate that calpain-2 is a potential therapeutic target in various forms of neurodegeneration, including traumatic brain injury and repeated concussions.
Collapse
Affiliation(s)
- Yubin Wang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (Y.W.); (Y.L.)
| | - Yan Liu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (Y.W.); (Y.L.)
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (Y.W.); (Y.L.)
| |
Collapse
|
7
|
The role of the Arp2/3 complex in shaping the dynamics and structures of branched actomyosin networks. Proc Natl Acad Sci U S A 2020; 117:10825-10831. [PMID: 32354995 DOI: 10.1073/pnas.1922494117] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Actomyosin networks give cells the ability to move and divide. These networks contract and expand while being driven by active energy-consuming processes such as motor protein walking and actin polymerization. Actin dynamics is also regulated by actin-binding proteins, such as the actin-related protein 2/3 (Arp2/3) complex. This complex generates branched filaments, thereby changing the overall organization of the network. In this work, the spatiotemporal patterns of dynamical actin assembly accompanying the branching-induced reorganization caused by Arp2/3 were studied using a computational model (mechanochemical dynamics of active networks [MEDYAN]); this model simulates actomyosin network dynamics as a result of chemical reactions whose rates are modulated by rapid mechanical equilibration. We show that branched actomyosin networks relax significantly more slowly than do unbranched networks. Also, branched networks undergo rare convulsive movements, "avalanches," that release strain in the network. These avalanches are associated with the more heterogeneous distribution of mechanically linked filaments displayed by branched networks. These far-from-equilibrium events arising from the marginal stability of growing actomyosin networks provide a possible mechanism of the "cytoquakes" recently seen in experiments.
Collapse
|
8
|
Liu X, Cui Y, Li X, Yang H. In-depth transcriptomic and proteomic analyses of the hippocampus and cortex in a rat model after cerebral ischemic injury and repair by Shuxuetong (SXT) injection. JOURNAL OF ETHNOPHARMACOLOGY 2020; 249:112362. [PMID: 31676400 DOI: 10.1016/j.jep.2019.112362] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/29/2019] [Accepted: 10/26/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND There is a lack of systematic descriptions and characterization of strokes and their effects in both the cerebral hippocampus and cortex. Shuxuetong (SXT) injection was reported to have good therapeutic effects in the clinic; therefore, it was selected as a drug intervention method for cerebral ischemia repair in rat models. The aim of this study was to understand the features of molecules and pathways and to reveal key processes of SXT repair. MATERIALS AND METHODS Evaluation of neurological deficit and infarct volume measurement was used to estimate the pharmacological effects of SXT injection on Ischemia-reperfusion(I/R) model rats. LC-MS/MS and RNA-Seq analysis were used to analyze the proteins and mRNA expression in the cerebral hippocampus and cortex 6 h and 24 h after ischemic injury and repair. A label-free approach (IBAQ) for proteomics analysis and FPKM based on gene read count for transcriptomics analysis were used to quantify the differences among the three experimental groups (Sham, Model and SXT-treated groups). Transcriptomics and proteomics analyses were verified by RT-qPCR and western blotting. RESULTS By combining LC-MS/MS and RNA-Seq, eight larger datasets (two time points and two tissues) were confidently identified in more than three biological replicates. An average of 4500 unique proteins and 8200 protein-coding genes were confidently identified. By combining the subcellular localization, hierarchical clustering, pathway enrichment analysis in the injury and repair phase, six core proteins and related genes that were significantly expressed were verified as candidates for cerebral ischemic injury by western blotting and quantitative real-time PCR. Meanwhile, the results indicated that there was better expression in the 6 h group by significant proteomics analysis during the development and progression of cerebral ischemia. Two primary co-enriched pathways, the PI3K-AKT and MAPK signaling pathways, and six related core candidates may play key roles in molecular mechanisms related to cerebral ischemic injury and repair by SXT injection. CONCLUSION Our data not only identified six core candidates and two key signaling pathways for cerebral ischemic injury and verification but also provided evidence for the explanation, prevention and treatment of cerebral ischemia by SXT injection. The results of the present study provide evidence for the explanation, prevention and treatment of cerebral ischemia by SXT injection.
Collapse
Affiliation(s)
- Xin Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yiran Cui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
9
|
Kastellakis G, Poirazi P. Synaptic Clustering and Memory Formation. Front Mol Neurosci 2019; 12:300. [PMID: 31866824 PMCID: PMC6908852 DOI: 10.3389/fnmol.2019.00300] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/25/2019] [Indexed: 01/12/2023] Open
Abstract
In the study of memory engrams, synaptic memory allocation is a newly emerged theme that focuses on how specific synapses are engaged in the storage of a given memory. Cumulating evidence from imaging and molecular experiments indicates that the recruitment of synapses that participate in the encoding and expression of memory is neither random nor uniform. A hallmark observation is the emergence of groups of synapses that share similar response properties and/or similar input properties and are located within a stretch of a dendritic branch. This grouping of synapses has been termed "synapse clustering" and has been shown to emerge in many different memory-related paradigms, as well as in in vitro studies. The clustering of synapses may emerge from synapses receiving similar input, or via many processes which allow for cross-talk between nearby synapses within a dendritic branch, leading to cooperative plasticity. Clustered synapses can act in concert to maximally exploit the nonlinear integration potential of the dendritic branches in which they reside. Their main contribution is to facilitate the induction of dendritic spikes and dendritic plateau potentials, which provide advanced computational and memory-related capabilities to dendrites and single neurons. This review focuses on recent evidence which investigates the role of synapse clustering in dendritic integration, sensory perception, learning, and memory as well as brain dysfunction. We also discuss recent theoretical work which explores the computational advantages provided by synapse clustering, leading to novel and revised theories of memory. As an eminent phenomenon during memory allocation, synapse clustering both shapes memory engrams and is also shaped by the parallel plasticity mechanisms upon which it relies.
