Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

C H A P T E R

17
Alcohol and the Brain
An Epigenetic Viewpoint
Ketan Marballi, Igor Ponomarev, R. Dayne Mayfield,
and R. Adron Harris
Waggoner Center for Alcohol and Addiction Research, University of Texas–Austin, USA

O U T L I N E

General Introduction 349 Alcohol and DNA Methylation 352


Alcohol and Histone Modification 354
Epigenetics: A General Overview 349
Alcohol and microRNAs 355
Epigenetic Modifications:
Summary and Future Directions 355
An Overview 350
DNA Methylation 350 Acknowledgments 356
Histone Modifications 351
Noncoding RNAs 352 References 356

Alcohol, Epigenetics, and Gene Expression:


An Overview 352

GENERAL INTRODUCTION studies. The brain is one of the few organs in which
changes in epigenetic status are not static, constantly
Alcoholism is a serious health problem worldwide. changing in response to different stimuli, including
It is a significant cause of chronic disease and injury, changes in environment, exposure to drugs, and syn-
and according to the World Health Organization it aptic activity (Crepaldi & Riccio, 2009). The aim of this
accounts for 4% of deaths worldwide every year (Yahn, review is to summarize the different epigenetic modifi-
Watterson, & Olive, 2013). Data from the Centers of cations in the brain that have been shown to play a role
Disease Control and Prevention have shown that alcohol in alcohol dependence.
is the third lifestyle-related cause of death in the United
States, and unhealthy alcohol habits strongly correlate
with substantial increases in morbidity (Lotfipour et al., EPIGENETICS: A GENERAL OVERVIEW
2013). Numerous research studies have shown that alco-
holism is a neurobiological disorder involving multiple The terms epigenetics/epigenome usually refer to
neurotransmitter circuitries, including the dopamine, potentially heritable changes in the DNA that occur
serotonin, glutamate, and GABA systems (Hillemacher, without modifying the actual DNA sequence, in contrast
2011). Epigenetic modifications such as DNA methyla- to mutations. These modifications take the form of dif-
tion influence expression of candidate genes in alco- ferent chemical groups that are added to the DNA or to
holism (Hillemacher, 2011). Chronic alcohol usage can the histone proteins that the DNA surrounds. Together,
cause global changes in gene expression, particularly in DNA and histones form chromatin that regulates gene
the brain, as supported by different in vitro and in vivo expression. Chromatin is made up of units called

Neurobiology of Alcohol Dependence



http://dx.doi.org/10.1016/B978-0-12-405941-2.00017-1
349 © 2014 Elsevier Inc. All rights reserved.
350 17.  Alcohol and the Brain

FIGURE 17.1  An overview of chromatin structure and epigenetic modifications. The cartoon shows an overview of the hierarchy of chroma-
tin structure, starting with a whole chromosome that is made up of the histone octamer, surrounded by linker DNA. Amino acid residues on
histones such as lysine act as substrates for the addition of different epigenetic modifications exemplified by acetylation and methylation. DNA
can be modified by the addition of methyl groups on cytosine nucleotides. These modifications will interact to ultimately affect gene expression.

nucleosomes, consisting of a histone core made up of a in the zygote and embryo, and also during germ cell lin-
complex of eight histone proteins and approximately 147 eage specification and organ-specific cell differentiation
base pairs of DNA that wrap around this complex. The (Cortessis et al., 2012).
octamer consists of two units each of histones H2A, H2B, Changes in environmental exposure, such as physical
H3, and H4, and histone H1 acts as a linker histone along activity, toxic agents, changes in diet, and social stressors
with the DNA. The histone core proteins have globular can all influence epigenetic regulation (Fleisch, Wright,
carboxy or C-terminal domains, whereas the amino ter- & Baccarelli, 2012). Epigenetics has been heralded as a
minal or N-terminal domains are more flexible and pro- mechanism that may be able to elucidate the etiology of
trude out from the nucleosome. The N-terminal region complex diseases such as obesity, diabetes, schizophre-
consists of amino acids such as lysine and arginine. Most nia, and disorders of addiction such as alcoholism, for
of the chemical epigenetic modifications occur in this which changes in DNA sequence alone fail to explain
region, including acetylation and methylation (Figure susceptibility and pathology (Fleisch et al., 2012). In
17.1). These modifications act as regulators of chroma- order to understand the role of epigenetics in alcohol-
tin accessibility, essentially impacting how amenable the ism, it is necessary to first review existing knowledge of
chromatin is to the process of transcription. They also known epigenetic modifications.
act as docking sites for other protein complexes that
can recognize these chemical groups and recruit other
chromatin modifiers (Dawson & Kouzarides, 2012). EPIGENETIC MODIFICATIONS:
These modifications can cause epigenetic changes that AN OVERVIEW
may impact the genome (the entirety of genetic material,
namely DNA) as well as the phenome (the sum total of Epigenetic control of gene expression can be regulated
expressed traits and characteristics). Epigenetic changes by two major pathways: through chemical modifications
control functions ranging from basic developmental such as DNA methylation or histone modifications, and
phenomena, such as regulating cell determination and via novel epigenetic regulators such as small noncoding
differentiation in both dividing as well as nondividing RNAs, all of which can alter gene transcription.
cells, to those that impact cellular processes on a global
level, including the interactions of environmental and
physiological conditions on the organism (Jablonka, DNA Methylation
2012). Epigenetic modifications can occur as early as The most well-characterized epigenetic DNA modi-
fertilization (when there is extensive removal of methyl fication is methylation, which usually occurs at the 5’
groups in paternal DNA), followed by similar changes position of cytosine residues (Nelson & Monteggia, 2011)

