Alcohol

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Genes, Brain and Behavior (2017) 16: 86–100 doi: 10.1111/gbb.

12344

Review

Reprogramming of mPFC transcriptome and function


in alcohol dependence

M. Heilig†,∗ , E. Barbier† , A. L. Johnstone‡ , formation of dynamic neuronal networks, or functional


J. Tapocik† , M. W. Meinhardt§ , S. Pfarr§ , ensembles that are shaped by transcriptional responses.
C. Wahlestedt‡ and W. H. Sommer§,¶ The epigenetic dysregulations identified by our molec-
ular studies are likely to alter this dynamic processing
† Center for Social and Affective Neuroscience, Department of
in multiple ways. In summary, epigenetic molecular
switches in the mPFC appear to be turned on as alco-
Clinical and Experimental Medicine, Linköping University, holism develops. Strategies to reverse these processes
Linköping, Sweden, ‡ Department of Psychiatry and Behavioral may offer targets for disease-modifying treatments.
Sciences, University of Miami Miller School of Medicine, Miami,
FL, USA, § Department of Psychopharmacology, and Keywords: Alcohol use disorder, animal model, BDNF, DNA
¶ Department of Addiction Medicine, Central Institute of Mental methylation, inhibitory control, mGlur2, miRNA, neuronal
Health, University of Heidelberg, Mannheim, Germany ensemble, post-dependent, transcriptome, viral vector
*Corresponding author: M. Heilig, Center for Social and Affec-
Received 5 July 2016, revised 30 August 2016, accepted for
tive Neuroscience, Department of Clinical and Experimental
publication 19 September 2016
Medicine, Linköping University, Linköping S-581 83, Sweden.
E-mail: markus.heilig@liu.se

Alcohol addiction: emergence of the syndrome


Despite its limited immediate reinforcement value, alco-
hol has a potent ability to induce neuroadaptations that Alcohol addiction [hereafter equated with alcohol depen-
promote its incentive salience, escalation of voluntary dence, moderate – severe alcohol use disorder (AUD)
alcohol intake and aversion-resistant alcohol seek- or simply ‘alcoholism’, the term preferred by Alcoholics
ing. A constellation of these traits, collectively called Anonymous] develops over years to decades. Most attempts
‘post-dependent’, emerges following brain exposure to capture this syndrome by different diagnostic systems are
to repeated cycles of intoxication and withdrawal. The based on symptoms observed or reported here-and-now.
medial prefrontal cortex (mPFC) and its subdivisions However, because of its chronic, progressive nature, it is also
exert top-down regulation of approach and avoidance informative to consider the development of this condition
behaviors, including those that lead to alcohol intake. over time. A prolonged, repeated brain alcohol exposure
Here, we review an emerging literature which indi- at considerable levels (above 150–250 mg/dl), interspersed
cates that a reprogramming of mPFC function occurs with alcohol-free intervals, is necessary albeit not sufficient
with prolonged exposure of the brain to cycles of alco- for alcoholism to develop (Heilig et al. 2010). The transition
hol intoxication and withdrawal. This reprogramming into the clinical disorder is slow and gradual, as illustrated
results in molecular dysregulations that contribute to by the fact that the time from meeting diagnostic criteria
the post-dependent syndrome. Convergent evidence for alcohol dependence to seeking treatment is on average
has identified neuroadaptations resulting in altered about a decade (Hasin et al. 2007).
glutamatergic and BDNF-mediated signaling, and for
Epidemiological studies have long indicated that only a
these pathways, direct evidence for a mechanistic role
minority of people exposed to addictive drugs develop addic-
has been obtained. Additional evidence points to a
tion (Anthony et al. 1994). While 87.6% of people aged 18 or
dysregulation of pathways involving calcium home-
ostasis and neurotransmitter release. Recent findings older in the U.S. have consumed alcohol in their lifetime, only
indicate that global DNA hypermethylation is a key 6.8% become affected by an alcohol use disorder (SAMHSA
factor in reprogramming the mPFC genome after a 2014). The way alcohol addiction evolves in a significant
history of dependence. As one of the results of this minority of people who expose their brains to alcohol has
epigenetic remodeling, several histone modifying epige- some important implications. It suggests that neuroadaptive
netic enzymes are repressed. Among these, PR-domain processes are triggered in these individuals, and that these
zinc-finger protein 2, a methyltransferase that selectively processes do not occur in the majority of exposed subjects
mono-methylates histone H3 at lysine 9 has been func- who do not develop a clinical syndrome. Alcohol-induced neu-
tionally validated to drive several of the molecular and roadaptations appear to persistently change brain functions
behavioral long-term consequences of alcohol depen- that affect the ability to control alcohol drinking. A biolog-
dence. Information processing within the mPFC involves ical understanding of these changes requires them to be

86 © 2016 John Wiley & Sons Ltd and International Behavioural and Neural Genetics Society
Reprogramming of mPFC transcriptome

modeled in experimental animals, where they can be stud- continue to consume alcohol following adulteration with qui-
ied at a molecular level. This is a challenging task, because nine to a higher extent than do non-dependent rats, showing
commonly used laboratory mice and rats do not voluntarily evidence of ‘compulsivity’. These behavioral consequences
consume amounts of alcohol sufficient to cause brain alco- of intermittent vapor exposure typically been observed 1–3
hol exposure levels approaching those that occur clinically. weeks after exposure has been terminated, in the absence
An additional challenge is the long duration of brain exposure of physical withdrawal. They have furthermore been studied
required for alcoholism to develop in patients. In rats, follow- up to 6 weeks following termination of exposure, and have
ing several months of voluntary consumption, some behav- appeared stable over this time (Meinhardt & Sommer 2015).
ioral traits believed to be relevant for alcoholism do emerge in The use of chronic intermittent vapor exposure has allowed
a significant minority of animals (Wolffgramm & Heyne 1995). us to study post-dependent neuroadaptations at a molecular
However, exposure levels in this model are still modest, and level. Our initial studies used a candidate-based approach,
the procedure is impractical because of its duration. Models and focused on the role of neuroadaptations involving
based on genetic selection can result in voluntary consump- corticotropin-releasing hormone and its receptors within
tion approaching adequate levels of exposure, but do so at the amygdala complex (Gehlert et al. 2007; Sommer et al.
the expense of other challenges. Perhaps most importantly, 2008). This work, which converged with that of other labora-
genetic selection leads to random allelic fixation throughout tories, has been reviewed previously (Heilig & Koob 2007).
the genome, resulting in a segregation of multiple behavioral In parallel, we have pursued transcriptome-wide approaches
and biological traits of uncertain relation to alcohol seeking as an unbiased discovery strategy, with the objective of
and taking (Sommer et al. 2006; Zhou et al. 2013). identifying novel mechanisms that might ultimately offer
In patients who ultimately develop alcohol addiction, treatment targets. A series of these studies have focused on
brain alcohol exposure occurs with a pattern that alternates the medial prefrontal cortex (mPFC), a brain area that exerts
between cycles of intoxication and abstinence. This type of important top-down control over structures that mediate
intermittent exposure is at the core of two animal models approach and positively reinforced drug seeking, such as
that have gained considerable popularity, every-second-day the Nucleus Accumbens (NAc), as well as structures that
intermittent access (Hopf et al. 2010; Simms et al. 2008; mediate aversion and negatively reinforced drug seeking
Wise 1973, 1975), and long-term drinking with repeated (Koob & Volkow 2010; Peters et al. 2009).
deprivation phases (Spanagel & Holter 1999; Vengeliene In the following, we first briefly review important struc-
et al. 2014). These models produce several behavioral traits tural features of the rodent mPFC, and their potential role
of considerable interest, most importantly an escalation of in the control of behaviors implicated in the control of alco-
voluntary alcohol consumption, and drinking that is resistant hol seeking and drinking. We then review studies that have
to aversive consequences such as quinine adulteration (‘aver- shown a wide-ranging and persistent reprogramming of the
sion resistant’, or ‘compulsive’ drinking). However, unless transcriptome within the mPFC in the post-dependent state,
exposure is for extended periods of time [3–4 months; and the molecular mechanisms we have been able to iden-
(Hopf et al. 2010)], the consequences of alcohol exposure tify in the course of these studies. An implication suggested
in both these models are limited, or of limited duration. by these findings is that molecular switches with broad and
This indicates that, to the extent they trigger neuroadaptive lasting functional consequences are turned on by a history
processes important for the evolution of alcohol addiction, of dependence, and that strategies to turn these switches
these models do so in an incomplete manner. back off may offer targets for novel, disease-modifying
The use of an intermittent brain exposure pattern can be treatments.
pushed a step further through the use of an approach pio-
neered in its basic form by Dora Goldstein, in her clas-
sic studies of alcohol withdrawal (Goldstein & Pal 1971). Functional neuroanatomy
Using this method, an intermittent pattern of brain alcohol of the PFC and control of behaviors
exposure can be imposed on rats through vapor chambers,
inducing blood-alcohol levels relevant for alcoholism (about Drug use despite adverse consequences is a core feature
150–250 mg/dl) that can be maintained for an extended of addiction. It is increasingly recognized that this behavior
period of time [4–7 weeks; (O’Dell et al. 2004; Rimondini does not simply reflect increased motivation to seek and take
et al. 2002, 2003, 2008)]. This results in the emergence of drugs, but rather a breakdown of the balance between sub-
a cluster of traits that persist for a long time after expo- cortical motivational processes and top-down executive con-
sure is terminated, and make up what we have collec- trol, exerted in large part by the prefrontal cortex (Goldstein
tively called ‘the post-dependent state’ (Heilig & Koob 2007; & Volkow 2002, 2011). The past decade has therefore seen
Heilig et al. 2010). Key characteristics of this syndrome, an increased interest in the prefrontal cortex and its role in
and its utility as a tool for drug discovery have recently addiction. This rise has coincided with seminal work that has
been reviewed (Meinhardt & Sommer 2015). In brief, expo- established the structural and functional diversity of rodent
sure using this approach results in escalation of subsequent prefrontal areas, their relation to the primate brain,and their
alcohol intake, both measured as home cage consumption role in attentional and emotional processing (Seamans et al.
on two-bottle free-choice access, and as rates of operant 2008; Uylings et al. 2003).
self-administration. Self-administration on a progressive ratio The prefrontal cortex can be separated into medial, lateral
schedule is also increased, supporting an increased moti- and orbital divisions. In rodents, the orbital and lateral divi-
vation to obtain alcohol. Furthermore, post-dependent rats sions are formed by the orbital frontal cortex and agranular

Genes, Brain and Behavior (2017) 16: 86–100 87


Heilig et al.

