1
|
Bahabry R, Jago SS, Hauser RM, Harmon J, Sheppard LD, Oyassan B, Lubin FD. Hippocampal gene expression changes associated with sequential behavioral training in a temporal lobe epilepsy rat model. Epilepsy Behav Rep 2025; 29:100735. [PMID: 39898299 PMCID: PMC11786087 DOI: 10.1016/j.ebr.2024.100735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 02/04/2025] Open
Abstract
The transcriptional mechanisms underlying impaired hippocampal-dependent memory seen in temporal lobe epilepsy (TLE) have been extensively studied in rodent models. While cognitive testing in these models often involves multiple behavioral tasks, the impact of sequential behavioral testing (SBT) on gene transcription changes in epilepsy remains poorly understood. This study utilized the Kainic Acid (KA) TLE rodent model to examine hippocampal gene expression changes influenced by SBT. Our findings indicate reduced anxiety-related behavior, along with impaired spatial and recognition memory and fear memory in epileptic animals. Quantitative PCR (qPCR) analysis revealed an increase in BDNF, dFosB, Tet2, and Tet3 expression in the epilepsy-SBT group compared to control-SBT, while there was a reduction in Npas4 and Egr4 expression. Immunohistochemistry (IHC) showed that in epileptic animals, performing SBT reversed the loss of 5-hydroxymethylcytosine (5-hmC) in the dorsal hippocampus compared to that seen in home-caged (HC) epileptic animals, and this reversal was neuron-driven. These findings highlight the complex interplay between gene transcription and epigenetic regulation during SBT enrichment in the context of epilepsy.
Collapse
Affiliation(s)
- Rudhab Bahabry
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Silvienne Sint Jago
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rebecca M. Hauser
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jonathan Harmon
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Leah Dinah Sheppard
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bellafaith Oyassan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Farah D. Lubin
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
2
|
Li H, Liuha X, Chen R, Xiao Y, Xu W, Zhou Y, Bai L, Zhang J, Zhao Y, Zhao Y, Wang L, Qin F, Chen Y, Han S, Wei Q, Li S, Zhang D, Bu Q, Wang X, Jiang L, Dai Y, Zhang N, Kuang W, Qin M, Wang H, Tian J, Zhao Y, Cen X. Pyruvate dehydrogenase complex E1 subunit α crotonylation modulates cocaine-associated memory through hippocampal neuron activation. Cell Rep 2024; 43:114529. [PMID: 39046876 DOI: 10.1016/j.celrep.2024.114529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/04/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
Neuronal activation is required for the formation of drug-associated memory, which is critical for the development, persistence, and relapse of drug addiction. Nevertheless, the metabolic mechanisms underlying energy production for neuronal activation remain poorly understood. In the study, a large-scale proteomics analysis of lysine crotonylation (Kcr), a type of protein posttranslational modification (PTM), reveals that cocaine promoted protein Kcr in the hippocampal dorsal dentate gyrus (dDG). We find that Kcr is predominantly discovered in a few enzymes critical for mitochondrial energy metabolism; in particular, pyruvate dehydrogenase (PDH) complex E1 subunit α (PDHA1) is crotonylated at the lysine 39 (K39) residue through P300 catalysis. Crotonylated PDHA1 promotes pyruvate metabolism by activating PDH to increase ATP production, thus providing energy for hippocampal neuronal activation and promoting cocaine-associated memory recall. Our findings identify Kcr of PDHA1 as a PTM that promotes pyruvate metabolism to enhance neuronal activity for cocaine-associated memory.
Collapse
Affiliation(s)
- Hongchun Li
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoyu Liuha
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Rong Chen
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuzhou Xiao
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Xu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Yuanyi Zhou
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Bai
- Histology and Imaging Platform, Core Facilities of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Zhang
- Histology and Imaging Platform, Core Facilities of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Zhao
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Zhao
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Wang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Qin
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaxing Chen
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuang Han
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingfan Wei
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shu Li
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dingwen Zhang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qian Bu
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; West China-Frontier PharmaTech Co., Ltd., Chengdu 610041, China
| | - Xiaojie Wang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linhong Jiang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Dai
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ni Zhang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weihong Kuang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Qin
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongbo Wang
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jingwei Tian
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yinglan Zhao
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaobo Cen
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Leon Kropf V, Albany CJ, Zoccarato A, Green HLH, Yang Y, Brewer AC. TET3 is a positive regulator of mitochondrial respiration in Neuro2A cells. PLoS One 2024; 19:e0294187. [PMID: 38227585 PMCID: PMC10790995 DOI: 10.1371/journal.pone.0294187] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/26/2023] [Indexed: 01/18/2024] Open
Abstract
Ten-Eleven-Translocase (TET) enzymes contribute to the regulation of the methylome via successive oxidation of 5-methyl cytosine (5mC) to derivatives which can be actively removed by base-excision-repair (BER) mechanisms in the absence of cell division. This is particularly important in post-mitotic neurons where changes in DNA methylation are known to associate with changes in neural function. TET3, specifically, is a critical regulator of both neuronal differentiation in development and mediates dynamic changes in the methylome of adult neurons associated with cognitive function. While DNA methylation is understood to regulate transcription, little is known of the specific targets of TET3-dependent catalytic activity in neurons. We report the results of an unbiased transcriptome analysis of the neuroblastoma-derived cell line; Neuro2A, in which Tet3 was silenced. Oxidative phosphorylation (OxPhos) was identified as the most significantly down-regulated functional canonical pathway, and these findings were confirmed by measurements of oxygen consumption rate in the Seahorse bioenergetics analyser. The mRNA levels of both nuclear- and mitochondrial-encoded OxPhos genes were reduced by Tet3-silencing, but we found no evidence for differential (hydroxy)methylation deposition at these gene loci. However, the mRNA expression of genes known to be involved in mitochondrial quality control were also shown to be significantly downregulated in the absence of TET3. One of these genes; EndoG, was identified as a direct target of TET3-catalytic activity at non-CpG methylated sites within its gene body. Accordingly, we propose that aberrant mitochondrial homeostasis may contribute to the decrease in OxPhos, observed upon Tet3-downregulation in Neuro2A cells.
