2014 - Kruman & Fowler - Impaires One Carbon Metabolism and DNa Methylation in Alcohol Toxicity

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

JOURNAL OF NEUROCHEMISTRY | 2014 | 129 | 770–780 doi: 10.1111/jnc.

12677

Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center,
Lubbock, Texas, USA

Abstract methyl groups for methylation, particularly DNA methylation


Excessive alcohol consumption is a prominent problem and one critical for epigenetic regulation of gene expression, and its
of the major causes of mortality and morbidity around the world. disturbance may compromise DNA methylation, thereby affect-
Long-term, heavy alcohol consumption is associated with a ing gene expression. OCM disturbance mediated by nutrient
number of deleterious health consequences, such as cancer, deficits is a well-known risk factor for various disorders and
heart and liver disease, a variety of neurological, cognitive, and developmental defects (e.g., neural tube defects). In this review,
behavioral deficits. Alcohol consumption is also associated with we summarize the role of OCM disturbance and associated
developmental defects. The causes of alcohol-induced toxicity epigenetic aberrations in chronic alcohol-induced toxicity.
are presently unclear. One of the mechanisms underlying alcohol Keywords: alcohol toxicity, DNA methylation, one-carbon
toxicity has to do with its interaction with folic acid/homocysteine metabolism.
or one-carbon metabolism (OCM). OCM is a major donor of J. Neurochem. (2014) 129, 770–780.

Millions of people worldwide have alcohol-use disorders modifications can significantly impact gene expression and
(Harper 2009). Numerous mechanisms may account for all thereby cellular function (Szyf 2011). Ethanol-related OCM
the effects of alcohol on both adult and developing dysfunction may therefore induce epigenetic modifications.
organisms. Yet, many gaps remain in our knowledge of This can explain why long-term excessive ethanol consump-
their molecular basis. One of mechanisms underlying alcohol tion adversely impacts nearly every organ of the body. Since
toxicity has to do with one-carbon metabolism (OCM) epigenetic alterations leading to modified physiological
impairment. OCM is a complex metabolic network of processes may be reversible, they can serve as targets for
pathways involved in the transfer of one-carbon groups and potential intervention (Choi and Friso 2010).
the main source of these groups for methylation reactions in Although alcohol is a known antagonist to OCM
the cell (Stover 2009). Hyperhomocysteinemia or elevated (Giovannucci 2004; Kharbanda 2009), the etiology of
homocysteine (Hcy) - an indicator of OCM dysfunction - is OCM disturbance in alcoholism is not entirely understood.
often observed in chronic alcoholism (Seitz and Stickel
2007).
Received August 19, 2013; revised manuscript received January 30,
The physiological importance of OCM is based on its two 2014; accepted February 3, 2014.
main functions: (i) de novo nucleotide biosynthesis, essential Address correspondence and reprint requests to Inna I. Kruman,
for DNA replication and repair and (ii) provision of methyl Department of Pharmacology and Neuroscience, Texas Tech University
groups for methylation, including that of DNA methylation. Health Sciences Center, Lubbock, TX 79430, USA.
The latter is an important epigenetic determinant in gene E-mail: inna.kruman@ttuhsc.edu
Abbreviations used: Dnmt, DNA methyltransferases; dNTP, deoxyri-
expression (Mason 2003; Villanueva et al. 2007). Other bonucleotide triphosphates; FASD, fetal alcohol spectrum disorders;
epigenetic phenomena include histone modifications, which HAT, histone acetyltransferase; Hcy, homocysteine; HDAC, histone
are in turn associated with chromatin remodeling. Chromatin deacetylase; HMTase, histone methyltransferase; MBD, methylated
is a fairly dynamic system for packaging and condensing DNA-binding domain proteins; MS, methionine synthase; MTHFR,
genomic DNA, critical for controlling the accessibility of methylene tetrahydrofolate reductase; MTRR, methionine synthase
reductase; NR2B, N-methyl D-aspartate receptor subtype 2B; OCM,
DNA for transcription. Since epigenetic mechanisms are one-carbon metabolism; ROS, reactive oxygen species; SAH, S-adenosyl
critical for gene expression and stably alter DNA function homocysteine; SAM, S-adenosyl methionine; THF, 5-methyl tetrahy-
without changing the sequence, structural chromatin drofolate; TSA, trichostatin A.

