51
|
Chen Y, Bang S, McMullen MF, Kazi H, Talbot K, Ho MX, Carlson G, Arnold SE, Ong WY, Kim SF. Neuronal Activity-Induced Sterol Regulatory Element Binding Protein-1 (SREBP1) is Disrupted in Dysbindin-Null Mice-Potential Link to Cognitive Impairment in Schizophrenia. Mol Neurobiol 2017; 54:1699-1709. [PMID: 26873854 PMCID: PMC4982840 DOI: 10.1007/s12035-016-9773-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/02/2016] [Indexed: 12/30/2022]
Abstract
Schizophrenia is a chronic debilitating neuropsychiatric disorder that affects about 1 % of the population. Dystrobrevin-binding protein 1 (DTNBP1 or dysbindin) is one of the Research Domain Constructs (RDoC) associated with cognition and is significantly reduced in the brain of schizophrenia patients. To further understand the molecular underpinnings of pathogenesis of schizophrenia, we have performed microarray analyses of the hippocampi from dysbindin knockout mice, and found that genes involved in the lipogenic pathway are suppressed. Moreover, we discovered that maturation of a master transcriptional regulator for lipid synthesis, sterol regulatory element binding protein-1 (SREBP1) is induced by neuronal activity, and is required for induction of the immediate early gene ARC (activity-regulated cytoskeleton-associated protein), necessary for synaptic plasticity and memory. We found that nuclear SREBP1 is dramatically reduced in dysbindin-1 knockout mice and postmortem brain tissues from human patients with schizophrenia. Furthermore, activity-dependent maturation of SREBP1 as well as ARC expression were attenuated in dysbindin-1 knockout mice, and these deficits were restored by an atypical antipsychotic drug, clozapine. Together, results indicate an important role of dysbindin-1 in neuronal activity induced SREBP1 and ARC, which could be related to cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
- Yong Chen
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sookhee Bang
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mary F McMullen
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hala Kazi
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Konrad Talbot
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mei-Xuan Ho
- Department of Anatomy and Neurobiology Research Programme, National University of Singapore, Singapore, 119260, Singapore
| | - Greg Carlson
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Steven E Arnold
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wei-Yi Ong
- Department of Anatomy and Neurobiology Research Programme, National University of Singapore, Singapore, 119260, Singapore.
| | - Sangwon F Kim
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
52
|
Singh P, Thakur MK. Histone Deacetylase 2 Inhibition Attenuates Downregulation of Hippocampal Plasticity Gene Expression during Aging. Mol Neurobiol 2017; 55:2432-2442. [PMID: 28364391 DOI: 10.1007/s12035-017-0490-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
Abstract
The brain undergoes several anatomical, biochemical, and molecular changes during aging, which subsequently result in downregulation of synaptic plasticity genes and decline of memory. However, the regulation of these genes during aging is not clearly understood. Previously, we reported that the expression of histone deacetylase (HDAC)2 was upregulated in the hippocampus of old mice and negatively correlated with the decline in recognition memory. As HDAC2 regulates key synaptic plasticity neuronal immediate early genes (IEGs), we have examined their expression and epigenetic regulation. We noted that the expression of neuronal IEGs decreased both at mRNA and protein level in the hippocampus of old mice. To explore the underlying regulation, we analyzed the binding of HDAC2 and level of histone acetylation at the promoter of neuronal IEGs. While the binding of HDAC2 was higher, H3K9 and H3K14 acetylation level was lower at the promoter of these genes in old as compared to young and adult mice. Further, we inhibited HDAC2 non-specifically by sodium butyrate and specifically by antisense oligonucleotide to recover epigenetic modification, expression of neuronal IEGs, and memory in old mice. Inhibition of HDAC2 increased histone H3K9 and H3K14 acetylation level at the promoter of neuronal IEGs, their expression, and recognition memory in old mice as compared to control. Thus, inhibition of HDAC2 can be used as a therapeutic target to recover decline in memory due to aging and associated neurological disorders.
Collapse
Affiliation(s)
- Padmanabh Singh
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - M K Thakur
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
53
|
Zhang YF, Li QQ, Qu J, Sun CM, Wang Y. Alterations of motor cortical microcircuit in a depressive-like mouse model produced by light deprivation. Neuroscience 2017; 341:79-94. [DOI: 10.1016/j.neuroscience.2016.11.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 01/01/2023]
|
54
|
A First-in-Class Small-Molecule that Acts as a Dual Inhibitor of HDAC and PDE5 and that Rescues Hippocampal Synaptic Impairment in Alzheimer's Disease Mice. Neuropsychopharmacology 2017; 42:524-539. [PMID: 27550730 PMCID: PMC5399234 DOI: 10.1038/npp.2016.163] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/27/2016] [Accepted: 07/30/2016] [Indexed: 12/26/2022]
Abstract
The targeting of two independent but synergistic enzymatic activities, histone deacetylases (HDACs, class I and HDAC6) and phosphodiesterase 5 (PDE5), has recently been validated as a potentially novel therapeutic approach for Alzheimer's disease (AD). Here we report the discovery of a new first-in-class small-molecule (CM-414) that acts as a dual inhibitor of PDE5 and HDACs. We have used this compound as a chemical probe to validate this systems therapeutics strategy, where an increase in the activation of cAMP/cGMP-responsive element-binding protein (CREB) induced by PDE5 inhibition, combined with moderate HDAC class I inhibition, leads to efficient histone acetylation. This molecule rescued the impaired long-term potentiation evident in hippocampal slices from APP/PS1 mice. Chronic treatment of Tg2576 mice with CM-414 diminished brain Aβ and tau phosphorylation (pTau) levels, increased the inactive form of GSK3β, reverted the decrease in dendritic spine density on hippocampal neurons, and reversed their cognitive deficits, at least in part by inducing the expression of genes related to synaptic transmission. Thus, CM-414 may serve as the starting point to discover balanced dual inhibitors with an optimal efficacy and safety profile for clinical testing on AD patients.
Collapse
|
55
|
β-Asarone Rescues Pb-Induced Impairments of Spatial Memory and Synaptogenesis in Rats. PLoS One 2016; 11:e0167401. [PMID: 27936013 PMCID: PMC5147873 DOI: 10.1371/journal.pone.0167401] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 11/14/2016] [Indexed: 11/19/2022] Open
Abstract
Chronic lead (Pb) exposure causes cognitive deficits. This study aimed to explore the neuroprotective effect and mechanism of β-asarone, an active component from Chinese Herbs Acorus tatarinowii Schott, to alleviate impairments of spatial memory and synaptogenesis in Pb-exposed rats. Both Sprague-Dawley developmental rat pups and adult rats were used in the study. Developmental rat pups were exposed to Pb throughout the lactation period and β-asarone (10, 40mg kg-1, respectively) was given intraperitoneally from postnatal day 14 to 21. Also, the adult rats were exposed to Pb from embryo stage to 11 weeks old and β-asarone (2.5, 10, 40mg kg-1, respectively) was given from 9 to 11 weeks old. The level of β-asarone in brain tissue was measured by High Performance Liquid Chromatography. The Morris water maze test and Golgi-Cox staining method were used to assess spatial memory ability and synaptogenesis. The protein expression of NR2B subunit of NMDA receptor, Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) and Wnt family member 7A (Wnt7a) in hippocampus, as well as mRNA expression of Arc/Arg3.1 and Wnt7a, was also explored. We found that β-asarone could pass through the blood brain barrier quickly. And β-asarone effectively attenuated Pb-induced reduction of spine density in hippocampal CA1 and dentate gyrus areas in a dose-dependent manner both in developmental and adult rats, meanwhile the Pb-induced impairments of learning and memory were partially rescued. In addition, β-asarone effectively up-regulated the protein expression of NR2B, Arc and Wnt7a, as well as the mRNA levels of Arc/Arg3.1 and Wnt7a, which had been suppressed by Pb exposure. The results suggest the neuroprotective properties of β-asarone against Pb-induced memory impairments, and the effect is possibly through the regulation of synaptogenesis, which is mediated via Arc/Arg3.1 and Wnt pathway.
Collapse
|
56
|
Lee S, Kang S, Kim J, Yoon S, Kim SH, Moon C. Enhanced expression of immediate-early genes in mouse hippocampus after trimethyltin treatment. Acta Histochem 2016; 118:679-684. [PMID: 27614947 DOI: 10.1016/j.acthis.2016.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 12/27/2022]
Abstract
Immediate-early genes (IEGs) are transiently and rapidly activated in response to various cellular stimuli. IEGs mediate diverse functions during pathophysiologic events by regulating cellular signal transduction. We investigated the temporal expression of several IEGs, including c-fos, early growth response protein-1 (Egr-1), and activity-regulated cytoskeleton-associated protein (Arc), in trimethyltin (TMT)-induced hippocampal neurodegeneration. Mice (7 weeks old, C57BL/6) administered TMT (2.6mg/kg intraperitoneally) presented severe neurodegenerative lesions in the dentate gyrus (DG) and showed behavioral seizure activity on days 1-4 post-treatment, after which the lesions and behavior recovered spontaneously over time. c-fos, Egr-1, and Arc mRNA and protein levels significantly increased in the mouse hippocampus after TMT treatment. Immunohistochemical analysis showed that nuclear c-fos expression increased mainly in the DG, whereas nuclear Egr-1 expression was increased extensively in cornu ammonis (CA) 1, CA3, and the DG after TMT treatment. Increased Arc levels were detected in the cellular somata/dendrites of the hippocampal subregions after TMT treatment. Therefore, we suggest that increased IEGs are associated with TMT-induced pathological events in mouse hippocampus.
