1
|
Arai M, Osanai H, Snell CC, Kitamura T, Ogawa SK. Combinative protein expression of immediate early genes c-Fos, Arc, and Npas4 along aversive- and reward-related neural networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.21.649441. [PMID: 40475597 PMCID: PMC12139746 DOI: 10.1101/2025.04.21.649441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2025]
Abstract
Expression of immediate early genes (IEGs) is critical for memory formation and has been widely used to identify the neural substrate of memory traces, termed memory engram cells. Functions of IEGs have been known to be different depending on their types. However, there is limited knowledge about the extent to which different types of IEGs are selectively or concurrently involved in the formation of memory engram. To address this question, we investigated the combinative expression of c-Fos, Arc, and Npas4 proteins using immunohistochemistry following aversive and rewarding experiences across subregions in the prefrontal cortex (PFC), basolateral amygdala (BLA), hippocampal dentate gyrus (DG), and retrosplenial cortex (RSC). Using an automated cell detection algorithm, we found that expression patterns of c-Fos, Npas4, and Arc varied across different brain areas, with a higher increase of IEG expressing cells in the PFC and posterior BLA than in the DG. The combinative expression patterns, along with their learning-induced changes, also differed across brain areas; the co-expression of IEGs increased in the PFC and BLA following learning whereas the increase was less pronounced in the DG and RSC. Furthermore, we demonstrate that different area-to-area functional connectivity networks were extracted by different IEGs. These findings provide insights into how different IEGs and their combinations identify engram cells, which will contribute to a deeper understanding of the functional significance of IEG-tagged memory engram cells.
Collapse
Affiliation(s)
- Mary Arai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hisayuki Osanai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chris C. Snell
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sachie K. Ogawa
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
2
|
Eom K, Kim D, Hyun JH. Engram and behavior: How memory is stored in the brain. Neurobiol Learn Mem 2025; 219:108047. [PMID: 40074071 DOI: 10.1016/j.nlm.2025.108047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/04/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
During the processing of information in humans, activated neurons behave in a specific way. The activity of these neurons leaves traces on the neurons, such as changes in synaptic or intrinsic properties. Formation of the memory traces is associated with molecular changes in the neurons. Hence, monitoring collective neural activities and following the trace of neural activities are important to neuroscience research. This collective or group of neurons is described as a 'neural ensemble', while the neural trace is described as a 'neural engram'. Both terms have been used and studied by neuroscientists for a long time. In this article, we discuss the development of these concepts, current research methods, and future areas of development.
Collapse
Affiliation(s)
- Kisang Eom
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Donguk Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jung Ho Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea; Biomedical Sciences & Engineering Major of Interdisciplinary Studies, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, Republic of Korea.
| |
Collapse
|
3
|
Nagahama K, Jung VH, Kwon HB. Cutting-edge methodologies for tagging and tracing active neuronal coding in the brain. Curr Opin Neurobiol 2025; 92:102997. [PMID: 40056794 DOI: 10.1016/j.conb.2025.102997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/09/2025] [Accepted: 02/14/2025] [Indexed: 03/10/2025]
Abstract
Decoding the neural substrates that underlie learning and behavior is a fundamental goal in neuroscience. Identifying "key players" at the molecular, cellular, and circuit levels has become possible with recent advancements in molecular technologies offering high spatiotemporal resolution. Immediate-early genes are effective markers of neural activity and plasticity, allowing for the identification of active cells involved in memory-based behavior. A calcium-dependent labeling system coupled with light or biochemical proximity labeling allows characterization of active cell ensembles and circuitry across broader brain regions within short time windows, particularly during transient behaviors. The integration of these systems expands the ability to address diverse research questions across behavioral paradigms. This review examines current molecular systems for activity-dependent labeling, highlighting their applications in identifying specific cell ensembles and circuits relevant to various scientific questions and further discuss their significance, along with future directions for the development of innovative methodologies.
Collapse
Affiliation(s)
- Kenichiro Nagahama
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Veronica Hyeyoon Jung
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hyung-Bae Kwon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
4
|
Rahimi K, Abbaszadeh M, Bakhtazad S, Ghotbeddin Z. Effects of dimethyl itaconate on expressions of NGFI-A and NGFI-B and inflammatory cytokines in the spinal cord in the formalin test. Brain Commun 2024; 6:fcae397. [PMID: 39568551 PMCID: PMC11577613 DOI: 10.1093/braincomms/fcae397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/06/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024] Open
Abstract
Neural sensitization can cause neuroinflammation, which is a type of inflammation that occurs in both the peripheral nervous system and central nervous system. The purpose of this study was to investigate the effect of dimethyl itaconate (DMI) on the expression of NGFI-A and NGFI-B and inflammatory cytokines in the spinal cord in the formalin test. The rats were divided into five groups: control, formalin, DMI 10 mg/kg + formalin, DMI 20 mg/kg + formalin and diclofenac sodium 10 mg/kg + formalin. We evaluated the impact of DMI on the spinal cords NGFI-A and NGFI-B expressions and inflammatory and anti-inflammatory cytokines [interleukin-1 beta (IL-1β), tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and interleukin-10 (IL-10)]. The findings indicate that DMI 10, DMI 20 and diclofenac sodium 10 mg/kg can relieve pain in rats during the formalin test. In addition, these substances were found to reduce the expression of NGFI-A and NGFI-B in the spinal cord. Moreover, DMI 10, DMI 20 and diclofenac sodium 10 mg/kg were observed to increase the expression of IL-10 while decreasing IL-1β, TNF-α and IL-6 in the spinal cord when compared with the formalin group. We have found that administering DMI can alleviate pain in rats during formalin test. Through our research, we have observed that DMI decreases the expression of NGFI-A and NGFI-B in the spinal cord. Furthermore, DMI has been shown to increase the levels of IL-10 while decreasing IL-1β, TNF-α and IL-6 in the spinal cord.
Collapse
Affiliation(s)
- Kaveh Rahimi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Abbaszadeh
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Sharareh Bakhtazad
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Zohreh Ghotbeddin
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Stem Cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
5
|
Rahmi U, Goenawan H, Sylviana N, Setiawan I, Putri ST, Andriyani S, Fitriana LA. Exercise induction at expression immediate early gene (c-Fos, ARC, EGR-1) in the hippocampus: a systematic review. Dement Neuropsychol 2024; 18:e20230015. [PMID: 38628561 PMCID: PMC11019719 DOI: 10.1590/1980-5764-dn-2023-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 11/06/2023] [Accepted: 11/17/2023] [Indexed: 04/19/2024] Open
Abstract
The immediate early gene exhibits activation markers in the nervous system consisting of ARC, EGR-1, and c-Fos and is related to synaptic plasticity, especially in the hippocampus. Immediate early gene expression is affected by physical exercise, which induces direct ARC, EGR-1, and c-Fos expression. Objective To assess the impact of exercise, we conducted a literature study to determine the expression levels of immediate early genes (ARC, c-Fos, and EGR-1). Methods The databases accessed for online literature included PubMed-Medline, Scopus, and ScienceDirect. The original English articles were selected using the following keywords in the title: (Exercise OR physical activity) AND (c-Fos) AND (Hippocampus), (Exercise OR physical activity) AND (ARC) AND (Hippocampus), (Exercise OR physical activity) AND (EGR-1 OR zif268) AND (Hippocampus). Results Physical exercise can affect the expression of EGR-1, c-Fos, and ARC in the hippocampus, an important part of the brain involved in learning and memory. High-intensity physical exercise can increase c-Fos expression, indicating neural activation. Furthermore, the expression of the ARC gene also increases due to physical exercise. ARC is a gene that plays a role in synaptic plasticity and regulation of learning and memory, changes in synaptic structure and increased synaptic connections, while EGR-1 also plays a role in synaptic plasticity, a genetic change that affects learning and memory. Overall, exercise or regular physical exercise can increase the expression of ARC, c-Fos, and EGR-1 in the hippocampus. This reflects the changes in neuroplasticity and synaptic plasticity that occur in response to physical activity. These changes can improve cognitive function, learning, and memory. Conclusion c-Fos, EGR-1, and ARC expression increases in hippocampal neurons after exercise, enhancing synaptic plasticity and neurogenesis associated with learning and memory.
Collapse
Affiliation(s)
- Upik Rahmi
- Universitas Pendidikan Indonesia, Department of Nursing, Bandung, West Java, Indonesia
- Universitas Padjadjaran, Department of Medicine, Bandung, West Java, Indonesia
| | - Hanna Goenawan
- Universitas Padjadjaran, Department of Medicine, Bandung, West Java, Indonesia
| | - Nova Sylviana
- Universitas Padjadjaran, Department of Medicine, Bandung, West Java, Indonesia
| | - Iwan Setiawan
- Universitas Padjadjaran, Department of Medicine, Bandung, West Java, Indonesia
| | - Suci Tuty Putri
- Universitas Pendidikan Indonesia, Department of Nursing, Bandung, West Java, Indonesia
| | - Septian Andriyani
- Universitas Pendidikan Indonesia, Department of Nursing, Bandung, West Java, Indonesia
| | - Lisna Anisa Fitriana
- Universitas Pendidikan Indonesia, Department of Nursing, Bandung, West Java, Indonesia
| |
Collapse
|
6
|
Granja-Galeano G, Dominguez-Rubio AP, Zappia CD, Wolfson M, Sanz-Blasco S, Aisemberg J, Zorrilla-Zubilete M, Fernandez N, Franchi A, Fitzsimons CP, Monczor F. CB1 receptor expression and signaling are required for dexamethasone-induced aversive memory consolidation. Neuropharmacology 2023; 239:109674. [PMID: 37541383 DOI: 10.1016/j.neuropharm.2023.109674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/12/2023] [Accepted: 07/27/2023] [Indexed: 08/06/2023]
Abstract
The molecular processes that underlie long-term memory formation involve signaling pathway activation by neurotransmitter release, which induces the expression of immediate early genes, such as Zif268, having a key role in memory formation. In this work, we show that the cannabinoid CB1 receptor signaling is necessary for the effects of dexamethasone on the behavioral response in an inhibitory avoidance task, on dexamethasone-induced ERK phosphorylation, and on dexamethasone-dependent Zif268 expression. Furthermore, we provide primary evidence for the mechanism responsible for this crosstalk between cannabinoid and glucocorticoid-mediated signaling pathways, showing that dexamethasone regulates endocannabinoid metabolism by inhibiting the activity of the Fatty acid amide hydrolase (FAAH), an integral membrane enzyme that hydrolyzes endocannabinoids and related amidated signaling lipids. Our results provide novel evidence regarding the role of the endocannabinoid system, and in particular of the CB1 receptor, as a mediator of the effects of glucocorticoids on the consolidation of aversive memories.