III.  GENE AND BEHAVIOR


Epigenetic Modifications: An Overview 351
located at CpG sites in the vertebrate genome. These corresponds with transcription activation mediated by
methylated cytosines can occur in CpG-enriched por- histone acetyl transferases (HATs), whereas deacetylation
tions of the genome and are referred to as CpG islands. corresponds with transcriptional repression carried out by
It has been suggested that CpG islands occur at promoter histone deacetylases (HDACs). Five families of HATs are
sites on 50% of the total genes present in the genome known, including HAT1, Gcn5/PCAF, MYST, p300 /CBP
(Hamilton, 2011). DNA methylation is believed to occur (Creb-Binding Protein), and Rtt109 (Yuan & Marmorstein,
at centromeres, telomeres, and repeat sequences and in 2013). Of these, the role of CBP has been well studied in
X-chromosome inactivation (Dawson & Kouzarides, the brain, where CBP+/- mice or mice that lack its homo-
2012). Methylation is controlled by enzymes termed logue p300 show deficits in memory formation and object
DNA methyltransferases (DNMTs). There are three major recognition (Chan & La Thangue, 2001; Oliveira, Wood,
DNMTs in mammals. DNMT1 is a maintenance methyl- McDonough, & Abel, 2007). HATs usually function as
ase that recognizes hemi-methylated DNA sequences and transcriptional coactivators and are recruited by transcrip-
methylates newly synthesized cytosines on the daughter tion factors serving in multi-subunit chromatin complex
strand after replication or repair has occurred. On the other regulation (Ellis, Atadja, & Johnstone, 2009). Antagonistic
hand, DNMT 3a, 3b are de novo methylases, which are to the action of HATs, histone deacetylation is usually
involved in establishing methylation signatures at desig- associated with a reduction in gene expression. HDACs
nated spots in the genome and are particularly important are divided into four families: class I (HDACs1–3 and 8),
during embryogenesis (Dawson & Kouzarides, 2012). class II (HDACs 4–10), class III (sirtuins 1–7), and class
DNA methylation acts as a signal for the recruitment of IV (HDAC 11). Class I HDACs are primarily localized
methyl binding proteins such as methyl CpG–binding in the nucleus, whereas Class II can localize to either the
protein 2 (MeCP2) and methyl binding domain–contain- nucleus or cytoplasm (Starkman, Sakharkar, & Pandey,
ing protein (MBD1). MBD1 contains an 80kDa methyl 2012). Both families seem to have a tissue-specific pattern
CpG–binding domain (MBD) that is required for its chro- of expression (Mandrekar, 2011). Class III HDACs require
mosomal binding (Nan, Meehan, & Bird, 1993; Nan, Tate, NAD, whereas Classes I, II, and IV require Zn+2 (Kelly
Li, & Bird, 1996). In general, hypermethylation of genes & Marks 2005; Lee, Kim, Kummar, Giaccone, & Trepel,
corresponds to transcription repression and is believed 2008). Altering HDAC expression has a significant impact
to be mediated by different methyl-binding proteins on brain function. For example, abrogating HDAC3 in the
(such as MeCP2, MBD1, MBD2, and MBD3) interacting dorsal hippocampus caused long term memory enhance-
with the hypermethylated DNA. One way these pro- ment, upregulation of histone H4K8 acetylation, and
teins can regulate gene expression is by binding to RNA increased expression of immediate early genes c-fos and
polymerase, transcription factors to control initiation/ Nr4a2 (McQuown et al., 2011). HDAC2 knockout mice
efficiency of transcription at hypermethylated promoters show enhanced memory as well as higher levels of H4K5,
(Ivanov, Kacevska, & Ingelman-Sundberg, 2012). Mice H4K12, and H2B acetylation in the hippocampus and
that express lower levels of methyl binding proteins show upregulation of genes, such as CaMKIIa and Creb, that
behavioral and social deficits, including deficits in learn- are important in memory formation (Guan et al., 2009).
ing and memory, signifying the importance of methyla- HDAC inhibitors have therefore been proposed as thera-
tion in the brain (Nelson & Monteggia, 2011). peutic interventions for treatment of different psychiatric
and addiction disorders (Starkman et al., 2012).
Methylation is another well-studied type of histone
Histone Modifications modification. Histone methylation (mono-, di-, or tri-
To date, approximately 16 different histone modifica- methylation) carried out by histone methyltransferases
tions have been identified. Key amongst these include (HMTs) occurs on lysine and arginine residues on his-
methylation, acetylation, sumoylation, phosphorylation, tones H3 and H4. It can function as a marker of activa-
and ubiquitination. These covalent modifications are car- tion/repression in the context of gene expression in a
ried out by a number of enzymes such as methyltransfer- residue-specific manner. Di- and tri-methylation of lysine
ases (HMTs), histone acteyltransferases (HATs), kinases, 4 act as a signal for active transcription (H3K4me2 and
phosphatases, and multiple others (Ivanov et al., 2012). H3K4me3), whereas tri-methylation of lysine 9 and lysine
These specific modifications can serve as signals for pro- 27 act as repressive signals (H3K9me3 and H3K27me3)
teins, such as the polycomb group of proteins (Niessen, (Peterson & Laniel, 2004; Sims & Reinberg, 2006).
Demmers, & Voncken, 2009), which act as transcriptional Increased histone H3 lysine 4 and 27 tri-methylation
repressors. Each of these modifications occurs at specific (Huang et al., 2007) were reported in the prefrontal cortex
amino acids. For example, lysine residues can be acety- of schizophrenia patients. H3 methylation and acetyla-
lated, methylated, ubiquitinated, or sumoylated. The main tion have been associated with alterations in glutamater-
sites of histone acetylation in histone H3 include lysine 4, 9, gic and GABAergic genes, thus pointing to the importance
14, and 28 (Mandrekar, 2011). Histone acetylation usually of histone modifications in neuronal function.

III.  GENE AND BEHAVIOR


352 17.  Alcohol and the Brain

Noncoding RNAs regulatory modifications, we can now delve deeper into


understanding their role in alcoholism.
Although enzymes and proteins involved in epi-
genetics such as HDACs or transcription factors can also
serve as epigenetic modulators, small or long noncoding ALCOHOL, EPIGENETICS, AND GENE
RNAs (ncRNAs), particularly miRNAs, have emerged as EXPRESSION: AN OVERVIEW
particularly strong candidates of epigenetic regulators. It
has been estimated that ∼76% of the genome is actively Although the brain is dynamically changing in term
transcribed, and only 3% codes for proteins. This sug- of synaptic connections, it mainly consists of differenti-
gests that noncoding RNAs likely constitute a significant ated neurons. Therefore it needs to employ approaches
number of transcripts and play a crucial role in regulat- that do not depend on cell division to maintain plastic-
ing gene expression. Long ncRNAs (>200 nucleotides in ity. One way the brain accomplishes this is through plas-
length) usually control gene expression by DNA meth- ticity in the chromatin, which can then regulate neuronal
ylation. X chromosome inactivation is an example of epi- function (Crepaldi & Riccio, 2009). The brain reacts to
genetic regulation mediated by long ncRNAs. Polycomb environmental changes, such as stress, toxins, and expo-
proteins can bind to the ncRNA XIST (which is expressed sure to alcohol and other drugs, by altering its chroma-
by the targeted X chromosome) and cause tri-meth- tin state, eventually causing synaptic alterations that in
ylation of histone H3 (H3K27), resulting in X chromo- turn regulate behavior. A complex web of epigenetic
some silencing (Kaikkonen, Lam, & Glass, 2011). Small modifications can modulate changes in gene expression
ncRNAs such as miRNAs, on the other hand, are around induced by alcohol and other drugs.
22 nucleotides in length. miRNAs can regulate multiple
developmental processes, including synaptic function
and neurogenesis (Papagiannakopoulos & Kosik, 2009; Alcohol and DNA Methylation
Shen & Temple, 2009). Generation of miRNAs is a com- Much of the evidence linking alcohol exposure and
plex process that requires multiple steps. The precursors DNA methylation comes from in vitro and peripheral
for miRNAs (known as pri-miRNAs) are usually several tissue studies. Both human lymphoblast cell line and
thousand nucleotides in length, contain numerous stem peripheral blood cell studies have been valuable in
and loop structures, and are generated through transcrip- refining our understanding of genes implicated in alco-
tion by RNA polymerase II. The pri-miRNA is then con- holism. There are allele-specific effects on DNA meth-
verted into a shorter pre-miRNA (about 70 nucleotides ylation of genes involved in brain function, such as the
in length) sequence by the microprocessor complex, con- serotonin transporter (5-HTT or SLC6A4). Individuals
sisting of the protein Drosha and its cofactor DiGeorge with a history of alcohol dependence had higher levels
syndrome critical region gene 8 (DGCR8) in the nucleus. of SLC6A4 expression, implying a synergism between
This pre-miRNA is now exported into the cytoplasm by genetic and epigenetic factors (Philibert, Sandhu, et al.,
the Exportin-5 (RAN GTP dependent transporter) where 2008). Peripheral blood studies have shown that severity
it is processed by Dicer into short, double-stranded of drinking has a negative correlation with methylation
miRNAs that are ∼22 nucleotides. The miRNA then of five defined CpG sites within the NMDA-receptor
binds the RNA-induced silencing complex and guides 2b (NR2B) promoter site, and NR2B expression was
it to a specific mRNA target (based on sequence com- increased in the first 24 hours of withdrawal (Biermann
plementarity) and cause translational/transcriptional et al., 2009). Chronic alcohol treatment caused a demeth-
repression based on the extent of sequence complemen- ylation of CpG islands and was associated with increased
tarity. With about 197 (human) and 294 (mouse) different NR2B expression in mouse cortical neurons (Marutha
miRNAs expressed in the brain (Ling et al., 2011; Shao Ravindran & Ticku, 2004). Ethanol exposure caused a 5%
et al., 2010), it comes as no surprise that miRNAs have reduction in global CpG methylation in mouse embry-
been shown to mediate a variety of processes and path- onic fibroblasts and elicited proteosomal degradation of
ways that can regulate behavior. For example, miRNAs DNA methyltransferases (DNMT1, 3a and 3b) as well
are involved in neural patterning (Ronshaugen, Biemar, as methyl CpG–binding proteins (MeCp-2 and MBD-2
Piel, Levine, & Lai, 2005), and perturbed production of and -3; Mukhopadhyay, Rezzoug, Kaikaus, Greene,
miRNAs (from deficient Dicer and DGCR8 expression) & Pisano, 2013). A majority of these examples suggest
can alter dendrite growth and cause learning and mem- hypomethylation to correspond with an increase in gene
ory deficits in vivo (Fenelon et al., 2011; Konopka et al., expression; however, this is not always true. Ethanol
2010). Overall, the brain is subject to extensive epigen- downregulated expression of several cell cycle regula-
etic control via multiple mechanisms involving DNA, tion genes by causing DNA hypomethylation in neural
histone modifications, and miRNA networks to regulate stem cell lines and in the mouse frontal cortex, an effect
gene expression. With a general understanding of these that was enhanced in the presence of TGF-β (Hicks,