Collapse
Affiliation(s)
- Sueun Lee
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Sohi Kang
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Juhwan Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Seongwook Yoon
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Sung-Ho Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Changjong Moon
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
57
|
Srivas S, Thakur MK. Epigenetic regulation of neuronal immediate early genes is associated with decline in their expression and memory consolidation in scopolamine-induced amnesic mice. Mol Neurobiol 2016; 54:5107-5119. [PMID: 27553230 DOI: 10.1007/s12035-016-0047-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/08/2016] [Indexed: 01/08/2023]
Abstract
Recently, we reported a correlation of scopolamine mediated decline in memory consolidation with increase in the expression of DNA methyltransferase 1 (DNMT1) and histone deacetylase 2 (HDAC2) in the mouse hippocampus. Memory consolidation is a protein synthesis-dependent process which involves the expression of synaptic plasticity genes, particularly neuronal immediate early genes (IEGs). However, the mechanism of regulation of these genes during decline in memory is poorly understood. Therefore, we have studied the epigenetic regulation of expression of neuronal IEGs in scopolamine-induced amnesic mice. Scopolamine significantly impaired memory consolidation as tested by radial arm maze, and the expression of neuronal IEGs was downregulated in the hippocampus as revealed by qRT-PCR and Western blotting. Further, methylated DNA immunoprecipitation (MeDIP) analysis showed increase in DNA methylation, while chromatin immunoprecipitation (ChIP) revealed decrease in H3K9/14 acetylation at the promoter of neuronal IEGs. Taken together, the present study shows that increased DNA methylation and decreased histone acetylation at the promoter of neuronal IEGs are associated with decline in their expression and memory consolidation during scopolamine-induced amnesia. These findings suggest that the epigenetic regulation through altered DNA methylation and histone acetylation might be explored further to develop potential therapeutic interventions for amnesia.
Collapse
Affiliation(s)
- Sweta Srivas
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221 005, India
| | - Mahendra K Thakur
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
58
|
Na Y, Park S, Lee C, Kim DK, Park JM, Sockanathan S, Huganir RL, Worley PF. Real-Time Imaging Reveals Properties of Glutamate-Induced Arc/Arg 3.1 Translation in Neuronal Dendrites. Neuron 2016; 91:561-73. [PMID: 27397520 DOI: 10.1016/j.neuron.2016.06.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/10/2016] [Accepted: 06/06/2016] [Indexed: 01/09/2023]
Abstract
The immediate early gene Arc (also Arg3.1) produces rapid changes in synaptic properties that are linked to de novo translation. Here we develop a novel translation reporter that exploits the rapid maturation and "flash" kinetics of Gaussia luciferase (Gluc) to visualize Arc translation. Following glutamate stimulation, discrete Arc-Gluc bioluminescent flashes representing sites of de novo translation are detected within 15 s at distributed sites in dendrites, but not spines. Flashes are episodic, lasting ∼20 s, and may be unitary or repeated at ∼minute intervals at the same sites. Analysis of flash amplitudes suggests they represent the quantal product of one or more polyribosomes, while inter-flash intervals appear random, suggesting they arise from a stochastic process. Surprisingly, glutamate-induced translation is dependent on Arc open reading frame. Combined observations support a model in which stalled ribosomes are reactivated to rapidly generate Arc protein.
Collapse
Affiliation(s)
- Youn Na
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Divison of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Sungjin Park
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| | - Changhee Lee
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Genetics, Harvard Medical School, Boston, MA 02446, USA
| | - Dong-Kyu Kim
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| | - Joo Min Park
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Cognition and Sociality, Institute for Basic Science, Daejeon 305-811, Korea
| | - Shanthini Sockanathan
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L Huganir
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul F Worley
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
59
|
Fernandes D, Carvalho AL. Mechanisms of homeostatic plasticity in the excitatory synapse. J Neurochem 2016; 139:973-996. [PMID: 27241695 DOI: 10.1111/jnc.13687] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 11/30/2022]
Abstract
Brain development, sensory information processing, and learning and memory processes depend on Hebbian forms of synaptic plasticity, and on the remodeling and pruning of synaptic connections. Neurons in networks implicated in these processes carry out their functions while facing constant perturbation; homeostatic responses are therefore required to maintain neuronal activity within functional ranges for proper brain function. Here, we will review in vitro and in vivo studies demonstrating that several mechanisms underlie homeostatic plasticity of excitatory synapses, and identifying participant molecular players. Emerging evidence suggests a link between disrupted homeostatic synaptic plasticity and neuropsychiatric and neurologic disorders. Hebbian forms of synaptic plasticity, such as long-term potentiation (LTP), induce long-lasting changes in synaptic strength, which can be destabilizing and drive activity to saturation. Conversely, homeostatic plasticity operates to compensate for prolonged activity changes, stabilizing neuronal firing within a dynamic physiological range. We review mechanisms underlying homeostatic plasticity, and address how neurons integrate distinct forms of plasticity for proper brain function. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".
Collapse
Affiliation(s)
- Dominique Fernandes
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,PDBEB-Doctoral Program in Experimental Biology and Biomedicine, Interdisciplinary Research Institute (III-UC), University of Coimbra, Coimbra, Portugal
| | - Ana Luísa Carvalho
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
60
|
Transcriptional regulation of long-term potentiation. Neurogenetics 2016; 17:201-210. [PMID: 27318935 DOI: 10.1007/s10048-016-0489-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/10/2016] [Indexed: 10/21/2022]
Abstract
Long-term potentiation (LTP), the persistent strengthening of synapses following high levels of stimulation, is a form of synaptic plasticity that has been studied extensively as a possible mechanism for learning and memory formation. The strengthening of the synapse that occurs during LTP requires cascades of complex molecular processes and the coordinated remodeling of pre-synaptic and post-synaptic neurons. Despite over four decades of research, our understanding of the transcriptional mechanisms and molecular processes underlying LTP remains incomplete. Identification of all the proteins and non-coding RNA transcripts expressed during LTP may provide greater insight into the molecular mechanisms involved in learning and memory formation.
Collapse
|
61
|
Mellström B, Kastanauskaite A, Knafo S, Gonzalez P, Dopazo XM, Ruiz-Nuño A, Jefferys JGR, Zhuo M, Bliss TVP, Naranjo JR, DeFelipe J. Specific cytoarchitectureal changes in hippocampal subareas in daDREAM mice. Mol Brain 2016; 9:22. [PMID: 26928278 PMCID: PMC4772309 DOI: 10.1186/s13041-016-0204-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/22/2016] [Indexed: 11/20/2022] Open
Abstract
Background Transcriptional repressor DREAM (downstream regulatory element antagonist modulator) is a Ca2+-binding protein that regulates Ca2+ homeostasis through gene regulation and protein-protein interactions. It has been shown that a dominant active form (daDREAM) is implicated in learning-related synaptic plasticity such as LTP and LTD in the hippocampus. Neuronal spines are reported to play important roles in plasticity and memory. However, the possible role of DREAM in spine plasticity has not been reported. Results Here we show that potentiating DREAM activity, by overexpressing daDREAM, reduced dendritic basal arborization and spine density in CA1 pyramidal neurons and increased spine density in dendrites in dentate gyrus granule cells. These microanatomical changes are accompanied by significant modifications in the expression of specific genes encoding the cytoskeletal proteins Arc, Formin 1 and Gelsolin in daDREAM hippocampus. Conclusions Our results strongly suggest that DREAM plays an important role in structural plasticity in the hippocampus. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0204-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Britt Mellström
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,National Biotechnology Center. CSIC, Darwin, 3. E-28049, Madrid, Spain.
| | - Asta Kastanauskaite
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,Cajal Institute, CSIC Madrid, Av Dr. Arce,37 E-28006, Madrid, Spain. .,Biomedical Technology Center, Politecnica University Madrid, Madrid, Spain.
| | - Shira Knafo
- Cajal Institute, CSIC Madrid, Av Dr. Arce,37 E-28006, Madrid, Spain. .,Present address: IkerBasque Basque Foundation for Science and BioCruces, Health Research Institute, Bizkaia, Spain.
| | - Paz Gonzalez
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,National Biotechnology Center. CSIC, Darwin, 3. E-28049, Madrid, Spain.
| | - Xose M Dopazo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,National Biotechnology Center. CSIC, Darwin, 3. E-28049, Madrid, Spain.
| | - Ana Ruiz-Nuño
- Neuronal Networks Group, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK.
| | - John G R Jefferys
- Neuronal Networks Group, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham, UK.
| | - Min Zhuo
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada. .,Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Tim V P Bliss
- MRC National Institutes for Medical Research, Mill Hill, London, UK.
| | - Jose R Naranjo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,National Biotechnology Center. CSIC, Darwin, 3. E-28049, Madrid, Spain.
| | - Javier DeFelipe
- Spanish Network for Biomedical Research in Neurodegenerative Diseases, CIBERNED, Madrid, Spain. .,Cajal Institute, CSIC Madrid, Av Dr. Arce,37 E-28006, Madrid, Spain. .,Biomedical Technology Center, Politecnica University Madrid, Madrid, Spain.
| |
Collapse
|
62
|
Essali N, Sanders J. Interdependent adrenergic receptor regulation of Arc and Zif268 mRNA in cerebral cortex. Neurosci Lett 2016; 612:38-42. [PMID: 26655475 PMCID: PMC4727989 DOI: 10.1016/j.neulet.2015.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 11/30/2022]
Abstract
Norepinephrine is a neurotransmitter that signals by stimulating the α1, α2 and β adrenergic receptor (AR). We determined the role of these receptors in regulating the immediate early genes, Activity Regulated Cytoskeleton Associated Protein (Arc) and Zif268 in the rat cerebral cortex. RX821002, an α2-AR antagonist, produced Arc and Zif268 elevations across cortical layers. Next we examined the effects of delivering RX821002 with an α1-AR antagonist, prazosin, and a β-AR antagonist, propranolol. RX821002 given with a prazosin and propranolol cocktail, or with each of these antagonists individually, decreased Arc and Zif268 to saline-treated control levels in most cortical layers. Arc and Zif268 levels were also similar to saline-treated control levels when rats were given a prazosin and propranolol cocktail alone, or when each of these antagonists were delivered individually. Taken together, these data reveal that α2-AR uniquely exert a tonic inibitory regulation of both Arc and Zif268 compared to α1 and β-AR. However, the ability of RX821002 to increase Arc and Zif268 is interdependent with α1 and β-AR signaling.