Collapse
Affiliation(s)
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
| |
Collapse
|
10
|
Morató X, Luján R, Gonçalves N, Watanabe M, Altafaj X, Carvalho AL, Fernández-Dueñas V, Cunha RA, Ciruela F. Metabotropic glutamate type 5 receptor requires contactin-associated protein 1 to control memory formation. Hum Mol Genet 2019; 27:3528-3541. [PMID: 30010864 DOI: 10.1093/hmg/ddy264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/09/2018] [Indexed: 12/31/2022] Open
Abstract
The hippocampus is a key brain region for memory formation. Metabotropic glutamate type 5 receptors (mGlu5R) are strongly expressed in CA1 pyramidal neurons and fine-tune synaptic plasticity. Accordingly, mGlu5R pharmacological manipulation may represent an attractive therapeutic strategy to manage hippocampal-related neurological disorders. Here, by means of a membrane yeast two-hybrid screening, we identified contactin-associated protein 1 (Caspr1), a type I transmembrane protein member of the neurexin family, as a new mGlu5R partner. We report that mGlu5R and Caspr1 co-distribute and co-assemble both in heterologous expression systems and in rat brain. Furthermore, downregulation of Caspr1 in rat hippocampal primary cultures decreased mGlu5R-mediated signaling. Finally, silencing Caspr1 expression in the hippocampus impaired the impact of mGlu5R on spatial memory. Our results indicate that Caspr1 plays a pivotal role controlling mGlu5R function in hippocampus-dependent memory formation. Hence, this new protein-protein interaction may represent novel target for neurological disorders affecting hippocampal glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Xavier Morató
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Luján
- IDINE, Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Nélio Gonçalves
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - Xavier Altafaj
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Ana Luísa Carvalho
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Víctor Fernández-Dueñas
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Rodrigo A Cunha
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
Wilkinson ST, Holtzheimer PE, Gao S, Kirwin DS, Price RB. Leveraging Neuroplasticity to Enhance Adaptive Learning: The Potential for Synergistic Somatic-Behavioral Treatment Combinations to Improve Clinical Outcomes in Depression. Biol Psychiatry 2019; 85:454-465. [PMID: 30528745 PMCID: PMC6380941 DOI: 10.1016/j.biopsych.2018.09.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/30/2018] [Accepted: 09/11/2018] [Indexed: 12/17/2022]
Abstract
Until recently, therapeutic development in psychiatry was targeted solely toward symptom reduction. While this is a worthwhile goal, it has yielded little progress in improved therapeutics in the last several decades in the field of mood disorders. Recent advancements in our understanding of pathophysiology suggests that an impairment of neuroplasticity may be a critical part of the development of neuropsychiatric disorders. Interventions that enhance or modulate neuroplasticity often reduce depressive symptoms when applied as stand-alone treatments. Unfortunately, when treatments are discontinued, the disease state often returns as patients relapse. However, treatments that enhance or modulate plasticity not only reduce symptom burden, but also may provide an opportune window wherein cognitive or behavioral interventions could be introduced to harness a state of enhanced neuroplasticity and lead to improved longer-term clinical outcomes. Here, we review the potential of synergistically combining plasticity-enhancing and behavioral therapies to develop novel translational treatment approaches for depression. After reviewing relevant neuroplasticity deficits in depression, we survey biological treatments that appear to reverse such deficits in humans, including N-methyl-D-aspartate receptor modulators (ketamine, D-cycloserine), electroconvulsive therapy, and transcranial brain stimulation. We then review evidence that either directly or indirectly supports the hypothesis that a robust enhancement of neuroplasticity through these methods might promote the uptake of cognitive and behavioral interventions to enhance longer-term treatment outcomes through a synergistic effect. We identify key missing pieces of evidence and discuss future directions to enhance this emerging line of research.
Collapse
Affiliation(s)
- Samuel T. Wilkinson
- Department of Psychiatry, Yale School of Medicine and Yale Psychiatric Hospital, New Haven, Connecticut
| | - Paul E. Holtzheimer
- National Center for PTSD, Executive Division, White River Junction VA Medical Center, White River Junction, Vermont;,Department of Psychiatry and Surgery, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Shan Gao
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David S. Kirwin
- Department of Psychiatry, Yale School of Medicine and Yale Psychiatric Hospital, New Haven, Connecticut
| | - Rebecca B. Price
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Kelley P, Evans MDR, Kelley J. Making Memories: Why Time Matters. Front Hum Neurosci 2018; 12:400. [PMID: 30386221 PMCID: PMC6198140 DOI: 10.3389/fnhum.2018.00400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 09/18/2018] [Indexed: 11/29/2022] Open
Abstract
In the last decade advances in human neuroscience have identified the critical importance of time in creating long-term memories. Circadian neuroscience has established biological time functions via cellular clocks regulated by photosensitive retinal ganglion cells and the suprachiasmatic nuclei. Individuals have different circadian clocks depending on their chronotypes that vary with genetic, age, and sex. In contrast, social time is determined by time zones, daylight savings time, and education and employment hours. Social time and circadian time differences can lead to circadian desynchronization, sleep deprivation, health problems, and poor cognitive performance. Synchronizing social time to circadian biology leads to better health and learning, as demonstrated in adolescent education. In-day making memories of complex bodies of structured information in education is organized in social time and uses many different learning techniques. Research in the neuroscience of long-term memory (LTM) has demonstrated in-day time spaced learning patterns of three repetitions of information separated by two rest periods are effective in making memories in mammals and humans. This time pattern is based on the intracellular processes required in synaptic plasticity. Circadian desynchronization, sleep deprivation, and memory consolidation in sleep are less well-understood, though there has been considerable progress in neuroscience research in the last decade. The interplay of circadian, in-day and sleep neuroscience research are creating an understanding of making memories in the first 24-h that has already led to interventions that can improve health and learning.
Collapse
Affiliation(s)
- Paul Kelley
- Sleep, Circadian and Memory Neuroscience, The Open University, Milton Keynes, United Kingdom
| | - M. D. R. Evans
- Sociology and Applied Statistics Program, University of Nevada, Reno, Reno, NV, United States
- Sociology, University of Nevada, Reno, Reno, NV, United States
| | - Jonathan Kelley
- Sociology, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
13
|
Saavedra A, Ballesteros JJ, Tyebji S, Martínez-Torres S, Blázquez G, López-Hidalgo R, Azkona G, Alberch J, Martín ED, Pérez-Navarro E. Proteolytic Degradation of Hippocampal STEP 61 in LTP and Learning. Mol Neurobiol 2018; 56:1475-1487. [PMID: 29948948 DOI: 10.1007/s12035-018-1170-1] [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: 04/13/2018] [Accepted: 05/31/2018] [Indexed: 10/14/2022]
Abstract
Striatal-enriched protein tyrosine phosphatase (STEP) modulates key signaling molecules involved in synaptic plasticity and neuronal function. It is postulated that STEP opposes the development of long-term potentiation (LTP) and that it exerts a restraint on long-term memory (LTM). Here, we examined whether STEP61 levels are regulated during hippocampal LTP and after training in hippocampal-dependent tasks. We found that after inducing LTP by high frequency stimulation or theta-burst stimulation STEP61 levels were significantly reduced, with a concomitant increase of STEP33 levels, a product of calpain cleavage. Importantly, inhibition of STEP with TC-2153 improved LTP in hippocampal slices. Moreover, we observed that after training in the passive avoidance and the T-maze spontaneous alternation task, hippocampal STEP61 levels were significantly reduced, but STEP33 levels were unchanged. Yet, hippocampal BDNF content and TrkB levels were increased in trained mice, and it is known that BDNF promotes STEP degradation through the proteasome. Accordingly, hippocampal pTrkBTyr816, pPLCγTyr783, and protein ubiquitination levels were increased in T-SAT trained mice. Remarkably, injection of the TrkB antagonist ANA-12 (2 mg/Kg, but not 0.5 mg/Kg) elicited LTM deficits and promoted STEP61 accumulation in the hippocampus. Also, STEP knockout mice outperformed wild-type animals in an age- and test-dependent manner. Summarizing, STEP61 undergoes proteolytic degradation in conditions leading to synaptic strengthening and memory formation, thus highlighting its role as a molecular constrain, which is removed to enable the activation of pathways important for plasticity processes.