Collapse
Affiliation(s)
- Valeria Leon Kropf
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Caraugh J. Albany
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Anna Zoccarato
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Hannah L. H. Green
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Youwen Yang
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Alison C. Brewer
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London British Heart Foundation Centre of Excellence, London, United Kingdom
| |
Collapse
|
4
|
Milton AL. Drug memory reconsolidation: from molecular mechanisms to the clinical context. Transl Psychiatry 2023; 13:370. [PMID: 38040677 PMCID: PMC10692359 DOI: 10.1038/s41398-023-02666-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023] Open
Abstract
Since its rediscovery at the beginning of the 21st Century, memory reconsolidation has been proposed to be a therapeutic target for reducing the impact of emotional memories that can go awry in mental health disorders such as drug addiction (substance use disorder, SUD). Addiction can be conceptualised as a disorder of learning and memory, in which both pavlovian and instrumental learning systems become hijacked into supporting drug-seeking and drug-taking behaviours. The past two decades of research have characterised the details of the molecular pathways supporting the reconsolidation of pavlovian cue-drug memories, with more recent work indicating that the reconsolidation of instrumental drug-seeking memories also relies upon similar mechanisms. This narrative review considers what is known about the mechanisms underlying the reconsolidation of pavlovian and instrumental memories associated with drug use, how these approaches have translated to experimental medicine studies, and the challenges and opportunities for the clinical use of reconsolidation-based therapies.
Collapse
Affiliation(s)
- Amy L Milton
- Department of Psychology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
5
|
Yin LT, Feng RR, Xie XY, Yang XR, Yang ZF, Hu JJ, Wu SF, Zhang C. Matrix metalloproteinase-9 overexpression in the hippocampus reduces alcohol-induced conditioned-place preference by regulating synaptic plasticity in mice. Behav Brain Res 2023; 442:114330. [PMID: 36746309 DOI: 10.1016/j.bbr.2023.114330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023]
Abstract
Extracellular matrix proteins appear to be necessary for the synaptic plasticity that underlies addiction memory. In the brain, matrix metalloproteinases (MMPs), especially matrix metalloproteinase-9 (MMP-9), have been recently implicated in processes involving alcohol reward and memory. Here, we showed for the first time, the positive effects of MMP-9 on alcohol-induced conditioned place preference (CPP) behavior and hippocampal neuron plasticity in C57BL/6 mice. Using recombinant adeno-associated viruses to overexpress MMP-9 in the hippocampus, we investigated the NMDAR, PSD-95, and cellular cytoskeleton proteins F-actin/G-actin in the modulation of alcohol reward behavior in mice exposed to CPP. We found that hippocampal infusions of MMP-9 decreased alcohol-induced place preference suggesting a reduction in alcohol reward. Western blot analysis demonstrated that protein expression of NMDA receptors (GluN1, GluN2A and GluN2B) in the hippocampus of alcohol-exposed mice were higher than that of the saline group. Further, the expression of these proteins was decreased in MMP-9 overexpressing mice. MMP-9 also regulated the ratio of F-actin/G-actin (dendritic spines cytoskeleton proteins), which might be the key mediator for behavioral changes in mice. Consequently, our results highlight new evidence that MMP-9 may play an important role in the molecular mechanism underlying alcohol reward and preference.