Collapse
Affiliation(s)
- Norah Essali
- College of Medicine, Texila American University, Georgetown, Guyana
| | - Jeff Sanders
- Department of Pharmacology and Experimental Neuroscience, 985800 Nebraska Medical Center, Omaha, NE 68198-5800, USA.
| |
Collapse
|
63
|
Structural, Synaptic, and Epigenetic Dynamics of Enduring Memories. Neural Plast 2016; 2016:3425908. [PMID: 26933513 PMCID: PMC4736770 DOI: 10.1155/2016/3425908] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 11/17/2022] Open
Abstract
Our memories are the records of the experiences we gain in our everyday life. Over time, they slowly transform from an initially unstable state into a long-lasting form. Many studies have been investigating from different aspects how a memory could persist for sometimes up to decades. In this review, we highlight three of the greatly addressed mechanisms that play a central role for a given memory to endure: the allocation of the memory to a given neuronal population and what brain areas are recruited for its storage; the structural changes that underlie memory persistence; and finally the epigenetic control of gene expression that might regulate and support memory perseverance. Examining such key properties of a memory is essential towards a finer understanding of its capacity to last.
Collapse
|
64
|
Adenosine A1 Receptor-Mediated Endocytosis of AMPA Receptors Contributes to Impairments in Long-Term Potentiation (LTP) in the Middle-Aged Rat Hippocampus. Neurochem Res 2015; 41:1085-97. [PMID: 26700433 DOI: 10.1007/s11064-015-1799-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/09/2015] [Accepted: 12/09/2015] [Indexed: 12/20/2022]
Abstract
Aging causes multiple changes in the mammalian brain, including changes in synaptic signaling. Previous reports have shown increased extracellular adenosine in the aging brain, and we recently reported that activation of adenosine A1 receptors (A1Rs) induces AMPA receptor (AMPAR) internalization in rat hippocampus. This study investigated whether aging-related changes in the rat hippocampus include altered surface expression of adenosine A1 and A2A receptors, and whether these changes correspond to changes in AMPAR surface expression and altered synaptic plasticity. We found reduced A1R surface expression in middle-aged rat hippocampus, and also reduced GluA1 and GluA2 AMPAR subunit surface expression. Using a chemically-induced LTP (cLTP) experimental protocol, we recorded fEPSPs in young (1 month old) and middle-aged (7-12 month old) rat hippocampal slices. There were significant impairments in cLTP in middle-aged slices, suggesting impaired synaptic plasticity. Since we previously showed that the A1R agonist N(6)-cyclopentyladenosine (CPA) reduced both A1Rs and GluA2/GluA1 AMPARs, we hypothesized that the observed impaired synaptic plasticity in middle-aged brains is regulated by A1R-mediated AMPAR internalization by clathrin-mediated endocytosis. Following cLTP, we found a significant increase in GluA1 and GluA2 surface expression in young rats, which was blunted in middle-aged brains or in young brains pretreated with CPA. Blocking A1Rs with 8-cyclopentyl-1,3-dipropylxanthine or AMPAR endocytosis with either Tat-GluA2-3Y peptide or dynasore (dynamin inhibitor) similarly enhanced AMPAR surface expression following cLTP. These data suggest that age-dependent alteration in adenosine receptor expression contributes to increased AMPAR endocytosis and impaired synaptic plasticity in aged brains.
Collapse
|
65
|
Association of SNPs in EGR3 and ARC with Schizophrenia Supports a Biological Pathway for Schizophrenia Risk. PLoS One 2015; 10:e0135076. [PMID: 26474411 PMCID: PMC4608790 DOI: 10.1371/journal.pone.0135076] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/17/2015] [Indexed: 02/07/2023] Open
Abstract
We have previously hypothesized a biological pathway of activity-dependent synaptic plasticity proteins that addresses the dual genetic and environmental contributions to schizophrenia. Accordingly, variations in the immediate early gene EGR3, and its target ARC, should influence schizophrenia susceptibility. We used a pooled Next-Generation Sequencing approach to identify variants across these genes in U.S. populations of European (EU) and African (AA) descent. Three EGR3 and one ARC SNP were selected and genotyped for validation, and three SNPs were tested for association in a replication cohort. In the EU group of 386 schizophrenia cases and 150 controls EGR3 SNP rs1877670 and ARC SNP rs35900184 showed significant associations (p = 0.0078 and p = 0.0275, respectively). In the AA group of 185 cases and 50 controls, only the ARC SNP revealed significant association (p = 0.0448). The ARC SNP did not show association in the Han Chinese (CH) population. However, combining the EU, AA, and CH groups revealed a highly significant association of ARC SNP rs35900184 (p = 2.353 x 10−7; OR [95% CI] = 1.54 [1.310–1.820]). These findings support previously reported associations between EGR3 and schizophrenia. Moreover, this is the first report associating an ARC SNP with schizophrenia and supports recent large-scale GWAS findings implicating the ARC complex in schizophrenia risk. These results support the need for further investigation of the proposed pathway of environmentally responsive, synaptic plasticity-related, schizophrenia genes.
Collapse
|
66
|
Liu WC, Rivers JW, White DJ. Vocal matching and intensity of begging calls are associated with a forebrain song circuit in a generalist brood parasite. Dev Neurobiol 2015; 76:615-25. [DOI: 10.1002/dneu.22348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/16/2015] [Accepted: 08/31/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Wan-Chun Liu
- Laboratory of Animal Behavior; The Rockefeller University; New York New York 10065
| | - James W. Rivers
- Department of Forest Ecosystems and Society; Oregon State University, Corvallis; Oregon 97331
| | - David J. White
- Psychology Department; Wilfrid Laurier University; Ontario Canada N2L 3C5
| |
Collapse
|
67
|
Bacopa monniera (CDRI-08) Upregulates the Expression of Neuronal and Glial Plasticity Markers in the Brain of Scopolamine Induced Amnesic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:837012. [PMID: 26413129 PMCID: PMC4564643 DOI: 10.1155/2015/837012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/20/2015] [Indexed: 12/18/2022]
Abstract
Preclinical studies on animal models have discerned the antiamnesic and memory-enhancing potential of Bacopa monniera (Brahmi) crude extract and standardized extracts. These studies primarily focus on behavioral consequences. However, lack of information on molecular underpinnings has limited the clinical trials of the potent herb in human subjects. In recent years, researchers highlight plasticity markers as molecular correlates of amnesia and being crucial to design therapeutic targets. In the present report, we have investigated the effect of a special extract of B. monniera (CDRI-08) on the expression of key neuronal (BDNF and Arc) and glial (GFAP) plasticity markers in the cerebrum of scopolamine induced amnesic mice. Pre- and postadministration of CDRI-08 ameliorated amnesic effect of scopolamine by decreasing acetyl cholinesterase activity and drastically upregulating the mRNA and protein expression of BDNF, Arc, and GFAP in mouse cerebrum. Interestingly, the plant extract per se elevated BDNF and Arc expression as compared to control but GFAP was unaltered. In conclusion, our findings provide the first molecular evidence for antiamnesic potential of CDRI-08 via enhancement of both neuronal and glial plasticity markers. Further investigations on detailed molecular pathways would encourage therapeutic application of the extract in memory disorders.
Collapse
|
68
|
Behavioral Tagging: A Translation of the Synaptic Tagging and Capture Hypothesis. Neural Plast 2015; 2015:650780. [PMID: 26380117 PMCID: PMC4562088 DOI: 10.1155/2015/650780] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/12/2015] [Indexed: 11/18/2022] Open
Abstract
Similar molecular machinery is activated in neurons following an electrical stimulus that induces synaptic changes and after learning sessions that trigger memory formation. Then, to achieve perdurability of these processes protein synthesis is required for the reinforcement of the changes induced in the network. The synaptic tagging and capture theory provided a strong framework to explain synaptic specificity and persistence of electrophysiological induced plastic changes. Ten years later, the behavioral tagging hypothesis (BT) made use of the same argument, applying it to learning and memory models. The hypothesis postulates that the formation of lasting memories relies on at least two processes: the setting of a learning tag and the synthesis of plasticity related proteins, which once captured at tagged sites allow memory consolidation. BT explains how weak events, only capable of inducing transient forms of memories, can result in lasting memories when occurring close in time with other behaviorally relevant experiences that provide proteins. In this review, we detail the findings supporting the existence of BT process in rodents, leading to the consolidation, persistence, and interference of a memory. We focus on the molecular machinery taking place in these processes and describe the experimental data supporting the BT in humans.
Collapse
|
69
|
Li Y, Pehrson AL, Waller JA, Dale E, Sanchez C, Gulinello M. A critical evaluation of the activity-regulated cytoskeleton-associated protein (Arc/Arg3.1)'s putative role in regulating dendritic plasticity, cognitive processes, and mood in animal models of depression. Front Neurosci 2015; 9:279. [PMID: 26321903 PMCID: PMC4530346 DOI: 10.3389/fnins.2015.00279] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 07/23/2015] [Indexed: 11/13/2022] Open
Abstract
Major depressive disorder (MDD) is primarily conceptualized as a mood disorder but cognitive dysfunction is also prevalent, and may limit the daily function of MDD patients. Current theories on MDD highlight disturbances in dendritic plasticity in its pathophysiology, which could conceivably play a role in the production of both MDD-related mood and cognitive symptoms. This paper attempts to review the accumulated knowledge on the basic biology of the activity-regulated cytoskeleton-associated protein (Arc or Arg3.1), its effects on neural plasticity, and how these may be related to mood or cognitive dysfunction in animal models of MDD. On a cellular level, Arc plays an important role in modulating dendritic spine density and remodeling. Arc also has a close, bidirectional relationship with postsynaptic glutamate neurotransmission, since it is stimulated by multiple glutamatergic receptor mechanisms but also modulates α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor internalization. The effects on AMPA receptor trafficking are likely related to Arc's ability to modulate phenomena such as long-term potentiation, long-term depression, and synaptic scaling, each of which are important for maintaining proper cognitive function. Chronic stress models of MDD in animals show suppressed Arc expression in the frontal cortex but elevation in the amygdala. Interestingly, cognitive tasks depending on the frontal cortex are generally impaired by chronic stress, while those depending on the amygdala are enhanced, and antidepressant treatments stimulate cortical Arc expression with a timeline that is reminiscent of the treatment efficacy lag observed in the clinic or in preclinical models. However, pharmacological treatments that stimulate regional Arc expression do not universally improve relevant cognitive functions, and this highlights a need to further refine our understanding of Arc on a subcellular and network level.
