DNA methylation and histone acetylation work in concert to regulate memory formation and synaptic plasticity

Similar documents
DNA and Histone Methylation in Learning and Memory

EPIGENETICS: A POSSIBLE MECHANISM OF MEMORY Aliza Grossman Rubenstein

Epigenetic Mechanisms in Cognition

Gene Expression DNA RNA. Protein. Metabolites, stress, environment

Epigenetics, Oestradiol and Hippocampal Memory Consolidation

Cognitive Enhancement Strategies. Florian Plattner, James A. Bibb

Histone Methylation Regulates Memory Formation

Biochemical Determinants Governing Redox Regulated Changes in Gene Expression and Chromatin Structure

Transcriptional and Epigenetic Mechanisms of Addiction

Epigenetic codes in cognition and behaviour

Chromatin-Based Regulation of Gene Expression

Chromatin Remodeling: A Novel Mechanism. of Psychotropic Drug Action

Involvement of DNA methylation in memory processing in the honey bee Gabrielle A. Lockett, Paul Helliwell and Ryszard Maleszka

Epigenetic Regulation of bdnf Gene Transcription in the Consolidation of Fear Memory

Chapter 11: Inherited Disorders of Human Memory Mental Retardation Syndromes. From Mechanisms of Memory, second edition By J. David Sweatt, Ph.D.

Social deficits in Shank3-deficient mouse models of autism are rescued by histone deacetylase (HDAC) inhibition

R. Piazza (MD, PhD), Dept. of Medicine and Surgery, University of Milano-Bicocca EPIGENETICS

Epigenetics. Lyle Armstrong. UJ Taylor & Francis Group. f'ci Garland Science NEW YORK AND LONDON

Epigenetic Regulation of BDNF Gene in Response to Stress

The Role of Smoking in Cocaine. Addiction

MeCP2 and psychostimulantinduced behavioral adaptations. Anne E. West, M.D., Ph.D. Department of Neurobiology Duke University Medical Center

Cellular Neurobiology BIPN140

Histone Deacetylase Inhibitors Enhance Memory and Synaptic Plasticity via CREB: CBP-Dependent Transcriptional Activation

Synaptic Plasticity and NO-cGMP-PKG Signaling Regulate Pre- and Postsynaptic Alterations at Rat Lateral Amygdala Synapses Following Fear Conditioning

mtorc2 controls actin polymerization required for consolidation of long-term memory

Are you the way you are because of the

HDAC1 Inhibitor Screening Assay Kit

Synaptic plasticityhippocampus. Neur 8790 Topics in Neuroscience: Neuroplasticity. Outline. Synaptic plasticity hypothesis

Epigenetic Regulation of Gene Expression in Physiological and Pathological Brain Processes

Epigenetics Armstrong_Prelims.indd 1 04/11/2013 3:28 pm

Role of Atypical Protein Kinases in Maintenance of Long-Term Memory and Synaptic Plasticity

DNA Methylation, Nuclear Structure, Gene Expression and Cancer

BIPN140 Lecture 12: Synaptic Plasticity (II)

Transcriptional control in Eukaryotes: (chapter 13 pp276) Chromatin structure affects gene expression. Chromatin Array of nuc

Neurobiology of Learning and Memory

Eukaryotic transcription (III)

Persistent improvement in synaptic and cognitive functions in an Alzheimer mouse model after rolipram treatment

Synaptic Plasticity During Learning

A form of long-lasting, learning-related synaptic plasticity in the hippocampus induced by heterosynaptic low-frequency pairing

SensoLyte 520 HDAC Activity Assay Kit *Fluorimetric*

Memory retention the synaptic stability versus plasticity dilemma

CASE 49. What type of memory is available for conscious retrieval? Which part of the brain stores semantic (factual) memories?

