You are on page 1of 18

Biomedicine & Pharmacotherapy 170 (2024) 115951

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Drug addiction and treatment: An epigenetic perspective


Arunkumar Singh Koijam a, Kabrambam Dasanta Singh a, Bunindro Singh Nameirakpam a,
Reena Haobam b, Yallappa Rajashekar a, *
a
Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India,
Takyelpat, Imphal 795001, Manipur, India
b
Department of Biotechnology, Manipur University, Canchipur, Imphal 795003, Manipur, India

A R T I C L E I N F O A B S T R A C T

Keywords:
Drug addiction is a complex disease affected by numerous genetic and environmental factors. Brain regions in
Addiction reward pathway, neuronal adaptations, genetic and epigenetic interactions causing transcriptional enhancement

Abbreviations: ADPr, ADP-ribose; Airn, antisense Igf2r RNA; ANP, Atrial natriuretic peptide; ATP, Adenosine triphosphate; AVP,, Vasopressin; BDNF, Brain-
derived neurotrophic factor; CamKIIa, Calcium/calmodulin-depencent kinase II a; cAMP, Cyclic AMP; CBP, CREB binding protein; CCAT1-L, Colon cancer associated
transcript 1 large; CCAT1-S, Colon cancer associated transcript 1 small; ccr2, C-C chemokine receptor 2; Cdk5, Cyclin-dependent kinase 5; cFos, cFos protooncogene;
CREB, cAMP response element binding; CRISPR-Cas9, Clustered Regularly Interspaced Short Palindromic Repeats/ Cas9; crRNA, CRISPR RNA; CYP1A1, Cytochrome
P450 Family 1 Subfamily A Member 1; D3R, Dopamine receptor D3; dCas9, Dead Cas9; DLK1, Delta-like 1 homolog; DNMTs, DNA Methyltransferases; DNMT1, DNA
Methyltransferase 1; DNMT3A, DNA Methyltransferase 3 A; DNMT3B, DNA Methyltransferase 3B; DPF, Double plant homeodomain finger; DRD2, Dopamine receptor
D2; DSB, Double strand break; DUBs, Deubiquitinating enzymes; EGR1, Early growth response; Ehmt1, Euchromatic histone-lysine-N-methyltransferases 1; Ehmt2,
Euchromatic histone-lysine-N-methyltransferases 2; FDA, Food and drug administration; FosB, FosB protooncogene; G9a, G9a gene or Ehmt2 gene; GAD1, Glutamate
Decarboxylase 1; GID, Gibberellin insensitive dwarf; GLP, G9a like protein; gRNA, guide RNA; GTP, Guanosine triphosphate; GWAS, Genome wide association study;
H2AK119, H2A lysine 119; H2AK119ub1, H2A lysine 119 monoubiquitylation; H2BK34ub, H2B lysine 34 ubiquitylation; H2AX, histone H2A variant; H2AXS139,
H2AX Serine 139; H2AXY142, H2AX tyrosine 142; H2BS10, H2B serine 10; H2BS32, H2B serine 32; H2BS36, H2B serine 36; H3R2me2a, H3 arginine 2 dimethylation
asymmetric; H3K122cr, H3 lysine 122 crotonylation; H3K4me3, Histone H3 trimethylation; H3K27ac, Histone H3 Lysine 27 acetylation; H3K27cr, Histone H3 Lysine
27 crotonylation; H3R17me1, H3 Arginine 17 monomethylation; H3K4, histone H3 lysine 4; H3K36, histone H3 lysine 36; H3K79, histone H3 lysine 79; H3K9,
histone H3 lysine 9; H3K27, histone H3 lysine 27; H3S10, H3 serine 10; H3S28, H3 serine 28; H3Y41, H3 tyrosine 41; H3T3, Histone H3 threonine 3; H3T6, H3
threonine 6; H3T11, H3 threonine 11; H3T45, H3 threonine 45; H3Q5ser, H3 glutamine 5 serotonylation; H3K9K14ac2, histone H3 lysine 9 lysine 14 diacetylation;
H3K4me3, H3 Lysine 4 trimethylation; H3K4me3Q5ser, H3 Lysine trimethylation; H3Q5dop, H3 glutamine 5 dopaminylation; H4H18, H2 histidine 18; H4H75, H4
histidine 75; H4K20, histone H3 lysine 20; H4K20me1, histone H4 lysine 20 monomethylation; H4S1, H4 serine 1; HATs, Histone acetyltransferases; HCT, Histone
crotonyltransferases; HDACs, Histone deacetylases; HDCRs, Histone decrotonylases; HDMs, Histone demethylases; HDR, Homology directed repair; HERP,, Homo­
cysteine-induced endoplasmic reticulum protein; HMTs, Histone methyltransferases; HOTAIR, HOX Transcript Antisense RNA; HP1, Heterochromatin protein 1; ICR,
Imprint control region; IEGs, Immediate early genes; IG-DMR, Intergenic differentially methylated region; KCr, Lysine crotonylation; LINE, Long interspersed ret­
rotransposable elements; Lnc RNAs, long non-coding RNAs; MEG3, Maternally expressed 3; miRNA, MicroRNA; MIAT, Myocardial Infarction Associated Transcript;
EMX2O, Empty spiracles homeobox 2 O; MEF2, Myocyte enhancer factor-2; MOF, K(lysine) acetyltransferase 8 (KAT8) gene; MAOA,, monoamine oxidase A;
MARylation, Mono-ADP-ribosylation; MBD, Methyl-CpG-binding domain; MeCP2, Methyl CpG binding protein 2; MEG3, Maternally expressed gene 3; MEF2,
Multiple myocyte-specific enhancer factor 2; MIAT, Myocardial infarction associated transcript; MSN, Medium spiny neurons; MT1, Metallothionein 1; MT2, Met­
allothionein 2; NAc, Nucleus Accumbens; NAD, Nicotinamide adenine dinucleotide; NEAT1, Nuclear paraspeckle assembly transcript 1; NEAT2, Nuclear paraspeckle
assembly transcript 2; NF-κB, Nuclear factor-kappa B; NGF, Nerve growth factor; NHEJ, Non-homologous end joining; NMDA, N-methyl D Aspartate; NR2B, NMDA
receptor 2B; OFC, Orbitofrontal cortex; OPRM1,, Mu opioid receptor 1; PAM, Protospacer Adjacent Motif; PARylation, Poly-ADP-ribosylation; PCAF, P300/CBP-
associated factor; PDYN, Prodynorphin; PFC, Prefrontal cortex; Pitx3, Pituitary homeobox 3; piRNAs, P-element-induced wimpy testis-interacting RNAs; PIWI, P-
element-induced wimpy testis; PIWIL1, PIWI like protein 1; PKA, Protein kinase A; Pomc, Proopiomelanocortin; PTMs, Post translational modifications; Raf-1, Raf1,
protooncogene; RRBS, Reduced representation bisulfite sequencing; ROS, Reactive oxygen species; RVDs, Repeat variable diresidues; SAHA, Suberoylanilide
hydroxamic acid; SAM, S-adenosyl methionine; sg RNA, single guide RNA; siRNAs, Short interfering RNAs; SNCA,, α-synuclein; Srcin1, Src kinase signaling inhibitor
1; SRF, Serum-response factor; SUMO, Small ubiquitin-related modifier; TAB-Seq, Tet-assisted bisulfite sequencing; TALENs, Transcription activator-like effector
nucleases; TET1, Ten Eleven Translocase 1; Tet3, Ten Eleven Translocase 3; TORC, Transducer of Regulated CREB; tracrRNA, Trans Activating crRNA; UHRF,
Ubiquitin-like, with PHD and RING finger domains; UTR, Untranslated region; VS, Ventral Striatum; VTA, Ventral Tegmental Area; WDR5, WD repeat containing
protein 5; WGBS, Whole genome bisulfite sequencing; ZFNs, Zinc-finger nucleases; ZFP, Zinc finger proteins.
* Corresponding author.
E-mail address: rajacftri77@gmail.com (Y. Rajashekar).

https://doi.org/10.1016/j.biopha.2023.115951
Received 16 September 2023; Received in revised form 23 November 2023; Accepted 27 November 2023
Available online 2 December 2023
0753-3322/© 2023 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

Epigenetic or repression of multiple genes induce different addiction phenotypes for varying duration. Addictive drug use
Epigenome editing complexes causes epigenetic alterations and similarly epigenetic changes induced by environment can promote addiction.
CRISPR-Cas9 Epigenetic mechanisms include DNA methylation and post-translational modifications like methylation, acety­
TALEN
lation, phosphorylation, ubiquitylation, sumoylation, dopaminylation and crotonylation of histones, and ADP-
ZFN
ribosylation. Non-coding RNAs also induce epigenetic changes. This review discusses these above areas and
stresses the need for exploring epidrugs as a treatment alternative and adjunct, considering the limited success of
current addiction treatment strategies. Epigenome editing complexes have lately been effective in eukaryotic
systems. Targeted DNA cleavage techniques such as CRISPR-Cas9 system, CRISPR-dCas9 complexes, transcrip­
tion activator-like effector nucleases (TALENs) and zinc-finger nucleases (ZFNs) have been exploited as targeted
DNA recognition or anchoring platforms, fused with epigenetic writer or eraser proteins and delivered by
transfection or transduction methods. Efficacy of epidrugs is seen in various neuropsychiatric conditions and
initial results in addiction treatment involving model organisms are remarkable. Epidrugs present a promising
alternative treatment for addiction.

1. Introduction emotional and sexual abuse, peer pressure, disease conditions etc [6].
Sex, age and ethnicity have reports of significant association while
Drug addiction is a complex disease of recurring psychological and socio-economic status, lower educational achievement, religion, marital
physiological impairment on persistent use of psychoactive drugs. It is status and region showed varying degrees of association with different
characterized by behavioural transition from positive reinforcement of addictive drugs [7]. Drug exposure induces expression changes in
drug rewarding effects to negative reinforcement of countering with­ numerous genes through epigenetic mechanisms within various brain
drawal symptoms, as presented in Fig. 1 [1]. Addiction is influenced by regions, accompanied by changes in structure, function and neuro­
multiple mechanisms of genetic, epigenetic, molecular and circuitry. They are retained for long durations spanning months or years
neuro-endocrine entities, environmental influences, behavioural, [8]. Familial interaction, genetic vulnerability, neuroendocrine changes
cognitive, emotional and motivational conditions, and brain responses and behaviours preceding addiction onset interlink through epigenetic
to reward and stress cues [2]. Genetics involve alterations in DNA, modifications in protective and risk polymorphisms on endogenous
genes, nucleotide sequences, expression changes arising thereof and its opioid, monoaminergic, oxytoninergic and hypothalamic pituitary ad­
heritability while epigenetics involve expression changes and herita­ renal stress response systems [9]. Considering the limited success of
bility due to regulatory effects arising out of changes in DNA and current addiction treatment strategies, reversible and inducible nature
DNA-associated proteins but not DNA sequence. Onset of drug use is of epigenetic mechanisms and initial remarkable results with model
fundamentally dependent upon the genes and environment to a lesser organisms offer tremendous possibilities of epigenetic treatment for
degree, while progression towards addiction is dependent significantly addiction.
upon genetic [3] and environmental factors [4,5]. While addiction and
its phenotypes have been associated with various genes, environmental
factors affecting addiction include family conditioning, physical,

Fig. 1. The cycle of Addiction and drug-induced neuronal effects. Intoxication/ Binge stage- characterized by rewarding sensation and positive reinforcement with
subsequent learning and memory formation. Reward mediating neurotransmitters and associated processes in Nucleus accumbens, Ventral and Dorsal striatum,
Global pallidus and Thalamus are engaged. Withdrawal/ Negative effect stage- characterized by negative emotions and stress with involvement of Amygdala, Bed
nucleus of stria terminalis, and ventral striatum. Preoccupation/ craving- characterized by processing conditioned reinforcement, contextual inputs and execution
involving Prefrontal cortex, Anterior cingulate, Orbitofrontal cortex, Hippocampus and Basolateral nucleus of amygdala [1]. Drug-induced short term and long term
effects are executed by processes involving neurotransmitters, neuronal proteins and gene expression changes specifically in neurons of the reward circuit.

2
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

2. Brain functions and neuronal changes in addiction properties in NAc via regulation of ΔFosB expression and also regulates
actin cytoskeletal remodelling for spinogenesis. ΔFosB overexpression
The Reward system or Mesolimbic system in the brain is the centre in NAc amplifies drug reward and self administration while transcrip­
for cognitive and physiological processing of reward [10]. Brain struc­ tional silencing has the reverse effects [23].
tures involved in Reward Pathway are crucial in determining the degree
of addiction, which include Prefrontal cortex (PFC), orbitofrontal cortex 3. Epigenetics and mechanisms
(OFC), Ventral Tegmental Area (VTA), Nucleus Accumbens (NAc),
Dorsal Striatum, Ventral striatum (VS), Amygdala and Hippocampus. Epigenetics involves the study of changes in gene expression and
Much of these structures are a mixture of distinct cell types with varied their heritability due to chemical modifications in DNA and DNA-
signaling and electrophysiological properties offering differential tran­ associated proteins but not DNA sequence changes. Epigenetic pro­
scription leading to a variety of drug responses [11]. Imaging studies cesses contribute to the unique differential gene expressions in various
have demonstrated that intoxication/ binge stage of addiction cycle are cells and correlate to nucleosomal changes regulating transcription and
primarily influenced by VTA and VS, while negative/ withdrawal stage biological processes. Changes in chromatin structure, particularly
is influenced by extended amygdala, and Anticipation/ preoccupation condensation and decondensation of nucleosomes, leads to concealment
stage is influenced by OFC, DS, PFC, basolateral amygdala and hippo­ or exposure of specific gene regions. These changes inhibit or allow
campus [1]. Drugs cause rewarding stimuli by activating the dopami­ access of transcriptional machinery to the chromatin scaffold and thus
nergic mesolimbic system leading to dopamine release in target nuclei gene expression is repressed or activated. A nucleosome is a basic DNA
[10]. Dopamine levels are increased in the NAc by almost all addictive compaction unit made of approximately 147 bp DNA coiled around a
drugs via indirect or direct action on dopaminergic neurons. Dopamine core histone octamer of two copies each of H2A, H2B, H3 and H4.
signaling in NAc is a crucial mechanism to drug reinforcement. Drugs Adjacent nucleosomes are linked together by 20–60 bp linker DNA and
increase dopamine release in brain and continued use decrease natural H1 histone. Histones of nucleosome have a carboxyl-terminal, a globular
production levels of dopamine, inducing drug dependency for main­ domain and a relatively disorganised amino-terminal bearing 20–35
taining normal dopamine levels. Of the various molecular mechanisms amino acid residues where majority of the post translational modifica­
of drug action, few prominent ones include the downregulation or tions (PTMs) occur. Nucleosomal restructuring after replication involves
upregulation of their receptors or transporters, changes in membrane daughter chromosomes receiving distributed parental histones at
permeability or ion conductance, inhibition of endogenous opioids and around the same dislodged locus and newly synthesized histones
peptides by feedback mechanism, effects on neurotransmitter systems replenishing to replicate covalent modification states of parental nu­
and neuronal plasticity of reward circuit [12]. Glutamatergic-mediated cleosomes [24]. Epigenetic mechanisms include methylation of DNA;
neuroadaptations in dopamine striato-thalamo-cortical and limbic acetylation, methylation, sumoylation, phosphorylation, ubiquitylation,
pathways are triggered by chronic exposure to drugs [13]. There are serotonylation, dopaminylation and crotonylation of histones; ADP
certain general effects of various drugs on NAc and VTA of brain. ribosylation and gene expression regulation by non-coding RNAs. Pro­
Alcohol and opiates stimulate DA production indirectly through inhi­ tein machineries affecting epigenetic modifications are broadly classi­
bition of GABAergic neurons of VTA. Stimulants, on the other hand, fied as Writers, Readers and Erasers, based on their respective catalytic
enhance DA transmission directly. Nicotine induces DA neuron firing activity of addition, recognition (through specific binding motifs and
through action on acetylcholine receptors. Cannabinoids dysregulate triggering various signaling pathways) and removal of various func­
normal endocannabinoid signaling of dopaminergic neurons to tional groups. Fig. 2 provides a brief illustration of various environ­
GABAergic and glutamatergic neurons through regressive suppression of mental factors and protein machineries affecting epigenome. Readers
inhibition and regressive suppression of excitation respectively [13]. contain various protein domains which bind to epigenetically modified
Neurotransmitters, their distribution, excitatory or inhibitory nature sites to direct or guide relevant protein complexes for downstream
and timespan in synapse also contribute to addiction phenotypes. effects.
Epigenetic processes, drug-induced neuronal and synaptic plasticity has Epigenetic changes can be induced by numerous chemical and
a crucial role towards learning, memory, addiction development and environmental factors [25]. Epigenetic inheritance in somatic cells is
relapse [14,15]. Drugs, neurotransmitters and neuronal proteins interact linked to long-term behavioural changes. Germ cells display parental
to cause alterations in gene expression particularly the immediate early imprinting and with progression from zygote to later embryonic stages,
genes (IEGs). Epigenetic changes may be induced in these genes based there is de-novo methylation. Imprinted genes exhibit specific DNA
on the degree and phase of drug exposure or during withdrawal [16]. methylation patterns and retain molecular memory of their germline but
Neurons acquire high plasticity via such epigenetic changes to store and are also contingent on epigenetic reprogramming post environmental
integrate new inputs or information. Neuronal changes in terms of exposures. The epigenome of primordial germ cells reset to regain
morphology and dendritic spine density of medium spiny neurons developmental totipotency where a limited number of genes can escape
(MSN) within the nucleus accumbens (NAc) and prefrontal cortex of rats the epimark erasure process [26]. Various drugs of abuse are known to
are induced by various drugs of abuse [17,18]. Methamphetamine induce intergenerational effects through epigenetic processes acting at
exposure induces the expression of synapsins, syntaxin 1 A and syn­ the level of gametogenesis or early embryogenesis [27–29] as detailed in
aptophysins which causes change in synaptic plasticity [19]. Various Section 4. Various factors account for epigenetic marks to be either
transcription factors have crucial roles in drug related synaptic plas­ tissue specific or tissue independent and studies that can provide a
ticity. They contain DNA-binding domains specific to sequences in consensus remains elusive.
promoter or enhancer regions of genes that promote or block RNA po­
lymerase II transcriptional complex recruitment. Apart from other 3.1. DNA methylation
functions, cAMP response element binding (CREB) mediates dendritic
spine formation and generates silent synapses vital to synaptic devel­ DNA Methylation has distinct role in tissue-specific gene expression,
opment linked with intensified drug seeking [20,21]. Nuclear X-chromosome inactivation, parental imprinting, chromatin organisa­
factor-kappa B (NF-κB) regulates synaptic plasticity, crucial for dendritic tion and overall organism development. There is extreme complexity in
spine induction in Nucleus Accumbens Medium Spiny Neurons [22]. patterns of DNA methylation and are correlated with genomic locations.
Multiple myocyte-specific enhancer factor 2 (MEF2) modulates tran­ The nucleobases Cytosine, Guanine and Adenine display natural enzy­
scription activation or repression of genes engaged in excitatory synapse matic methylation tendency which modifies into 5-methylcytosine, 5-
regulation and cell survival. It may negatively regulate neuronal plas­ hydroxymethylcytosine, N7Methylguanine, N4-methylcytosine and N6-
ticity. Serum-response factor (SRF) mediates cocaine rewarding methyladenine. 5-methylcytosine is the most commonly found DNA

