Memory and neuromodulation: A perspective of DNA methylation

https://doi.org/10.1016/j.neubiorev.2019.12.022Get rights and content

Highlights

  • Neuromodulation enhances cognition through altered methylation profile.

  • DNMTs, MeCP2 and DNA demethylases are major components underlying neuromodulation effects.

  • Neuroepigentic studies are warranted to characterize diverse effects of neuromodulation on memory.

Abstract

Neuromodulation techniques have shown promising efficacy on memory function and understanding the epigenetic mechanisms contributing to these processes would shed light on the molecular outcomes essential for cognition. In this review, we highlight some epigenetic mechanisms underlying neuromodulation and regulatory effects of neuronal activity-induced DNA methylation on genes that are highly involved in memory formation. Next, we examine the evidence to support DNA methyltransferase 3a, methyl-CpG binding protein 2, and DNA demethylase as possible memory modulation targets. Finally, we report the recent developments in the field of neuromodulation and explore the potential of these techniques for future neuroepigenetic research.

Introduction

In recent years, neuroscientists have made significant progress in elucidating how memory is formed in the complex neural network. One of the current hypotheses of memory formation suggests that genes are divided into memory-suppressing and memory-enhancing genes. For memory consolidation to occur, memory-suppressing genes are transcriptionally inactivated, whereas memory-enhancing genes are expressed, thus tilting the balance towards the expression of memory-enhancing proteins (Abel and Kandel, 1998). Memory formation starts with a transient external stimulus, which then becomes sustained over a relatively long period of time, even throughout one’s lifetime. The stimulus must be able to induce certain molecular and cellular responses robustly and persistently to maintain these effects. One of the many hypotheses in memory research suggest that the memory relies on the stimulus resulting in perpetual gene transcription and protein expression, as the major effectors of the cellular responses. After Bliss and Lomo’s discovery in 1973 of long-term potentiation (LTP) that explained how synaptic transmission mediated memory formation, researchers have been trying to answer the question of how memories propagate (Bliss and Lomo, 1973). Eric Kandel and his team, who are pioneers in the field, discovered that during the late phase of LTP (L-LTP), which is recognized as the prerequisite of long-term memory formation, gene transcription becomes necessary, as shown by the subsequent activation of cAMP-response element binding (CREB) protein, a transcription factor that mediates the expression of genes that facilitate L-LTP (Kandel, 2001). Recent studies suggest that DNA methyltransferase (DNMT) is required in the early induction of LTP in the hippocampus, and the subsequent methylation of certain genes may mediate memory formation (Levenson et al., 2006; Miller and Sweatt, 2007). Given the bidirectional control it exerts on gene expressions, DNA methylation is thought to serve as the driving force of upregulating the transcription of memory-enhancing genes such as brain-derived neurotrophic factor (Bdnf), reelin and activity-regulated cytoskeleton-associated protein (Arc) while reducing the production of memory-suppressing gene transcripts such as calcineurin (CaN) and protein phosphatase 1 (PP1) (see below). These changes are often accompanied by altered expressions of DNMTs and DNA demethylases, and they are essential to successful memory formation. In light of this, memory researchers have invested much effort into investigating epigenetic mechanisms of how gene expression modulates behavior and memory propagation, which has given rise to the subfield in epigenetics called neuroepigenetics (Day and Sweatt, 2011).

DNA methylation, which governs a wide array of gene expressions in a self-perpetuating manner, contributes to various cellular functions. Research from the past decades has contributed to an increasing body of evidence that suggests the intricate epigenetic network may serve as the foundation of cellular events contributing to memory formation. However, the temporal and regional specificity of DNA methylation in the neural circuitry remains a concern in memory research. Neuromodulatory surgical techniques, which involve the delivery of an electrical current to produce therapeutic relief, have shown immense potential in treating memory-related and neuropsychiatric disorders (Perlmutter and Mink, 2006; Merkl et al., 2009; Chang et al., 2018; Hadar et al., 2018). Given that previous studies have validated the ability of neuronal activity to effectively alter DNA methylation (Nelson et al., 2008; Guo et al., 2011a), it is reasonable that researchers would begin to explore the mechanism of neuromodulation techniques, such as transcranial direct current stimulation (tDCS) (Podda et al., 2016), electroconvulsive therapy (ECT) (Ma et al., 2009), vagus nerve stimulation (VNS) (Sanders et al., 2019), and deep brain stimulation (DBS) (Pohodich et al., 2018), from an epigenetic perspective. By researching these neuromodulation techniques in in vitro studies that mimic cellular changes under neuronal activity, we can gain important insights on the potential therapeutic application of these current methods. The therapeutic effects of neuromodulation techniques have been shown to be mediated by epigenetic mechanisms. A deeper understanding of the underlying epigenetic mechanism of these techniques may pave way for their application in neurological disorders associated with epigenetics. In this review, we highlight the major findings in neuroepigenetic research, which have revealed the intertwined network connecting DNA methylation and cognitive abilities. We also discuss DNA methyltransferase 3a (DNMT3a), methyl-CpG binding protein 2 (MeCP2), and DNA demethylase, which are three components highly involved in DNA methylation-dependent epigenetic regulation. Lastly, we discuss how neuromodulation holds much promise as an epigenetic modulator, and examine the current research to gain insights that may help to further clarify the role of DNA methylation in memory modulation.