III.  GENE AND BEHAVIOR


Alcohol, Epigenetics, and Gene Expression: An Overview 353
Middleton, & Miller, 2010). Other factors such as gender that alcohol usage affects levels of DNMT1 mRNA (con-
and ethnicity can also affect methylation status in alco- sistent with previous data by Ponomarev et al., 2012),
hol dependence. For example, methylation status of the decreased levels of apolipoprotein B mRNA-editing
monoamine oxidase A (MAOA) gene significantly cor- enzyme catalytic polypeptide-like (APOBEC-3C), and
related with both alcohol and nicotine dependence in increased levels of 10-11-translocation methylcytosine
women but not men (Philibert, Gunter, Beach, Brody, & dioxygenase (TET) mRNA (the latter two both involved
Madan, 2008). Certain studies have shown that ethanol in the process of demethylation) in the frontal cortex of
can cause hypermethlyation. Most recently, a genome- psychotic patients (Guidotti et al., 2013). Thus studies
wide methylation study in alcoholism showed that there using human brain tissue provide evidence that meth-
were a higher number of genes that were hypermethyl- ylation can be a key epigenetic mediator in alcoholism
ated between European American cases versus controls and other psychiatric disorders.
(HTR3A, NCAM1, DRD4, MBD3, HTR2B, and GRIN1) In addition to studies in postmortem human brains, in
and between African American cases versus controls vivo studies of animal models of alcoholism supported a
(GABRB3 and POMC; Zhang et al., 2013). In addition, role for methylation. For example, administration of the
three CpGs that were upstream of the μ-opioid recep- DNMT1 inhibitor 5-azacitidine caused a 20% decrease in
tor gene (OPRM1, involved in mediating the rewarding alcohol intake in mice (Warnault, Darcq, Levine, Barak,
effects of alcohol) had significantly higher methylation & Ron, 2013). Prenatal ethanol exposure in pregnant rat
levels in alcohol-dependent versus control individuals dams produced significant changes in the epigenetic
(Zhang et al., 2012). Overall, the majority of studies sup- profile of hypothalamic proopiomelanocortin (POMC)
port the role of alcohol mediating changes in brain gene neurons in their male offsprings, as shown by increased
expression via DNA methylation. levels of DNMT1 and methyl CpG–binding protein 2
In addition to in vitro models, studies using human (MeCP2), in addition to increased POMC gene methyla-
postmortem tissue have been invaluable in providing tion and decreased POMC mRNA levels (Bekdash,
insight into the role of alcohol on regulating gene expres- Zhang, & Sarkar, 2013). Diet is a major contributor to the
sion in the brain. Ponomarev, Wang, Zhang, Harris, and methylation process. Alcohol-induced hypermethyl-
Mayfield (2012) identified global changes in epigenetic ation in rat hippocampus and prefrontal cortex was
modifications from postmortem brain samples of alco- decreased by treatment with choline, although choline
holics versus matched controls. Alcohol abuse correlated increased methylation in control rats (Otero, Thomas,
with gene expression changes in different brain regions, Saski, Xia, & Kelly, 2012), showing differential effects of
including the frontal cortex, basolateral, and central choline on methylation based on prior exposure to alco-
nucleus of the amygdala (Ponomarev et al., 2012). Long hol. These changes were reversed by addition of choline
terminal repeat (LTR) regions containing retrotranspo- in the mother’s diet during gestation, highlighting the
sons (endogenous retroviruses, or ERV), which are con- role of dietary supplements in regulating methylation.
stitutively densely methylated, were found to be Other dietary regulators include vitamins such as folic
hypomethylated and showed increased expression in acid (Williams, Berry, & Beerstecher, 1949), which can
alcoholic brains compared to controls. Also, most of the contribute to the formation of S-adenosyl methionine
genes upregulated in the frontal cortex were GC rich in (SAM), the primary methyl group in DNA methylation
content, and DNMT1 was shown to be downregulated (Hamid, Wani, & Kaur, 2009). SAM has been shown to
in all three brain regions, correlating with the hypometh- attenuate effects of alcohol on gene expression. A recent
ylation observed. Given the significant prevalence of study found that rats fed an alcohol diet showed hyper-
LTRs in the human genome, the study concluded that methylation and concomitant upregulation of genes
global methylation changes are correlated with alcohol (Cd14, Hspa1a, Irf1, Irak1, Irak2, Map3k7, Myd88, Pparα,
dependence. A recent study utilized methylated DNA Ripk2, Tollip, and Traf6) compared to rats fed a com-
immunoprecipitation hybridized on microarrays com- bined alcohol and SAM diet (Khachatoorian et al., 2013).
posed of genomic promoter regions in frontal cortices of To summarize, postmortem data suggests that alco-
alcoholics versus controls (Manzardo, Henkhaus, & hol causes DNA hypomethylation, whereas some of the
Butler, 2012). No differences in global methylation were in vivo data point to DNA hypermethylation. These
found between the two groups. Only about 20% of the opposing effects seem to occur in a cell/tissue/organ-
promoters showed differential methylation, with about specific manner and may also be temporal in nature. The
half being hypermethylated in alcoholics, for example, dogma of increased DNA methylation being synony-
histone gene HIST2H2AC. Another group studied the mous with gene repression is changing and giving rise
role of alcohol abuse in gene expression changes in psy- to a far more complex pattern of gene regulation via
chiatric disorders, given the frequent occurrence of alco- methylation. One important point to consider is the
hol abuse as a comorbid factor in psychiatric patients potential difference in methylation patterns between
(Bolton, Robinson, & Sareen, 2009). Results suggested peripheral and brain tissues. Barker, Zhang, Wang,