Collapse
Affiliation(s)
- Li-Tian Yin
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| | - Rui-Rui Feng
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiao-Yan Xie
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiao-Rong Yang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhuan-Fang Yang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jia-Jia Hu
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Shu-Fen Wu
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ce Zhang
- Key Laboratory for Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
6
|
Li J, Wu Y, Xue T, He J, Zhang L, Liu Y, Zhao J, Chen Z, Xie M, Xiao B, Ye Y, Qin S, Tang Q, Huang M, Zhu H, Liu N, Guo F, Zhang L, Zhang L. Cdc42 signaling regulated by dopamine D2 receptor correlatively links specific brain regions of hippocampus to cocaine addiction. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166569. [PMID: 36243293 DOI: 10.1016/j.bbadis.2022.166569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Hippocampus plays critical roles in drug addiction. Cocaine-induced modifications in dopamine receptor function and the downstream signaling are important regulation mechanisms in cocaine addiction. Rac regulates actin filament accumulation while Cdc42 stimulates the formation of filopodia and neurite outgrowth. Based on the region specific roles of small GTPases in brain, we focused on the hippocampal subregions to detect the regulation of Cdc42 signaling in long-term morphological and behavioral adaptations to cocaine. METHODS Genetically modified mouse models of Cdc42, dopamine receptor D1 (D1R) and D2 (D2R) and expressed Cdc42 point mutants that are defective in binding to and activation of its downstream effector molecules PAK and N-WASP were generated, respectively, in CA1 or dentate gyrus (DG) subregion. RESULTS Cocaine induced upregulation of Cdc42 signaling activity. Cdc42 knockout or mutants blocked cocaine-induced increase in spine plasticity in hippocampal CA1 pyramidal neurons, leading to a decreased conditional place preference (CPP)-associated memories and spatial learning and memory in water maze. Cdc42 knockout or mutants promoted cocaine-induced loss of neurogenesis in DG, leading to a decreased CPP-associated memories and spatial learning and memory in water maze. Furthermore, by using D1R knockout, D2R knockout, and D2R/Cdc42 double knockout mice, we found that D2R, but not D1R, regulated Cdc42 signaling in cocaine-induced neural plasticity and behavioral changes. CONCLUSIONS Cdc42 acts downstream of D2R in the hippocampus and plays an important role in cocaine-induced neural plasticity through N-WASP and PAK-LIMK-Cofilin, and Cdc42 signaling pathway correlatively links specific brain regions (CA1, dentate gyrus) to cocaine-induced CPP behavior.
Collapse
Affiliation(s)
- Juan Li
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, School of Basic Medical Sciences, Center for Orthopaedic Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue Wu
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tao Xue
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jing He
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lei Zhang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yutong Liu
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, School of Basic Medical Sciences, Center for Orthopaedic Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jinlan Zhao
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenzhong Chen
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Minjuan Xie
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Xiao
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yingshan Ye
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Sifei Qin
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qingqiu Tang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Mengfan Huang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hangfei Zhu
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - N Liu
- Institute of Comparative Medicine & Laboratory Animal Center, Elderly Health Services Research Center, Southern Medical University, Guangzhou 510515, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, Cincinnati, OH, USA
| | - Lin Zhang
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province, School of Basic Medical Sciences, Center for Orthopaedic Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
7
|
Pratt KJB, Shea JM, Remesal-Gomez L, Bieri G, Smith LK, Couthouis J, Chen CP, Roy IJ, Gontier G, Villeda SA. Loss of neuronal Tet2 enhances hippocampal-dependent cognitive function. Cell Rep 2022; 41:111612. [PMID: 36351399 PMCID: PMC10032941 DOI: 10.1016/j.celrep.2022.111612] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/04/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022] Open
Abstract
DNA methylation has emerged as a critical modulator of neuronal plasticity and cognitive function. Notwithstanding, the role of enzymes that demethylate DNA remain to be fully explored. Here, we report that loss of ten-eleven translocation methylcytosine dioxygenase 2 (Tet2), which catalyzes oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), in adult neurons enhances cognitive function. In the adult mouse hippocampus, we detected an enrichment of Tet2 in neurons. Viral-mediated neuronal overexpression and RNA interference of Tet2 altered dendritic complexity and synaptic-plasticity-related gene expression in vitro. Overexpression of neuronal Tet2 in adult hippocampus, and loss of Tet2 in adult glutamatergic neurons, resulted in differential hydroxymethylation associated with genes involved in synaptic transmission. Functionally, overexpression of neuronal Tet2 impaired hippocampal-dependent memory, while loss of neuronal Tet2 enhanced memory. Ultimately, these data identify neuronal Tet2 as a molecular target to boost cognitive function.
Collapse
Affiliation(s)
- Karishma J B Pratt
- Department of Anatomy, University of California San Francisco, 513 Parnassus Avenue, Box 0452, San Francisco, CA 94143, USA; Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jeremy M Shea
- Department of Anatomy, University of California San Francisco, 513 Parnassus Avenue, Box 0452, San Francisco, CA 94143, USA
| | - Laura Remesal-Gomez
- Department of Anatomy, University of California San Francisco, 513 Parnassus Avenue, Box 0452, San Francisco, CA 94143, USA
| | - Gregor Bieri
- Department of Anatomy, University of California San Francisco, 513 Parnassus Avenue, Box 0452, San Francisco, CA 94143, USA
| | - Lucas K Smith
- Department of Anatomy, University of California San Francisco, 513 Parnassus Avenue, Box 0452, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christopher P Chen
- Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Irena J Roy
- Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Geraldine Gontier
- Department of Anatomy, University of California San Francisco, 513 Parnassus Avenue, Box 0452, San Francisco, CA 94143, USA.