Collapse
Affiliation(s)
- Yan Li
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Alan L Pehrson
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Jessica A Waller
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Elena Dale
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Connie Sanchez
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Maria Gulinello
- Behavioral Core Facility, Department of Neuroscience, Albert Einstein College of Medicine Bronx, NY, USA
| |
Collapse
|
70
|
Mosher J, Zhang W, Blumhagen RZ, D'Alessandro A, Nemkov T, Hansen KC, Hesselberth JR, Reis T. Coordination between Drosophila Arc1 and a specific population of brain neurons regulates organismal fat. Dev Biol 2015. [PMID: 26209258 DOI: 10.1016/j.ydbio.2015.07.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The brain plays a critical yet incompletely understood role in regulating organismal fat. We performed a neuronal silencing screen in Drosophila larvae to identify brain regions required to maintain proper levels of organismal fat. When used to modulate synaptic activity in specific brain regions, the enhancer-trap driver line E347 elevated fat upon neuronal silencing, and decreased fat upon neuronal activation. Unbiased sequencing revealed that Arc1 mRNA levels increase upon E347 activation. We had previously identified Arc1 mutations in a high-fat screen. Here we reveal metabolic changes in Arc1 mutants consistent with a high-fat phenotype and an overall shift toward energy storage. We find that Arc1-expressing cells neighbor E347 neurons, and manipulating E347 synaptic activity alters Arc1 expression patterns. Elevating Arc1 expression in these cells decreased fat, a phenocopy of E347 activation. Finally, loss of Arc1 prevented the lean phenotype caused by E347 activation, suggesting that Arc1 activity is required for E347 control of body fat. Importantly, neither E347 nor Arc1 manipulation altered energy-related behaviors. Our results support a model wherein E347 neurons induce Arc1 in specific neighboring cells to prevent excess fat accumulation.
Collapse
Affiliation(s)
- Jeremy Mosher
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Medical School, Aurora, CO 80045, United States
| | - Wei Zhang
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Medical School, Aurora, CO 80045, United States
| | - Rachel Z Blumhagen
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Medical School, Aurora, CO 80045, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Medical School, Aurora, CO 80045, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Medical School, Aurora, CO 80045, United States
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Medical School, Aurora, CO 80045, United States
| | - Jay R Hesselberth
- Department of Biochemistry and Molecular Genetics, University of Colorado Medical School, Aurora, CO 80045, United States
| | - Tânia Reis
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Medical School, Aurora, CO 80045, United States.
| |
Collapse
|
71
|
Knipper M, Panford-Walsh R, Singer W, Rüttiger L, Zimmermann U. Specific synaptopathies diversify brain responses and hearing disorders: you lose the gain from early life. Cell Tissue Res 2015; 361:77-93. [PMID: 25843689 PMCID: PMC4487345 DOI: 10.1007/s00441-015-2168-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/05/2015] [Indexed: 01/08/2023]
Abstract
Before hearing onset, inner hair cell (IHC) maturation proceeds under the influence of spontaneous Ca(2+) action potentials (APs). The temporal signature of the IHC Ca(2+) AP is modified through an efferent cholinergic feedback from the medial olivocochlear bundle (MOC) and drives the IHC pre- and post-synapse phenotype towards low spontaneous (spike) rate (SR), high-threshold characteristics. With sensory experience, the IHC pre- and post-synapse phenotype matures towards the instruction of low-SR, high-threshold and of high-SR, low-threshold auditory fiber characteristics. Corticosteroid feedback together with local brain-derived nerve growth factor (BDNF) and catecholaminergic neurotransmitters (dopamine) might be essential for this developmental step. In this review, we address the question of whether the control of low-SR and high-SR fiber characteristics is linked to various degrees of vulnerability of auditory fibers in the mature system. In particular, we examine several IHC synaptopathies in the context of various hearing disorders and exemplified shortfalls before and after hearing onset.
Collapse
Affiliation(s)
- Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | | | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| | - Ulrike Zimmermann
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Straße 5, 72076 Tübingen, Germany
| |
Collapse
|
72
|
Martín V, Vale C, Rubiolo JA, Roel M, Hirama M, Yamashita S, Vieytes MR, Botana LM. Chronic Ciguatoxin Treatment Induces Synaptic Scaling through Voltage Gated Sodium Channels in Cortical Neurons. Chem Res Toxicol 2015; 28:1109-19. [DOI: 10.1021/tx500506q] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Víctor Martín
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Carmen Vale
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Juan A. Rubiolo
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Maria Roel
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Masahiro Hirama
- Department
of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan
| | - Shuji Yamashita
- Department
of Chemistry, Graduate School of Science, Tohoku University, Sendai 980-8578, Japan
| | - Mercedes R. Vieytes
- Departamento
de Fisiología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Luís M. Botana
- Departamento
de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| |
Collapse
|
73
|
Abstract
Accumulating data, including those from large genetic association studies, indicate that alterations in glutamatergic synapse structure and function represent a common underlying pathology in many symptomatically distinct cognitive disorders. In this review, we discuss evidence from human genetic studies and data from animal models supporting a role for aberrant glutamatergic synapse function in the etiology of intellectual disability (ID), autism spectrum disorder (ASD), and schizophrenia (SCZ), neurodevelopmental disorders that comprise a significant proportion of human cognitive disease and exact a substantial financial and social burden. The varied manifestations of impaired perceptual processing, executive function, social interaction, communication, and/or intellectual ability in ID, ASD, and SCZ appear to emerge from altered neural microstructure, function, and/or wiring rather than gross changes in neuron number or morphology. Here, we review evidence that these disorders may share a common underlying neuropathy: altered excitatory synapse function. We focus on the most promising candidate genes affecting glutamatergic synapse function, highlighting the likely disease-relevant functional consequences of each. We first present a brief overview of glutamatergic synapses and then explore the genetic and phenotypic evidence for altered glutamate signaling in ID, ASD, and SCZ.
Collapse
Affiliation(s)
- Lenora Volk
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| | | | | | | |
Collapse
|
74
|
Almaguer-Melian W, Mercerón-Martínez D, Pavón-Fuentes N, Alberti-Amador E, Leon-Martinez R, Ledón N, Delgado Ocaña S, Bergado Rosado JA. Erythropoietin Promotes Neural Plasticity and Spatial Memory Recovery in Fimbria-Fornix-Lesioned Rats. Neurorehabil Neural Repair 2015; 29:979-88. [PMID: 25847024 DOI: 10.1177/1545968315572389] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Erythropoietin (EPO) upregulates the mitogen activated protein kinase (MAPK) cascade, a central signaling pathway in cellular plastic mechanisms, and is critical for normal brain development. OBJECTIVE We hypothesized that EPO could modulate the plasticity mechanisms supporting spatial memory recovery in fimbria-fornix-transected animals. METHODS Fimbria-fornix was transected in 3 groups of rats. Seven days later, EPO was injected daily for 4 consecutive days within 10 minutes after training on a water maze task. RESULTS Our results show that EPO injections 10 minutes after training produced a substantial spatial memory recovery in fimbria-fornix-lesioned animals. In contrast, an EPO injection shortly after fimbria-fornix lesion surgery does not promote spatial-memory recovery. Neither does daily EPO injection 5 hours after the water maze performance. EPO, on the other hand, induced the expression of plasticity-related genes like arc and bdnf, but this effect was independent of training or lesion. CONCLUSIONS This finding supports our working hypothesis that EPO can modulate transient neuroplastic mechanisms triggered by training in lesioned animals. Consequently, we propose that EPO administration can be a useful trophic factor to promote neural restoration when given in combination with training.
Collapse
Affiliation(s)
| | | | | | | | | | - Nuris Ledón
- Centro de Inmunología Molecular, La Habana, Cuba
| | | | | |
Collapse
|
75
|
Vecsey CG, Park AJ, Khatib N, Abel T. Effects of sleep deprivation and aging on long-term and remote memory in mice. ACTA ACUST UNITED AC 2015; 22:197-202. [PMID: 25776037 PMCID: PMC4371173 DOI: 10.1101/lm.036590.114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Sleep deprivation (SD) following hippocampus-dependent learning in young mice impairs memory when tested the following day. Here, we examined the effects of SD on remote memory in both young and aged mice. In young mice, we found that memory is still impaired 1 mo after training. SD also impaired memory in aged mice 1 d after training, but, by a month after training, sleep-deprived and control aged animals performed similarly, primarily due to remote memory decay in the control aged animals. Gene expression analysis supported the finding that SD has similar effects on the hippocampus in young and aged mice.
Collapse
Affiliation(s)
- Christopher G Vecsey
- Department of Biology, Swarthmore College, Swarthmore, Pennsylvania, USA Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alan J Park
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nora Khatib
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
76
|
Byers CE, Barylko B, Ross JA, Southworth DR, James NG, Taylor CA, Wang L, Collins KA, Estrada A, Waung M, Tassin TC, Huber KM, Jameson DM, Albanesi JP. Enhancement of dynamin polymerization and GTPase activity by Arc/Arg3.1. Biochim Biophys Acta Gen Subj 2015; 1850:1310-8. [PMID: 25783003 DOI: 10.1016/j.bbagen.2015.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 02/17/2015] [Accepted: 03/09/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND The Activity-regulated cytoskeleton-associated protein, Arc, is an immediate-early gene product implicated in various forms of synaptic plasticity. Arc promotes endocytosis of AMPA type glutamate receptors and regulates cytoskeletal assembly in neuronal dendrites. Its role in endocytosis may be mediated by its reported interaction with dynamin 2, a 100 kDa GTPase that polymerizes around the necks of budding vesicles and catalyzes membrane scission. METHODS Enzymatic and turbidity assays are used in this study to monitor effects of Arc on dynamin activity and polymerization. Arc oligomerization is measured using a combination of approaches, including size exclusion chromatography, sedimentation analysis, dynamic light scattering, fluorescence correlation spectroscopy, and electron microscopy. RESULTS We present evidence that bacterially-expressed His6-Arc facilitates the polymerization of dynamin 2 and stimulates its GTPase activity under physiologic conditions (37°C and 100mM NaCl). At lower ionic strength Arc also stabilizes pre-formed dynamin 2 polymers against GTP-dependent disassembly, thereby prolonging assembly-dependent GTP hydrolysis catalyzed by dynamin 2. Arc also increases the GTPase activity of dynamin 3, an isoform of implicated in dendrite remodeling, but does not affect the activity of dynamin 1, a neuron-specific isoform involved in synaptic vesicle recycling. We further show in this study that Arc (either His6-tagged or untagged) has a tendency to form large soluble oligomers, which may function as a scaffold for dynamin assembly and activation. CONCLUSIONS AND GENERAL SIGNIFICANCE The ability of Arc to enhance dynamin polymerization and GTPase activation may provide a mechanism to explain Arc-mediated endocytosis of AMPA receptors and the accompanying effects on synaptic plasticity.