Subregion-specific p300 conditional knock-out mice exhibit long-term memory impairments

DNA Methylation and Demethylation as Targets for Anticancer Therapy

SUPPLEMENTARY INFORMATION

Chromatin Structure & Gene activity part 2

BIPN 140 Problem Set 6

GESTATIONAL AND LACTATIONAL α-linolenic ACID AVAILABILITY INDUCES EPIGENETIC CHANGES IN THE BRAIN OF MOUSE OFFSPRING

Contrasting Effects of Histone Deacetylase Inhibitors on Reward and Aversive Olfactory Memories in the Honey Bee

Behavioral epigenetics

Jayanti Tokas 1, Puneet Tokas 2, Shailini Jain 3 and Hariom Yadav 3

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION

BIPN 140 Problem Set 6

INTERACTION DRUG BODY

NNZ-2566 in Rett Syndrome and Autism Spectrum Disorders Role and Update

The Neurobiology of Learning and Memory

An Overview of the Molecular Basis of Epigenetics

Fragile X Syndrome. Genetics, Epigenetics & the Role of Unprogrammed Events in the expression of a Phenotype

Developmental Downregulation of Histone Posttranslational Modifications Regulates Visual Cortical Plasticity

Cancer and Gene Regulation

Epigenetics: The Future of Psychology & Neuroscience. Richard E. Brown Psychology Department Dalhousie University Halifax, NS, B3H 4J1

Epigenetic Inheritance

mirna Dr. S Hosseini-Asl

Supplemental information Acid-sensing ion channel 1a contributes to hippocampal LTP inducibility through multiple mechanisms

EPIGENETIC MECHANISMS IN MEMORY FORMATION

Not IN Our Genes - A Different Kind of Inheritance.! Christopher Phiel, Ph.D. University of Colorado Denver Mini-STEM School February 4, 2014

HDAC1 Inhibitor Screening Assay Kit

Transcription and chromatin. General Transcription Factors + Promoter-specific factors + Co-activators

Epigenetic Mechanisms

TREATMENT-SPECIFIC ABNORMAL SYNAPTIC PLASTICITY IN EARLY PARKINSON S DISEASE

Transcription Regulation And Gene Expression in Eukaryotes Cycle G2 (lecture 13709) FS 2014 P Matthias & RG Clerc

EPIGENOMICS PROFILING SERVICES

Neuronal Plasticity, Learning and Memory. David Keays Institute of Molecular Pathology

International Journal of. Peptides. Hindawi Publishing Corporation. Submit your manuscripts at. Anatomy Research International

SUPPLEMENTARY INFORMATION

Changes in DNA methylation and histone modification during epigenetic transitions

Memory Systems II How Stored: Engram and LTP. Reading: BCP Chapter 25

CREB Binding Protein Is Required for Both Short-Term and Long-Term Memory Formation

Sotos syndrome and the NSD1 gene. Jamie Masliah Gene4cs 564

Ch. 18 Regulation of Gene Expression

Bidirectional modifications in synaptic efficacy, exemplified

Molecular Biology of Memory Storage: The Persistence of Memory

EpiQuik Circulating Acetyl Histone H3K18 ELISA Kit (Colorimetric)

Molecular Cell Biology. Prof. D. Karunagaran. Department of Biotechnology. Indian Institute of Technology Madras

Where Splicing Joins Chromatin And Transcription. 9/11/2012 Dario Balestra

Rishabh Kala 1, Harsh N. Shah 1, Samantha L. Martin 1 and Trygve O. Tollefsbol 1,2,3,4,5*

Is Intrinsic Hyperexcitability in CA3 the Culprit for Seizures in Rett Syndrome?

Epigenetic regulation of macrophage polarization and inflammation by DNA methylation in obesity

Behavioral Neuroscience: Fear thou not. Rony Paz

Epigenetics & cancer. Present by : Sanaz Zebardast Under supervision : Dr. Gheibi. 31 December 2016

SUPPLEMENTARY INFORMATION

Feedback Education and Neuroscience. Pankaj Sah

Memory reconsolidation mediates the strengthening of memories by additional learning Lee, Jonathan

Michael Brenner, Ph.D. Professor, Department of Neurobiology Evelyn F. McKnight Brain Institute Phone:

Cellular Mechanisms of Learning and the Biological Basis of Individuality

Histones modifications and variants

Consolidation of CS and US Representations in Associative Fear Conditioning

Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment

Epigenetics: A historical overview Dr. Robin Holliday

Transcription:

Available online at www.sciencedirect.com Neurobiology of Learning and Memory 89 (2008) 599 603 Brief Report DNA methylation and histone acetylation work in concert to regulate memory formation and synaptic plasticity Courtney A. Miller *, Susan L. Campbell, J. David Sweatt Department of Neurobiology, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1074 Shelby Building, 1825 University Boulevard, Birmingham, USA Received 14 June 2007; revised 18 July 2007; accepted 23 July 2007 Available online 18 September 2007 www.elsevier.com/locate/ynlme Abstract A clear understanding is developing concerning the importance of epigenetic-related molecular mechanisms in transcription-dependent long-term memory formation. Chromatin modification, in particular histone acetylation, is associated with transcriptional activation, and acetylation of histone 3 (H3) occurs in Area CA1 of the hippocampus following contextual fear conditioning training. Conversely, DNA methylation is associated with transcriptional repression, but is also dynamically regulated in Area CA1 following training. We recently reported that inhibition of the enzyme responsible for DNA methylation, DNA methyltransferase (DNMT), in the adult rat hippocampus blocks behavioral memory formation. Here, we report that DNMT inhibition also blocks the concomitant memory-associated H3 acetylation, without affecting phosphorylation of its upstream regulator, extracellular signal-regulated kinase (ERK). Interestingly, the DNMT inhibitor-induced deficit in memory consolidation, along with deficits in long-term potentiation, can be rescued by pharmacologically increasing levels of histone acetylation prior to DNMT inhibition. These observations suggest that DNMT activity is not only necessary for memory and plasticity, but that DNA methylation may work in concert with histone modifications to regulate plasticity and memory formation in the adult rat hippocampus. Ó 2007 Elsevier Inc. All rights reserved. Keywords: DNA methylation; Acetylation; Histone; Chromatin; Epigenetics; Hippocampus; Learning; Consolidation; HDAC * Corresponding author. Fax: +1 205 975 5097. E-mail address: cmiller@nrc.uab.edu (C.A. Miller). Both memory consolidation and an in vitro analog, long-term potentiation (LTP), require a cascade of signaling events that include activation of NMDA receptors, protein kinases and transcription factors; events which lead to changes in gene transcription. Recent evidence indicates that regulation of chromatin structure also serves as an additional level of control in this cascade. In particular, memory formation has been shown to be associated with histone acetylation (Alarcon et al., 2004; Guan et al., 2002; Korzus, Rosenfeld, & Mayford, 2004; Levenson et al., 2004; Vecsey et al., 2007; Wood et al., 2005). This process of histone acetylation relaxes chromatin structure, making it more accessible to transcriptional machinery (Lunyak et al., 2002; Turner, 2002; Varga-Weisz & Becker, 1998). Most recently, we have reported that an epigeneticsrelated mechanism, DNA (cytosine-5) methylation, is also important for synaptic plasticity and memory formation in the adult nervous system (Levenson et al., 2006; Miller & Sweatt, 2007). Methylation, a covalent chemical modification of DNA, is catalyzed by DNA (cytosine-5) methyltransferases (DNMTs) and, together with other modifications affecting chromatin structure, DNA methylation serves to regulate gene transcription. DNA methylation has been studied extensively in development, and has long been considered a static process following cell differentiation (Santos, Mazzola, & Carvalho, 2005). However, high DNMT mrna levels persist into adulthood in the brain and we have found that DNMT inhibition alters gene methylation in vitro and in vivo and prevents the induction of LTP and the consolidation of memory (Levenson et al., 2006; Miller & Sweatt, 2007). These observations suggest that DNA methylation is in fact rapidly and dynamically regulated in the adult nervous system. In the current study, 1074-7427/$ - see front matter Ó 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.nlm.2007.07.016

