151
|
Kredlow MA, Eichenbaum H, Otto MW. Memory creation and modification: Enhancing the treatment of psychological disorders. AMERICAN PSYCHOLOGIST 2018; 73:269-285. [PMID: 29494172 PMCID: PMC5897133 DOI: 10.1037/amp0000185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Modification of the ongoing influence of maladaptive cognitive, emotional, and behavioral patterns is a fundamental feature of many psychological treatments. Accordingly, a clear understanding of the nature of memory adaptation and accommodation to therapeutic learning becomes an important issue for (1) understanding the impact of clinical interventions, and (2) considering innovations in treatment strategies. In this article, we consider advances in the conceptualization of memory processes and memory modification research relative to clinical treatment. We review basic research on the formation of memories, the way in which new learning is integrated within memory structures, and strategies to influence the nature and degree to which new learning is integrated. We then discuss cognitive/behavioral and pharmacological strategies for influencing memory formation in relation to disorder prevention or treatment. Our goal is to foster awareness of current strategies for enhancing therapeutic learning and to encourage research on potential new avenues for memory enhancement in service of the treatment of mental health disorders. (PsycINFO Database Record
Collapse
Affiliation(s)
| | | | - Michael W Otto
- Department of Psychological and Brain Sciences, Boston University
| |
Collapse
|
152
|
Scheggia D, Zamberletti E, Realini N, Mereu M, Contarini G, Ferretti V, Managò F, Margiani G, Brunoro R, Rubino T, De Luca MA, Piomelli D, Parolaro D, Papaleo F. Remote memories are enhanced by COMT activity through dysregulation of the endocannabinoid system in the prefrontal cortex. Mol Psychiatry 2018. [PMID: 28630452 DOI: 10.1038/mp.2017.126] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prefrontal cortex (PFC) is a crucial hub for the flexible modulation of recent memories (executive functions) as well as for the stable organization of remote memories. Dopamine in the PFC is implicated in both these processes and genetic variants affecting its neurotransmission might control the unique balance between cognitive stability and flexibility present in each individual. Functional genetic variants in the catechol-O-methyltransferase (COMT) gene result in a different catabolism of dopamine in the PFC. However, despite the established role played by COMT genetic variation in executive functions, its impact on remote memory formation and recall is still poorly explored. Here we report that transgenic mice overexpressing the human COMT-Val gene (COMT-Val-tg) present exaggerated remote memories (>50 days) while having unaltered recent memories (<24 h). COMT selectively and reversibly modulated the recall of remote memories as silencing COMT Val overexpression starting from 30 days after the initial aversive conditioning normalized remote memories. COMT genetic overactivity produced a selective overdrive of the endocannabinoid system within the PFC, but not in the striatum and hippocampus, which was associated with enhanced remote memories. Indeed, acute pharmacological blockade of CB1 receptors was sufficient to rescue the altered remote memory recall in COMT-Val-tg mice and increased PFC dopamine levels. These results demonstrate that COMT genetic variations modulate the retrieval of remote memories through the dysregulation of the endocannabinoid system in the PFC.
Collapse
Affiliation(s)
- D Scheggia
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - E Zamberletti
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - N Realini
- Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - M Mereu
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy.,Dipartimento di Scienze del Farmaco, Universita' degli Studi di Padova, Largo Meneghetti, Padova, Italy
| | - G Contarini
- Dipartimento di Scienze del Farmaco, Universita' degli Studi di Padova, Largo Meneghetti, Padova, Italy
| | - V Ferretti
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - F Managò
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - G Margiani
- Department of Biomedical Sciences, Università di Cagliari, Cagliari, Italy
| | - R Brunoro
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - T Rubino
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - M A De Luca
- Department of Biomedical Sciences, Università di Cagliari, Cagliari, Italy
| | - D Piomelli
- Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy.,Department of Anatomy and Neurobiology, Pharmacology and Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - D Parolaro
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - F Papaleo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
153
|
Dubey H, Gulati K, Ray A. Recent studies on cellular and molecular mechanisms in Alzheimer’s disease: focus on epigenetic factors and histone deacetylase. Rev Neurosci 2018; 29:241-260. [DOI: 10.1515/revneuro-2017-0049] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/28/2017] [Indexed: 01/06/2023]
Abstract
AbstractAlzheimer’s disease (AD) is one of the most common neurodegenerative disorders mainly affecting elderly people. It is characterized by progressive loss of memory and cognitive function. More than 95% of AD cases are related to sporadic or late-onset AD (LOAD). The etiology of LOAD is still unclear. It has been reported that environmental factors and epigenetic alterations play a significant role in AD pathogenesis. Furthermore, recently, genome-wide association studies (GWAS) identified 10 novel risk genes:ABCA7,APOE,BIN1,CD2AP,CD33,CLU,CR1,MS4A6A,MS4A4E, andPICALM, which play an important role for LOAD. In this review, the therapeutic approaches of AD by epigenetic modifications have been discussed. Nowadays, HDAC inhibitors have clinically proven its activity for epigenetic modifications. Furthermore, we try to establish the relationship between HDAC inhibitors and above mentioned LOAD risk genes. Finally, we are hoping that this review may open new area of research for AD treatment.
Collapse
|
154
|
Gong Z, Zhou Q. Dnmt3a in the dorsal dentate gyrus is a key regulator of fear renewal. Sci Rep 2018; 8:5093. [PMID: 29572461 PMCID: PMC5865109 DOI: 10.1038/s41598-018-23533-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/14/2018] [Indexed: 12/13/2022] Open
Abstract
Renewal of extinguished fear memory in an altered context is widely believed to be a major limiting issue for exposure therapy in treating various psychiatric diseases. Effective prevention of fear renewal will significantly improve the efficacy of exposure therapy. DNA methyltransferase (DNMTs) mediated epigenetic processes play critical roles in long term memory, but little is known about their functions in fear memory extinction or renewal. Here we investigated whether DNMTs regulate fear renewal after extinction. We found that elevated Dnmt3a level in the dorsal dentate gyrus (dDG) of hippocampus was associated with the absence of fear renewal in an altered context after extinction training. Overexpression and knockdown of Dnmt3a in the dDG regulated the occurrence of fear renewal in a bi-directional manner. In addition, Dnmt3a overexpression was associated with elevated expression of c-Fos in the dDG during extinction training. Furthermore, we found that renewal of remote fear memory can be prevented, and the absence of renewal was concurrent with an elevated Dnmt3a level. Our results indicate that Dnmt3a in the dDG is a key regulator of fear renewal after extinction, and Dnmt3a may play a critical role in controlling fear memory return and thus has therapeutic values.
Collapse
Affiliation(s)
- Zhiting Gong
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
155
|
Akers KG, Chérasse Y, Fujita Y, Srinivasan S, Sakurai T, Sakaguchi M. Concise Review: Regulatory Influence of Sleep and Epigenetics on Adult Hippocampal Neurogenesis and Cognitive and Emotional Function. Stem Cells 2018; 36:969-976. [DOI: 10.1002/stem.2815] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 02/02/2018] [Accepted: 02/17/2018] [Indexed: 12/20/2022]
Affiliation(s)
| | - Yoan Chérasse
- International Institute for Integrative Sleep Medicine, University of Tsukuba; Tsukuba Ibaraki Japan
| | - Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine; Osaka University; Suita Osaka Japan
| | - Sakthivel Srinivasan
- International Institute for Integrative Sleep Medicine, University of Tsukuba; Tsukuba Ibaraki Japan
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine, University of Tsukuba; Tsukuba Ibaraki Japan
| | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine, University of Tsukuba; Tsukuba Ibaraki Japan
| |
Collapse
|
156
|
Uchida S, Shumyatsky GP. Epigenetic regulation of Fgf1 transcription by CRTC1 and memory enhancement. Brain Res Bull 2018; 141:3-12. [PMID: 29477835 PMCID: PMC6128695 DOI: 10.1016/j.brainresbull.2018.02.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 01/30/2018] [Accepted: 02/20/2018] [Indexed: 01/06/2023]
Abstract
Recent evidence demonstrates that epigenetic regulation of gene transcription is critically involved in learning and memory. Here, we discuss the role of histone acetylation and DNA methylation, which are two best understood epigenetic processes in memory processes. More specifically, we focus on learning-strength-dependent changes in chromatin on the fibroblast growth factor 1 (Fgf1) gene and on the molecular events that modulate regulation of Fgf1 transcription, required for memory enhancement, with the specific focus on CREB-regulated transcription coactivator 1 (CRTC1).
Collapse
Affiliation(s)
- Shusaku Uchida
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Gleb P Shumyatsky
- Department of Genetics, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA.
| |
Collapse
|
157
|
Homeostatic Plasticity in the Hippocampus Facilitates Memory Extinction. Cell Rep 2018; 22:1451-1461. [DOI: 10.1016/j.celrep.2018.01.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 11/25/2017] [Accepted: 01/09/2018] [Indexed: 11/19/2022] Open
|
158
|
Tago T, Toyohara J. Advances in the Development of PET Ligands Targeting Histone Deacetylases for the Assessment of Neurodegenerative Diseases. Molecules 2018; 23:E300. [PMID: 29385079 PMCID: PMC6017260 DOI: 10.3390/molecules23020300] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 01/29/2018] [Accepted: 01/29/2018] [Indexed: 12/05/2022] Open
Abstract
Epigenetic alterations of gene expression have emerged as a key factor in several neurodegenerative diseases. In particular, inhibitors targeting histone deacetylases (HDACs), which are enzymes responsible for deacetylation of histones and other proteins, show therapeutic effects in animal neurodegenerative disease models. However, the details of the interaction between changes in HDAC levels in the brain and disease progression remain unknown. In this review, we focus on recent advances in development of radioligands for HDAC imaging in the brain with positron emission tomography (PET). We summarize the results of radiosynthesis and biological evaluation of the HDAC ligands to identify their successful results and challenges. Since 2006, several small molecules that are radiolabeled with a radioisotope such as carbon-11 or fluorine-18 have been developed and evaluated using various assays including in vitro HDAC binding assays and PET imaging in rodents and non-human primates. Although most compounds do not readily cross the blood-brain barrier, adamantane-conjugated radioligands tend to show good brain uptake. Until now, only one HDAC radioligand has been tested clinically in a brain PET study. Further PET imaging studies to clarify age-related and disease-related changes in HDACs in disease models and humans will increase our understanding of the roles of HDACs in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan.