Collapse
Affiliation(s)
- Christopher E Byers
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Barbara Barylko
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Justin A Ross
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 9681, United States
| | - Daniel R Southworth
- Department of Chemistry and Chemical Biology, University of California, San Francisco, CA 94158, United States
| | - Nicholas G James
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 9681, United States
| | - Clinton A Taylor
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Lei Wang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Katie A Collins
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Armando Estrada
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Maggie Waung
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Tara C Tassin
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Kimberly M Huber
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - David M Jameson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 9681, United States
| | - Joseph P Albanesi
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| |
Collapse
|
77
|
A Neuronal Activity-Dependent Dual Function Chromatin-Modifying Complex Regulates Arc Expression. eNeuro 2015; 2:eN-NWR-0020-14. [PMID: 26464965 PMCID: PMC4586916 DOI: 10.1523/eneuro.0020-14.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 01/18/2015] [Accepted: 01/22/2015] [Indexed: 11/21/2022] Open
Abstract
Chromatin modification is an important epigenetic mechanism underlying neuroplasticity. Histone methylation and acetylation have both been shown to modulate gene expression, but the machinery responsible for mediating these changes in neurons has remained elusive. Here we identify a chromatin-modifying complex containing the histone demethylase PHF8 and the acetyltransferase TIP60 as a key regulator of the activity-induced expression of Arc, an important mediator of synaptic plasticity. Clinically, mutations in PHF8 cause X-linked mental retardation while TIP60 has been implicated in the pathogenesis of Alzheimer's disease. Within minutes of increased synaptic activity, this dual function complex is rapidly recruited to the Arc promoter, where it specifically counteracts the transcriptionally repressive histone mark H3K9me2 to facilitate the formation of the transcriptionally permissive H3K9acS10P, thereby favoring transcriptional activation. Consequently, gain-of-function of the PHF8-TIP60 complex in primary rat hippocampal neurons has a positive effect on early activity-induced Arc gene expression, whereas interfering with the function of this complex abrogates it. A global proteomics screen revealed that the majority of common interactors of PHF8 and TIP60 were involved in mRNA processing, including PSF, an important molecule involved in neuronal gene regulation. Finally, we proceeded to show, using super-resolution microscopy, that PHF8 and TIP60 interact at the single molecule level with PSF, thereby situating this chromatin modifying complex at the crossroads of transcriptional activation. These findings point toward a mechanism by which an epigenetic pathway can regulate neuronal activity-dependent gene transcription, which has implications in the development of novel therapeutics for disorders of learning and memory.
Collapse
|
78
|
Khodadad A, Adelson PD, Lifshitz J, Thomas TC. The time course of activity-regulated cytoskeletal (ARC) gene and protein expression in the whisker-barrel circuit using two paradigms of whisker stimulation. Behav Brain Res 2015; 284:249-56. [PMID: 25682931 DOI: 10.1016/j.bbr.2015.01.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/21/2014] [Accepted: 01/20/2015] [Indexed: 11/25/2022]
Abstract
Immediate early genes have previously demonstrated a rapid increase in gene expression after various behavioral paradigms. The main focus of this article is to identify a molecular marker of circuit activation after manual whisker stimulation or exploration of a novel environment. To this end, we investigated the dynamics of ARC transcription in adult male rats during whisker somatosensation throughout the whisker barrel circuit. At various time points, tissue was biopsied from the ventral posterior medial nucleus (VPM) of the thalamus, primary somatosensory barrel field (S1BF) cortex and hippocampus for quantification using real-time PCR and western blot. Our results show that there were no significant differences in ARC gene or protein expression in the VPM after both types of stimulation. However, manual whisker stimulation resulted in increased ARC gene expression at 15, 30, 60 and 300 min in the S1BF, and 15 min in the hippocampus (p<0.05). Also, exploration of a novel environment resulted in increased ARC mRNA expression at 15 and 30 min in the S1BF and at 15 min in the hippocampus (p<0.05). The type of stimulation (manual versus exploration of a novel environment) influenced the magnitude of ARC gene expression in the S1BF (p<0.05). These data are the first to demonstrate that ARC is a specific, quantifiable and input dependent molecular marker of circuit activation which can serve to quantify the impact of brain injury and subsequent rehabilitation on whisker sensation.
Collapse
Affiliation(s)
- Aida Khodadad
- Barrow Neurological Institute at Phoenix Children's Hospital- Phoenix, AZ; Department of Child Health, University of Arizona College of Medicine-Phoenix, AZ; Department of Neuroscience, University of Strasbourg, France.
| | - P David Adelson
- Barrow Neurological Institute at Phoenix Children's Hospital- Phoenix, AZ; Department of Child Health, University of Arizona College of Medicine-Phoenix, AZ; Neuroscience Program, Arizona State University, Tempe, AZ; School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ.
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital- Phoenix, AZ; Department of Child Health, University of Arizona College of Medicine-Phoenix, AZ; Phoenix VA Healthcare System- Phoenix, AZ; Neuroscience Program, Arizona State University, Tempe, AZ.
| | - Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital- Phoenix, AZ; Department of Child Health, University of Arizona College of Medicine-Phoenix, AZ; Phoenix VA Healthcare System- Phoenix, AZ.
| |
Collapse
|
79
|
Calais JB, Ojopi EB, Morya E, Sameshima K, Ribeiro S. Experience-dependent upregulation of multiple plasticity factors in the hippocampus during early REM sleep. Neurobiol Learn Mem 2015; 122:19-27. [PMID: 25626078 DOI: 10.1016/j.nlm.2015.01.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/26/2014] [Accepted: 01/05/2015] [Indexed: 11/16/2022]
Abstract
Sleep is beneficial to learning, but the underlying mechanisms remain controversial. The synaptic homeostasis hypothesis (SHY) proposes that the cognitive function of sleep is related to a generalized rescaling of synaptic weights to intermediate levels, due to a passive downregulation of plasticity mechanisms. A competing hypothesis proposes that the active upscaling and downscaling of synaptic weights during sleep embosses memories in circuits respectively activated or deactivated during prior waking experience, leading to memory changes beyond rescaling. Both theories have empirical support but the experimental designs underlying the conflicting studies are not congruent, therefore a consensus is yet to be reached. To advance this issue, we used real-time PCR and electrophysiological recordings to assess gene expression related to synaptic plasticity in the hippocampus and primary somatosensory cortex of rats exposed to novel objects, then kept awake (WK) for 60 min and finally killed after a 30 min period rich in WK, slow-wave sleep (SWS) or rapid-eye-movement sleep (REM). Animals similarly treated but not exposed to novel objects were used as controls. We found that the mRNA levels of Arc, Egr1, Fos, Ppp2ca and Ppp2r2d were significantly increased in the hippocampus of exposed animals allowed to enter REM, in comparison with control animals. Experience-dependent changes during sleep were not significant in the hippocampus for Bdnf, Camk4, Creb1, and Nr4a1, and no differences were detected between exposed and control SWS groups for any of the genes tested. No significant changes in gene expression were detected in the primary somatosensory cortex during sleep, in contrast with previous studies using longer post-stimulation intervals (>180 min). The experience-dependent induction of multiple plasticity-related genes in the hippocampus during early REM adds experimental support to the synaptic embossing theory.
Collapse
Affiliation(s)
- Julien Braga Calais
- Laboratório de Neurociências (LIM/27), Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (USP), Departamento e Instituto de Psiquiatria, São Paulo, Brazil; Laboratório Cesar Timo-Iaria, Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Elida Benquique Ojopi
- Laboratório de Neurociências (LIM/27), Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (USP), Departamento e Instituto de Psiquiatria, São Paulo, Brazil
| | - Edgard Morya
- Laboratório Cesar Timo-Iaria, Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, São Paulo, Brazil; Edmond and Lily Safra International Institute of Neuroscience of Natal (ELS-IINN), Natal, Brazil
| | - Koichi Sameshima
- Laboratório Cesar Timo-Iaria, Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, São Paulo, Brazil; Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil.
| | - Sidarta Ribeiro
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte (UFRN), Natal, Brazil.
| |
Collapse
|
80
|
Hurley SW, Johnson AK. The role of the lateral hypothalamus and orexin in ingestive behavior: a model for the translation of past experience and sensed deficits into motivated behaviors. Front Syst Neurosci 2014; 8:216. [PMID: 25431553 PMCID: PMC4230038 DOI: 10.3389/fnsys.2014.00216] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/13/2014] [Indexed: 01/23/2023] Open
Abstract
The hypothalamus has been recognized for its involvement in both maintaining homeostasis and mediating motivated behaviors. The present article discusses a region of the hypothalamus known as the lateral hypothalamic area (LHA). It is proposed that brain nuclei within the LHA including the dorsal region of the lateral hypothalamus (LHAd) and perifornical area (PeF) provide a link between neural systems that regulate homeostasis and those that mediate appetitive motivated behaviors. Functional and immunohistochemical data indicate that the LHA promotes many motivated behaviors including food intake, water intake, salt intake, and sexual behavior. Anatomical tracing experiments demonstrate that the LHA is positioned to receive inputs from brain areas involved in regulating body fluid and energy homeostasis. Regions within the LHA send dense projections to the ventral tegmental area (VTA), providing a pathway for the LHA to influence dopaminergic systems generally recognized to be involved in motivated behaviors and their reinforcement. Furthermore, the LHA contains neurons that synthesize orexin/hypocretin, a neuropeptide that promotes many appetitive motivated behaviors. The LHA also receives inputs from brain areas involved in reward-related learning and orexin neuron activation can become conditioned to environmental stimuli that are associated with rewards. Therefore, it is hypothesized that the LHA integrates signaling from areas that regulate body fluid and energy balance and reward-related learning. In turn, this information is “fed into” mesolimbic circuitry to influence the performance of motivated behaviors. This hypothesis may foster experiments that will result in an improved understanding of LHA function. An improved understanding of LHA function may aid in treating disorders that are associated with an excess or impairment in the expression of ingestive behavior including obesity, anorexia, impairments in thirst, salt gluttony, and salt deficiency.