insular cortex, while the medial division encompasses the muscimol did not affect context-induced reinstatement of
anterior cingulate cortex (ACC), the prelimbic and the infral- alcohol seeking (Willcocks & McNally 2013).
imbic subdivisions of the mPFC (Uylings et al. 2003). The The functional ensemble that is activated within the infral-
mPFC receives dopaminergic input from the ventral tegmen- imbic mPFC upon recall of an alcohol memory comprises
tal area (VTA), as well as other inputs from limbic and sen- about 10% of infralimbic neurons (Pfarr et al. 2015). This is a
sory areas, including amygdala and hippocampus (Hoover & larger population than that identified by their c-Fos response
Vertes 2007). Subdivisions of the PFC, including the agranu- after similar reinstatement experiments for cocaine or heroin
lar insular cortex, are highly interconnected. This allows them seeking (Bossert et al. 2011; Koya et al. 2009). It can be spec-
to evaluate external and internal states, integrate these with ulated that alcohol cues engage a wider network than other
the motivational salience of specific stimuli – including those drugs, because the more complex actions of alcohol gener-
associated with alcohol or other drugs – and exert executive ate more extensive internal representations. Because abla-
control in selecting approach or avoidance behaviors via pro- tion of the ensemble activated by alcohol cues resulted in
jections to the NAc, amygdala and other targets. Although increased alcohol seeking, the function of this ensemble is
a matter of relative density rather than clear-cut boundaries, likely to inhibit relapse-like behavior. However, because global
the projection patterns of the prelimbic and infralimbic mPFC inactivation of the infralimbic cortex did not affect reinstate-
subdivisions differ, with the former predominantly projecting ment of alcohol seeking, neither when reversible (Willcocks
to the NAc core and the latter predominantly to the shell & McNally 2013) nor permanent (Pfarr et al. 2015), this region
(Vertes 2004). does not seem to generally inhibit reinstatement of alcohol
Anatomical, cellular and functional differences between seeking. Furthermore, the stress-responsive subset of neu-
mPFC subdivisions have given rise to a widely held dichoto- rons within the infralimbic cortex must be different from that
mous view of pre- and infralimbic mPFC function in the activated by alcohol cues, even though both promote the
control of both fear and drug responses (Moorman et al. same behavioral output, i.e. reinstatement of lever pressing.
2015). According to this view, dorsal (largely prelimbic) mPFC Our findings provide further evidence that neural mech-
promotes conditioned approach to both drug and natu- anisms underlying drug seeking differ between drug
ral rewards, whereas the ventral (infralimbic) mPFC sup- classes, and rely on distinct interactions of mPFC circuits
presses it. A similar dichotomy has been thought to exist that are not easily accounted for by a simple ‘prelim-
in controlling the expression and suppression of condi- bic – go/infralimbic – no-go’ model. This view is also
tioned fear responses, respectively. Some support for a supported by recent in vivo multiarray recording experi-
‘prelimbic – go/infralimbic – no-go’ dichotomy in the control ments. The use of this approach has shown that prelimbic
of conditioned appetitive and aversive behaviors has also and infralimbic neurons signal contextual information that
been provided by human neuroimaging findings. These have promotes sucrose seeking in response to discriminative
shown that the activity of dorsal ACC and ventromedial stimuli, irrespective of whether this involves initiation or sup-
PFC correlates with initiation and inhibition of drug and fear pression of behavioral responses (Moorman & Aston-Jones
related behaviors, respectively (Peters et al. 2009). However, 2015). A more complex model of functional compartmental-
because the human PFC has undergone a vast expansion dur- ization of the mPFC proposes that the prelimbic subdivision
ing evolution, it is likely that functional homologs of rodent is mainly involved in the learning of rules, whereas the
mPFC subdivisions are more intermixed within the larger infralimbic supports flexibility of this response and the ability
human mPFC volume. to shift toward new strategies when contingencies change
Evidence has emerged in recent years to challenge the sim- (Heidbreder & Groenewegen 2003; Seamans et al. 2008).
ple ‘go/no-go’ model for the functional contributions of pre-
vs. infralimbic mPFC to drug seeking, respectively. This has
in particular been the case outside the context of cocaine
or fear conditioning with simple discrete cues (Gourley & A reprogrammed mPFC transcriptome
Taylor 2016). A striking example was provided for heroin in the post-dependent state
seeking, where selective deletion of a neuronal ensemble
activated during context-induced reinstatement resulted in For some time, our work has been guided by the hypothesis
suppressed, rather than increased reinstatement respond- that long-term changes in gene expression within key brain
ing on subsequent testing (Bossert et al. 2011, 2012). In structures are a likely mechanism underlying the persistent
contrast, it was found that ablation of a neuronal ensem- behavioral changes observed in the post-dependent state. To
ble activated within the infralimbic cortex by alcohol cues examine this hypothesis, we carried out transcriptome profil-
promoted alcohol seeking. The effect of inactivating the ing of several brain structures from post-dependent and con-
tagged ensemble was specific for the infralimbic cortex, trol rats, and found the most pronounced differences within
while manipulations of the prelimbic cortex were ineffec- the mPFC (Meinhardt et al. 2013). Low magnitudes of dif-
tive. The effect was also selective for cue-induced reinstate- ferential gene expression are typically found in models of
ment, while stress-induced reinstatement was unaffected. psychiatric disorders, largely due to the heterogeneous com-
Importantly, non-selective inactivation of infralimbic neurons position of the brain tissue samples (Reimers et al. 2005). As
was also without consequence for reinstatement behavior a result, the specificity achieved through commonly used sta-
(Pfarr et al. 2015). The latter finding is in agreement with tistical corrections for false discovery rates is at the expense
the observation that a reversible non-selective inactivation of of a low sensitivity, i.e. a low probability that true positives
the infralimbic cortex using a combination of baclofen and will be detected. To facilitate discovery, we therefore chose a

88 Genes, Brain and Behavior (2017) 16: 86–100


Reprogramming of mPFC transcriptome

different strategy, in which we have initially screened for tran- by mGluR2s is a candidate mechanism underlying some of
scripts with nominally (i.e. uncorrected) significant differential these neuroadaptations. Based on these observations, an
expression, and have then sought to confirm candidates of extensive functional validation effort was undertaken, which
interest through independent, wet analysis. This is a strategy is described in detail in the next section.
that is standard in clinical analytics, where optimal results are As a second knowledge-based strategy, a broader, unbi-
commonly achieved through the combined use of separate ased Gene Ontology (GO) analysis was carried out (Tapocik
screening and validation steps, allowing these to prioritize et al. 2013). Overall, 90 of the 165 differentially regulated
sensitivity and specificity, respectively. genes belonged to significantly overrepresented GO cate-
Our initial mPFC screen identified 165 transcripts with nom- gories. Among the top categories of genes with differen-
inally differential expression that persisted 3 weeks follow- tial expression in the mPFC identified as significant by the
ing a history of alcohol dependence (Tapocik et al. 2013). GO analysis were two related categories, associated with
Among the differentially expressed transcripts detected, 105 ion channels and synaptic transmission, respectively. These
were significantly upregulated and 60 significantly downregu- categories are functionally related in that they both have as
lated, arguing against a global repression or cell death due to members genes that influence transmitter release. Specifi-
non-specific toxicity as major factors underlying the findings. cally, several synaptic proteins involved in vesicle docking and
As expected, the fold-changes in gene expression were rela- transmitter release were downregulated, including synapsin
tively small, consistent with what has typically been found II (Syn2) and synaptotagmin 7 (Syt7). This was also the case
for brain expression in psychiatric disorders such as alco- for several ion channels that influence transmitter release
hol dependence (Reimers et al. 2005; Sommer et al. 2005). through effects on membrane potentials and Ca++ flux, such
Among the differentially expressed transcripts identified in as, e.g. the pore-forming alpha 1B subunit of the N-type
this initial screen, 37 had previously been associated with voltage-dependent calcium channel. We have subsequently
alcohol use, abuse and dependence (Tapocik et al. 2013). discovered differential expression of genes encoding addi-
Because a known effect of alcohol is to influence the exci- tional calcium channel and synaptic protein transcripts in an
tation – inhibition balance controlled by gamma aminobutyric RNA sequencing screen of mPFC gene expression in the
acid (GABA) and glutamate, a first, focused knowledge-based
post-dependent model. These findings were confirmed by
follow-up analysis used Gene-Set Enriched Analysis (Sub-
qPCR, and subjected to functional validation (Barbier et al.
ramanian et al. 2005) to examine the potential presence of
2015), in work that is discussed in a subsequent section.
persistent neuroadaptations in GABAergic or glutamatergic
A third category of differentially expressed genes iden-
mPFC neurons. For this purpose, two marker gene sets were
tified by the GO analysis was associated with neuronal
used that have previously been described as highly divergent
plasticity. Within this category, several members of the
between GABAergic and glutamatergic neurons (Sugino
Early Growth Response gene family (Egr1, 2 and 4) were
et al. 2006). The results of this analysis indicated a marked
downregulated together with other transcription factors of
enrichment of downregulated glutamatergic marker genes
the immediate early gene class (e.g. of the Fos, Jun or Nr4a
in the mPFC of post-dependent rats. Several transcripts
families). Downregulation of these transcripts was in each
corresponding to these genes were confirmed by indepen-
case independently confirmed by qPCR of bulk mPFC tissue
dent quantitative polymerase chain reaction (qPCR) or in situ
hybridization analysis (Meinhardt et al. 2013). Among them, extracts or by in situ hybridization; the downregulation of
Grm2, which encodes the metabotropic glutamate receptor Egr2 and Egr4 was also confirmed in identified projection
2 (mGluR2), was markedly downregulated in post-dependent neurons from the infralimbic mPFC to the NAc, isolated
rats, as well as in postmortem tissue from mPFC of human using laser capture microscopy (Meinhardt et al. 2013).
alcoholics. This experiment showed that modest expression changes
The mGluR2 has recently been implicated in control detected in bulk extracts of neuronal tissue may reflect
of alcohol consumption in some Wistar rat populations, much more robust expression differences, 10- to 500-fold,
including the genetically selected Indiana alcohol preferring within specific neuronal populations. A further transcript
P-rat line (Wood et al. 2016; Zhou et al. 2013), and is con- in the category of plasticity-associated genes that was of
sidered a promising candidate medication target (Augier functional interest was brain-derived growth factor (Bdnf ),
et al. 2016; Holmes et al. 2013). In situ hybridization was which was robustly downregulated in the array analysis,
used to localize the downregulation of mGluR2 expression and confirmed by independent qPCR analysis of bulk mPFC
in greater anatomical detail. This analysis once again con- tissue as well as in situ hybridization (Tapocik et al. 2013).
firmed a marked downregulation of mGluR2 expression, The Bdnf and several of the plasticity-associated transcripts
but also showed that this downregulation was restricted to were significantly linked when the array dataset was eval-
the infralimbic mPFC; expression in the prelimbic area was uated using the Ingenuity Pathway Analysis software (IPA,
unaffected. The specificity of infralimbic mGluR2 repression QIAGEN; Methodological details on IPA can be found at
in post-dependent rats was further highlighted by the obser- http://www.ingenuity.com/science/knowledge_base.html.).
vation that the expression of a closely related metabotropic Collectively, the expression data, the bioinformatics analysis,
glutamate receptor subtype, mGluR3, was unaffected by a and prior data indicating a role for Bdnf within corticostriatal
history of dependence, both in the pre- and the infralimbic projections in regulating excessive alcohol intake (Jeanblanc
areas. These findings suggested that the infralimbic mPFC et al. 2006, 2009; Logrip et al. 2009; McGough et al. 2004)
is a key structure affected by alcohol-induced neuroadap- made Bdnf a high priority candidate for a functional validation
tations, and that presynaptic control of glutamate release effort, described in a subsequent section.

Genes, Brain and Behavior (2017) 16: 86–100 89


Heilig et al.