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan.
| |
Collapse
|
159
|
Hirata Y, Sasaki T, Kanki H, Choong CJ, Nishiyama K, Kubo G, Hotei A, Taniguchi M, Mochizuki H, Uesato S. New 5-Aryl-Substituted 2-Aminobenzamide-Type HDAC Inhibitors with a Diketopiperazine Group and Their Ameliorating Effects on Ischemia-Induced Neuronal Cell Death. Sci Rep 2018; 8:1400. [PMID: 29362442 PMCID: PMC5780423 DOI: 10.1038/s41598-018-19664-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/05/2018] [Indexed: 01/05/2023] Open
Abstract
We previously synthesized new 5-thienyl-substituted 2-aminobenzamide-type HDAC1, 2 inhibitors with the (4-ethyl-2,3-dioxopiperazine-1-carboxamido) methyl group. K-560 (1a) protected against neuronal cell death in a Parkinson’s disease model by up-regulating the expression of XIAP. This finding prompted us to design new K-560-related compounds. We examined the structure activity relationship (SAR) for the neuronal protective effects of newly synthesized and known K-560 derivatives after cerebral ischemia. Among them, K-856 (8), containing the (4-methyl-2,5-dioxopiperazin-1-yl) methyl group, exhibited a promising neuronal survival activity. The SAR study strongly suggested that the attachment of a monocyclic 2,3- or 2,5-diketopiperazine group to the 2-amino-5-aryl (but not 2-nitro-5-aryl) scaffold is necessary for K-560-related compounds to exert a potent neuroprotective effect.
Collapse
Affiliation(s)
- Yoshiyuki Hirata
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan.,Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
| | - Hideaki Kanki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Chi-Jing Choong
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Kumiko Nishiyama
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Genki Kubo
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Ayana Hotei
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Masahiko Taniguchi
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Shinichi Uesato
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan. .,Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan.
| |
Collapse
|
160
|
An X, Yang P, Chen S, Zhang F, Yu D. An Additional Prior Retrieval Alters the Effects of a Retrieval-Extinction Procedure on Recent and Remote Fear Memory. Front Behav Neurosci 2018; 11:259. [PMID: 29358910 PMCID: PMC5766663 DOI: 10.3389/fnbeh.2017.00259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 12/19/2017] [Indexed: 11/19/2022] Open
Abstract
Several studies have shown that the isolated retrieval of a consolidated fear memory can induce a labile phase, during which extinction training can prevent the reinstatement, a form of relapse in which fear response to a fear-provoking context returns when a mild shock is presented. However, fear memory retrieval may also have another opposing result: the enhancement of fear memory. This implies that the fear memory trace can be modified by a brief retrieval. Unclear is whether the fear-impairing effect of retrieval-extinction (RE) is altered by a prior brief retrieval. The present study investigated the responses of recent and remote fear memories to the RE procedure after the presentation of an additional prior retrieval (priRet). We found that a single RE procedure effectively blocked the reinstatement of 2-day recent contextual fear memory. The memory-impairing effect of the RE procedure on recent fear was not observed when priRet was presented 6 or 24 h before the RE procedure. In contrast to the 2-day recent memory, the RE procedure failed to block the reinstatement of 36-day remote fear memory but successfully disrupted the return of remote fear memory after priRet. This memory-disruptive effect on remote memory did not occur when priRet was performed in a novel context. Nimodipine administration revealed that the blockade of priRet-induced processes recovered the effects of the RE procedure on both recent and remote fear memories. Our findings suggest that the susceptibility of recent and remote fear memories to RE procedures can be altered by an additional retrieval.
Collapse
Affiliation(s)
- Xianli An
- School of Educational Science, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, China
| | - Ping Yang
- School of Educational Science, Yangzhou University, Yangzhou, China
| | - Siguang Chen
- School of Educational Science, Yangzhou University, Yangzhou, China
| | - Fenfen Zhang
- School of Educational Science, Yangzhou University, Yangzhou, China
| | - Duonan Yu
- Jiangsu Key Laboratory of Experimental and Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, China.,RNA Center, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou, China
| |
Collapse
|
161
|
Abstract
Scientific advances in the last decades uncovered that memory is not a stable, fixed entity. Apparently stable memories may become transiently labile and susceptible to modifications when retrieved due to the process of reconsolidation. Here, we review the initial evidence and the logic on which reconsolidation theory is based, the wide range of conditions in which it has been reported and recent findings further revealing the fascinating nature of this process. Special focus is given to conceptual issues of when and why reconsolidation happen and its possible outcomes. Last, we discuss the potential clinical implications of memory modifications by reconsolidation.
Collapse
Affiliation(s)
- Josue Haubrich
- Department of Psychology, McGill University, Montreal, Canada
- Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Karim Nader
- Department of Psychology, McGill University, Montreal, Canada.
| |
Collapse
|
162
|
Wey HY, Gilbert TM, Zürcher NR, She A, Bhanot A, Taillon BD, Schroeder FA, Wang C, Haggarty SJ, Hooker JM. Insights into neuroepigenetics through human histone deacetylase PET imaging. Sci Transl Med 2017; 8:351ra106. [PMID: 27510902 DOI: 10.1126/scitranslmed.aaf7551] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 07/07/2016] [Indexed: 12/14/2022]
Abstract
Epigenetic dysfunction is implicated in many neurological and psychiatric diseases, including Alzheimer's disease and schizophrenia. Consequently, histone deacetylases (HDACs) are being aggressively pursued as therapeutic targets. However, a fundamental knowledge gap exists regarding the expression and distribution of HDACs in healthy individuals for comparison to disease states. Here, we report the first-in-human evaluation of neuroepigenetic regulation in vivo. Using positron emission tomography with [(11)C]Martinostat, an imaging probe selective for class I HDACs (isoforms 1, 2, and 3), we found that HDAC expression is higher in cortical gray matter than in white matter, with conserved regional distribution patterns within and between healthy individuals. Among gray matter regions, HDAC expression was lowest in the hippocampus and amygdala. Through biochemical profiling of postmortem human brain tissue, we confirmed that [(11)C]Martinostat selectively binds HDAC isoforms 1, 2, and 3, the HDAC subtypes most implicated in regulating neuroplasticity and cognitive function. In human stem cell-derived neural progenitor cells, pharmacologic-level doses of Martinostat induced changes in genes closely associated with synaptic plasticity, including BDNF (brain-derived neurotrophic factor) and SYP (synaptophysin), as well as genes implicated in neurodegeneration, including GRN (progranulin), at the transcript level, in concert with increased acetylation at both histone H3 lysine 9 and histone H4 lysine 12. This study quantifies HDAC expression in the living human brain and provides the foundation for gaining unprecedented in vivo epigenetic information in health and disease.
Collapse
Affiliation(s)
- Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Tonya M Gilbert
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Nicole R Zürcher
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Angela She
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Anisha Bhanot
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Brendan D Taillon
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Fredrick A Schroeder
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Changing Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
163
|
Schmauss C. The roles of class I histone deacetylases (HDACs) in memory, learning, and executive cognitive functions: A review. Neurosci Biobehav Rev 2017; 83:63-71. [DOI: 10.1016/j.neubiorev.2017.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 01/06/2023]
|
164
|
Liu C, Sun X, Wang Z, Le Q, Liu P, Jiang C, Wang F, Ma L. Retrieval-Induced Upregulation of Tet3 in Pyramidal Neurons of the Dorsal Hippocampus Mediates Cocaine-Associated Memory Reconsolidation. Int J Neuropsychopharmacol 2017; 21:255-266. [PMID: 29106571 PMCID: PMC5838812 DOI: 10.1093/ijnp/pyx099] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/27/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Memory retrieval refers to reexposure to information previously encoded and stored in the brain. Following retrieval, a once-consolidated memory destabilizes and undergoes reconsolidation, during which gene expression changes to restabilize memory. Investigating epigenetic regulation during reconsolidation could provide insights into normal memory formation and pathological memory associated with psychiatric disorders. METHODS We used cocaine-induced conditioned place preference to assess the cocaine-associated memory of mice and used chemogenetic methods to manipulate the activity of the pyramidal neurons in the dorsal hippocampus. We isolated the ribosome-associated transcripts from the excitatory neurons in the dorsal hippocampus by RiboTag purification to identify the potential epigenetic regulators, and we specifically knocked down gene expression in pyramidal neurons with a Cre-dependent lentivirus. RESULTS Chemogenetically silencing the activity of the pyramidal neurons in the dorsal hippocampus immediately after memory retrieval markedly impaired memory reconsolidation, and the ribosome-associated mRNA level of the ten-eleven translocation (Tet) family methylcytosine dioxygenase Tet3, but not Tet1 or Tet2, was dramatically upregulated 10 minutes after memory retrieval. The protein level of Tet3 in the dorsal hippocampus but not in the anterior cingulate cortex was dramatically increased 1 hour after memory retrieval. Specifically, knockdown of Tet3 in pyramidal neurons in the dorsal hippocampus decreased the activation of pyramidal neurons and impaired the reconsolidation of cocaine-associated memory. CONCLUSIONS Our findings highlight the new function of the DNA demethylation regulator Tet3 in pyramidal neurons of the dorsal hippocampus in regulating the reconsolidation of cocaine-associated memory.