Collapse
Affiliation(s)
- Gina Granja-Galeano
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, 1113, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, 1113, Argentina
| | - Ana Paula Dominguez-Rubio
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA/CONICET). Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - C Daniel Zappia
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, 1113, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, 1113, Argentina
| | - Manuel Wolfson
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA/CONICET). Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sara Sanz-Blasco
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, 1113, Argentina
| | - Julieta Aisemberg
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA/CONICET). Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Zorrilla-Zubilete
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA/CONICET). Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Fernandez
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, 1113, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, 1113, Argentina
| | - Ana Franchi
- Laboratorio de Fisiopatología de la Preñez y el Parto, Centro de Estudios Farmacológicos y Botánicos (CEFyBO-UBA/CONICET). Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos P Fitzsimons
- Center for Neuroscience, Faculty of Science, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Federico Monczor
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, 1113, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, 1113, Argentina.
| |
Collapse
|
7
|
Openshaw RL, Thomson DM, Bristow GC, Mitchell EJ, Pratt JA, Morris BJ, Dawson N. 16p11.2 deletion mice exhibit compromised fronto-temporal connectivity, GABAergic dysfunction, and enhanced attentional ability. Commun Biol 2023; 6:557. [PMID: 37225770 DOI: 10.1038/s42003-023-04891-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/01/2023] [Indexed: 05/26/2023] Open
Abstract
Autism spectrum disorders are more common in males, and have a substantial genetic component. Chromosomal 16p11.2 deletions in particular carry strong genetic risk for autism, yet their neurobiological impact is poorly characterised, particularly at the integrated systems level. Here we show that mice reproducing this deletion (16p11.2 DEL mice) have reduced GABAergic interneuron gene expression (decreased parvalbumin mRNA in orbitofrontal cortex, and male-specific decreases in Gad67 mRNA in parietal and insular cortex and medial septum). Metabolic activity was increased in medial septum, and in its efferent targets: mammillary body and (males only) subiculum. Functional connectivity was altered between orbitofrontal, insular and auditory cortex, and between septum and hippocampus/subiculum. Consistent with this circuit dysfunction, 16p11.2 DEL mice showed reduced prepulse inhibition, but enhanced performance in the continuous performance test of attentional ability. Level 1 autistic individuals show similarly heightened performance in the equivalent human test, also associated with parietal, insular-orbitofrontal and septo-subicular dysfunction. The data implicate cortical and septal GABAergic dysfunction, and resulting connectivity changes, as the cause of pre-attentional and attentional changes in autism.
Collapse
Affiliation(s)
- Rebecca L Openshaw
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK
| | - David M Thomson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Greg C Bristow
- Department of Biomedical and Life Sciences, Lancaster University, Lancaster, LA1 4YW, UK
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Emma J Mitchell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Brian J Morris
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir James Black Building, Glasgow, G12 8QQ, UK.
| | - Neil Dawson
- Department of Biomedical and Life Sciences, Lancaster University, Lancaster, LA1 4YW, UK.
| |
Collapse
|
8
|
Gilissen SRJ, Hennes M, Arckens L. Analysis of Immediate Early Gene Expression Levels to Interrogate Changes in Cortical Neuronal Activity Patterns upon Vision Loss. Methods Mol Biol 2023; 2636:55-70. [PMID: 36881295 DOI: 10.1007/978-1-0716-3012-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Mapping immediate early gene (IEG) expression levels to characterize changes in neuronal activity patterns has become a golden standard in neuroscience research. Due to straightforward detection methods such as in situ hybridization and immunohistochemistry, changes in IEG expression can be easily visualized across brain regions and in response to physiological and pathological stimulation. Based on in-house experience and existing literature, zif268 represents itself as the IEG of choice to investigate the neuronal activity dynamics induced by sensory deprivation. In the monocular enucleation mouse model of partial vision loss, zif268 in situ hybridization can be implemented to study cross-modal plasticity by charting the initial decline and subsequent rise in neuronal activity in visual cortical territory deprived of direct retinal visual input. Here, we describe a protocol for high-throughput radioactive zif268 in situ hybridization as a readout for cortical neuronal activity dynamics in response to partial vision loss in mice.
Collapse
Affiliation(s)
- Sara R J Gilissen
- Department of Biology & Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Maroussia Hennes
- Department of Biology & Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Lutgarde Arckens
- Department of Biology & Leuven Brain Institute, KU Leuven, Leuven, Belgium.
- Laboratory of Neuroplasticity and Neuroproteomics, Katholieke Universiteit Leuven, Leuven, Belgium.
| |
Collapse
|
9
|
Iwakura Y, Kawahara-Miki R, Kida S, Sotoyama H, Gabdulkhaev R, Takahashi H, Kunii Y, Hino M, Nagaoka A, Izumi R, Shishido R, Someya T, Yabe H, Kakita A, Nawa H. Elevation of EGR1/zif268, a Neural Activity Marker, in the Auditory Cortex of Patients with Schizophrenia and its Animal Model. Neurochem Res 2022; 47:2715-2727. [PMID: 35469366 DOI: 10.1007/s11064-022-03599-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023]
Abstract
The family of epidermal growth factor (EGF) including neuregulin-1 are implicated in the neuropathology of schizophrenia. We established a rat model of schizophrenia by exposing perinatal rats to EGF and reported that the auditory pathophysiological traits of this model such as prepulse inhibition, auditory steady-state response, and mismatch negativity are relevant to those of schizophrenia. We assessed the activation status of the auditory cortex in this model, as well as that in patients with schizophrenia, by monitoring the three neural activity-induced proteins: EGR1 (zif268), c-fos, and Arc. Among the activity markers, protein levels of EGR1 were significantly higher at the adult stage in EGF model rats than those in control rats. The group difference was observed despite an EGF model rat and a control rat being housed together, ruling out the contribution of rat vocalization effects. These changes in EGR1 levels were seen to be specific to the auditory cortex of this model. The increase in EGR1 levels were detectable at the juvenile stage and continued until old ages but displayed a peak immediately after puberty, whereas c-fos and Arc levels were nearly indistinguishable between groups at all ages with an exception of Arc decrease at the juvenile stage. A similar increase in EGR1 levels was observed in the postmortem superior temporal cortex of patients with schizophrenia. The commonality of the EGR1 increase indicates that the EGR1 elevation in the auditory cortex might be one of the molecular signatures of this animal model and schizophrenia associating with hallucination.
Collapse
Affiliation(s)
- Yuriko Iwakura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan.
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, 1-757 Asahimachi-Dori, Chuo-ku, Niigata City, Niigata, 951-8585, Japan.
| | | | - Satoshi Kida
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Bioscience, Faculty of Life Science, Tokyo University of Agriculture, Tokyo, Japan
| | - Hidekazu Sotoyama
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Ramil Gabdulkhaev
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hitoshi Takahashi
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yasuto Kunii
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Mizuki Hino
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Atsuko Nagaoka
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Ryuta Izumi
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Risa Shishido
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Toshiyuki Someya
- Department of Psychiatry, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirooki Yabe
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
10
|
Bristow GC, Thomson DM, Openshaw RL, Mitchell EJ, Pratt JA, Dawson N, Morris BJ. 16p11 Duplication Disrupts Hippocampal-Orbitofrontal-Amygdala Connectivity, Revealing a Neural Circuit Endophenotype for Schizophrenia. Cell Rep 2021; 31:107536. [PMID: 32320645 DOI: 10.1016/j.celrep.2020.107536] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/18/2020] [Accepted: 03/28/2020] [Indexed: 02/07/2023] Open
Abstract
Chromosome 16p11.2 duplications dramatically increase risk for schizophrenia, but the mechanisms remain largely unknown. Here, we show that mice with an equivalent genetic mutation (16p11.2 duplication mice) exhibit impaired hippocampal-orbitofrontal and hippocampal-amygdala functional connectivity. Expression of schizophrenia-relevant GABAergic cell markers (parvalbumin and calbindin) is selectively decreased in orbitofrontal cortex, while somatostatin expression is decreased in lateral amygdala. When 16p11.2 duplication mice are tested in cognitive tasks dependent on hippocampal-orbitofrontal connectivity, performance is impaired in an 8-arm maze "N-back" working memory task and in a touchscreen continuous performance task. Consistent with hippocampal-amygdala dysconnectivity, deficits in ethologically relevant social behaviors are also observed. Overall, the cellular/molecular, brain network, and behavioral alterations markedly mirror those observed in schizophrenia patients. Moreover, the data suggest that 16p11.2 duplications selectively impact hippocampal-amygdaloid-orbitofrontal circuitry, supporting emerging ideas that dysfunction in this network is a core element of schizophrenia and defining a neural circuit endophenotype for the disease.
Collapse
Affiliation(s)
- Greg C Bristow
- Department of Biomedical and Life Sciences, University of Lancaster, Lancaster LA1 4YW, UK
| | - David M Thomson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Rebecca L Openshaw
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow G12 8QQ, UK
| | - Emma J Mitchell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Neil Dawson
- Department of Biomedical and Life Sciences, University of Lancaster, Lancaster LA1 4YW, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow G12 8QQ, UK.
| |
Collapse
|
11
|
c-Fos and FosB/ΔFosB colocalizations in selected forebrain structures after olanzapine, amisulpride, aripiprazole, and quetiapine single administration in rats preconditioned by two different mild stressors sequences. Endocr Regul 2020; 54:43-52. [PMID: 32597143 DOI: 10.2478/enr-2020-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE Olanzapine (OLA), amisulpride (AMI), aripiprazole (ARI), and quetiapine (QUE) belong to antipsychotics, which administration represents still most reliable way for the treatment of schizophrenic and bipolar disorders. The intention of the present study was to explore whether the acute administration of a particular antipsychotic, indicated by the presence of c-Fos, will: a) stimulate neurons already activated by a long lasting homogeneous or heterogeneous stress preconditioning, indicated by the FosB/ΔFosB (ΔFosB) expression, or b) have a stimulatory effect only on a not activated, so called silent neurons. The pattern of ΔFosB and c-Fos spatial relationship was investigated in three forebrain structures, including the septal ventrolateral nucleus (seVL), the striatal dorsolateral area (stDL), and the shell of the nucleus accumbens (shell). METHODS The rats were divided into 10 groups and exposed to two types of stressors. Half of them was exposed to a sequence of homogeneous stressor - handling (HDL) and the other half to a heterogeneous stressor (CMS) daily for 20 days. CMS consisted of five types of stressors: crowding, air-puff, wet bedding, predator stress, and forced swimming applied in an unexpected order. On the 21st day of the experiment, the rats were free of the stress exposure and on the 22nd day, both groups of animals receive a single intraperitoneal injection of vehicle (4% DMSO in saline, 0.1 ml/100 g) or OLA (5 mg/kg), AMI (20 mg/kg), ARI (10 mg/kg), and QUE (15 mg/kg). 90 min after the drugs administration the animals were transcardially perfused, brains removed, cut into 30 µm thick coronal sections, and double stained: first with ΔFosB antibody linked with Alexa488 fluorescent dye and second with c-Fos antibody linked to Alexa596 one. Quantitative evaluation of ΔFosB and c-Fos colocalizations was performed on fluorescence photomicrographs transformed into a final picture containing only yellow, green, and red colored circles. RESULTS The data of this investigation demonstrate that ΔFosB and c-Fos colocalizations occurred in each of the three areas investigated, i.e. seVL, stDL, and shell ones, in both HDL as well as CMS preconditioned rats. The levels of ΔFosB and c-Fos colocalizations varied in the individual forebrain areas studied. From the total 22 areas measured, level of c-Fos colocalization prevailed over ΔFosB in 18 ones. However, neither c-Fos nor ΔFosB reached 100% level of colocalization in any of the forebrain areas investigated. CONCLUSION The present findings indicate that ΔFosB and c-Fos colocalizations occurred in each of the three areas investigated, i.e. seVL, stDL, and shell, in both HDL and CMS preconditioned rats, whereas the parallel occurrence of free c-Fos as well as c-Fos colocalized with ΔFosB might speak out for a possible involvement of the c-Fos activated by antipsychotics applied in dual, i.e. short- and long-lasting, functions.