Collapse
Affiliation(s)
- Seth W Hurley
- Department of Psychology, University of Iowa Iowa City, IA, USA
| | - Alan Kim Johnson
- Department of Psychology, University of Iowa Iowa City, IA, USA ; Department of Pharmacology, University of Iowa Iowa City, IA, USA ; Department of Health and Human Physiology, University of Iowa Iowa City, IA, USA ; François M. Abboud Cardiovascular Center, University of Iowa Iowa City, IA, USA
| |
Collapse
|
81
|
Akane H, Saito F, Shiraki A, Takeyoshi M, Imatanaka N, Itahashi M, Murakami T, Shibutani M. Downregulation of immediate-early genes linking to suppression of neuronal plasticity in rats after 28-day exposure to glycidol. Toxicol Appl Pharmacol 2014; 279:150-62. [DOI: 10.1016/j.taap.2014.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 05/05/2014] [Accepted: 05/28/2014] [Indexed: 11/26/2022]
|
82
|
l-DOPA reverses the impairment of Dentate Gyrus LTD in experimental parkinsonism via β-adrenergic receptors. Exp Neurol 2014; 261:377-85. [PMID: 25058044 DOI: 10.1016/j.expneurol.2014.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 05/30/2014] [Accepted: 07/08/2014] [Indexed: 11/20/2022]
Abstract
Parkinson's disease (PD) patients exhibit motor and non-motor symptoms that severely affect quality of life. Cognitive alterations in PD subjects have been related to both structural and functional hippocampal changes. Here we investigated the effects of the 6-hydroxydopamine (6-OHDA) lesion in the Medial Forebrain Bundle (MFB) on the hippocampus focusing on the Dentate Gyrus (DG). In vivo microdialysis measurements revealed that the 6-OHDA injection disrupts both dopaminergic and noradrenergic transmission in rat DG. In vitro electrophysiological recordings showed that these neurochemical alterations were accompanied by impairment of long-term depression (LTD) at medial perforant path/DG synapses. Furthermore, this alteration was reversed by l-DOPA treatment. Notably, the therapeutic effect of l-DOPA on LTD was blocked by the antagonism of β-noradrenergic receptors, but not by dopamine D1 or D2 receptor antagonists. Thus, while the dopaminergic transmission does not seem to be implicated in this therapeutic effect of l-DOPA, the noradrenergic system plays a central role in the synaptic dysfunction of the DG in experimental PD. Our work provides new evidence on the role of catecholamines in DG synaptic plasticity and sheds light on the possible synaptic mechanisms underlying cognitive deficits in PD. Furthermore, our results indicate that l-DOPA exerts a therapeutic effect on the parkinsonian brain through different, coexistent, mechanisms.
Collapse
|
83
|
Vousden DA, Epp J, Okuno H, Nieman BJ, van Eede M, Dazai J, Ragan T, Bito H, Frankland PW, Lerch JP, Henkelman RM. Whole-brain mapping of behaviourally induced neural activation in mice. Brain Struct Funct 2014; 220:2043-57. [PMID: 24760545 DOI: 10.1007/s00429-014-0774-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
Abstract
The ability to visualize behaviourally evoked neural activity patterns across the rodent brain is essential for understanding the distributed brain networks mediating particular behaviours. However, current imaging methods are limited in their spatial resolution and/or ability to obtain brain-wide coverage of functional activity. Here, we describe a new automated method for obtaining cellular-level, whole-brain maps of behaviourally induced neural activity in the mouse. This method combines the use of transgenic immediate-early gene reporter mice to visualize neural activity; serial two-photon tomography to image the entire brain at cellular resolution; advanced image processing algorithms to count the activated neurons and align the datasets to the Allen Mouse Brain Atlas; and statistical analysis to identify the network of activated brain regions evoked by behaviour. We demonstrate the use of this approach to determine the whole-brain networks activated during the retrieval of fear memories. Consistent with previous studies, we identified a large network of amygdalar, hippocampal, and neocortical brain regions implicated in fear memory retrieval. Our proposed methods can thus be used to map cellular networks involved in the expression of normal behaviours as well as to investigate in depth circuit dysfunction in mouse models of neurobiological disease.
Collapse
Affiliation(s)
- Dulcie A Vousden
- Mouse Imaging Centre, The Hospital for Sick Children, 25 Orde St., Toronto, ON M5T 3H7, Canada,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Distinct and simultaneously active plasticity mechanisms in mouse hippocampus during different phases of Morris water maze training. Brain Struct Funct 2014; 220:1273-90. [PMID: 24562414 DOI: 10.1007/s00429-014-0722-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 01/28/2014] [Indexed: 01/30/2023]
Abstract
Although the Morris water maze (MWM) is the most frequently used protocol to examine hippocampus-dependent learning in mice, not much is known about the spatio-temporal dynamics of underlying plasticity processes. Here, we studied molecular and cellular hippocampal plasticity mechanisms during early and late phases of spatial learning in the MWM. Quantitative in situ hybridization for the immediate early genes zif268 and Homer1a (H1a) revealed phase-dependent differences in their expression between areas CA1 and CA3. During the initial learning phase, CA1 expression levels of the molecular plasticity marker H1a, but not of the activity reporter gene zif268, were related to task proficiency; whereas no learning-specific changes could be detected in CA3. Simultaneously, the ratio of surface-expressed NMDAR subunits NR2A and NR2B was downregulated as measured by acute slice biotinylation assay, while the total number of surface NMDARs was unaltered. When intrinsic 'somatic' and synaptic plasticity in the CA1-region of hippocampal slices were examined, we found that early learning promotes intrinsic neuronal plasticity as manifested by a reduction of spike frequency adaptation and postburst afterhyperpolarization. At the synaptic level, however, maintenance of long-term potentiation (LTP) in all learning groups was impaired which is most likely due to 'intrinsic' learning-induced LTP which occluded any further electrically induced LTP. Late learning, in contrast, was characterized by re-normalized H1a, NR2A and NR2B expression and neuronal firing, yet a further strengthening of learning-induced LTP. Together, our data support a precisely timed cascade of complex molecular and subcellular transformations occurring from early to late MWM learning.
Collapse
|
85
|
Dyrvig M, Christiansen SH, Woldbye DPD, Lichota J. Temporal gene expression profile after acute electroconvulsive stimulation in the rat. Gene 2014; 539:8-14. [PMID: 24518690 DOI: 10.1016/j.gene.2014.01.072] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 01/17/2014] [Accepted: 01/25/2014] [Indexed: 01/20/2023]
Abstract
Electroconvulsive therapy (ECT) remains one of the most effective treatments of major depression. It has been suggested that the mechanisms of action involve gene expression. In recent decades there have been several investigations of gene expression following both acute and chronic electroconvulsive stimulation (ECS). These studies have focused on several distinct gene targets but have generally included only few time points after ECS for measuring gene expression. Here we measured gene expression of three types of genes: Immediate early genes, synaptic proteins, and neuropeptides at six time points following an acute ECS. We find significant increases for c-Fos, Egr1, Neuritin 1 (Nrn 1), Bdnf, Snap29, Synaptotagmin III (Syt 3), Synapsin I (Syn 1), and Psd95 at differing time points after ECS. For some genes these changes are prolonged whereas for others they are transient. Npy expression significantly increases whereas the gene expression of its receptors Npy1r, Npy2r, and Npy5r initially decreases. These decreases are followed by a significant increase for Npy2r, suggesting anticonvulsive adaptations following seizures. In summary, we find distinct changes in mRNA quantities that are characteristic for each gene. Considering the observed transitory and inverse changes in expression patterns, these data underline the importance of conducting measurements at several time points post-ECS.
Collapse
Affiliation(s)
- Mads Dyrvig
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Søren H Christiansen
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - David P D Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jacek Lichota
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
86
|
Viola H, Ballarini F, Martínez MC, Moncada D. The tagging and capture hypothesis from synapse to memory. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 122:391-423. [PMID: 24484708 DOI: 10.1016/b978-0-12-420170-5.00013-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The synaptic tagging and capture theory (STC) was postulated by Frey and Morris in 1997 and provided a strong framework to explain how to achieve synaptic specificity and persistence of electrophysiological-induced plasticity changes. Ten years later, the same argument was applied on learning and memory models to explain the formation of long-term memories, resulting in the behavioral tagging hypothesis (BT). These hypotheses are able to explain how a weak event that induces transient changes in the brain can establish long-lasting phenomena through a tagging and capture process. In this framework, it was postulated that the weak event sets a tag that captures plasticity-related proteins/products (PRPs) synthesized by an independent strong event. The tagging and capture processes exhibit symmetry, and therefore, PRPs can be captured if they are synthesized either before or after the setting of the tag. In summary, the hypothesis provides a wide framework that gives a solid explanation of how lasting changes occur and how the interaction between different events leads to promotion, reinforcement, or impairment of such changes. In this chapter, we will summarize the postulates of STC hypothesis, the common features between synaptic plasticity and memory, as well as a detailed compilation of the findings supporting the existence of BT process. At the end, we pose some questions related to BT mechanism and LTM formation, which probably will be answered in the near future.