Collapse
Affiliation(s)
- Cao Liu
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, School of Basic Medical
Sciences and Institutes of Brain Science, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Xue Sun
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, School of Basic Medical
Sciences and Institutes of Brain Science, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Zhilin Wang
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, School of Basic Medical
Sciences and Institutes of Brain Science, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Qiumin Le
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, School of Basic Medical
Sciences and Institutes of Brain Science, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Peipei Liu
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, School of Basic Medical
Sciences and Institutes of Brain Science, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Changyou Jiang
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, School of Basic Medical
Sciences and Institutes of Brain Science, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Feifei Wang
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, School of Basic Medical
Sciences and Institutes of Brain Science, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China,Feifei Wang, PhD, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China ()
| | - Lan Ma
- The State Key Laboratory of Medical Neurobiology and Pharmacology Research Center, School of Basic Medical
Sciences and Institutes of Brain Science, and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China,Correspondence: Lan Ma, PhD, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China ()
| |
Collapse
|
165
|
Disrupting nNOS-PSD-95 coupling in the hippocampal dentate gyrus promotes extinction memory retrieval. Biochem Biophys Res Commun 2017; 493:862-868. [DOI: 10.1016/j.bbrc.2017.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 08/31/2017] [Accepted: 09/02/2017] [Indexed: 11/20/2022]
|
166
|
Tang Y, Lin YH, Ni HY, Dong J, Yuan HJ, Zhang Y, Liang HY, Yao MC, Zhou QG, Wu HY, Chang L, Luo CX, Zhu DY. Inhibiting Histone Deacetylase 2 (HDAC2) Promotes Functional Recovery From Stroke. J Am Heart Assoc 2017; 6:e007236. [PMID: 28982677 PMCID: PMC5721897 DOI: 10.1161/jaha.117.007236] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/18/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Stroke is a leading cause of long-term disability worldwide. However, current therapies that promote functional recovery from stroke are limited to physical rehabilitation. No pharmacological therapy is available. Thus, understanding the role of histone deacetylase 2 (HDAC2) in the pathophysiological process of stroke-induced functional loss may provide a novel strategy for stroke recovery. METHODS AND RESULTS Focal stroke was induced by photothrombosis. LV-HDAC2-shRNA-GFP, LV-GFP, Ad-HDAC2-Flag, or Ad-inactive-HDAC2-Flag was microinjected into the peri-infarct area immediately after stroke. HDAC inhibitors were microinjected into the peri-infarct area 4 to 10 days after stroke. Grid-walking task and cylinder task were conducted to assess motor function. Golgi-Cox staining, chromatin immunoprecipitation, and electrophysiology were used to reveal the mechanisms underlying stroke recovery. Knockdown or knockout of HDAC2 promoted stroke recovery, whereas overexpression of HDAC2 worsened stroke-induced functional impairment. More importantly, trichostatin A, a pan-HDAC inhibitor, promoted functional recovery from stroke in WT mice when used in the delayed phase, but it was ineffective in Hdac2 conditional knockout (Hdac2 CKO) mice. Treatment with suberoylanilide hydroxamic acid, a selective HDAC1 and HDAC2 inhibitor, in the delayed phase of stroke produced sustained functional recovery in mice via epigenetically enhancing neuroplasticity of surviving neurons in the peri-infarct zone. CONCLUSIONS Our novel findings provide evidence that HDAC2 is a crucial target for functional recovery from stroke. As there are clinically available HDAC inhibitors, our findings could be directly translated into clinical research of stroke.
Collapse
Affiliation(s)
- Ying Tang
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yu-Hui Lin
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Huan-Yu Ni
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jian Dong
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Hong-Jin Yuan
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yu Zhang
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Hai-Ying Liang
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Meng-Cheng Yao
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Qi-Gang Zhou
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Hai-Yin Wu
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Lei Chang
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Chun-Xia Luo
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Dong-Ya Zhu
- Institution of Stem Cells and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Precision Medicine of Cardiovascular Disease, Nanjing, China
| |
Collapse
|
167
|
Luyten L, Beckers T. A preregistered, direct replication attempt of the retrieval-extinction effect in cued fear conditioning in rats. Neurobiol Learn Mem 2017; 144:208-215. [PMID: 28765085 PMCID: PMC5931313 DOI: 10.1016/j.nlm.2017.07.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/11/2017] [Accepted: 07/28/2017] [Indexed: 11/19/2022]
Abstract
In 2009, Monfils and colleagues proposed a behavioral procedure that was said to result in a permanent attenuation of a previously established fear memory, thereby precluding a possible return of fear after extinction (Monfils, Cowansage, Klann, & LeDoux, 2009). By presenting a single retrieval trial one hour before standard extinction training, they found an enduring reduction of fear. The retrieval-extinction procedure holds great clinical potential, particularly for anxiety patients, but the findings are not undisputed, and several conceptual replications have failed to reproduce the effect. These failures have largely been attributed to small procedural differences. This preregistered study is the first endeavor to exactly replicate three key experiments of the original report by Monfils et al. (2009), thereby gauging the robustness of their seminal findings. Despite adhering to the original procedures as closely as possible, we did not find any evidence for reduced return of fear with the retrieval-extinction procedure relative to regular extinction training, as assessed through spontaneous recovery, reinstatement and renewal. Behavior of animals in the control condition (extinction only) was comparable to that in the original studies and provided an adequate baseline to reveal differences with the retrieval-extinction condition. Our null findings indicate that the effect sizes in the original paper may have been inflated and question the legitimacy of previously proposed moderators of the retrieval-extinction effect. We argue that direct experimental evaluation of purported moderators of the retrieval-extinction effect will be key to shed more light on its nature and prerequisites.
Collapse
Affiliation(s)
- Laura Luyten
- Centre for the Psychology of Learning and Experimental Psychopathology, KU Leuven, Belgium.
| | - Tom Beckers
- Centre for the Psychology of Learning and Experimental Psychopathology, KU Leuven, Belgium
| |
Collapse
|
168
|
Psychedelic Drugs in Biomedicine. Trends Pharmacol Sci 2017; 38:992-1005. [PMID: 28947075 DOI: 10.1016/j.tips.2017.08.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/11/2017] [Accepted: 08/11/2017] [Indexed: 12/18/2022]
Abstract
Psychedelic drugs, such as lysergic acid diethylamide (LSD), mescaline, and psilocybin, exert profound effects on brain and behavior. After decades of difficulties in studying these compounds, psychedelics are again being tested as potential treatments for intractable biomedical disorders. Preclinical research of psychedelics complements human neuroimaging studies and pilot clinical trials, suggesting these compounds as promising treatments for addiction, depression, anxiety, and other conditions. However, many questions regarding the mechanisms of action, safety, and efficacy of psychedelics remain. Here, we summarize recent preclinical and clinical data in this field, discuss their pharmacological mechanisms of action, and outline critical areas for future studies of psychedelic drugs, with the goal of maximizing the potential benefits of translational psychedelic biomedicine to patients.
Collapse
|
169
|
Kokare DM, Kyzar EJ, Zhang H, Sakharkar AJ, Pandey SC. Adolescent Alcohol Exposure-Induced Changes in Alpha-Melanocyte Stimulating Hormone and Neuropeptide Y Pathways via Histone Acetylation in the Brain During Adulthood. Int J Neuropsychopharmacol 2017; 20:758-768. [PMID: 28575455 PMCID: PMC5581492 DOI: 10.1093/ijnp/pyx041] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 04/15/2017] [Accepted: 05/30/2017] [Indexed: 12/18/2022] Open
Abstract
Background Adolescent intermittent ethanol exposure causes long-lasting alterations in brain epigenetic mechanisms. Melanocortin and neuropeptide Y signaling interact and are affected by ethanol exposure in the brain. Here, the persistent effects of adolescent intermittent ethanol on alpha-melanocyte stimulating hormone, melanocortin 4 receptor, and neuropeptide Y expression and their regulation by histone acetylation mechanisms were investigated in adulthood. Methods Male rats were exposed to adolescent intermittent ethanol (2 g/kg, i.p.) or volume-matched adolescent intermittent saline from postnatal days 28 to 41 and allowed to grow to postnatal day 92. Anxiety-like behaviors were measured by the elevated plus-maze test. Brain regions from adult rats were used to examine changes in alpha-melanocyte stimulating hormone, melanocortin 4 receptor, and neuropeptide Y expression and the histone acetylation status of their promoters. Results Adolescent intermittent ethanol-exposed adult rats displayed anxiety-like behaviors and showed increased pro-opiomelanocortin mRNA levels in the hypothalamus and increased melanocortin 4 receptor mRNA levels in both the amygdala and hypothalamus compared with adolescent intermittent saline-exposed adult rats. The alpha-Melanocyte stimulating hormone and melanocortin 4 receptor protein levels were increased in the central and medial nucleus of the amygdala, paraventricular nucleus, and arcuate nucleus of the hypothalamus in adolescent intermittent ethanol-exposed compared with adolescent intermittent saline-exposed adult rats. Neuropeptide Y protein levels were decreased in the central and medial nucleus of the amygdala of adolescent intermittent ethanol-exposed compared with adolescent intermittent saline-exposed adult rats. Histone H3K9/14 acetylation was decreased in the neuropeptide Y promoter in the amygdala but increased in the melanocortin 4 receptor gene promoter in the amygdala and the melanocortin 4 receptor and pro-opiomelanocortin promoters in the hypothalamus of adolescent intermittent ethanol-exposed adult rats compared with controls. Conclusions Increased melanocortin and decreased neuropeptide Y activity due to changes in histone acetylation in emotional brain circuitry may play a role in adolescent intermittent ethanol-induced anxiety phenotypes in adulthood.