Collapse
|
12
|
Brito I, Britto LRG, Ferrari EAM. Retrieval of contextual aversive memory and induction of Zenk expression in the hippocampus of pigeons. Brain Res Bull 2019; 153:341-349. [PMID: 31586459 DOI: 10.1016/j.brainresbull.2019.09.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/08/2019] [Accepted: 09/27/2019] [Indexed: 02/05/2023]
Abstract
The hippocampus has a fundamental role in many learning and memory processes, which include the formation and retrieval of context-fear associations, as evidenced by studies in rodents and birds. The present paper has analyzed contextual memory and Zenk expression in the hippocampus of the pigeon after fear conditioning. Pigeons were trained under four conditions: with 3 tone-shock associations (Paired), with shock and tone presented randomly (Unpaired), with exposure to the experimental chamber without stimulation (Control) and with only daily handling (Naive). The testing was conducted 24 h after training. All sessions were digitally recorded. The level of freezing expressed by the Paired and Unpaired groups differed significantly from that of the control group during both training and test sessions. Pigeons from the Paired group revealed a significantly greater density of Zenk positive nuclei in the ventromedial region of the hippocampus than did the Unpaired, Control and Naive groups. These data suggest that Zenk-mediated processes of synaptic plasticity in the hippocampus are induced during the retrieval of conditioned fear memory in the pigeon.
Collapse
Affiliation(s)
- Ivana Brito
- School of Arts, Sciences and Humanities, São Paulo University, São Paulo, Brazil.
| | | | | |
Collapse
|
13
|
Saalfield J, Spear L. Fos activation patterns related to acute ethanol and conditioned taste aversion in adolescent and adult rats. Alcohol 2019; 78:57-68. [PMID: 30797833 DOI: 10.1016/j.alcohol.2019.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/22/2019] [Accepted: 02/13/2019] [Indexed: 12/27/2022]
Abstract
Studies in rats have revealed marked age differences in sensitivity to the aversive properties of ethanol, with a developmental insensitivity to ethanol aversion that is most pronounced during pre- and early adolescence, declining thereafter to reach the enhanced aversive sensitivity of adults. The adolescent brain undergoes significant transitions throughout adolescence, including in regions linked with drug reward and aversion; however, it is unknown how ontogenetic changes within this reward/aversion circuitry contribute to developmental differences in aversive sensitivity. The current study examined early adolescent (postnatal day [P]28-30) and adult (P72-74) Sprague-Dawley male rats for conditioned taste aversion (CTA) after doses of 0, 1.0, or 2.5 g/kg ethanol, and patterns of neuronal activation in response to ethanol using Fos-like immunohistochemistry (Fos+) to uncover regions where age differences in activation are associated with ethanol aversion. An adolescent-specific ethanol-induced increase in Fos+ staining was seen within the nucleus accumbens shell and core. An age difference was also noted within the Edinger-Westphal nucleus (EW) following administration of the lower dose of ethanol, with 1 g/kg ethanol producing CTA in adults but not in adolescents and inducing a greater EW Fos response in adults than adolescents. Regression analysis revealed that greater numbers of Fos+ neurons within the EW and insula (Ins) were related to lower consumption of the conditioned stimulus (CS) on test day (reflecting greater CTA). Some regionally specific age differences in Fos+ were noted under baseline conditions, with adolescents displaying fewer Fos+ neurons than adults within the prelimbic (PrL) cortex, but more than adults in the bed nucleus of the stria terminalis (BNST). In the BNST (but not PrL), ethanol-induced increases in Fos-immunoreactivity (IR) were evident at both ages. Increased ethanol-induced activity within critical appetitive brain regions (NAc core and shell) supports a role for greater reward-related activation during adolescence, possibly along with attenuated responsiveness to ethanol in EW and Ins in the age-typical resistance of adolescents to the aversive properties of ethanol.
Collapse
|
14
|
c-Fos expression response to olanzapine, amisulpride, aripiprazole, and quetiapine single administration in the rat forebrain: Effect of a mild stress preconditioning. Neurochem Int 2019; 126:187-194. [PMID: 30905743 DOI: 10.1016/j.neuint.2019.03.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/26/2019] [Accepted: 03/18/2019] [Indexed: 12/20/2022]
Abstract
Antipsychotics have been shown to stimulate different forebrain areas, whereas some of them are sensitive to stress. In the present study, effect of a single administration of olanzapine (OLA), amisulpride (AMI), aripiprazole (ARI), and quetiapine (QUE) on the activity of cells in the striatal dorsolateral (stDL) area, the periventricular zone (peVZ), the septal ventrolateral (seVL) nucleus, and the accumbens nucleus shell (shACC) and core (coACC) was investigated in male rats preconditioned with a mild stress complex (CMS) for 20 days. The objective of the study was to extend the anatomical-functional knowledge on the mechanism of selected antipsychotics with the goals: 1) to analyze the ability of the selected antipsychotics to induce c-Fos protein expression in the above mentioned forebrain structures and to map the pattern of their topography and 2) to find out whether longer-lasting mild stress preconditioning may modify the impact of the selected antipsychotics on the activity of cells in the forebrain areas in adult rats. Ten groups of rats were used. CMS complex contained five stressors: cage crowding, air-puff noising, wet bedding, predator stress, and forced swimming. AMI (20 mg/kg), OLA (5 mg/kg), QUE (15 mg/kg), and ARI (10 mg/kg/b.w.) were administered intraperitoneally and 90 min later the animals transcardially perfused by fixative. c-Fos was visualized by ABC complex. In unstressed animals, OLA and ARI elevated c-Fos expression in all areas studied, AMI and QUE in all areas except stDL, seVL and coACC, shACC FL-2 (shACC posterior level), respectively. CMS potentiated the effect of AMI in coACC, and QUE in shACC FL-2 and suppressed the effect of AMI in peVZ, and ARI in peVZ and seVL. The present data provide new insights into activity of cells in response to CMS challenge, which might be helpful in understanding the diverse clinical effects of atypical antipsychotics.
Collapse
|
15
|
Yagi S, Galea LAM. Sex differences in hippocampal cognition and neurogenesis. Neuropsychopharmacology 2019; 44:200-213. [PMID: 30214058 PMCID: PMC6235970 DOI: 10.1038/s41386-018-0208-4] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022]
Abstract
Sex differences are reported in hippocampal plasticity, cognition, and in a number of disorders that target the integrity of the hippocampus. For example, meta-analyses reveal that males outperform females on hippocampus-dependent tasks in rodents and in humans, furthermore women are more likely to experience greater cognitive decline in Alzheimer's disease and depression, both diseases characterized by hippocampal dysfunction. The hippocampus is a highly plastic structure, important for processing higher order information and is sensitive to the environmental factors such as stress. The structure retains the ability to produce new neurons and this process plays an important role in pattern separation, proactive interference, and cognitive flexibility. Intriguingly, there are prominent sex differences in the level of neurogenesis and the activation of new neurons in response to hippocampus-dependent cognitive tasks in rodents. However, sex differences in spatial performance can be nuanced as animal studies have demonstrated that there are task, and strategy choice dependent sex differences in performance, as well as sex differences in the subregions of the hippocampus influenced by learning. This review discusses sex differences in pattern separation, pattern completion, spatial learning, and links between adult neurogenesis and these cognitive functions of the hippocampus. We emphasize the importance of including both sexes when studying genomic, cellular, and structural mechanisms of the hippocampal function.
Collapse
Affiliation(s)
- Shunya Yagi
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Liisa A M Galea
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
16
|
LaVallee J, Grant T, D'Angelo-Early S, Kletsov S, Berry NA, Abt KM, Bloch CP, Muscedere ML, Adams KW. Refining the nuclear localization signal within the Egr transcriptional coregulator NAB2. FEBS Lett 2018; 593:107-118. [PMID: 30411343 DOI: 10.1002/1873-3468.13288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/29/2018] [Accepted: 11/02/2018] [Indexed: 01/09/2023]
Abstract
NAB1 and 2 are coregulators for early growth response (Egr) transcription factors. The NAB1 nuclear localization signal (NLS) was previously described as a bipartite NLS of sequence R(X2 )K(X11 )KRXK. The sequence is conserved in NAB2 as K(X2 )R(X11 )KKXK; however, whether it functions as the NAB2 NLS has not been tested. We show that the KKXK motif in NAB2 is necessary and sufficient to mediate nuclear localization. Mutation of the KKXK motif to AAXA causes cytoplasmic localization of NAB2, while Lys/Arg-to-Ala mutations of the upstream K(X2 )R motif have no effect. Fusion of the KKXK motif to cytoplasmic protein eIF2Bε causes nuclear localization. Altogether, this study refines our knowledge of the NAB2 NLS, demonstrating that KKXK343-346 is necessary and sufficient for nuclear localization.