The expression changes within the mPFC of post- history of dependence was assessed in infralimbic mPFC
dependent rats have been replicated in multiple batches neurons that project to the NAc shell (NAcSh). Extracel-
of animals in different laboratory settings. Downregulation lular glutamate levels in the target area of this projection
of Egr1 and Bdnf was also observed in the PFC of mice were measured using in vivo microdialysis in freely moving
immediately following chronic intermittent alcohol exposure, rats. Given its role as a presynaptic autoreceptor, stimula-
but was not detected 8 h into withdrawal (Melendez et al. tion of mGluR2 was expected to reduce glutamate release.
2012). When the expression analysis in this model was As expected, systemic administration of the mGluR2/3 ago-
extended 7 days into abstinence, only a limited overlap with nist LY379268 decreased extracellular glutamate levels in
the rat studies was found within the mPFC (Smith et al. dialysate obtained from the NAcSh of control rats. This effect
2016). Differences between species, procedures and time of the agonist was absent in post-dependent rats. Thus,
points analyzed could each contribute to the differences in downregulation of mGluR2 expression in the mPFC-NAcSh
findings. The time-course is a particularly important factor pathway resulted in a loss of mGluR2 function (Meinhardt
to assess, given recent findings of highly dynamic changes et al. 2013).
in dopamine D1 receptor and dopamine transporter expres- Next, behavioral consequences of suppressed mGluR2
sion in the striatum of post-dependent rats across different expression within this circuit were assessed. Post-dependent
phases following initiation of abstinences (Hirth et al. 2016). rats showed markedly higher response rates following
Limited human data are available to assess the extent to cue-induced reinstatement of extinguished alcohol seeking,
which our rat expression data translate to the human situa- a widely used model of relapse (Sanchis-Segura & Spanagel
tion. Early microarray studies from prefrontal cortex of alco- 2006; Shaham et al. 2003). A rescue of infralimbic mGluR2
holics found robust downregulation of the immediate early expression in the mPFC of post-dependent rats using local
genes (IEG) JunB and Nr4a1 (Iwamoto et al. 2004). A subse- injections of a lentiviral construct, was found to result in
quent report used expression data from rodent and human a rescue of cue-induced alcohol seeking. Following viral
PFC to enrich the results of a genome-wide association study injections, reinstatement response rates of post-dependent
(GWAS) of alcoholism, and identified Jun, Fosl2, Nr4a3 and animals declined by about 40%, bringing them into the
Bdnf as significant hits (Zhao et al. 2012). Recently, a more range of non-dependent controls (Meinhardt et al. 2013).
comprehensive study found that the expression of multiple Together, these experiments provided consistent evidence
ion channels within the PFC was associated with lifetime that a profound, anatomically restricted mGluR2 pathol-
alcohol consumption. Glutamatergic and GABAergic recep- ogy in the mPFC causally contributes to excessive alcohol
tors were among the most central elements of these altered seeking in post-dependent rats; and that restoring mGluR2
gene networks (Farris et al. 2015). Another report that inte- function in this region is sufficient for regaining control over
grated a GWAS of alcohol dependence with human and ani- alcohol-seeking behavior.
mal gene expression data found members of the synaptotag- It will be important to examine possible functional conse-
min and metabotropic glutamate receptor families among its quences of a post-dependent mGluR2 repression for other
top hits (Levey et al. 2014). A brain bank that provides care- behaviors controlled by the mPFC. One useful test for
fully phenotyped postmortem brain tissue samples matched this purpose is the attentional set shifting task (ASST), a
for age and postmortem interval, the Tissue Resource Center rodent equivalent to the human Wisconsin Card Sorting
of the University of South Wales, was used for the following Test (WCST); both assess an aspect of executive function
work (Sheedy et al. 2008). The Grm2 expression was deter- commonly referred to as cognitive flexibility, and their per-
mined in the anterior cingulate cortex (ACC), a brain region formance depends largely on mPFC in both primates and
that is anatomically and functionally related to the rodent rodents (Berg 1948; Brown & Tait 2016). In alcohol-dependent
mPFC (Uylings et al. 2003). Within the ACC, a significant, patients, impaired performance on the WCST is a predic-
2.6-fold decrease in Grm2 transcript levels was found in alco- tor of relapse (Wicks et al. 2001). In mice, experiments have
holics compared with controls (Meinhardt et al. 2013). shown performance deficits in the ASST after 1 week of absti-
In summary, there is convergent evidence for long-lasting nence from chronic intermittent alcohol exposure (Kroener
reprogramming of gene expression networks in the pre- et al. 2012). Patch clamp recordings in brain slices from these
frontal cortex by a history of alcohol dependence, with mice showed increased presynaptic transmitter release, con-
some candidate genes receiving support from both human sistent with impaired mGluR2 autoreceptor function.
and rodent studies. This holds particularly true for the The mechanism underlying reduced mGluR2 expression in
metabotropic glutamate receptor mGluR2 and for the the mPFC of post-dependent rats is presently not known. The
neurotrophin Bdnf . Both these potentially offer molecular Grm2 was not detected by a DNA methylation screen (see
below), but could be a target for other types of direct epige-
targets for novel alcoholism therapeutics, and therefore
netic regulation that have not been assessed in those stud-
merit a detailed functional analysis.
ies. For instance, treatment with L-acetylcarnitine, a drug that
promotes histone acetylation and is approved as a medica-
tion for the treatment of neuropathic pain, leads to increased
Functional role of infralimbic mGluR2 receptor K27-acetylation of H3 histones bound to the Grm2 promoter
repression as well as Bdnf promoter in the PFC, and this is associ-
ated with respective increased expression of the respective
In a series of experiments, the functional consequences of gene in this region, as well as rapid antidepressant actions in
the suppressed mGluR2 expression observed following a rodent models of depression (Nasca et al. 2013). Also, mice

90 Genes, Brain and Behavior (2017) 16: 86–100


Reprogramming of mPFC transcriptome

with a deletion of the 5-HT2A receptor show repressive his- these dysregulations is unknown. Human studies offer lim-
tone marks at the Grm2 promoter, and this is associated with ited opportunities to isolate the consequences of brain alco-
a pronounced downregulation of prefrontal Grm2 expres- hol exposure from those of preexisting vulnerability factors,
sion (Kurita et al. 2013). Regulation of mGluR2 expression by or of environmental influences associated with alcoholism
alcohol exposure could also be indirect, resulting from the such as head trauma or nutritional deficiency. Animal mod-
downregulated expression of transcription factors belonging els offer an important complementary tool for this type of
to the Egr-family (Meinhardt et al. 2013). The Grm2 promoter analysis.
contains binding sites for this family of transcription factors, The smaller number of miRNAs (about 1800 in the rat
and viral mediated Egr1 overexpression can induce mGluR2 genome) and their lower complexity make the analysis of
expression (Kurita et al. 2013). Furthermore, Egr2, which has their expression more tractable than that of mRNA tran-
a specific binding site within the Grm2 promoter, can itself scripts. Microarray-based screening for differential miRNA
be subject to epigenetic regulation via hypermethylation of expression in the mPFC of individual post-dependent rats
intron 1 (Swanberg et al. 2009; Unoki & Nakamura 2003). It resulted in 41 candidates (Tapocik et al. 2013). Among these,
is thus possible that a network of IEG is dysregulated by epi- 17 were upregulated and 24 were decreased, again suggest-
genetic mechanisms, ultimately leading to reduced Grm2 in ing that non-specific global processes such as cell death were
the mPFC of post-dependent rats. unlikely to account for the observations. Principal component
Small molecule mGluR2/3 agonists are available, and are analysis indicated that a history of dependence accounted for
able to reduce self-administration and drug seeking for sev- a higher proportion of the variance in miRNA than in mRNA
eral drugs of abuse, including alcohol, nicotine, cocaine, expression. Accordingly, hierarchical clustering of miRNA
methamphetamine and heroin (Baptista et al. 2004; Bossert expression segregated post-dependent and control animals
et al. 2006; Cannella et al. 2013; Crawford et al. 2013; Liechti more robustly than that of mRNA expression. Thus, the effect
et al. 2007; Peters & Kalivas 2006; Zhao et al. 2006). These size of post-dependent neuroadaptations is higher at the
studies indicate that presynaptic group II mGluRs act as gate- miRNA than at the mRNA level.
keepers that regulate glutamate release at synapses rele- The bioinformatic analysis identified 89 mPFC mRNA tran-
vant for addictive behaviors. Available orthosteric agonists are scripts as being potentially regulated by miRNAs. These were
unable to discriminate between mGluR2 and mGluR3, but subjected to a pathway analysis using the Ingenuity soft-
positive allosteric modulators (PAMs) selective for mGluR2 ware tool. A major pathway that emerged from this anal-
have recently been developed (Augier et al. 2016; Holmes ysis was associated with neuronal plasticity (Fig. 1). This
et al. 2013; Johnson & Lovinger 2015; Nickols & Conn 2014). network contained multiple genes thought to be involved in
Using these PAMs, several investigators have been able to neuropsychiatric disorders and addiction, including Bdnf , Fos,
demonstrate that mGluR2 activation can reduce drug intake Per2 and Arc, as well as the nuclear receptors/transcription
and drug-seeking behaviors (Caprioli et al. 2015; Justinova factors Nr4a1 and Nr4a3. The 15 differentially expressed
et al. 2015; Nikiforuk et al. 2010). Clinical trials are currently mRNAs within this network were targeted by 26 of the
ongoing to evaluate whether these promising data can be miRNAs found in the miRNA microarray analysis, with sev-
translated to humans (Salih et al. 2015). eral of the miRNAs converging on the same mRNA targets
(Tapocik et al. 2013). This observation is in agreement with
prior observations that multiple miRNAs can act synergis-
microRNA reprogramming of the mPFC tically to repress gene expression (Nachmani et al. 2010).
Several candidate miRNA–mRNAs pairs identified by these
analyses above were chosen for confirmation by qPCR. For
Expression regulation by microRNAs (miRNAs) represents an
the miRNAs selected for confirmation, results were highly
appealing candidate mechanism for post-dependent neuroad-
consistent with the microarray data; for the selected mRNAs,
aptation of mPFC function. The miRNAs are small (∼22–30
confirmation was obtained in most, but not all cases. Notably,
nucleotide in length), highly conserved noncoding RNAs that
an experimentally robust and functionally compelling expres-
modulate gene expression at the posttranscriptional level.
sion pair that emerged from this analysis was that of Bdnf
After being processed into a mature form, they bind to miRNA
and miR-206.
recognition elements in the 3′ -untranslated region (UTR) of
their target transcripts and cause translational degradation
or repression (Guo et al. 2010). The miRNA regulation of
mRNA expression is inherently divergent, because recog- A functional role for miRNA-mediated BDNF
nition elements are shared between multiple transcripts. A repression in the mPFC
limited set of miRNAs could therefore account for a substan-
tial proportion of differential gene expression observed in the Dysregulation of BDNF expression within the mPFC
post-dependent state at the mRNA level. offers a potential mechanism for the persistent neuroad-
The miRNAs may play an important role in neuroadaptive aptations observed in the post-dependent state. The
processes triggered by chronic drug exposure (Pietrzykowski BDNF signaling leads to phosphorylation of the cAMP
2010), including tolerance to alcohol (Pietrzykowski et al. response element-binding protein (CREB) and upregula-
2008). Postmortem analysis has also identified upregulation tion of CREB target genes, including the activity-regulated
of multiple miRNAs in the frontal cortex of alcohol-dependent cytoskeleton-associated (ARC) protein. The ARC is of partic-
patients (Lewohl et al. 2011), but the functional role of ular interest for long-term neuroadaptive processes, because

Genes, Brain and Behavior (2017) 16: 86–100 91


Heilig et al.