Collapse
Affiliation(s)
- Dadasaheb M Kokare
- Center for Alcohol Research in Epigenetics, Department of Psychiatry (Dr Kokare, Mr Kyzar, and Drs Zhang, Sakharkar, and Pandey), and Department of Anatomy and Cell Biology (Dr Pandey), University of Illinois at Chicago, Chicago; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois (Mr Kyzar and Drs Zhang, Sakharkar, and Pandey)
| | - Evan J Kyzar
- Center for Alcohol Research in Epigenetics, Department of Psychiatry (Dr Kokare, Mr Kyzar, and Drs Zhang, Sakharkar, and Pandey), and Department of Anatomy and Cell Biology (Dr Pandey), University of Illinois at Chicago, Chicago; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois (Mr Kyzar and Drs Zhang, Sakharkar, and Pandey)
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics, Department of Psychiatry (Dr Kokare, Mr Kyzar, and Drs Zhang, Sakharkar, and Pandey), and Department of Anatomy and Cell Biology (Dr Pandey), University of Illinois at Chicago, Chicago; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois (Mr Kyzar and Drs Zhang, Sakharkar, and Pandey)
| | - Amul J Sakharkar
- Center for Alcohol Research in Epigenetics, Department of Psychiatry (Dr Kokare, Mr Kyzar, and Drs Zhang, Sakharkar, and Pandey), and Department of Anatomy and Cell Biology (Dr Pandey), University of Illinois at Chicago, Chicago; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois (Mr Kyzar and Drs Zhang, Sakharkar, and Pandey)
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry (Dr Kokare, Mr Kyzar, and Drs Zhang, Sakharkar, and Pandey), and Department of Anatomy and Cell Biology (Dr Pandey), University of Illinois at Chicago, Chicago; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois (Mr Kyzar and Drs Zhang, Sakharkar, and Pandey)
| |
Collapse
|
170
|
Antipsychotic-induced Hdac2 transcription via NF-κB leads to synaptic and cognitive side effects. Nat Neurosci 2017; 20:1247-1259. [PMID: 28783139 DOI: 10.1038/nn.4616] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022]
Abstract
Antipsychotic drugs remain the standard for schizophrenia treatment. Despite their effectiveness in treating hallucinations and delusions, prolonged exposure to antipsychotic medications leads to cognitive deficits in both schizophrenia patients and animal models. The molecular mechanisms underlying these negative effects on cognition remain to be elucidated. Here we demonstrate that chronic antipsychotic drug exposure increases nuclear translocation of NF-κB in both mouse and human frontal cortex, a trafficking event triggered via 5-HT2A-receptor-dependent downregulation of the NF-κB repressor IκBα. This upregulation of NF-κB activity led to its increased binding at the Hdac2 promoter, thereby augmenting Hdac2 transcription. Deletion of HDAC2 in forebrain pyramidal neurons prevented the negative effects of antipsychotic treatment on synaptic remodeling and cognition. Conversely, virally mediated activation of NF-κB signaling decreased cortical synaptic plasticity via HDAC2. Together, these observations may aid in developing therapeutic strategies to improve the outcome of schizophrenia treatment.
Collapse
|
171
|
Hennig KM, Fass DM, Zhao WN, Sheridan SD, Fu T, Erdin S, Stortchevoi A, Lucente D, Cody JD, Sweetser D, Gusella JF, Talkowski ME, Haggarty SJ. WNT/β-Catenin Pathway and Epigenetic Mechanisms Regulate the Pitt-Hopkins Syndrome and Schizophrenia Risk Gene TCF4. MOLECULAR NEUROPSYCHIATRY 2017; 3:53-71. [PMID: 28879201 DOI: 10.1159/000475666] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/07/2017] [Indexed: 12/11/2022]
Abstract
Genetic variation within the transcription factor TCF4 locus can cause the intellectual disability and developmental disorder Pitt-Hopkins syndrome (PTHS), whereas single-nucleotide polymorphisms within noncoding regions are associated with schizophrenia. These genetic findings position TCF4 as a link between transcription and cognition; however, the neurobiology of TCF4 remains poorly understood. Here, we quantitated multiple distinct TCF4 transcript levels in human induced pluripotent stem cell-derived neural progenitors and differentiated neurons, and PTHS patient fibroblasts. We identify two classes of pharmacological treatments that regulate TCF4 expression: WNT pathway activation and inhibition of class I histone deacetylases. In PTHS fibroblasts, both of these perturbations upregulate a subset of TCF4 transcripts. Finally, using chromatin immunoprecipitation sequencing in conjunction with genome-wide transcriptome analysis, we identified TCF4 target genes that may mediate the effect of TCF4 loss on neuroplasticity. Our studies identify new pharmacological assays, tools, and targets for the development of therapeutics for cognitive disorders.
Collapse
Affiliation(s)
- Krista M Hennig
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel M Fass
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts, USA
| | - Wen-Ning Zhao
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven D Sheridan
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Ting Fu
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Serkan Erdin
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexei Stortchevoi
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Diane Lucente
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jannine D Cody
- Chromosome 18 Clinical Research Center, Department of Pediatrics, University of Texas Health Sciences Center, San Antonio, Texas, USA.,The Chromosome 18 Registry and Research Society, San Antonio, Texas, USA
| | - David Sweetser
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Divisions of Pediatric Hematology/Oncology and Medical Genetics, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James F Gusella
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael E Talkowski
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts, USA.,Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
172
|
Kwapis JL, Jarome TJ, Ferrara NC, Helmstetter FJ. Updating Procedures Can Reorganize the Neural Circuit Supporting a Fear Memory. Neuropsychopharmacology 2017; 42:1688-1697. [PMID: 28139682 PMCID: PMC5518901 DOI: 10.1038/npp.2017.23] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 11/08/2022]
Abstract
Established memories undergo a period of vulnerability following retrieval, a process termed 'reconsolidation.' Recent work has shown that the hypothetical process of reconsolidation is only triggered when new information is presented during retrieval, suggesting that this process may allow existing memories to be modified. Reconsolidation has received increasing attention as a possible therapeutic target for treating disorders that stem from traumatic memories, yet little is known about how this process changes the original memory. In particular, it is unknown whether reconsolidation can reorganize the neural circuit supporting an existing memory after that memory is modified with new information. Here, we show that trace fear memory undergoes a protein synthesis-dependent reconsolidation process following exposure to a single updating trial of delay conditioning. Further, this reconsolidation-dependent updating process appears to reorganize the neural circuit supporting the trace-trained memory, so that it better reflects the circuit supporting delay fear. Specifically, after a trace-to-delay update session, the amygdala is now required for extinction of the updated memory but the retrosplenial cortex is no longer required for retrieval. These results suggest that updating procedures could be used to force a complex, poorly defined memory circuit to rely on a better-defined neural circuit that may be more amenable to behavioral or pharmacological manipulation. This is the first evidence that exposure to new information can fundamentally reorganize the neural circuit supporting an existing memory.
Collapse
Affiliation(s)
- Janine L Kwapis
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Timothy J Jarome
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Nicole C Ferrara
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Fred J Helmstetter
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| |
Collapse
|
173
|
Delgado-Morales R, Agís-Balboa RC, Esteller M, Berdasco M. Epigenetic mechanisms during ageing and neurogenesis as novel therapeutic avenues in human brain disorders. Clin Epigenetics 2017; 9:67. [PMID: 28670349 PMCID: PMC5493012 DOI: 10.1186/s13148-017-0365-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/11/2017] [Indexed: 12/26/2022] Open
Abstract
Ageing is the main risk factor for human neurological disorders. Among the diverse molecular pathways that govern ageing, epigenetics can guide age-associated decline in part by regulating gene expression and also through the modulation of genomic instability and high-order chromatin architecture. Epigenetic mechanisms are involved in the regulation of neural differentiation as well as in functional processes related to memory consolidation, learning or cognition during healthy lifespan. On the other side of the coin, many neurodegenerative diseases are associated with epigenetic dysregulation. The reversible nature of epigenetic factors and, especially, their role as mediators between the genome and the environment make them exciting candidates as therapeutic targets. Rather than providing a broad description of the pathways epigenetically deregulated in human neurological disorders, in this review, we have focused on the potential use of epigenetic enzymes as druggable targets to ameliorate neural decline during normal ageing and especially in neurological disorders. We will firstly discuss recent progress that supports a key role of epigenetic regulation during healthy ageing with an emphasis on the role of epigenetic regulation in adult neurogenesis. Then, we will focus on epigenetic alterations associated with ageing-related human disorders of the central nervous system. We will discuss examples in the context of psychiatric disorders, including schizophrenia and posttraumatic stress disorders, and also dementia or Alzheimer's disease as the most frequent neurodegenerative disease. Finally, methodological limitations and future perspectives are discussed.
Collapse
Affiliation(s)
- Raúl Delgado-Morales
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), 3rd Floor, Hospital Duran i Reynals, Av. Gran Via 199-203, 08908L'Hospitalet, Barcelona, Catalonia Spain.,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Roberto Carlos Agís-Balboa
- Psychiatric Diseases Research Group, Galicia Sur Health Research Institute, Complexo Hospitalario Universitario de Vigo (CHUVI), SERGAS, CIBERSAM, Vigo, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), 3rd Floor, Hospital Duran i Reynals, Av. Gran Via 199-203, 08908L'Hospitalet, Barcelona, Catalonia Spain.,Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - María Berdasco
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), 3rd Floor, Hospital Duran i Reynals, Av. Gran Via 199-203, 08908L'Hospitalet, Barcelona, Catalonia Spain
| |
Collapse
|
174
|
Opening a New Time Window for Treatment of Stroke by Targeting HDAC2. J Neurosci 2017; 37:6712-6728. [PMID: 28592694 DOI: 10.1523/jneurosci.0341-17.2017] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/10/2017] [Accepted: 06/01/2017] [Indexed: 11/21/2022] Open
Abstract
Narrow therapeutic window limits treatments with thrombolysis and neuroprotection for most stroke patients. Widening therapeutic window remains a critical challenge. Understanding the key mechanisms underlying the pathophysiological events in the peri-infarct area where secondary injury coexists with neuroplasticity over days to weeks may offer an opportunity for expanding the therapeutic window. Here we show that ischemia-induced histone deacetylase 2 (HDAC2) upregulation from 5 to 7 d after stroke plays a crucial role. In this window phase, suppressing HDAC2 in the peri-infarct cortex of rodents by HDAC inhibitors, knockdown or knock-out of Hdac2 promoted recovery of motor function from stroke via epigenetically enhancing cells survival and neuroplasticity of surviving neurons as well as reducing neuroinflammation, whereas overexpressing HDAC2 worsened stroke-induced functional impairment of both WT and Hdac2 conditional knock-out mice. More importantly, inhibiting other isoforms of HDACs had no effect. Thus, the intervention by precisely targeting HDAC2 in this window phase is a novel strategy for the functional recovery of stroke survivors.SIGNIFICANCE STATEMENT Narrow time window phase impedes current therapies for stroke patients. Understanding the key mechanisms underlying secondary injury may open a new window for pharmacological interventions to promote recovery from stroke. Our study indicates that ischemia-induced histone deacetylase 2 upregulation from 5 to 7 d after stroke mediates the secondary functional loss by reducing survival and neuroplasticity of peri-infarct neurons as well as augmenting neuroinflammation. Thus, precisely targeting histone deacetylase 2 in the window phase provides a novel therapeutic strategy for stroke recovery.