770 © 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780
Alcohol toxicity and DNA methylation 771

It may result from the deficiency of folate, an essential Acetaldehyde is further metabolized to acetate by aldehyde
vitamin involved in OCM. Well documented in alcoholics, dehydrogenase 2. Acetaldehyde and ROS damage cellular
folate deficiency may be caused by poor diet, malabsorption, macromolecules such as lipids, proteins, and DNA and
increased urinary excretion, or a combination of these factors contribute to alcohol-produced cell and tissue destruction
(Halsted and Medici 2011). OCM dysfunction, however, (Deng and Deitrich 2008) when acting in concert as factors
may result from a direct interaction of ethanol or its involved in alcohol toxicity.
metabolites with OCM enzymes, such as methionine
synthase (MS), and this is not associated with reduced
One-carbon metabolism and risk factors for its
availability of folate (Barak et al. 1993; Villanueva and
disturbance
Halsted 2004). Given that MS is critical for OCM function,
its inactivation by ethanol may play a significant role in OCM (Fig. 1) is a complex biochemical pathway responsible
alcohol-induced OCM impairment. for generating DNA nucleotides, precursors for DNA
Toxins that may directly impact OCM are generated from biosynthesis and repair, as well as production and utilization
ethanol metabolism by alcohol dehydrogenase (ADH), of S-adenosyl methionine (SAM), the universal methyl donor
cytochrome P450 2E1, or catalase. These enzymes convert in cellular methylation reactions (Stover 2004). B vitamins
ethanol to acetaldehyde (Wu et al. 2010; Cederbaum 2012). including folic acid and its derivatives, folates together with
Cytochrome P450 2E1 (Cyp2E1) is one of P450 isozymes OCM enzymes such as MS, methylene tetrahydrofolate
inducible by ethanol. Cyp2E1 is a major component of the reductase (MTHFR) and cystathinine beta-synthase (Singh
microsomal ethanol oxidizing system in the liver (Carpenter and Jaiswal 2013) represent essential OCM components.
et al. 1996; Lieber 2004), it is also inducible by ethanol in MTHFR converts 5-methyltetrahydrofolate (5-methylTHF)
other tissues including brain (Zerilli et al. 1995; Warner and to 5-methylTHF. Once absorbed into the bloodstream, folate
Gustafsson 1994; Zimatkin et al. 2006; Deitrich 2011). As molecules are taken up by the liver and retained or released
demonstrated in vitro studies, in the brain, where ADH into blood or bile as 5-methylTHF, the main circulating form
activity is low, catalase and Cyp2E1 generate 60 to 70% and of folate, which is then transported into various organs by
5 to 20% of acetaldehyde, respectively (Zimatkin et al. active transporters. MS converts Hcy to methionine utilizing
2006). Acetaldehyde production is the first step of ethanol 5-methylTHF as a donor of one-carbon moiety to methylate
metabolism, which is accompanied by formation of reactive Hcy. This yields methionine and tetrahydrofolate (THF).
oxygen species (ROS) which has been shown to occur in Methionine is converted to the direct methyl donor, SAM
brain as well (Montoliu et al. 1994; Haorah et al. 2008). (Stover 2009). The methyl groups methylate biomolecules
Given that 8-hydroxy-20 -deoxyguanosine is a major product (histone proteins and DNA), whereas catalyzed by DNA
of oxidative DNA damage, its formation in mouse brains methyltransferases (Dnmts) (Fuso 2013). SAM donates
following alcohol exposure also confirms that alcohol gen- methyl groups and converts to S-adenosyl-homocysteine
erates ROS in the brain (Hirano 2011; Fowler et al. 2012). (SAH) which is hydrolyzed to Hcy. Then Hcy is catabolyzed

Fig. 1 Ethanol interferes with one-carbon metab-


olism. CBS, cystathionine b-synthase; DHF,
dihydrofolate; dNTP, deoxyribonucleotide triphos-
phate; DMG, dimethylglycine; dTMP, deoxyt-
hymidine monophosphate; dUMP, deoxyuridine
monophosphate; Hcy, homocysteine; MS, methio-
nine synthase; MTHFR, methylene THF reductase;
5-methylTHF, SAH, S-adenosyl homocysteine;
SHMT, serine hydroxymethyltransferase.