Figure 1: Ingenuity pathway analysis showing one of the top gene networks that is downregulated in the mPFC by history of
alcohol dependence. Green color indicates decreased expression in post-dependent compared with control rats. Red arrows indicate
genes regulated by DNA methylation. Blue arrows indicate genes regulated by Prdm2.

it is directly involved in remodeling of neuronal connectivity of BDNF occurs (An et al. 2008). The 3′ -UTR of the long Bdnf
via dendritic spine formation. For instance, physical modu- transcript contains three predicted target sites for miR-206,
lation of spine density by ARC is required for consolidation and is repressed by this miRNA in the course of muscle cell
of long-term synaptic potentiation (reviewed in Bramham differentiation (Miura et al. 2012).
2008). Previous research has also shown that ARC medi- Under normal circumstances, miR-206 does not seem to
ates alcohol-induced neuroadaptations within the amygdala be expressed in the brain at significant levels, but its expres-
(Pandey et al. 2008). sion is induced by accumulation of amyloid in a transgenic
The Bdnf gene has a unique, highly conserved organiza- mouse model of Alzheimers disease, where inhibition of
tion. In rodents, it is composed of at least eight 5′ -exons, miR-206 activity resulted in a rescue of low BDNF levels and
each with its own promoter, that are spliced to a common spine density numbers (Lee et al. 2012). The miR-206 induc-
3′ -terminal exon containing the coding frame and two alter- tion may therefore be a response to neuronal insult. Based
native 3′ -UTRs (Aid et al. 2007; Timmusk et al. 1993). The on these findings, a chain of events can be hypothesized to
promoters for exons I and II mediate constitutive expression, occur as post-dependent neuroadaptations develop. Accord-
while the promoters for exons IV and VI have been shown to ing to this hypothesis, miR-206 is induced in mPFC neurons
share properties with IEG and seem to mediate much of the by cycles of intoxication and withdrawal, represses BDNF
activity-dependent gene regulation of Bdnf (Hansson et al. expression, and results in decreased expression and activity
2006, 2011; Jiang et al. 2008). The short and long 3′ -UTR Bdnf of ARC. Decreased activity of ARC then contributes to last-
transcripts are involved in different cellular functions. Short ing changes in the connectivity of mPFC neurons with their
3′ -UTR mRNAs are restricted to cell bodies, while long 3′ -UTR projection targets, and impaired top-down regulation of moti-
mRNAs are also localized in dendrites, where local synthesis vated behaviors.

92 Genes, Brain and Behavior (2017) 16: 86–100


Reprogramming of mPFC transcriptome

A series of studies was carried out to evaluate this hypoth- However, some data additionally suggest that a global
esis (Tapocik et al. 2014). In situ hybridization confirmed a per- potentiation of BDNF activity may in fact have a therapeutic
sistent induction of miR-206 in the mPFC of post-dependent potential. Specifically, a hemizygous deletion of BDNF was
rats. This induction was region specific, as no increase was reported to result in increased alcohol consumption in mice
seen in the VTA, NAc or the amygdala. When the activity of (Hensler et al. 2003), while systemic injections of a fusion
miR-206 was examined in vitro using a luciferase reporter protein that delivers Rack1 to the brain and promotes BDNF
assay, a 60% repression of the Bdnf reporter construct was activity resulted in decreased drinking. These findings pro-
found. The three miR-206 target sites in the Bdnf 3′ -UTR vide a rationale for developing small molecule BDNF mimics
contributed synergistically to this effect, whereby the pres- as therapeutics for alcohol addiction.
ence of two of the sites, site 2 (located at bp 392–398)
and 3 (bp 1294–1300) is necessary. Functional in vivo val-
idation of these results was obtained by adeno-associated Global hypermethylation of the mPFC
virus (AAV)-mediated overexpression of miR-206 in the mPFC and synaptic transmission
of rats that did not have any history of alcohol depen-
dence. Vector-mediated miR-206 expression resulted in a Similar to miRNAs, epigenetic mechanisms (i.e. DNA methy-
significant reduction of BDNF levels within the mPFC. This lation and histone modification) represent an attractive
was accompanied by a significant escalation of alcohol candidate mechanism to drive alcohol-induced long-term
self-administration rates in these animals, mimicking what is neuroadaptation. Like miRNAs, DNA methylation and his-
observed following a history of dependence. tone modification can regulate multiple genes and therefore
Together, these findings provide consistent support for the be one of the main upstream regulators of alcohol-induced
hypothesis that miR-206 expression is induced in the mPFC transcriptomic reprogramming. Moreover, DNA methylation
of post-dependent rats, and causally contributes to one of has the ability to change dynamically in response to external
the core elements of the resulting behavioral syndrome, factors. For instance, stress (Murgatroyd et al. 2009; Weaver
escalation of alcohol self-administration, through repression et al. 2002) and exposure to drugs of abuse (Tian et al.
of BDNF. This notion has subsequently received indepen- 2012; Wong et al. 2011) regulate methylation patterns in the
dent support by data from the laboratory of Dorit Ron. brain. Both human and animal studies suggest that alcohol
Using a mouse model, this group showed that another can also modify methylation patterns in the brain and more
miRNA, miR-30a-5p, mediates a transition to escalated drink- specifically in the mPFC. Manzardo and colleagues found
ing through repression of BDNF expression within the mPFC no global methylation differences between alcoholics and
(Darcq et al. 2015). control subjects. However, they found different methyla-
A role for BDNF in addiction-related behaviors and neu- tion patterns at the gene promoter level between control
roadaptations has previously been reported in studies with and alcoholic subjects, suggesting a ‘gene specific effect’
cocaine, but these effects have been highly dependent on (Manzardo et al. 2012). In accordance with this study, Wang
the brain region examined (reviewed in McGinty et al. 2010). and his collaborators recently observed different methylation
Specifically, BDNF injections into the mPFC have been found patterns between mPFC tissue of controls and subjects with
to result in decreased cocaine self-administration and sup- alcoholism (Wang et al. 2016). Contrary to Manzardo’s study,
pression of both cue- and priming-induced relapse to cocaine which measured methylation level at the promoter region
seeking. In contrast, BDNF administration into the VTA or the only, Wang et al., extended their analysis to the full gene
NAc has largely produced the opposite effect. The mPFC find- sequence and measured methylation at individual CpGs.
ings on alcohol intake are generally in agreement with the They found that the majority of the differentially methylated
cocaine studies in which BDNF has been administered into CpGs identified in male alcoholics were hypermethylated
this brain area. The cocaine studies suggest the possibility and were dispersed over different gene regions. These
that BDNF may simply be a general mediator of drug-induced results suggest that alcohol-induced hypermethylation in
plasticity, with different functional effects depending on the non-promoter regions that comprise, for example, enhancer
circuitry in which it acts. Based on the presented mPFC and insulator regions, may regulate splicing and gene expres-
data alone, it is therefore difficult to predict the net effect of sion by influencing the binding of regulatory proteins (Jones
interventions with global effects on BDNF activity for alco- 2012). More experiments are needed in order to better
hol addiction. Indeed, opposite effects of escalating alco- understand this mechanism.
hol intake have been reported on cortical and striatal BDNF Although studies of DNA methylation in human post-
expression, i.e. decreased expression in the mPFC and mortem brain tissues are informative, this approach does not
increase in the striatum (Jeanblanc et al. 2006, 2009; Logrip allow for determination of the functional consequences of
et al. 2009; McGough et al. 2004). It is unclear whether this differential DNA methylation. Animal models offer an impor-
reflects the actions of locally produced BDNF within this tant complement to these studies, as they allow assessment
structure, BDNF provided by inputs to the striatum such as of dependence-induced DNA methylation changes to be car-
those originating from the prefrontal cortex, or both. At a min- ried out in relevant brain regions. More importantly, they
imum, findings to date support the notion that remodeling of allow the evaluation of the functional impact of DNA methy-
mPFC output connectivity is an important mechanism in the lation changes, as it is possible to manipulate the enzymes
transition to excessive alcohol drinking, and that BDNF is an that regulate DNA methylation levels and assess the behav-
important mediator of this remodeling. ioral consequences. For example, manipulation of DNA

Genes, Brain and Behavior (2017) 16: 86–100 93


Heilig et al.