Collapse
|
175
|
Wang D, Liu C, Li Z, Wang Y, Wang W, Wu X, Wang K, Miao W, Li L, Peng L. Regulation of Histone Acetylation on Expression Profiles of Potassium Channels During Cardiomyocyte Differentiation From Mouse Embryonic Stem Cells. J Cell Biochem 2017; 118:4460-4467. [PMID: 28464250 DOI: 10.1002/jcb.26102] [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: 03/09/2017] [Accepted: 04/28/2017] [Indexed: 11/06/2022]
Abstract
The cardiomyocyte differentiation from mouse embryonic stem cells (mESCs) is a dynamic and complex process that involved in the precision regulation of histone acetylation. The formation of action potential (AP) in mature cardiomyocytes is based on the expression pattern of Na+ , Ca2+ , and K+ ion channels, in which the slow delayed rectifier potassium current (IKs ), the rapid delayed rectifier potassium current (IKr ) and the inwardly rectifying Kir current (IK1 ) mainly contribute to repolarization for AP in different species. However, the expression status of potassium channels conducted IKs , IKr , and IK1 in cardiomyocyte differentiation are not fully defined. Here, we investigated the expression pattern of the slow delayed rectifier potassium channel and the rapid delayed rectifier potassium channel using a model of mouse cardiomyocyte differentiation under different conditions of histone acetylation. We found that expression levels of both the delayed rectifier potassium channel and the inwardly rectifying potassium channel were more sensitive to histone hyperacetylation during differentiation from mESCs into cardiomyocytes. Especially, histone H4 hyperacetylation induced by Class I HDACs inhibitors promoted the expression profiles of potassium channels (Kcnj2, Kcnj3, Kcnj5, Kcnj11, and Kcnh2) in the process. Our results provide a clue for expression status of potassium channels which may be essential to forming functional cardiomyocyte in the cardiac lineage commitment of mESC. J. Cell. Biochem. 118: 4460-4467, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Duo Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Chang Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhigang Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yumei Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Wenjing Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiujuan Wu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Kang Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Wei Miao
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Li Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Luying Peng
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai 200092, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
176
|
Lee JLC, Nader K, Schiller D. An Update on Memory Reconsolidation Updating. Trends Cogn Sci 2017; 21:531-545. [PMID: 28495311 DOI: 10.1016/j.tics.2017.04.006] [Citation(s) in RCA: 292] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 10/19/2022]
Abstract
The reactivation of a stored memory in the brain can make the memory transiently labile. During the time it takes for the memory to restabilize (reconsolidate) the memory can either be reduced by an amnesic agent or enhanced by memory enhancers. The change in memory expression is related to changes in the brain correlates of long-term memory. Many have suggested that such retrieval-induced plasticity is ideally placed to enable memories to be updated with new information. This hypothesis has been tested experimentally, with a translational perspective, by attempts to update maladaptive memories to reduce their problematic impact. We review here progress on reconsolidation updating studies, highlighting their translational exploitation and addressing recent challenges to the reconsolidation field.
Collapse
Affiliation(s)
- Jonathan L C Lee
- School of Psychology, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Karim Nader
- Department of Psychology, McGill University, Department of Psychology,1205 Dr Penfield Avenue, Montreal, QC H3A 1B1, Canada.
| | - Daniela Schiller
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai and Friedman Brain Institute, New York, NY 10029, USA.
| |
Collapse
|
177
|
Context and Auditory Fear are Differentially Regulated by HDAC3 Activity in the Lateral and Basal Subnuclei of the Amygdala. Neuropsychopharmacology 2017; 42:1284-1294. [PMID: 27924874 PMCID: PMC5437888 DOI: 10.1038/npp.2016.274] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/04/2016] [Accepted: 12/02/2016] [Indexed: 01/17/2023]
Abstract
Histone acetylation is a fundamental epigenetic mechanism that is dynamically regulated during memory formation. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) compete to modulate histone acetylation, allowing for rapid changes in acetylation in response to a learning event. HDACs are known to be powerful negative regulators of memory formation, but it is not clear whether this function depends on HDAC enzymatic activity per se. Here, we tested whether the enzymatic activity of an individual Class I HDAC, HDAC3, has a role in fear memory formation in subregions of the hippocampus and amygdala. We found that fear conditioning drove expression of the immediate early genes cFos and Nr4a2 in the hippocampus, which coincided with reduced HDAC3 occupancy at these promoters. Using a dominant-negative, deacetylase-dead point mutant virus (AAV-HDAC3(Y298H)-v5), we found that selectively blocking HDAC3 deacetylase activity in either the dorsal hippocampus or basal nucleus of the amygdala enhanced context fear without affecting tone fear. Blocking HDAC3 activity in the lateral nucleus of the amygdala, on the other hand, enhanced tone, but not context fear memory. These results show for the first time that the enzymatic activity of HDAC3 functions to negatively regulate fear memory formation. Further, HDAC3 activity regulates different aspects of fear memory in the basal and lateral subregions of the amygdala. Thus, the deacetylase activity of HDAC3 is a powerful negative regulator of fear memory formation in multiple subregions of the fear circuit.
Collapse
|
178
|
Ripoli C. Engrampigenetics: Epigenetics of engram memory cells. Behav Brain Res 2017; 325:297-302. [DOI: 10.1016/j.bbr.2016.11.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/24/2016] [Accepted: 11/26/2016] [Indexed: 12/21/2022]
|
179
|
Ju LS, Yang JJ, Lei L, Xia JY, Luo D, Ji MH, Martynyuk AE, Yang JJ. The Combination of Long-term Ketamine and Extinction Training Contributes to Fear Erasure by Bdnf Methylation. Front Cell Neurosci 2017; 11:100. [PMID: 28473755 PMCID: PMC5398013 DOI: 10.3389/fncel.2017.00100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 03/27/2017] [Indexed: 12/13/2022] Open
Abstract
A combination of antidepressant drugs and psychotherapy exhibits more promising efficacy in treating fear disorders than either treatment alone, but underlying mechanisms of such treatments remain largely unknown. Here we investigated the role of DNA methylation of the brain-derived neurotrophic factor (Bdnf) gene in the therapeutic effects of ketamine in combination with extinction training in a mouse model of post-traumatic stress disorder (PTSD) induced by inescapable electric foot shocks (IFS). Male mice received ketamine for 22 consecutive days starting 1 h after the IFS (long-term ketamine treatment) or 2 h prior to the extinction training on days 15 and 16 after the IFS (short-term ketamine treatment). The Open Field (OF) and Elevated Plus Maze (EPM) tests were conducted on days 18 and 20. The spontaneous recovery and fear renewal tests were performed on day 23. Mice, subjected to IFS, exhibited anxiety-like behavior and fear relapse, accompanied by the increased levels of DNA methyltransferases, hyper-methylation of Bdnf gene, and decreased BDNF mRNA expression in the medial prefrontal cortex (mPFC) and hippocampus (HIP). Long-term treatment with ketamine combined with extinction training alleviated the IFS-induced abnormalities. These results suggest that long-term ketamine treatment in combination with extinction training may ameliorate fear relapse in the murine model of PTSD, at least in part, by normalizing DNA methylation of Bdnf gene.
Collapse
Affiliation(s)
- Ling-Sha Ju
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast UniversityNanjing, China
| | - Jiao-Jiao Yang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast UniversityNanjing, China.,Department of Anesthesiology and the McKnight Brain Institute, University of Florida College of MedicineGainesville, FL, USA
| | - Lei Lei
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast UniversityNanjing, China
| | - Jiang-Yan Xia
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast UniversityNanjing, China
| | - Dan Luo
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast UniversityNanjing, China
| | - Mu-Huo Ji
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast UniversityNanjing, China
| | - Anatoly E Martynyuk
- Department of Anesthesiology and the McKnight Brain Institute, University of Florida College of MedicineGainesville, FL, USA
| | - Jian-Jun Yang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast UniversityNanjing, China
| |
Collapse
|
180
|
Auchter AM, Shumake J, Gonzalez-Lima F, Monfils MH. Preventing the return of fear using reconsolidation updating and methylene blue is differentially dependent on extinction learning. Sci Rep 2017; 7:46071. [PMID: 28397861 PMCID: PMC5387397 DOI: 10.1038/srep46071] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/08/2017] [Indexed: 12/23/2022] Open
Abstract
Many factors account for how well individuals extinguish conditioned fears, such as genetic variability, learning capacity and conditions under which extinction training is administered. We predicted that memory-based interventions would be more effective to reduce the reinstatement of fear in subjects genetically predisposed to display more extinction learning. We tested this hypothesis in rats genetically selected for differences in fear extinction using two strategies: (1) attenuation of fear memory using post-retrieval extinction training, and (2) pharmacological enhancement of the extinction memory after extinction training by low-dose USP methylene blue (MB). Subjects selectively bred for divergent extinction phenotypes were fear conditioned to a tone stimulus and administered either standard extinction training or retrieval + extinction. Following extinction, subjects received injections of saline or MB. Both reconsolidation updating and MB administration showed beneficial effects in preventing fear reinstatement, but differed in the groups they targeted. Reconsolidation updating showed an overall effect in reducing fear reinstatement, whereas pharmacological memory enhancement using MB was an effective strategy, but only for individuals who were responsive to extinction.