600 C.A. Miller et al. / Neurobiology of Learning and Memory 89 (2008) 599 603 we examined the possibility that DNA methylation influences memory consolidation, in part, by modulating chromatin structure. To confirm our previous finding that DNMT activity is necessary for memory consolidation (Miller & Sweatt, 2007), we gave animals intra-ca1 infusions of the DNMT inhibitor, 5-aza-2-deoxycytidine (5-AZA) immediately after contextual fear conditioning, a hippocampus-dependent task. Infusions were administered post-training to avoid state-dependent effects of the drug. Infusion needle tips in the brains of animals examined were located well within Area CA1 in all cannulated animals (Supplementary Figure S1). Confirming our earlier result, when memory was assessed 24 h later, animals infused with 5-AZA displayed significantly less freezing than their vehicle-treated counterparts (F(1, 15) = 10.21, P <.01; Fig. 1a), indicating that hippocampal DNMT activity is indeed necessary for memory consolidation (Miller & Sweatt, 2007). We and others have also previously reported that acetylation of histone 3 (H3) in Area CA1 is associated with consolidation of contextual fear memory and that memory can be enhanced by pre-treatment with histone deacetylase (HDAC) inhibitors, which pharmacologically raise levels of histone acetylation (Levenson et al., 2004; Vecsey et al., 2007; Wood et al., 2005; Fischer, Sananabenesi, Wang, Dobbin, & Tsai, 2007). In addition, changes in DNA methylation can affect levels of histone acetylation (Becker, Ruppert, & Schutz, 1987; Collins et al., 2004; Cross, Meehan, Nan, & Bird, 1997; Jones et al., 1998; Nan, Campoy, & Bird, 1997; Nan et al., 1998) and we have demonstrated in vitro that DNMT inhibition significantly attenuates PKC-induced H3 acetylation (Levenson et al., 2006). Therefore, we hypothesized that the blockade of memory consolidation by 5-AZA could be partially due to modulation of histone acetylation. To test this, we delivered intra-ca1 infusions of 5-AZA or vehicle immediately after contextual fear conditioning (C/S + 5-AZA or VEH) and assessed levels of acetylated H3 (AcH3) one hour later. Controls were infused (C + 5-AZA or VEH) after exposure to the novel context alone. There were no differences between vehicle and 5-AZA treatment for the context only controls (P >.05), indicating that 5-AZA alone does not have an effect on basal acetylation of H3. Context plus shock vehicle-treated animals showed the expected increase in AcH3 (P <.05 for all comparisons), but context plus shock 5-AZA-infused animals did not (Fig. 1b). This demonstrates that intra-ca1 5-AZA treatment immediately following fear conditioning blocks increased H3 acetylation, which may account in part for the 5-AZA induced memory deficit (Fig. 1a). No differences were observed in H4 acetylation for any of the groups (P >.05 for all comparisons), confirming and extending our previous findings (Levenson et al., 2004). We examined contextual fear conditioning-associated ERK2 phosphorylation as a control for the non-specific effects of 5-AZA. There was no ERK activation in vehicle and 5-AZA treated animals with context only exposure, as Fig. 1. DNMT inhibition blocks memory consolidation and histone acetylation. (a) Intra-CA1 infusion of 5-AZA immediately following contextual fear conditioning blocked consolidation, as evidenced by a lack of freezing at the 24 h test (N = 7, C/S + VEH; N = 9, C/S + 5-AZA). * P <.01. (b) Intra-CA1 infusion of 5-AZA immediately following contextual fear conditioning (C/S + 5-AZA) blocked AcH3 one hr post-training, but had no effect on context only controls (C + 5-AZA; N = 7 per group). * P <.05. (c) Intra-CA1 infusion of 5-AZA had no effect on the ERK phosphorylation induced by contextual fear conditioning. (N = 7 per group) * differs from all others except C/S + 5-AZA, P <.001. # Differs from all others except C/S + VEH, P <.001. Error bars represent SEM.