Section snippets

Role of DNA methylation in memory consolidation and retrieval

Understanding the role of DNA methylation in learning and memory remains key to deciphering the evidence obtained from neuromodulation studies. DNA methylation is a reversible epigenetic mechanism associated with gene silencing. This process depends on DNA methyltransferase (DNMT), which functions by attaching a methyl group donated by S-adenosyl-l-methionine to cytosine nucleotides residing in a specific dinucleotide region called CpG island to form 5-methylcytosine (5-mC), which consequently

DNA methyltransferase 3a (DNMT3a)

In view of the interaction between neuronal activity and the DNA methylation landscape, it is important to highlight the molecular machinery involved in these highly dynamic processes to establish their proposed functionality in neuromodulation (Fig. 1). One of the enzymes substantially involved in DNA methylation is DNMT. Recent studies have shed light on the role of DNMT in modulating the methylation pattern, and hence, in mediating memory formation and maintenance. Among the types of DNMT,

Neuromodulation techniques

Neuromodulation involves the delivery of an electrical current in specific brain regions to restore normal circuit connections and brain functions. Neuromodulation techniques have recently given rise to promising therapeutic options due to their efficacy in a wide range of neuropsychiatric and neurodegenerative disorders, such as major depressive disorder (Mayberg et al., 2005; Merkl et al., 2009; Neyazi et al., 2018), Rett syndrome (Hao et al., 2015; Lu et al., 2016), Alzheimer’s disease (

Conclusion

Much effort has been devoted to revealing the molecular basis behind memory formation. It is becoming clear that neuronal activity and DNA methylation are both pivotal components that bridge environmental stimuli and cognition. Neuromodulation techniques, such as neuronal activation and altered neurotransmission, have been demonstrated to have great efficacy in treating learning and memory-related disorders. Although the usage of these techniques in neuropsychiatric disorders is not a novel

Declaration of Competing Interest

All authors declare no conflicts of interest.

Acknowledgements

The scientific work was funded by grants from the Hong Kong Research Grant Council (RGC-ECS 27104616), and The University of Hong Kong URC Supplementary Funding (102009728) awarded to LWL.

References (156)

  • P. Follesa et al.

    Vagus nerve stimulation increases norepinephrine concentration and the gene expression of BDNF and bFGF in the rat brain

    Brain Res.

    (2007)
  • M.S. George et al.

    Vagus nerve stimulation: a new tool for brain research and therapy

    Biol. Psychiatry

    (2000)
  • K. Gulmez Karaca et al.

    Adult hippocampal MeCP2 preserves the genomic responsiveness to learning required for long-term memory formation

    Neurobiol. Learn. Mem.

    (2018)
  • J.U. Guo et al.

    Hydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brain

    Cell

    (2011)
  • C. Hamani et al.

    Deep brain stimulation reverses anhedonic-like behavior in a chronic model of depression: role of serotonin and brain derived neurotrophic factor

    Biol. Psychiatry

    (2012)
  • N. Hidaka et al.

    Involvement of alpha4beta2 nicotinic acetylcholine receptors in working memory impairment induced by repeated electroconvulsive seizures in rats

    Epilepsy Res.

    (2013)
  • G.A. Kaas et al.

    TET1 controls CNS 5-methylcytosine hydroxylation, active DNA demethylation, gene transcription, and memory formation

    Neuron

    (2013)
  • S.L. Kigar et al.

    Gadd45b is an epigenetic regulator of juvenile social behavior and alters local pro-inflammatory cytokine production in the rodent amygdala

    Brain Behav. Immun.

    (2015)
  • J.M. Levenson et al.

    Evidence that DNA (cytosine-5) methyltransferase regulates synaptic plasticity in the hippocampus

    J. Biol. Chem.

    (2006)
  • H. Lu et al.

    Loss and gain of MeCP2 cause similar hippocampal circuit dysfunction that is rescued by deep brain stimulation in a Rett syndrome mouse model

    Neuron

    (2016)
  • G. Malleret et al.