3
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

Fig. 2. Environmental factors and Epigenetic regulatory mechanisms. Various environmental factors modulate gene expression and corresponding phenotypes
through epigenetic mechanisms. Proteins/ enzymes grouped under Writers add chemical moieties or epigenetic tags to DNA or histones which can be interpreted by
Readers to help various factors to mediate cellular processes and Erasers remove the epigenetic tags. For instance, histone acetylation relaxes chromatin, activate
transcription and increase gene expression.

methylation in humans, followed by a tenfold lower level of 5-hydroxy­ birth, synaptic pruning and synaptogenesis coincides with CpH
methylcytosine while other methylations are rare or limited to pro­ methylation accumulation in brain [35]. Human primary astrocytes
karyotes [30]. Methylation mainly occurs at cytosine residues exposed to cocaine have been reported to change CpG and non CpG
particularly at the CpG dinucleotides. CpG dinucleotides are concen­ methylation levels of mtDNA [36]. Methylation changes may occur at a
trated in genomic regions called CpG islands (regions of about 1 kb with specific locus or globally or both and may be an epigenetic driver of
more than 55% GC) and CpG shores (0–2 kb from promoter) and to a various conditions [37]. Demethylation and de-novo methylation
lesser degree in CpG shelves (2–4 kb from promoter) and open sea (other executed by Demethylases and DNA Methyltransferases (DNMTs) during
regions in gene promoter). The human genome is estimated to contain embryonic development allow establishment of unique and stable
around 29000 CpG islands with locations near transcription start site or methylation patterns in genes of differentiated tissues [33]. The brain
first exon [31]. Methylation of CpG islands and CpG shores is associated exhibits one of the maximum methylation levels in comparison to other
with underexpression of genes [32]. In dividing cells, methylation of the human parts [38,39] mCH frequency were found to be highest in adult
gene region downstream of first exon known as gene body, have been neurons while glial mCH level was observed to be same as cortical tis­
correlated with overexpression while the same is not exhibited by sues of foetus. Out of the total methylation in the genome of adult
slow-dividing and non-dividing cells [33]. Generally, hypomethylation human neurons, about 53% is accounted to mCH while about 47% is
occurs in the active gene regulatory elements and methylation di­ accounted to mCG [39]. Rizzardi et al. [40] reported that differentially
minishes the binding affinity of transcription factors (TFs) thereby often methylated regions (DMRs) within neuronal population are greatly
resulting in underexpression of the relevant gene. Interestingly, analysis enhanced for heritability of addictive behaviour. While DNA methyl­
with methylation-sensitive SELEX (systematic evolution of ligands by ation patterns vary between tissues and within cells, methylation studies
exponential enrichment) involving 542 human TFs have shown that CpG involving peripheral tissues other than cells in various brain regions,
methylated sequences are preferential for many TFs [34]. Methylation of have indicated towards possible usage as biomarkers of addiction
cytosine is also reported in CpH (where H= A, C or T) dinucleotides [41–43]. Pilot scale study involving 27 participants with substance use
restricting mostly to neurons, muscles and embryonic stem cells. After disorder have demonstrated differential methylation of 5532 CGs with

4
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

1074 CG hypermethylation and 4458 CG hypomethylation in T-cells to condensation of chromatin and repression of genes [55].
[41]. A similar observation of 186 differentially methylated positions
was observed in a study involving 23 cocaine use disorder participants 3.2. Histone methylation
and 24 healthy controls [43]. Hypermethylation of specific genes like
OPRM1 has also been observed in opioid use disorder individuals [42]. Histone methylation involves incorporating methyl groups donated
DNA methylation is the catalytic transfer of a methyl group from S- by SAM in the lysine and arginine residues present at histone tails.
adenosyl methionine (SAM) to a cytosine or adenine residue. Three Histone tails do not exhibit change in overall charge but hydrophobicity,
prominent DNMTs viz. DNMT1, DNMT3A and DNMT3B methylate DNA. basicity and anionic affinity are modified on histone methylation. His­
DNMT1 maintains de-novo DNA methylation, with its deletion inducing tone methylation and demethylation processes are catalyzed by Histone
global demethylation and death in human embryonic stem cells [44], or methyltransferases (HMTs) and Histone demethylases (HDMs) respec­
overexpression causing embryonic lethality [33]. DNMT3A regulates tively (represented in Fig. 3). Histone methylation regulate gene
genome-wide de-novo methylation and methylation patterns during expression at multiple transcriptional levels including chromatin ar­
parental imprinting, embryonic and haematopoietic stem cell differen­ chitecture and loci-specific regulation by loosening or binding their tails
tiation. DNMT3B is the main de-novo DNA methyltransferase actively around the DNA thus granting or blocking access to cell-specific tran­
expressed during implantation and embryonic stages. DNMT3A and scription factors, initiation factors, elongation factors and associated
DNMT3B have been largely linked to structural changes in DNA ac­ proteins [56]. Lysine 4 of histone H3 can be mono, di or tri methylated
counting to long-term memory formation [45]. Three different protein [57]. In many cell types, H3K4me1 is linked to gene repression.
families viz. methyl-CpG-binding domain (MBD) proteins, ubiquitin-like H3Kme2/3 recruits chromatin readers or interactors such as PHD fingers
containing plant homeodomain and RING finger domain (UHRF) pro­ of ING1 and ING2 [58]. Gene activation is promoted by H3Kme3
teins and Zinc finger proteins recognize DNA methylations and aid in through transcription activation effector molecules [57]. Complex
reading by acting along with various protein complexes. Chromatin combinatorial possibilities of histone methylation regulate gene
remodelers such as methylases and histone deacetylases are recruited to expression and phenotypic outcomes including disease development.
methylated DNA by MBD proteins, but they also tend to bind to regu­ For instance, monomethylation on lysine 20 of histone H4 (H4K20me1)
latory regions and unmethylated DNA [46]. On repeated cocaine leads to chromatin contraction [59,60]. H4K20me1 may also facilitate
exposure, DNMT3A and DNMT3B overexpression was observed. Further, chromatin openness and subsequently promote transcription [61] while
in the caudate putamen of rats, increased methylation of PP1Cb gene, H3R17me1 induces transcriptional activation [59]. Lysine methylation
and subsequent repression of PP1Cb gene was observed along with its at H3K4, H3K36 and H3K79 induces transcriptional activation while
MeCP2 binding [47]. An isoform of DNMT3A is the DNMT3A2 and its H3K9, H3K27 and H4K20 cause transcriptional silencing [62].
overexpression in NAc shell of rats but not NAc core, has been reported H3K9me3 and H3K9me2 have been associated with transcriptionally
following acute cocaine administration. While cocaine-reinforcing ef­ silent, while H3K4me3 and H3K4me2 are associated with actively
fects are not mediated by DNMT3A2, it regulates cocaine-cue memories transcribing genes [63]. Intermittent subcutaneous methamphetamine
driving reinstatement to seek cocaine, following early abstinence and injection in mice leading to methamphetamine-induced behaviour,
incubation of cocaine craving [48]. The same study [48] used primary caused elevated mRNA expression of chemokine receptor CCR2, which
striatal cultures to demonstrate the activation of DNMT3A2 by dopa­ in turn has been attributed to an increase in trimethylation of H3 histone
mine D1 receptor signaling and the induction of IEGs viz. Arc, FosB and at lysine 4 (H3K4me3) in CCR2 promoter [64]. Increased H3K4me3 has
Egr2 is impaired by DNMT3A2 knockdown. Demethylation during been correlated with methamphetamine-associated memory. Knock­
developmental processes and cellular differentiation occurs through two down of the methyltransferase MLL1 and subsequent reduction of
mechanisms viz. Passive DNA demethylation and Active DNA deme­ H3K4me3 disrupt methamphetamine-associated memory while knock­
thylation. Replication-dependent 5mC dilution or Passive DNA deme­ down of the histone demethylase, KDM5C leading to H3K4 hyper­
thylation results from lack of functional maintenance methylation. methylation prevents expression of methamphetamine-associated
Replication-independent Active DNA demethylation is an indirect pro­ memory [65]. Numerous biological processes such as transcription, cell
cess which brings about 5mC replacement with unmodified cytosine cycle, DNA repair, differentiation, stress response, long-term memory
preceded by enzyme-mediated 5mC modification [49]. Another set of formation and aging are influenced by histone methylation [56,66].
enzymes called Ten eleven translocation (TET) proteins are responsible
for oxidation of 5-methylcytosine and potentially remove DNA 3.3. Histone acetylation
methylation [50,51]. TET1, TET2 and TET3 oxidise 5-methylcytosine
into 5-hydroxymethylcytosine, 5-formylcytosine and 5-carboxylcyto­ Histone acetylation exhibits transfer of acetyl group from donor
sine. TET1 but not TET2 and TET3, has been demonstrated to be Acetyl CoA to ε-amino group (–NH3 +) of lysine of histones. Acetylation
downregulated in the nucleus accumbens following repated cocaine especially lysine in N-terminal histone tails reduce DNA-histone contacts
administration [52]. Further, differential levels of 5-hydroxymethylcy­ by neutralizing the electrostatic interaction between positively charged
tosine were reported at 11,511 regions across autosomes following amino groups on lysine and negatively charged DNA. This loosens the
repeated cocaine exposure [52]. TET1 and TET3 have also been asso­ nucleosomal structure for easy access to transcriptional machinery and
ciated with morphine seeking behaviour as epigenetic modulators and thus gene expression is activated. Lysine deacetylation on the other
present options in addiction treatment [51]. hand, has the reverse effect of chromatin condensation and transcription
Paternal epigenome is globally demethylated before the first cell inhibition [62]. Two groups of counteracting enzymes viz. histone
division while maternal epigenome gets passively demethylated with acetyltransferases (HATs) and histone deacetylases (HDACs) regulate
subsequent cleavage [53]. Epigenetic marks in Imprint control region acetylation by adding or removing acetyl groups respectively (Fig. 3).
(ICR) of genes regulate genomic imprinting and are heritable resulting in HATs are classified on the basis of sub-cellular location as Type A HATs
monoallelic, parent of origin dependent gene expression. Maternal (nuclear localization) that acetylate histones and chromatin associated
methylation is predominant in ICRs with consequence of inheritance proteins and Type B HATs (predominantly cytoplasmic localization)
and reprogramming through maternal line [54]. Transcription factors which acetylate newly synthesized histones. The 18 membered HDAC
which can be repressors or activators, interact with methylated cyto­ superfamily has four classes, broadly grouped into two protein families:
sines. CRE sequence methylation promotes C/EBPα binding to DNA Zinc-dependent metalloproteins (Class I, Class II and Class IV HDACs)
consequently activating a promoter set specialised for differentiation of and NAD+ dependent proteins (Class III HDACs) [67]. Histone acety­
adipocytes. Methylated CpG dinucleotide bind to methyl-binding lation has been observed on all the core histones in human viz. lysine
domain (MBD) proteins and induces deacetylation of histones leading residues at positions 5, 9, 13, 15, 36, 74, 75, 95, 118, 127 and 129 in

5
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

Fig. 3. Histone post translational modifications, PTM enzymes and substrates. Histone ADP-ribosylation- Reversible single or multiple ADP-ribose addition on lysine
(K)/ arginine (R)/ aspartic acid (D)/ glutamic acid (E)/ cysteine (C)/ serine (S) causing Mono-ADP-ribosylation (MARylation) and Poly-ADP-ribosylation (PAR­
ylation). Sumoylation- Adding SUMO to Lysine residue in histones in a 3-step process of activation, conjugation and ligation. Ubiquitylation- Adding ubiquitin to
lysine residues in histone. Phosphorylation- adds phosphate from ATP/GTP to serine, tyrosine and threonine in histones. Methyltransferase adds methyl group from
SAM to lysine and arginine in histones. Acetylation- Counteracting HATs and HDACs add and remove acetyl groups respectively in lysine of histones.

histone H2A; lysine at 5, 11, 12, 15, 16, 20, 23, 24, 43, 46, 57, 85, 108, lysine 9 and 14 at H3. Cdk5, Bdnf and Fosb gene promoters displayed
116, 120 and 125 in histone H2B; lysine at 4, 9, 14, 18, 23, 27, 36, 37, increased acetylation at H3K9, H3K14 but not H4K5, H4K8, H4K12,
56, 64, 79, 115 and 122 in histone H3; and lysine at 5, 8, 12, 16, 20, 31, H4K16, following chronic cocaine administration [70]. CREB-binding
77, 79 and 91 in histone H4 [68,69]. Chronic exposure to psychosti­ protein (CBP) which is a histone acetyltransferase is responsible for
mulants has been associated with increased histone acetylation at pro­ mediating transcriptional activation by histone acetylation and
moters of various genes involved in synaptic transmission including recruitment of basal transcription machinery. It regulates expression of
Cdk5, CamKIIa, NFkB subunits [70,71]. Although a few inconsistencies genes required for memory and long-term plasticity [73]. CBP deletion
have been reported [72,73], acetylation of lysine at 5, 8, 12 and 16 of H4 in mice NAc has been correlated with impaired cocaine sensitivity and
at Fos and Fosb is increased with acute cocaine administration but not at context-cocaine associated memory [73]. In a study [74], the NAc of

6
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

chronic cocaine treated mice or saline were taken to map the histones by protein kinases while phosphatases cause counteractive
genome-wide promoter interaction of modified histones using ChIP and dephosphorylation (Fig. 3). Histone phosphorylation in general, has a
antibodies directed against polyacetylation of H3 and H4. Chronic transcriptional activation effect due to repulsion between negatively
cocaine administration induced hyperacetylation of histones H3 and H4 charged phosphates in histones and DNA backbone as well as HAT
at 959 and 692 gene promoters respectively while hypoacetylating 83 recruitment, specific transcription factor association and removal of
and 123 gene promoters respectively [74]. Autoacetylation of P300 and proteins or PTMs engaged in gene silencing [83]. In human H2AX his­
acetyltransferase action has been reported to be upregulated by NEAT1 tone variant, phosphorylation has been reported on serine at 139 and
along with changes in H3K27ac and H3K27cr [75]. Maurice et al., [76] tyrosine at 142 (H2AXS139 and H2AXY142). Similarly, serine at 32 and
demonstrated that P300/CBP-associated factor (PCAF) homozygous 36 position in H2B (H2BS32, H2BS36); threonine at 3, 6, 11, 45 (H3T3,
knockout mice have memory and learning impairments, indicating the H3T6, H3T11, H3T45), serine at 10 and 28 (H3S10, H3S28) and tyrosine
inseparable role of HATs. At 2 months maturity, these knockout mice at 41 (H3Y41) in H3, and serine at 1, histidine at 18 and 75 (H4S1,
indicated short term memory deficits that were correlated to hippo­ H4H18 and H4H75) in H4 histones are phosphorylated [84]. It has been
campal changes, Fos levels and activation of MAP kinase. Memory and linked to various cellular processes such as transcription regulation
learning worsened with age but differences based on gender were not involving H2BS36, H2BY37, H3S10, H3S28, H2BS32, H3T6, H3T11,
observed [76]. Considering the extensive overlapping of learning and H3Y41, H4S1 and H4S47; meiosis involving H2BS10 and H4S1; mitosis
memory with neural processes underlying addiction [77], HATs may involving H3S10 and H3S28; DNA damage involving H2AX S139 and
play a role through PCAF. In striatal neurons, D2-like receptors blockade H4S1; apoptosis involving H2AXS139, H2BS10 and H3T45 [85]. Pro­
cause phospho-acetylation of H3 (H3pS10-acK14) through moter regions of rapidly inducible ‘immediate early’ genes like c-fos
cAMP-dependent PKA and NMDA receptor signaling [78]. HDACs can be exhibit histone phosphorylation [85]. H2AX phosphorylation is a tran­
recruited by MBD proteins to methylated DNA, but also tend to bind to sient modification resulting from DNA damage. Although there are no
regulatory regions and unmethylated DNA through nucleosome direct experimental evidences of H2AX phosphorylation with drugs,
remodeling deacetylase (NuRD/Mi-2) complex members [46]. In­ certain drugs like opioids are known to induce DNA damage in the NAc
hibitors of HDAC (HDACi) such as Valproic acid and Magnesium val­ and PFC in heroin exposed mice [86]. MeCP2 phosphorylation at Ser421
proate have been approved by FDA and used for treatment of neurologic is known to be induced by psychostimulants, and this phosphorylation
disorders and seizures. Valproic acid acts on gamma amino butyric acid has role in limiting circuit plasticities in NAc which relate to addictive
(GABA) levels and most HDACs [79]. The expression of Class III HDACs behaviours [87].
like SIRT1 and SIRT2 within mouse NAc is increased on chronic cocaine
exposure. Chronic morphine exposure causes SIRT1 overexpression 3.5. Histone ubiquitylation
[80]. Rewarding effects of both cocaine and morphine were enhanced by
viral-mediated SIRT1 and SIRT2 overexpression in NAc of mice. On the Histone ubiquitylation or ubiquitination is a PTM involving enzy­
other hand, drug reward decreased in mice with SIRT1 knockdown in matic covalent ligation of an ubiquitin protein to histones wherein an
NAc. SIRT1-induced behavioural effects are regulated through various isopeptide bond gets established between ε-amino group (–NH+ 3 ) of
synaptic proteins in NAc and SIRT1-induced dendritic spines on medium lysine in histones and carboxylic acid group of the last glycine (glycine
spiny neurons of NAc [80]. In a study [81] involving rats withdrawn 76) in ubiquitin. Three groups of enzymes process ubiquitylation viz.
from methamphetamine self administration and footshocks for several Ubiquitin-activating enzymes (E1s), Ubiquitin-conjugating enzymes
weeks, HDAC1 mRNA significantly decreased in the dorsal striatum of (E2s) and Ubiquitin ligases (E3s). The structure of chromatin is affected
sensitive rats as against control rats. Sensitive rats displayed a significant by ubiquitinylation through mechanisms such as regulating DNA access
decline in the mRNA levels of HDAC2 and HDAC8 as compared to to transcriptional machinery, providing binding site for transcriptional
control and resistant rats. In comparison to control rats, the dorsal activators or inhibitors, trans-histone cross talk and associating other
striatum of sensitive rats showed a significant decline in HDAC class IIa, PTMs. Although all histones including H1 can be ubiquitinylated, H2A,
IIb and IV. HDAC9 mRNA was significantly downregulated in sensitive H2B and H3 are most commonly ubiquitinylated in the nucleus. Ubiq­
rats compared to control and resistant rats. The dorsal striatum of uitinylation can be reversed by ubiquitin-specific peptidases called
resistant rats showed significant increase in the mRNA of the lysine Deubiquitinating enzymes (DUBs) (Fig. 3). H2A ubiquitination has a
acetyltransferases KAT4 and KAT5 in comparison to sensitive and con­ repressive effect on transcription while H2B ubiquitination has an
trol rats [81]. Downregulation was observed for SIRT2, SIRT3 and SIRT4 activating or permissive effect on transcription [88]. Ubiquitination has
in the dorsal striatum of shock resistant and shock sensitive rats as been reported for H2AK119ub1, H2AK13/15ub1, H2AK127/129ub1,
compared to controls. The effects seen on few epigenetic enzymes in the H2BK120ub1, H3K18/23ub1, H3K14ub1 and H3K23/36/37ub1 [89].
NAc is suggestive of the fact that dorsal striatum participates in H2B is ubiquitinated by PAF1 complex in the course of transcription
behavioural manifestations of forced abstinence following compulsive elongation. PAF1C controls ubiquitination of H2BK120ub and
meth administration [81]. Cadet et al., [82] have reported that mRNAs H2BK34ub. H2BK120ub is required for H3K79me2/3 and H3K4me2/3.
of HDACs were differentially expressed in the NAc of rats with H2BK34ub enhances H4K16ac via MOF action [90].
compulsive methamphetamine and abstinent rats. These were analysed
on the basis of responses to footshocks during the period of metham­ 3.6. Histone sumoylation
phetamine self administration. In the study [82], HDAC1, HDAC3,
HDAC6, and HDAC8 expressions in NAc were observed to be signifi­ Histone Sumoylation is another reversible PTM where isopeptide
cantly different between shock resistant (SR) and shock sensitive (SS) bond is formed between the ε-amino group (–NH+ 3 ) of lysine in histones
rats. SS rats displayed significantly higher SIRT1 and SIRT7 expression and C-terminal Glycine of small ubiquitin-related modifier (SUMO)
compared to SR rats. SS rats also exhibited significantly higher mRNA thereby covalently attaching SUMO group. Humans express four distinct
levels compared to both SR and control rats [82]. HDAC inhibitors are SUMO isoforms viz. SUMO1, SUMO2, SUMO3 and SUMO4. Sumoylation
potential epidrugs for addiction treatment and are discussed in later process is achieved through an activating heterodimer enzyme (E1:
section. Aos1/Uba2), a conjugating enzyme (E2: Ubc9) and SUMO ligase (E3)
(Fig. 3). Sumoylation occurs on all histone types including variants H2A.
3.4. Histone phosphorylation Z and H2A.X. affecting transcriptional repression by recruiting HDAC
and HP1. A study [91] demonstrated that sumoylation has been linked
Histone phosphorylation involves catalytically adding phosphate to various processes such as chromatin organization, protein-protein or
group from ATP or GTP to serine, tyrosine and threonine amino acids of protein-DNA interactions, transcriptional regulation, nuclear-cytoplasm