Collapse
Affiliation(s)
- Ana Saavedra
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús J Ballesteros
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shiraz Tyebji
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
| | - Sara Martínez-Torres
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Gloria Blázquez
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rosa López-Hidalgo
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Grupo de Patología Celular y Molecular del Alcohol, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Garikoitz Azkona
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eduardo D Martín
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Instituto Cajal, CSIC, Madrid, Spain
| | - Esther Pérez-Navarro
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
14
|
Cui Y, Liu X, Li X, Yang H. In-Depth Proteomic Analysis of the Hippocampus in a Rat Model after Cerebral Ischaemic Injury and Repair by Danhong Injection (DHI). Int J Mol Sci 2017; 18:ijms18071355. [PMID: 28672812 PMCID: PMC5535848 DOI: 10.3390/ijms18071355] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/17/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023] Open
Abstract
Stroke is the second most common cause of death worldwide. A systematic description and characterization of the strokes and the effects induced in the hippocampus have not been performed so far. Here, we analysed the protein expression in the hippocampus 24 h after cerebral ischaemic injury and repair. Drug intervention using Danhong injection (DHI), which has been reported to have good therapeutic effects in a clinical setting, was selected for our study of cerebral ischaemia repair in rat models. A larger proteome dataset and total 4091 unique proteins were confidently identified in three biological replicates by combining tissue extraction for rat hippocampus and LC-MS/MS analysis. A label-free approach was then used to quantify the differences among the four experimental groups (Naive, Sham, middle cerebral artery occlusion (MCAO) and MCAO + DHI groups) and showed that about 2500 proteins on average were quantified in each of the experiment group. Bioinformatics analysis revealed that in total 280 unique proteins identified above were differentially expressed (P < 0.05). By combining the subcellular localization, hierarchical clustering and pathway information with the results from injury and repair phase, 12 significant expressed proteins were chosen and verified with respect to their potential as candidates for cerebral ischaemic injury by Western blot. The primary three signalling pathways of the candidates related may be involved in molecular mechanisms related to cerebral ischaemic injury. In addition, a glycogen synthase kinase-3β (Gsk-3β) inhibitor of the candidates with the best corresponding expression trends between western blotting (WB) and label-free quantitative results were chosen for further validation. The results of Western blot analysis of protein expression and 2,3,5- chloride three phenyl tetrazole (TTC) staining of rat brains showed that DHI treatment and Gsk-3β inhibitor are both able to confer protection against ischaemic injury in rat MCAO model. The observations of the present study provide a novel understanding regarding the regulatory mechanism of cerebral ischaemic injury.
Collapse
Affiliation(s)
- Yiran Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700 Beijing, China.
| | - Xin Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700 Beijing, China.
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, 100700 Beijing, China.
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700 Beijing, China.
| |
Collapse
|
15
|
Differential role of calpain-dependent protein cleavage in intermediate and long-term operant memory in Aplysia. Neurobiol Learn Mem 2016; 137:134-141. [PMID: 27913293 DOI: 10.1016/j.nlm.2016.11.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/28/2016] [Accepted: 11/29/2016] [Indexed: 01/06/2023]
Abstract
In addition to protein synthesis, protein degradation or protein cleavage may be necessary for intermediate (ITM) and long-term memory (LTM) to remove molecular constraints, facilitate persistent kinase activity and modulate synaptic plasticity. Calpains, a family of conserved calcium dependent cysteine proteases, modulate synaptic function through protein cleavage. We used the marine mollusk Aplysia californica to investigate the in vivo role of calpains during intermediate and long-term operant memory formation using the learning that food is inedible (LFI) paradigm. A single LFI training session, in which the animal associates a specific netted seaweed with the failure to swallow, generates short (30min), intermediate (4-6h) and long-term (24h) memory. Using the calpain inhibitors calpeptin and MDL-28170, we found that ITM requires calpain activity for induction and consolidation similar to the previously reported requirements for persistent protein kinase C activity in intermediate-term LFI memory. The induction of LTM also required calpain activity. In contrast to ITM, calpain activity was not necessary for the molecular consolidation of LTM. Surprisingly, six hours after LFI training we found that calpain activity was necessary for LTM, although this is a time at which neither persistent PKC activity nor protein synthesis is required for the maintenance of long-term LFI memory. These results demonstrate that calpains function in multiple roles in vivo during associative memory formation.
Collapse
|
16
|
Tan WH, Bird LM. Angelman syndrome: Current and emerging therapies in 2016. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2016; 172:384-401. [PMID: 27860204 DOI: 10.1002/ajmg.c.31536] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a loss of the maternally-inherited UBE3A; the paternal UBE3A is silenced in neurons by a mechanism involving an antisense transcript (UBE3A-AS) at the unmethylated paternal locus. We reviewed all published information on the clinical trials that have been completed as well as the publicly available information on ongoing trials of therapies in AS. To date, all clinical trials that strove to improve neurodevelopment in AS have been unsuccessful. Attempts at hypermethylating the maternal locus through dietary compounds were ineffective. The results of an 8-week open-label trial using minocycline as a matrix metalloproteinase-9 inhibitor were inconclusive, while a subsequent randomized placebo-controlled trial suggested that treatment with minocycline for 8 weeks did not result in any neurodevelopmental gains. A 1-year randomized placebo-controlled trial using levodopa to alter the phosphorylation of calcium/calmodulin-dependent kinase II did not lead to any improvement in neurodevelopment. Topoisomerase inhibitors and antisense oligonucleotides are being developed to directly inhibit UBE3A-AS. Artificial transcription factors are being developed to "super activate" UBE3A or inhibit UBE3A-AS. Other strategies targeting specific pathways are briefly discussed. We also reviewed the medications that are currently used to treat seizures and sleep disturbances, which are two of the more common complications of AS. © 2016 Wiley Periodicals, Inc.
Collapse
|
17
|
|
18
|
Chen X, Du YM, Xu F, Liu D, Wang YL. Propofol Prevents Hippocampal Neuronal Loss and Memory Impairment in Cerebral Ischemia Injury Through Promoting PTEN Degradation. J Mol Neurosci 2016; 60:63-70. [DOI: 10.1007/s12031-016-0791-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 06/28/2016] [Indexed: 01/07/2023]
|
19
|
Abstract
Fear memory is the best-studied form of memory. It was thoroughly investigated in the past 60 years mostly using two classical conditioning procedures (contextual fear conditioning and fear conditioning to a tone) and one instrumental procedure (one-trial inhibitory avoidance). Fear memory is formed in the hippocampus (contextual conditioning and inhibitory avoidance), in the basolateral amygdala (inhibitory avoidance), and in the lateral amygdala (conditioning to a tone). The circuitry involves, in addition, the pre- and infralimbic ventromedial prefrontal cortex, the central amygdala subnuclei, and the dentate gyrus. Fear learning models, notably inhibitory avoidance, have also been very useful for the analysis of the biochemical mechanisms of memory consolidation as a whole. These studies have capitalized on in vitro observations on long-term potentiation and other kinds of plasticity. The effect of a very large number of drugs on fear learning has been intensively studied, often as a prelude to the investigation of effects on anxiety. The extinction of fear learning involves to an extent a reversal of the flow of information in the mentioned structures and is used in the therapy of posttraumatic stress disorder and fear memories in general.
Collapse
Affiliation(s)
- Ivan Izquierdo
- National Institute of Translational Neuroscience, National Research Council of Brazil, and Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristiane R. G. Furini
- National Institute of Translational Neuroscience, National Research Council of Brazil, and Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jociane C. Myskiw
- National Institute of Translational Neuroscience, National Research Council of Brazil, and Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
20
|
Calpain-1 and Calpain-2: The Yin and Yang of Synaptic Plasticity and Neurodegeneration. Trends Neurosci 2016; 39:235-245. [PMID: 26874794 DOI: 10.1016/j.tins.2016.01.007] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 01/09/2023]
Abstract
Many signaling pathways participate in both synaptic plasticity and neuronal degeneration. While calpains participate in these phenomena, very few studies have evaluated the respective roles of the two major calpain isoforms in the brain, calpain-1 and calpain-2. We review recent studies indicating that calpain-1 and calpain-2 exhibit opposite functions in both synaptic plasticity and neurodegeneration. Calpain-1 activation is required for the induction of long-term potentiation (LTP) and is generally neuroprotective, while calpain-2 activation limits the extent of potentiation and is neurodegenerative. This duality of functions is related to their associations with different PDZ-binding proteins, resulting in differential subcellular localization, and offers new therapeutic opportunities for a number of indications in which these proteases have previously been implicated.