Collapse
Affiliation(s)
- Jacquelyn LaVallee
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Terrain Grant
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | | | - Sergey Kletsov
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Nicole A Berry
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Kimberly M Abt
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Christopher P Bloch
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | | | - Kenneth W Adams
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| |
Collapse
|
17
|
Kyrke-Smith M, Williams JM. Bridging Synaptic and Epigenetic Maintenance Mechanisms of the Engram. Front Mol Neurosci 2018; 11:369. [PMID: 30344478 PMCID: PMC6182070 DOI: 10.3389/fnmol.2018.00369] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/18/2018] [Indexed: 12/27/2022] Open
Abstract
How memories are maintained, and how memories are lost during aging or disease, are intensely investigated issues. Arguably, the reigning theory is that synaptic modifications allow for the formation of engrams during learning, and sustaining engrams sustains memory. Activity-regulated gene expression profiles have been shown to be critical to these processes, and their control by the epigenome has begun to be investigated in earnest. Here, we propose a novel theory as to how engrams are sustained. We propose that many of the genes that are currently believed to underlie long-term memory are actually part of a “plasticity transcriptome” that underpins structural and functional modifications to neuronal connectivity during the hours to days following learning. Further, we hypothesize that a “maintenance transcriptome” is subsequently induced that includes epigenetic negative regulators of gene expression, particularly histone deacetylases. The maintenance transcriptome negatively regulates the plasticity transcriptome, and thus the plastic capability of a neuron, after learning. In this way, the maintenance transcriptome would act as a metaplasticity mechanism that raises the threshold for change in neurons within an engram, helping to ensure the connectivity is stabilized and memory is maintained.
Collapse
Affiliation(s)
- Madeleine Kyrke-Smith
- Department of Anatomy, The Brain Health Research Centre, Brain Research New Zealand - Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand.,Department of Psychology, The Brain Health Research Centre, Brain Research New Zealand - Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand.,Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, United States
| | - Joanna M Williams
- Department of Anatomy, The Brain Health Research Centre, Brain Research New Zealand - Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
Gallo FT, Katche C, Morici JF, Medina JH, Weisstaub NV. Immediate Early Genes, Memory and Psychiatric Disorders: Focus on c-Fos, Egr1 and Arc. Front Behav Neurosci 2018; 12:79. [PMID: 29755331 PMCID: PMC5932360 DOI: 10.3389/fnbeh.2018.00079] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/10/2018] [Indexed: 01/08/2023] Open
Abstract
Many psychiatric disorders, despite their specific characteristics, share deficits in the cognitive domain including executive functions, emotional control and memory. However, memory deficits have been in many cases undervalued compared with other characteristics. The expression of Immediate Early Genes (IEGs) such as, c-fos, Egr1 and arc are selectively and promptly upregulated in learning and memory among neuronal subpopulations in regions associated with these processes. Changes in expression in these genes have been observed in recognition, working and fear related memories across the brain. Despite the enormous amount of data supporting changes in their expression during learning and memory and the importance of those cognitive processes in psychiatric conditions, there are very few studies analyzing the direct implication of the IEGs in mental illnesses. In this review, we discuss the role of some of the most relevant IEGs in relation with memory processes affected in psychiatric conditions.
Collapse
Affiliation(s)
- Francisco T Gallo
- Instituto de Fisiología y Biofísica Bernardo Houssay, Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Cynthia Katche
- Instituto de Biología Celular y Neurociencias (IBCN) Dr. Eduardo de Robertis, Facultad de Medicina, CONICET, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Juan F Morici
- Instituto de Fisiología y Biofísica Bernardo Houssay, Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Jorge H Medina
- Instituto de Biología Celular y Neurociencias (IBCN) Dr. Eduardo de Robertis, Facultad de Medicina, CONICET, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos (UBA), Buenos Aires, Argentina
| | - Noelia V Weisstaub
- Instituto de Fisiología y Biofísica Bernardo Houssay, Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|
19
|
Synapse development organized by neuronal activity-regulated immediate-early genes. Exp Mol Med 2018; 50:1-7. [PMID: 29628504 PMCID: PMC5938016 DOI: 10.1038/s12276-018-0025-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
Classical studies have shown that neuronal immediate-early genes (IEGs) play important roles in synaptic processes critical for key brain functions. IEGs are transiently activated and rapidly upregulated in discrete neurons in response to a wide variety of cellular stimuli, and they are uniquely involved in various aspects of synapse development. In this review, we summarize recent studies of a subset of neuronal IEGs in regulating synapse formation, transmission, and plasticity. We also discuss how the dysregulation of neuronal IEGs is associated with the onset of various brain disorders and pinpoint key outstanding questions that should be addressed in this field. Immediate-early genes (IEGs), genes that are rapidly and transiently activated by cellular stimuli, regulate the interactions between neurons and key brain functions. Ji Won Um and colleagues at Daegu Gyeongbuk Institute of Science and Technology in South Korea review recent studies on three IEGs that are activated by neuronal activity and highlight their contribution to neuronal excitability and cognitive behaviors. These genes rely on different molecular mechanisms to regulate neuronal receptors and the structure of synapses. Research in mice lacking any one of these IEGs reveals their contribution to learning and memory as well as to some behavioral abnormalities associated with neuropsychiatric disorders. Further research into the activity of IEGs will advance our understanding of how a neuron’s environment influences brain development and disease.
Collapse
|
20
|
Eagle AL, Gajewski PA, Robison AJ. Role of hippocampal activity-induced transcription in memory consolidation. Rev Neurosci 2018; 27:559-73. [PMID: 27180338 DOI: 10.1515/revneuro-2016-0010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 03/26/2016] [Indexed: 01/15/2023]
Abstract
Experience-dependent changes in the strength of connections between neurons in the hippocampus (HPC) are critical for normal learning and memory consolidation, and disruption of this process drives a variety of neurological and psychiatric diseases. Proper HPC function relies upon discrete changes in gene expression driven by transcription factors (TFs) induced by neuronal activity. Here, we describe the induction and function of many of the most well-studied HPC TFs, including cyclic-AMP response element binding protein, serum-response factor, AP-1, and others, and describe their role in the learning process. We also discuss the known target genes of many of these TFs and the purported mechanisms by which they regulate long-term changes in HPC synaptic strength. Moreover, we propose that future research in this field will depend upon unbiased identification of additional gene targets for these activity-dependent TFs and subsequent meta-analyses that identify common genes or pathways regulated by multiple TFs in the HPC during learning or disease.
Collapse
|
21
|
Zhu QL, Luo Y, Xue QS, Zhang FJ, Yu BW. Different doses of sevoflurane facilitate and impair learning and memory function through activation of the ERK pathway and synthesis of ARC protein in the rat hippocampus. Brain Res 2017; 1678:174-179. [PMID: 29074343 DOI: 10.1016/j.brainres.2017.10.019] [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: 06/04/2017] [Revised: 10/04/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sevoflurane has been shown to stimulate or depress memory in adult rats; however, the cellular mechanism of this bidirectional effect has not been fully investigated. METHODS We used an intra-hippocampal microinfusion of U0126 to suppress ERK activation. Male SD rats were randomly assigned to four groups: Sham, 0.11%SEV, 0.3%SEV and 0.3%+U0126. They received bilateral injections of U0126 or saline. Rats were anesthetized, and Inhibitory Avoidance (IA) training was performed immediately after anesthesia. The memory retention latency was observed 24 h later. In another experiment, the hippocampus was removed 45 min after IA training to assess ARC expression, the synapsin 1 protein levels and the phosphorylation level of ERK. RESULTS Treatment with 0.11%SEV led to rapid phosphorylation of ERK, while 0.3%SEV inhibited phosphorylation; the latter change was reversed by the microinfusion of U0126 in the hippocampus. The memory latency result had similar tendencies. The local infusion of U0126 abolished the 0.3%SEV-induced memory impairment and ERK inhibition. Selective upregulations of ARC and synapsin 1 proteins were observed in the 0.3%SEV group compared with the 0.11%SEV group. CONCLUSIONS The results indicate that different doses of sevoflurane trigger synaptic plasticity-related cytoskeleton proteins through the ERK signaling pathway. This novel modulation by inhalational agents may help to reduce their side-effects on memory function.
Collapse
Affiliation(s)
- Qian-Lin Zhu
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Sheng Xue
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fu-Jun Zhang
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bu-Wei Yu
- Department of Anesthesiology, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Yu LN, Sun LH, Wang M, Yan M. Research progress of the role and mechanism of extracellular signal-regulated protein kinase 5 (ERK5) pathway in pathological pain. J Zhejiang Univ Sci B 2017; 17:733-741. [PMID: 27704743 DOI: 10.1631/jzus.b1600188] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Extracellular signal-regulated protein kinase 5 (ERK5), also known as big mitogen-activated protein kinase 1 (MAPK1), is an important member of ERK family, which is a subfamily of the large MAPK family. ERK5 is expressed in many tissues, including the dorsal root ganglion (DRG) neurons and the spinal cord. In this review, we focus on elaborating ERK5-associated pathway in pathological pain, in which the ERK5/CREB (cyclic adenosine monophosphate (cAMP)-response element-binding protein) pathway plays a crucial role in the transduction of pain signal and contributes to pain hypersensitivity. ERK5 activation in the spinal dorsal horn occurs mainly in microglia. The activation of ERK5 can be mediated by N-methyl-D-aspartate (NMDA) receptors. We also elaborate the relationship between ERK5 activation and nerve growth factor-tyrosine kinase A (NGF-TrkA), and the connection between ERK5 activation and brain-derived neurotrophic factor (BDNF) in pathological pain in detail.
Collapse
Affiliation(s)
- Li-Na Yu
- Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Li-Hong Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221000, China
| | - Min Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221000, China
| | - Min Yan
- Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.,Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221000, China
| |
Collapse
|
23
|
Gröger N, Mannewitz A, Bock J, de Schultz TF, Guttmann K, Poeggel G, Braun K. Infant avoidance training alters cellular activation patterns in prefronto-limbic circuits during adult avoidance learning: I. Cellular imaging of neurons expressing the synaptic plasticity early growth response protein 1 (Egr1). Brain Struct Funct 2017; 222:3639-3651. [DOI: 10.1007/s00429-017-1423-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/03/2017] [Indexed: 12/24/2022]
|
24
|
Basinou V, Park JS, Cederroth CR, Canlon B. Circadian regulation of auditory function. Hear Res 2017; 347:47-55. [PMID: 27665709 PMCID: PMC5364078 DOI: 10.1016/j.heares.2016.08.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/25/2016] [Indexed: 12/11/2022]
Abstract
The circadian system integrates environmental cues to regulate physiological functions in a temporal fashion. The suprachiasmatic nucleus, located in the hypothalamus, is the master clock that synchronizes central and peripheral organ clocks to orchestrate physiological functions. Recently, molecular clock machinery has been identified in the cochlea unravelling the potential involvement in the circadian regulation of auditory functions. Here, we present background information on the circadian system and review the recent findings that introduce circadian rhythms to the auditory field. Understanding the mechanisms by which circadian rhythms regulate auditory function will provide fundamental knowledge on the signalling networks that control vulnerability and resilience to auditory insults.