C.A. Miller et al. / Neurobiology of Learning and Memory 89 (2008) 599 603 601 expected (P >.05). However, context plus shock vehicleand 5-AZA-treated animals both showed an identical elevation in perk2 (P <.05; Fig. 1c). This observation suggests that 5-AZA infusion does not have non-specific effects on a wide variety of cellular and molecular processes upstream of ERK/MAPK activation in the hippocampus. In light of the ability of DNMT inhibition to block memory-associated histone acetylation (Fig. 1b), we next investigated the possibility that the deficits in memory consolidation induced by intra-ca1 infusions of 5-AZA could be blocked by pharmacologically maintaining histone acetylation by inhibiting HDACs. To test this, we gave animals intra-ca1 infusions of the HDAC inhibitor sodium butyrate (NaB). Thirty minutes later, we trained animals for contextual fear conditioning and immediately followed this with an infusion of 5-AZA. Pre-training NaB infusions had no effect on freezing behavior observed during training, suggesting that NaB does not affect an animal s ability to perceive and respond to footshock (P >.05; Fig. 2a). In addition, freezing levels at training were equal across all four groups of animals (vehicle = 46.5% ± 3.2, NaB = 51.8% ± 6.8, 5-AZA = 41.0% ± 4.0, NaB + 5- AZA = 42.6% ± 6.9; F (3,21) = 0.71, P >.05). Twenty-four hours after training, we tested animals for their freezing behavior. Post hoc analyses revealed a confirmation of our earlier results (Fig. 1a), again demonstrating a memory consolidation deficit induced by 5-AZA compared to vehicle-treated context plus shock controls (Fig. 2b, P <.05). Animals treated with NaB alone showed levels of freezing equivalent to vehicle-treated controls (P >.05). This indicates that with this training procedure and dose of NaB, NaB does not affect baseline memory formation. It is worth commenting on the lack of HDAC inhibitor-induced memory enhancement in the current experiment (Fig. 2b), versus what has been previously reported (Levenson et al., 2004; Vecsey et al., 2007; Fischer et al., 2007). The differing result is likely due to two factors: our use of a low concentration and dose of NaB delivered in a site-specific (intra-ca1), rather than systemic fashion, and our use of a 3-shock fear conditioning protocol, rather than the single shock protocol utilized by Levenson et al. (2004), Fischer, Sananabenesi, Wang, Dobbin, and Tsai (2007), and Vecsey et al. (2007). The current experiments were designed to specifically probe for an interaction between DNMTs and histone acetylation at normal levels of learning. Interestingly, animals that were treated with NaB plus 5- AZA displayed freezing equivalent to the vehicle and NaBtreated animals (P <.05 for all comparisons; Fig. 2b). b Fig. 2. HDAC inhibition prevents the memory deficit induced by DNMT inhibition. (a) Pre-training intra-ca1 infusions of NaB do not affect an animal s ability to perceive and respond to footshock during training. The VEH group refers to animals that received pre-training vehicle infusions, but contains animals that then received either vehicle or 5- AZA post-training, after the percent freezing during training measurements were taken. The NaB group refers to animals that received pretraining NaB infusions, but contains animals that received either vehicle or 5-AZA post-training. (b) Intra-CA1 infusion of NaB prior to training blocked the memory consolidation deficit produced by 5-AZA infusion. (N = 7 8 per group) * P <.05. (c) The rescue effect of HDAC inhibition on memory for contextual fear conditioning is long-lasting; present at not only 24 h (Fig. 2A), but also seven days later. * P <.05. Error bars represent SEM.