| | - Saul A Villeda
- Department of Anatomy, University of California San Francisco, 513 Parnassus Avenue, Box 0452, San Francisco, CA 94143, USA; Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA; Department of Physical Therapy and Rehabilitation Science, San Francisco, CA 94143, USA; Bakar Aging Research Institute, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
Anier K, Somelar K, Jaako K, Alttoa M, Sikk K, Kokassaar R, Kisand K, Kalda A. Psychostimulant-induced aberrant DNA methylation in an in vitro model of human peripheral blood mononuclear cells. Clin Epigenetics 2022; 14:89. [PMID: 35842682 PMCID: PMC9288712 DOI: 10.1186/s13148-022-01303-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/30/2022] [Indexed: 11/14/2022] Open
Abstract
Background Several reports have provided crucial evidence in animal models that epigenetic modifications, such as DNA methylation, may be involved in psychostimulant-induced stable changes at the cellular level in the brain. Epigenetic editors DNA methyltransferases (DNMTs) and ten-eleven translocation enzymes (TETs) coordinate expression of gene networks, which then manifest as long-term behavioural changes. However, the extent to which aberrant DNA methylation is involved in the mechanisms of substance use disorder in humans is unclear. We previously demonstrated that cocaine modifies gene transcription, via DNA methylation, throughout the brain and in peripheral blood cells in mice. Results We treated human peripheral blood mononuclear cells (PBMCs) from healthy male donors (n = 18) in vitro with psychostimulants (amphetamine, cocaine). After treatment, we assessed mRNA levels and enzymatic activities of TETs and DNMTs, conducted genome-wide DNA methylation assays and next-generation sequencing. We found that repeated exposure to psychostimulants decreased mRNA levels and enzymatic activity of TETs and 5-hydroxymethylation levels in PBMCs. These data were in line with observed hyper- and hypomethylation and mRNA expression of marker genes (IL-10, ATP2B4). Additionally, we evaluated whether the effects of cocaine on epigenetic editors (DNMTs and TETs) and cytokines interleukin-6 (IL-6) and IL-10 could be reversed by the DNMT inhibitor decitabine. Indeed, decitabine eliminated cocaine’s effect on the activity of TETs and DNMTs and decreased cytokine levels, whereas cocaine increased IL-6 and decreased IL-10. Conclusions Our data suggest that repeated psychostimulant exposure decreases TETs’ enzymatic activity in PBMCs. Co-treatment with decitabine reversed TETs’ levels and modulated immune response after repeated cocaine exposure. Further investigation is needed to clarify if TET could represent a putative biomarker of psychostimulant use and if DNMT inhibition could have therapeutic potential. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01303-w.
Collapse
Affiliation(s)
- Kaili Anier
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Kelli Somelar
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia.
| | - Külli Jaako
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Margret Alttoa
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Kerli Sikk
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Raul Kokassaar
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Kai Kisand
- Department of Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Anti Kalda
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| |
Collapse
|
9
|
Qi S, Tan SM, Wang R, Higginbotham JA, Ritchie JL, Ibarra CK, Arguello AA, Christian RJ, Fuchs RA. Optogenetic inhibition of the dorsal hippocampus CA3 region during early-stage cocaine-memory reconsolidation disrupts subsequent context-induced cocaine seeking in rats. Neuropsychopharmacology 2022; 47:1473-1483. [PMID: 35581381 PMCID: PMC9205994 DOI: 10.1038/s41386-022-01342-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/11/2022] [Accepted: 05/07/2022] [Indexed: 11/09/2022]
Abstract
The dorsal hippocampus (DH) is key to the maintenance of cocaine memories through reconsolidation into long-term memory stores after retrieval-induced memory destabilization. Here, we examined the time-dependent role of the cornu ammonis 3 DH subregion (dCA3) in cocaine-memory reconsolidation by utilizing the temporal and spatial specificity of optogenetics. eNpHR3.0-eYFP- or eYFP-expressing male Sprague-Dawley rats were trained to lever press for cocaine infusions in a distinct context and received extinction training in a different context. Rats were then re-exposed to the cocaine-paired context for 15 min to destabilize cocaine memories (memory reactivation) or remained in their home cages (no-reactivation). Optogenetic dCA3 inhibition for one hour immediately after memory reactivation reduced c-Fos expression (index of neuronal activation) in dCA3 stratum pyramidale (SP) glutamatergic and GABAergic neurons and in stratum lucidum (SL) GABAergic neurons during reconsolidation. Furthermore, dCA3 inhibition attenuated drug-seeking behavior (non-reinforced lever presses) selectively in the cocaine-paired context three days later (recall test), relative to no photoinhibition. This behavioral effect was eNpHR3.0-, memory-reactivation, and time-dependent, indicating a memory-reconsolidation deficit. Based on this observation and our previous finding that protein synthesis in the DH is not necessary for cocaine-memory reconsolidation, we postulate that recurrent pyramidal neuronal activity in the dCA3 may maintain labile cocaine memories prior to protein synthesis-dependent reconsolidation elsewhere, and SL/SP interneurons may facilitate this process by limiting extraneous neuronal activity. Interestingly, SL c-Fos expression was reduced at recall concomitant with impairment in cocaine-seeking behavior, suggesting that SL neurons may also facilitate cocaine-memory retrieval by inhibiting non-engram neuronal activity.
Collapse
Affiliation(s)
- Shuyi Qi
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA
| | - Shi Min Tan
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA
| | - Rong Wang
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA
| | - Jessica A Higginbotham
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA
| | - Jobe L Ritchie
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA
| | - Christopher K Ibarra
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA
| | - Amy A Arguello
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA
| | - Robert J Christian
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA
| | - Rita A Fuchs
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, Washington, USA.