    Inducible and reversible enhancement of learning, memory, and long-term potentiation by genetic inhibition of calcineurin

    Cell

    (2001)
  • D.A. Malone et al.

    Deep brain stimulation of the ventral capsule/ventral striatum for treatment-resistant depression

    Biol. Psychiatry

    (2009)
  • A. Mann et al.

    Chronic deep brain stimulation in an Alzheimer’s disease mouse model enhances memory and reduces pathological hallmarks

    Brain Stimul.

    (2018)
  • H.S. Mayberg et al.

    Deep brain stimulation for treatment-resistant depression

    Neuron

    (2005)
  • R.R. Meehan et al.

    Identification of a mammalian protein that binds specifically to DNA containing methylated CpGs

    Cell

    (1989)
  • A. Merkl et al.

    Antidepressant electroconvulsive therapy: mechanism of action, recent advances and limitations

    Exp. Neurol.

    (2009)
  • C.A. Miller et al.

    Covalent modification of DNA regulates memory formation

    Neuron

    (2007)
  • R.E. Amir et al.

    Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2

    Nat. Genet.

    (1999)
  • J. Ausio

    MeCP2 and the enigmatic organization of brain chromatin. Implications for depression and cocaine addiction

    Clin. Epigenet.

    (2016)
  • N. Axmacher et al.

    Interactions between medial temporal lobe, prefrontal cortex, and inferior temporal regions during visual working memory: a combined intracranial EEG and functional magnetic resonance imaging study

    J. Neurosci.

    (2008)
  • F. Bahner et al.

    Hippocampal-dorsolateral prefrontal coupling as a species-conserved cognitive mechanism: a human translational imaging study

    Neuropsychopharmacology

    (2015)
  • F.R. Bambico et al.

    Neuroplasticity-dependent and -independent mechanisms of chronic deep brain stimulation in stressed rats

    Transl. Psychiatry

    (2015)
  • S. Barbash et al.

    Length-dependent gene misexpression is associated with Alzheimer’s disease progression

    Sci. Rep.

    (2017)
  • G.R. Barker et al.

    The different effects on recognition memory of perirhinal kainate and NMDA glutamate receptor antagonism: implications for underlying plasticity mechanisms

    J. Neurosci.

    (2006)
  • T. Bienvenu et al.

    Molecular genetics of Rett syndrome: when DNA methylation goes unrecognized

    Nat. Rev. Genet.

    (2006)
  • T.V. Bliss et al.

    Long-lasting potentiation of synaptic transmission in the dentate area of the anaesthetized rabbit following stimulation of the perforant path

    J. Physiol.

    (1973)
  • P. Boon et al.

    Deep brain stimulation in patients with refractory temporal lobe epilepsy

    Epilepsia

    (2007)
  • C. Castellano et al.

    NMDA receptors and learning and memory processes

    Curr. Drug Targets

    (2001)
  • M. Chahrour et al.

    MeCP2, a key contributor to neurological disease, activates and represses transcription

    Science

    (2008)
  • A.Y. Chan et al.

    Effect of neurostimulation on cognition and mood in refractory epilepsy

    Epilepsia Open

    (2018)
  • C.H. Chang et al.

    Brain stimulation in alzheimer’s disease

    Front. Psychiatry

    (2018)
  • W.G. Chen et al.

    Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2

    Science

    (2003)
  • Z. Chen et al.

    Accumulated quiescent neural stem cells in adult hippocampus of the mouse model for the MECP2 duplication syndrome

    Sci. Rep.

    (2017)
  • L. Chouliaras et al.

    DNMT3A moderates cognitive decline in subjects with mild cognitive impairment: replicated evidence from two mild cognitive impairment cohorts

    Epigenomics

    (2015)
  • C.M. Connor et al.

    DNA methylation changes in schizophrenia and bipolar disorder

    Epigenetics

    (2008)
  • I. Corripio et al.

    Clinical improvement in a treatment-resistant patient with schizophrenia treated with deep brain stimulation

    Biol. Psychiatry

    (2016)
  • J.J. Day et al.

    DNA methylation regulates associative reward learning

    Nat. Neurosci.

    (2013)
  • J.O. de Jong et al.

    Epigenetic effects of electroconvulsive seizures

    J. ECT

    (2014)
  • J.V. Deng et al.

    MeCP2 in the nucleus accumbens contributes to neural and behavioral responses to psychostimulants

    Nat. Neurosci.

    (2010)
  • D. Denys et al.

    Deep brain stimulation of the nucleus accumbens for treatment-refractory obsessive-compulsive disorder

    Arch. Gen. Psychiatry

    (2010)
  • Cited by (0)

    View full text