Collapse
Affiliation(s)
- Allison M Auchter
- The University of Texas at Austin, Department of Psychology, Austin, TX 78712, USA
| | - Jason Shumake
- The University of Texas at Austin, Department of Psychology, Austin, TX 78712, USA.,Institute for Mental Health Research, The University of Texas at Austin, USA
| | | | - Marie H Monfils
- The University of Texas at Austin, Department of Psychology, Austin, TX 78712, USA.,Institute for Mental Health Research, The University of Texas at Austin, USA
| |
Collapse
|
181
|
Ding X, Liu S, Tian M, Zhang W, Zhu T, Li D, Wu J, Deng H, Jia Y, Xie W, Xie H, Guan JS. Activity-induced histone modifications govern Neurexin-1 mRNA splicing and memory preservation. Nat Neurosci 2017; 20:690-699. [PMID: 28346453 DOI: 10.1038/nn.4536] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 02/26/2017] [Indexed: 12/12/2022]
Abstract
Epigenetic mechanisms regulate the formation, consolidation and reconsolidation of memories. However, the signaling path from neuronal activation to epigenetic modifications within the memory-related brain circuit remains unknown. We report that learning induces long-lasting histone modifications in hippocampal memory-activated neurons to regulate memory stability. Neuronal activity triggers a late-onset shift in Nrxn1 splice isoform choice at splicing site 4 by accumulating a repressive histone marker, H3K9me3, to modulate the splicing process. Activity-dependent phosphorylation of p66α via AMP-activated protein kinase recruits HDAC2 and Suv39h1 to establish repressive histone markers and changes the connectivity of the activated neurons. Removal of Suv39h1 abolished the activity-dependent shift in Nrxn1 splice isoform choice and reduced the stability of established memories. We uncover a cell-autonomous process for memory preservation in which memory-related neurons initiate a late-onset reduction of their rewiring capacities through activity-induced histone modifications.
Collapse
Affiliation(s)
- Xinlu Ding
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China.,Centre for Brain Inspired Computing Research, Tsinghua University, Beijing, China
| | - Sanxiong Liu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China.,Centre for Brain Inspired Computing Research, Tsinghua University, Beijing, China
| | - Miaomiao Tian
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China.,Centre for Brain Inspired Computing Research, Tsinghua University, Beijing, China
| | - Wenhao Zhang
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tao Zhu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China.,Centre for Brain Inspired Computing Research, Tsinghua University, Beijing, China
| | - Dongdong Li
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China.,Centre for Brain Inspired Computing Research, Tsinghua University, Beijing, China
| | - Jiawei Wu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - HaiTeng Deng
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yichang Jia
- School of Medicine, Tsinghua University, Beijing, China
| | - Wei Xie
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hong Xie
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China.,Centre for Brain Inspired Computing Research, Tsinghua University, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| | - Ji-Song Guan
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China.,Centre for Brain Inspired Computing Research, Tsinghua University, Beijing, China
| |
Collapse
|
182
|
Gershman SJ, Monfils MH, Norman KA, Niv Y. The computational nature of memory modification. eLife 2017; 6:e23763. [PMID: 28294944 PMCID: PMC5391211 DOI: 10.7554/elife.23763] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 03/13/2017] [Indexed: 11/25/2022] Open
Abstract
Retrieving a memory can modify its influence on subsequent behavior. We develop a computational theory of memory modification, according to which modification of a memory trace occurs through classical associative learning, but which memory trace is eligible for modification depends on a structure learning mechanism that discovers the units of association by segmenting the stream of experience into statistically distinct clusters (latent causes). New memories are formed when the structure learning mechanism infers that a new latent cause underlies current sensory observations. By the same token, old memories are modified when old and new sensory observations are inferred to have been generated by the same latent cause. We derive this framework from probabilistic principles, and present a computational implementation. Simulations demonstrate that our model can reproduce the major experimental findings from studies of memory modification in the Pavlovian conditioning literature.
Collapse
Affiliation(s)
- Samuel J Gershman
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, United States
| | - Marie-H Monfils
- Department of Psychology, University of Texas, Austin, United States
| | - Kenneth A Norman
- Princeton Neuroscience Institute and Department of Psychology, Princeton University, Princeton, United States
| | - Yael Niv
- Princeton Neuroscience Institute and Department of Psychology, Princeton University, Princeton, United States
| |
Collapse
|
183
|
HDAC7 Ubiquitination by the E3 Ligase CBX4 Is Involved in Contextual Fear Conditioning Memory Formation. J Neurosci 2017; 37:3848-3863. [PMID: 28283560 DOI: 10.1523/jneurosci.2773-16.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 12/18/2022] Open
Abstract
Histone acetylation, an epigenetic modification, plays an important role in long-term memory formation. Recently, histone deacetylase (HDAC) inhibitors were demonstrated to promote memory formation, which raises the intriguing possibility that they may be used to rescue memory deficits. However, additional research is necessary to clarify the roles of individual HDACs in memory. In this study, we demonstrated that HDAC7, within the dorsal hippocampus of C57BL6J mice, had a late and persistent decrease after contextual fear conditioning (CFC) training (4-24 h), which was involved in long-term CFC memory formation. We also showed that HDAC7 decreased via ubiquitin-dependent degradation. CBX4 was one of the HDAC7 E3 ligases involved in this process. Nur77, as one of the target genes of HDAC7, increased 6-24 h after CFC training and, accordingly, modulated the formation of CFC memory. Finally, HDAC7 was involved in the formation of other hippocampal-dependent memories, including the Morris water maze and object location test. The current findings facilitate an understanding of the molecular and cellular mechanisms of HDAC7 in the regulation of hippocampal-dependent memory.SIGNIFICANCE STATEMENT The current findings demonstrated the effects of histone deacetylase 7 (HDAC7) on hippocampal-dependent memories. Moreover, we determined the mechanism of decreased HDAC7 in contextual fear conditioning (CFC) through ubiquitin-dependent protein degradation. We also verified that CBX4 was one of the HDAC7 E3 ligases. Finally, we demonstrated that Nur77, as one of the important targets for HDAC7, was involved in CFC memory formation. All of these proteins, including HDAC7, CBX4, and Nur77, could be potential therapeutic targets for preventing memory deficits in aging and neurological diseases.
Collapse
|
184
|
Scott H, Smith AE, Barker GR, Uney JB, Warburton EC. Contrasting roles for DNA methyltransferases and histone deacetylases in single-item and associative recognition memory. NEUROEPIGENETICS 2017; 9:1-9. [PMID: 28367410 PMCID: PMC5364272 DOI: 10.1016/j.nepig.2017.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/27/2017] [Accepted: 02/27/2017] [Indexed: 12/17/2022]
Abstract
Recognition memory enables us to judge whether we have encountered a stimulus before and to recall associated information, including where the stimulus was encountered. The perirhinal cortex (PRh) is required for judgment of stimulus familiarity, while hippocampus (HPC) and medial prefrontal cortex (mPFC) are additionally involved when spatial information associated with a stimulus needs to be remembered. While gene expression is known to be essential for the consolidation of long-term recognition memory, the underlying regulatory mechanisms are not fully understood. Here we investigated the roles of two epigenetic mechanisms, DNA methylation and histone deacetylation, in recognition memory. Infusion of DNA methyltransferase inhibitors into PRh impaired performance in novel object recognition and object-in-place tasks while infusions into HPC or mPFC impaired object-in-place performance only. In contrast, inhibition of histone deacetylases in PRh, but not mPFC, enhanced recognition memory. These results support the emerging role of epigenetic processes in learning and memory.
Collapse
Affiliation(s)
- Hannah Scott
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Anna E. Smith
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Gareth R. Barker
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - James B. Uney
- School of Clinical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - E. Clea Warburton
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
185
|
Webb WM, Sanchez RG, Perez G, Butler AA, Hauser RM, Rich MC, O'Bierne AL, Jarome TJ, Lubin FD. Dynamic association of epigenetic H3K4me3 and DNA 5hmC marks in the dorsal hippocampus and anterior cingulate cortex following reactivation of a fear memory. Neurobiol Learn Mem 2017; 142:66-78. [PMID: 28232238 DOI: 10.1016/j.nlm.2017.02.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/28/2017] [Accepted: 02/15/2017] [Indexed: 12/20/2022]
Abstract
Epigenetic mechanisms such as DNA methylation and histone methylation are critical regulators of gene transcription changes during memory consolidation. However, it is unknown how these epigenetic modifications coordinate control of gene expression following reactivation of a previously consolidated memory. Here, we found that retrieval of a recent contextual fear conditioned memory increased global levels of H3 lysine 4-trimethylation (H3K4me3) and DNA 5-hydroxymethylation (5hmC) in area CA1 of the dorsal hippocampus. Further experiments revealed increased levels of H3K4me3 and DNA 5hmC within a CpG-enriched coding region of the Npas4, but not c-fos, gene. Intriguingly, retrieval of a 30-day old memory increased H3K4me3 and DNA 5hmC levels at a CpG-enriched coding region of c-fos, but not Npas4, in the anterior cingulate cortex, suggesting that while these two epigenetic mechanisms co-occur following the retrieval of a recent or remote memory, their gene targets differ depending on the brain region. Additionally, we found that in vivo siRNA-mediated knockdown of the H3K4me3 methyltransferase Mll1 in CA1 abolished retrieval-induced increases in DNA 5hmC levels at the Npas4 gene, suggesting that H3K4me3 couples to DNA 5hmC mechanisms. Consistent with this, loss of Mll1 prevented retrieval-induced increases in Npas4 mRNA levels in CA1 and impaired fear memory. Collectively, these findings suggest an important link between histone methylation and DNA hydroxymethylation mechanisms in the epigenetic control of de novo gene transcription triggered by memory retrieval.
Collapse
Affiliation(s)
- William M Webb
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Richard G Sanchez
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Gabriella Perez
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Anderson A Butler
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Rebecca M Hauser
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Megan C Rich
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Aidan L O'Bierne
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Timothy J Jarome
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Farah D Lubin
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
186
|
Hemstedt TJ, Lattal KM, Wood MA. Reconsolidation and extinction: Using epigenetic signatures to challenge conventional wisdom. Neurobiol Learn Mem 2017; 142:55-65. [PMID: 28119018 DOI: 10.1016/j.nlm.2017.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/15/2017] [Accepted: 01/16/2017] [Indexed: 12/17/2022]
Abstract
Epigenetic mechanisms have the potential to give rise to lasting changes in cell function that ultimately can affect behavior persistently. This concept is especially interesting with respect to fear reconsolidation and fear memory extinction. These two behavioral approaches are used in the laboratory to investigate how fear memory can be attenuated, which becomes important when searching for therapeutic intervention to treat anxiety disorders and post-traumatic stress disorder. Here we review the role of several key epigenetic mechanisms in reconsolidation and extinction of learned fear and their potential to persistently alter behavioral responses to conditioned cues. We also briefly discuss how epigenetic mechanisms may establish persistent behaviors that challenge our definitions of extinction and reconsolidation.