Collapse
Affiliation(s)
- Haydée Viola
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis", Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina; Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fabricio Ballarini
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis", Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Cecilia Martínez
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis", Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Diego Moncada
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis", Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina; Neurophysiology of Learning and Memory Research Group, Leibniz-Institut für Neurobiologie, Magdeburg, Germany
| |
Collapse
|
87
|
Knipper M, Van Dijk P, Nunes I, Rüttiger L, Zimmermann U. Advances in the neurobiology of hearing disorders: Recent developments regarding the basis of tinnitus and hyperacusis. Prog Neurobiol 2013; 111:17-33. [DOI: 10.1016/j.pneurobio.2013.08.002] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/20/2013] [Accepted: 08/20/2013] [Indexed: 10/26/2022]
|
88
|
Gautam A, Wadhwa R, Thakur MK. Involvement of hippocampal Arc in amnesia and its recovery by alcoholic extract of Ashwagandha leaves. Neurobiol Learn Mem 2013; 106:177-84. [DOI: 10.1016/j.nlm.2013.08.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/19/2013] [Accepted: 08/21/2013] [Indexed: 12/19/2022]
|
89
|
Roy A, Jana M, Corbett GT, Ramaswamy S, Kordower JH, Gonzalez FJ, Pahan K. Regulation of cyclic AMP response element binding and hippocampal plasticity-related genes by peroxisome proliferator-activated receptor α. Cell Rep 2013; 4:724-37. [PMID: 23972989 DOI: 10.1016/j.celrep.2013.07.028] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/28/2013] [Accepted: 07/18/2013] [Indexed: 01/19/2023] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a transcription factor that regulates genes involved in fatty acid catabolism. Here, we provide evidence that PPARα is constitutively expressed in nuclei of hippocampal neurons and, surprisingly, controls calcium influx and the expression of various plasticity-related genes via direct transcriptional regulation of cyclic AMP response element binding (CREB). Accordingly, Pparα-null, but not Pparβ-null, mice are deficient in CREB and memory-associated proteins and have decreased spatial learning and memory. Small hairpin RNA knockdown of PPARα in the hippocampus suppressed CREB and NR2A, rendering wild-type animals markedly poor in consolidating spatial memory, whereas introduction of PPARα to the hippocampus of Pparα-null mice increased hippocampal CREB and NR2A and improved spatial learning and memory. Through detailed analyses of CREB and NR2A activity, as well as spatial learning and memory in bone marrow chimeric animals lacking PPARα in the CNS, we uncover a mechanism for transcriptional control of Creb and associated plasticity genes by PPARα.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
90
|
Kádár E, Huguet G, Aldavert-Vera L, Morgado-Bernal I, Segura-Torres P. Intracranial self stimulation upregulates the expression of synaptic plasticity related genes and Arc protein expression in rat hippocampus. GENES BRAIN AND BEHAVIOR 2013; 12:771-9. [PMID: 23898803 DOI: 10.1111/gbb.12065] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/31/2013] [Accepted: 07/25/2013] [Indexed: 11/30/2022]
Abstract
Post-training lateral hypothalamus (LH) intracranial self stimulation (ICSS) has a reliable enhancing effect on explicit memory formation evaluated in hippocampus-dependent tasks such as the Morris water maze. In this study, the effects of ICSS on gene expression in the hippocampus are examined 4.5 h post treatment by using oligonucleotide microarray and real-time PCR, and by measuring Arc protein levels in the different layers of hippocampal subfields through immunofluorescence. The microarray data analysis resulted in 65 significantly regulated genes in rat ICSS hippocampi compared to sham, including cAMP-mediated signaling as one of the most significantly enriched Database for Annotation, Visualization and Integrated Discovery (DAVID) functional categories. In particular, expression of CREB-dependent synaptic plasticity related genes (c-Fos, Arc, Bdnf, Ptgs-2 and Crem and Icer) was regulated in a time-dependent manner following treatment administration. Immunofluorescence results showed that ICSS treatment induced a significant increase in Arc protein expression in CA1 and DG hippocampal subfields. This empirical evidence supports our hypothesis that the effect of ICSS on improved or restored memory functions might be mediated by increased hippocampal expression of activity-dependent synaptic plasticity related genes, including Arc protein expression, as neural mechanisms related to memory consolidation.
Collapse
Affiliation(s)
- E Kádár
- Departament de Biologia, Universitat de Girona, Girona, Spain
| | | | | | | | | |
Collapse
|
91
|
Li M, Liu WJ, Lu B, Wang YH, Liu JG. Differential expression of Arc in the mesocorticolimbic system is involved in drug and natural rewarding behavior in rats. Acta Pharmacol Sin 2013; 34:1013-24. [PMID: 23708554 DOI: 10.1038/aps.2013.28] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 03/06/2013] [Indexed: 12/25/2022] Open
Abstract
AIM To investigate the different effects of heroin and milk in activating the corticostriatal system that plays a critical role in reward reinforcement learning. METHODS Male SD rats were trained daily for 15 d to self-administer heroin or milk tablets in a classic runway drug self-administration model. Immunohistochemical assay was used to quantify Arc protein expression in the medial prefrontal cortex (mPFC), the nucleus accumbens (NAc), the dorsomedial striatum (DMS) and the ventrolateral striatum (VLS) in response to chronic self-administration of heroin or milk tablets. NMDA receptor antagonist MK801 (0.1 mg/kg) or dopamine D1 receptor antagonist SCH23390 (0.03 mg/kg) were intravenously injected at the same time as heroin was infused intravenously. RESULTS Runway training with heroin resulted in robust enhancement of Arc expression in the mPFC, the NAc and the DMS on d 1, 7, and 15, and in the VLS on d 1 and d 7. However, runway training with milk led to increased Arc expression in the mPFC, the NAc and the DMS only on d 7 and/or d 15 but not on d 1. Moreover, runway training with milk failed to induce increased Arc protein in the VLS. Both heroin-seeking behavior and Arc protein expression were blocked by MK801 or SCH23390 administration. CONCLUSION The VLS is likely to be critically involved in drug-seeking behavior. The NMDA- and D1 receptor-dependent Arc expression is important in drug-seeking behavior.
Collapse
|
92
|
Molteni R, Macchi F, Riva MA. Gene expression profiling as functional readout of rodent models for psychiatric disorders. Cell Tissue Res 2013; 354:51-60. [DOI: 10.1007/s00441-013-1648-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 04/24/2013] [Indexed: 10/26/2022]
|
93
|
Christensen DZ, Thomsen MS, Mikkelsen JD. Reduced basal and novelty-induced levels of activity-regulated cytoskeleton associated protein (Arc) and c-Fos mRNA in the cerebral cortex and hippocampus of APPswe/PS1ΔE9 transgenic mice. Neurochem Int 2013; 63:54-60. [PMID: 23598246 DOI: 10.1016/j.neuint.2013.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 03/26/2013] [Accepted: 04/06/2013] [Indexed: 02/04/2023]
Abstract
Activity-regulated cytoskeletal-associated protein (Arc) and c-Fos are immediate early gene (IEG) products induced by novelty in the hippocampus and involved in the consolidation of synaptic plasticity and long-term memory. We investigated whether induction of arc and c-fos after exposure to a novel open field environment was compromised in different neocortical areas and the hippocampal formation in APP/PS1ΔE9 transgenic mice characterized by pronounced accumulation and deposition of beta amyloid (Aβ). Notably, the basal level of Arc and c-fos mRNA in the neocortex was significantly lower in APP/PS1ΔE9 compared to wild-type mice. Novelty exposure induced an increase in Arc and c-Fos mRNA in the medial prefrontal cortex (mPFC), parietal cortex, and hippocampal formation in both APP/PS1ΔE9 transgenic and wild-type mice. However, novelty-induced IEG expression did not reach the same levels in APP/PS1ΔE9 as in the wild-type mice. In contrast, synaptophysin levels did not differ between mutant and wild type mice, suggesting that the observed effect was not due to a general decrease in the number of presynapses. These data suggest a reduction in basal and novelty-induced neuronal activity in a transgenic mouse model of Alzheimer's disease, which is most pronounced in cortical regions, indicating that a decreased functional response in IEG expression could be partly responsible for the cognitive deficits observed in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Ditte Z Christensen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Denmark
| | | | | |
Collapse
|
94
|
Zhang F, Zhu Q, Xue Q, Luo Y, Yu B. Extra-cellular signal-regulated kinase (ERK) is inactivated associating hippocampal ARC protein up-regulation in sevoflurane induced bidirectional regulation of memory. Neurochem Res 2013; 38:1341-7. [PMID: 23572329 DOI: 10.1007/s11064-013-1027-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Revised: 03/04/2013] [Accepted: 03/23/2013] [Indexed: 11/27/2022]
Abstract
Low dose sevoflurane is demonstrated to have neuronal excitatory effects in the central nervous system. Activity-regulated cytoskeleton protein (Arc) can be rapidly expressed in the hippocampus for the modulation of synaptic plasticity. The extracellular signal-regulated kinase (ERK) pathway is also involved in learning and memory by mediating signals and modifications. This study aims at exploring the mechanism of sevoflurane on memory by connecting the ERK pathway, Arc and inhibitory avoidance (IA) behavioral training. SD rats were randomly assigned to three groups (sham, 0.11 and 0.3 % SEV). Anesthesia was given by target dose of sevoflurane for 45 min and IA (0.4 mA, 2 s) was conducted on every subject immediately after inhalation. The memory retention latency was observed 24 h after. Another serial of rats were sacrificed for Western-blot and polymerase chain reaction (PCR) examination of hippocampal tissue after first IA. 24 h IA performance was compared among groups. The 0.11 % SEV group displayed an elevation of memory retention, while the 0.3 % SEV group had decreased memory retention, both showed statistical differences from the sham (air) group. PCR analysis of Arc mRNA levels showed that subanesthetic doses of sevoflurane did not change Arc transcription levels between groups. However, 0.11 % sevoflurane significantly increased Arc protein in the hippocampus, while 0.3 % sevoflurane reversed this (*P < 0.05, compared with the sham group). There was no difference in total ERK between groups. Expression of phosphorylated ERK was significantly increased upon exposure to sevoflurane in a does dependent manner. ERK was down-regulated with hippocampal ARC expression in sevoflurane induced bidirectional regulation of memory, potentially at a translational level of modification.