Collapse
|
21
|
Eom HS, Park HR, Jo SK, Kim YS, Moon C, Kim SH, Jung U. Ionizing Radiation Induces Altered Neuronal Differentiation by mGluR1 through PI3K-STAT3 Signaling in C17.2 Mouse Neural Stem-Like Cells. PLoS One 2016; 11:e0147538. [PMID: 26828720 PMCID: PMC4734671 DOI: 10.1371/journal.pone.0147538] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 01/04/2016] [Indexed: 01/02/2023] Open
Abstract
Most studies of IR effects on neural cells and tissues in the brain are still focused on loss of neural stem cells. On the other hand, the effects of IR on neuronal differentiation and its implication in IR-induced brain damage are not well defined. To investigate the effects of IR on C17.2 mouse neural stem-like cells and mouse primary neural stem cells, neurite outgrowth and expression of neuronal markers and neuronal function-related genes were examined. To understand this process, the signaling pathways including PI3K, STAT3, metabotrophic glutamate receptor 1 (mGluR1) and p53 were investigated. In C17.2 cells, irradiation significantly increased the neurite outgrowth, a morphological hallmark of neuronal differentiation, in a dose-dependent manner. Also, the expression levels of neuronal marker proteins, β-III tubulin were increased by IR. To investigate whether IR-induced differentiation is normal, the expression of neuronal function-related genes including synaptophysin, a synaptic vesicle forming proteins, synaptotagmin1, a calcium ion sensor, γ-aminobutyric acid (GABA) receptors, inhibitory neurotransmitter receptors and glutamate receptors, excitatory neurotransmitter receptors was examined and compared to that of neurotrophin-stimulated differentiation. IR increased the expression of synaptophysin, synaptotagmin1 and GABA receptors mRNA similarly to normal differentiation by stimulation of neurotrophin. Interestingly, the overall expression of glutamate receptors was significantly higher in irradiated group than normal differentiation group, suggesting that the IR-induced neuronal differentiation may cause altered neuronal function in C17.2 cells. Next, the molecular mechanism of the altered neuronal differentiation induced by IR was studied by investigating signaling pathways including p53, mGluR1, STAT3 and PI3K. Increases of neurite outgrowth, neuronal marker and neuronal function-related gene expressions by IR were abolished by inhibition of p53, mGluR-1, STAT3 or PI3K. The inhibition of PI3K blocked both p53 signaling and STAT3-mGluR1 signaling but inhibition of p53 did not affect STAT3-mGluR1 signaling in irradiated C17.2 cells. Finally, these results of the IR-induced altered differentiation in C17.2 cells were verified in ex vivo experiments using mouse primary neural stem cells. In conclusion, the results of this study demonstrated that IR is able to trigger the altered neuronal differentiation in undifferentiated neural stem-like cells through PI3K-STAT3-mGluR1 and PI3K-p53 signaling. It is suggested that the IR-induced altered neuronal differentiation may play a role in the brain dysfunction caused by IR.
Collapse
Affiliation(s)
- Hyeon Soo Eom
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Hae Ran Park
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sung Kee Jo
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Sung-Ho Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Uhee Jung
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
22
|
Pharmacological therapies for Angelman syndrome. Wien Med Wochenschr 2016; 167:205-218. [PMID: 26758979 DOI: 10.1007/s10354-015-0408-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/15/2015] [Indexed: 12/16/2022]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a loss of the maternally inherited UBE3A; the paternal UBE3A is silenced in neurons by a mechanism involving an antisense transcript (UBE3A-AS). We reviewed the published information on clinical trials that have been completed as well as the publicly available information on ongoing trials of therapies for AS. Attempts at hypermethylating the maternal locus through dietary compounds were ineffective. The results of a clinical trial using minocycline as a matrix metalloproteinase-9 inhibitor were inconclusive; another clinical trial is underway. Findings from a clinical trial using L-dopa to alter phosphorylation of calcium/calmodulin-dependent kinase II are awaited. Topoisomerase inhibitors and antisense oligonucleotides are being developed to directly inhibit UBE3A-AS. Other strategies targeting specific pathways are briefly discussed. We also reviewed the medications that are currently used to treat seizures and sleep disturbances, which are two of the more debilitating manifestations of AS.
Collapse
|
23
|
Abstract
INTRODUCTION Angelman syndrome (AS) is a neurodevelopmental disorder caused by deficiency of maternally inherited UBE3A, an ubiquitin E3 ligase. Despite recent progress in understanding the mechanism underlying UBE3A imprinting, there is no effective treatment. Further investigation of the roles played by UBE3A in the central nervous system (CNS) is needed for developing effective therapies. AREA COVERED This review covers the literature related to genetic classifications of AS, recent discoveries regarding the regulation of UBE3A imprinting, alterations in cell signaling in various brain regions and potential therapeutic approaches. Since a large proportion of AS patients exhibit comorbid autism spectrum disorder (ASD), potential common molecular bases are discussed. EXPERT OPINION Advances in understanding UBE3A imprinting provide a unique opportunity to induce paternal UBE3A expression, thus targeting the syndrome at its 'root.' However, such efforts have yielded less-than-expected rescue effects in AS mouse models, raising the concern that activation of paternal UBE3A after a critical period cannot correct all the CNS defects that developed in a UBE3A-deficient environment. On the other hand, targeting abnormal downstream cell signaling pathways has provided promising rescue effects in preclinical research. Thus, combined reinstatement of paternal UBE3A expression with targeting abnormal signaling pathways should provide better therapeutic effects.
Collapse
Affiliation(s)
- Xiaoning Bi
- a Department of Basic Medical Sciences, COMP , Western University of Health Sciences , Pomona , CA , USA
| | - Jiandong Sun
- a Department of Basic Medical Sciences, COMP , Western University of Health Sciences , Pomona , CA , USA
| | - Angela X Ji
- a Department of Basic Medical Sciences, COMP , Western University of Health Sciences , Pomona , CA , USA
| | - Michel Baudry
- b Graduate College of Biomedical Sciences , Western University of Health Sciences , Pomona , CA , USA
| |
Collapse
|
24
|
Dillon DG. The neuroscience of positive memory deficits in depression. Front Psychol 2015; 6:1295. [PMID: 26441703 PMCID: PMC4561348 DOI: 10.3389/fpsyg.2015.01295] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/13/2015] [Indexed: 12/17/2022] Open
Abstract
Adults with unipolar depression typically show poor episodic memory for positive material, but the neuroscientific mechanisms responsible for this deficit have not been characterized. I suggest a simple hypothesis: weak memory for positive material in depression reflects disrupted communication between the mesolimbic dopamine pathway and medial temporal lobe (MTL) memory systems during encoding. This proposal draws on basic research showing that dopamine release in the hippocampus is critical for the transition from early- to late-phase long-term potentiation (LTP) that marks the conversion of labile, short-term memories into stable, long-term memories. Neuroimaging and pharmacological data from healthy humans paint a similar picture: activation of the mesolimbic reward circuit enhances encoding and boosts retention. Unipolar depression is characterized by anhedonia-loss of pleasure-and reward circuit dysfunction, which is believed to reflect negative effects of stress on the mesolimbic dopamine pathway. Thus, I propose that the MTL is deprived of strengthening reward signals in depressed adults and memory for positive events suffers accordingly. Although other mechanisms are important, this hypothesis holds promise as an explanation for positive memory deficits in depression.