III.  GENE AND BEHAVIOR


354 17.  Alcohol and the Brain

Taylor, and Zhang (2013) compared effects of alcohol- cell cycle regulation that were downregulated in the pres-
induced DNA methylation of the serotonin receptor 3a ence of ethanol (Zhou et al., 2011). This downregulation
(Htr3a) gene in mice using nine different brain regions was similar to the actions of the methylation inhibitor
and trunk blood. Alcohol consumption did not alter 5-azacitidine (5-AzaC), and sites altered by methylation
methylation of the Htr3a promoter CpGs in six of the were also shown to be binding sites for transcription fac-
nine brain regions (Barker et al., 2013). However, changes tors (Zhou et al., 2011). Overall, in vitro data suggests inter-
in promoter methylation (both increases and decreases) play of histone methylation and acetylation modifications.
were observed that were brain region, blood, and CpG This is indicative of a more integrated process of control-
specific. In some brain regions, such as the dorsomedial ling gene expression whereby a single histone modifica-
striatum, methylation inversely correlated with Htr3a tion does not predicate the direction of gene expression,
expression. This points to not only variations in gene but instead a combination of different modifications
expression between brain and peripheral tissue, but dif- directs gene expression and cellular function.
ferential brain regional variation as well. Given that
DNA methylation and methyl-binding proteins have In Vivo/Animal Models
been implicated in complex neuronal processes such as Alcohol exposure in rats increased H3K9 and H4K8
long term potentiation (Nelson & Monteggia, 2011), acetylation in the brain (Pandey, Ugale, Zhang, Tang, &
methylation may serve as a means of fine-tuning gene Prakash, 2008). Alcohol induced anxiety-like behavior,
expression and may effectively enable different regions with a concomitant decrease in HDAC activity. It also
of the brain to specifically modulate behavior. increased levels of CREB-binding protein (CBP) and
neuropeptide Y (NPY), coinciding with the increased
Alcohol and Histone Modification histone acetylation. However, anxiety-like behavior dur-
ing alcohol withdrawal showed the opposite effects,
In Vitro Models that is, decreased acetylation and decreased expression
Alcohol-induced histone modifications have been of these proteins in the amygdala. The HDAC inhibitor
reported in the brain, liver, and gastrointestinal system Trichostatin A (TSA) reversed these effects during with-
(Shukla et al., 2008). Alcohol treatment induced acetylation drawal, restoring mRNA and protein levels of NPY and
at lysine 9 in histone H3 (H3AcK9) in primary rat hepato- preventing the anxiety-like behavior (Pandey et al., 2008).
cyte cultures (Park, Miller, & Shukla, 2003). Knockdown of TSA also increased ethanol consumption in C57BL/6NCrl
the GCN5 HAT caused global changes in gene expression mice, and ethanol intake affected gene expression levels
(891 transcripts differentially expressed) in hepatoma cells in the nucleus accumbens (NAc), including several genes
transfected with alcohol dehydrogenase 1 (Choudhury involved in chromatin remodeling (Wolstenholme et al.,
et al., 2011). SLC44A2 (a putative choline transporter 2011). A recent study by Warnault, Darcq, Levine, Barak
gene) was upregulated in response to alcohol treatment and Ron (2013) showed that administering the DNMT
(Choudhury et al., 2011). In human SK-N-MC neuroblas- inhibitor (5-AzaC) decreased alcohol drinking, whereas
toma cells and fetal-derived primary neurons exposed to the HDAC inhibitor SAHA reduced binge-like drink-
ethanol, an upregulation of HDAC activity, induction of ing in a dose-specific manner (Warnault et al., 2013). In
HDAC1,3 gene expression, and concomitant increase in line with these findings, excessive alcohol consumption
expression of the serotonin gene were observed (Agudelo, increased DNMT1 levels and decreased histone H4 acety-
Yoo, & Nair, 2012), and these changes were reversed by lation in the NAc of mice (Warnault et al., 2013). The NAc
treatment with the HDAC inhibitor trichostatin A (TSA). seems to be a critical region in controlling alcohol depen-
Studies using human SH-SY5Y neuroblastoma cells dence, with higher levels of glutamate being observed
showed that prolonged ethanol exposure significantly in the NAc of alcohol-dependent patients compared to
increased H3K27me3 and H3K9Ac levels in the promoter controls and glutamate and glutamine levels in the NAc
region of the prodynorphin (PDYN) gene (D’Addario and cingular cortex, respectively, showing a positive cor-
et al., 2011). A chronic intermittent ethanol paradigm relation with drinking behavior (Bauer et al., 2013).
increased levels of the NMDA-receptor 2B (NR2B) gene Two major studies have modeled epigenetics in fetal
and increased H3K9Ac levels near the NR2B promoter alcohol spectrum disorders (FASD). Both these studies
in mouse cortical neurons. Levels of G9a, Suv39 h1, and use ethanol exposure in the time period of postnatal days
HDAC1–3 in the chromatin of the NR2B gene promoter two to 12, which models the third trimester of human
decreased and were believed to contribute to the H3K9Ac pregnancy, in which severe cerebellar damage (one of
increase reported earlier (Qiang, Denny, Lieu, Carreon, & the hallmarks of FASD) occurs (Green, 2004). Postnatal
Li, 2011). Neural stem cells exposed to a binge-like etha- day seven rats that had previously been exposed to etha-
nol dose showed decreased migration, differentiation, and nol showed a decrease in histone acetyltransferase
gene expression analysis, revealing a number of genes CBP levels and a concomitant decrease in acetylation
involved in glutamate transmission, axon extension, and of H3 and H4 in the cerebella (Guo et al., 2011).