602 C.A. Miller et al. / Neurobiology of Learning and Memory 89 (2008) 599 603 Fig. 3. HDAC inhibition prevents the LTP deficit induced by DNMT inhibition. (a) LTP was rescued by application of the HDAC inhibitor, TSA, prior to 5-AZA. (N = 10 17 per group; calibration is 1 mv and 5 ms). (b) Input-output synaptic relation is normal in slices pretreated with 5-Aza, TSA or co-application of 5-Aza and TSA relative to their respective vehicles. Error bars represent SEM. These same effects on memory were present when we tested the animals again 1 week later (P <.05; Fig. 2c). This demonstrates that preserved and/or enhanced histone acetylation in Area CA1 during the memory consolidation period immediately after training is sufficient to overcome the memory deficit produced by DNMT inhibition, and is consistent with the hypothesis that regulation of histone acetylation is one mechanism of action of DNA methylation in the adult nervous system. We next investigated the possibility that, as with memory consolidation, h-burst stimulus-induced LTP could be rescued by treating slices with an HDAC inhibitor prior to application of the DNMT inhibitor. For this experiment, we used Trichostatin A (TSA), an HDAC inhibitor that can be used at a lower concentration than NaB in vitro in a dilute aqueous solution. We have previously demonstrated that both TSA and NaB enhance LTP and histone acetylation in acute hippocampal slices (Levenson et al., 2004). h-burst stimulation resulted in the induction of robust LTP (Fig. 3a). Confirming our earlier findings, exposure of slices to 5-AZA resulted in an immediate and significant reduction in LTP (P <.01), while application of TSA resulted in an enhancement of LTP (P <.01) (Levenson et al., 2004; Levenson et al., 2006). Slices that were pre-treated with TSA, followed by 5-AZA 20 min later, displayed LTP that was equivalent to slices treated with vehicle (P >.05; Fig. 3a). We also examined basal synaptic transmission and found no significant difference between slices treated with 5-Aza, TSA or co-application of 5-Aza and TSA compared to their respective vehicles for the input output function (P >.05; Fig. 3b). Together these data demonstrate that the deficit in synaptic plasticity produced by DNMT inhibition can be overcome to a significant extent by enhancing levels of histone acetylation, consistent with what we observed in our behavioral memory studies. DNA methylation is associated with transcriptional repression and, indeed, we have recently reported that inhibition of the DNA methyltransferase enzyme not only blocks memory consolidation, but also results in the aberrant transcription of a memory suppressor gene, protein phosphatase 1 (PP1). In the absence of normal DNMT activity, memory-associated PP1 gene methylation no longer occurs, resulting in its aberrant transcription and interference with memory formation. In the present study, we were able to rescue the DNMT inhibitor-induced memory deficit by elevating histone acetylation levels with HDAC inhibition. Interestingly, Vecsey et al. (2007) have recently reported that HDAC inhibition does not result in global increases in acetylation. Rather, HDAC inhibition increases the acetylation associated with just a subset of genes. Therefore, it seems likely that NaB overcomes the memory deficit produced by 5-AZA by increasing the transcriptional activity of memory promoter genes, such as Nr4a1, whose transcription is upregulated by HDAC inhibition (Vecsey et al., 2007). These genes may then overcome the memory suppressing effects of genes like PP1 that were made aberrantly active by the DNMT inhibition, ultimately resulting in normal memory formation. This is the first study to present evidence suggesting that DNA methylation, once thought to be a static process after cellular differentiation, might dynamically regulate memory formation in the adult nervous system through modulation of chromatin structure. We have confirmed that DNA methyltransferase activity is necessary for memory consolidation in the hippocampus and demonstrated that DNMT inhibition blocks memory-associated histone acetylation. In addition, modifying chromatin structure with an HDAC inhibitor is sufficient to rescue the memory and synaptic plasticity deficits induced by DNA methyltransferase interference. The current findings indicate that DNA methylation and histone acetylation play an impor-