Collapse
Affiliation(s)
- Daniel G Dillon
- Motivated Learning and Memory Laboratory, Center for Depression, Anxiety and Stress Research, McLean Hospital, Harvard Medical School , Belmont, MA, USA
| |
Collapse
|
25
|
Colciago A, Casati L, Negri-Cesi P, Celotti F. Learning and memory: Steroids and epigenetics. J Steroid Biochem Mol Biol 2015; 150:64-85. [PMID: 25766520 DOI: 10.1016/j.jsbmb.2015.02.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 12/19/2022]
Abstract
Memory formation and utilization is a complex process involving several brain structures in conjunction as the hippocampus, the amygdala and the adjacent cortical areas, usually defined as medial temporal lobe structures (MTL). The memory processes depend on the formation and modulation of synaptic connectivity affecting synaptic strength, synaptic plasticity and synaptic consolidation. The basic neurocognitive mechanisms of learning and memory are shortly recalled in the initial section of this paper. The effect of sex hormones (estrogens, androgens and progesterone) and of adrenocortical steroids on several aspects of memory processes are then analyzed on the basis of animal and human studies. A specific attention has been devoted to the different types of steroid receptors (membrane or nuclear) involved and on local metabolic transformations when required. The review is concluded by a short excursus on the steroid activated epigenetic mechanisms involved in memory formation.
Collapse
Affiliation(s)
- Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences, Section of Biomedicine and Endocrinology, Via Balzaretti 9, 20133 Milano, Italy
| | - Lavinia Casati
- Department of Medical Biotechnologies and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
| | - Paola Negri-Cesi
- Department of Pharmacological and Biomolecular Sciences, Section of Biomedicine and Endocrinology, Via Balzaretti 9, 20133 Milano, Italy
| | - Fabio Celotti
- Department of Pharmacological and Biomolecular Sciences, Section of Biomedicine and Endocrinology, Via Balzaretti 9, 20133 Milano, Italy
| |
Collapse
|
26
|
Kastellakis G, Cai DJ, Mednick SC, Silva AJ, Poirazi P. Synaptic clustering within dendrites: an emerging theory of memory formation. Prog Neurobiol 2015; 126:19-35. [PMID: 25576663 PMCID: PMC4361279 DOI: 10.1016/j.pneurobio.2014.12.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 12/29/2014] [Accepted: 12/29/2014] [Indexed: 11/30/2022]
Abstract
It is generally accepted that complex memories are stored in distributed representations throughout the brain, however the mechanisms underlying these representations are not understood. Here, we review recent findings regarding the subcellular mechanisms implicated in memory formation, which provide evidence for a dendrite-centered theory of memory. Plasticity-related phenomena which affect synaptic properties, such as synaptic tagging and capture, synaptic clustering, branch strength potentiation and spinogenesis provide the foundation for a model of memory storage that relies heavily on processes operating at the dendrite level. The emerging picture suggests that clusters of functionally related synapses may serve as key computational and memory storage units in the brain. We discuss both experimental evidence and theoretical models that support this hypothesis and explore its advantages for neuronal function.
Collapse
Affiliation(s)
- George Kastellakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology, Hellas (FORTH), P.O. Box 1385, GR 70013 Heraklion, Greece
| | - Denise J Cai
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, 2554 Gonda Center, Los Angeles, CA 90095, United States
| | - Sara C Mednick
- Department of Psychology, University of California, 900 University Avenue, Riverside, CA 92521, United States
| | - Alcino J Silva
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, 2554 Gonda Center, Los Angeles, CA 90095, United States
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology, Hellas (FORTH), P.O. Box 1385, GR 70013 Heraklion, Greece.
| |
Collapse
|
27
|
Ménard C, Gaudreau P, Quirion R. Signaling pathways relevant to cognition-enhancing drug targets. Handb Exp Pharmacol 2015; 228:59-98. [PMID: 25977080 DOI: 10.1007/978-3-319-16522-6_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aging is generally associated with a certain cognitive decline. However, individual differences exist. While age-related memory deficits can be observed in humans and rodents in the absence of pathological conditions, some individuals maintain intact cognitive functions up to an advanced age. The mechanisms underlying learning and memory processes involve the recruitment of multiple signaling pathways and gene expression, leading to adaptative neuronal plasticity and long-lasting changes in brain circuitry. This chapter summarizes the current understanding of how these signaling cascades could be modulated by cognition-enhancing agents favoring memory formation and successful aging. It focuses on data obtained in rodents, particularly in the rat as it is the most common animal model studied in this field. First, we will discuss the role of the excitatory neurotransmitter glutamate and its receptors, downstream signaling effectors [e.g., calcium/calmodulin-dependent protein kinase II (CaMKII), protein kinase C (PKC), extracellular signal-regulated kinases (ERK), mammalian target of rapamycin (mTOR), cAMP response element-binding protein (CREB)], associated immediate early gene (e.g., Homer 1a, Arc and Zif268), and growth factors [insulin-like growth factors (IGFs) and brain-derived neurotrophic factor (BDNF)] in synaptic plasticity and memory formation. Second, the impact of the cholinergic system and related modulators on memory will be briefly reviewed. Finally, since dynorphin neuropeptides have recently been associated with memory impairments in aging, it is proposed as an attractive target to develop novel cognition-enhancing agents.
Collapse
Affiliation(s)
- Caroline Ménard
- Douglas Mental Health University Institute, McGill University, Perry Pavilion, 6875 LaSalle Boulevard, Montreal, QC, Canada, H4H 1R3
| | | | | |
Collapse
|
28
|
Mercado III E. Learning-Related Synaptic Reconfiguration in Hippocampal Networks: Memory Storage or Waveguide Tuning? AIMS Neurosci 2015. [DOI: 10.3934/neuroscience.2015.1.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
29
|
Rudy JW. Variation in the persistence of memory: An interplay between actin dynamics and AMPA receptors. Brain Res 2014; 1621:29-37. [PMID: 25511990 DOI: 10.1016/j.brainres.2014.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 10/24/2022]
Abstract
William James noted that memories could persist from minutes to weeks. This essay attempts to explain this variation by situating the explanation in the biochemistry of dendritic spines. Two outcomes are critical to generate the synaptic basis of memory: (1) the actin cytoskeleton in the spine must be degraded to permit (2) additional AMPA receptors (GluA1s) to enter new "hot spots" in the postsynaptic density. These initial outcomes can support short-lasting memories. The threshold for these events is low but the underlying synaptic changes cannot resist the endocytic processes that remove the added AMPA receptors. For the memory to persist the degraded actin cytoskeleton must be rebuilt and the vacated "hot spots" refilled with GluA2 receptors. A primary claim is that it is the stabilization of an enlarged actin cytoskeleton that is the target outcome that consolidates the synaptic basis of memory (see Lynch et al., 2007). The stabilized actin cytoskeleton has properties that enable it to garner the synaptic proteins it needs to self sustain the potentiated state and to benefit from activation of memory modulation systems. This article is part of a Special Issue entitled Brain and Memory.
Collapse
Affiliation(s)
- Jerry W Rudy
- Department of Psychology and Neuroscience University of Colorado, Boulder, CO 80309, United States.
| |
Collapse
|
30
|
Rudy JW. Actin dynamics and the evolution of the memory trace. Brain Res 2014; 1621:17-28. [PMID: 25498985 DOI: 10.1016/j.brainres.2014.12.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 12/24/2022]
Abstract
The goal of this essay is to link the regulation of actin dynamics to the idea that the synaptic changes that support long-term potentiation and memory evolve in temporally overlapping stages-generation, stabilization, and consolidation. Different cellular/molecular processes operate at each stage to change the spine cytoarchitecture and, in doing so, alter its function. Calcium-dependent processes that degrade the actin cytoskeleton network promote a rapid insertion of AMPA receptors into the post synaptic density, which increases a spine's capacity to express a potentiated response to glutamate. Other post-translation events then begin to stabilize and expand the actin cytoskeleton by increasing the filament actin content of the spine and reorganizing it to be resistant to depolymerizing events. Disrupting actin polymerization during this stabilization period is a terminal event-the actin cytoskeleton shrinks and potentiated synapses de-potentiate and memories are lost. Late-arriving, new proteins may consolidate changes in the actin cytoskeleton. However, to do so requires a stabilized actin cytoskeleton. The now enlarged spine has properties that enable it to capture other newly transcribed mRNAs or their protein products and thus enable the synaptic changes that support LTP and memory to be consolidated and maintained. This article is part of a Special Issue entitled SI: Brain and Memory.