III.  GENE AND BEHAVIOR


Summary and Future Directions 355
Inhibition of the lysine dimethyltransferase (G9a) prior changes seen in postmortem brain samples of alcoholics
to ethanol treatment in postnatal day seven mouse pups (Lewohl et al., 2011). Recently, Gorini, Nunez, Roberts,
rescued alcohol-mediated neurodegeneration and nor- and Mayfield (2013) identified region-specific changes in
malized the increased levels of H3K9me2 and H3K27me2 miRNA levels in the cerebral cortex and midbrain using
in the developing mouse brain (Subbanna et al., 2013). a chronic intermittent ethanol exposure paradigm in
A majority of the in vivo studies discussed previously vivo and revealed miRNA 146b as a top alcohol-respon-
point to histone acetylation and transcriptional comodu- sive miRNA (Gorini et al., 2013). This miRNA has been
lators, such as CBP, in regulation of alcohol-mediated shown to negatively regulate Toll-like receptor-4 signal-
epigenetic effects. ing (Curtale et al., 2013). miRNAs have emerged as key
mediators of gene and protein expression, and future
Human Postmortem Brain Studies research will determine the long-term effects of alcohol
Human postmortem studies have also shown changes usage on brain miRNA expression and epigenetic regu-
in histone gene expression in alcoholics. For example, lation in the brain.
CBP and CREB mRNA levels decreased in alcoholic
brains (Ponomarev et al., 2012). On the other hand,
expression of genes involved in transcription corepres- SUMMARY AND FUTURE DIRECTIONS
sor complexes (TCC) increased in alcoholics (Liu et al.,
2006; Ponomarev et al., 2012). Thus several of the changes The field of epigenetics has rapidly advanced over the
observed in human postmortem brains agree with the past few years. The ENCyclopedia Of DNA Elements
findings from animal models of alcoholism. The study (ENCODE) project has been key in catapulting our
of the interrelationship between epigenetics and alcohol understanding of the genome and epigenome. It has
in human brain tissue is still in its infancy, and further been calculated that about 56% of the genome is highly
studies are warranted to analyze the complex epigenetic enriched in modified histones, supporting a significant
modifications involved in the alcoholic brain. role for epigenetics in constitutive gene expression and
therefore normal cellular function. Given the complex-
ity of epigenetic regulators and their modifications, an
Alcohol and microRNAs intricate array of processes for regulation of gene expres-
microRNAs (miRNAs) are short, noncoding RNAs sion has emerged. Improved techniques for analyzing
that are capable of controlling chromatin modification, epigenetic changes have further fueled growth of this
DNA methylation, mRNA degradation, and repres- field, including bisulphate sequencing, ChIP-seq and
sion of mRNA translation (Bartel, 2009). One of the first Ribo-seq, profile proteins associated with histone marks,
reports of ethanol action on miRNA showed that at doses and transcriptome assays (Telese, Gamliel, Skowronska-
representative of chronic alcohol users ethanol caused a Krawczyk, Garcia-Bassets, & Rosenfeld, 2013).
downregulation in levels of miR-21, -335, -9, and -153 Alcoholism is a complex disease that is subject to
in neurosphere cultures (Sathyan, Golden, & Miranda, multiple genetic and environmental factors. It also
2007). These miRNAs have specific functions in control- appears to be comorbid with other disorders such as
ling proliferation and apoptosis, suggesting that ethanol psychiatric illness and cancer. Multiple epigenetic
may alter these functions indirectly by altering miRNA mechanisms control gene expression, usually in a cell/
expression. Furthermore, miR-9 was upregulated in tissue-specific manner (Figure 17.2). For example, based
rat adult brains following ethanol exposure, whereas
expression of specific BK-channel (calcium- and voltage-
gated channels) splice variants was downregulated
(Pietrzykowski et al., 2008). Increased expression of miR-
375 was reported in pharyngeal/laryngeal tumors that
correlated with alcohol use (Avissar, McClean, Kelsey,
& Marsit, 2009). Studies of alcoholic liver disease have
shown that ethanol can produce overexpression of
miR-212 and cause gut leakiness via regulation of tight
junction proteins, such as ZO-1, which is a miR-212 tar-
get (Tanget et al., 2008). In addition, ethanol-induced
miR-199 can target ET-1 and HIF-1 in endothelial cells
(Yeligar, Tsukamoto, & Kalra, 2009). Microarray studies
FIGURE 17.2  Factors contributing to epigenetic changes. The
have shown that ethanol exposure causes global changes
figure depicts different factors contributing to epigenetic modifications
in miRNA expression in primary cortical neurons (Guo, that can alter gene expression in a cell/tissue-specific manner, leading
Chen, Carreon, & Qiang, 2012) in agreement with similar to changes in behavior.