Collapse
Affiliation(s)
- Thekla J Hemstedt
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, Irvine, CA, USA
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA; Center for the Neurobiology of Learning and Memory, Irvine, CA, USA.
| |
Collapse
|
187
|
Bartlett AA, Singh R, Hunter RG. Anxiety and Epigenetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 978:145-166. [PMID: 28523545 DOI: 10.1007/978-3-319-53889-1_8] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Anxiety disorders are highly prevalent psychiatric disorders often comorbid with depression and substance abuse. Twin studies have shown that anxiety disorders are moderately heritable. Yet, genome-wide association studies (GWASs) have failed to identify gene(s) significantly associated with diagnosis suggesting a strong role for environmental factors and the epigenome. A number of anxiety disorder subtypes are considered "stress related." A large focus of research has been on the epigenetic and anxiety-like behavioral consequences of stress. Animal models of anxiety-related disorders have provided strong evidence for the role of stress on the epigenetic control of the hypothalamic-pituitary-adrenal (HPA) axis and of stress-responsive brain regions. Neuroepigenetics may continue to explain individual variation in susceptibility to environmental perturbations and consequently anxious behavior. Behavioral and pharmacological interventions aimed at targeting epigenetic marks associated with anxiety may prove fruitful in developing treatments.
Collapse
Affiliation(s)
- Andrew A Bartlett
- Department of Psychology, University of Massachusetts, 100 Morrissey Blvd, Boston, MA, 02125, USA
| | - Rumani Singh
- Department of Psychology, University of Massachusetts, 100 Morrissey Blvd, Boston, MA, 02125, USA
| | - Richard G Hunter
- Department of Psychology, University of Massachusetts, 100 Morrissey Blvd, Boston, MA, 02125, USA.
| |
Collapse
|
188
|
Rusconi F, Grillo B, Toffolo E, Mattevi A, Battaglioli E. NeuroLSD1: Splicing-Generated Epigenetic Enhancer of Neuroplasticity. Trends Neurosci 2017; 40:28-38. [DOI: 10.1016/j.tins.2016.11.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 11/18/2016] [Indexed: 12/17/2022]
|
189
|
Prospects for Medications to Reverse Causative Epigenetic Processes in Neuropsychiatry Disorders. Neuropsychopharmacology 2017; 42:367-368. [PMID: 27909326 PMCID: PMC5143508 DOI: 10.1038/npp.2016.219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
190
|
Kroes MCW, Schiller D, LeDoux JE, Phelps EA. Translational Approaches Targeting Reconsolidation. Curr Top Behav Neurosci 2016; 28:197-230. [PMID: 27240676 PMCID: PMC5646834 DOI: 10.1007/7854_2015_5008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Maladaptive learned responses and memories contribute to psychiatric disorders that constitute a significant socio-economic burden. Primary treatment methods teach patients to inhibit maladaptive responses, but do not get rid of the memory itself, which explains why many patients experience a return of symptoms even after initially successful treatment. This highlights the need to discover more persistent and robust techniques to diminish maladaptive learned behaviours. One potentially promising approach is to alter the original memory, as opposed to inhibiting it, by targeting memory reconsolidation. Recent research shows that reactivating an old memory results in a period of memory flexibility and requires restorage, or reconsolidation, for the memory to persist. This reconsolidation period allows a window for modification of a specific old memory. Renewal of memory flexibility following reactivation holds great clinical potential as it enables targeting reconsolidation and changing of specific learned responses and memories that contribute to maladaptive mental states and behaviours. Here, we will review translational research on non-human animals, healthy human subjects, and clinical populations aimed at altering memories by targeting reconsolidation using biological treatments (electrical stimulation, noradrenergic antagonists) or behavioural interference (reactivation-extinction paradigm). Both approaches have been used successfully to modify aversive and appetitive memories, yet effectiveness in treating clinical populations has been limited. We will discuss that memory flexibility depends on the type of memory tested and the brain regions that underlie specific types of memory. Further, when and how we can most effectively reactivate a memory and induce flexibility is largely unclear. Finally, the development of drugs that can target reconsolidation and are safe for use in humans would optimize cross-species translations. Increasing the understanding of the mechanism and limitations of memory flexibility upon reactivation should help optimize efficacy of treatments for psychiatric patients.
Collapse
Affiliation(s)
- Marijn C W Kroes
- Department of Psychology, Centre for Neural Science, New York University, New York, NY, 10003, USA
| | - Daniela Schiller
- Department of Psychiatry and Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, 10029, USA.
| | - Joseph E LeDoux
- Department of Psychology, Centre for Neural Science, New York University, New York, NY, 10003, USA
- Nathan Kline Institute, Orangeburg, NY, 10962, USA
| | - Elizabeth A Phelps
- Department of Psychology, Centre for Neural Science, New York University, New York, NY, 10003, USA
- Nathan Kline Institute, Orangeburg, NY, 10962, USA
| |
Collapse
|
191
|
Enhancing dopaminergic signaling and histone acetylation promotes long-term rescue of deficient fear extinction. Transl Psychiatry 2016; 6:e974. [PMID: 27922638 PMCID: PMC5315560 DOI: 10.1038/tp.2016.231] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 01/04/2023] Open
Abstract
Extinction-based exposure therapy is used to treat anxiety- and trauma-related disorders; however, there is the need to improve its limited efficacy in individuals with impaired fear extinction learning and to promote greater protection against return-of-fear phenomena. Here, using 129S1/SvImJ mice, which display impaired fear extinction acquisition and extinction consolidation, we revealed that persistent and context-independent rescue of deficient fear extinction in these mice was associated with enhanced expression of dopamine-related genes, such as dopamine D1 (Drd1a) and -D2 (Drd2) receptor genes in the medial prefrontal cortex (mPFC) and amygdala, but not hippocampus. Moreover, enhanced histone acetylation was observed in the promoter of the extinction-regulated Drd2 gene in the mPFC, revealing a potential gene-regulatory mechanism. Although enhancing histone acetylation, via administering the histone deacetylase (HDAC) inhibitor MS-275, does not induce fear reduction during extinction training, it promoted enduring and context-independent rescue of deficient fear extinction consolidation/retrieval once extinction learning was initiated as shown following a mild conditioning protocol. This was associated with enhanced histone acetylation in neurons of the mPFC and amygdala. Finally, as a proof-of-principle, mimicking enhanced dopaminergic signaling by L-dopa treatment rescued deficient fear extinction and co-administration of MS-275 rendered this effect enduring and context-independent. In summary, current data reveal that combining dopaminergic and epigenetic mechanisms is a promising strategy to improve exposure-based behavior therapy in extinction-impaired individuals by initiating the formation of an enduring and context-independent fear-inhibitory memory.
Collapse
|
192
|
Taguchi YH. Principal component analysis based unsupervised feature extraction applied to publicly available gene expression profiles provides new insights into the mechanisms of action of histone deacetylase inhibitors. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.nepig.2016.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
193
|
Hirano Y, Ihara K, Masuda T, Yamamoto T, Iwata I, Takahashi A, Awata H, Nakamura N, Takakura M, Suzuki Y, Horiuchi J, Okuno H, Saitoe M. Shifting transcriptional machinery is required for long-term memory maintenance and modification in Drosophila mushroom bodies. Nat Commun 2016; 7:13471. [PMID: 27841260 PMCID: PMC5114576 DOI: 10.1038/ncomms13471] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 10/06/2016] [Indexed: 01/08/2023] Open
Abstract
Accumulating evidence suggests that transcriptional regulation is required for maintenance of long-term memories (LTMs). Here we characterize global transcriptional and epigenetic changes that occur during LTM storage in the Drosophila mushroom bodies (MBs), structures important for memory. Although LTM formation requires the CREB transcription factor and its coactivator, CBP, subsequent early maintenance requires CREB and a different coactivator, CRTC. Late maintenance becomes CREB independent and instead requires the transcription factor Bx. Bx expression initially depends on CREB/CRTC activity, but later becomes CREB/CRTC independent. The timing of the CREB/CRTC early maintenance phase correlates with the time window for LTM extinction and we identify different subsets of CREB/CRTC target genes that are required for memory maintenance and extinction. Furthermore, we find that prolonging CREB/CRTC-dependent transcription extends the time window for LTM extinction. Our results demonstrate the dynamic nature of stored memory and its regulation by shifting transcription systems in the MBs. Transcriptional regulation is necessary for maintaining long-term memories (LTM) but the mechanistic details are not completely defined. Here the authors identify transcriptional machinery and histone modifiers required for LTM maintenance in Drosophila and show that transcriptional regulation for LTM maintenance is distinct from that for LTM formation.
Collapse
Affiliation(s)
- Yukinori Hirano
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Japan Science and Technology Agency, PRESTO, 4-4-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Kunio Ihara
- Center of Gene Research, Nagoya University, Huro-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Tomoko Masuda
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo 156-0057, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application, Department of Reprogramming Science, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto 606-8507, Japan.,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto 606-8507, Japan.,AMED-CREST, AMED 1-7-1 Otemach, Chiyodaku, Tokyo 100-0004, Japan
| | - Ikuko Iwata
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Aya Takahashi
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroko Awata
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Naosuke Nakamura
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Kyoto Sangyo University, Motoyama, Kamigamo, Kita-ku, Kyoto City 603-8555, Japan
| | - Mai Takakura
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yusuke Suzuki
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Junjiro Horiuchi
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo 156-0057, Japan
| | - Hiroyuki Okuno
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Minoru Saitoe
- Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo 156-0057, Japan
| |
Collapse
|
194
|
Ganai SA, Ramadoss M, Mahadevan V. Histone Deacetylase (HDAC) Inhibitors - emerging roles in neuronal memory, learning, synaptic plasticity and neural regeneration. Curr Neuropharmacol 2016; 14:55-71. [PMID: 26487502 PMCID: PMC4787286 DOI: 10.2174/1570159x13666151021111609] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 08/23/2015] [Accepted: 10/08/2015] [Indexed: 11/22/2022] Open
Abstract
Epigenetic regulation of neuronal signalling through histone acetylation dictates transcription programs that govern neuronal memory, plasticity and learning paradigms. Histone Acetyl Transferases (HATs) and Histone Deacetylases (HDACs) are antagonistic enzymes that regulate gene expression through acetylation and deacetylation of histone proteins around which DNA is wrapped inside a eukaryotic cell nucleus. The epigenetic control of HDACs and the cellular imbalance between HATs and HDACs dictate disease states and have been implicated in muscular dystrophy, loss of memory, neurodegeneration and autistic disorders. Altering gene expression profiles through inhibition of HDACs is now emerging as a powerful technique in therapy. This review presents evolving applications of HDAC inhibitors as potential drugs in neurological research and therapy. Mechanisms that govern their
expression profiles in neuronal signalling, plasticity and learning will be covered. Promising and exciting possibilities of HDAC inhibitors in memory formation, fear conditioning, ischemic stroke and neural regeneration have been detailed.