Collapse
Affiliation(s)
- Jerry W Rudy
- Department of Psychology and Neuroscience, University of Colorado, 345 UCB, Boulder, CO 80309, USA.
| |
Collapse
|
31
|
Multiple cellular cascades participate in long-term potentiation and in hippocampus-dependent learning. Brain Res 2014; 1621:73-81. [PMID: 25482663 DOI: 10.1016/j.brainres.2014.11.033] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 11/15/2014] [Indexed: 02/02/2023]
Abstract
Since its discovery by Bliss and Lomo, the phenomenon of long-term potentiation (LTP) has been extensively studied, as it was viewed as a potential cellular mechanism of learning and memory. Over the years, many signaling cascades have been implicated in its induction, consolidation and maintenance, raising questions regarding its real significance. Here, we review several of the most commonly studie signaling cascades and discuss how they converge on a common set of mechanisms likely to be involved in the maintenance of LTP. We further argue that the existence of cross-talks between these different signaling cascades can not only account for several discrepancies in the literature, but also account for the existence of different forms of LTP, which can be engaged by different types of stimulus parameters under different experimental conditions. Finally, we discuss how the understanding of the diversity of LTP mechanisms can help us understand the diversity of the types of learning and memory. This article is part of a Special Issue entitled SI: Brain and Memory.
Collapse
|
32
|
Galantamine prevents long-lasting suppression of excitatory synaptic transmission in CA1 pyramidal neurons of soman-challenged guinea pigs. Neurotoxicology 2014; 44:270-8. [PMID: 25064080 DOI: 10.1016/j.neuro.2014.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/08/2014] [Accepted: 07/15/2014] [Indexed: 12/27/2022]
Abstract
Galantamine, a drug currently approved for the treatment of Alzheimer's disease, has recently emerged as an effective pretreatment against the acute toxicity and delayed cognitive deficits induced by organophosphorus (OP) nerve agents, including soman. Since cognitive deficits can result from impaired glutamatergic transmission in the hippocampus, the present study was designed to test the hypothesis that hippocampal glutamatergic transmission declines following an acute exposure to soman and that this effect can be prevented by galantamine. To test this hypothesis, spontaneous excitatory postsynaptic currents (EPSCs) were recorded from CA1 pyramidal neurons in hippocampal slices obtained at 1h, 24h, or 6-9 days after guinea pigs were injected with: (i) 1×LD50 soman (26.3μg/kg, s.c.); (ii) galantamine (8mg/kg, i.m.) followed 30min later by 1×LD50 soman, (iii) galantamine (8mg/kg, i.m.), or (iv) saline (0.5ml/kg, i.m.). In soman-injected guinea pigs that were not pretreated with galantamine, the frequency of EPSCs was significantly lower than that recorded from saline-injected animals. There was no correlation between the severity of soman-induced acute toxicity and the magnitude of soman-induced reduction of EPSC frequency. Pretreatment with galantamine prevented the reduction of EPSC frequency observed at 6-9 days after the soman challenge. Prevention of soman-induced long-lasting reduction of hippocampal glutamatergic synaptic transmission may be an important determinant of the ability of galantamine to counter cognitive deficits that develop long after an acute exposure to the nerve agent.
Collapse
|
33
|
Ménard C, Quirion R, Bouchard S, Ferland G, Gaudreau P. Glutamatergic signaling and low prodynorphin expression are associated with intact memory and reduced anxiety in rat models of healthy aging. Front Aging Neurosci 2014; 6:81. [PMID: 24847259 PMCID: PMC4019859 DOI: 10.3389/fnagi.2014.00081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/16/2014] [Indexed: 11/13/2022] Open
Abstract
The LOU/C/Jall (LOU) rat strain is considered a model of healthy aging due to its increased longevity, maintenance of stable body weight (BW) throughout life and low incidence of age-related diseases. However, aging LOU rat cognitive and anxiety status has yet to be investigated. In the present study, male and female LOU rat cognitive performances (6-42 months) were assessed using novel object recognition and Morris Water Maze tasks. Recognition memory remained intact in all LOU rats up to 42 months of age. As for spatial memory, old LOU rat performed similarly as young animals for learning acquisition, reversal learning, and retention. While LOU rat BW remained stable despite aging, 20-month-old ad-libitum-fed (OAL) male Sprague Dawley rats become obese. We determined if long-term caloric restriction (LTCR) prevents age-related BW increase and cognitive deficits in this rat strain, as observed in the obesity-resistant LOU rats. Compared to young animals, recognition memory was impaired in OAL but intact in 20-month-old calorie-restricted (OCR) rats. Similarly, OAL spatial learning acquisition was impaired but LTCR prevented the deficits. Exacerbated stress responses may favor age-related cognitive decline. In the elevated plus maze and open field tasks, LOU and OCR rats exhibited high levels of exploratory activity whereas OAL rats displayed anxious behaviors. Expression of prodynorphin (Pdyn), an endogenous peptide involved in stress-related memory impairments, was increased in the hippocampus of OAL rats. Group 1 metabotropic glutamate receptor 5 and immediate early genes Homer 1a and Arc expression, both associated with successful cognitive aging, were unaltered in aging LOU rats but lower in OAL than OCR rats. Altogether, our results, supported by principal component analysis and correlation matrix, suggest that intact memory and low anxiety are associated with glutamatergic signaling and low Pdyn expression in the hippocampus of non-obese aging rats.