methyltransferase in a rat model suggests that DNA methy- that drive compulsive-like behavior observed in alcohol
lation is involved in alcohol addiction. Systemic inhibition of post-dependent rat (Vendruscolo et al. 2012).
DNA methyltransferase activity (DNMT) activity decreased
excessive alcohol drinking and seeking behaviors in rodents
(Warnault et al. 2013). Similarly, intraventricular infusion of Fine-tuning histone function: PRDM2
the DNMT inhibitor RG-108 significantly decreased alcohol
self-administration in post-dependent but not in control In addition to direct methylation of DNA, environmentally
rats (Barbier et al. 2015). Interestingly, micro-infusion of sensitive epigenetic mechanisms can also manifest as post-
RG-108 into the mPFC was sufficient to inhibit escalation translational modifications to unstructured histone tails (Feil
in alcohol intake in post-dependent rats, suggesting that & Fraga 2011). Histone tail modifications include acetyla-
alcohol-induced hypermethylation in this region is involved in tion, methylation and phosphorylation, among others. While
increased alcohol intake. there are three enzymes that catalyze DNA methylation
In order to identify methylation-dependent gene expres- (DNMT1, DNMT3A, DNMT3B), there are numerous epige-
sion changes that occur after a history of alcohol depen- netic enzymes that oversee the addition, interpretation and
dence, whole transcriptome analysis was performed using removal of various histone modifications (Borrelli et al. 2008).
RNA-seq (Barbier et al. 2015). Seven hundred and eighty-four For example, there are over 50 human epigenetic enzymes
genes with significant expression changes in the mPFC that catalyze the addition of methyl groups to histones (his-
of post-dependent compared with control rats were identi- tone methyltransferases), and approximately 30 enzymes
fied. Similar to what was previously found (Meinhardt et al. that remove these methyl groups (histone demethylases)
2013; Tapocik et al. 2013), bioinformatics analysis identified (Copeland et al. 2009). Histone methylation can either facil-
within this list two overrepresented categories of biologi- itate or repress transcription depending on the particular
cal functions related to gene expression and neurotransmis- amino acid residue that is modified. For example, methyla-
sion. Specifically, a history of alcohol dependence decreased tion at histone H3 lysine 4 (H3K4) is typically associated with
expression of a gene network that includes proteins reg- transcriptional activation, while methylation at H3K27 con-
ulating synaptic vesicle formation and function (Fig. 1). A denses chromatin and inhibits gene expression. This ‘histone
subset of seven genes coding for synaptic proteins (synap- code’ is further complicated by the fact that lysine can be
totagmin 1: Syt1, synaptotagmin 2: Syt2, P/Q type chan- mono-, di- or tri-methylated, and the degree of methylation
nel: Cacna1a, T-type calcium channel: Cacan1i and the can differentially influence gene expression. Each epigenetic
serine/threonine-protein kinase 1 and 2: Wnk1 and Wnk2, enzyme specifically catalyzes modification of a particular
and Potassium Channel, voltage-gated Shaw related sub- amino acid on a particular histone, and further exhibits
specificity toward the degree of methylation at that site.
family C, member 1: Kcnc1) was selected for subsequent
Despite the fact that many of these epigenetic enzymes are
analysis. Expression of these genes was not influenced by
expressed in the brain, little is known about their neurological
alcohol exposure in other brain regions known to play a
function.
role in addiction (Nac, amygdala and hippocampus). These
RNA-seq analysis showed that 22 epigenetic enzyme
data suggest a persistent dysregulation of synaptic trans-
mRNAs were dysregulated in mPFC tissue from post-
mission in the mPFC after a history of alcohol dependence,
dependent rats (Barbier et al. 2015). One of these enzymes,
which is consistent with the hypothesis of mPFC hypoac-
histone methyltransferase PR-domain containing 2 (PRDM2),
tivity associated with drug addiction (Goldstein & Volkow
is strongly enriched in the brain compared with periph-
2011).
eral tissues and is also selectively expressed in neurons.
Importantly, we found that micro-infusion of RG-108 into Further, qPCR showed that downregulation of Prdm2 in
the mPFC restored the expression of four out of the seven alcohol-dependent rats could be restored by treatment with
transcripts (Syt1, Syt2, Cacna1a and Wnk2), indicating a the DNA methyltransferase inhibitor RG-108, suggesting
regulatory effect of alcohol-induced DNA hypermethylation that PRDM2 is part of an epigenetic pathway downstream
on those genes. Pyrosequencing analysis did not show of DNA methylation changes. While PRDM2 had previously
altered methylation on the promoter region of Cacna1a, sug- been implicated in brain cancer, no studies had examined a
gesting an indirect regulation of this transcript’s expression role for PRDM2 in psychiatric conditions.
by DNA methylation. In contrast, chronic intermittent alcohol In order to test whether PRDM2 contributes to alcohol-
exposure did induce hypermethylation on exon 1 of Syt2. seeking behavior, a lentiviral-mediated siRNA knockdown
There is now evidence suggesting that, similar to promoter approach was used to downregulate Prdm2 expression in
hypermethylation, DNA methylation on exon 1 is associated the mPFC of alcohol-naïve rats (Barbier et al. 2015). The
with gene silencing (Brenet et al. 2011). Together, these Prdm2 knockdown was sufficient to cause escalated alcohol
results suggest a direct regulation of Syt2 by alcohol-induced self-administration, aversion-resistant drinking in a quinine
DNA hypermethylation. Interestingly, it was also found that adulteration task, and increased sensitivity to stress-induced
although Syt2 knockdown in non-dependent rats does not reinstatement, essentially resulting in a phenocopy of the
alter alcohol consumption, it increases tolerance to quinine post-dependent state. These effects were specific for alcohol
adulteration. Aversion-resistant alcohol intake has been intake, as Prdm2 knockdown did not affect sucrose-seeking
previously interpreted to indicate compulsive motivation to behavior.
consume alcohol (Wolffgramm et al. 2000). Therefore, Target genes of PRDM2 that could underlie the observed
decreased Syt2 may be part of the molecular events alcohol-seeking behaviors, were identified using a tripartite

94 Genes, Brain and Behavior (2017) 16: 86–100


Reprogramming of mPFC transcriptome

Figure 2: Post-dependent reprogramming of transcriptome responses in the mPFC affects local cue-responsive neuronal
networks. Local neuronal networks characterized by coherent activity play a critical role in mPFC function. We hypothesize that such
functional ensembles are loosely formed by many neurons during early learning (task acquisition) and as performance consolidates these
networks become more stable, comprised of discrete sets of neurons. A distinct feature associated with coherent neuronal activity is
stimulus-induced expression of Fos (here represented by blue labeled neurons across most of the mPFC after presentation of a cue)
and of other IEG, which in turn regulate downstream effector genes. These transcriptional responses are coordinated by epigenetic
mechanisms and are crucial for shaping future outcomes of neural responses from the mPFC that ultimately determine the level
of control over behavior. A history of alcohol dependence – such in the post-dependent state – causes epigenetic dysregulation and
blunted IEG responses. This molecular reprogramming is likely impacting on neuronal communication and consequently on ensemble
formation, which either could result in fewer neurons participating or lessened stimulus-driven transcriptional activity in the ensembles.
This may underlie reduced behavioral flexibility and consequently increased risk of relapse.

approach. The PRDM2 is known to specifically catalyze the genes to those most likely to be functionally involved in
addition of a single methyl group at histone H3, lysine 9 the ability of the Prdm2 knockdown to create a phenocopy
(H3K9me1) resulting in transcriptional activation (Barski et al. of the post-dependent state. There were four convergent
2007; Kim et al. 2003). The H3K9me1 is decreased in the target genes identified by our ChIP-seq, RNA-seq and Prdm2
mPFC of post-dependent rats, consistent with Prdm2 down- knockdown studies: Bsn, Cacna1i, Cacna1d and Wnk2. All
regulation. Therefore, we first used chromatin immunopre- four genes were downregulated, consistent with PRDM2
cipitation followed by DNA sequencing (ChIP-seq) to identify downregulation, decreased H3K9me1, and subsequent tran-
the genomic regions differentially regulated by H3K9me1 in scriptional repression in alcohol dependence. Cacna1i and
control as compared to post-dependent mPFC. These results Cacna1d encode subunits of voltage-gated calcium chan-
were overlaid with the corresponding RNA-seq data in order nels, while Bsn encodes Bassoon, a protein that supports
to narrow down the list of H3K9me1 target genes to those calcium channel activity by anchoring the channels to the
that are also transcriptionally dysregulated as a consequence presynaptic terminal. The WNK2 is a kinase that regulates
of protracted alcohol exposure. Pathway analysis of these cation-chloride transporters, and along with Cacna1i and
genes showed a significant enrichment of biological functions Cacna1d, was part of the gene network we found to be
related to synaptic communication, specifically regulation of regulated by DNA methylation in dependent rats (Barbier
voltage-gated calcium signaling (Barbier et al. 2015). et al. 2015); see Fig. 1). Calcium channels have previously
Finally, laser capture microdissection was used to quantify been implicated in altered patterns of cortical excitability
the expression of known alcohol-associated genes in mPFC that may underlie excessive alcohol intake and increase the
neurons of alcohol-naïve rats subjected to Prdm2 siRNA propensity for relapse. For instance, nonhuman primates
compared to scramble control. This third arm of our tripartite that chronically self-administer alcohol display plasticity in
approach allowed us to narrow the list of H3K9me1 target T-type calcium channel currents that is sensitive to periods

Genes, Brain and Behavior (2017) 16: 86–100 95


Heilig et al.

of intoxication and withdrawal (Carden et al. 2006; Welsh in loss of structural plasticity that is required for long-term
et al. 2011). Furthermore, our findings are in accordance maintenance of synaptic plasticity (Tapocik et al. 2013, 2014).
with prior postmortem studies, which have shown changes Repression of synaptic proteins and Ca++ channels is likely to
in the expression pattern of genes involved in synaptic result in impaired transmitter release from affected cell pop-
neurotransmission (Liu et al. 2006, 2007). ulations, leading to a loss of inhibitory control over alcohol
The fact that this same gene network was implicated intake despite adverse consequences (Barbier et al. 2015).
in distinct experimental approaches emphasizes the valid- In conclusion, neuronal populations within the mPFC form
ity and central importance of this epigenetically regulated dynamic ensembles that control alcohol-related behaviors
signaling pathway in mediating dependence-associated neu- in a stimulus-specific manner (Fig. 2). Formation of these
roadaptive changes within the mPFC. Interestingly, func- ensembles and their output depends on rapid transcriptional
tional enrichment analysis of miRNAs significantly enriched responses mediated by immediate early gene networks, as
for target genes within the H3K9me1 ChIP-seq results well as more lasting epigenetic mechanisms that shape the
showed miR-206 as the number one hit (WebGestalt adj. transcriptional program of these cells. A prolonged exposure
P = 0.0004), relating these recent findings to our earlier stud- to cycles of alcohol intoxication and withdrawal results in
ies (Tapocik et al. 2014). Therefore, a plausible hypothesis persistent dysregulation of gene expression and function
to test in future experiments is that PRDM2 directly medi- within these circuits. This dysregulation is in large part medi-
ates miR-206 dysregulation in alcohol dependence, or that ated through a broad range of epigenetic mechanisms that
Prdm2 knockdown or miR-206 overexpression both cause include induction of miR-expression, altered DNA methyla-
escalation of alcohol self-administration by targeting overlap- tion and altered activity of epigenetic enzymes that modify
ping gene networks. Altogether, this line of experimental histone function. The resulting functional impairment inter-
inquiry showed that PRDM2-mediated histone modifications feres with fundamental mechanisms of top-down control
are an essential component of genomic reprogramming in of motivational and emotional processes. The breakdown
the mPFC that contributes to neuroadaptive changes under- of top-down control promotes the risk of relapse in alco-
lying a transition into alcohol addiction. Because epigenetic holism, but presently we have only an incomplete under-
enzymes such as PRDM2 are emerging as novel drug- standing of the exact circuit-level mechanisms through which
gable targets with potential to treat complex human disease this occurs. We hypothesize that future research leading to
(Copeland et al. 2009), this work underscores the potential an improved understanding of mPFC function and its dys-
for epigenetic-targeted therapies to impact substance abuse regulation could contribute to the development of treatment
disorders. responsive biomarkers, and the development of improved,
personalized therapies (Heilig et al. 2016).