- Alcohol and Drug Abuse Research Program, Washington State University, Pullman, Washington, USA.
| |
Collapse
|
10
|
Shrestha P, Klann E. Spatiotemporally resolved protein synthesis as a molecular framework for memory consolidation. Trends Neurosci 2022; 45:297-311. [PMID: 35184897 PMCID: PMC8930706 DOI: 10.1016/j.tins.2022.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 01/25/2023]
Abstract
De novo protein synthesis is required for long-term memory consolidation. Dynamic regulation of protein synthesis occurs via a complex interplay of translation factors and modulators. Many components of the protein synthesis machinery have been targeted either pharmacologically or genetically to establish its requirement for memory. The combination of ligand/light-gating and genetic strategies, that is, chemogenetics and optogenetics, has begun to reveal the spatiotemporal resolution of protein synthesis in specific cell types during memory consolidation. This review summarizes current knowledge of the macroscopic and microscopic neural substrates for protein synthesis in memory consolidation. In addition, we highlight future directions for determining the localization and timing of de novo protein synthesis for memory consolidation with tools that permit unprecedented spatiotemporal precision.
Collapse
Affiliation(s)
- Prerana Shrestha
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA.
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10012, USA; NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
11
|
Kaplan G, Xu H, Abreu K, Feng J. DNA Epigenetics in Addiction Susceptibility. Front Genet 2022; 13:806685. [PMID: 35145550 PMCID: PMC8821887 DOI: 10.3389/fgene.2022.806685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/06/2022] [Indexed: 12/22/2022] Open
Abstract
Addiction is a chronically relapsing neuropsychiatric disease that occurs in some, but not all, individuals who use substances of abuse. Relatively little is known about the mechanisms which contribute to individual differences in susceptibility to addiction. Neural gene expression regulation underlies the pathogenesis of addiction, which is mediated by epigenetic mechanisms, such as DNA modifications. A growing body of work has demonstrated distinct DNA epigenetic signatures in brain reward regions that may be associated with addiction susceptibility. Furthermore, factors that influence addiction susceptibility are also known to have a DNA epigenetic basis. In the present review, we discuss the notion that addiction susceptibility has an underlying DNA epigenetic basis. We focus on major phenotypes of addiction susceptibility and review evidence of cell type-specific, time dependent, and sex biased effects of drug use. We highlight the role of DNA epigenetics in these diverse processes and propose its contribution to addiction susceptibility differences. Given the prevalence and lack of effective treatments for addiction, elucidating the DNA epigenetic mechanism of addiction vulnerability may represent an expeditious approach to relieving the addiction disease burden.
Collapse
|
12
|
Chen L, Yan H, Wang Y, He Z, Leng Q, Huang S, Wu F, Feng X, Yan J. The Mechanisms and Boundary Conditions of Drug Memory Reconsolidation. Front Neurosci 2021; 15:717956. [PMID: 34421529 PMCID: PMC8377231 DOI: 10.3389/fnins.2021.717956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Drug addiction can be seen as a disorder of maladaptive learning characterized by relapse. Therefore, disrupting drug-related memories could be an approach to improving therapies for addiction. Pioneering studies over the last two decades have revealed that consolidated memories are not static, but can be reconsolidated after retrieval, thereby providing candidate pathways for the treatment of addiction. The limbic-corticostriatal system is known to play a vital role in encoding the drug memory engram. Specific structures within this system contribute differently to the process of memory reconsolidation, making it a potential target for preventing relapse. In addition, as molecular processes are also active during memory reconsolidation, amnestic agents can be used to attenuate drug memory. In this review, we focus primarily on the brain structures involved in storing the drug memory engram, as well as the molecular processes involved in drug memory reconsolidation. Notably, we describe reports regarding boundary conditions constraining the therapeutic potential of memory reconsolidation. Furthermore, we discuss the principles that could be employed to modify stored memories. Finally, we emphasize the challenge of reconsolidation-based strategies, but end with an optimistic view on the development of reconsolidation theory for drug relapse prevention.
Collapse
Affiliation(s)
- Liangpei Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - He Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Yufang Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Ziping He
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Qihao Leng
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shihao Huang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Feilong Wu
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiangyang Feng
- Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China.,Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
13
|
4R Tau Modulates Cocaine-Associated Memory through Adult Dorsal Hippocampal Neurogenesis. J Neurosci 2021; 41:6753-6774. [PMID: 34099513 DOI: 10.1523/jneurosci.2848-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
The development, persistence and relapse of drug addiction require drug memory that generally develops with drug administration-paired contextual stimuli. Adult hippocampal neurogenesis (AHN) contributes to cocaine memory formation; however, the underlying mechanism remains unclear. Male mice hippocampal expression of Tau was significantly decreased during the cocaine-associated memory formation. Genetic overexpression of four microtubule-binding repeats Tau (4R Tau) in the mice hippocampus disrupted cocaine memory by suppressing AHN. Furthermore, 4R Tau directly interacted with phosphoinositide 3-kinase (PI3K)-p85 and impaired its nuclear translocation and PI3K-AKT signaling, processes required for hippocampal neuron proliferation. Collectively, 4R Tau modulates cocaine memory formation by disrupting AHN, suggesting a novel mechanism underlying cocaine memory formation and provide a new strategy for the treatment of cocaine addiction.SIGNIFICANCE STATEMENT Drug memory that generally develops with drug-paired contextual stimuli and drug administration is critical for the development, persistence and relapse of drug addiction. Previous studies have suggested that adult hippocampal neurogenesis (AHN) plays a role in cocaine memory formation. Here, we showed that Tau was significantly downregulated in the hippocampus in the cocaine memory formation. Tau knock-out (KO) promoted AHN in the hippocampal dentate gyrus (DG), resulting in the enhanced memory formation evoked by cocaine-cue stimuli. In contrast, genetically overexpressed 4R Tau in the hippocampus disrupted cocaine-cue memory by suppressing AHN. In addition, 4R Tau interacted directly with phosphoinositide 3-kinase (PI3K)-p85 and hindered its nuclear translocation, eventually repressing PI3K-AKT signaling, which is essential for hippocampal neuronal proliferation.