7
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

transport, genome stability, protein stability, stress response, cell cycle contributes to vulnerability and relapse. Administration of cocaine
progression, apoptosis and cellular localization [92,93]. acutely enhances dopamine production in NAc synapses and cause
dopaminylation of glutamine 5 at histone H3 (H3Q5dop). H3Q5dop in
3.7. Histone ADP ribosylation VTA is strongly associated with cocaine-induced transcriptional plas­
ticity. H3Q5dop attenuation in the VTA following prolonged cocaine
ADP-ribosylation is a reversible PTM characterized by enzymatic access decreases the release of dopamine in ventral striatum. H3Q5dop
transfer of ADP-ribose (ADPr) from donor nicotinamide adenine dinu­ reduction in VTA impairs cocaine seeking [107]. H3Q5dop and dopa­
cleotide (NAD+) to amino acid side chains with nucleophilic nitrogen, mine increases following acute cocaine exposure, resulting in under­
oxygen or sulphur (such as lysine, arginine, aspartic acid, glutamic acid, expression of DRD2 and finally leads to cocaine withdrawal. Further,
cysteine and serine) on histone protein forming N-, O- or S- glycosidic during cocaine withdrawal, Src kinase signaling inhibitor 1 (Srcin1 or
linkage to ribose and releasing nicotinamide. It is affected by ADP- p140CAP) reduces H3R2me2a binding. This is followed by inhibition of
ribosyltransferases that covalently attaches ADPr and ADP- dopaminylation and diminished NAc Src signaling, cocaine reward and
ribosylglycohydrolases which remove ADPr (as represented in Fig. 3). motivation [108].
There are two ways in which ADP-ribosylation can occur viz. Mono-
ADP-ribosylation (MARylation) and Poly-ADP-ribosylation (PAR­ 3.10. Histone crotonylation
ylation). ADP-ribosylation provides numerous options to change the
functions and properties of substrates (histones) due to the fact that Histone crotonylation was first identified in the core histones (H2A,
ADPr can cause hydrogen bonding and hydrophobic interactions as it H2B, H3 and H4) by Tan et al., [68] and has been associated with gene
contains an adenine ring, two ribose moieties and bears two negative activation. Crotonyl-CoA is the substrate for crotonylation. Much like
charges at physiological pH. ADP-ribosylation regulates numerous key acetylation, crotonylation at lysine residue of histones neutralize its
processes such as transcriptional regulation, DNA repair, cellular positive charge weakening DNA interactions and loosening chromatin
transport, immune response, cell survival etc. [94]. PARylation by for access to various DNA-interacting proteins. It is thus suggested to
PARP1 are vital for viability in neurons, learning and memory. Transient have a role in transcription activation of genes [109]. Lysine crotony­
decondensation of chromatin is induced by PARylation leading to lation (KCr) has been reported to involve in replication stress response.
transcription of genetic components required for long term memory Replication stress reversibly regulates crotonylation and ubiquitination
[95]. Cocaine-induced PARylation has been reported in several studies of H2A at lysine 119 (H2AK119). Nucleosome destabilisation capability
[96–100]. A study [96] demonstrated the global increase in PARP1 and of Kcr has been suggested to involve in chromatin to nucleoprotamine
Poly(ADP-ribose) or PAR in mice NAc following repeated cocaine transition in the course of spermatogenesis [110]. Crotonylation occurs
exposure. Further, H1 and H3 were selectively PARylated with chronic on all the five histones in humans viz. lysine residues at positions 33, 63,
cocaine use, indicating a permissive chromatin structure. PARP1 has 84, 89, 96, 158 and 167 in histone H1; lysine at 36, 118, 119 and 125 in
also been associated with cocaine-paired cues and acts by binding at histone H2A; lysine at 5, 11, 12, 15, 16, 20, 23 and 34 in histone H2B;
promoter of a putative transposase inhibitor [97] PARP1 is thus a po­ lysine at 4, 9, 18, 23, 27 and 56 in histone H3; and lysine at 5, 8 and 12 in
tential target for cocaine relapse prevention. PARP1 is known to be histone H4 [68,69]. Crotonylation of 122 lysine in H3 (H3K122cr)
post-transcriptionally regulated by miR-125b and miR-124. Acute and causes hindrance in water-mediated hydrogen bond formation with
chronic cocaine exposure, both in cultured neurons and in mice, DNA backbone thereby reducing DNA-histone interaction and eventu­
persistently downregulates miR-125b and miR-124, which in turn in­ ally favouring transcriptional activation. Although crotonylation mainly
creases PARP1 expression [98,99]. PARP1 acts through positive or promotes transcription, crotonylation at H3K9 and H3K27 on
negative regulation of promoters and enhancers along with transcrip­ Pro-growth genes and endocytosis-related genes respectively, have been
tional modulation via structural changes in chromatin. PARP1 enzyme reported to inhibit transcription [111]. MOF, CBP and p300 are the main
action is crucial for activation of NF-κB [101]. The regulation of gene histone crotonyltransferases (HCT) in the mammalian system while
expression is attributed to the competition between histone H1 and Class I HDACs are the main histone decrotonylases (HDCRs). Decroto­
PARP1 to bind to specific gene promoters [102,103]. PARP1 also con­ nylation is also processed by Sirt1 via multiple targets [109]. While
tributes to open chromatin formation through prevention of H3K4me3 Bromodomain proteins are also identified Kcr readers, Double plant
demethylation. Binding of KDM5B to chromatin is inhibited by its homeodomain finger (DPF) domain has been identified as histone
PARylation and this enhances RNA polymerase II binding at promoter Kcr-preferential reader [69,112]. Lysine crotonylation level is suggested
regions of specific genes with positive regulation [104]. to be impacted by intracellular crotonyl-CoA concentration, dynamics of
crotonyltransferase and decrotonylase, and ratio of
3.8. Histone Serotonylation Acetyl-CoA/crotonyl-CoA [69]. Kcr has been reported to occur in
non-neuronal and neuronal cells in spinal cord, medulla oblongata, tri­
Histone serotonylation is the PTM involving the addition of serotonyl geminal ganglia and dorsal root ganglia. Kcr has been suggested to be
group to histones. The 5th position of Histone 3 occupied by glutamine is involved in regulation of neuropathic pain and neuroinflammation
of specific importance in terms of serotonylation (H3Q5ser) as it has a [113] which are also associated with chronic opioid use [114].
permissive role in gene activation in conjunction with H3K4me3 during
differentiation of neurons. Cellular differentiation is associated with 3.11. Non-coding RNA induced epigenetic changes
increased levels of H3K4me3Q5ser. Mutant RN46A-B14 cells incapable
of H3Q5ser demonstrated inability for cellular differentiation [105]. Non-coding RNAs represent the group of RNAs which do not code for
WDR5 is an H3Q5ser reader whose interaction mediates epigenetic any functional protein but are engaged in post transcriptional gene
regulation and tumour development [106]. regulation. They include long non-coding RNAs (lncRNAs), P-element-
induced wimpy testis (PIWI)-interacting RNAs (piRNAs), microRNAs
3.9. Histone dopaminylation (miRNAs) and short interfering RNAs (siRNAs). They play a vital role in
epigenetic regulation. Various drugs of abuse regulate the expression of
Histone dopaminylation is another PTM wherein dopamine mole­ non-coding RNAs. Non-coding RNAs are crucial regulators of chromatin
cules are directly attached to glutamine in histones by Transglutaminase regulating proteins and transcription factors which in turn affect
2 and was first reported by Lepack et al., [107]. In association with neuronal adaptations and subsequent memory formation [115].
cocaine administration, dopaminylation of histones in the VTA play a lncRNAs are more than 200 nucleotide long RNA which are spliced,
major role in dysregulated gene expression within neurons and capped at 5′ and polyadenylated. With over 30,000 lncRNAs known in

8
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

humans and mice, lncRNAs such as MIAT, MEG3, Airn, CCAT1-L, family members cluster (let-7 f/ miR-98) and clusters miR-1/133a,
CCAT1-S, H19, HOTAIR etc. have been associated with epigenetic miR-182/96/183, and miR-216a/217, indicating a coordinate regula­
regulation of various genes and cellular processes [116]. Heroin abusers tion controlling similar mRNA targets [132]. miR-125b and miR-124
display upregulated MIAT, MEG3, NEAT1 and NEAT2 in NAc. NEAT1, post-transcriptionally regulate PARP1 during acute and chronic
NEAT2 and MIAT are known to link with nuclear domains and thus cocaine exposure [88,89]. Nicotine is known to promote the expression
likely affect gene expression changes [117]. MEG3 is a cAMP-sensitive of miR-140 and miR-504, while alcohol upregulates miR-9, miR-497 and
lncRNA which is maternally imprinted and expressed in specific neu­ miR-302b. miR-23b and let-7 is upregulated while miR-190 and
rons. Few splice variants of MEG3 act as transcriptional co-activators miR-133b is downregulated by opioids. Neuronal differentiation is
[118]. GWAS study have also associated MEG3 with risk for heroin regulated by morphine and one of the mechanisms is through down­
addiction [119]. mRNA levels of many synaptic genes and synaptic regulation of miR133b. Being a target of miR133B, the transcription
density is likely regulated by NEAT2 [118]. factor Pitx3, which activate dopamine transporter and tyrosine hy­
piRNAs, miRNAs and siRNAs are associated with chromatin- droxylase, is overexpressed [133].
mediated gene silencing, rearrangement of DNA and mRNA transcript
regulation. piRNAs are 26–31 nucleotide long, small non-coding RNAs 4. Addiction-induced epigenetic alteration and epigenetic
that interact with PIWI proteins. piRNAs are involved in regulation of alteration-induced addiction
chromatin, altering specific sites on a gene and silencing transposon.
They interact in clusters at euchromatin-heterochromatin junctions and Addiction or repeated exposure to various psychoactive drugs is
contain copies of ancient fragmented transposon. The piRNA in­ known to induce epigenetic effects. Numerous genes are influenced by
teractions and its pathway have also been associated with transgene addictive drugs directly or indirectly at an epigenetic level, which in
silencing, transcriptional silencing, formation of heterochromatin, turn induce expression changes that contribute to addiction phenotypes.
modification of histones and HP1α [120]. piRNAs inhibit transposon in Parental health, stress, exposure to various chemicals, diet and envi­
mice through DNA methylation of CpG islands in transposable elements. ronments influence the epigenetic pattern of offspring development,
The repressive H3K9me3 is maintained at high levels in long inter­ health and disease outcomes [134]. Cigarette and alcohol consumption
spersed retrotransposable elements (LINE) region of germ cells through during pregnancy induce alteration in DNA methylation and microRNA
piRNA pathway [121]. PIWIL1 activation in piRNA-PIWIL1 pathway expression [135,136]. Significant increase in CYP1A1 expression of
can cause specific localized change in hypermethylation while losing placenta along with differential methylation of critical xenobiotic
hypomethylation globally [122]. Endo-siRNAs such as RDR2-dependent response element has been observed following in-utero tobacco expo­
siRNAs can silence retroelements, transposons and DNA methylation sure [137]. Germline and sperm epigenomes have been reported to
[123]. piRNA is known to regulate target miRNA genes positively and change due to paternal alcohol use and exposure to chemicals like
target mRNA genes negatively [124]. chromium chloride and vinclozolin [134]. Long term epigenetic modi­
miRNA is a group of 21–23 nucleotide long small noncoding RNA fications contributing to addictive phenotype in later life can be induced
molecules capable of repressing translation and gene silencing through by prenatal exposure to cannabinoids [138]. Various studies involving
specific complementary binding of the 3′ untranslated region (UTR) of paternal cocaine administration have demonstrated enhancement in
mRNAs. MicroRNA mediated translational regulation induces synaptic drug susceptibility and related reward-based behaviours in their off­
remodeling that is likely involved in transition from recreational drug springs through epigenetics processes [27–29]. Paternal voluntary
use to compulsive use. Various miRNAs target functional protein com­ administration of cocaine has demonstrated to cause epigenetic germ­
plexes and epigenetic enzymes. For instance, miR-200a targets 3′ UTR of line reprogramming with increased Brain-derived neurotrophic factor
HDAC4 mRNA repressing HDAC4 expression and upregulating acety­ (Bdnf) mRNA and BDNF protein in medial prefrontal cortex of only male
lation levels of H3 histones. MiR-27a-5p directly targets Tet3 gene offsprings. This was also associated with H3 acetylation at BDNF pro­
involved in paternal methylation reprogramming [125]. MiR-29b is moters [27]. Further studies with similar cocaine exposure also indi­
known to target DNMT3A and DNMT3B directly and DNMT1 indirectly cated hippocampal epigenetic remodeling resulting in NMDA receptor
to eventually induce global DNA hypomethylation. miRNAs are also –dependent memory formation and impairments in synaptic plasticity of
under epigenetic control as they are transcribed by specifically assigned male offsprings only [29]. Analysis involving targeted next-generation
mRNA genes and undergo post-transcriptional processing [126]. bisulphite sequencing (TNGBS) of human astrocytes exposed to
Cocaine upregulates MIAT, MEG3, NEAT2 and EMX2O in NAc along cocaine demonstrated changes in levels of DNMT1, DNMT3a, DNMT3b,
with miR-324, miR-369, miR-212, miR-181a, Ago2, miR-7, miR-8, miR- TET1, TET2 and TET3 along with lower mtDNA methylation levels [36].
142 and let-7 families, while downregulating miR-124, let-7d, miR- A study demonstrated the overexpression of HDAC3 in Drd1 but not
200c, miR125a-5p, miR-200b, miR770–5p and miR200a. miR-324 and Drd2 cells of mice NAc, after chronic cocaine exposure. Mice with dis­
miR-369 associated with cocaine administration, modulates MEF2 and rupted HDAC3 enzyme activity within the NAc displayed changes in
FosB transcription factors involved in memory development and reward gene expression and synaptic plasticity following cocaine administra­
[115]. Long term cocaine administration induces upregulation of tion. However, there was no effect on cocaine-induced behaviour [139].
miR-212 in the dorsal striatum resulting in reduced motivation. miR-212 Cocaine exposure induces overexpression of MeCP2, HDAC2 and
intensifies Raf-1 action sensitizing adenylyl cyclase and overexpression HDAC11 accounting to increased acetylation of histones in NAc [140]
of Transducer of Regulated CREB (TORC), eventually increasing CREB while significantly downregulating GLP (Ehmt1) and G9a (Ehmt2) in
signaling [127]. MiR-212 interacts with MeCP2 in regulating cocaine NAc [11]. Chronic cocaine exposure also caused hyperacetylation at
administration through striatal brain-derived neurotrophic factor CamKIIa gene. Chronic alcohol and cocaine administration induces
(BDNF) [128]. Ago2 is involved in generating miRNA and miRNA regulatory changes of various genes like FosB, Cdk5 and Bdnf at chro­
mediated gene silencing. Drd2-expressing neurons devoid of Ago2 im­ matin level [119]. A study [70] observed hyperacetylation of H4 histone
pairs motivation for cocaine self administration in mice [129]. Chronic at cFos promoter on acute cocaine administration but not chronic
cocaine inhibits let-7d and miR-124 while inducing miR-181a. let-7d administration while histone H3 at bdnf and cdk5 promoters displayed
and miR-124 markedly downregulates D3R and BDNF respectively, hyperacetylation on chronic cocaine administration but not acute
indicating a complex network with cocaine-sensitive plasticity genes administration. DNMTs significantly regulate synaptic plasticity induc­
[130]. miR181a negatively regulates GluA2 post transcription, along tion in hippocampus while their inhibition changed methylation states
with involvement in cocaine-induced synaptic plasticity [131]. Cocaine in promoter region of BDNF and reelin genes which have been associated
exposure induces differential expression of miRNA clusters that encode with synaptic plasticity induction [141]. Cocaine administration in­
miR8 family members (miR-429/200a/b and miR-141/200c), let-7 duces DNMT3A overexpression causing enhanced dendritic spine