Collapse
Affiliation(s)
- Fujun Zhang
- Department of Anesthesiology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | | | | | | | | |
Collapse
|
95
|
Scheffner M, Kumar S. Mammalian HECT ubiquitin-protein ligases: biological and pathophysiological aspects. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:61-74. [PMID: 23545411 DOI: 10.1016/j.bbamcr.2013.03.024] [Citation(s) in RCA: 217] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/18/2013] [Accepted: 03/22/2013] [Indexed: 10/27/2022]
Abstract
Members of the HECT family of E3 ubiquitin-protein ligases are characterized by a C-terminal HECT domain that catalyzes the covalent attachment of ubiquitin to substrate proteins and by N-terminal extensions of variable length and domain architecture that determine the substrate spectrum of a respective HECT E3. Since their discovery in 1995, it has become clear that deregulation of distinct HECT E3s plays an eminent role in human disease or disease-related processes including cancer, cardiovascular and neurological disorders, viral infections, and immune response. Thus, a detailed understanding of the structure-function aspects of HECT E3s as well as the identification and characterization of the substrates and regulators of HECT E3s is critical in developing new approaches in the treatment of respective diseases. In this review, we summarize what is currently known about mammalian HECT E3s, with a focus on their biological functions and roles in pathophysiology.This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.
Collapse
Affiliation(s)
- Martin Scheffner
- Department of Biology, Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany.
| | | |
Collapse
|
96
|
The reduced cochlear output and the failure to adapt the central auditory response causes tinnitus in noise exposed rats. PLoS One 2013; 8:e57247. [PMID: 23516401 PMCID: PMC3596376 DOI: 10.1371/journal.pone.0057247] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 01/18/2013] [Indexed: 01/15/2023] Open
Abstract
Tinnitus is proposed to be caused by decreased central input from the cochlea, followed by increased spontaneous and evoked subcortical activity that is interpreted as compensation for increased responsiveness of central auditory circuits. We compared equally noise exposed rats separated into groups with and without tinnitus for differences in brain responsiveness relative to the degree of deafferentation in the periphery. We analyzed (1) the number of CtBP2/RIBEYE-positive particles in ribbon synapses of the inner hair cell (IHC) as a measure for deafferentation; (2) the fine structure of the amplitudes of auditory brainstem responses (ABR) reflecting differences in sound responses following decreased auditory nerve activity and (3) the expression of the activity-regulated gene Arc in the auditory cortex (AC) to identify long-lasting central activity following sensory deprivation. Following moderate trauma, 30% of animals exhibited tinnitus, similar to the tinnitus prevalence among hearing impaired humans. Although both tinnitus and no-tinnitus animals exhibited a reduced ABR wave I amplitude (generated by primary auditory nerve fibers), IHCs ribbon loss and high-frequency hearing impairment was more severe in tinnitus animals, associated with significantly reduced amplitudes of the more centrally generated wave IV and V and less intense staining of Arc mRNA and protein in the AC. The observed severe IHCs ribbon loss, the minimal restoration of ABR wave size, and reduced cortical Arc expression suggest that tinnitus is linked to a failure to adapt central circuits to reduced cochlear input.
Collapse
|
97
|
Xie L, Korkmaz KS, Braun K, Bock J. Early life stress-induced histone acetylations correlate with activation of the synaptic plasticity genes Arc and Egr1 in the mouse hippocampus. J Neurochem 2013; 125:457-64. [PMID: 23413876 DOI: 10.1111/jnc.12210] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 01/30/2013] [Accepted: 02/07/2013] [Indexed: 11/30/2022]
Abstract
Early life stress (ELS) programs the developing organism and influences the development of brain and behavior. We tested the hypothesis that ELS-induced histone acetylations might alter the expression of synaptic plasticity genes that are critically involved in the establishment of limbic brain circuits. Maternal separation (MS) from postnatal day 14-16 was applied as ELS and two immediate early genes underlying experience-induced synaptic plasticity, Arc and early growth response 1 (Egr1) were analyzed. We show here that repeated ELS induces a rapid increase of Arc and Egr1 in the mouse hippocampus. Furthermore, immunoblotting revealed that these changes are paralleled by histone modifications, reflected by increased acetylation levels of H3 and H4. Most importantly, using native Chromatin immunoprecipitation quantitative PCR (nChIP-qPCR), we show for the first time a correlation between elevated histone acetylation and increased Arc and Egr1 expression in response to ELS. These rapid epigenetic changes are paralleled by increases of dendritic complexity and spine number of hippocampal CA3 pyramidal neurons in ELS animals at weaning age. Our results are in line with our working hypothesis that ELS induces activation of synaptic plasticity genes, mediated by epigenetic mechanisms. These events are assumed to represent early steps in the adaption of neuronal networks to a stressful environment.
Collapse
Affiliation(s)
- Lan Xie
- Research Group Structural Plasticity, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | | | | | | |
Collapse
|
98
|
Kerrigan TL, Randall AD. A new player in the "synaptopathy" of Alzheimer's disease - arc/arg 3.1. Front Neurol 2013; 4:9. [PMID: 23407382 PMCID: PMC3570765 DOI: 10.3389/fneur.2013.00009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 01/26/2013] [Indexed: 01/08/2023] Open
Abstract
Alzheimer’s disease (AD) is increasingly referred to as a “synaptopathy.” This moniker reflects the loss or damage of synapses that occurs as the disease progresses, which in turn produces functional degeneration of specific neuronal circuits and consequent aberrant activity in neural networks. Accumulating evidence supports the functional importance of the early-expression activity-regulated cytoskeletal (Arc) gene in regulating memory consolidation. Interestingly, AD patients express anomalously high levels of Arc protein. Arc physically associates with presenilin1, a pivotal protease for the generation of Amyloid β (Aβ) peptides. Arc expression itself is disrupted in the vicinity of Aβ oligomers and plaques. Such alterations result in the interruption of neuronal network integration in vivo. It is not clear what the impacts of these alterations are on the functional neurophysiology of transgenic mouse models of AD-associated amyloidopathy. Our group and others have described alterations to neuronal excitability and thus intrinsic firing within these transgenic mice models. This brief review will emphasize the rising role of Arc and its involvement in neurophysiological alterations of current AD models.
Collapse
Affiliation(s)
- Talitha L Kerrigan
- School of Physiology and Pharmacology, Faculty of Medical and Veterinary Sciences, University of Bristol Bristol, UK
| | | |
Collapse
|
99
|
Besnard A, Laroche S, Caboche J. Comparative dynamics of MAPK/ERK signalling components and immediate early genes in the hippocampus and amygdala following contextual fear conditioning and retrieval. Brain Struct Funct 2013; 219:415-30. [PMID: 23389809 DOI: 10.1007/s00429-013-0505-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 01/06/2013] [Indexed: 12/01/2022]
Abstract
Over the past few years multiple studies have attempted to uncover molecular signatures of memory reconsolidation when compared to consolidation. In the present study we used immunocytochemical detection of the MAPK/ERK1/2 pathway, to track activated neuronal circuits in the hippocampus and amygdala recruited during the consolidation and reconsolidation of a contextual fear conditioning (CFC) memory. We report selective differences in magnitude and temporal dynamics of activated ERK1/2 signalling in different subregions of these two structures between the post-training and post-retrieval periods, except in the dentate gyrus, where the patterns of activation were similar. We then focused on this brain area to dissect out the patterns of downstream ERK1/2 signalling components, including the phosphorylation of MSK-1 and histone H3 on ser10, along with the induction of the Immediate Early Genes (IEGs) Arc/Arg3.1, c-Fos and Zif268/Egr1 following CFC training and retrieval. We found that the completion of the nucleosomal response as well as the induction of IEGs shorter during the reconsolidation period as compared to consolidation. Our results shed new light on the cellular mechanisms underlying the consolidation and reconsolidation processes engaged following CFC training and retrieval and further extend the notion that memory reconsolidation is not mechanistically a repetition of consolidation. In addition, we provide evidence that the strength of a previously established CFC memory is characterized by distinct patterns of ERK1/2 activation in different hippocampal and amygdalar subfields upon CFC memory recall. Our results emphasize the differences between consolidation and reconsolidation processes in relation to contextual fear memories.
Collapse
Affiliation(s)
- Antoine Besnard
- INSERM, UMRS 952, Physiopathologie des Maladies du Système Nerveux Central, 9 Quai Saint Bernard, 75005, Paris, France
| | | | | |
Collapse
|
100
|
Singer W, Geisler HS, Knipper M. The Geisler method: tracing activity-dependent cGMP plasticity changes upon double detection of mRNA and protein on brain slices. Methods Mol Biol 2013; 1020:223-33. [PMID: 23709037 DOI: 10.1007/978-1-62703-459-3_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We recently demonstrated that an increase of guanosine 3',5'-cyclic monophosphate (cGMP) signaling could protect the inner ear from noise-induced hair cell damage. Noise exposure not only damages hair cells but also alters the central responsiveness to sound leading to plasticity changes. cGMP signaling has long been known to play a crucial role for plasticity changes and long-term potentiation (LTP). To get a first insight into the role of cGMP for noise-induced plasticity changes we aimed to co-trace the mRNA and protein of plasticity-related genes as, e.g., the immediate early gene Arc (activity-regulated cytoskeletal protein) with markers for the cGMP pathway. We developed a method that permits the simultaneous monitoring of mRNA and protein through light microscopy to visualize gene expression in neurons and synapses of its processes. Accordingly, different from previous fluorescence-based assays that detect, e.g., fluorochrome-labeled Arc antibodies and Arc mRNA, we describe here a methodology that allows the detection of mRNA and protein of synaptic genes using nonfluorescent stable tracers for high-resolution observation of activity-dependent plasticity changes using light microscopy even after weeks or months.
Collapse
Affiliation(s)
- Wibke Singer
- Department of Otolaryngology, Hearing Research Centre Tübingen (THRC), Molecular Physiology of Hearing, Universität Tübingen, Tübingen, Germany
| | | | | |
Collapse
|