III.  GENE AND BEHAVIOR


356 17.  Alcohol and the Brain

on human postmortem tissue studies DNA and histone NIH, NIAAA grant from the Integrative Neuroscience Initiative on
methylation seem to be the predominant epigenetic Alcoholism to RAH (AA013518), a K award to IP (AA017234), and a
DOD grant to RAH (W81XWH-11-1-0245).
modifications in the brain, whereas acetylation, meth-
ylation, and phosphorylation have been observed in the
liver. This does not necessarily imply that epigenetic References
changes in the brain are limited to methylation, but it Agudelo, M., Yoo, C., & Nair, M. P. (2012). Alcohol-induced serotoner-
may instead reflect a limitation of current methodology gic modulation: The role of histone deacetylases. Alcohol, 46(7),
to detect all modifications in tissue sensitive to degra- 635–642.
dation. Support for this comes from studies in neurons Avissar, M., McClean, M. D., Kelsey, K. T., & Marsit, C. J. (2009).
MicroRNA expression in head and neck cancer associates with
in vitro in which acetylation was detected (Qiang
alcohol consumption and survival. Carcinogenesis, 30(12),
et al., 2011). 2059–2063.
It is important to keep in mind that alcoholism, as a Barker, J. M., Zhang, Y., Wang, F., Taylor, J. R., & Zhang, H. (2013).
complex disease, involves multiple interconnected path- Ethanol induced Htr3a promoter methylation changes in mouse
ways. Metabolites of alcohol, such as acetaldehyde, can blood and brain. Alcoholism, Clinical and Experimental Research,
cause DNA damage (Langevin, Crossan, Rosado, 37(Suppl. 1), E101–107.
Bartel, D. P. (2009). MicroRNAs: Target recognition and regulatory
Arends, & Patel, 2011; Rulten, Hodder, Ripley, Stephens, functions. Cell, 136(2), 215–233.
& Mayne, 2008), and neuroimmune regulation is another Bauer, J., Pedersen, A., Scherbaum, N., Bening, J., Patschke, J., & Kugel, H.
important pathway implicated in alcoholism (Blednov (2013). Craving in alcohol-dependent patients after detoxification
et al. 2012; Crews, Zou, & Qin, 2011; Liu et al., 2004). The is related to glutamatergic dysfunction in the nucleus accumbens
combined effects of alcohol on epigenetic and inflamma- and the anterior cingulate cortex. Neuropsychopharmacology, 38(8),
1401–1408.
tory processes are examples of pathways that may be
Bekdash, R. A., Zhang, C., & Sarkar, D. K. (2013). Gestational choline
interconnected to modulate genetic regulation on a supplementation normalized fetal alcohol-induced alterations in
global scale without affecting DNA sequence. The conse- histone modifications, DNA methylation, and proopiomelanocor-
quences of these interactions may be far reaching, tin (POMC) gene expression in beta-endorphin-producing POMC
affecting not only the brain but also other organs in neurons of the hypothalamus. Alcoholism, Clinical and Experimental
which inflammatory mechanisms could be potentially Research, 37(7), 1133–1142.
Biermann, T., Reulbach, U., Lenz, B., Frieling, H., Muschler, M., &
damaging.
Hillemacher, T. (2009). N-methyl-D-aspartate 2b receptor subtype
The ultimate goal of epigenetic research is to discover (NR2B) promoter methylation in patients during alcohol with-
successful therapies for addictions such as alcoholism drawal. Journal of Neural Transmission, 116(5), 615–622.
and other complex diseases. As noted by Ponomarev Blednov, Y. A., Ponomarev, I., Geil, C., Bergeson, S., Koob, G. F., &
(2013), current literature shows increased levels of his- Harris, R. A. (2012). Neuroimmune regulation of alcohol consump-
tone acetylation due to acute alcohol exposure, and tion: Behavioral validation of genes obtained from genomic stud-
ies. Addiction Biology, 17(1), 108–120.
withdrawal is characterized by increased deacetylation, Bolton, J. M., Robinson, J., & Sareen, J. (2009). Self-medication of mood
therefore providing a strong rationale for the use of disorders with alcohol and drugs in the National Epidemiologic
HDAC modulators as therapeutic targets. However, cur- Survey on alcohol and related conditions. Journal of Affective
rent studies also suggest brain region–specific changes Disorders, 115(3), 367–375.
in epigenetic modifications, reinforcing the need for tar- Chan, H. M., & La Thangue, N. B. (2001). P300/CBP proteins: HATs for
transcriptional bridges and scaffolds. Journal of Cell Science, 114(Pt
geted therapy for specific regions or even specific neuro-
13), 2363–2373.
nal subpopulations. Dietary and other environmental Choudhury, M., Pandey, R. S., Clemens, D. L., Davis, J. W., Lim, R. W.,
factors are additional considerations in designing poten- & Shukla, S. D. (2011). Knock down of GCN5 histone acetyltrans-
tial therapeutic agents to target epigenetic regulation of ferase by siRNA decreases ethanol-induced histone acetylation
alcoholism. and affects differential expression of genes in human hepatoma
The human genome is highly enriched in modified cells. Alcohol, 45(4), 311–324.
Cortessis, V. K., Thomas, D. C., Levine, A. J., Breton, C. V., Mack, T. M.,
DNA and histones, providing a prime template for epi-
& Siegmund, K. D. (2012). Environmental epigenetics: Prospects
genetics in regulating constitutive gene expression and for studying epigenetic mediation of exposure-response relation-
therefore normal cellular function. Alcohol’s role in ships. Human Genetics, 131(10), 1565–1589.
altering these mechanisms will no doubt continue to fuel Crepaldi, L., & Riccio, A. (2009). Chromatin learns to behave.
significant advancements in the genetics of alcohol Epigenetics, 4(1), 23–26.
addiction, highlighting pathways for gene modulation Crews, F. T., Zou, J., & Qin, L. (2011). Induction of innate immune genes
in brain create the neurobiology of addiction. Brain, Behavior, and
without any change in DNA sequence. Immunity, 25(Suppl. 1), S4–S12.
Curtale, G., Mirolo, M., Renzi, T. A., Rossato, M., Bazzoni, F., & Locati,
Acknowledgments M. (2013). Negative regulation of Toll-like receptor 4 signaling by
The authors would like to express their sincere gratitude towards Dr. IL-10-dependent microRNA-146b. Proceedings of the National
Jody Mayfield for her valuable editorial comments and suggestions. Academy of Sciences of the United States of America, 110(28),
We would also like to acknowledge funding support provided by a 11499–11504.