9
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

density in NAc [11]. Offsprings of mothers exposed to cocaine exhibited treat addiction and withdrawal of various psychoactive drugs vary
higher DNA methylation and overexpression of DNMTs [119]. Acute depending on the type of drug. Common treatment medications include
exposure of ethanol in mice caused lower expression of GAD1, HDAC2 buprenorphine, methadone, extended release naltrexone and lofexidine
and HDAC11 leading to hypoacetylation of histone at GAD1 and HDAC2 for opioid addiction treatment. Bupropion, varenicline and nicotine
promoters. Further MT1, MT2, EGR1 expression increased and associa­ replacement therapies are used for nicotine addiction treatment. For
tion was observed with increased H3K4me3 levels at MT2 promoter and alcohol addiction treatment disulfiram, naltrexone and acamprosate are
decreased H3K27me3 levels MT1 promoter [142]. Long-term ethanol used [154]. Medications generally mimic compounds that provide
consumption induces significant DNA hypomethylation due to intermediary relief or prolong reward sensation or treat withdrawal
decreased methionine and SAM production caused by excess ROS for­ symptoms, whose dosage can be gradually reduced to finally abstaining
mation following ethanol metabolism. Ethanol also causes site-specific it. Limited success of current treatment strategies with 40–60% relapse
methylation, phosphorylation and acetylation of histones [143]. His­ rate [155] calls for newer effective alternatives and adjuncts like epi­
tone H3 and H4 acetylation increased in amygdala following acute drugs. Epigenetic drugs or epidrugs are molecular complexes which
alcohol exposure whereas withdrawal decreased acetylation [144]. harness the potential of targeting epigenetic effector domains and the
Chronic alcohol use induces DNA demethylation of NMDA NR2B gene possibility to change epigenetic marks within the human genome.
with elevated NR2B expression in the cortex of mice. DNA hyper­ Presently, epidrugs specifically aimed for addiction treatment are very
methylation of PDYN have been observed in cultured neurons and cortex limited. RG108 is a DNA methylation inhibitor acting through direct
of alcoholics. Their lymphocytes exhibited increased DNA methylation binding and inhibition of DNMT1 enzyme [67]. Rat models injected
in α-synuclein (SNCA), homocysteine-induced endoplasmic reticulum with RG108 within NAc demonstrated significant attenuation of
protein (HERP), monoamine oxidase A (MAOA) and vasopressin (AVP) cue-induced cocaine seeking after upto 1 month treatment. Similar trials
genes while ANP gene was hypomethylated. Withdrawal of alcohol with S-adenosyl methionine (SAM) displayed opposite effects with long
displayed DNA hypermethylation of NGF gene promoter in blood. term cue-induced cocaine seeking [156]. Addictive behaviours of
Hypomethylation of imprinted IG-DMR region of DLK1 gene has been cocaine-dependent rodents have been reported to be inhibited by
observed in sperms obtained from alcoholics [119]. Alcohol probably administering L-methionine. Multiple piRNAs regulating Vav2 genes, in
induces epigenetic effects in human, as significant global hyper­ PFC following cocaine treatment have been reversed by L-methionine
methylation was observed among alcoholics than in normal controls treatment [124]. Methionine administration has also been reported to
[145]. PPMG1 gene hypermethylation was found to be associated with reverse demethylation of c-MYB binding site, inhibit cocaine condi­
alcohol use disorder [146]. Significant association was also reported for tioned place preference (CPP) and induce Bdnf IV expression by cocaine
MAOA gene DNA methylation and nicotine and alcohol dependence in CPP [157]. Methionine treatment reverses the reduced CpG methylation
women [147]. Cigarette smoking downregulates DNMT3B in lung can­ in the c-Fos gene promoter and c-Fos expression induced by cocaine
cer cells and causes demethylation of metastatic genes [148]. Meth­ [158]. However, FDA-approved epidrugs are in use for various cancer
amphetamine use induces histone H3 methylation (H3K4me3) on C-C and neuropsychiatric disease treatment as presented in Table 1. Many
chemokine receptor 2 (ccr2) gene promoter and knocking out this gene in epidrugs with different targets and action mechanisms are under study
mice results in impaired sensitization to methamphetamine [149]. at various experimental stages and presented in Table 2. Epidrugs have
Chronic methamphetamine has been reported to induce expression of evolved upto three generations with better understanding [159]. Epi­
SIRT1, SIRT2, HDAC1, HDAC2, repressor element 1 silencing tran­ drugs may range from targeting or interacting individual gene to set of
scription factor (REST) and REST corepressor proteins in dorsal striatum genes involved in one or more pathways. Most of the FDA-approved
of rats [150]. Hypoacetylation of H4K5, H4K12 and H4K16 at promoter epidrugs have shown remarkable efficiency as adjuncts to existing
or enhancer regions of GluA1 and GluA2 genes has been observed on medications. FDA-approved atypical antipsychotics such as clozapine,
chronic methamphetamine exposure [150]. Prenatal drug exposure olanzapine and quetiapine have reports of inducing epigenetic effects.
changes synaptic plasticity, neural development and phenotypes along Sulpiride and Clozapine activate DNA demethylation while the latter
with transgenerational inheritance pointing towards the role of epige­ also induces acetylation of H3K9 in PFC [160]. 5-azacitidine has been
netic marks in addiction [151]. Rat sperms of F1 offsprings exposed to reported to prevent excess intake of alcohol in mice through its action of
alcohol at fetal stages exhibited hypermethylation at Pomc gene and reducing DNA methylation by inhibiting DNMT. FDA-approved SAHA,
male germline transmitted the same upto F3 generation [152]. Epige­ HDAC inhibitor, has also been observed to inhibit alcohol-seeking
netic changes in the germline of male rats voluntarily administering motivation [161]. Preclinical studies with HDAC inhibitors viz Tri­
cocaine induce histone H3 acetylation (H3K9K14ac2) at Bdnf promoter chostatin A and Phenylbutyrate, have reported reduction in motivation
of medial PFC brain region and their male offsprings exhibited reduced and cocaine intake in self-administering rats [162]. Pretreatment with
cocaine intake [153]. Chronic opioid exposure induces hyper­ Valproic acid, an HDACi, in mice failed to reduce ethanol-induced
methylation in OPRM1 gene promoter with variations among different conditioned place preference (CPP) but increased the level of BDNF in
ethnic groups [119]. Many non-coding RNAs have been associated with ventral striatum, indicating its modulatory effect on neutrophic mech­
addiction and related phenotypes as discussed above. Enhanced resis­ anisms and addiction-like phenotype [163]. Another study [164] on
tance to conventional treatment modalities and vulnerability to relapse twenty individuals with cocaine-use disorder found that valproic acid
have been observed in people with drug use disorder, with epigenetic treatment was unable to reduce spontaneous and cue-induced cocaine
modifications mediated by addictive drugs [138]. High relapse rate craving. In mice, valproic acid has been reported to inhibit
among addicts also necessitates establishing its connection with long methamphetamine-induced behavioural sensitization when it is acutely
lasting and stable epigenetic changes. The undeniable evidences pre­ or repeatedly co-administered with methamphetamine [165]. Valproic
sented by the above studies linking addiction and epigenetic changes acid co-treatment in methamphetamine administered rats prevented
simply direct the exploration of epidrugs for possible addiction methamphetamine-mediated hypoacetylation of H4K5, H4K12 and
treatment. H4K16 at GluA1, GluA2 and GluN1 genes through HDAC1 and HDAC2
inhibition [150]. Valproic acid has shown to block
5. Prospects of epidrugs in addiction treatment methamphetamine-induced underexpression of the glutamate receptors
viz alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid re­
Current addiction treatment varies from medications, treating co- ceptors (AMPARs) and N-methyl-D-aspartate receptor (NMDAR) [150].
morbid conditions, counselling, behavioural therapies, exercise, yoga, Another HDAC inhibitor, RGFP966 caused cocaine CPP and
food supplements, vocational training to socialization depending on the drug-seeking behaviour to become extinct, particularly through HDAC3
drug/ addictive substance to be recovered from. Current medications to inhibition. RGFP966 significantly enhanced acetylation at lysine 14 on

10
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

Table 1
FDA approved epidrugs and their characteristics [182–184].
Epidrug Type FDA Approved Disease Approved Characteristics
Epidrug

DNA Methyltransferases inhibitors Decitabine Myelodysplastic syndrome Anti-cancer, anti-metabolite drug, cytotoxic to cancer cells.
(DNMTi) 5-Azacytidine Myelodysplastic syndrome Cytidine analog, incorporation in nucleic acids causing faulty
methylation
Hydralazine Hypertension Antihypertensive, inhibits DNMT1 activity, adverse immune
reactions observed
Procainamide Ventricular arrhythmias Inhibits DNMT1 activity
Tazemetostat Epitheloid sarcoma, refractory or Inhibits zeste homolog 2
relapsed follicular lymphoma
Histone Demethylase inhibitors (HDMi) Tranylcypromine Major depression disorder Irreversibly inhibits monoamine oxidase, antidepressant.
Deferiprone Treatment of iron Iron chelator
overload in thalassemia
Deferasirox Treatment of iron Iron chelator
overload
Histone methyltransferase inhibitors Phenelzine Depression Irreversibly inhibits monoamine oxidase.
(HMTi)
Histone Deacetylase inhibitor (HDACi) Vorinostat/SAHA Cutaneous T-cell lymphoma Inhibits all class I and II HDAC proteins
Panobinostat Multiple myeloma Inhibits all HDACs
Nicotinamide Pellagra, As food additives. Inhibits Sir2 protein non-competitively, inhibits deacetylation
action of sirtuins through reaction with an intermediate
Belinostat Peripheral T-cell lymphoma Inhibits HDACs and cell proliferation
Romidepsin Cutaneous T-cell lymphoma Strong inhibitor of HDAC1 and HDAC2, less potent inhibitor of
HDAC4 and HDAC6
Valproic acid Neurologic disorders, Antidepressive Anti-epileptic, anticancer, chemosensitizer, inhibit sodium
channels, increases GABA availability in synapse of brain
Magnesium Seizures Selective inhibition of HDAC2, under tests for treating
Valproate chemoresistance
Carbamazepine Epilepsy, trigeminal neuralgia, episodes Anti-epileptic, inhibits HDAC3 and HDAC7
of bipolar disorder
Bromodomain & Extra terminal motif Dinaciclib Different types of cancer Cyclin dependent kinase inhibitor
proteins inhibitors (BETi)
Poly(ADP-ribose) Olaparib Breast cancer, Ovarian cancer, Prostrate Inhibits PARP-1, PARP-2, PARP-3
polymerase cancer
(PARP1) inhibitor (PARPi) Niraparib Prostrate cancer Inhibits PARP-1, PARP-2
Rucaparib Metastatic castrate-resistant prostate Inhibits PARP-1, PARP-2, PARP-3
cancer
Talazoparib Metastatic castrate-resistant prostate Inhibits PARP-1, PARP-2
cancer
Non-coding RNAs Patisiran Familial amyloid polyneuropathy Direct sequence-specific degradation of Transthyretin (TTR) mRNA
in the liver
Givosiran Hepatic porphyria siRNA directed against 5-aminolevulinic acid synthase 1
Pegatanib Macular degeneration Binds and inhibits extracellular Vascular Endothelial Growth Factor
(VEGF165)
Protein arginine streptomycin and Rheumatoid arthritis inhibition of the binding of interleukin 1-beta to its cell surface
deiminase inhibitor methotrexate receptor
(PADi)

H3 histone (H3K14Ac) within NAc, hippocampus and infra-limbic cor­ epigenetic sites for a desired effect is imperative, it requires extensive
tex, and H3K14Ac has been correlated with learning. H4K8Ac is known and advanced studies ranging from in-silico, in-vitro to in-vivo systems.
to be regulated by HDAC3, and on RGFP966 treatment, acetylation of High throughput technologies for studying the complex milieu of epi­
H4 histone at lysine 8 (H4K8Ac) increased in infra-limbic cortex but not marks at genome-wide level and advanced artificial intelligence tools for
in hippocampus and NAc [166]. BIX-01294 is an investigational epidrug decoding epigenome data are expected to provide deeper insights.
inhibiting histone H3K9-specific methyltransferases G9a and to a lesser Various next generation sequencing platforms such as Whole genome
extent, Glp84. It induces autophagy, can attenuate neuronal gene bisulfite sequencing (WGBS), reduced representation bisulfite
expression and direct infusion into ventral striatum causes increased sequencing (RRBS), targeted bisulfite sequencing, Tet-assisted bisulfite
cocaine reward phenotype [167]. Epigenetic changes associated with sequencing (TAB-Seq) and RNA sequencing (RNA-Seq), need to be
use of various investigational epidrugs and psychotropic drugs have extensively exploited to produce more effective data relating to epige­
already been reported [168]. netics and gene expression in addiction. Data from such studies will
The realization of epidrugs as alternative or adjunct is not without eventually pave the epigenetic approaches to addiction treatment. There
challenges. The numerous mechanisms of actions in epigenetics is also a need for cost effective tools and processes that can reliably and
involving an even more numerous epigenetic effectors add to the effectively screen epidrugs. Site-specific and targeted delivery of epi­
complexity of addiction, but at the same time, also presents multiple drugs to exert their effects without any unwanted side-effects, non-tar­
approaches through which therapeutic models can be addressed. In spite geted or otherwise global epigenetic effects is another big challenge
of the knowledge of various compounds with epigenetic activity or towards epidrug realization. However, this has been largely overcome
epidrugs, the transition from preclinical studies to therapeutics of with recent developments in epigenome engineering tools such as Zinc
addiction is limited by issues of blood-brain barrier, tissue specific finger nucleases (ZFNs), Transcription activator-like effector nucleases
epigenetic landscape and global or non-targeted epigenetic effects (TALENs) and CRISPR/Cas 9, with characteristics (Table 3) that enable
among others. The limited understanding of epigenome and variations targeting specific epigenetic mark to single gene [169]. Targeted DNA
in epigenetic landscapes affected by addiction across cells, tissues, or­ cleavage techniques such as CRISPR-Cas9 system, transcription
gans and individuals is also a big hurdle. While striking the key activator-like effector nucleases (TALENs) and zinc-finger nucleases

11
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

Table 2 Table 3
Epidrugs in experimental stages for various disease conditions [182,183]. A list of characteristics of the epigenetic engineering tools CRISPR-Cas9, TALENs
Epidrug Type Experimental Epidrugs
and ZFNs.
Characteristics CRISPR-Cas9 TALENs ZFNs
DNA Methyltransferases inhibitors Epigallocatechin-3-gallate, Zebularine,
(DNMTi) Equol, Genistein, SGI-110, Procaine, Target site 22 bp+ PAM 30–40 bp 9–18 bp
Nanaomycin A, Disulfiram, Lomeguatrib, RG- length sequence
108, SGI-1027, MG98, CP-4200, Hinokitiol, Target range Any genomic site Upto 18742 Limited by ZFP
DC_517, DC-05 preceding PAM human genes combinations
Histone Methyltransferases (HMTi) UNC0321, UNC0224, EPZ-6438, DZNep, sequence based on TALEN
GSK343, Chaetocin, BIX-01338, BIX-01294, plasmid library
UNCO638, EPZ005687, GSK126, EPZ-5676, (Ho et al.,2018)
EPZ004777, SGC0946, E72, A-366, Off target Low High Moderate
UNC1999, CPI360, UNC0965, BIX-01337, Dimerization Not required Required Required
EI1, GSK503, BCI-121, LLY-507, EPZ015666, Recognition/ DNA-RNA DNA-Protein DNA-Protein
AZ505 ditrifluoroacetate, GSK3326595, Interaction/
MS023, UNC0642, JNJ-64619178, CM-579, type
EED226, MI-503, EPZ015866, MI-463, MI- Endonuclease Cas9 FokI FokI
538, MS049, CPI-169, BRD9539, LLY-283, Design and Relatively Easy Labour intensive Labour intensive
EML741, OTS186935, SGC3027, CPI-1205, assembly
Domatinostat (4SC-202), Tazemetostat. Specificity Moderate, High, few Moderate, more
Histone Demethylase inhibitors Pargyline, Clorgyline, Bizine, GSK2879552, comparatively more mismatches mismatches
(HDMi) KDMC-C70, JIB-04, ORY-1001, SID mismatches tolerated tolerated
85736331, Namoline, CBB1007, Methylstat, tolerated
GSKJ4, GSKJ1, QC6352, SP2509, KDOAM- Degenerate No Yes Yes
25, T-448, Daminozide, CPI-455, recognition
NCGC00244536, NCGC00247743, GSK-J2, Precision of Moderate High Moderate
Corin, GSK690, PBIT, S 2101, T-3775440 genome
hydrochloride editing
Proteins binding to methylated UNC669, UNC1215 Additional Yes Yes Yes
histones (PMHi) complex
Histone Acetyltransferase inhibitor Gallic acid, Garcinol, Anacardic acid, linkage
(HATi) Procyanidin, MB-3, CTK7A, Plumbagin, DNA No Yes Not known
Embelin methylation
Histone Deacetylase inhibitor Givinostat, AR-42, Entinostat, Apicidin, sensitivity
(HDACi) Pracinostat, Abexinostat, Resminostat, Design and Easy Labour intensive Complex and
CUDC-101, Toxoflavin, Inauhzin, Cambinol, assembly labour intensive
Salermide, Trichostatin A, CG-1521, OSU-
HDAC-42, HC-toxin, Plitidepsin,
Tasquinimod, Sodium butyrate, fusing FokI cleavage domain to DNA binding domain of zinc finger
Mocetinostat, Tefinostat, CHR-3996,
protein (ZFP), which can to recognise and cleave specific DNA se­
Curcumin, QUISINOSTAT, Sodium
phenylbutyrate, Pivanex, quences. Conjugated Cys2His2 motifs make up zinc fingers. Site speci­
Butyroyloxymethyl-diethyl phosphate, ficity is achieved through the 3–4 base pair recognition by DNA binding
Resveratrol, Dacinostat, Psammaplin A, ITF- domains in ZFPs which may be arranged in series for longer specific
A, ITF-B, OSU-HDAC-44, Ricolinostat,
locus, and this is the challenging aspect. ZFN dimers are required for
Tubastatin A, RGFP966, TMP195,
Fimepinostat, LMK-235, ACY-738, PCI-
double-strand DNA breaks. Their design involves a flanking pair of
34051, Nexturastat, CAY10603, ACY-775, unique sequence recognising ZFNs within which target spacer sequence
WT-161, MC1568, RGFP109, Citarinostat, of about 5–7 base pairs. ZFN cleavage induces DNA repair process by
Scriptaid, Tucidinostat (Chidamide), either non-homologous end-joining or homology-directed repair, during
Santacruzamate A, EDO-S101, Oxamflatin,
which, any desired target modification is effectively introduced [170].
HPOB, BML-210, Pomiferin, Domatinostat,
BG45, Bufexamac, Sinapinic acid, FT895, Zinc finger proteins can be clustered to recognize unique DNA sequences
CHDI-390576. of both strands in a target sequence. They can be fused with various
Proteins binding to acetylated CPI203, RVX-208, I-BET-726 epieffector molecules which can be activators or inhibitors of tran­
histones (PAHi) scription so as to form Zinc finger complexes.
Phosphorylation (Histone Kinase Ruxolitinib, KU-55933, VE-821
inhibitors)
Poly(ADP-ribose) Polymerase AMF-26, Talazoparib, Ilimaquinone,
inhibitors (PARPi) Veliparib, Olaparib, Niraparib, Rucaparib 5.2. Transcription activator-like effector nucleases (TALENs) / TALE-
Ubiquitin Signaling inhibitor PTC209, GW7647, PRT4165, ML323 complexes
Bromodomain & Extra terminal OTX15, JQ1, Thienodiazepine JQ1, I-BET762
motif proteins inhibitors (BETi) (GSK525762), I-BET151 (GSK1210151A), TALENs are gene editing protein complexes much like ZFNs. Typi­
GS-5829, CPI-0610, TEN-010, OTX-015,
BMS-986158, INCB057643 and ZEN003694
cally, a TALEN consists of a 12–28 repeat DNA binding domain, Fok1
Protein Arginine Deiminases (PADi) YW3–56, YW4–03, YW4–15, D-o-F-amidine, nuclease, a nuclear localization signal and a target gene transcription-
Cl-amidine, GSK484 activation domain. The DNA binding domains are proteins with a rela­
tively conserved 33–35 amino acid length, with polymorphisms at 12
and 13 positions called repeat variable diresidues (RVDs). Each RVD
(ZFNs) (represented in Fig. 4) have been exploited as targeted DNA
recognises a single base pair and determine nucleotide specificity. Thus
recognition or anchoring platforms, fused with epigenetic writer or
a chain of TALEs with aligned RVDs can recognise and effectively act on
eraser proteins (epieffectors) and delivered by transfection or trans­
a target sequence with precision. Fok1 dimerizes to generate double
duction methods.
strand breaks. TALEs can also be associated with various other proteins
or enzymes or epieffectors for better efficiency and precision. TALE-
5.1. Zinc finger nucleases (ZFNs) DNMT3ACD-3 L, TALE-CIB1; TALE/dCas9- KRAB, DNMT3ACD,
DNMT3L etc. are some remarkable targeted methylation tools with
Zinc finger nucleases (ZFNs) are engineered endonucleases made by varying efficacies. Similarly, TALE-TET1CD, TALE-TET1, etc. are