Collapse
Affiliation(s)
- Vasiliki Basinou
- Department of Physiology and Pharmacology, Laboratory of Experimental Audiology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jung-Sub Park
- Department of Physiology and Pharmacology, Laboratory of Experimental Audiology, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Otolaryngology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon, 16499, South Korea
| | - Christopher R Cederroth
- Department of Physiology and Pharmacology, Laboratory of Experimental Audiology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Laboratory of Experimental Audiology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
25
|
Duclot F, Kabbaj M. The Role of Early Growth Response 1 (EGR1) in Brain Plasticity and Neuropsychiatric Disorders. Front Behav Neurosci 2017; 11:35. [PMID: 28321184 PMCID: PMC5337695 DOI: 10.3389/fnbeh.2017.00035] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/21/2017] [Indexed: 12/11/2022] Open
Abstract
It is now clearly established that complex interactions between genes and environment are involved in multiple aspects of neuropsychiatric disorders, from determining an individual's vulnerability to onset, to influencing its response to therapeutic intervention. In this perspective, it appears crucial to better understand how the organism reacts to environmental stimuli and provide a coordinated and adapted response. In the central nervous system, neuronal plasticity and neurotransmission are among the major processes integrating such complex interactions between genes and environmental stimuli. In particular, immediate early genes (IEGs) are critical components of these interactions as they provide the molecular framework for a rapid and dynamic response to neuronal activity while opening the possibility for a lasting and sustained adaptation through regulation of the expression of a wide range of genes. As a result, IEGs have been tightly associated with neuronal activity as well as a variety of higher order processes within the central nervous system such as learning, memory and sensitivity to reward. The immediate early gene and transcription factor early growth response 1 (EGR1) has thus been revealed as a major mediator and regulator of synaptic plasticity and neuronal activity in both physiological and pathological conditions. In this review article, we will focus on the role of EGR1 in the central nervous system. First, we will summarize the different factors influencing its activity. Then, we will analyze the amount of data, including genome-wide, that has emerged in the recent years describing the wide variety of genes, pathways and biological functions regulated directly or indirectly by EGR1. We will thus be able to gain better insights into the mechanisms underlying EGR1's functions in physiological neuronal activity. Finally, we will discuss and illustrate the role of EGR1 in pathological states with a particular interest in cognitive functions and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Florian Duclot
- Department of Biomedical Sciences, Florida State UniversityTallahassee, FL, USA; Program in Neuroscience, Florida State UniversityTallahassee, FL, USA
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State UniversityTallahassee, FL, USA; Program in Neuroscience, Florida State UniversityTallahassee, FL, USA
| |
Collapse
|
26
|
Adams KW, Kletsov S, Lamm RJ, Elman JS, Mullenbrock S, Cooper GM. Role for Egr1 in the Transcriptional Program Associated with Neuronal Differentiation of PC12 Cells. PLoS One 2017; 12:e0170076. [PMID: 28076410 PMCID: PMC5226839 DOI: 10.1371/journal.pone.0170076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/28/2016] [Indexed: 11/17/2022] Open
Abstract
PC12 cells are a well-established model to study how differences in signal transduction duration can elicit distinct cell behaviors. Epidermal growth factor (EGF) activates transient ERK signaling in PC12 cells that lasts 30–60 min, which in turn promotes proliferation; nerve growth factor (NGF) activates more sustained ERK signaling that lasts 4–6 h, which in turns induces neuronal differentiation. Data presented here extend a previous study by Mullenbrock et al. (2011) that demonstrated that sustained ERK signaling in response to NGF induces preferential expression of a 69-member gene set compared to transient ERK signaling in response to EGF and that the transcription factors AP-1 and CREB play a major role in the preferential expression of several genes within the set. Here, we examined whether the Egr family of transcription factors also contributes to the preferential expression of the gene set in response to NGF. Our data demonstrate that NGF causes transient induction of all Egr family member transcripts, but a corresponding induction of protein was detected for only Egr1 and 2. Chromatin immunoprecipitation experiments provided clearest evidence that, after induction, Egr1 binds 12 of the 69 genes that are preferentially expressed during sustained ERK signaling. In addition, Egr1 expression and binding upstream of its target genes were both sustained in response to NGF versus EGF within the same timeframe that its targets are preferentially expressed. These data thus provide evidence that Egr1 contributes to the transcriptional program activated by sustained ERK signaling in response to NGF, specifically by contributing to the preferential expression of its target genes identified here.
Collapse
Affiliation(s)
- Kenneth W Adams
- Department of Biological Sciences, Bridgewater State University, Bridgewater, Massachusetts, United States of America
| | - Sergey Kletsov
- Department of Biological Sciences, Bridgewater State University, Bridgewater, Massachusetts, United States of America
| | - Ryan J Lamm
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Jessica S Elman
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Steven Mullenbrock
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Geoffrey M Cooper
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
| |
Collapse
|
27
|
Dossat AM, Jourdi H, Wright KN, Strong CE, Sarkar A, Kabbaj M. Viral-mediated Zif268 expression in the prefrontal cortex protects against gonadectomy-induced working memory, long-term memory, and social interaction deficits in male rats. Neuroscience 2017; 340:243-257. [PMID: 27816701 PMCID: PMC5154846 DOI: 10.1016/j.neuroscience.2016.10.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/17/2016] [Accepted: 10/25/2016] [Indexed: 01/28/2023]
Abstract
In humans, some males experience reductions in testosterone levels, as a natural consequence of aging or in the clinical condition termed hypogonadism, which are associated with impaired cognitive performance and mood disorder(s). Some of these behavioral deficits can be reversed by testosterone treatment. Our previous work in rats reported that sex differences in the expression of the transcription factor Zif268, a downstream target of testosterone, within the medial prefrontal cortex (mPFC) mediates sex differences in social interaction. In the present study, we aimed to examine the effects of gonadectomy (GNX) in male rats on mPFC Zif268 expression, mood and cognitive behaviors. We also examined whether reinstitution of Zif268 in GNX rats will correct some of the behavioral deficits observed following GNX. Our results show that GNX induced a downregulation of Zif268 protein in the mPFC, which was concomitant with impaired memory in the y-maze and spontaneous object recognition test, reduced social interaction time, and depression-like behaviors in the forced swim test. Reinstitution of mPFC Zif268, using a novel adeno-associated-viral (AAV) construct, abrogated GNX-induced working memory and long-term memory impairments, and reductions in social interaction time, but not GNX-induced depression-like behaviors. These findings suggest that mPFC Zif268 exerts beneficial effects on memory and social interaction, and could be a potential target for novel treatments for behavioral impairments observed in hypogonadal and aged men with declining levels of gonadal hormones.
Collapse
Affiliation(s)
- Amanda M Dossat
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, United States
| | - Hussam Jourdi
- Department of Biology, Faculty of Science, University of Balamand, Souk-El-Gharb, Mount-Lebanon, Lebanon
| | - Katherine N Wright
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, United States
| | - Caroline E Strong
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, United States
| | - Ambalika Sarkar
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, United States
| | - Mohamed Kabbaj
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, United States.
| |
Collapse
|
28
|
Tochiki KK, Maiarú M, Norris C, Hunt SP, Géranton SM. The mitogen and stress-activated protein kinase 1 regulates the rapid epigenetic tagging of dorsal horn neurons and nocifensive behaviour. Pain 2016; 157:2594-2604. [PMID: 27482631 PMCID: PMC5065054 DOI: 10.1097/j.pain.0000000000000679] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/15/2022]
Abstract
Phosphorylation of histone H3 at serine 10 (p-H3S10) is a marker of active gene transcription. Using cognitive models of neural plasticity, p-H3S10 was shown to be downstream of extracellular signal-regulated kinase (ERK) signalling in the hippocampus. In this study, we show that nociceptive signalling after peripheral formalin injection increased p-H3S10 expression in the ipsilateral dorsal horn. This increase was maximal 30 minutes after formalin injection and occurred mainly within p-ERK-positive neurons. Spinal p-H3S10-enhanced expression was also observed in neurokinin 1 receptor (NK1R), c-Fos, and Zif268 positive neurons and was inhibited by ablation of serotonergic descending controls. The mitogen and stress-activated protein kinase 1 (MSK1) is downstream of ERK and can induce p-H3S10. We found that, after formalin injection, most phospho-MSK1 (p-MSK1)-positive cells (87% ± 3%) expressed p-ERK and the majority of p-H3S10-positive cells (85% ± 5%) expressed p-MSK1. Inhibition of ERK activity with the MEK inhibitor SL327 reduced formalin-induced p-ERK, p-MSK1, and p-H3S10, demonstrating that spinal p-MSK1 and p-H3S10 were at least partly downstream of ERK signalling. Crucially, pharmacological blockade of spinal MSK1 activity with the novel MSK1 inhibitor SB727651A inhibited formalin-induced spinal p-H3S10 and nocifensive behaviour. These findings are the first to establish the involvement of p-H3S10 and its main kinase, MSK1, in ERK regulation of nociception. Given the general importance of ERK signalling in pain processing, our results suggest that p-H3S10 could play a role in the response to injury.
Collapse
Affiliation(s)
- Keri K. Tochiki
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Maria Maiarú
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Caspar Norris
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Stephen P. Hunt
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Sandrine M. Géranton
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
29
|
Impaired hippocampal synaptic plasticity and NR2A/2B expression ratio in remifentanil withdrawal rats. Neurotoxicology 2016; 53:115-123. [PMID: 26777139 DOI: 10.1016/j.neuro.2016.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/05/2015] [Accepted: 01/09/2016] [Indexed: 11/20/2022]
Abstract
Remifentanil is a kind of synthetic opioid which has gained wide clinical acceptance by anesthesiologists. In this study, we attempted to test whether withdrawal effects on learning mechanisms can be triggered by repeated low-dose remifentanil treatment. Male Sprague-Dawley (SD) rats were subjected to remifentanil (50μg/kgs.c.) twice per day at 12h intervals for 15 days. When the animals of remifentanil group were withdrawn from remifentanil at 10h after the last injection, changes in open field test, Morris water maze test (MWM) and synaptic efficacy were examined in each group. We demonstrated that repeated exposure to 50μg/kg remifentanil produced enhanced locomotor activity indicating that a remifentanil addiction animal model in rats was established. MWM results showed that exposure to remifentanil had no influence on the spatial cognition. After withdrawal of remifentanil rats showed impaired spatial cognition. In electrophysiology test, remifentanil group rats showed a trend for a rightward shift of input/output relationship and significant deficits in maintenance of STP and LTP. Immunohistochemistry results demonstrated increased NR2A/NR2B ratio that should be included depression of LTP. In the whole-cell patch-clamp recording, after elimination from remifentanil incubation, mEPSC frequency was down regulated in hippocampal CA1 neurons, indicating that basal synaptic transmission were affected by remifentanil withdrawal. Taken together, the current findings demonstrate that the remifentanil withdrawn rats exhibit obvious impairment of hippocampus-dependent memory and synaptic plasticity. Increased hippocampal NR2A/NR2B expression ratio and the changes of basal synaptic transmission may participate in the impairment of LTP.