Collapse
|
14
|
Jiang F, Zheng W, Wu C, Li Y, Shen F, Liang J, Li M, Zhang J, Sui N. Double dissociation of inhibitory effects between the hippocampal TET1 and TET3 in the acquisition of morphine self-administration in rats. Addict Biol 2021; 26:e12875. [PMID: 32031744 DOI: 10.1111/adb.12875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 12/02/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
The development of opioid addiction involves DNA methylation. Accordingly, the DNA demethylation, induced by ten-eleven translocation (Tet) enzymes, may represent a novel approach to prevent opioid addiction. The present study examined the role of TET1 and TET3 in the development of morphine-seeking behavior in rats. We showed that 1 day of morphine self-administration (SA) training upregulated TET3 but not TET1 expression in the hippocampal CA1. With 7 days of morphine SA training, the expression of TET3 in the CA1 returned to the baseline level, while the TET1 expression was downregulated. No change of TET1 and TET3 in the nucleus accumbens shell was observed in morphine SA trained rats, or in the yoked morphine rats, or in rats trained for saccharin SA. Furthermore, we found that knocking down TET3 expression in the CA1 accelerated the acquisition of morphine SA, while overexpression of the catalytic domain of TET1 in the CA1 attenuated the acquisition. Together, these findings suggest that TET1 and TET3 in the CA1 are important epigenetic modulators involved in the morphine-seeking behavior and provide a new strategy in the treatment of opioid addiction.
Collapse
Affiliation(s)
- Feng‐Ze Jiang
- CAS Key Laboratory of Mental Health, Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Wei Zheng
- CAS Key Laboratory of Mental Health, Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Chao Wu
- CAS Key Laboratory of Mental Health, Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Yonghui Li
- CAS Key Laboratory of Mental Health, Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Fang Shen
- CAS Key Laboratory of Mental Health, Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Jing Liang
- CAS Key Laboratory of Mental Health, Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Ming Li
- Department of Psychology University of Nebraska—Lincoln Lincoln Nebraska USA
| | - Jian‐Jun Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| | - Nan Sui
- CAS Key Laboratory of Mental Health, Institute of Psychology Beijing China
- Department of Psychology University of Chinese Academy of Sciences Beijing China
| |
Collapse
|
15
|
Bender BN, Torregrossa MM. Molecular and circuit mechanisms regulating cocaine memory. Cell Mol Life Sci 2020; 77:3745-3768. [PMID: 32172301 PMCID: PMC7492456 DOI: 10.1007/s00018-020-03498-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/04/2020] [Accepted: 03/02/2020] [Indexed: 01/27/2023]
Abstract
Risk of relapse is a major challenge in the treatment of substance use disorders. Several types of learning and memory mechanisms are involved in substance use and have implications for relapse. Associative memories form between the effects of drugs and the surrounding environmental stimuli, and exposure to these stimuli during abstinence causes stress and triggers drug craving, which can lead to relapse. Understanding the neural underpinnings of how these associations are formed and maintained will inform future advances in treatment practices. A large body of research has expanded our knowledge of how associative memories are acquired and consolidated, how they are updated through reactivation and reconsolidation, and how competing extinction memories are formed. This review will focus on the vast literature examining the mechanisms of cocaine Pavlovian associative memories with an emphasis on the molecular memory mechanisms and circuits involved in the consolidation, reconsolidation, and extinction of these memories. Additional research elucidating the specific signaling pathways, mechanisms of synaptic plasticity, and epigenetic regulation of gene expression in the circuits involved in associative learning will reveal more distinctions between consolidation, reconsolidation, and extinction learning that can be applied to the treatment of substance use disorders.
Collapse
Affiliation(s)
- Brooke N Bender
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
16
|
Creighton SD, Stefanelli G, Reda A, Zovkic IB. Epigenetic Mechanisms of Learning and Memory: Implications for Aging. Int J Mol Sci 2020; 21:E6918. [PMID: 32967185 PMCID: PMC7554829 DOI: 10.3390/ijms21186918] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
The neuronal epigenome is highly sensitive to external events and its function is vital for producing stable behavioral outcomes, such as the formation of long-lasting memories. The importance of epigenetic regulation in memory is now well established and growing evidence points to altered epigenome function in the aging brain as a contributing factor to age-related memory decline. In this review, we first summarize the typical role of epigenetic factors in memory processing in a healthy young brain, then discuss the aspects of this system that are altered with aging. There is general agreement that many epigenetic marks are modified with aging, but there are still substantial inconsistencies in the precise nature of these changes and their link with memory decline. Here, we discuss the potential source of age-related changes in the epigenome and their implications for therapeutic intervention in age-related cognitive decline.