12
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

Fig. 4. Epigenetic editing tools. (A) A Zinc finger nuclease (ZFN) dimer bound to DNA. Endonuclease FokI is fused to a cluster of three Zinc finger proteins wherein
Fok1 dimerizes at target site (B) A Zinc finger complex. Zinc finger proteins recognising specific target nucleotide sequences is fused to one or more epieffector
molecules instead of Fok1. (C) Transcription activator-like effector nucleases (TALENs). Right and left strings of TALEs recognize specific DNA sequence targeting the
site for Fok1 cleavage. (D) Transcription activator-like effector complex. Right and left strings of TALEs fused with specific epieffectors recognize specific DNA
sequence for desired epigenetic alterations. (E) Clustered regularly interspaced short palindromic repeats/Cas9 (CRISPR/Cas9). crRNA of guide RNA binds to target
DNA region. RuvB and HNH domains of Cas9 cleave DNA adjacent to PAM. (F) Clustered regularly interspaced short palindromic repeats/dCas9. Cleavage inac­
tivated dCas9 is fused to epieffectors depending on desired epigenetic alteration. crRNA provides the target DNA specificity.

demethylation tools employed for various studies [171]. TALE com­ dCas9-SUNTag-GCN4-VP64-scFv, and, (iii) the three system dCas9 tool,
plexes however pose delivery issues using viral systems due to its size consisting of coupling or dimerization system, receptor-coupled system
[172] and other alternatives need to be exploited. and split dCas9 system. In coupling or dimerization system, a light factor
(PhyB, PIF- activated by light) or chemical factor (ABI, PYL1- activated
5.3. Clustered regularly interspaced short palindromic repeats/ Cas9 by abscisic acid; GAI, GID- activated by gibberellins) fuse separately to
(CRISPR/Cas 9) dCas9 and epieffector, and activate the later when coupled with their
drive. In Receptor-coupled system, epieffectors are activated by the
CRISPR/Cas 9 is a highly efficient genome and epigenome editing coupling of ligand and receptor. In Split dCas9 system, epieffectors are
system comprised of guide RNA (gRNA) or single guide RNA (sg RNA) activated by the connection of two cleft dCas9 parts in the target
and Cas 9 nuclease. The gRNA consists of two components viz. (i) sequence [173]. Overall, CRISPR-based epigenome editing tool requires
CRISPR RNA (crRNA) - of about 20 bp for binding to the target DNA a target DNA binding protein, a gRNA sequence and an effector protein.
sequence and Protospacer Adjacent Motif (PAM) sequence required for Site-specific DNA methylation manipulation in-vivo has been achieved
binding with Cas9, and (ii) Non coding Trans Activating crRNA using deactivated Cas9 fused to enzymes which methylate or demeth­
(tracrRNA) required for Cas9 cleavage. PAM is a 2–6 bp DNA sequence ylate DNA. A few targeted methylation dCas9 complexes include dCas9-
adjacent to DNA cleavage target and required for cleavage by Cas DNMT3ACD, dCas9-DNMT3A, dCas9- DNMT3ACD3 L, dCas9-Sun
nuclease. Target DNA cleavage is processed by Cas9 through its domains Tag-DNMT3A, dCas9-Sun TagDNMT3ACD, dCas9- MQ1, dCas9-split
viz. RuvC and HNH causing double strand break (DSB). DSBs are M.Sss etc. while targeted demethylation dCas9 complexes include
repaired by Homology directed repair (HDR) or Non-homologous end dCas9- TET1CD, dCas9- TET1CD, MS2-TET1CD, dCasTET1CD,
joining (NHEJ). A slight variation of CRISPR/Cas9 system is the dCas9 dCas9-Sun Tag-TET1CD, Gal4- ROS1 CD [171]. For instance, Shayevitch
complex system where catalytically dead Cas9 (dCas9) is fused with et al., [174] studied by co-transfecting cells with dCas9-DNMT3A-3 L
various EpiEffector enzymes, and is reported to be highly efficient in and single guide RNA (sgRNA) expression plasmids or dCas9-TET1 and
eukaryotic epigenome editing [173]. A dCas9 conserves DNA-binding sgRNA expression plasmids. In this, sgRNA guides dCas9 fusion protein
property while losing its cleavage activity. dCas9 epigenome editing to targeted site of genome allowing DNMT3A-3 L or TET1 for
tools have evolved with three generations viz. (i) dCas9 fused to a site-specific methylation or demethylation respectively. The promising
transcriptional activator or repressor (CRISPRa/i), e.g. dCas9-TET, aspect of CRISPR based genome and epigenome editing has the need to
dCas9-DNMT (ii) dCas9 fused to transcriptional activators and multi­ overcome the basic hurdles of crossing the blood brain barrier,
ple epieffectors such as VPR, VP64, MS2, SAM, SunTag, scRNA etc. e.g. non-specific cell targeting and in-vivo instability. Study [169] providing

13
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

remarkable evidence towards the role of epigenetic remodeling of gene understanding of the different factors of addiction. With the continuous
and subsequent control of stress response and reward, was demonstrated advancement in epigenetic understanding, epidrug developments, pre­
through targeting epigenetic marks at histone H3 viz H3 lysine 9/14 cision epigenome editing complexes, and safe and efficient delivery
acetylation (H3K9/14ac) and H3 lysine 9 dimethylation (H3K9me2) of systems, the control of epigenetic switch of addiction is in the next leap.
Cdk5 locus in NAc, in- vivo, employing engineered zinc finger proteins The prevention and reversal of addiction phenotypes with appropriate
and viral-mediated delivery. Resilience to social stress and epidrugs and combinatorial treatment is expected soon.
cocaine-induced locomotor behaviour increased with Cdk5-targeted
H3K9/14ac while Cdk5-targeted H3K9me2 impaired both conditioned Author statement
place preference and locomotor behaviour, but stress-induced social
avoidance behaviour was unaffected [169]. Targeted epigenetic editing A.S.K., R.H. and Y.R. conceptualized the study; A.S.K. performed
of H3K36me3 at Srsf11 in mice using dCas9 complex has been suc­ literature searches and wrote the first draft, A.S.K., K.D.S., B.S.N., R.H.
cessfully achieved in establishing its alternative splicing and cocaine and Y.R reviewed and edited the manuscript in its final form. All authors
rewarding behaviour [175]. Experimental evidence of suppression of contributed to and have approved the final manuscript.
cocaine-induced behaviour by Nr4a1 through epigenetic regulation was
demonstrated using dCas9 system fused to VP64 transcriptional acti­ CRediT authorship contribution statement
vation domain (dCas9-VP64), driven by a neuron specific promoter and
a single-guide RNA (sgRNA) targeting the Nr4a1 promoter [176]. A Koijam Arunkumar Singh: Conceptualization, Data curation,
modified CRISPR/dCas9 approach was also used by Teague and col­ Formal analysis, Investigation, Validation, Writing – original draft.
leagues to demonstrate the counteractive reinforcement effects of Nameirakpam Bunindro Singh: Formal analysis, Software, Validation,
cocaine by CREB-Zfp189 interaction within the NAc Drd2 + neurons Visualization. Singh Kabrambam Dasanta: Data curation, Formal
[177]. The repertoire of prospective CRISPR/Cas target sequences has analysis, Resources, Software, Writing – original draft. Haobam Reena:
recently been greatly expanded by the advanced engineered PAMs. With Funding acquisition, Supervision, Writing – review & editing. Raja­
web-based resources predicting the possible off-targets and efficacy of shekar Yallappa: Conceptualization, Funding acquisition, Project
gRNA, the crucial step of target sequence and corresponding gRNA se­ administration, Supervision, Writing – review & editing.
lection has become easier [178]. Interesting advancements in
CRISPR-Cas9 delivery at targeted cells including brain, are gradually in Declaration of Competing Interest
the horizon. Zou et al., [179] has reported unique nanocapsule-based
CRISPR-Cas9 delivery system that is non-invasive, facilitates BBB The authors declare that they have no known competing financial
penetration and safe for glioblastoma gene therapy. interests or personal relationships that could have appeared to influence
Epigenetic therapy has also been approached from various aspects the work reported in this paper.
including agents with multiple high affinity epigenetic targets wherein
one is an epigenetic enzyme and others being unrelated cellular targets Data Availability
[180]. This approach reduces adverse drug reactions, potential drug
resistance and simplifies treatment [181]. In the light of such de­ Data will be made available on request.
velopments, advancement in targeted epigenetic remodelling or DNA
cleavage techniques with fused or engineered epigenetic enzymes,
Acknowledgements
small-molecule epigenetic modulators, nanoparticle-based, vir­
al-mediated, peptide-based, and receptor-based drug delivery systems
Financial support from the DBT-RA Program in Biotechnology and
with ability to cross blood-brain barrier is expected for addiction
Life Sciences to A.S.K. is gratefully acknowledged. We thank the Di­
treatment. Limitations in understanding addiction-associated epigenetic
rector, Institute of Bioresources and Sustainable Development, Imphal,
changes include differential epigenetic alterations by different drugs,
for supporting this study. The IBSD manuscript number is IBSD/MS/
non-uniformity in study designs, obtaining participants with similar age,
2020/01/089.
sex and shared environmental factors, high quality post-mortem brain
tissues, uncertain epigenetic modifications, lack of clarity in time
References
duration that epigenetic changes can persist, and limited knowledge on
how epigenetic mechanisms and landscape works. Deeper studies [1] G.F. Koob, N.D. Volkow, Neurocircuitry of addiction, Neuropsychopharmacology
addressing these limitations and determination of whether the variety of 35 (2010) 217–238, https://doi.org/10.1038/npp.2009.110.
addiction-induced or epidrug-induced modifications and epimarks are [2] L. Strathearn, C.E. Mertens, L. Mayes, H. Rutherford, P. Rajhans, G. Xu, M.
N. Potenza, S. Kim, Pathways relating the neurobiology of attachment to drug
intergenerational or transgenerational are required. It would be addiction, Front. Psychiatry 10 (2019), 737, https://doi.org/10.3389/
worthwhile to study individual and co-ordinated effects of various fpsyt.2019.00737.
environmental factors, genetic predisposition, protein or enzymatic in­ [3] N. Hiroi, S. Agatsuma, Genetic susceptibility to substance dependence, Mol.
Psychiatry 10 (2005) 336–344, https://doi.org/10.1038/sj.mp.4001622.
teractions, microRNAs, gene expressions, genetic penetrance of risk [4] S.H. Rhee, J.K. Hewitt, S.E. Young, R.P. Corley, T.J. Crowley, M.C. Stallings,
gene/ alleles/ variants, vulnerable epigenetic marks and epigenetic Genetic and environmental influences on substance initiation, use and problem
penetrance of the epimarks for achieving effective addiction treatment use in adolescents, Arch. Gen. Psychiatry 60 (2003) 1256–1264, https://doi.org/
10.1001/archpsyc.60.12.1256.
strategies.
[5] K.S. Kendler, E. Schmitt, S.H. Aggen, C.A. Prescott, Genetic and environmental
influences on alcohol, caffeine, cannabis, and nicotine use from early adolescence
6. Concluding remarks to middle adulthood, Arch. Gen. Psychiatry 65 (2008) 674–682, https://doi.org/
10.1001/archpsyc.65.6.674.
[6] M. Whitesell, A. Bachand, J. Peel, M. Brown, Familial, social and individual
The complex nature of drug addiction with multiple factors and their factors contributing to risk for adolescent substance use, J. Addict. (2013),
varied combinatorial effects has been an unresolved loop. Proper iden­ 579310, https://doi.org/10.1155/2013/579310.
[7] J.C. Anthony, L.A. Warner, R.C. Kessler, Comparative epidemiology of
tification of epigenetic drug targets, effective epigenetic alterations,
dependence on tobacco, alcohol, controlled substances and inhalants: Basic
correction or prevention of epigenetic marks, precision delivery and findings from the National Comorbidity Survey, Exp. Clin. Psychopharmacol. 2
minimal interaction with unintended regions are some key character­ (1994) 244–268, https://doi.org/10.1037/1064-1297.2.3.244.
istics that are expected from epidrugs. At present, epidrugs for addiction [8] E.E. Zumbrun, J.M. Sido, P.S. Nagarkatti, M. Nagarkatti, Epigenetic regulation of
immunological alterations following prenatal exposure to marijuana
treatment is at its infancy that needs to be proven against various con­ cannabinoids and its long term consequences in offspring, J. Neuroimmune
cerns and future studies should be directed towards the collective Pharmacol. 10 (2015) 245–254, https://doi.org/10.1007/s11481-015-9586-0.