Collapse
Affiliation(s)
| | | | - Vijayalakshmi Mahadevan
- School of Chemical & Biotechnology SASTRA University Tirumalaisamudram, Thanjavur - 613 401 India.
| |
Collapse
|
195
|
Bitanihirwe BKY, Mauney SA, Woo TUW. Weaving a Net of Neurobiological Mechanisms in Schizophrenia and Unraveling the Underlying Pathophysiology. Biol Psychiatry 2016; 80:589-98. [PMID: 27113498 PMCID: PMC5017894 DOI: 10.1016/j.biopsych.2016.03.1047] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/14/2016] [Accepted: 03/03/2016] [Indexed: 12/26/2022]
Abstract
Perineuronal nets (PNNs) are enigmatic structures composed of extracellular matrix molecules that encapsulate the soma, dendrites, and axon segments of neurons in a lattice-like fashion. Although most PNNs condense around parvalbumin-expressing gamma-aminobutyric acidergic interneurons, some glutamatergic pyramidal cells in the brain are also surrounded by PNNs. Experimental findings suggest pivotal roles of PNNs in the regulation of synaptic formation and function. Also, an increasing body of evidence links PNN abnormalities to schizophrenia. The number of PNNs progressively increases during postnatal development until plateauing around the period of late adolescence and early adulthood, which temporally coincides with the age of onset of schizophrenia. Given the established role of PNNs in modulating developmental plasticity, the PNN represents a possible candidate for altering the onset and progression of schizophrenia. Similarly, the reported function of PNNs in regulating the trafficking of glutamate receptors places them in a critical position to modulate synaptic pathology, considered a cardinal feature of schizophrenia. We discuss the physiologic role of PNNs in neural function, synaptic assembly, and plasticity as well as how they interface with circuit/system mechanisms of cognition. An integrated understanding of these neurobiological processes should provide a better basis to elucidate how PNN abnormalities influence brain function and contribute to the pathogenesis of neurodevelopmental disorders such as schizophrenia.
Collapse
Affiliation(s)
- Byron K Y Bitanihirwe
- Department of International Health, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Sarah A Mauney
- Program in Cellular Neuropathology, McLean Hospital, Belmont
| | - Tsung-Ung W Woo
- Program in Cellular Neuropathology, McLean Hospital, Belmont; Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
196
|
Choi KY, Kim YK. Plasticity-augmented psychotherapy for refractory depressive and anxiety disorders. Prog Neuropsychopharmacol Biol Psychiatry 2016; 70:134-47. [PMID: 27072378 DOI: 10.1016/j.pnpbp.2016.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/28/2022]
Abstract
Psychotherapy and pharmacotherapy have been the mainstays of treatment for depression and anxiety disorders during the last century. However, treatment response has not improved in the last few decades, with only half of all patients responding satisfactorily to typical antidepressants. To fulfill the needs of the remaining patients, new treatments with better efficacy are in demand. The addition of psychotherapy to antidepressant treatment has been shown to be superior to pharmacotherapy alone. However, the time costs of psychotherapy limit its use for clinicians and patients. Advancements in neuroscience have contributed to an improved understanding of the pathogenesis of depressive and anxiety disorders. In particular, recent advances in the field of fear conditioning have provided valuable insight into the treatment of refractory depressive and anxiety disorders. In this review, we studied the reconsolidation-updating paradigm and the concept of epigenetic modification, which has been shown to permanently attenuate remote fear memory. This has implications for drug-augmented, e.g. antidepressant and valproic acid, psychotherapy. Future research on more sophisticated psychotherapy techniques will increase the desirability of this treatment modality for both clinicians and patients.
Collapse
Affiliation(s)
- Kwang-Yeon Choi
- Department of Psychiatry, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
197
|
Zhu Z, Wang Y, Cao Z, Chen B, Cai H, Wu Y, Rao Y. Cue-independent memory impairment by reactivation-coupled interference in human declarative memory. Cognition 2016; 155:125-134. [DOI: 10.1016/j.cognition.2016.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 06/23/2016] [Accepted: 06/27/2016] [Indexed: 11/29/2022]
|
198
|
Koh DXP, Sng JCG. HDAC1 negatively regulates Bdnf and Pvalb required for parvalbumin interneuron maturation in an experience-dependent manner. J Neurochem 2016; 139:369-380. [PMID: 27534825 DOI: 10.1111/jnc.13773] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 06/24/2016] [Accepted: 08/10/2016] [Indexed: 01/29/2023]
Abstract
During early postnatal development, neuronal circuits are sculpted by sensory experience provided by the external environment. This experience-dependent regulation of circuitry development consolidates the balance of excitatory-inhibitory (E/I) neurons in the brain. The cortical barrel-column that innervates a single principal whisker is used to provide a clear reference frame for studying the consolidation of E/I circuitry. Sensory deprivation of S1 at birth disrupts the consolidation of excitatory-inhibitory balance by decreasing inhibitory transmission of parvalbumin interneurons. The molecular mechanisms underlying this decrease in inhibition are not completely understood. Our findings show that epigenetic mechanisms, in particular histone deacetylation by histone deacetylases, negatively regulate the expression of brain-derived neurotrophic factor (Bdnf) and parvalbumin (Pvalb) genes during development, which are required for the maturation of parvalbumin interneurons. After whisker deprivation, increased histone deacetylase 1 expression and activity led to increased histone deacetylase 1 binding and decreased histone acetylation at Bdnf promoters I-IV and Pvalb promoter, resulting in the repression of Bdnf and Pvalb gene transcription. The decrease in Bdnf expression further affected parvalbumin interneuron maturation at layer II/III in S1, demonstrated by decreased parvalbumin expression, a marker for parvalbumin interneuron maturation. Knockdown of HDAC1 recovered Bdnf and Pvalb gene transcription and also prevented the decrease of inhibitory synapses accompanying whisker deprivation.
Collapse
Affiliation(s)
- Dawn X P Koh
- National University of Singapore, Graduate School of Integrative Sciences and Engineering, Singapore, Singapore.,Singapore Institute of Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore.,Department of Pharmacology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Judy C G Sng
- Singapore Institute of Clinical Sciences (SICS), A*STAR, Brenner Centre for Molecular Medicine, Singapore, Singapore. .,Department of Pharmacology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore.
| |
Collapse
|
199
|
Blouin AM, Sillivan SE, Joseph NF, Miller CA. The potential of epigenetics in stress-enhanced fear learning models of PTSD. ACTA ACUST UNITED AC 2016; 23:576-86. [PMID: 27634148 PMCID: PMC5026205 DOI: 10.1101/lm.040485.115] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/14/2016] [Indexed: 11/29/2022]
Abstract
Prolonged distress and dysregulated memory processes are the core features of post-traumatic stress disorder (PTSD) and represent the debilitating, persistent nature of the illness. However, the neurobiological mechanisms underlying the expression of these symptoms are challenging to study in human patients. Stress-enhanced fear learning (SEFL) paradigms, which encompass both stress and memory components in rodents, are emerging as valuable preclinical models of PTSD. Rodent models designed to study the long-term mechanisms of either stress or fear memory alone have identified a critical role for numerous epigenetic modifications to DNA and histone proteins. However, the epigenetic modifications underlying SEFL remain largely unknown. This review will provide a brief overview of the epigenetic modifications implicated in stress and fear memory independently, followed by a description of existing SEFL models and the few epigenetic mechanisms found to date to underlie SEFL. The results of the animal studies discussed here highlight neuroepigenetics as an essential area for future research in the context of PTSD through SEFL studies, because of its potential to identify novel candidates for neurotherapeutics targeting stress-induced pathogenic memories.
Collapse
Affiliation(s)
- Ashley M Blouin
- Department of Metabolism and Aging and Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Stephanie E Sillivan
- Department of Metabolism and Aging and Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Nadine F Joseph
- Department of Metabolism and Aging and Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Courtney A Miller
- Department of Metabolism and Aging and Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| |
Collapse
|
200
|
Jones CE, Monfils MH. Post-retrieval extinction in adolescence prevents return of juvenile fear. ACTA ACUST UNITED AC 2016; 23:567-75. [PMID: 27634147 PMCID: PMC5026207 DOI: 10.1101/lm.043281.116] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/13/2016] [Indexed: 11/24/2022]
Abstract
Traumatic experiences early in life can contribute to the development of mood and anxiety disorders that manifest during adolescence and young adulthood. In young rats exposed to acute fear or stress, alterations in neural development can lead to enduring behavioral abnormalities. Here, we used a modified extinction intervention (retrieval+extinction) during late adolescence (post-natal day 45 [p45]), in rats, to target auditory Pavlovian fear associations acquired as juveniles (p17 and p25). The effects of adolescent intervention were examined by assessing freezing as adults during both fear reacquisition and social transmission of fear from a cagemate. Rats underwent testing or training at three time points across development: juvenile (p17 or p25), adolescent (p45), and adult (p100). Retrieval+extinction during late adolescence prevented social reinstatement and recovery over time of fears initially acquired as juveniles (p17 and p25, respectively). Adolescence was the only time point tested here where retrieval+extinction prevented fear recall of associations acquired 20+ days earlier.
Collapse
Affiliation(s)
- Carolyn E Jones
- Department of Psychology, The University of Texas at Austin, Austin, Texas 78712-1043, USA
| | - Marie-H Monfils
- Department of Psychology, The University of Texas at Austin, Austin, Texas 78712-1043, USA
| |
Collapse
|