Collapse
Affiliation(s)
- Samantha D. Creighton
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
| | - Gilda Stefanelli
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
| | - Anas Reda
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S, Canada;
| | - Iva B. Zovkic
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; (S.D.C.); (G.S.)
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S, Canada;
| |
Collapse
|
17
|
Taujanskaitė U, Cahill EN, Milton AL. Targeting drug memory reconsolidation: a neural analysis. Curr Opin Pharmacol 2020; 56:7-12. [PMID: 32961367 DOI: 10.1016/j.coph.2020.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 10/23/2022]
Abstract
Addiction can be conceptualised as a disorder of maladaptive learning and memory. Therefore, maladaptive drug memories supporting drug-seeking and relapse behaviours may present novel treatment targets for therapeutic approaches based upon reconsolidation-blockade. It is known that different structures within the limbic corticostriatal system contribute differentially to different types of maladaptive drug memories, including pavlovian associations between environmental cues and contexts with the drug high, and instrumental memories underlying drug-seeking. Here, we review the mechanisms underlying drug memory reconsolidation in the amygdala, striatum, and hippocampus, noting similarities and differences, and opportunities for future research.
Collapse
Affiliation(s)
- Uršulė Taujanskaitė
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Emma N Cahill
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Amy L Milton
- Department of Psychology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
18
|
Li X, Marshall PR, Leighton LJ, Zajaczkowski EL, Wang Z, Madugalle SU, Yin J, Bredy TW, Wei W. The DNA Repair-Associated Protein Gadd45γ Regulates the Temporal Coding of Immediate Early Gene Expression within the Prelimbic Prefrontal Cortex and Is Required for the Consolidation of Associative Fear Memory. J Neurosci 2019; 39:970-983. [PMID: 30545945 PMCID: PMC6363930 DOI: 10.1523/jneurosci.2024-18.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/11/2018] [Accepted: 12/04/2018] [Indexed: 02/04/2023] Open
Abstract
We have identified a member of the growth arrest and DNA damage (Gadd45) protein family, Gadd45γ, which is known to be critically involved in DNA repair, as a key player in the regulation of immediate early gene (IEG) expression underlying the consolidation of associative fear memory in adult male C57BL/6 mice. Gadd45γ temporally influences learning-induced IEG expression in the prelimbic prefrontal cortex (PLPFC) through its interaction with DNA double-strand break (DSB)-mediated changes in DNA methylation. Our findings suggest a two-hit model of experience-dependent IEG activity and learning that comprises (1) a first wave of IEG expression governed by DSBs and followed by a rapid increase in DNA methylation, and (2) a second wave of IEG expression associated with the recruitment of Gadd45γ and active DNA demethylation at the same site, which is necessary for memory consolidation.SIGNIFICANCE STATEMENT How does the pattern of immediate early gene transcription in the brain relate to the storage and accession of information, and what controls these patterns? This paper explores how Gadd45γ, a gene that is known to be involved with DNA modification and repair, regulates the temporal coding of IEGs underlying associative learning and memory. We reveal that, during fear learning, Gadd45γ serves to act as a coordinator of IEG expression and subsequent memory consolidation by directing temporally specific changes in active DNA demethylation at the promoter of plasticity-related IEGs.
Collapse
Affiliation(s)
- Xiang Li
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Paul R Marshall
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Laura J Leighton
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Esmi L Zajaczkowski
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Ziqi Wang
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Sachithrani U Madugalle
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Jiayu Yin
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Timothy W Bredy
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Wei Wei
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
19
|
Marshall PR, Bredy TW. Neuroepigenetic mechanisms underlying fear extinction: emerging concepts. Psychopharmacology (Berl) 2019; 236:133-142. [PMID: 30506235 PMCID: PMC7293886 DOI: 10.1007/s00213-018-5084-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/16/2018] [Indexed: 12/11/2022]
Abstract
An understanding of how memory is acquired and how it can be modified in fear-related anxiety disorders, with the enhancement of failing memories on one side and a reduction or elimination of traumatic memories on the other, is a key unmet challenge in the fields of neuroscience and neuropsychiatry. The latter process depends on an important form of learning called fear extinction, where a previously acquired fear-related memory is decoupled from its ability to control behaviour through repeated non-reinforced exposure to the original fear-inducing cue. Although simple in description, fear extinction relies on a complex pattern of brain region and cell-type specific processes, some of which are unique to this form of learning and, for better or worse, contribute to the inherent instability of fear extinction memory. Here, we explore an emerging layer of biology that may compliment and enrich the synapse-centric perspective of fear extinction. As opposed to the more classically defined role of protein synthesis in the formation of fear extinction memory, a neuroepigenetic view of the experience-dependent gene expression involves an appreciation of dynamic changes in the state of the entire cell: from a transient change in plasticity at the level of the synapse, to potentially more persistent long-term effects within the nucleus. A deeper understanding of neuroepigenetic mechanisms and how they influence the formation and maintenance of fear extinction memory has the potential to enable the development of more effective treatment approaches for fear-related neuropsychiatric conditions.