14
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

[9] M.L. Gerra, M.C. Gerra, L. Tadonio, P. Pellegrini, C. Marchesi, E. Mattfeld, methylation on DNA binding specificities of human transcription factors, Sci. (N.
G. Gerra, P. Ossola, Early parent-child interactions and substance use disorder: An Y., N. Y. ) 356 (2017), eaaj2239, https://doi.org/10.1126/science.aaj2239.
attachment perspective on a biopsychosocial entanglement, Neurosci. Biobehav. [35] A. de Mendoza, D. Poppe, S. Buckberry, J. Pflueger, C.B. Albertin, T. Daish,
Rev. 131 (2021) 560–580, https://doi.org/10.1016/j.neubiorev.2021.09.052. S. Bertrand, E. de la Calle-Mustienes, J.L. Gómez-Skarmeta, J.R. Nery, J.R. Ecker,
[10] R.G. Lewis, E. Florio, D. Punzo, E. Borrelli, The brain’s reward system in health B. Baer, C.W. Ragsdale, F. Grützner, H. Escriva, B. Venkatesh, O. Bogdanovic,
and disease, Adv. Exp. Med. Biol. 1344 (2021) 57–69, https://doi.org/10.1007/ R. Lister, The emergence of the brain non-CpG methylation system in vertebrates,
978-3-030-81147-1_4. Nat. Ecol. Evol. 5 (2021) 369–378, https://doi.org/10.1038/s41559-020-01371-
[11] I. Maze, E.J. Nestler, The epigenetic landscape of addiction, Ann. N. Y. Acad. Sci. 2.
1216 (2011) 99–113, https://doi.org/10.1111/j.1749-6632.2010.05893.x. [36] M. Doke, V. Jeganathan, J.P. McLaughlin, T. Samikkannu, HIV-1 Tat and cocaine
[12] S. Gupta, P. Kulhara, Cellular and molecular mechanisms of drug dependence: An impact mitochondrial epigenetics: effects on DNA methylation, Epigenetics 16
overview and update, Indian J. Psychiatry 49 (2007) 85–90, https://doi.org/ (2021) 980–999, https://doi.org/10.1080/15592294.2020.1834919.
10.4103/0019-5545.33253. [37] N.T. Tavares, S. Gumauskaitė, J. Lobo, C. Jerónimo, R. Henrique, DNA
[13] N.D. Volkow, M. Michaelides, R. Baler, The neuroscience of drug reward and methylation biomarkers for prediction of response to platinum-based
addiction, Physiol. Rev. 99 (2019) 2115–2140, https://doi.org/10.1152/ chemotherapy: where do we stand? Cancers 14 (2022) 2918, https://doi.org/
physrev.00014.2018. 10.3390/cancers14122918.
[14] K.M. Lattal, M.A. Wood, Epigenetics and persistent memory: implications for [38] M. Ehrlich, M.A. Gama-Sosa, L.H. Huang, R.M. Midgett, K.C. Kuo, R.A. McCune,
reconsolidation and silent extinction beyond the zero, Nat. Neurosci. 16 (2013) C. Gehrke, Amount and distribution of 5-methylcytosine in human DNA from
124–129, https://doi.org/10.1038/nn.3302. different types of tissues of cells, Nucleic Acids Res 10 (1982) 2709–2721,
[15] C.C. Wong, J. Mill, C. Fernandes, Drugs and addiction: an introduction to https://doi.org/10.1093/nar/10.8.2709.
epigenetics, Addiction 106 (2011) 480–489, https://doi.org/10.1111/j.1360- [39] R. Lister, E.A. Mukamel, J.R. Nery, M. Urich, C.A. Puddifoot, N.D. Johnson,
0443.2010.03321.x. J. Lucero, Y. Huang, A.J. Dwork, M.D. Schultz, M. Yu, J. Tonti-Filippini, H. Heyn,
[16] R.M. Bastle, J.L. Neisewander, Epigenetics and drug abuse (editors), in: W. S. Hu, J.C. Wu, A. Rao, M. Esteller, C. He, F.G. Haghighi, T.J. Sejnowski, M.
M. Meil, C.L. Ruby (Eds.), Recent advances in drug addiction research and clinical M. Behrens, J.R. Ecker, Global epigenomic reconfiguration during mammalian
applications, IntechOpen, 2016, pp. 21–45, https://doi.org/10.5772/63952 brain development, Science 341 (2013), 1237905, https://doi.org/10.1126/
(editors). science.1237905.
[17] H.W. Shen, S. Toda, K. Moussawi, A. Bouknight, D.S. Zahm, P.W. Kalivas, Altered [40] L.F. Rizzardi, P.F. Hickey, V. Rodriguez DiBlasi, R. Tryggvadóttir, C.M. Callahan,
dendritic spine plasticity in cocaine-withdrawn rats, J. Neurosci. 29 (2009) A. Idrizi, K.D. Hansen, A.P. Feinberg, Neuronal brain-region-specific DNA
2876–2884, https://doi.org/10.1523/JNEUROSCI.5638-08.2009. methylation and chromatin accessibility are associated with neuropsychiatric
[18] T.E. Robinson, B. Kolb, Structural plasticity associated with exposure to drugs of trait heritability, Nat. Neurosci. 22 (2019) 307–316, https://doi.org/10.1038/
abuse, Neuropharmacology 47 (Suppl 1) (2004) 33–46, https://doi.org/10.1016/ s41593-018-0297-8.
j.neuropharm.2004.06.025. [41] E. Lax, G. Warhaftig, D. Ohana, R. Maayan, Y. Delayahu, P. Roska, A.
[19] J.L. Cadet, C. Brannock, S. Jayanthi, I.N. Krasnova, Transcriptional and M. Ponizovsky, A. Weizman, G. Yadid, M. Szyf, A DNA methylation signature of
epigenetic substrates of methamphetamine addiction and withdrawal: evidence addiction in t cells and its reversal with DHEA intervention, Front. Mol. Neurosci.
from a long-access self-administration model in the rat, Mol. Neurobiol. 51 (2015) 11 (2018), 322, https://doi.org/10.3389/fnmol.2018.00322.
696–717, https://doi.org/10.1007/s12035-014-8776-8. [42] E. Lax, M. Szyf, The role of dna methylation in drug addiction: implications for
[20] Y.H. Huang, O.M. Schluter, Y. Dong, Silent synapses speak up, updates of the diagnostic and therapeutics, Prog. Mol. Biol. Transl. Sci. 157 (2018) 93–104,
neural rejuvenation hypothesis of drug addiction, Neuroscientist 21 (2015) https://doi.org/10.1016/bs.pmbts.2018.01.003.
451–459, https://doi.org/10.1177/1073858415579405. [43] C. Camilo, M. Maschietto, H.C. Vieira, A.C. Tahira, G.R. Gouveia, A.C. Feio Dos
[21] H. Marie, W. Morishita, X. Yu, N. Calakos, R.C. Malenka, Generation of silent Santos, A.B. Negrão, M. Ribeiro, R. Laranjeira, H. Vallada, H. Brentani, Genome-
synapses by acute in vivo expression of CaMKIV and CREB, Neuron 45 (2005) wide DNA methylation profile in the peripheral blood of cocaine and crack
741–752, https://doi.org/10.1016/j.neuron.2005.01.039. dependents, Rev. Bras. De. Psiquiatr. 41 (2019) 485–493, https://doi.org/
[22] C. Engelmann, R. Haenold, Transcriptional control of synaptic plasticity by 10.1590/1516-4446-2018-0092.
transcription factor NF-kappaB, Neural Plast. 2016 (2016), 7027949, https://doi. [44] J. Liao, R. Karnik, H. Gu, M.J. Ziller, K. Clement, A.M. Tsankov, V. Akopian, C.
org/10.1155/2016/7027949. A. Gifford, J. Donaghey, C. Galonska, R. Pop, D. Reyon, S.Q. Tsai, W. Mallard, J.
[23] A. Eagle, B. Al Masraf, A.J. Robison, Transcriptional and epigenetic regulation of K. Joung, J.L. Rinn, A. Gnirke, A. Meissner, Targeted disruption of DNMT1,
reward circuitry in drug addiction, in: T. Mary (Ed.), Neural mechanisms of DNMT3A and DNMT3B in human embryonic stem cells, Nat. Genet. 47 (2015)
addiction, Cambridge: Academic Press, 2019, pp. 23–34, https://doi.org/ 469–478, https://doi.org/10.1038/ng.3258.
10.1016/B978-0-12-812202-0.00003-8. [45] M. Gagliardi, M. Strazzullo, M.R. Matarazzo, DNMT3B Functions: Novel insights
[24] A. Bošković, O.J. Rando, Transgenerational Epigenetic Inheritance, Ann. Rev. from human disease, Front. Cell. Dev. Biol. 6 (2018), 140, https://doi.org/
Genet. 52 (2018) 21–41, https://doi.org/10.1146/annurev-genet-120417- 10.3389/fcell.2018.00140.
031404. [46] Q. Du, P.L. Luu, C. Stirzaker, S.J. Clark, Methyl-CpG-binding domain proteins:
[25] P.R. Nielsen, D. Nietlispach, H.R. Mott, J. Callaghan, A. Bannister, T. Kouzarides, readers of the epigenome, Epigenomics 7 (2015) 1051–1073, https://doi.org/
A.G. Murzin, N.V. Murzina, E.D. Laue, Structure of the HP1 chromodomain bound 10.2217/epi.15.39.
to histone H3 methylated at lysine 9, Nature 416 (2002) 103–107, https://doi. [47] S. Pol Bodetto, D. Carouge, M. Fonteneau, J.B. Dietrich, J. Zwiller, P. Anglard,
org/10.1038/nature722. Cocaine represses protein phosphatase-1Cβ through DNA methylation and
[26] E. Li, Chromatin modification and epigenetic reprogramming in mammalian Methyl-CpG Binding Protein-2 recruitment in adult rat brain,
development, Nat. Rev. Genet. 3 (2002) 662–673, https://doi.org/10.1038/ Neuropharmacology 73 (2013) 31–40, https://doi.org/10.1016/j.
nrg887. neuropharm.2013.05.005.
[27] F.M. Vassoler, S.L. White, H.D. Schmidt, G. Sadri-Vakili, R.C. Pierce, Epigenetic [48] N. Cannella, A.M.M. Oliveira, T. Hemstedt, T. Lissek, E. Buechler, H. Bading,
inheritance of a cocaine-resistance phenotype, Nat. Neurosci. 16 (2013) 42–47, R. Spanagel, Dnmt3a2 in the nucleus accumbens shell is required for
https://doi.org/10.1038/nn.3280. reinstatement of cocaine seeking, J. Neurosci. 38 (2018) 7516–7528, https://doi.
[28] S.L. White, F.M. Vassoler, H.D. Schmidt, R.C. Pierce, M.E. Wimmer, Enhanced org/10.1523/JNEUROSCI.0600-18.2018.
anxiety in the male offspring of sires that self-administered cocaine, Addict. Biol. [49] H. Wu, Y. Zhang, Reversing DNA methylation: Mechanisms genomics and
21 (2016) 802–810, https://doi.org/10.1111/adb.12258. biological functions, Cell 156 (2014) 45–68, https://doi.org/10.1016/j.
[29] M.E. Wimmer, L.A. Briand, B. Fant, L.A. Guercio, A.C. Arreola, H.D. Schmidt, cell.2013.12.019.
S. Sidoli, Y. Han, B.A. Garcia, R.C. Pierce, Paternal cocaine taking elicits [50] K.D. Rasmussen, K. Helin, Role of TET enzymes in DNA methylation,
epigenetic remodeling and memory deficits in male progeny, Mol. Psychiatry 22 development, and cancer, Genes Dev. 30 (2016) 733–750, https://doi.org/
(2017) 1641–1650, https://doi.org/10.1038/mp.2017.8. 10.1101/gad.276568.115.
[30] V. López, A.F. Fernández, M.F. Fraga, The role of 5-hydroxymethylcytosine in [51] F.Z. Jiang, W. Zheng, C. Wu, Y. Li, F. Shen, J. Liang, M. Li, J.J. Zhang, N. Sui,
development, aging and age-related diseases, Ageing Res. Rev. 37 (2017) 28–38, Double dissociation of inhibitory effects between the hippocampal TET1 and
https://doi.org/10.1016/j.arr.2017.05.002. TET3 in the acquisition of morphine self-administration in rats, Addict. Biol. 26
[31] A. Majchrzak-Celińska, A. Warych, M. Szoszkiewicz, Novel approaches to (2021), e12875, https://doi.org/10.1111/adb.12875.
epigenetic therapies: From drug combinations to epigenetic editing, Genes 12 [52] J. Feng, N. Shao, K.E. Szulwach, V. Vialou, J. Huynh, C. Zhong, T. Le,
(2021) 208, https://doi.org/10.3390/genes12020208. D. Ferguson, M.E. Cahill, Y. Li, J.W. Koo, E. Ribeiro, B. Labonte, B.M. Laitman,
[32] R.A. Irizarry, C. Ladd-Acosta, B. Wen, Z. Wu, C. Montano, P. Onyango, H. Cui, D. Estey, V. Stockman, P. Kennedy, T. Couroussé, I. Mensah, G. Turecki, K.
K. Gabo, M. Rongione, M. Webster, H. Ji, J. Potash, S. Sabunciyan, A.P. Feinberg, F. Faull, G. Ming, H. Song, G. Fan, P. Casaccia, L. Shen, P. Jin, E.J. Nestler, Role of
The human colon cancer methylome shows similar hypo- and hypermethylation Tet1 and 5-hydroxymethylcytosine in cocaine action, Nat. Neurosci. 18 (2015)
at conserved tissue-specific CpG island shores, Nat. Genet. 41 (2009) 178–186, 536–544, https://doi.org/10.1038/nn.3976.
https://doi.org/10.1038/ng.298. [53] M.A. Surani, K. Hayashi, P. Hajkova, Genetic and epigenetic regulators of
[33] L. Moore, T. Le, G. Fan, DNA Methylation and its basic function, pluripotency, Cell 128 (2007) 747–762, https://doi.org/10.1016/j.
Neuropsychopharmacology 38 (2013) 23–38, https://doi.org/10.1038/ cell.2007.02.010.
npp.2012.112. [54] S. Seisenberger, J.R. Peat, T.A. Hore, F. Santos, W. Dean, W. Reik,
[34] Y. Yin, E. Morgunova, A. Jolma, E. Kaasinen, B. Sahu, S. Khund-Sayeed, P.K. Das, Reprogramming DNA methylation in the mammalian life cycle: Building and
T. Kivioja, K. Dave, F. Zhong, K.R. Nitta, M. Taipale, A. Popov, P.A. Ginno, breaking epigenetic barriers, Philos. Trans. R. Soc. Lond. B. Biol. Sci. 368 (2013)
S. Domcke, J. Yan, D. Schübeler, C. Vinson, J. Taipale, Impact of cytosine 20110330, https://doi.org/10.1098/rstb.2011.0330.