III.  GENE AND BEHAVIOR


REFERENCES 357
D’Addario, C., Johansson, S., Candeletti, S., Romualdi, P., Ogren, S. O., Kelly, W. K., & Marks, P. A. (2005). Drug insight: Histone deacetylase
& Terenius, L. (2011). Ethanol and acetaldehyde exposure induces inhibitors–development of the new targeted anticancer agent
specific epigenetic modifications in the prodynorphin gene pro- suberoylanilide hydroxamic acid. Nature Clinical Practice and
moter in a human neuroblastoma cell line. The FASEB Journal: Oncology, 2(3), 150–157.
Official Publication of the Federation of American Societies for Khachatoorian, R., Dawson, D., Maloney, E. M., Wang, J., French, B. A.,
Experimental Biology, 25(3), 1069–1075. & French, S. W. (2013). SAMe treatment prevents the ethanol-
Dawson, M. A., & Kouzarides, T. (2012). Cancer epigenetics: From induced epigenetic alterations of genes in the Toll-like receptor
mechanism to therapy. Cell, 150(1), 12–27. pathway. Experimental and Molecular Pathology, 94(1), 243–246.
Ellis, L., Atadja, P. W., & Johnstone, R. W. (2009). Epigenetics in cancer: Konopka, W., Kiryk, A., Novak, M., Herwerth, M., Parkitna, J. R., &
Targeting chromatin modifications. Molecular Cancer Therapeutics, Wawrzyniak, M. (2010). MicroRNA loss enhances learning and
8(6), 1409–1420. memory in mice. The Journal of Neuroscience: The Official Journal of
Fenelon, K., Mukai, J., Xu, B., Hsu, P. K., Drew, L. J., & Karayiorgou, M. the Society for Neuroscience, 30(44), 14835–14842.
(2011). Deficiency of Dgcr8, a gene disrupted by the 22q11.2 micro- Langevin, F., Crossan, G. P., Rosado, I. V., Arends, M. J., & Patel, K. J.
deletion, results in altered short-term plasticity in the prefrontal (2011). Fancd2 counteracts the toxic effects of naturally produced
cortex. Proceedings of the National Academy of Sciences of the United aldehydes in mice. Nature, 475(7354), 53–58.
States of America, 108(11), 4447–4452. Lee, M. J., Kim, Y. S., Kummar, S., Giaccone, G., & Trepel, J. B. (2008).
Fleisch, A. F., Wright, R. O., & Baccarelli, A. A. (2012). Environmental Histone deacetylase inhibitors in cancer therapy. Current Opinion
epigenetics: A role in endocrine disease? Journal of Molecular in Oncology, 20(6), 639–649.
Endocrinology, 49(2), R61–R67. Lewohl, J. M., Nunez, Y. O., Dodd, P. R., Tiwari, G. R., Harris, R. A., &
Gorini, G., Nunez, Y. O., Roberts, A. J., Mayfield, R. D. (2013). Mayfield, R. D. (2011). Up-regulation of microRNAs in brain of
Unpublished data. human alcoholics. Alcoholism, Clinical and Experimental Research,
Green, J. T. (2004). The effects of ethanol on the developing cerebellum 35(11), 1928–1937.
and eyeblink classical conditioning. Cerebellum, 3(3), 178–187. Ling, K. H., Brautigan, P. J., Hahn, C. N., Daish, T., Rayner, J. R., &
http://dx.doi.org/10.1080/14734220410017338. Cheah, P. S. (2011). Deep sequencing analysis of the develop-
Guan, J. S., Haggarty, S. J., Giacometti, E., Dannenberg, J. H., Joseph, ing mouse brain reveals a novel microRNA. BMC Genomics,
N., & Gao, J. (2009). HDAC2 negatively regulates memory forma- 12, 176.
tion and synaptic plasticity. Nature, 459(7243), 55–60.
Liu, J., Lewohl, J. M., Dodd, P. R., Randall, P. K., Harris, R. A., &
Guidotti, A., Dong, E., Gavin, D. P., Veldic, M., Zhao, W., & Bhaumik,
Mayfield, R. D. (2004). Gene expression profiling of individual
D. K. (2013). DNA methylation/demethylation network expres-
cases reveals consistent transcriptional changes in alcoholic human
sion in psychotic patients with a history of alcohol abuse.
brain. Journal of Neurochemistry, 90(5), 1050–1058.
Alcoholism, Clinical and Experimental Research, 37(3), 417–424.
Liu, J., Lewohl, J. M., Harris, R. A., Iyer, V. R., Dodd, P. R., & Randall, P.
Guo, W., Crossey, E. L., Zhang, L., Zucca, S., George, O. L., & Valenzuela,
K. (2006). Patterns of gene expression in the frontal cortex discrimi-
C. F. (2011). Alcohol exposure decreases CREB binding protein
nate alcoholic from nonalcoholic individuals. Neuro-
expression and histone acetylation in the developing cerebellum.
psychopharmacology, 31(7), 1574–1582.
PLoS One, 6(5), e19351.
Lotfipour, S., Howard, J., Roumani, S., Hoonpongsimanont, W.,
Guo, Y., Chen, Y., Carreon, S., & Qiang, M. (2012). Chronic intermittent
Chakravarthy, B., & Anderson, C. L. (2013). Increased detection of
ethanol exposure and its removal induce a different miRNA
alcohol consumption and at-risk drinking with computerized alco-
expression pattern in primary cortical neuronal cultures.
hol screening. The Journal of Emergency Medicine, 44(4), 861–866.
Alcoholism, Clinical and Experimental Research, 36(6), 1058–1066.
Hamid, A., Wani, N. A., & Kaur, J. (2009). New perspectives on folate Mandrekar, P. (2011). Epigenetic regulation in alcoholic liver disease.
transport in relation to alcoholism-induced folate malabsorption– World Journal of Gastroenterology: WJG, 17(20), 2456–2464.
association with epigenome stability and cancer development. The Manzardo, A. M., Henkhaus, R. S., & Butler, M. G. (2012). Global DNA
FASEB Journal: Official Publication of the Federation of American promoter methylation in frontal cortex of alcoholics and controls.
Societies for Experimental Biology, 276(8), 2175–2191. Gene, 498(1), 5–12.
Hamilton, J. P. (2011). Epigenetics: Principles and practice. Digestive Marutha Ravindran, C. R., & Ticku, M. K. (2004). Changes in methyla-
Diseases, 29(2), 130–135. tion pattern of NMDA receptor NR2B gene in cortical neurons after
Hicks, S. D., Middleton, F. A., & Miller, M. W. (2010). Ethanol-induced chronic ethanol treatment in mice. Brain Research and Molecular
methylation of cell cycle genes in neural stem cells. Journal of Brain Research, 121(1–2), 19–27.
Neurochemistry, 114(6), 1767–1780. McQuown, S. C., Barrett, R. M., Matheos, D. P., Post, R. J., Rogge, G. A.,
Hillemacher, T. (2011). Biological mechanisms in alcohol dependence— & Alenghat, T. (2011). HDAC3 is a critical negative regulator of
new perspectives. Alcohol and Alcoholism, 46(3), 224–230. long-term memory formation. The Journal of Neuroscience: The
Huang, H. S., Matevossian, A., Whittle, C., Kim, S. Y., Schumacher, A., Official Journal of the Society for Neuroscience, 31(2), 764–774.
& Baker, S. P. (2007). Prefrontal dysfunction in schizophrenia Mukhopadhyay, P., Rezzoug, F., Kaikaus, J., Greene, R. M., & Pisano,
involves mixed-lineage leukemia 1-regulated histone methyla- M. M. (2013). Alcohol modulates expression of DNA methyltran-
tion at GABAergic gene promoters. The Journal of Neuroscience: ferases and methyl CpG-/CpG domain-binding proteins in murine
The Official Journal of the Society for Neuroscience, 27(42), embryonic fibroblasts. Reproductive Toxicology, 37, 40–48.
11254–11262. Nan, X., Meehan, R. R., & Bird, A. (1993). Dissection of the methyl-CpG
Ivanov, M., Kacevska, M., & Ingelman-Sundberg, M. (2012). binding domain from the chromosomal protein MeCP2. Nucleic
Epigenomics and interindividual differences in drug response. Acids Research, 21(21), 4886–4892.
Clinical Pharmacology and Therapeutics, 92(6), 727–736. Nan, X., Tate, P., Li, E., & Bird, A. (1996). DNA methylation specifies
Jablonka, E. (2012). Epigenetic variations in heredity and evolution. chromosomal localization of MeCP2. Molecular and Cellular Biology,
Clinical Pharmacology and Therapeutics, 92(6), 683–688. 16(1), 414–421.
Kaikkonen, M. U., Lam, M. T., & Glass, C. K. (2011). Non-coding RNAs Nelson, E. D., & Monteggia, L. M. (2011). Epigenetics in the mature
as regulators of gene expression and epigenetics. Cardiovascular mammalian brain: Effects on behavior and synaptic transmission.
Research, 90(3), 430–440. Neurobiology of Learning and Memory, 96(1), 53–60.