Collapse
Affiliation(s)
- Caroline Ménard
- Neuroscience Division, Douglas Mental Health University Institute Research Center Montreal, QC, Canada ; Department of Psychiatry, McGill University Montreal, QC, Canada ; Laboratory of Neuroendocrinology of Aging, Centre Hospitalier de l'Université de Montréal Research Center Montreal, QC, Canada ; Department of Medicine, University of Montreal Montreal, QC, Canada
| | - Rémi Quirion
- Neuroscience Division, Douglas Mental Health University Institute Research Center Montreal, QC, Canada ; Department of Psychiatry, McGill University Montreal, QC, Canada
| | - Sylvain Bouchard
- Faculty of Medicine, University of Montreal Montreal, QC, Canada
| | - Guylaine Ferland
- Hôpital du Sacré-Coeur de Montréal Research Center Montreal, QC, Canada ; Department of Nutrition, University of Montreal Montreal, QC, Canada
| | - Pierrette Gaudreau
- Laboratory of Neuroendocrinology of Aging, Centre Hospitalier de l'Université de Montréal Research Center Montreal, QC, Canada ; Department of Medicine, University of Montreal Montreal, QC, Canada
| |
Collapse
|
34
|
Hagenfeld D, Kathagen N, Prehm P. Adsorption of Glycosaminoglycans to the Cell Surface Is Responsible for Cellular Donnan Effects. J Cell Biochem 2014; 115:1334-41. [DOI: 10.1002/jcb.24791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/17/2014] [Indexed: 01/02/2023]
Affiliation(s)
- Daniel Hagenfeld
- Section of Periodontology; Department of Conservative Dentistry, Clinic for Oral, Dental and Maxillofacial Diseases, University Hospital Heidelberg; Heidelberg Germany
| | - Nadine Kathagen
- Muenster University Hospital; Institute of Physiological Chemistry and Pathobiochemistry; Waldeyerstr. 15 D-48129 Muenster Germany
| | - Peter Prehm
- Muenster University Hospital; Institute of Physiological Chemistry and Pathobiochemistry; Waldeyerstr. 15 D-48129 Muenster Germany
- Hylitis; Rudolf-Harbig-Str. 5 D-48301 Nottuln Germany
| |
Collapse
|
35
|
Calpain-generated natural protein fragments as short-lived substrates of the N-end rule pathway. Proc Natl Acad Sci U S A 2014; 111:E817-26. [PMID: 24550490 DOI: 10.1073/pnas.1401639111] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Calpains are Ca(2+)-dependent intracellular proteases. We show here that calpain-generated natural C-terminal fragments of proteins that include G protein-coupled receptors, transmembrane ion channels, transcriptional regulators, apoptosis controllers, kinases, and phosphatases (Phe-GluN2a, Lys-Ica512, Arg-Ankrd2, Tyr-Grm1, Arg-Atp2b2, Glu-Bak, Arg-Igfbp2, Glu-IκBα, and Arg-c-Fos), are short-lived substrates of the Arg/N-end rule pathway, which targets destabilizing N-terminal residues. We also found that the identity of a fragment's N-terminal residue can change during evolution, but the residue's destabilizing activity is virtually always retained, suggesting selection pressures that favor a short half-life of the calpain-generated fragment. It is also shown that a self-cleavage of a calpain can result in an N-end rule substrate. Thus, the autoprocessing of calpains can control them by making active calpains short-lived. These and related results indicate that the Arg/N-end rule pathway mediates the remodeling of oligomeric complexes by eliminating protein fragments that are produced in these complexes through cleavages by calpains or other nonprocessive proteases. We suggest that this capability of the Arg/N-end rule pathway underlies a multitude of its previously known but mechanistically unclear functions.
Collapse
|
36
|
|
37
|
Khan ZU, Martín-Montañez E, Navarro-Lobato I, Muly EC. Memory deficits in aging and neurological diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 122:1-29. [PMID: 24484696 DOI: 10.1016/b978-0-12-420170-5.00001-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Memory is central to our ability to perform daily life activities and correctly function in society. Improvements in public health and medical treatment for a variety of diseases have resulted in longer life spans; however, age-related memory impairments have been significant sources of morbidity. Loss in memory function is not only associated with aging population but is also a feature of neurodegenerative diseases such as Alzheimer's disease and other psychiatric and neurological disorders. Here, we focus on current understanding of the impact of normal aging on memory and what is known about its mechanisms, and further review pathological mechanisms behind the cause of dementia in Alzheimer's disease. Finally, we discuss schizophrenia and look into abnormalities in circuit function and neurotransmitter systems that contribute to memory impairment in this illness.
Collapse
Affiliation(s)
- Zafar U Khan
- Laboratory of Neurobiology at CIMES, University of Málaga, Málaga, Spain; Department of Medicine at Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Elisa Martín-Montañez
- Laboratory of Neurobiology at CIMES, University of Málaga, Málaga, Spain; Department of Pharmacology at Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Irene Navarro-Lobato
- Laboratory of Neurobiology at CIMES, University of Málaga, Málaga, Spain; Department of Medicine at Faculty of Medicine, University of Málaga, Málaga, Spain
| | - E Chris Muly
- Atlanta Department of Veterans Affairs Medical Center, Decatur, Georgia, USA; Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia, USA; Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Atlanta, Georgia, USA
| |
Collapse
|
38
|
Jensen G, Ward RD, Balsam PD. Information: theory, brain, and behavior. J Exp Anal Behav 2013; 100:408-31. [PMID: 24122456 DOI: 10.1002/jeab.49] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 08/26/2013] [Indexed: 01/15/2023]
Abstract
In the 65 years since its formal specification, information theory has become an established statistical paradigm, providing powerful tools for quantifying probabilistic relationships. Behavior analysis has begun to adopt these tools as a novel means of measuring the interrelations between behavior, stimuli, and contingent outcomes. This approach holds great promise for making more precise determinations about the causes of behavior and the forms in which conditioning may be encoded by organisms. In addition to providing an introduction to the basics of information theory, we review some of the ways that information theory has informed the studies of Pavlovian conditioning, operant conditioning, and behavioral neuroscience. In addition to enriching each of these empirical domains, information theory has the potential to act as a common statistical framework by which results from different domains may be integrated, compared, and ultimately unified.
Collapse
|
39
|
Kelley P, Whatson T. Making long-term memories in minutes: a spaced learning pattern from memory research in education. Front Hum Neurosci 2013; 7:589. [PMID: 24093012 PMCID: PMC3782739 DOI: 10.3389/fnhum.2013.00589] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/01/2013] [Indexed: 12/23/2022] Open
Abstract
Memory systems select from environmental stimuli those to encode permanently. Repeated stimuli separated by timed spaces without stimuli can initiate Long-Term Potentiation (LTP) and long-term memory (LTM) encoding. These processes occur in time scales of minutes, and have been demonstrated in many species. This study reports on using a specific timed pattern of three repeated stimuli separated by 10 min spaces drawn from both behavioral and laboratory studies of LTP and LTM encoding. A technique was developed based on this pattern to test whether encoding complex information into LTM in students was possible using the pattern within a very short time scale. In an educational context, stimuli were periods of highly compressed instruction, and spaces were created through 10 min distractor activities. Spaced Learning in this form was used as the only means of instruction for a national curriculum Biology course, and led to very rapid LTM encoding as measured by the high-stakes test for the course. Remarkably, learning at a greatly increased speed and in a pattern that included deliberate distraction produced significantly higher scores than random answers (p < 0.00001) and scores were not significantly different for experimental groups (one hour spaced learning) and control groups (four months teaching). Thus learning per hour of instruction, as measured by the test, was significantly higher for the spaced learning groups (p < 0.00001). In a third condition, spaced learning was used to replace the end of course review for one of two examinations. Results showed significantly higher outcomes for the course using spaced learning (p < 0.0005). The implications of these findings and further areas for research are briefly considered.
Collapse
Affiliation(s)
- Paul Kelley
- Science + Technology in LearningWhitley Bay, UK
| | - Terry Whatson
- Life, Health and Chemical Sciences, The Open UniversityMilton Keynes, UK
| |
Collapse
|
40
|
Baudry M, Bi X, Aguirre C. Progesterone-estrogen interactions in synaptic plasticity and neuroprotection. Neuroscience 2013; 239:280-94. [PMID: 23142339 PMCID: PMC3628409 DOI: 10.1016/j.neuroscience.2012.10.051] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 10/23/2012] [Accepted: 10/26/2012] [Indexed: 01/01/2023]
Abstract
17ß-Estradiol and progesterone exert a number of physiological effects throughout the brain due to interactions with several types of receptors belonging to the traditional family of intracellular hormonal receptors as well as to membrane-bound receptors. In particular, both hormones elicit rapid modifications of neuronal excitability that have been postulated to underlie their effects on synaptic plasticity and learning and memory. Likewise, both hormones have been shown to be neuroprotective under certain conditions, possibly due to the activation of pro-survival pathways and the inhibition of pro-apoptotic cascades. Because of the similarities in their cellular effects, there have been a number of questions raised by numerous observations that progesterone inhibits the effects of estrogen. In this manuscript, we first review the interactions between 17ß-estradiol (E2) and progesterone (P4) in synaptic plasticity, and conclude that, while E2 exerts a clear and important role in long-term potentiation of synaptic transmission in hippocampal neurons, the role of P4 is much less clear, and could be accounted by the direct or indirect regulation of GABAA receptors. We then discuss the neuroprotective roles of both hormones, in particular against excitotoxicity. In this case, the neuroprotective effects of these hormones are very similar to those of the neurotrophic factor BDNF. Interestingly, P4 antagonizes the effects of E2, possibly through the regulation of estrogen receptors or of proteins associated with the receptors or interactions with signaling pathways activated by E2. Overall, this review emphasizes the existence of common molecules and pathways that participate in the regulation of both synaptic plasticity and neurodegeneration.