15
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

[55] Y.A. Medvedeva, A.M. Khamis, I.V. Kulakovskiy, W. Ba-Alawi, M.S.I. Bhuyan, β clearance via the epigenetic regulation of endocytosis -related genes expression,
H. Kawaji, T. Lassmann, M. Harbers, A.R.R. Forrest, V.B. Bajic, Effects of cytosine Cell Mol. Life Sci. (15) (2019) 3005–3018, https://doi.org/10.1007/s00018-019-
methylation on transcription factor binding sites, B. M. C. Genom. 15 (2014), 03074-9.
119, https://doi.org/10.1186/1471-2164-15-119. [76] T. Maurice, F. Duclot, J. Meunier, G. Naert, L. Givalois, J. Meffre, A. Célérier,
[56] T. Bartke, M. Vermeulen, B. Xhemalce, S.C. Robson, M. Mann, T. Kouzarides, C. Jacquet, V. Copois, N. Mechti, K. Ozato, C. Gongora, Altered memory
Nucleosome interacting proteins regulated by DNA and histone methylation, Cell capacities and response to stress in p300/CBP-associated factor (PCAF) histone
143 (2010) 470–484, https://doi.org/10.1016/j.cell.2010.10.012. acetylase knockout mice, Neuropsychopharmacol 33 (2008) 1584–1602, https://
[57] X. Shi, T. Hong, K.L. Walter, M. Ewalt, E. Michishita, T. Hung, D. Carney, P. Peña, doi.org/10.1038/sj.npp.1301551.
F. Lan, M.R. Kaadige, N. Lacoste, C. Cayrou, F. Davrazou, A. Saha, B.R. Cairns, D. [77] T.J. Gould, Addiction and cognition, Addict. Sci. Clin. Pract. 5 (2010) 4–14.
E. Ayer, T.G. Kutateladze, Y. Shi, J. Côté, K.F. Chua, O. Gozani, ING2 PHD domain [78] J. Li, Y. Guo, F.A. Schroeder, R.M. Youngs, T.W. Schmidt, C. Ferris, C. Konradi,
links histone H3 lysine 4 methylation to active gene repression, Nature 442 S. Akbarian, Dopamine D2-like antagonists induce chromatin remodeling in
(2006) 96–99, https://doi.org/10.1038/nature04835. striatal neurons through cyclic AMP-protein kinase A and NMDA receptor
[58] J. Cheng, R. Blum, C. Bowman, D. Hu, A. Shilatifard, S. Shen, B.D. Dynlacht, signaling, J. Neurochem 90 (2004) 1117–1131, https://doi.org/10.1111/j.1471-
A role for H3K4 monomethylation in gene repression and partitioning of 4159.2004.02569.x.
chromatin readers, Mol. Cell 53 (2014) 979–992, https://doi.org/10.1016/j. [79] L. Ni, L. Wang, C. Yao, Z. Ni, F. Liu, C. Gong, X. Zhu, X. Yan, S.S. Watowich, D.
molcel.2014.02.032. A. Lee, S. Zhu, The histone deacetylase inhibitor valproic acid inhibits NKG2D
[59] A. la Torre, F. Lo Vecchio, A. Greco, Epigenetic mechanisms of aging and aging- expression in natural killer cells through suppression of STAT3 and HDAC3, Sci.
associated diseases, Cells 12 (2023) 1163, https://doi.org/10.3390/ Rep. 7 (2017), 45266, https://doi.org/10.1038/srep45266.
cells12081163. [80] D. Ferguson, J.W. Koo, J. Feng, E. Heller, J. Rabkin, M. Heshmati, W. Renthal,
[60] S.J.D. Tjalsma, M. Hori, Y. Sato, A. Bousard, A. Ohi, A.C. Raposo, J. Roensch, R. Neve, X. Liu, N. Shao, V. Sartorelli, L. Shen, E.J. Nestler, Essential role of SIRT1
A. Le Saux, J. Nogami, K. Maehara, T. Kujirai, T. Handa, S. Bagés-Arnal, signaling in the nucleus accumbens in cocaine and morphine action, J. Neurosci.
Y. Ohkawa, H. Kurumizaka, S.T. da Rocha, J.J. Żylicz, H. Kimura, E. Heard, 33 (2013) 16088–16098, https://doi.org/10.1523/JNEUROSCI.1284-13.2013.
H4K20me1 and H3K27me3 are concurrently loaded onto the inactive X [81] J.L. Cadet, R. Patel, S. Jayanthi, Compulsive methamphetamine taking and
chromosome but dispensable for inducing gene silencing, EMBO Rep. 22 (2021), abstinence in the presence of adverse consequences: Epigenetic and
e51989, https://doi.org/10.15252/embr.202051989. transcriptional consequences in the rat brain, Pharmacol. Biochem. Behav. 179
[61] M. Shoaib, Q. Chen, X. Shi, N. Nair, C. Prasanna, R. Yang, D. Walter, K. (2019) 98–108, https://doi.org/10.1016/j.pbb.2019.02.009.
S. Frederiksen, H. Einarsson, J.P. Svensson, C.F. Liu, K. Ekwall, M. Lerdrup, [82] J.L. Cadet, B. Ladenheim, I. Krasnova, S. Jayanthi, Differential expression of
L. Nordenskiöld, C.S. Sørensen, Histone H4 lysine 20 mono-methylation directly mRNAs coding for histone deacetylases (HDACs) in the nucleus accumbens of
facilitates chromatin openness and promotes transcription of housekeeping genes, compulsive methamphetamine takers and abstinent rats, J. Drug Alcohol Res. 5
Nat. Commun. 12 (2021), 4800, https://doi.org/10.1038/s41467-021-25051-2. (2016) 1–9, https://doi.org/10.4303/jdar/235998.
[62] E.L. Greer, Y. Shi, Histone methylation: a dynamic mark in health disease and [83] T.G. Bredfeldt, C.L. Walker, Epigenetics, in: C.A. McQueen (Ed.), Comprehensive
inheritance, Nat. Rev. Genet. 13 (2012) 343–357, https://doi.org/10.1038/ Toxicology (2nd edn), Elsevier, 2010, pp. 335–358, https://doi.org/10.1016/
nrg3173. B978-0-08-046884-6.00219-0.
[63] S. Gupta, S.Y. Kim, S. Artis, D.L. Molfese, A. Schumacher, J.D. Sweatt, R.E. Paylor, [84] T. Banerjee, D. Chakravarti, A peek into the complex realm of histone
F.D. Lubin, Histone methylation regulates memory formation, J. Neurosci. 30 phosphorylation, Mol. Cell. Biol. 31 (2011) 4858–4873, https://doi.org/10.1128/
(2010) 3589–3599, https://doi.org/10.1523/JNEUROSCI.3732-09.2010. MCB.05631-11.
[64] D. Ikegami, M. Narita, S. Imai, K. Miyashita, R. Tamura, M. Narita, S. Takagi, [85] D. Rossetto, N. Avvakumov, J. Côté, Histone phosphorylation: a chromatin
A. Yokomizo, H. Takeshima, T. Ando, K. Igarashi, J. Kanno, N. Kuzumaki, modification involved in diverse nuclear events, Epigenetics 7 (2012) 1098–1108,
T. Ushijima, T. Suzuki, Epigenetic modulation at the CCR2 gene correlates with https://doi.org/10.4161/epi.21975.
the maintenance of behavioral sensitization to methamphetamine, Addict. Biol. [86] Y. Wang, A. Singh, G. Li, S. Yue, K. Hertel, Z.J. Wang, Opioid induces increased
15 (2010) 358–361, https://doi.org/10.1111/j.1369-1600.2010.00219.x. DNA damage in prefrontal cortex and nucleus accumbens, Pharmacol. Biochem.
[65] A. Aguilar-Valles, T. Vaissière, E.M. Griggs, M.A. Mikaelsson, I.F. Takács, E. Behav. 224 (2023), 173535, https://doi.org/10.1016/j.pbb.2023.173535.
J. Young, G. Rumbaugh, C.A. Miller, Methamphetamine-associated memory is [87] J.V. Deng, Y. Wan, X. Wang, S. Cohen, W.C. Wetsel, M.E. Greenberg, P.J. Kenny,
regulated by a writer and an eraser of permissive histone methylation, Biol. N. Calakos, A.E. West, MeCP2 phosphorylation limits psychostimulant-induced
Psychiatry 76 (2014) 57–65, https://doi.org/10.1016/j.biopsych.2013.09.014. behavioral and neuronal plasticity, J. Neurosci. 34 (2014) 4519–4527, https://
[66] S. Heerboth, K. Lapinska, N. Snyder, M. Leary, S. Rollinson, S. Sarkar, Use of doi.org/10.1523/JNEUROSCI.2821-13.2014.
epigenetic drugs in disease: an overview, Genet. Epigenet. 6 (2014) 9–19, https:// [88] B.A. Alhamwe, R. Khalaila, J. Wolf, V. von Bülow, H. Harb, F. Alhamdan, C.S. Hii,
doi.org/10.4137/GEG.S12270. S.L. Prescott, A. Ferrante, H. Renz, H. Garn, D.P. Potaczek, Histone modifications
[67] N. Chessum, K. Jones, E. Pasqua, M. Tucker, Recent advances in cancer and their role in epigenetics of atopy and allergic diseases, Allergy Asthma Clin.
therapeutics, in: G. Lawton, D.R. Witty (Eds.), Progress in Medicinal Chemistry, Immunol. 14 (2018), 39, https://doi.org/10.1186/s13223-018-0259-4.
Vol. 54, Elsevier, 2015, pp. 1–63, https://doi.org/10.1016/bs. [89] J.J. Chen, D. Stermer, J.C. Tanny, Decoding histone ubiquitylation, Front. Cell
pmch.2014.11.002-. Dev. Biol. 10 (2022), 968398, https://doi.org/10.3389/fcell.2022.968398.
[68] M. Tan, H. Luo, S. Lee, F. Jin, J.S. Yang, E. Montellier, T. Buchou, Z. Cheng, [90] A.E. Hare, J.D. Parvin, Processes that regulate the ubiquitination of chromatin
S. Rousseaux, N. Rajagopal, Z. Lu, Z. Ye, Q. Zhu, J. Wysocka, Y. Ye, S. Khochbin, and chromatin-associated proteins, in: M. Summers (Ed.), Ubiquitin Proteasome
B. Ren, Y. Zhao, Identification of 67 histone marks and histone lysine System - Current insights into mechanism cellular regulation and disease,
crotonylation as a new type of histone modification, Cell 146 (2011) 1016–1028, Intechopen, 2019, pp. 1–22, https://doi.org/10.5772/intechopen.82567.
https://doi.org/10.1016/j.cell.2011.08.008. [91] P. Krumova, J.H. Weishaupt, Sumoylation in neurodegenerative diseases, Cell.
[69] G. Jiang, C. Li, M. Lu, K. Lu, H. Li, Protein lysine crotonylation: past, present, Mol. Life Sci. 70 (2013) 2123–2138, https://doi.org/10.1007/s00018-012-1158-
perspective, Cell Death Dis. 12 (2021), 703, https://doi.org/10.1038/s41419- 3.
021-03987-z. [92] N.G. Simonet, G. Rasti, A. Vaquero, The histone code and disease:
[70] A. Kumar, K.H. Choi, W. Renthal, N.M. Tsankova, D.E. Theobald, H.T. Truong, S. Posttranslational modification as potential prognostic factors for clinical
J. Russo, Q. LaPlant, T.S. Sasaki, K.N. Whistler, R.L. Neve, D.W. Self, E.J. Nestler, diagnosis, in: J.L. García-Giménez (Ed.), Epigenetic biomarkers and diagnostics,
Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity Academic Press, Cambridge, 2016, pp. 417–445, https://doi.org/10.1016/B978-
in striatum, Neuron 48 (2005) 303–314, https://doi.org/10.1016/j. 0-12-801899-6.00021-8.
neuron.2005.09.023. [93] M.D. Rubio, K. Wood, V. Haroutunian, J.H. Meador-Woodruff, Dysfunction of the
[71] L. Wang, Z. Lv, Z. Hu, J. Sheng, B. Hui, J. Sun, L. Ma, Chronic cocaine-induced H3 ubiquitin proteasome and ubiquitin-like systems in schizophrenia,
acetylation and transcriptional activation of CaMKIIalpha in the nucleus Neuropsychopharmacology 38 (2013) 1910–1920, https://doi.org/10.1038/
accumbens is critical for motivation for drug reinforcement, npp.2013.84.
Neuropsychopharmacology 35 (2010) 913–928, https://doi.org/10.1038/ [94] G. Manco, G. Lacerra, E. Porzio, G. Catara, ADP-Ribosylation post-translational
npp.2009.193. modification: An overview with a focus on rna biology and new pharmacological
[72] I. Maze, H.E. Covington, 3rd, D.M. Dietz, Q. LaPlant, W. Renthal, S.J. Russo, perspectives, Biomolecules 12 (2022) 443, https://doi.org/10.3390/
M. Mechanic, E. Mouzon, R.L. Neve, S.J. Haggarty, Y. Ren, S.C. Sampath, Y. biom12030443.
L. Hurd, P. Greengard, A. Tarakhovsky, A. Schaefer, E.J. Nestler, Essential role of [95] K.N. Scobie, D. Damez-Werno, H. Sun, N. Shao, A. Gancarz, C.H. Panganiban,
the histone methyltransferase G9a in cocaine-induced plasticity, Science 327 C. Dias, J. Koo, P. Caiafa, L. Kaufman, R.L. Neve, D.M. Dietz, L. Shen, E.J. Nestler,
(2010) 213–216, https://doi.org/10.1126/science.1179438. Essential role of polyADP-ribosylation in cocaine action, Proc. Natl. Acad. Sci. U.
[73] M. Malvaez, E. Mhillaj, D.P. Matheos, M. Palmery, M.A. Wood, CBP in the nucleus S. A. 111 (2014) 2005–2010, https://doi.org/10.1073/pnas.1319703111.
accumbens regulates cocaine-induced histone acetylation and is critical for [96] C.D. Striplin, P.W. Kalivas, Correlation between behavioral sensitization to
cocaine-associated behaviors, J. Neurosci. 31 (2011) 16941–16948, https://doi. cocaine and G protein ADP-ribosylation in the ventral tegmental area, Brain Res
org/10.1523/JNEUROSCI.2747-11.2011. 579 (1992) 181–186, https://doi.org/10.1016/0006-8993(92)90049-f.
[74] W. Renthal, A. Kumar, G. Xiao, M. Wilkinson, H.E. Covington, 3rd, I. Maze, [97] E. Lax, A. Friedman, R. Massart, R. Barnea, L. Abraham, D. Cheishvili, M. Zada,
D. Sikder, A.J. Robison, Q. LaPlant, D.M. Dietz, S.J. Russo, V. Vialou, H. Ahdoot, T. Bareli, G. Warhaftig, L. Visochek, M. Suderman, M. Cohen-Armon,
S. Chakravarty, T.J. Kodadek, A. Stack, M. Kabbaj, E.J. Nestler, Genome-wide M. Szyf, G. Yadid, PARP-1 is required for retrieval of cocaine-associated memory
analysis of chromatin regulation by cocaine reveals a novel role for sirtuins, by binding to the promoter of a novel gene encoding a putative transposase
Neuron 62 (2009) 335–348, https://doi.org/10.1016/j.neuron.2009.03.026. inhibitor, Mol. Psychiatry 22 (2017) 570–579, https://doi.org/10.1038/
[75] Z. Wang, Y. Zhao, N. Xu, S. Zhang, S. Wang, Y. Mao, Y. Zhu, B. Li, Y. Jiang, mp.2016.119.
Y. Tan, W. Xie, B.B. Yang, Y. Zhang, NEAT1 regulates neuroglial cell mediating A

16
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

[98] S. Dash, M. Balasubramaniam, T. Rana, A. Godino, E.G. Peck, J.S. Goodwin, [120] J.C. Peng, H. Lin, Beyond transposons: The epigenetic and somatic functions of
F. Villalta, E.S. Calipari, E.J. Nestler, C. Dash, J. Pandhare, Poly (ADP-ribose) the Piwi-piRNA mechanism, Curr. Opin. Cell Biol. 25 (2013) 190–194, https://
polymerase-1 (PARP-1) induction by cocaine is post-transcriptionally regulated doi.org/10.1016/j.ceb.2013.01.010.
by miR-125b, eNeuro 4 (2017), ENEURO.0089-17.2017, https://doi.org/ [121] D. Pezic, S.A. Manakov, R. Sachidanandam, A.A. Aravin, piRNA pathway targets
10.1523/ENEURO.0089-17.2017. active LINE1 elements to establish the repressive H3K9me3 mark in germ cells,
[99] S. Dash, M. Balasubramaniam, F.J. Martínez-Rivera, A. Godino, E.G. Peck, Genes Dev. 28 (2014) 1410–1428, https://doi.org/10.1101/gad.240895.114.
S. Patnaik, M. Suar, E.S. Calipari, E.J. Nestler, F. Villalta, C. Dash, J. Pandhare, [122] M. Litwin, A. Szczepańska-Buda, A. Piotrowska, P. Dzięgiel, W. Witkiewicz, The
Cocaine-regulated microRNA miR-124 controls poly (ADP-ribose) polymerase-1 meaning of PIWI proteins in cancer development, Oncol. Lett. 13 (2017)
expression in neuronal cells, Sci. Rep. 10 (2020), 11197, https://doi.org/ 3354–3362, https://doi.org/10.3892/ol.2017.5932.
10.1038/s41598-020-68144-6. [123] I.R. Henderson, S.E. Jacobsen, Epigenetic inheritance in plants, Nature 447
[100] S. Dash, C. Dash, J. Pandhare, Activation of proline metabolism maintains ATP (2007) 418–424, https://doi.org/10.1038/nature05917.
levels during cocaine-induced polyADP-ribosylation, Amino Acids 53 (2021) [124] K. Zhang, G. Ji, M. Zhao, Y. Wang, Candidate l-methionine target piRNA
1903–1915, https://doi.org/10.1007/s00726-021-03065-w. regulatory networks analysis response to cocaine-conditioned place preference in
[101] Y. Ke, J. Zhang, X. Lv, X. Zeng, X. Ba, Novel insights into PARPs in gene mice, Brain Behav. 11 (2021), e2272, https://doi.org/10.1002/brb3.2272.
expression: regulation of RNA metabolism, Cell. Mol. Life Sci. 76 (2019) [125] J.Y. Zhong, R.R. Cui, X. Lin, F. Xu, T. Zhu, F. Li, F. Wu, E. Zhou, L. Yi, L.Q. Yuan,
3283–3299, https://doi.org/10.1007/s00018-019-03120-6. Aberrant DNA methylation of synaptophysin is involved in adrenal cortisol-
[102] R. Gupte, Z. Liu, W.L. Kraus, PARPs and ADP-ribosylation: recent advances producing adenoma, Aging 11 (2019) 5232–5245, https://doi.org/10.18632/
linking molecular functions to biological outcomes, Genes Dev. 31 (2017) aging.102119.
101–126, https://doi.org/10.1101/gad.291518.116. [126] E. Dai, X. Yu, Y. Zhang, F. Meng, S. Wang, X. Liu, D. Liu, J. Wang, X. Li, W. Jiang,
[103] M.P. Marjanović, K. Crawford, I. Ahel, PARP, transcription and chromatin EpimiR: a database of curated mutual regulation between miRNAs and epigenetic
modeling, Semin. Cell Dev. Biol. 63 (2017) 102–113, https://doi.org/10.1016/j. modifications, Database (2014) bau023, https://doi.org/10.1093/database/
semcdb.2016.09.014. bau023.
[104] R. Krishnakumar, W.L. Kraus, PARP-1 regulates chromatin structure and [127] J.A. Hollander, H.I. Im, A.L. Amelio, J. Kocerha, P. Bali, Q. Lu, D. Willoughby,
transcription through a KDM5B-dependent pathway, Mol. Cell. 39 (2010) C. Wahlestedt, M.D. Conkright, P.J. Kenny, Striatal microRNA controls cocaine
736–749, https://doi.org/10.1016/j.molcel.2010.08.014. intake through CREB signalling, Nature 466 (2010) 197–202, https://doi.org/
[105] L.A. Farrelly, R.E. Thompson, S. Zhao, A.E. Lepack, Y. Lyu, N.V. Bhanu, B. Zhang, 10.1038/nature09202.
Y.E. Loh, A. Ramakrishnan, K.C. Vadodaria, K.J. Heard, G. Erikson, T. Nakadai, R. [128] H.I. Im, J.A. Hollander, P. Bali, P.J. Kenny, MeCP2 controls BDNF expression and
M. Bastle, B.J. Lukasak, H. Zebroski 3rd, N. Alenina, M. Bader, O. Berton, R. cocaine intake through homeostatic interactions with microRNA-212, Nat.
G. Roeder, H. Molina, F.H. Gage, L. Shen, B.A. Garcia, H. Li, T.W. Muir, I. Maze, Neurosci. 13 (2010) 1120–1127, https://doi.org/10.1038/nn.2615.
Histone serotonylation is a permissive modification that enhances TFIID binding [129] A. Schaefer, H.I. Im, M.T. Venø, C.D. Fowler, A. Min, A. Intrator, J. Kjems, P.
to H3K4me3, Nature 567 (2019) 535–539, https://doi.org/10.1038/s41586-019- J. Kenny, D. O’Carroll, P. Greengard, Argonaute 2 in dopamine 2 receptor-
1024-7. expressing neurons regulates cocaine addiction, J. Exp. Med. 207 (2010)
[106] J. Zhao, W. Chen, Y. Pan, Y. Zhang, H. Sun, H. Wang, F. Yang, Y. Liu, N. Shen, 1843–1851, https://doi.org/10.1084/jem.20100451.
X. Zhang, X. Mo, J. Zang, Structural insights into the recognition of histone H3Q5 [130] V. Chandrasekar, J.L. Dreyer, microRNAs miR-124, let-7d and miR-181a regulate
serotonylation by WDR5, Sci. Adv. 7 (2021), eabf4291, https://doi.org/10.1126/ cocaine-induced plasticity, Mol. Cell Neurosci. 42 (2009) 350–362, https://doi.
sciadv.abf4291. org/10.1016/j.mcn.2009.08.009.
[107] A.E. Lepack, C.T. Werner, A.F. Stewart, S.L. Fulton, P. Zhong, L.A. Farrelly, A.C. [131] R. Saba, P.H. Störchel, A. Aksoy-Aksel, F. Kepura, G. Lippi, T.D. Plant, G.
W. Smith, A. Ramakrishnan, Y. Lyu, R.M. Bastle, J.A. Martin, S. Mitra, R. M. Schratt, Dopamine-regulated microRNA MiR-181a controls GluA2 surface
M. O’Connor, Z.J. Wang, H. Molina, G. Turecki, L. Shen, Z. Yan, E.S. Calipari, D. expression in hippocampal neurons, Mol. Cell Biol. 32 (2012) 619–632, https://
M. Dietz, P.J. Kenny, I. Maze, Dopaminylation of histone H3 in ventral tegmental doi.org/10.1128/MCB.05896-11.
area regulates cocaine seeking, Science 368 (2020) 197–201, https://doi.org/ [132] J.E. Eipper-Mains, D.D. Kiraly, D. Palakodeti, R.E. Mains, B.A. Eipper, B.
10.1126/science.aaw8806. R. Graveley, MicroRNA-Seq reveals cocaine-regulated expression of striatal
[108] K. Blum, M.S. Gold, J.L. Cadet, D. Baron, A. Bowirrat, P.K. Thanos, R. Brewer, R. microRNAs, RNA 17 (2011) 1529–1543, https://doi.org/10.1261/rna.2775511.
D. Badgaiyan, M.C. Gondré-Lewis, Dopaminylation in psychostimulant use [133] F.M. Sanchez-Simon, X.X. Zhang, H.H. Loh, P.Y. Law, R.E. Rodriguez, Morphine
disorder protects against psychostimulant seeking behavior by normalizing regulates dopaminergic neuron differentiation via miR-133b, Mol. Pharmacol. 78
Nucleus Accumbens (NAc) Dopamine expression, Curr. Psychopharmacol. 11 (2010) 935–942, https://doi.org/10.1124/mol.110.066837.
(2022) 11–17, https://doi.org/10.2174/2211556009666210108112737. [134] R.R. Kanherkar, N. Bhatia-Dey, A.B. Csoka, Epigenetics across the human lifespan,
[109] A. Ntorla, J.R. Burgoyne, The regulation and function of histone crotonylation, Front. Cell. Dev. Biol. 2 (2014) 49, https://doi.org/10.3389/fcell.2014.00049.
Front. Cell Dev. Biol. 9 (2021), 624914, https://doi.org/10.3389/ [135] V.S. Knopik, M.A. Maccani, S. Francazio, J.E. McGeary, The epigenetics of
fcell.2021.624914. maternal cigarette smoking during pregnancy and effects on child development,
[110] E. Montellier, F. Boussouar, S. Rousseaux, K. Zhang, T. Buchou, F. Fenaille, Dev. Psychopathol. 24 (2012) 1377–1390, https://doi.org/10.1017/
H. Shiota, A. Debernardi, P. Héry, S. Curtet, M. Jamshidikia, S. Barral, H. Holota, S0954579412000776.
A. Bergon, F. Lopez, P. Guardiola, K. Pernet, J. Imbert, C. Petosa, M. Tan, Y. Zhao, [136] C.R. Zhang, M.F. Ho, M.C.S. Vega, T.H. Burne, S. Chong, Prenatal ethanol
M. Gérard, S. Khochbin, Chromatin-to-nucleoprotamine transition is controlled exposure alters adult hippocampal VGLUT2 expression with concomitant changes
by the histone H2B variant TH2B, Genes Dev. 27 (2013) 1680–1692, https://doi. in promoter DNA methylation H3K4 trimethylation and miR-467b-5p levels,
org/10.1101/gad.220095.113. Epigenetics Chromatin 8 (2015), 40, https://doi.org/10.1186/s13072-015-0032-
[111] K. Li, Z. Wang, Histone crotonylation-centric gene regulation, Epigenetics 6.
Chromatin 14 (2021), 10, https://doi.org/10.1186/s13072-021-00385-9. [137] M. Suter, A. Abramovici, L. Showalter, L, M. Hu, C.D. Shope, M. Varner,
[112] X. Xiong, T. Panchenko, S. Yang, S. Zhao, P. Yan, W. Zhang, W. Xie, Y. Li, Y. Zhao, K. Aagaard-Tillery, In utero tobacco exposure epigenetically modifies placental
C.D. Allis, H. Li, Selective recognition of histone crotonylation by double PHD CYP1A1 expression, Metabolism 59 (2010) 1481–1490, https://doi.org/10.1016/
fingers of MOZ and DPF2, Nat. Chem. Biol. 12 (2016) 1111–1118, https://doi. j.metabol.2010.01.013.
org/10.1038/nchembio.2218. [138] P. Biliński, A. Wojtyła, L. Kapka-Skrzypczak, R. Chwedorowicz, M. Cyranka,
[113] Y. Zou, X.H. Bai, L.C. Kong, F.F. Xu, T.Y. Ding, P.F. Zhang, F.L. Dong, Y.J. Ling, B. T. Studziński, Epigenetic regulation in drug addiction, Ann. Agric. Environ. Med.
C. Jiang, Involvement of histone lysine crotonylation in the regulation of nerve- 19 (2012) 491–496.
injury-induced neuropathic pain, Front. Immunol. 13 (2022), 885685, https:// [139] R.R. Campbell, E.A. Kramár, L. Pham, J.H. Beardwood, A.S. Augustynski, A.
doi.org/10.3389/fimmu.2022.885685. J. López, O.S. Chitnis, G. Delima, J. Banihani, D.P. Matheos, M.A. Wood, HDAC3
[114] C.M. Cahill, A.M. Taylor, Neuroinflammation-a co-occurring phenomenon linking Activity within the nucleus accumbens regulates cocaine-induced plasticity and
chronic pain and opioid dependence, Curr. Opin. Behav. Sci. 13 (2017) 171–177, behavior in a cell-type-specific manner, J. Neurosci. 41 (2021) 2814–2827,
https://doi.org/10.1016/j.cobeha.2016.12.003. https://doi.org/10.1523/JNEUROSCI.2829-20.2021.
[115] G.C. Sartor, G. St Laurent 3rd, C. Wahlestedt, The emerging role of non-coding [140] L. Host, J.B. Dietrich, D. Carouge, D. Aunis, J. Zwiller, Cocaine self-administration
RNAs in drug addiction, Front. Genet. 3 (2012) 106, https://doi.org/10.3389/ alters the expression of chromatin-remodelling proteins; modulation by histone
fgene.2012.00106. deacetylase inhibition, J. Psychopharmacol. 25 (2011) 222–229, https://doi.org/
[116] C. Wang, L. Wang, Y. Ding, X. Lu, G. Zhang, J. Yang, H. Zheng, H. Wang, Y. Jiang, 10.1177/0269881109348173.
L. Xu, LncRNA structural characteristics in epigenetic regulation, Int. J. Mol. Sci. [141] J.M. Levenson, T.L. Roth, F.D. Lubin, C.A. Miller, I.C. Huang, P. Desai, L.
18 (2017) 2659, https://doi.org/10.3390/ijms18122659. M. Malone, J.D. Sweatt, Evidence that DNA (cytosine-5) methyltransferase
[117] D.N. Albertson, C.J. Schmidt, G. Kapatos, M.J. Bannon, Distinctive profiles of regulates synaptic plasticity in the hippocampus, J. Biol. Chem. 281 (2006)
gene expression in the human nucleus accumbens associated with cocaine and 15763–15773, https://doi.org/10.1074/jbc.M511767200.
heroin abuse, Neuropsychopharmacology 31 (2006) 2304–2312, https://doi.org/ [142] A. Finegersh, G.E. Homanics, Acute ethanol alters multiple histone modifications
10.1038/sj.npp.1301089. at model gene promoters in the cerebral cortex, Alcohol. Clin. Exp. Res. 38 (2014)
[118] S.K. Michelhaugh, L. Lipovich, J. Blythe, H. Jia, G. Kapatos, M.J. Bannon, Mining 1865–1873, https://doi.org/10.1111/acer.12465.
Affymetrix microarray data for long non-coding RNAs: altered expression in the [143] S. Zakhari, Alcohol metabolism and epigenetics changes, Alcohol Res 35 (2013)
nucleus accumbens of heroin abusers, J. Neurochem. 116 (2011) 459–466, 6–16.
https://doi.org/10.1111/j.1471-4159.2010.07126.x. [144] J.C. Peng, H. Lin, Beyond transposons: The epigenetic and somatic functions of
[119] D.A. Nielsen, A. Utrankar, J.A. Reyes, D.D. Simons, T.R. Kosten, Epigenetics of the Piwi-piRNA mechanism, Curr. Opin. Cell Biol. 25 (2013) 190–194, https://
drug abuse: predisposition or response, Pharmacogenomics 13 (2012) doi.org/10.1016/j.ceb.2013.01.010.
1149–1160, https://doi.org/10.2217/pgs.12.94.