Collapse
|
30
|
Bojovic O, Bramham CR, Tjølsen A. Stimulation-induced expression of immediate early gene proteins in the dorsal horn is increased in neuropathy. Scand J Pain 2016; 10:43-51. [PMID: 28361770 DOI: 10.1016/j.sjpain.2015.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 09/05/2015] [Accepted: 09/08/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Peripheral neuropathic pain is described as a pain state caused by an injury or dysfunction of the nervous system, and could have clinical manifestations such as hyperalgesia, allodynia and spontaneous pain. The development of neuropathic pain may depend on long-term forms of neuronal plasticity in the spinal cord (SC). Expression of the immediate early gene proteins (IEGPs) Arc, Zif268, and c-Fos are implicated in establishment of long-term potentiation (LTP) induced by conditioning stimulation (CS) of primary afferent fibres. However, the impact of the neuropathic state (Bennett's model) on CS-induced expression of IEGPs has not been studied. The aim of this study was to compare the levels of Arc, c-Fos and Zif268 immunoreactivity prior to and after conditioning stimulation in animals with developed neuropathic pain, with sham operated, non-ligated controls. METHODS Twenty-four animals were divided equally into the neuropathic and non-neuropathic groups. Neuropathic pain was induced in all animals by conducting a loose ligation of the sciatic nerve with Chromic Catgut 4.0 sutures 7 days prior to conditioning stimulation or sham operation. The loose ligation was performed by placing sutures around the sciatic nerve compressing the nerve slightly just enough to reduce but not completely diminish the perineural circulation. A state of neuropathy was confirmed by a significant decrease in mechanical withdrawal threshold measured by von Frey's fibres. Immunohistochemical analysis was performed on transverse sections obtained from the L3-L5 segments of the SC at 2 and 6h post-CS and IEGP positive cells were counted in lamina I and II of the dorsal horn. During statistical analyses, the groups were compared by means of analysis of variance (univariate general linear model). If significant differences were found, each set of animals was compared with the sham group with post hoc Tukey's multiple comparison test. RESULTS Strikingly, all IEGPs exhibited a significant increase in immunoreactivity at both time points compared to time-matched, sham operated controls. Maximal IEGP expression was found 2h after CS in neuropathic rats, and there was a smaller but still significant increase 6h after CS. The unstimulated side of the dorsal horn in stimulated animals did not show any significant change of the number of IEGP positive cells and was approximately at the same level as sham operated animals. The number of IEGP positive cells in sham operated controls (non-neuropathic and non-stimulated animals) showed same immunoreactivity in 2 and 6h post sham operation. CONCLUSIONS AND IMPLICATIONS The neurophysiological process of neuropathic pain development is complex and needs to be studied further in order to clarify its nature and components. This present study is meant to reveal a step towards further understanding the role of Arc, c-Fos and Zif268 in neuropathic pain. Moreover, this study might contribute to the knowledge base for further research on better therapeutic possibilities for neuropathic pain.
Collapse
Affiliation(s)
- Ognjen Bojovic
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009Bergen, Norway
| | - Arne Tjølsen
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009Bergen, Norway
| |
Collapse
|
31
|
Plasticity-Related PKMζ Signaling in the Insular Cortex Is Involved in the Modulation of Neuropathic Pain after Nerve Injury. Neural Plast 2015; 2015:601767. [PMID: 26457205 PMCID: PMC4592717 DOI: 10.1155/2015/601767] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/16/2015] [Accepted: 04/17/2015] [Indexed: 12/22/2022] Open
Abstract
The insular cortex (IC) is associated with important functions linked with pain and emotions. According to recent reports, neural plasticity in the brain including the IC can be induced by nerve injury and may contribute to chronic pain. Continuous active kinase, protein kinase Mζ (PKMζ), has been known to maintain the long-term potentiation. This study was conducted to determine the role of PKMζ in the IC, which may be involved in the modulation of neuropathic pain. Mechanical allodynia test and immunohistochemistry (IHC) of zif268, an activity-dependent transcription factor required for neuronal plasticity, were performed after nerve injury. After ζ-pseudosubstrate inhibitory peptide (ZIP, a selective inhibitor of PKMζ) injection, mechanical allodynia test and immunoblotting of PKMζ, phospho-PKMζ (p-PKMζ), and GluR1 and GluR2 were observed. IHC demonstrated that zif268 expression significantly increased in the IC after nerve injury. Mechanical allodynia was significantly decreased by ZIP microinjection into the IC. The analgesic effect lasted for 12 hours. Moreover, the levels of GluR1, GluR2, and p-PKMζ were decreased after ZIP microinjection. These results suggest that peripheral nerve injury induces neural plasticity related to PKMζ and that ZIP has potential applications for relieving chronic pain.
Collapse
|
32
|
Bojovic O, Panja D, Bittins M, Bramham CR, Tjølsen A. Time course of immediate early gene protein expression in the spinal cord following conditioning stimulation of the sciatic nerve in rats. PLoS One 2015; 10:e0123604. [PMID: 25860146 PMCID: PMC4393234 DOI: 10.1371/journal.pone.0123604] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 02/20/2015] [Indexed: 12/20/2022] Open
Abstract
Long-term potentiation induced by conditioning electrical stimulation of afferent fibers is a widely studied form of synaptic plasticity in the brain and the spinal cord. In the spinal cord dorsal horn, long-term potentiation is induced by a series of high-frequency trains applied to primary afferent fibers. Conditioning stimulation (CS) of sciatic nerve primary afferent fibers also induces expression of immediate early gene proteins in the lumbar spinal cord. However, the time course of immediate early gene expression and the rostral-caudal distribution of expression in the spinal cord have not been systematically studied. Here, we examined the effects of sciatic nerve conditioning stimulation (10 stimulus trains, 0.5 ms stimuli, 7.2 mA, 100 Hz, train duration 2 s, 8 s intervals between trains) on cellular expression of immediate early genes, Arc, c-Fos and Zif268, in anesthetized rats. Immunohistochemical analysis was performed on sagittal sections obtained from Th13- L5 segments of the spinal cord at 1, 2, 3, 6 and 12 h post-CS. Strikingly, all immediate early genes exhibited a monophasic increase in expression with peak increases detected in dorsal horn neurons at 2 hours post-CS. Regional analysis showed peak increases at the location between the L3 and L4 spinal segments. Both Arc, c-Fos and Zif268 remained significantly elevated at 2 hours, followed by a sharp decrease in immediate early gene expression between 2 and 3 hours post-CS. Colocalization analysis performed at 2 hours post-CS showed that all c-Fos and Zif268 neurons were positive for Arc, while 30% and 43% of Arc positive neurons were positive for c-Fos and Zif268, respectively. The present study identifies the spinal cord level and time course of immediate early gene (IEGP) expression of relevance for analysis of IEGPs function in neuronal plasticity and nociception.
Collapse
Affiliation(s)
- Ognjen Bojovic
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
- * E-mail:
| | - Debabrata Panja
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Margarethe Bittins
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Clive R. Bramham
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Arne Tjølsen
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| |
Collapse
|
33
|
Nuclear factor kappa B-dependent Zif268 expression in hippocampus is required for recognition memory in mice. Neurobiol Learn Mem 2015; 119:10-7. [DOI: 10.1016/j.nlm.2014.12.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 12/23/2014] [Accepted: 12/30/2014] [Indexed: 12/28/2022]
|
34
|
Ménard C, Gaudreau P, Quirion R. Signaling pathways relevant to cognition-enhancing drug targets. Handb Exp Pharmacol 2015; 228:59-98. [PMID: 25977080 DOI: 10.1007/978-3-319-16522-6_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aging is generally associated with a certain cognitive decline. However, individual differences exist. While age-related memory deficits can be observed in humans and rodents in the absence of pathological conditions, some individuals maintain intact cognitive functions up to an advanced age. The mechanisms underlying learning and memory processes involve the recruitment of multiple signaling pathways and gene expression, leading to adaptative neuronal plasticity and long-lasting changes in brain circuitry. This chapter summarizes the current understanding of how these signaling cascades could be modulated by cognition-enhancing agents favoring memory formation and successful aging. It focuses on data obtained in rodents, particularly in the rat as it is the most common animal model studied in this field. First, we will discuss the role of the excitatory neurotransmitter glutamate and its receptors, downstream signaling effectors [e.g., calcium/calmodulin-dependent protein kinase II (CaMKII), protein kinase C (PKC), extracellular signal-regulated kinases (ERK), mammalian target of rapamycin (mTOR), cAMP response element-binding protein (CREB)], associated immediate early gene (e.g., Homer 1a, Arc and Zif268), and growth factors [insulin-like growth factors (IGFs) and brain-derived neurotrophic factor (BDNF)] in synaptic plasticity and memory formation. Second, the impact of the cholinergic system and related modulators on memory will be briefly reviewed. Finally, since dynorphin neuropeptides have recently been associated with memory impairments in aging, it is proposed as an attractive target to develop novel cognition-enhancing agents.
Collapse
Affiliation(s)
- Caroline Ménard
- Douglas Mental Health University Institute, McGill University, Perry Pavilion, 6875 LaSalle Boulevard, Montreal, QC, Canada, H4H 1R3
| | | | | |
Collapse
|
35
|
Prior high corticosterone exposure reduces activation of immature neurons in the ventral hippocampus in response to spatial and nonspatial memory. Hippocampus 2014; 25:329-44. [DOI: 10.1002/hipo.22375] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2014] [Indexed: 12/21/2022]
|
36
|
ZióŁkowska B, Gieryk A, Solecki W, PrzewŁocki R. Temporal and anatomic patterns of immediate-early gene expression in the forebrain of C57BL/6 and DBA/2 mice after morphine administration. Neuroscience 2014; 284:107-124. [PMID: 25290009 DOI: 10.1016/j.neuroscience.2014.09.069] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
Abstract
Although morphine was previously reported to produce an instant induction of c-fos in the striatum, our recent studies have demonstrated that the expression of numerous immediate early genes (IEGs) is significantly elevated at delayed time-points (several hours) after morphine administration. To better dissect the time-course of opioid-produced IEG induction, we used in situ hybridization to examine the expression of the IEGs c-fos, zif268 and arc in the mouse forebrain at several time-points after acute morphine injection. To link drug-produced behavioral changes with the activity of specific neuronal complexes, this study was performed comparatively in the C57BL/6 and DBA/2 mouse strains, which differ markedly in their locomotor responses to opioids and opioid reward. Our study demonstrates that morphine produces two episodes of IEG induction, which are separate in time (30 min vs. 4-6 h) and which have different neuroanatomic distribution. At 30 min, one or more IEGs were induced in circumscribed subregions of the dorsal striatum (dStr) and of the nucleus accumbens (NAc) shell, as well as in the lateral septum. The observed inter-strain differences in IEG expression at 30 min support earlier proposals that activation of the dorsomedial striatum may mediate morphine-elicited locomotor stimulation (both effects were present only in the C57BL/6 strain). In contrast, NAc shell activation does not appear to be linked to morphine-elicited changes in locomotor behavior. The second IEG induction (of arc and of zif268) was more widespread, involving most of the dStr and the cortex. The second IEG induction peaked earlier in the DBA/2 mice than in the C57BL/6 mice (4 h compared with 6 h) and displayed no apparent relation to locomotor behavior. This delayed episode of IEG activation, which has largely been overlooked thus far, may contribute to the development of long-term effects of opioids such as tolerance, dependence and/or addiction.