III.  GENE AND BEHAVIOR


358 17.  Alcohol and the Brain

Niessen, H. E., Demmers, J. A., & Voncken, J. W. (2009). Talking to chro- model of the fetal cerebral cortical neuroepithelium. The Journal of
matin: Post-translational modulation of polycomb group function. Neuroscience: The Official Journal of the Society for Neuroscience,
Epigenetics Chromatin, 2(1), 10. 27(32), 8546–8557.
Oliveira, A. M., Wood, M. A., McDonough, C. B., & Abel, T. (2007). Shao, N. Y., Hu, H. Y., Yan, Z., Xu, Y., Hu, H., & Menzel, C. (2010).
Transgenic mice expressing an inhibitory truncated form of p300 Comprehensive survey of human brain microRNA by deep
exhibit long-term memory deficits. Learning and Memory, 14(9), sequencing. BMC Genomics, 11, 409.
564–572. Shen, Q., & Temple, S. (2009). Fine control: MicroRNA regulation of
Otero, N. K., Thomas, J. D., Saski, C. A., Xia, X., & Kelly, S. J. (2012). adult neurogenesis. Nature Neuroscience, 12(4), 369–370.
Choline supplementation and DNA methylation in the hippocam- Shukla, S. D., Velazquez, J., French, S. W., Lu, S. C., Ticku, M. K., &
pus and prefrontal cortex of rats exposed to alcohol during devel- Zakhari, S. (2008). Emerging role of epigenetics in the actions of
opment. Alcoholism, Clinical and Experimental Research, 36(10), alcohol. Alcoholism, Clinical and Experimental Research, 32(9),
1701–1709. 1525–1534.
Pandey, S. C., Ugale, R., Zhang, H., Tang, L., & Prakash, A. (2008). Brain Sims, R. J., III & Reinberg, D. (2006). Histone H3 Lys 4 methylation:
chromatin remodeling: A novel mechanism of alcoholism. Caught in a bind? Genes and Development, 20(20), 2779–2786.
The Journal of Neuroscience: The Official Journal of the Society for Starkman, B. G., Sakharkar, A. J., & Pandey, S. C. (2012). Epigenetics-
Neuroscience, 28(14), 3729–3737. beyond the genome in alcoholism. Alcohol Research, 34(3), 293–305.
Papagiannakopoulos, T., & Kosik, K. S. (2009). MicroRNA-124: Subbanna, S., Shivakumar, M., Umapathy, N. S., Saito, M., Mohan, P. S.,
Micromanager of neurogenesis. Cell Stem Cell, 4(5), 375–376. & Kumar, A. (2013). G9a-mediated histone methylation regulates
Park, P. H., Miller, R., & Shukla, S. D. (2003). Acetylation of histone H3 ethanol-induced neurodegeneration in the neonatal mouse brain.
at lysine 9 by ethanol in rat hepatocytes. Biochemical and Biophysical Neurobiology of Diseases, 54, 475–485.
Research Communications, 306(2), 501–504. Tang, Y., Banan, A., Forsyth, C. B., Fields, J. Z., Lau, C. K., & Zhang, L.
Peterson, C. L., & Laniel, M. A. (2004). Histones and histone modifica- J. (2008). Effect of alcohol on miR-212 expression in intestinal epi-
tions. Current Biology, 14(14), R546–R551. thelial cells and its potential role in alcoholic liver disease.
Philibert, R. A., Sandhu, H., Hollenbeck, N., Gunter, T., Adams, W., & Alcoholism, Clinical and Experimental Research, 32(2), 355–364.
Madan, A. (2008). The relationship of 5HTT (SLC6A4) methylation Telese, F., Gamliel, A., Skowronska-Krawczyk, D., Garcia-Bassets, I., &
Rosenfeld, M. G. (2013). “Seq-ing” insights into the epigenetics of
and genotype on mRNA expression and liability to major depres-
neuronal gene regulation. Neuron, 77(4), 606–623.
sion and alcohol dependence in subjects from the Iowa Adoption
Warnault, V., Darcq, E., Levine, A., Barak, S., & Ron, D. (2013).
Studies. American Journal of Medical Genetics. Part B, Neuropsychiatric
Chromatin remodeling—a novel strategy to control excessive alco-
Genetics: The Official Publication of the International Society of
hol drinking. Translational Psychiatry, 3, e231.
Psychiatric Genetics, 147B(5), 543–549.
Williams, R. J., Berry, L. J., & Beerstecher, E. (1949). Individual meta-
Philibert, R. A., Gunter, T. D., Beach, S. R., Brody, G. H., & Madan, A.
bolic patterns, alcoholism, genetotrophic diseases. Proceedings of
(2008). MAOA methylation is associated with nicotine and alcohol
the National Academy of Sciences of the United States of America, 35(6),
dependence in women. American Journal of Medical Genetics. Part B,
265–271.
Neuropsychiatric Genetics: The Official Publication of the International
Wolstenholme, J. T., Warner, J. A., Capparuccini, M. I., Archer, K. J.,
Society of Psychiatric Genetics, 147B(5), 565–570.
Shelton, K. L., & Miles, M. F. (2011). Genomic analysis of individual
Pietrzykowski, A. Z., Friesen, R. M., Martin, G. E., Puig, S. I., Nowak,
differences in ethanol drinking: Evidence for non-genetic factors in
C. L., & Wynne, P. M. (2008). Posttranscriptional regulation of BK
C57BL/6 mice. PLoS One, 6(6), e21100.
channel splice variant stability by miR-9 underlies neuroadapta- Yahn, S. L., Watterson, L. R., & Olive, M. F. (2013). Safety and efficacy of
tion to alcohol. Neuron, 59(2), 274–287. acamprosate for the treatment of alcohol dependence. Substance
Ponomarev, I., Wang, S., Zhang, L., Harris, R. A., & Mayfield, R. D. Abuse, 6, 1–12.
(2012). Gene coexpression networks in human brain identify epi- Yeligar, S., Tsukamoto, H., & Kalra, V. K. (2009). Ethanol-induced expres-
genetic modifications in alcohol dependence. The Journal of sion of ET-1 and ET-BR in liver sinusoidal endothelial cells and
Neuroscience: The Official Journal of the Society for Neuroscience, 32(5), human endothelial cells involves hypoxia-inducible factor-1alpha
1884–1897. and microrNA-199. Journal of Immunology, 183(8), 5232–5243.
Ponomarev, I. (2013). Epigenetic control of gene expression in the alco- Yuan, H., & Marmorstein, R. (2013). Histone acetyltransferases: Rising
holic brain. Alcohol Research: Current Reviews, 35(1), 69. ancient counterparts to protein kinases. Biopolymers, 99(2), 98–111.
Qiang, M., Denny, A., Lieu, M., Carreon, S., & Li, J. (2011). Histone http://dx.doi.org/10.1002/bip.22128.
H3K9 modifications are a local chromatin event involved in etha- Zhang, H., Herman, A. I., Kranzler, H. R., Anton, R. F., Simen, A. A., &
nol-induced neuroadaptation of the NR2B gene. Epigenetics, 6(9), Gelernter, J. (2012). Hypermethylation of OPRM1 promoter region
1095–1104. in European Americans with alcohol dependence. Journal of Human
Ronshaugen, M., Biemar, F., Piel, J., Levine, M., & Lai, E. C. (2005). The Genetics, 57(10), 670–675.
Drosophila microRNA iab-4 causes a dominant homeotic transfor- Zhang, H., Herman, A. I., Kranzler, H. R., Anton, R. F., Zhao, H., &
mation of halteres to wings. Genes and Development, 19(24), Zheng, W. (2013). Array-based profiling of DNA methylation
2947–2952. changes associated with alcohol dependence. Alcoholism, Clinical
Rulten, S. L., Hodder, E., Ripley, T. L., Stephens, D. N., & Mayne, L. V. and Experimental Research, 37(Suppl. 1), E108–E115.
(2008). Alcohol induces DNA damage and the Fanconi anemia D2 Zhou, F. C., Balaraman, Y., Teng, M., Liu, Y., Singh, R. P., & Nephew,
protein implicating FANCD2 in the DNA damage response path- K. P. (2011). Alcohol alters DNA methylation patterns and inhibits
ways in brain. Alcoholism, Clinical and Experimental Research, 32(7), neural stem cell differentiation. Alcoholism, Clinical and Experimental
1186–1196. Research, 35(4), 735–746.
Sathyan, P., Golden, H. B., & Miranda, R. C. (2007). Competing interac-
tions between micro-RNAs determine neural progenitor survival
and proliferation after ethanol exposure: Evidence from an ex vivo

III.  GENE AND BEHAVIOR

You might also like