17
A.S. Koijam et al. Biomedicine & Pharmacotherapy 170 (2024) 115951

[145] D. Bonsch, B. Lenz, U. Reulbach, J. Kornhuber, S. Bleich, Homocysteine [164] M.S. Reid, V. Thakkar, Valproate treatment and cocaine cue reactivity in cocaine
associated genomic DNA hypermethylation in patients with chronic alcoholism, dependent individuals, Drug Alcohol Depend. 102 (2009) 144–150, https://doi.
J. Neural Transm. 111 (2004) 1611–1616, https://doi.org/10.1007/s00702-004- org/10.1016/j.drugalcdep.2009.02.010.
0232-x. [165] R. Coccurello, A. Caprioli, O. Ghirardi, A. Virmani, Valproate and acetyl-L-
[146] B. Ruggeri, C. Nymberg, E. Vuoksimaa, A. Lourdusamy, C.P. Wong, F. carnitine prevent methamphetamine-induced behavioral sensitization in mice,
M. Carvalho, T. Jia, A. Cattrell, C. Macare, T. Banaschewski, G.J. Barker, A. Ann. N. Y. Acad. Sci. (2007) 260–275, https://doi.org/10.1196/annals.1403.019.
L. Bokde, U. Bromberg, C. Büchel, P.J. Conrod, M. Fauth-Bühler, H. Flor, [166] M. Malvaez, S.C. McQuown, G.A. Rogge, M. Astarabadi, V. Jacques, S. Carreiro, J.
V. Frouin, J. Gallinat, H. Garavan, P. Gowland, A. Heinz, B. Ittermann, J. R. Rusche, M.A. Wood, HDAC3-selective inhibitor enhances extinction of cocaine-
L. Martinot, F. Nees, Z. Pausova, T. Paus, M. Rietschel, T. Robbins, M.N. Smolka, seeking behavior in a persistent manner, Proc. Natl. Acad. Sci. U. S. A. 110 (2013)
R. Spanagel, G. Bakalkin, J. Mill, W.H. Sommer, R.J. Rose, J. Yan, F. Aliev, 2647–2652, https://doi.org/10.1073/pnas.1213364110.
D. Dick, J. Kaprio, S. Desrivières, G. Schumann, IMAGEN Consortium, Association [167] C.J. Peter, S. Akbarian, Balancing histone methylation activities in psychiatric
of protein phosphatase PPM1G with alcohol use disorder and brain activity disorders, Trends Mol. Med. 17 (2011) 372–379, https://doi.org/10.1016/j.
during behavioral control in a genome-wide methylation analysis, 543-452, Am. molmed.2011.02.003.
J. Psychiatry 172 (2015) (2015), https://doi.org/10.1176/appi. [168] D. Kringel, S. Malkusch, J. Lötsch, Drugs and epigenetic molecular functions. A
ajp.2014.14030382. pharmacological data scientometric analysis, Int. J. Mol. Sci. 22 (2021) 7250,
[147] R.A. Philibert, T.D. Gunter, S.R. Beach, G.H. Brody, A. Madan, MAOA methylation https://doi.org/10.3390/ijms22147250.
is associated with nicotine and alcohol dependence in women, Am. J. Med. Genet. [169] E.A. Heller, P.J. Hamilton, D.D. Burek, S.I. Lombroso, C.J. Pena, R.L. Neve, E.
B Neuropsychiatr. Genet. 147B (2008) 565–570, https://doi.org/10.1002/ajmg. J. Nestler, Targeted epigenetic remodeling of the Cdk5 gene in nucleus
b.30778. accumbens regulates cocaine- and stress-evoked behaviour, J. Neurosci. 36
[148] H. Liu, Y. Zhou, S.E. Boggs, S.A. Belinsky, J. Liu, Cigarette smoke induces (2016) 4690–4697, https://doi.org/10.1523/JNEUROSCI.0013-16.2016.
demethylation of prometastatic oncogene synuclein-gamma in lung cancer cells [170] T. Gaj, C.A. Gersbach, C.F. Barbas 3rd, ZFN, TALEN, and CRISPR/Cas-based
by downregulation of DNMT3B, Oncogene 26 (2007) 5900–5910, https://doi. methods for genome engineering, Trends Biotechnol. 31 (2013) 397–405, https://
org/10.1038/sj.onc.1210400. doi.org/10.1016/j.tibtech.2013.04.004.
[149] A. Godino, S. Jayanthi, J.L. Cadet, Epigenetic landscape of amphetamine and [171] Y. Lei, Y.H. Huang, M.A. Goodell, DNA methylation and de-methylation using
methamphetamine addiction in rodents, Epigenetics 10 (2015) 574–580, https:// hybrid site-targeting proteins, Genome Biol. 19 (2018), 187, https://doi.org/
doi.org/10.1080/15592294.2015.1055441. 10.1186/s13059-018-1566-2.
[150] S. Jayanthi, M.T. McCoy, B. Chen, J.P. Britt, S. Kourrich, H.J. Yau, B. Ladenheim, [172] M. Holkers, I. Maggio, J. Liu, J.M. Janssen, F. Miselli, C. Mussolino, A. Recchia,
I.N. Krasnova, A. Bonci, J.L. Cadet, Methamphetamine downregulates striatal T. Cathomen, M.A. Gonçalves, Differential integrity of TALE nuclease genes
glutamate receptors via diverse epigenetic mechanisms, Biol. Psychiatry 76 following adenoviral and lentiviral vector gene transfer into human cells, Nucleic
(2014) 47–56, https://doi.org/10.1016/j.biopsych.2013.09.034. Acids Res 41 (2013), e63, https://doi.org/10.1093/nar/gks1446.
[151] D. De Sa Nogueira, K. Merienne, K. Befort, Neuroepigenetics and addictive [173] F. Maroufi, A. Maali, M. Abdollahpour-Alitappeh, M.H. Ahmadi, M. Azad,
behaviors: Where do we stand? Neurosci. Biobehav. Rev. 106 (2019) 58–72, CRISPR-mediated modification of DNA methylation pattern in the new era of
https://doi.org/10.1016/j.neubiorev.2018.08.018. cancer therapy, Epigenomics 12 (2020) 1845–1859, https://doi.org/10.2217/epi-
[152] D. Govorko, R.A. Bekdash, C. Zhang, D.K. Sarkar, Male germline transmits fetal 2020-0110.
alcohol adverse effect on hypothalamic proopiomelanocortin gene across [174] R. Shayevitch, D. Askayo, I. Keydar, G. Ast, The importance of DNA methylation
generations, Biol. Psychiatry 72 (2012) 378–388, https://doi.org/10.1016/j. of exons on alternative splicing, RNA 24 (2018) 1351–1362, https://doi.org/
biopsych.2012.04.006. 10.1261/rna.064865.117.
[153] F.M. Vassoler, D.J. Oliver, C. Wyse, A. Blau, M. Shtutman, J.R. Turner, E. [175] S.J. Xu, S.I. Lombroso, D.K. Fischer, M.D. Carpenter, D.M. Marchione, P.
M. Byrnes, Transgenerational attenuation of opioid self-administration as a J. Hamilton, C.J. Lim, R.L. Neve, B.A. Garcia, M.E. Wimmer, R.C. Pierce, E.
consequence of adolescent morphine exposure, Neuropharmacology 113 (2017) A. Heller, Chromatin-mediated alternative splicing regulates cocaine-reward
271–280, https://doi.org/10.1016/j.neuropharm.2016.10.006. behaviour, Neuron 109 (2021) 2943–2966.e8, https://doi.org/10.1016/j.
[154] NIDA, Treatment and Recovery. https://nida.nih.gov/publications/drugs-brains- neuron.2021.08.008.
behavior-science-addiction/treatment-recovery, 2023, (accessed on November 12 [176] M.D. Carpenter, Q. Hu, A.M. Bond, S.I. Lombroso, K.S. Czarnecki, C.J. Lim,
2023). H. Song, M.E. Wimmer, R.C. Pierce, E.A. Heller, Nr4a1 suppresses cocaine-
[155] A.T. McLellan, D.C. Lewis, C.P. O’Brien, H.D. Kleber, Drug dependence, a chronic induced behavior via epigenetic regulation of homeostatic target genes, Nat.
medical illness: implications for treatment, insurance, and outcomes evaluation, Commun. 11 (2020), 504, https://doi.org/10.1038/s41467-020-14331-y.
JAMA 284 (2000) 1689–1695, https://doi.org/10.1001/jama.284.13.1689. [177] C.D. Teague, J.A. Picone, W.J. Wright, C.J. Browne, G.M. Silva, R. Futamura,
[156] R. Massart, R. Barnea, Y. Dikshtein, M. Suderman, O. Meir, M. Hallett, A. Minier-Toribio, M.E. Estill, A. Ramakrishnan, F.J. Martinez-Rivera, A. Godino,
P. Kennedy, E.J. Nestler, M. Szyf, G. Yadid, Role of DNA methylation in the E.M. Parise, K.H. Schmidt, N.V. Pulido, Z.S. Lorsch, J.H. Kim, L. Shen, R.L. Neve,
nucleus accumbens in incubation of cocaine craving, J. Neurosci. 35 (2015) Y. Dong, E.J. Nestler, P.J. Hamilton, CREB binding at the Zfp189 promoter within
8042–8058, https://doi.org/10.1523/JNEUROSCI.3053-14.2015. medium spiny neuron subtypes differentially regulates behavioral and
[157] W. Tian, J. Wang, K. Zhang, H. Teng, C. Li, M. Szyf, Z.S. Sun, M. Zhao, physiological adaptations over the course of cocaine use, Biol. Psychiatry 93
Demethylation of c-MYB binding site mediates upregulation of Bdnf IV in cocaine- (2023) 502–511, https://doi.org/10.1016/j.biopsych.2022.07.022.
conditioned place preference, Sci. Rep. 6 (2016), 22087, https://doi.org/ [178] J.G. Doench, N. Fusi, M. Sullender, M. Hegde, E.W. Vaimberg, K.F. Donovan,
10.1038/srep22087. I. Smith, Z. Tothova, C. Wilen, R. Orchard, H.W. Virgin, J. Listgarten, D.E. Root,
[158] K.N. Wright, F. Hollis, F. Duclot, A.M. Dossat, C.E. Strong, T.C. Francis, R. Mercer, Optimized sgRNA design to maximize activity and minimize off-target effects of
J. Feng, D.M. Dietz, M.K. Lobo, E.J. Nestler, M. Kabbaj, Methyl supplementation CRISPR-Cas9, Nat. Biotechnol. 34 (2016) 184–191, https://doi.org/10.1038/
attenuates cocaine-seeking behaviors and cocaine-induced c-Fos activation in a nbt.3437.
DNA methylation-dependent manner, J. Neurosci. 35 (2015) 8948–8958, https:// [179] Y. Zou, X. Sun, Q. Yang, M. Zheng, O. Shimoni, W. Ruan, Y. Wang, D. Zhang,
doi.org/10.1523/JNEUROSCI.5227-14.2015. J. Yin, X. Huang, W. Tao, J.B. Park, X.J. Liang, K.W. Leong, B. Shi, Blood-brain
[159] M. Montalvo-Casimiro, R. González-Barrios, M.A. Meraz-Rodriguez, V.T. Juárez- barrier-penetrating single CRISPR-Cas9 nanocapsules for effective and safe
González, C. Arriaga-Canon, L.A. Herrera, Epidrug repurposing: Discovering new glioblastoma gene therapy, Sci. Adv. 8 (2022), eabm8011, https://doi.org/
faces of old acquaintances in cancer therapy, Front. Oncol. 10 (2020), 605386, 10.1126/sciadv.abm8011.
https://doi.org/10.3389/fonc.2020.605386. [180] A.R. De Lera, A. Ganesan, Two-hit wonders: the expanding universe of
[160] E. Dong, M. Nelson, D.R. Grayson, E. Costa, A. Guidotti, Clozapine and sulpiride multitargeting epigenetic agents, Curr. Opin. Chem. Biol. 57 (2020) 135–154,
but not haloperidol or olanzapine activate brain DNA demethylation, Proc. Natl. https://doi.org/10.1016/j.cbpa.2020.05.009.
Acad. Sci. U. S. A. 105 (2008) 13614–13619, https://doi.org/10.1073/ [181] D. Tomaselli, A. Lucidi, D. Rotili, A. Mai, Epigenetic polypharmacology: a new
pnas.0805493105. frontier for epi-drug discovery, Med. Res. Rev. 40 (2020) 190–244, https://doi.
[161] V. Warnault, E. Darcq, A. Levine, S. Barak, D. Ron, Chromatin remodeling-a novel org/10.1002/med.21600.
strategy to control excessive alcohol drinking, Transl. Psychiatry 3 (2013), e231, [182] S.G. Shah, T. Mandloi, P. Kunte, A. Natu, M. Rashid, D. Reddy, N. Gadewal,
https://doi.org/10.1038/tp.2013.4. S. Gupta, HISTome2: a database of histone proteins, modifiers for multiple
[162] P. Romieu, L. Host, S. Gobaille, G. Sandner, D. Aunis, J. Zwiller, Histone organisms and epidrugs, Epigenetics Chromatin 13 (2020), 31, https://doi.org/
deacetylase inhibitors decrease cocaine but not sucrose self-administration in rats, 10.1186/s13072-020-00354-8.
J. Neurosci. 28 (2008) 9342–9348, https://doi.org/10.1523/JNEUROSCI.0379- [183] R. Guo, J. Li, J. Hu, Q. Fu, Y. Yan, S. Xu, X. Wang, F. Jiao, Combination of
08.2008. epidrugs with immune checkpoint inhibitors in cancer immunotherapy: From
[163] M.A. Dos Santos, S.S. Júnior, Escudeiro, G.S. Vasconcelos, N.C.B. Matos, M.R. theory to therapy, Int. Immunopharmacol. (2023), 110417, https://doi.org/
M. de Souza, M.C.A. Patrocínio, L.P. Dantas, D. Macêdo, S.M.M. Vasconcelos, The 10.1016/j.intimp.2023.110417.
interplay between ventro striatal BDNF levels and the effects of valproic acid on [184] M.G. Gladkova, E. Leidmaa, E.A. Anderzhanova, Epidrugs in the therapy of
the acquisition of ethanol-induced conditioned place preference in mice, central nervous system disorders: a way to drive on? Cells 12 (2023) 1464,
Neurosci. Lett. 660 (2017) 86–89, https://doi.org/10.1016/j.neulet.2017.09.009. https://doi.org/10.3390/cells12111464.

18

You might also like