Integrating molecular changes with mPFC


function References
Aid, T., Kazantseva, A., Piirsoo, M., Palm, K. & Timmusk, T. (2007)
Our work over the past 5 years has provided evidence that Mouse and rat BDNF gene structure and expression revisited. J
prolonged brain alcohol exposure causes a lasting repro- Neurosci Res 85, 525–535.
gramming of mPFC transcriptional activity and function. An, J.J., Gharami, K., Liao, G.Y., Woo, N.H., Lau, A.G., Vanevski, F.,
Torre, E.R., Jones, K.R., Feng, Y., Lu, B. & Xu, B. (2008) Distinct
It remains to be determined by what exact mechanisms
role of long 3′ UTR BDNF mRNA in spine morphology and synaptic
that these molecular changes impact the control of alco- plasticity in hippocampal neurons. Cell 134, 175–187.
hol seeking. Alcohol cues induce neural activity through- Anthony, J.C., Warner, L.A. & Kessler, R.C. (1994) Comparative epi-
out the entire mPFC, as shown by our c-fos studies (Pfarr demiology of dependence on tobacco, alcohol, controlled sub-
et al. 2015) and this process is altered in the post-dependent stances, and inhalants: basic findings from the National Comorbid-
state (Meinhardt et al. 2013). Although only the deletion of ity Survey. Exp Clin Psychopharmacol 2, 244–268.
Fos-positive neurons within the infralimbic subregion has Augier, E., Dulman, R.S., Rauffenbart, C., Augier, G., Cross, A.J.
& Heilig, M. (2016) The mGluR2 positive allosteric modulator,
affected cue-induced reinstatement, activated neurons in AZD8529 and cue-induced relapse to alcohol seeking in rats. Neu-
other mPFC subregions may serve other behavioral pro- ropsychopharmacology. 2016 Jul 20. doi: 10.1038/npp.2016.107.
cesses. The emerging picture is that mPFC neurons can form [Epub ahead of print]
highly dynamic networks. These seem to cross anatomical Baptista, M.A., Martin-Fardon, R. & Weiss, F. (2004) Preferential
boundaries, and differentially contribute to behavioral output effects of the metabotropic glutamate 2/3 receptor agonist
according to contextual demands at the time. LY379268 on conditioned reinstatement versus primary reinforce-
ment: comparison between cocaine and a potent conventional
Similar to the functional studies, the epigenetic dysregula- reinforcer. J Neurosci 24, 4723–4727.
tion identified by our molecular studies affects both infralim- Barbier, E., Tapocik, J.D., Juergens, N., Pitcairn, C., Borich, A., Schank,
bic and prelimbic subregions, and is likely to alter the dynamic J.R., Sun, H., Schuebel, K., Zhou, Z., Yuan, Q., Vendruscolo, L.F.,
communication within and between mPFC networks in mul- Goldman, D. & Heilig, M. (2015) DNA methylation in the medial
tiple ways. A loss of autoreceptor function normally provided prefrontal cortex regulates alcohol-induced behavior and plasticity.
by mGluR2 in corticostriatal projection neurons impairs their J Neurosci 35, 6153–6164.
Barski, A., Cuddapah, S., Cui, K., Roh, T.Y., Schones, D.E., Wang, Z.,
capacity for synaptic plasticity, as showed by the loss of post-
Wei, G., Chepelev, I. & Zhao, K. (2007) High-resolution profiling of
synaptic long-term depression in animals that do not express histone methylations in the human genome. Cell 129, 823–837.
mGluR2 due to a stop-codon mutation (Meinhardt et al. 2013; Berg, E.A. (1948) A simple objective technique for measuring flexibil-
Zhou et al. 2013). Loss of BDNF expression is likely to result ity in thinking. J Gen Psychol 39, 15–22.

96 Genes, Brain and Behavior (2017) 16: 86–100


Reprogramming of mPFC transcriptome

Borrelli, E., Nestler, E.J., Allis, C.D. & Sassone-Corsi, P. (2008) Decod- Gourley, S.L. & Taylor, J.R. (2016) Going and stopping: dichotomies
ing the epigenetic language of neuronal plasticity. Neuron 60, in behavioral control by the prefrontal cortex. Nat Neurosci 19,
961–974. 656–664.
Bossert, J.M., Gray, S.M., Lu, L. & Shaham, Y. (2006) Activation of Guo, H., Ingolia, N.T., Weissman, J.S. & Bartel, D.P. (2010) Mammalian
group II metabotropic glutamate receptors in the nucleus accum- microRNAs predominantly act to decrease target mRNA levels.
bens shell attenuates context-induced relapse to heroin seeking. Nature 466, 835–840.
Neuropsychopharmacology 31, 2197–2209. Hansson, A.C., Sommer, W.H., Metsis, M., Stromberg, I., Agnati,
Bossert, J.M., Stern, A.L., Theberge, F.R., Cifani, C., Koya, E., Hope, L.F. & Fuxe, K. (2006) Corticosterone actions on the hippocampal
B.T. & Shaham, Y. (2011) Ventral medial prefrontal cortex neuronal brain-derived neurotrophic factor expression are mediated by exon
ensembles mediate context-induced relapse to heroin. Nat Neu- IV promoter. J Neuroendocrinol 18, 104–114.
rosci 14, 420–422. Hansson, A.C., Rimondini, R., Heilig, M., Mathe, A.A. & Sommer,
Bossert, J.M., Stern, A.L., Theberge, F.R., Marchant, N.J., Wang, W.H. (2011) Dissociation of antidepressant-like activity of esci-
H.L., Morales, M. & Shaham, Y. (2012) Role of projections from talopram and nortriptyline on behaviour and hippocampal BDNF
ventral medial prefrontal cortex to nucleus accumbens shell in expression in female rats. J Psychopharmacol 25, 1378–1387.
context-induced reinstatement of heroin seeking. J Neurosci 32, Hasin, D.S., Stinson, F.S., Ogburn, E. & Grant, B.F. (2007) Prevalence,
4982–4991. correlates, disability, and comorbidity of DSM-IV alcohol abuse
Bramham, C.R. (2008) Local protein synthesis, actin dynamics, and and dependence in the United States: results from the National
LTP consolidation. Curr Opin Neurobiol 18, 524–531. Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen
Brenet, F., Moh, M., Funk, P., Feierstein, E., Viale, A.J., Socci, N.D. & Psychiatry 64, 830–842.
Scandura, J.M. (2011) DNA methylation of the first exon is tightly Heidbreder, C.A. & Groenewegen, H.J. (2003) The medial prefrontal
linked to transcriptional silencing. PLoS One 6, e14524. cortex in the rat: evidence for a dorso-ventral distinction based upon
Brown, V.J. & Tait, D.S. (2016) Attentional set-shifting across species. functional and anatomical characteristics. Neurosci Biobehav Rev
Curr Top Behav Neurosci 28, 363–395. 27, 555–579.
Cannella, N., Halbout, B., Uhrig, S., Evrard, L., Corsi, M., Corti, C., Heilig, M. & Koob, G.F. (2007) A key role for corticotropin-releasing
Deroche-Gamonet, V., Hansson, A.C. & Spanagel, R. (2013) The factor in alcohol dependence. Trends Neurosci 30, 399–406.
mGluR2/3 agonist LY379268 induced anti-reinstatement effects in Heilig, M., Egli, M., Crabbe, J.C. & Becker, H.C. (2010) Acute with-
rats exhibiting addiction-like behavior. Neuropsychopharmacology drawal, protracted abstinence and negative affect in alcoholism: are
38, 2048–2056. they linked? Addict Biol 15, 169–184.
Caprioli, D., Venniro, M., Zeric, T., Li, X., Adhikary, S., Madangopal, R., Heilig, M., Sommer, W.H. & Spanagel, R. (2016) The need for treat-
Marchant, N.J., Lucantonio, F., Schoenbaum, G., Bossert, J.M. & ment responsive translational biomarkers in alcoholism research.
Shaham, Y. (2015) Effect of the novel positive allosteric modulator Curr Top Behav Neurosci 28, 151–171.
of metabotropic glutamate receptor 2 AZD8529 on incubation of Hensler, J.G., Ladenheim, E.E. & Lyons, W.E. (2003) Ethanol con-
methamphetamine craving after prolonged voluntary abstinence in sumption and serotonin-1A (5-HT1A) receptor function in heterozy-
a rat model. Biol Psychiatry 78, 463–473. gous BDNF (+/−) mice. J Neurochem 85, 1139–1147.
Carden, W.B., Alexander, G.M., Friedman, D.P., Daunais, J.B., Grant, Hirth, N., Meinhardt, M.W., Noori, H.R., Salgado, H., Torres-Ramirez,
K.A., Mu, J. & Godwin, D.W. (2006) Chronic ethanol drinking O., Uhrig, S., Broccoli, L., Vengeliene, V., Rossmanith, M.,
reduces native T-type calcium current in the thalamus of nonhuman Perreau-Lenz, S., Kohr, G., Sommer, W.H., Spanagel, R. & Hans-
primates. Brain Res 1089, 92–100. son, A.C. (2016) Convergent evidence from alcohol-dependent
Copeland, R.A., Solomon, M.E. & Richon, V.M. (2009) Protein methyl- humans and rats for a hyperdopaminergic state in protracted
transferases as a target class for drug discovery. Nat Rev Drug abstinence. Proc Natl Acad Sci U S A 113(11), 3024–3029.
Discov 8, 724–732. Holmes, A., Spanagel, R. & Krystal, J.H. (2013) Glutamatergic targets
Crawford, J.T., Roberts, D.C. & Beveridge, T.J. (2013) The group II for new alcohol medications. Psychopharmacology 229, 539–554.
metabotropic glutamate receptor agonist, LY379268, decreases Hoover, W.B. & Vertes, R.P. (2007) Anatomical analysis of afferent
methamphetamine self-administration in rats. Drug Alcohol projections to the medial prefrontal cortex in the rat. Brain Struct
Depend 132, 414–419. Funct 212, 149–179.
Darcq, E., Warnault, V., Phamluong, K., Besserer, G.M., Liu, F. & Ron, Hopf, F.W., Chang, S.J., Sparta, D.R., Bowers, M.S. & Bonci, A. (2010)
D. (2015) MicroRNA-30a-5p in the prefrontal cortex controls the Motivation for alcohol becomes resistant to quinine adulteration
transition from moderate to excessive alcohol consumption. Mol after 3 to 4 months of intermittent alcohol self-administration.
Psychiatry 20, 1219–1231. Alcohol Clin Exp Res 34, 1565–1573.
Farris, S.P., Arasappan, D., Hunicke-Smith, S., Harris, R.A. & Mayfield, Iwamoto, K., Bundo, M., Washizuka, S., Kakiuchi, C. & Kato, T. (2004)
R.D. (2015) Transcriptome organization for chronic alcohol abuse in Expression of HSPF1 and LIM in the lymphoblastoid cells derived
human brain. Mol Psychiatry 20, 1438–1447. from patients with bipolar disorder and schizophrenia. J Hum Genet
Feil, R. & Fraga, M.F. (2011) Epigenetics and the environment: emerg- 49, 227–231.
ing patterns and implications. Nat Rev Genet 13, 97–109. Jeanblanc, J., He, D.Y., McGough, N.N., Logrip, M.L., Phamluong, K.,
Gehlert, D.R., Cippitelli, A., Thorsell, A., Le, A.D., Hipskind, P.A., Ham- Janak, P.H. & Ron, D. (2006) The dopamine D3 receptor is part of a
douchi, C., Lu, J., Hembre, E.J., Cramer, J., Song, M., McKinzie, homeostatic pathway regulating ethanol consumption. J Neurosci
D., Morin, M., Ciccocioppo, R. & Heilig, M. (2007) 3-(4-Chloro- 26, 1457–1464.
2-morpholin-4-yl-thiazol-5-yl)-8-(1-ethylpropyl)-2,6-dimethyl-imidazo Jeanblanc, J., He, D.Y., Carnicella, S., Kharazia, V., Janak, P.H. & Ron,
[1,2-b]pyridazine: a novel brain-penetrant, orally available D. (2009) Endogenous BDNF in the dorsolateral striatum gates
corticotropin-releasing factor receptor 1 antagonist with efficacy in alcohol drinking. J Neurosci 29, 13494–13502.
animal models of alcoholism. J Neurosci 27, 2718–2726. Jiang, X., Tian, F., Du, Y., Copeland, N.G., Jenkins, N.A., Tessarollo, L.,
Goldstein, D.B. & Pal, N. (1971) Alcohol dependence produced in mice Wu, X., Pan, H., Hu, X.Z., Xu, K., Kenney, H., Egan, S.E., Turley, H.,
by inhalation of ethanol: grading the withdrawal reaction. Science Harris, A.L., Marini, A.M. & Lipsky, R.H. (2008) BHLHB2 controls
172, 288–290. Bdnf promoter 4 activity and neuronal excitability. J Neurosci 28,
Goldstein, R.Z. & Volkow, N.D. (2002) Drug addiction and its under- 1118–1130.
lying neurobiological basis: neuroimaging evidence for the involve- Johnson, K.A. & Lovinger, D.M. (2015) Metabotropic glutamate recep-
ment of the frontal cortex. Am J Psychiatry 159, 1642–1652. tor 2 positive allosteric modulators: closing the gate on drug abuse?
Goldstein, R.Z. & Volkow, N.D. (2011) Dysfunction of the prefrontal Biol Psychiatry 78, 436–438.
cortex in addiction: neuroimaging findings and clinical implications. Jones, P.A. (2012) Functions of DNA methylation: islands, start sites,
Nat Rev Neurosci 12, 652–669. gene bodies and beyond. Nat Rev Genet 13, 484–492.