Collapse
Affiliation(s)
- B ZióŁkowska
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland.
| | - A Gieryk
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - W Solecki
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Łojasiewicza 4, 30-348 Kraków, Poland
| | - R PrzewŁocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Łojasiewicza 4, 30-348 Kraków, Poland
| |
Collapse
|
37
|
Xu Y, Pan J, Sun J, Ding L, Ruan L, Reed M, Yu X, Klabnik J, Lin D, Li J, Chen L, Zhang C, Zhang H, O'Donnell JM. Inhibition of phosphodiesterase 2 reverses impaired cognition and neuronal remodeling caused by chronic stress. Neurobiol Aging 2014; 36:955-70. [PMID: 25442113 DOI: 10.1016/j.neurobiolaging.2014.08.028] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 12/22/2022]
Abstract
Chronic stress and neuronal vulnerability have recently been recognized as factors contributing to cognitive disorders. One way to modify neuronal vulnerability is through mediation of phosphodiesterase 2 (PDE2), an enzyme that exerts its action on cognitive processes via the control of intracellular second messengers, cGMP and, to a lesser extent, cAMP. This study explored the effects of a PDE2 inhibitor, Bay 60-7550, on stress-induced learning and memory dysfunction in terms of its ramification on behavioral, morphologic, and molecular changes. Bay 60-7550 reversed stress-induced cognitive impairment in the Morris water maze, novel object recognition, and location tasks (object recognition test and/or object location test), effects prevented by treatment with 7-NI, a selective inhibitor of neuronal nitric oxide synthase; MK801, a glutamate receptor (NMDAR) inhibitor; myr-AIP, a CaMKII inhibitor; and KT5823, a protein kinase G inhibitor. Bay 60-7550 also ameliorated stress-induced structural remodeling in the CA1 of the hippocampus, leading to increases in dendritic branching, length, and spine density. However, the neuroplasticity initiated by Bay 60-7550 was not seen in the presence of 7-NI, MK801, myr-AIP, or KT5823. PDE2 inhibition reduced stress-induced extracellular-regulated protein kinase activation and attenuated stress-induced decreases in transcription factors (e.g., Elk-1, TORC1, and CREB phosphorylation) and plasticity-related proteins (e.g., Egr-1 and brain-derived neurotrophic factor). Pretreatment with inhibitors of NMDA, CaMKII, neuronal nitric oxide synthase, and protein kinase G (or protein kinase A) blocked the effects of Bay 60-7550 on cGMP or cAMP signaling. These findings indicate that the effect of PDE2 inhibition on stress-induced memory impairment is potentially mediated via modulation of neuroplasticity-related NMDAR-CaMKII-cGMP/cAMP signaling.
Collapse
Affiliation(s)
- Ying Xu
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Jianchun Pan
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Jiao Sun
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Lianshu Ding
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Lina Ruan
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Miranda Reed
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Xuefeng Yu
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Jonathan Klabnik
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - Dan Lin
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Jianxin Li
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Ling Chen
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Chong Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Hanting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - James M O'Donnell
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
38
|
Cellular registration without behavioral recall of olfactory sensory input under general anesthesia. Anesthesiology 2014; 120:890-905. [PMID: 24694846 DOI: 10.1097/aln.0000000000000137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Previous studies suggest that sensory information is "received" but not "perceived" under general anesthesia. Whether and to what extent the brain continues to process sensory inputs in a drug-induced unconscious state remain unclear. METHODS One hundred seven rats were randomly assigned to 12 different anesthesia and odor exposure paradigms. The immunoreactivities of the immediate early gene products c-Fos and Egr1 as neural activity markers were combined with behavioral tests to assess the integrity and relationship of cellular and behavioral responsiveness to olfactory stimuli under a surgical plane of ketamine-xylazine general anesthesia. RESULTS The olfactory sensory processing centers could distinguish the presence or absence of experimental odorants even when animals were fully anesthetized. In the anesthetized state, the c-Fos immunoreactivity in the higher olfactory cortices revealed a difference between novel and familiar odorants similar to that seen in the awake state, suggesting that the anesthetized brain functions beyond simply receiving external stimulation. Reexposing animals to odorants previously experienced only under anesthesia resulted in c-Fos immunoreactivity, which was similar to that elicited by familiar odorants, indicating that previous registration had occurred in the anesthetized brain. Despite the "cellular memory," however, odor discrimination and forced-choice odor-recognition tests showed absence of behavioral recall of the registered sensations, except for a longer latency in odor recognition tests. CONCLUSIONS Histologically distinguishable registration of sensory processing continues to occur at the cellular level under ketamine-xylazine general anesthesia despite the absence of behavioral recognition, consistent with the notion that general anesthesia causes disintegration of information processing without completely blocking cellular communications.
Collapse
|
39
|
Veyrac A, Besnard A, Caboche J, Davis S, Laroche S. The transcription factor Zif268/Egr1, brain plasticity, and memory. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 122:89-129. [PMID: 24484699 DOI: 10.1016/b978-0-12-420170-5.00004-0] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The capacity to remember our past experiences and organize our future draws on a number of cognitive processes that allow our brain to form and store neural representations that can be recalled and updated at will. In the brain, these processes require mechanisms of neural plasticity in the activated circuits, brought about by cellular and molecular changes within the neurons activated during learning. At the cellular level, a wealth of experimental data accumulated in recent years provides evidence that signaling from synapses to nucleus and the rapid regulation of the expression of immediate early genes encoding inducible, regulatory transcription factors is a key step in the mechanisms underlying synaptic plasticity and the modification of neural networks required for the laying down of memories. In the activated neurons, these transcriptional events are thought to mediate the activation of selective gene programs and subsequent synthesis of proteins, leading to stable functional and structural remodeling of the activated networks, so that the memory can later be reactivated upon recall. Over the past few decades, novel insights have been gained in identifying key transcriptional regulators that can control the genomic response of synaptically activated neurons. Here, as an example of this approach, we focus on one such activity-dependent transcription factor, Zif268, known to be implicated in neuronal plasticity and memory formation. We summarize current knowledge about the regulation and function of Zif268 in different types of brain plasticity and memory processes.
Collapse
Affiliation(s)
- Alexandra Veyrac
- CNRS, Centre de Neurosciences Paris-Sud, UMR 8195, Orsay, France; Centre de Neurosciences Paris-Sud, Univ Paris-Sud, UMR 8195, Orsay, France
| | - Antoine Besnard
- Harvard Stem Cell Institute, Harvard Medical School, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jocelyne Caboche
- INSERM, UMRS 952, Physiopathologie des Maladies du Système Nerveux Central, Paris, France; CNRS, UMR7224, Physiopathologie des Maladies du Système Nerveux Central, Paris, France; UPMC University Paris 6, Paris, France
| | - Sabrina Davis
- CNRS, Centre de Neurosciences Paris-Sud, UMR 8195, Orsay, France; Centre de Neurosciences Paris-Sud, Univ Paris-Sud, UMR 8195, Orsay, France
| | - Serge Laroche
- CNRS, Centre de Neurosciences Paris-Sud, UMR 8195, Orsay, France; Centre de Neurosciences Paris-Sud, Univ Paris-Sud, UMR 8195, Orsay, France
| |
Collapse
|
40
|
Han S, Hong S, Mo J, Lee D, Choi E, Choi JS, Sun W, Lee HW, Kim H. Impaired extinction of learned contextual fear memory in early growth response 1 knockout mice. Mol Cells 2014; 37:24-30. [PMID: 24552706 PMCID: PMC3907009 DOI: 10.14348/molcells.2014.2206] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/29/2013] [Accepted: 11/25/2013] [Indexed: 01/27/2023] Open
Abstract
Inductive expression of early growth response 1 (Egr-1) in neurons is associated with many forms of neuronal activity. However, only a few Egr-1 target genes are known in the brain. The results of this study demonstrate that Egr-1 knockout (KO) mice display impaired contextual extinction learning and normal fear acquisition relative to wild-type (WT) control animals. Genome-wide microarray experiments revealed 368 differentially expressed genes in the hippocampus of Egr-1 WT exposed to different phases of a fear conditioning paradigm compared to gene expression profiles in the hippocampus of KO mice. Some of genes, such as serotonin receptor 2C (Htr2c), neuropeptide B (Npb), neuronal PAS domain protein 4 (Npas4), NPY receptor Y1 (Npy1r), fatty acid binding protein 7 (Fabp7), and neuropeptide Y (Npy) are known to regulate processing of fearful memories, and promoter analyses demonstrated that several of these genes contained Egr-1 binding sites. This study provides a useful list of potential Egr-1 target genes which may be regulated during fear memory processing.
Collapse
Affiliation(s)
- Seungrie Han
- Department of Anatomy and Neuroscience, College of Medicine, Korea University, Seoul 136-705,
Korea
| | - Soontaek Hong
- Department of Anatomy and Neuroscience, College of Medicine, Korea University, Seoul 136-705,
Korea
| | - Jiwon Mo
- Department of Anatomy and Neuroscience, College of Medicine, Korea University, Seoul 136-705,
Korea
| | - Dongmin Lee
- Department of Anatomy and Neuroscience, College of Medicine, Korea University, Seoul 136-705,
Korea
| | | | | | - Woong Sun
- Department of Anatomy and Neuroscience, College of Medicine, Korea University, Seoul 136-705,
Korea
| | - Hyun Woo Lee
- Department of Anatomy and Neuroscience, College of Medicine, Korea University, Seoul 136-705,
Korea
| | - Hyun Kim
- Department of Anatomy and Neuroscience, College of Medicine, Korea University, Seoul 136-705,
Korea
| |
Collapse
|
41
|
Abstract
This review highlights recent discoveries that have shaped the emerging viewpoints in the field of epigenetic influences in the central nervous system (CNS), focusing on the following questions: (i) How is the CNS shaped during development when precursor cells transition into morphologically and molecularly distinct cell types, and is this event driven by epigenetic alterations?; ii) How do epigenetic pathways control CNS function?; (iii) What happens to "epigenetic memory" during aging processes, and do these alterations cause CNS dysfunction?; (iv) Can one restore normal CNS function by manipulating the epigenome using pharmacologic agents, and will this ameliorate aging-related neurodegeneration? These and other still unanswered questions remain critical to understanding the impact of multifaceted epigenetic machinery on the age-related dysfunction of CNS.