Collapse
Affiliation(s)
- M Baudry
- GCBS and COMP, Western University of Health Sciences, Pomona, CA, USA.
| | | | | |
Collapse
|
41
|
Baudry M, Bi X. Learning and memory: an emergent property of cell motility. Neurobiol Learn Mem 2013; 104:64-72. [PMID: 23707799 DOI: 10.1016/j.nlm.2013.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 04/29/2013] [Accepted: 04/30/2013] [Indexed: 01/24/2023]
Abstract
In this review, we develop the argument that the molecular/cellular mechanisms underlying learning and memory are an adaptation of the mechanisms used by all cells to regulate cell motility. Neuronal plasticity and more specifically synaptic plasticity are widely recognized as the processes by which information is stored in neuronal networks engaged during the acquisition of information. Evidence accumulated over the last 25 years regarding the molecular events underlying synaptic plasticity at excitatory synapses has shown the remarkable convergence between those events and those taking place in cells undergoing migration in response to extracellular signals. We further develop the thesis that the calcium-dependent protease, calpain, which we postulated over 25 years ago to play a critical role in learning and memory, plays a central role in the regulation of both cell motility and synaptic plasticity. The findings discussed in this review illustrate the general principle that fundamental cell biological processes are used for a wide range of functions at the level of organisms.
Collapse
Affiliation(s)
- Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA.
| | | |
Collapse
|
42
|
Integrin dynamics produce a delayed stage of long-term potentiation and memory consolidation. J Neurosci 2012; 32:12854-61. [PMID: 22973009 DOI: 10.1523/jneurosci.2024-12.2012] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Memory consolidation theory posits that newly acquired information passes through a series of stabilization steps before being firmly encoded. We report here that in rat and mouse, hippocampus cell adhesion receptors belonging to the β1-integrin family exhibit dynamic properties in adult synapses and that these contribute importantly to a previously unidentified stage of consolidation. Quantitative dual immunofluorescence microscopy showed that induction of long-term potentiation (LTP) by theta burst stimulation (TBS) activates β1 integrins, and integrin-signaling kinases, at spine synapses in adult hippocampal slices. Neutralizing antisera selective for β1 integrins blocked these effects. TBS-induced integrin activation was brief (<7 min) and followed by an ∼45 min period during which the adhesion receptors did not respond to a second application of TBS. Brefeldin A, which blocks integrin trafficking to the plasma membrane, prevented the delayed recovery of integrin responses to TBS. β1 integrin-neutralizing antisera erased LTP when applied during, but not after, the return of integrin responsivity. Similarly, infusions of anti-β1 into rostral mouse hippocampus blocked formation of long-term, object location memory when started 20 min after learning but not 40 min later. The finding that β1 integrin neutralization was effective in the same time window for slice and behavioral experiments strongly suggests that integrin recovery triggers a temporally discrete, previously undetected second stage of consolidation for both LTP and memory.
Collapse
|
43
|
Shevelkin AV, Efimova OI, Nikitin VP, Anokhin KV, Sherstnev VV. Specific Changes in c-fos Expression and Colocalization with DNA in Identified Neuronal Nuclei of Edible Snail Following Neurotransmitter Application. Bull Exp Biol Med 2012; 153:734-7. [DOI: 10.1007/s10517-012-1813-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
44
|
Ménard C, Quirion R. Group 1 metabotropic glutamate receptor function and its regulation of learning and memory in the aging brain. Front Pharmacol 2012; 3:182. [PMID: 23091460 PMCID: PMC3469824 DOI: 10.3389/fphar.2012.00182] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/23/2012] [Indexed: 12/22/2022] Open
Abstract
Normal aging is generally characterized by a slow decline of cognitive abilities albeit with marked individual differences. Several animal models have been studied to explore the molecular and cellular mechanisms underlying this phenomenon. The excitatory neurotransmitter glutamate and its receptors have been closely linked to spatial learning and hippocampus-dependent memory processes. For decades, ionotropic glutamate receptors have been known to play a critical role in synaptic plasticity, a form of adaptation regulating memory formation. Over the past 10 years, several groups have shown the importance of group 1 metabotropic glutamate receptor (mGluR) in successful cognitive aging. These G-protein-coupled receptors are enriched in the hippocampal formation and interact physically with other proteins in the membrane including glutamate ionotropic receptors. Synaptic plasticity is crucial to maintain cognitive abilities and long-term depression (LTD) induced by group 1 mGluR activation, which has been linked to memory in the aging brain. The translation and synthesis of proteins by mGluR-LTD modulate ionotropic receptor trafficking and expression of immediate early genes related to cognition. Fragile X syndrome, a genetic form of autism characterized by memory deficits, has been associated to mGluR receptor malfunction and aberrant activation of its downstream signaling pathways. Dysfunction of mGluR could also be involved in neurodegenerative disorders like Alzheimer’s disease (AD). Indeed, beta-amyloid, the main component of insoluble senile plaques and one of the hallmarks of AD, occludes mGluR-dependent LTD leading to diminished functional synapses. This review highlights recent findings regarding mGluR signaling, related synaptic plasticity, and their potential involvement in normal aging and neurological disorders.
Collapse
Affiliation(s)
- Caroline Ménard
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University Montreal, QC, Canada
| | | |
Collapse
|
45
|
Baudry M, Kramar E, Xu X, Zadran H, Moreno S, Lynch G, Gall C, Bi X. Ampakines promote spine actin polymerization, long-term potentiation, and learning in a mouse model of Angelman syndrome. Neurobiol Dis 2012; 47:210-5. [PMID: 22525571 DOI: 10.1016/j.nbd.2012.04.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 03/29/2012] [Accepted: 04/06/2012] [Indexed: 12/20/2022] Open
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder largely due to abnormal maternal expression of the UBE3A gene leading to the deletion of E6-associated protein. AS subjects have severe cognitive impairments for which there are no therapeutic interventions. Mouse models (knockouts of the maternal Ube3a gene: 'AS mice') of the disorder have substantial deficits in long-term potentiation (LTP) and learning. Here we report a clinically plausible pharmacological treatment that ameliorates both deficits. AS mice were injected ip twice daily for 5 days with vehicle or the ampakine CX929; drugs of this type enhance fast EPSCs by positively modulating AMPA receptors. Theta burst stimulation (TBS) produced a normal enhancement of field EPSPs in hippocampal slices prepared from vehicle-treated AS mice but LTP decreased steadily to baseline; however, LTP in slices from ampakine-treated AS mice stabilized at levels found in wild-type controls. TBS-induced actin polymerization within dendritic spines, an essential event for stabilizing LTP, was severely impaired in slices from vehicle-treated AS mice but not in those from ampakine-treated AS mice. Long-term memory scores in a fear conditioning paradigm were reduced by 50% in vehicle-treated AS mice but were comparable to values for littermate controls in the ampakine-treated AS mice. We propose that AS is associated with a profound defect in activity-driven spine cytoskeletal reorganization, resulting in a loss of the synaptic plasticity required for the encoding of long-term memory. Notably, the spine abnormality along with the LTP and learning impairments can be reduced by a minimally invasive drug treatment.
Collapse
Affiliation(s)
- Michel Baudry
- Department of Biological Science, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|