Genes, Brain and Behavior (2017) 16: 86–100 97


Heilig et al.

Justinova, Z., Panlilio, L.V., Secci, M.E., Redhi, G.H., Schindler, ethanol exposure and early withdrawal in C57BL/6J mice. Addict
C.W., Cross, A.J., Mrzljak, L., Medd, A., Shaham, Y. & Gold- Biol 17, 351–364.
berg, S.R. (2015) The novel metabotropic glutamate receptor Miura, P., Amirouche, A., Clow, C., Belanger, G. & Jasmin, B.J. (2012)
2 positive allosteric modulator, AZD8529, decreases nicotine Brain-derived neurotrophic factor expression is repressed during
self-administration and relapse in squirrel monkeys. Biol Psychiatry myogenic differentiation by miR-206. J Neurochem 120, 230–238.
78, 452–462. Moorman, D.E. & Aston-Jones, G. (2015) Prefrontal neurons encode
Kim, K.C., Geng, L. & Huang, S. (2003) Inactivation of a histone context-based response execution and inhibition in reward seeking
methyltransferase by mutations in human cancers. Cancer Res 63, and extinction. Proc Natl Acad Sci U S A 112, 9472–9477.
7619–7623. Moorman, D.E., James, M.H., McGlinchey, E.M. & Aston-Jones,
Koob, G.F. & Volkow, N.D. (2010) Neurocircuitry of addiction. Neu- G. (2015) Differential roles of medial prefrontal subregions in the
ropsychopharmacology 35, 217–238. regulation of drug seeking. Brain Res 1628, 130–146.
Koya, E., Golden, S.A., Harvey, B.K., Guez-Barber, D.H., Berkow, Murgatroyd, C., Patchev, A.V., Wu, Y., Micale, V., Bockmuhl, Y.,
A., Simmons, D.E., Bossert, J.M., Nair, S.G., Uejima, J.L., Marin, Fischer, D., Holsboer, F., Wotjak, C.T., Almeida, O.F. & Spengler,
M.T., Mitchell, T.B., Farquhar, D., Ghosh, S.C., Mattson, B.J. & D. (2009) Dynamic DNA methylation programs persistent adverse
Hope, B.T. (2009) Targeted disruption of cocaine-activated nucleus effects of early-life stress. Nat Neurosci 12, 1559–1566.
accumbens neurons prevents context-specific sensitization. Nat Nachmani, D., Lankry, D., Wolf, D.G. & Mandelboim, O. (2010) The
Neurosci 12, 1069–1073. human cytomegalovirus microRNA miR-UL112 acts synergistically
Kroener, S., Mulholland, P.J., New, N.N., Gass, J.T., Becker, H.C. & with a cellular microRNA to escape immune elimination. Nat
Chandler, L.J. (2012) Chronic alcohol exposure alters behavioral Immunol 11, 806–813.
and synaptic plasticity of the rodent prefrontal cortex. PLoS One Nasca, C., Xenos, D., Barone, Y., Caruso, A., Scaccianoce, S.,
7, e37541. Matrisciano, F., Battaglia, G., Mathe, A.A., Pittaluga, A., Lionetto,
Kurita, M., Moreno, J.L., Holloway, T., Kozlenkov, A., Mocci, G., L., Simmaco, M. & Nicoletti, F. (2013) L-acetylcarnitine causes rapid
Garcia-Bea, A., Hanks, J.B., Neve, R., Nestler, E.J., Russo, S.J. & antidepressant effects through the epigenetic induction of mGlu2
Gonzalez-Maeso, J. (2013) Repressive epigenetic changes at the receptors. Proc Natl Acad Sci U S A 110, 4804–4809.
mGlu2 promoter in frontal cortex of 5-HT2A knockout mice. Mol Nickols, H.H. & Conn, P.J. (2014) Development of allosteric modula-
Pharmacol 83, 1166–1175. tors of GPCRs for treatment of CNS disorders. Neurobiol Dis 61,
Lee, S.T., Chu, K., Jung, K.H., Kim, J.H., Huh, J.Y., Yoon, H., Park, D.K., 55–71.
Lim, J.Y., Kim, J.M., Jeon, D., Ryu, H., Lee, S.K., Kim, M. & Roh, Nikiforuk, A., Popik, P., Drescher, K.U., van Gaalen, M., Relo, A.L.,
J.K. (2012) miR-206 regulates brain-derived neurotrophic factor in Mezler, M., Marek, G., Schoemaker, H., Gross, G. & Bespalov,
Alzheimer disease model. Ann Neurol 72, 269–277. A. (2010) Effects of a positive allosteric modulator of group II
Levey, D.F. et al. (2014) Genetic risk prediction and neurobiological metabotropic glutamate receptors, LY487379, on cognitive flexibil-
understanding of alcoholism. Transl Psychiatry 4, e391. ity and impulsive-like responding in rats. J Pharmacol Exp Ther 335,
Lewohl, J.M., Nunez, Y.O., Dodd, P.R., Tiwari, G.R., Harris, R.A. & 665–673.
Mayfield, R.D. (2011) Up-regulation of microRNAs in brain of human O’Dell, L.E., Roberts, A.J., Smith, R.T. & Koob, G.F. (2004) Enhanced
alcoholics. Alcohol Clin Exp Res 35, 1928–1937. alcohol self-administration after intermittent versus continuous
Liechti, M.E., Lhuillier, L., Kaupmann, K. & Markou, A. (2007) alcohol vapor exposure. Alcohol Clin Exp Res 28, 1676–1682.
Metabotropic glutamate 2/3 receptors in the ventral tegmental Pandey, S.C., Zhang, H., Ugale, R., Prakash, A., Xu, T. & Misra, K.
area and the nucleus accumbens shell are involved in behaviors (2008) Effector immediate-early gene arc in the amygdala plays a
relating to nicotine dependence. J Neurosci 27, 9077–9085. critical role in alcoholism. J Neurosci 28, 2589–2600.
Liu, J., Lewohl, J.M., Harris, R.A., Iyer, V.R., Dodd, P.R., Randall, Peters, J. & Kalivas, P.W. (2006) The group II metabotropic glu-
P.K. & Mayfield, R.D. (2006) Patterns of gene expression in the tamate receptor agonist, LY379268, inhibits both cocaine- and
frontal cortex discriminate alcoholic from nonalcoholic individuals. food-seeking behavior in rats. Psychopharmacology 186, 143–149.
Neuropsychopharmacology 31, 1574–1582. Peters, J., Kalivas, P.W. & Quirk, G.J. (2009) Extinction circuits for fear
Liu, J., Lewohl, J.M., Harris, R.A., Dodd, P.R. & Mayfield, R.D. (2007) and addiction overlap in prefrontal cortex. Learn Mem 16, 279–288.
Altered gene expression profiles in the frontal cortex of cirrhotic Pfarr, S., Meinhardt, M.W., Klee, M.L., Hansson, A.C., Vengeliene, V.,
alcoholics. Alcohol Clin Exp Res 31, 1460–1466. Schonig, K., Bartsch, D., Hope, B.T., Spanagel, R. & Sommer, W.H.
Logrip, M.L., Janak, P.H. & Ron, D. (2009) Escalating ethanol intake (2015) Losing control: excessive alcohol seeking after selective
is associated with altered corticostriatal BDNF expression. J Neu- inactivation of cue-responsive neurons in the infralimbic cortex. J
rochem 109, 1459–1468. Neurosci 35, 10750–10761.
Manzardo, A.M., Henkhaus, R.S. & Butler, M.G. (2012) Global DNA Pietrzykowski, A.Z. (2010) The role of microRNAs in drug addiction: a
promoter methylation in frontal cortex of alcoholics and controls. big lesson from tiny molecules. Int Rev Neurobiol 91, 1–24.
Gene 498, 5–12. Pietrzykowski, A.Z., Friesen, R.M., Martin, G.E., Puig, S.I., Nowak,
McGinty, J.F., Whitfield, T.W. Jr. & Berglind, W.J. (2010) Brain-derived C.L., Wynne, P.M., Siegelmann, H.T. & Treistman, S.N. (2008) Post-
neurotrophic factor and cocaine addiction. Brain Res 1314, transcriptional regulation of BK channel splice variant stability by
183–193. miR-9 underlies neuroadaptation to alcohol. Neuron 59, 274–287.
McGough, N.N., He, D.Y., Logrip, M.L., Jeanblanc, J., Phamluong, Reimers, M., Heilig, M. & Sommer, W.H. (2005) Gene discovery
K., Luong, K., Kharazia, V., Janak, P.H. & Ron, D. (2004) RACK1 in neuropharmacological and behavioral studies using Affymetrix
and brain-derived neurotrophic factor: a homeostatic pathway that microarray data. Methods 37, 219–228.
regulates alcohol addiction. J Neurosci 24, 10542–10552. Rimondini, R., Arlinde, C., Sommer, W. & Heilig, M. (2002)
Meinhardt, M.W. & Sommer, W.H. (2015) Postdependent state in rats Long-lasting increase in voluntary ethanol consumption and tran-
as a model for medication development in alcoholism. Addict Biol scriptional regulation in the rat brain after intermittent exposure to
20, 1–21. alcohol. FASEB J 16, 27–35.
Meinhardt, M.W., Hansson, A.C., Perreau-Lenz, S., Bauder-Wenz, Rimondini, R., Sommer, W. & Heilig, M. (2003) A temporal threshold
C., Stahlin, O., Heilig, M., Harper, C., Drescher, K.U., Spanagel, for induction of persistent alcohol preference: behavioral evidence
R. & Sommer, W.H. (2013) Rescue of infralimbic mGluR2 deficit in a rat model of intermittent intoxication. J Stud Alcohol 64,
restores control over drug-seeking behavior in alcohol dependence. 445–449.
J Neurosci 33, 2794–2806. Rimondini, R., Sommer, W.H., Dall’Olio, R. & Heilig, M. (2008)
Melendez, R.I., McGinty, J.F., Kalivas, P.W. & Becker, H.C. (2012) Brain Long-lasting tolerance to alcohol following a history of depen-
region-specific gene expression changes after chronic intermittent dence. Addict Biol 13, 26–30.