Collapse
Affiliation(s)
- Paul R Marshall
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Timothy W Bredy
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
20
|
Zhang YX, Akumuo RC, España RA, Yan CX, Gao WJ, Li YC. The histone demethylase KDM6B in the medial prefrontal cortex epigenetically regulates cocaine reward memory. Neuropharmacology 2018; 141:113-125. [PMID: 30165076 PMCID: PMC6170674 DOI: 10.1016/j.neuropharm.2018.08.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/23/2018] [Accepted: 08/24/2018] [Indexed: 02/07/2023]
Abstract
Epigenetic remodeling contributes to synaptic plasticity via modification of gene expression, which underlies cocaine-induced long-term memory. A prevailing hypothesis in drug addiction is that drugs of abuse rejuvenate developmental machinery to render reward circuitry highly plastic and thus engender drug memories to be highly stable. Identification and reversal of these pathological pathways are therefore critical for cocaine abuse treatment. Previous studies revealed an interesting finding in which the mRNA of histone lysine demethylase, KDM6B, is upregulated in the medial prefrontal cortex (mPFC) during early cocaine withdrawal. However, whether and how it contributes to drug-seeking behavior remain unknown. Here we used a conditioned place preference paradigm to investigate the potential role of KDM6B in drug-associated memory. We found that KDM6B protein levels selectively increased in the mPFC during cocaine withdrawal. Notably, systemic injection of KDM6B inhibitor, GSK-J4, disrupted both reconsolidation of cocaine-conditioned memory and cocaine-primed reinstatement, suggesting dual effects of KDM6B in cocaine reward memory. In addition, we found that NMDAR expression and function were both enhanced during early cocaine withdrawal in mPFC. Injection of GSK-J4 selectively reversed this cocaine-induced increase of NR2A expression and synaptic function, suggesting that mal-adaptation of cocaine-induced synaptic plasticity in mPFC largely underlies KDM6B-mediated cocaine-associated memory. Altogether, these data suggest that KDM6B plays an essential role in cocaine-associated memory, which mainly acts through enhancing cocaine-induced synaptic plasticity in the mPFC. Our findings revealed a novel role of KDM6B in cocaine-associated memory and inhibition of KDM6B is a potential strategy to alleviate drug-seeking behavior.
Collapse
Affiliation(s)
- Yu-Xiang Zhang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Rita C Akumuo
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Rodrigo A España
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Chun-Xia Yan
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| | - Yan-Chun Li
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
21
|
Ploense KL, Li X, Baker-Andresen D, Carr AE, Woodward N, Bagley J, Szumlinski KK, Bredy TW, Kippin TE. Prolonged-access to cocaine induces distinct Homer2 DNA methylation, hydroxymethylation, and transcriptional profiles in the dorsomedial prefrontal cortex of Male Sprague-Dawley rats. Neuropharmacology 2018; 143:299-305. [PMID: 30268522 DOI: 10.1016/j.neuropharm.2018.09.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/16/2022]
Abstract
Repeated cocaine administration induces many long-term structural and molecular changes in the dorsal medial prefrontal cortex (dmPFC) and are known to underlie aspects of cocaine-seeking behavior. DNA methylation is a key long-lasting epigenetic determinant of gene expression and is implicated in neuroplasticity, however, the extent to which this epigenetic modification is involved in the neuroplasticity associated with drug addiction has received limited attention. Here, we examine the relation between DNA methylation and gene expression within the dorsal medial prefrontal cortex (dmPFC) following limited cocaine self-administration (1 h/day), prolonged cocaine self-administration (6 h/day), and saline self-administration (1 h/day). Rats were fitted with intravenous catheters and allowed to lever press for saline or cocaine (0.25 mg/kg/0.1 mL infusion) in the different access conditions for 20 days. Prolonged-access rats exhibited escalation in cocaine intake over the course of training, while limited-access rats did not escalate cocaine intake. Additionally, limited-access and prolonged-access rats exhibited unique Homer2 epigenetic profiles and mRNA expression. In prolonged-access rats, Homer2 mRNA levels in the dmPFC were increased, which was accompanied by decreased DNA methylation and p300 binding within the Homer2 promoter. Limited-access animals exhibited decreased DNA methylation, decreased DNA hydroxymethylation, and increased p300 binding within the Homer2 promoter. These data indicate that distinct epigenetic profiles are induced by limited-versus prolonged-access self-administration conditions that contribute to transcriptional profiles and lend support to the notion that covalent modification of DNA is implicated in addiction-like changes in cocaine-seeking behavior.
Collapse
Affiliation(s)
- Kyle L Ploense
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA; Institute for Collaborative Biotechnology, University of California, Santa Barbara, CA, USA; Department of Chemistry & Biochemistry, University of California, Santa Barbara, CA, USA.
| | - Xiang Li
- Queensland Brain Institute, University of Queensland, St Lucia, QLD, Australia
| | | | - Amanda E Carr
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA
| | - Nick Woodward
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA
| | - Jared Bagley
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA; Department of Psychology, Binghamton University, Binghamton, NY, USA
| | - Karen K Szumlinski
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA; Neuroscience Research Institute, University of California, Santa Barbara, CA, USA; Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Timothy W Bredy
- Queensland Brain Institute, University of Queensland, St Lucia, QLD, Australia
| | - Tod E Kippin
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA; Neuroscience Research Institute, University of California, Santa Barbara, CA, USA; Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, CA, USA; Institute for Collaborative Biotechnology, University of California, Santa Barbara, CA, USA
| |
Collapse
|