C.A. Miller et al. / Neurobiology of Learning and Memory 89 (2008) 599 603 603 tant and interacting role in memory consolidation and synaptic plasticity. We speculate that future studies of the adult CNS will likely reveal that these two epigeneticsrelated modifications effect memory-associated processes through a complex set of bidirectional interactions. Acknowledgments The authors thank an anonymous reviewer for his or her helpful comments on the manuscript. This work was supported by the NIMH, NINDS, NARSAD, NIH Blueprint for Neuroscience Research, American Health Assistance Foundation and the Evelyn F. McKnight Research Foundation. C.A.M. is a Civitan Emerging Scholar. Appendix A. Supplementary data Supplementary data associated with this article can be found, in the online version, at doi:10.1016/ j.nlm.2007.07.016. References Alarcon, J. M., Malleret, G., Touzani, K., Vronskaya, S., Ishii, S., Kandel, E. R., et al. (2004). Chromatin acetylation, memory, and LTP are impaired in CBP mice: A model for the cognitive deficit in Rubinstein-Taybi syndrome and its amelioration. Neuron, 42, 947 959. Becker, P. B., Ruppert, S., & Schutz, G. (1987). Genomic footprinting reveals cell type-specific DNA binding of ubiquitous factors. Cell, 51, 435 443. Collins, A. L., Levenson, J. M., Vilaythong, A. P., Richman, R., Armstrong, D. L., Noebels, J. L., et al. (2004). Mild overexpression of MeCP2 causes a progressive neurological disorder in mice. Human Molecular Genetics, 13, 2679 2689. Cross, S. H., Meehan, R. R., Nan, X., & Bird, A. (1997). A component of the transcriptional repressor MeCP1 shares a motif with DNA methyltransferase and HRX proteins. Nature Genetics, 16, 256 259. Fischer, A., Sananabenesi, F., Wang, X., Dobbin, M., & Tsai, L. H. (2007). Recovery of learning and memory is associated with chromatin remodelling. Nature, 447, 178 182. Guan, Z., Giustetto, M., Lomvardas, S., Kim, J. H., Miniaci, M. C., Schwartz, J. H., et al. (2002). Integration of long-term-memoryrelated synaptic plasticity involves bidirectional regulation of gene expression and chromatin structure. Cell, 111, 483 493. Jones, P. L., Veenstra, G. J., Wade, P. A., Vermaak, D., Kass, S. U., Landsberger, N., et al. (1998). Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nature Genetics, 19, 187 191. Korzus, E., Rosenfeld, M. G., & Mayford, M. (2004). CBP histone acetyltransferase activity is a critical component of memory consolidation. Neuron, 42, 961 972. Levenson, J. M., O Riordan, K. J., Brown, K. D., Trinh, M. A., Molfese, D. L., et al. (2004). Regulation of histone acetylation during memory formation in the hippocampus. Journal of Biological Chemistry, 279, 40545 40559. Levenson, J. M., Roth, T. L., Lubin, F. D., Miller, C. A., Huang, I., Desai, P., et al. (2006). Evidence that DNA (cytosine-5) methyltransferase regulates synaptic plasticity in the hippocampus. Journal of Biological Chemistry, 281, 15763 15773. Lunyak, V. V., Burgess, R., Prefontaine, G. G., Nelson, C., Sze, S. H., Chenoweth, J., et al. (2002). Corepressor-dependent silencing of chromosomal regions encoding neuronal genes. Science, 298, 1747 1752. Miller, C. A., & Sweatt, J. D. (2007). Covalent modification of DNA regulates memory formation. Neuron, 53, 857 869. Nan, X., Campoy, F. J., & Bird, A. (1997). MeCP2 is a transcriptional repressor with abundant binding sites in genomic chromatin. Cell, 88, 471 481. Nan, X., Ng, H. H., Johnson, C. A., Laherty, C. D., Turner, B. M., Eisenman, R. N., et al. (1998). Transcriptional repression by the methyl-cpg-binding protein MeCP2 involves a histone deacetylase complex. Nature, 393, 386 389. Santos, K. F., Mazzola, T. N., & Carvalho, H. F. (2005). The prima donna of epigenetics: The regulation of gene expression by DNA methylation. Brazilian Journal of Medical and Biological Research, 38, 1531 1541. Turner, B. M. (2002). Cellular memory and the histone code. Cell, 111, 285 291. Varga-Weisz, P. D., & Becker, P. B. (1998). Chromatin-remodeling factors: Machines that regulate? Current Opinion in Cell Biology, 10, 346 353. Vecsey, C. G., Hawk, J. D., Lattal, K. M., Stein, J. M., Fabian, S. A., Attner, M. A., et al. (2007). Histone deacetylase inhibitors enhance memory and synaptic plasticity via CREB: CBP-dependent transcriptional activation. Journal of Neurosience, 27, 6128 6140. Wood, M. A., Kaplan, M. P., Park, A., Blanchard, E. J., Oliveira, A. M., Lombardi, T. L., et al. (2005). Transgenic mice expressing a truncated form of CREB-binding protein (CBP) exhibit deficits in hippocampal synaptic plasticity and memory storage. Learning and Memory, 12, 111 119.