98 Genes, Brain and Behavior (2017) 16: 86–100


Reprogramming of mPFC transcriptome

Salih, H., Anghelescu, I., Kezic, I., Sinha, V., Hoeben, E., Van Nueten, Timmusk, T., Palm, K., Metsis, M., Reintam, T., Paalme, V., Saarma,
L., De Smedt, H. & De Boer, P. (2015) Pharmacokinetic and pharma- M. & Persson, H. (1993) Multiple promoters direct tissue-specific
codynamic characterisation of JNJ-40411813, a positive allosteric expression of the rat BDNF gene. Neuron 10, 475–489.
modulator of mGluR2, in two randomised, double-blind phase-I Unoki, M. & Nakamura, Y. (2003) Methylation at CpG islands in intron
studies. J Psychopharmacol 29, 414–425. 1 of EGR2 confers enhancer-like activity. FEBS Lett 554, 67–72.
SAMHSA (2014). National Survey on Drug Use and Health (NSDUH). Uylings, H.B.M., Groenewegen, H.J. & Kolb, B. (2003) Do rats have a
Serious Mental Health Challenges among Older Adolescents and prefrontal cortex? Behav Brain Res 146, 3–17.
Young Adults. Lipari RN, Hedden SL.The CBHSQ Report. Rockville Vendruscolo, L.F., Barbier, E., Schlosburg, J.E., Misra, K.K., Whitfield,
(MD): Substance Abuse and Mental Health Services Administra- T.W. Jr., Logrip, M.L., Rivier, C., Repunte-Canonigo, V., Zorrilla, E.P.,
tion (US); 2013–2014 May 6. Sanna, P.P., Heilig, M. & Koob, G.F. (2012) Corticosteroid-dependent
Sanchis-Segura, C. & Spanagel, R. (2006) Behavioural assessment of plasticity mediates compulsive alcohol drinking in rats. J Neurosci
drug reinforcement and addictive features in rodents: an overview. 32, 7563–7571.
Addict Biol 11, 2–38. Vengeliene, V., Bilbao, A. & Spanagel, R. (2014) The alcohol depri-
Seamans, J.K., Lapish, C.C. & Durstewitz, D. (2008) Comparing the vation effect model for studying relapse behavior: a comparison
prefrontal cortex of rats and primates: insights from electrophysi- between rats and mice. Alcohol 48, 313–320.
ology. Neurotox Res 14, 249–262. Vertes, R.P. (2004) Differential projections of the infralimbic and
Shaham, Y., Shalev, U., Lu, L., De Wit, H. & Stewart, J. (2003) The prelimbic cortex in the rat. Synapse 51, 32–58.
reinstatement model of drug relapse: history, methodology and Wang, F., Xu, H., Zhao, H., Gelernter, J. & Zhang, H. (2016) DNA
major findings. Psychopharmacology 168, 3–20. co-methylation modules in postmortem prefrontal cortex tissues
Sheedy, D., Garrick, T., Dedova, I., Hunt, C., Miller, R., Sundqvist, N. of European Australians with alcohol use disorders. Sci Rep 6,
& Harper, C. (2008) An Australian Brain Bank: a critical investment 19430.
with a high return!. Cell Tissue Bank 9, 205–216. Warnault, V., Darcq, E., Levine, A., Barak, S. & Ron, D. (2013) Chro-
Simms, J.A., Steensland, P., Medina, B., Abernathy, K.E., Chandler, matin remodeling – a novel strategy to control excessive alcohol
L.J., Wise, R. & Bartlett, S.E. (2008) Intermittent access to 20% drinking. Transl Psychiatry 3, e231.
ethanol induces high ethanol consumption in Long-Evans and Weaver, I.C., Szyf, M. & Meaney, M.J. (2002) From maternal care to
Wistar rats. Alcohol Clin Exp Res 32, 1816–1823. gene expression: DNA methylation and the maternal programming
Smith, M.L., Lopez, M.F., Archer, K.J., Wolen, A.R., Becker, H.C. & of stress responses. Endocr Res 28, 699.
Miles, M.F. (2016) Time-course analysis of brain regional expres- Welsh, J.P., Han, V.Z., Rossi, D.J., Mohr, C., Odagiri, M., Daunais,
sion network responses to chronic intermittent ethanol and with- J.B. & Grant, K.A. (2011) Bidirectional plasticity in the primate
drawal: implications for mechanisms underlying excessive ethanol inferior olive induced by chronic ethanol intoxication and sustained
consumption. PLoS One 11, e0146257. abstinence. Proc Natl Acad Sci U S A 108, 10314–10319.
Sommer, W., Arlinde, C. & Heilig, M. (2005) The search for candidate Wicks, S., Hammar, J., Heilig, M. & Wisen, O. (2001) Factors affecting
genes of alcoholism: evidence from expression profiling studies. the short-term prognosis of alcohol dependent patients undergoing
Addict Biol 10, 71–79. inpatient detoxification. Subst Abus 22, 235–245.
Sommer, W., Hyytia, P. & Kiianmaa, K. (2006) The alcohol-preferring Willcocks, A.L. & McNally, G.P. (2013) The role of medial prefrontal
AA and alcohol-avoiding ANA rats: neurobiology of the regulation cortex in extinction and reinstatement of alcohol-seeking in rats.
of alcohol drinking. Addict Biol 11, 289–309. Eur J Neurosci 37, 259–268.
Sommer, W.H., Rimondini, R., Hansson, A.C., Hipskind, P.A., Gehlert, Wise, R.A. (1973) Voluntary ethanol intake in rats following expo-
D.R., Barr, C.S. & Heilig, M.A. (2008) Upregulation of voluntary sure to ethanol on various schedules. Psychopharmacologia 29,
alcohol intake, behavioral sensitivity to stress, and amygdala crhr1 203–210.
expression following a history of dependence. Biol Psychiatry 63, Wise, R.A. (1975) Maximization of ethanol intake in the rat. Adv Exp
139–145. Med Biol 59, 279–294.
Spanagel, R. & Holter, S.M. (1999) Long-term alcohol self- Wolffgramm, J. & Heyne, A. (1995) From controlled drug intake to loss
administration with repeated alcohol deprivation phases: an of control: the irreversible development of drug addiction in the rat.
animal model of alcoholism? Alcohol Alcohol 34, 231–243. Behav Brain Res 70, 77–94.
Subramanian, A., Tamayo, P., Mootha, V.K., Mukherjee, S., Ebert, Wolffgramm, J., Galli, G., Thimm, F. & Heyne, A. (2000) Animal
B.L., Gillette, M.A., Paulovich, A., Pomeroy, S.L., Golub, T.R., Lan- models of addiction: models for therapeutic strategies? J Neural
der, E.S. & Mesirov, J.P. (2005) Gene set enrichment analysis: a Transm 107, 649–668.
knowledge-based approach for interpreting genome-wide expres- Wong, C.C., Mill, J. & Fernandes, C. (2011) Drugs and addiction: an
sion profiles. Proc Natl Acad Sci U S A 102, 15545–15550. introduction to epigenetics. Addiction 106, 480–489.
Sugino, K., Hempel, C.M., Miller, M.N., Hattox, A.M., Shapiro, P., Wu, Wood, C.M., Nicolas, C.S., Choi, S.L., Roman, E., Nylander, I.,
C., Huang, Z.J. & Nelson, S.B. (2006) Molecular taxonomy of major Fernandez-Teruel, A., Kiianmaa, K., Bienkowski, P., de Jong, T.R.,
neuronal classes in the adult mouse forebrain. Nat Neurosci 9, Colombo, G., Chastagnier, D., Wafford, K.A., Collingridge, G.L.,
99–107. Wildt, S.J., Conway-Campbell, B.L., Robinson, E.S. & Lodge, D.
Swanberg, S.E., Nagarajan, R.P., Peddada, S., Yasui, D.H. & LaSalle, (2016) Prevalence and influence of cys407* Grm2 mutation in
J.M. (2009) Reciprocal co-regulation of EGR2 and MECP2 is dis- Hannover-derived Wistar rats: mGlu2 receptor loss links to alcohol
rupted in Rett syndrome and autism. Hum Mol Genet 18, 525–534. intake, risk taking and emotional behaviour. Neuropharmacology
Tapocik, J.D., Solomon, M., Flanigan, M., Meinhardt, M., Barbier, E., 14, pii: S0028-3908(16)30092-2.
Schank, J.R., Schwandt, M., Sommer, W.H. & Heilig, M. (2013) Zhao, Y., Dayas, C.V., Aujla, H., Baptista, M.A., Martin-Fardon, R.
Coordinated dysregulation of mRNAs and microRNAs in the rat & Weiss, F. (2006) Activation of group II metabotropic glutamate
medial prefrontal cortex following a history of alcohol dependence. receptors attenuates both stress and cue-induced ethanol-seeking
Pharmacogenomics J 13, 286–296. and modulates c-fos expression in the hippocampus and amygdala.
Tapocik, J.D., Barbier, E., Flanigan, M., Solomon, M., Pincus, A., J Neurosci 26, 9967–9974.
Pilling, A., Sun, H., Schank, J.R., King, C. & Heilig, M. (2014) Zhao, Z., Guo, A.Y., van den Oord, E.J., Aliev, F., Jia, P., Edenberg,
microRNA-206 in rat medial prefrontal cortex regulates BDNF H.J., Riley, B.P., Dick, D.M., Bettinger, J.C., Davies, A.G., Grotewiel,
expression and alcohol drinking. J Neurosci 34, 4581–4588. M.S., Schuckit, M.A., Agrawal, A., Kramer, J., Nurnberger, J.I.
Tian, W., Zhao, M., Li, M., Song, T., Zhang, M., Quan, L., Li, S. & Jr., Kendler, K.S., Webb, B.T. & Miles, M.F. (2012) Multi-species
Sun, Z.S. (2012) Reversal of cocaine-conditioned place preference data integration and gene ranking enrich significant results in an
through methyl supplementation in mice: altering global DNA alcoholism genome-wide association study. BMC Genomics 13
methylation in the prefrontal cortex. PLoS One 7, e33435. (Suppl. 8), S16.

Genes, Brain and Behavior (2017) 16: 86–100 99


Heilig et al.

Zhou, Z., Karlsson, C., Liang, T., Xiong, W., Kimura, M., Tapocik, J.D., laboratory receives funding by Deutsche Forschungsgemein-
Yuan, Q., Barbier, E., Feng, A., Flanigan, M., Augier, E., Enoch, schaft center grants (SFB636, SFB1134, subprojects D7 and
M.A., Hodgkinson, C.A., Shen, P.H., Lovinger, D.M., Edenberg, B04, respectively). Work in the Wahlestedt laboratory was
H.J., Heilig, M. & Goldman, D. (2013) Loss of metabotropic glu- supported by US National Institute of Health NIAAA R01 no.
tamate receptor 2 escalates alcohol consumption. Proc Natl Acad 1R01AA023781-01A1. Related epigenomics and RNA work in the
Sci U S A 110, 16963–16968. Wahlestedt laboratory has received funding by US National Insti-
tute of Health awards DA035592, MH084880 and NS071674.

Acknowledgments
Work in the Heilig laboratory reviewed here is supported by
the Swedish Science Council (Dnr 2013-7434). The Sommer

100 Genes, Brain and Behavior (2017) 16: 86–100


Copyright of Genes, Brain & Behavior is the property of Wiley-Blackwell and its content
may not be copied or emailed to multiple sites or posted to a listserv without the copyright
holder's express written permission. However, users may print, download, or email articles for
individual use.

You might also like