Collapse
Affiliation(s)
- Yue-Qiang Zhao
- />Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400 USA
- />Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - I. King Jordan
- />School of Biology, Georgia Institute of Technology, Atlanta, GA USA
- />PanAmerican Bioinformatics Institute, Santa Marta, Magdalena Colombia
| | - Victoria V. Lunyak
- />Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400 USA
| |
Collapse
|
42
|
Affiliation(s)
- Robert Jaffard
- Laboratoire de Neurosciences Comportementales et Cognitives, Université de Bordeaux I, France
| | - Martine Meunier
- Laboratoire de Neurosciences Comportementales et Cognitives, Université de Bordeaux I, France
| |
Collapse
|
43
|
Rocha L. Interaction between electrical modulation of the brain and pharmacotherapy to control pharmacoresistant epilepsy. Pharmacol Ther 2013; 138:211-28. [DOI: 10.1016/j.pharmthera.2013.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 01/07/2013] [Indexed: 12/15/2022]
|
44
|
Bepari AK, Watanabe K, Yamaguchi M, Tamamaki N, Takebayashi H. Visualization of odor-induced neuronal activity by immediate early gene expression. BMC Neurosci 2012; 13:140. [PMID: 23126335 PMCID: PMC3538715 DOI: 10.1186/1471-2202-13-140] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 10/31/2012] [Indexed: 12/15/2022] Open
Abstract
Background Sensitive detection of sensory-evoked neuronal activation is a key to mechanistic understanding of brain functions. Since immediate early genes (IEGs) are readily induced in the brain by environmental changes, tracing IEG expression provides a convenient tool to identify brain activity. In this study we used in situ hybridization to detect odor-evoked induction of ten IEGs in the mouse olfactory system. We then analyzed IEG induction in the cyclic nucleotide-gated channel subunit A2 (Cnga2)-null mice to visualize residual neuronal activity following odorant exposure since CNGA2 is a key component of the olfactory signal transduction pathway in the main olfactory system. Results We observed rapid induction of as many as ten IEGs in the mouse olfactory bulb (OB) after olfactory stimulation by a non-biological odorant amyl acetate. A robust increase in expression of several IEGs like c-fos and Egr1 was evident in the glomerular layer, the mitral/tufted cell layer and the granule cell layer. Additionally, the neuronal IEG Npas4 showed steep induction from a very low basal expression level predominantly in the granule cell layer. In Cnga2-null mice, which are usually anosmic and sexually unresponsive, glomerular activation was insignificant in response to either ambient odorants or female stimuli. However, a subtle induction of c-fos took place in the OB of a few Cnga2-mutants which exhibited sexual arousal. Interestingly, very strong glomerular activation was observed in the OB of Cnga2-null male mice after stimulation with either the neutral odor amyl acetate or the predator odor 2, 3, 5-trimethyl-3-thiazoline (TMT). Conclusions This study shows for the first time that in vivo olfactory stimulation can robustly induce the neuronal IEG Npas4 in the mouse OB and confirms the odor-evoked induction of a number of IEGs. As shown in previous studies, our results indicate that a CNGA2-independent signaling pathway(s) may activate the olfactory circuit in Cnga2-null mice and that neuronal activation which correlates to behavioral difference in individual mice is detectable by in situ hybridization of IEGs. Thus, the in situ hybridization probe set we established for IEG tracing can be very useful to visualize neuronal activity at the cellular level.
Collapse
Affiliation(s)
- Asim K Bepari
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | |
Collapse
|
45
|
Long-term decrease in immediate early gene expression after electroconvulsive seizures. J Neural Transm (Vienna) 2012; 120:259-66. [PMID: 22875635 DOI: 10.1007/s00702-012-0861-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 06/29/2012] [Indexed: 10/28/2022]
Abstract
Electroconvulsive therapy (ECT) is a well-established psychiatric treatment for severe depression. Despite its clinical utility, post-ECT memory deficits are a common side effect. Neuronal plasticity and memory consolidation are intimately related to the expression of immediate early genes (IEG), such as Egr1, Fos and Arc. Changes in IEG activation have been postulated to underlie long-term neuronal adaptations following electroconvulsive seizures (ECS), an animal model of ECT. To test this hypothesis, we used real-time PCR to examine the effect of acute and chronic ECS (8 sessions, one every other day) on the long-term (>24 h) expression of IEG Egr1, Fos and Arc in the hippocampus, a brain region implicated both in the pathophysiology of depression as well as in memory function. We observed a transient increase in Egr1 and Fos expression immediately after ECS, followed by a long-term decrease of IEG levels after both acute and chronic ECS. A separate group of animals, submitted to the same chronic ECS protocol and then subjected to open field or passive avoidance tasks, confirmed robust memory deficits 2 weeks after the last chronic ECS. The possible role of IEG downregulation on long-term learning deficits observed following ECS are discussed.
Collapse
|
46
|
|
47
|
Eriksen GS, Jacobsen LM, Mahmood A, Pedersen LM, Gjerstad J. Inhibition of fatty acid amide hydrolase (FAAH) reduces spinal nociceptive responses and expression of spinal long-term potentiation (LTP). Brain Res Bull 2012; 87:234-7. [DOI: 10.1016/j.brainresbull.2011.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/25/2011] [Accepted: 11/15/2011] [Indexed: 01/11/2023]
|
48
|
Ranieri F, Podda MV, Riccardi E, Frisullo G, Dileone M, Profice P, Pilato F, Di Lazzaro V, Grassi C. Modulation of LTP at rat hippocampal CA3-CA1 synapses by direct current stimulation. J Neurophysiol 2012; 107:1868-80. [PMID: 22236710 DOI: 10.1152/jn.00319.2011] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transcranial direct current stimulation (tDCS) can produce a lasting polarity-specific modulation of cortical excitability in the brain, and it is increasingly used in experimental and clinical settings. Recent studies suggest that the after-effects of tDCS are related to molecular mechanisms of activity-dependent synaptic plasticity. Here we investigated the effect of DCS on the induction of one of the most studied N-methyl-d-aspartate receptor-dependent forms of long-term potentiation (LTP) of synaptic activity at CA3-CA1 synapses in the hippocampus. We show that DCS applied to rat brain slices determines a modulation of LTP that is increased by anodal and reduced by cathodal DCS. Immediate early genes, such as c-fos and zif268 (egr1/NGFI-A/krox24), are rapidly induced following neuronal activation, and a specific role of zif268 in the induction and maintenance of LTP has been demonstrated. We found that both anodal and cathodal DCS produce a marked subregion-specific increase in the expression of zif268 protein in the cornus ammonis (CA) region, whereas the same protocols of stimulation produce a less pronounced increase in c-fos protein expression in the CA and in dentate gyrus regions of the hippocampus. Brain-derived neurotrophic factor expression was also investigated, and it was found to be reduced in cathodal-stimulated slices. The present data demonstrate that it is possible to modulate LTP by using DCS and provide the rationale for the use of DCS in neurological diseases to promote the adaptive and suppress the maladaptive forms of brain plasticity.
Collapse
Affiliation(s)
- F Ranieri
- Istituto di Neurologia, Università Cattolica, L.go A. Gemelli 8, 00168 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
While there is ample agreement that the cognitive role of sleep is explained by sleep-dependent synaptic changes, consensus is yet to be established as to the nature of these changes. Some researchers believe that sleep promotes global synaptic downscaling, leading to a non-Hebbian reset of synaptic weights that is putatively necessary for the acquisition of new memories during ensuing waking. Other investigators propose that sleep also triggers experience-dependent, Hebbian synaptic upscaling able to consolidate recently acquired memories. Here, I review the molecular and physiological evidence supporting these views, with an emphasis on the calcium signaling pathway. I argue that the available data are consistent with sleep promoting experience-dependent synaptic embossing, understood as the simultaneous non-Hebbian downscaling and Hebbian upscaling of separate but complementary sets of synapses, heterogeneously activated at the time of memory encoding and therefore differentially affected by sleep.
Collapse
Affiliation(s)
- Sidarta Ribeiro
- Brain Institute, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil.
| |
Collapse
|
50
|
Dolan S, Hastie P, Crossan C, Nolan AM. Co-induction of cyclooxygenase-2 [correction of cyclooxyenase-2] and early growth response gene (Egr-1) in spinal cord in a clinical model of persistent inflammation and hyperalgesia. Mol Pain 2011; 7:91. [PMID: 22112635 PMCID: PMC3256114 DOI: 10.1186/1744-8069-7-91] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 11/23/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study characterised the effects of persistent peripheral inflammation of the foot on pain and spinal cord expression of cyclooxygenase-1 and -2 (COX-1 and COX-2) and early growth response gene 1 (Egr-1), known markers of neuronal plasticity, in a clinical model of naturally-occurring inflammatory disease and hyperalgesia in sheep ('footrot'), before and after routine treatment (parenteral treatment with antibiotics and antiseptic footbathing). The temporal pattern of expression of COX-1, COX-2 and Egr-1 mRNA and protein were analysed using real-time PCR and Western blotting. RESULTS Animals affected with persistent peripheral inflammation displayed significant hyperalgesia and lameness (a proxy indicator of spontaneous pain) restricted to the inflamed limb. Hyperalgesia and lameness were significantly attenuated 1 day after treatment, and resolved further by day 7 and day 3, respectively. COX-2 but not COX-1, protein expression was up-regulated in spinal cord from lame animals on day 0, before treatment. Following treatment and attenuation of pain behaviours, levels of COX-2 returned to control levels. Significant induction of Egr-1 mRNA and protein were observed in spinal cord from lame animals. Three days after treatment, levels of Egr-1 mRNA returned to control levels, however, Egr-1 protein remained elevated. CONCLUSION Elevated levels of spinal COX-2 and Egr-1 protein correlate with the presence of pain and hyperalgesia, and may underlie persistent pain, although a direct causal link has still to be established. Understanding the temporal pattern of expression of key mediators in clinical pain states may lead to better strategies to manage pain.
Collapse
Affiliation(s)
- Sharron Dolan
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Cowcaddens Road, Glasgow, G4 0BA, UK.
| | | | | | | |
Collapse
|