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


772 I. I. Kruman and A.-K. Fowler

via the trans-sulfuration pathway initiated by cystathionine b Methylating function of OCM through SAM formation
synthase. Hcy accumulation increases SAH because of the is indispensable for various CNS reactions including syn-
reversibility of the reaction converting SAH to Hcy; the thesis of neurotransmitters and phospholipids and methyla-
equilibrium favors SAH synthesis, whereas the hydrolytic tion of DNA and proteins (Stanger et al. 2009). The
direction is favored only if Hcy is efficiently lowered (Stover importance of OCM for the CNS was initially demonstrated
2009). SAH is a strong DNA methyltransferase inhibitor, in studies that revealed an increased risk of neural tube
reinforcing DNA hypomethylation (Chiang et al. 1996; Lu defects under conditions of folate deficiency during preg-
2000). Alteration in OCM can lead to a decrease in the SAM/ nancy (Laurence et al. 1981; Blom et al. 2006). Epidemio-
SAH ratio, eventually causing aberrant methylation resulting logical studies have demonstrated that OCM disturbance is
in modified gene expression (Fuso and Scarpa 2011). also involved in pathogenesis of neurodegenerative and
Biosynthesis of the deoxynucleotides in OCM involves neuropsychiatric diseases, including stroke, Parkinson’s and
serine and is catalyzed by serine hydroxymethyltransferase 1 Alzheimer’s diseases, dementia, and depression (Reynolds
(SHMT) (Fig. 1). For this synthesis, one-carbon moiety is 2002; He et al. 2004; Irizarry et al. 2005; Lamberti et al.
obtained from THF, yielding 5,10-methyleneTHF, a 2005; Choi and Friso 2010). Animal and in vitro studies
common substrate to methylation and synthesis of DNA supported direct impact of folate deficiency and hyperhom-
precursors - deoxyribonucleotide triphosphates (dNTPs). ocysteinemia in neurotoxicity (Kruman et al. 2002; Fuso
Insufficiency of folate and other vitamins such as B6, and et al. 2012).
B12 compromises the steady-state level of dNTPs which
may lead to misincorporation of uracil into the DNA
DNA methylation
backbone in place of thymine (Singh and Jaiswal 2013).
Persistent uracil misincorporation results in futile cycles of DNA methylation is the most extensively studied epigenetic
DNA repair, chromosome breakage and genomic instability mechanism, important for regulation of transcription, histone
(Stover 2009). This scenario is also possible in conditions of modification, and genomic stability. DNA methylation
‘methyl trap’. Once 5,10-methyleneTHF is converted by involves a covalent modification of DNA by addition of a
MTHFR to 5-methylTHF, the reverse reaction cannot occur, methyl group to cytosine base at the CpG dinucleotide
and the only way for the recycling of 5-methylTHF to THF - residues which can cause a stable alteration of DNA function
and thus participation in dNTP biosynthesis - is through MS. without changing the sequence (reviewed in Choi and Friso
However, MS inactivation which can be caused, for example, 2010; Szyf 2011). DNA methylation is a heritable epigenetic
by the oxidation of its coenzyme, vitamin B12 (Lim and mechanism by which a methylated cytosine in CpG
Song 2012) or by ethanol-produced damage to MS (Barak accurately replicates its methylation pattern (Gruenbaum
et al. 1993; Villanueva and Halsted 2004), prevents the et al. 1981; Bohacek et al., 2013) and participates in
conversion of 5-methylTHF to THF, resulting in trapping of establishing differentiated cell types during embryogenesis
the cellular folate as 5-methylTHF. This leads to a cellular (Razin et al. 1984; Szyf 2011; Lister et al. 2009). In
pseudo-folate deficiency, where despite adequate amounts of vertebrates, the CpG dinucleotide sequence is a principal
folate, OCM is impaired in a manner that is identical to that target of DNA methylation, as it is preferentially recognized
seen in true folate deficiency (Stover 2009). The loss of MS by Dnmts (Gruenbaum et al. 1982). Five different Dnmts
activity will also compromise conversion of Hcy to methi- catalyze DNA methylation at the CpG sequence, responsible
onine, since MS catalyzes this conversion. Hcy, however, for maintenance (Dnmt1) and de novo (Dnmt 3a and 3b)
can be remethylated via an alternative mechanism using methylation. The function of Dnmt2 is not yet clear.
betaine, supplied from dietary choline, in the kidney and Knockout studies in mice have shown that de novo DNA
liver. Thus, OCM regulates two vital processes - DNA methylation is essential for development, since Dnmt3a-
synthesis and DNA methylation. Consequently, disturbance deficient animals die several weeks after birth and Dnmt3b-
of OCM enzyme activity or deficiency in its vitamins affects deficient animals die in utero (Okano et al. 1999). Changes
both methylation and DNA synthesis, which may lead to in Dnmt activity or expression can affect global DNA
disease (Singh and Jaiswal 2013). Particularly, elevated methylation. Dnmt1 expression is known to decrease with
blood- and CSF levels of Hcy, and reduced SAM in CSF and age, which may alter DNA methylation status and DNA
brain tissue are associated with neurological diseases and methylation-mediated gene expression (Li et al. 2010). This
mood disorders (D’Anci and Rosenberg 2004). OCM which can be associated with pathogenesis of age-associated
is essential for CNS function is different from OCM in the diseases. DNA methylation, however, not always occurs in
liver or kidney, where remethylation can be performed the context of CpG. Recent data suggest that DNA methyl-
through betaine-homocysteine-methyltransferase which is ation occurs in other dinucleotide sequences in regions
not detected in the brain (Sunden et al. 1997). In addition, beyond the traditional promoters (Lister et al. 2009; Fuso
in contrast to the liver and kidney, the brain exhibits a reduced et al. 2010; McGowan et al. 2011). This, in turn, suggests
activity of the trans-sulfuration pathway (Miller 2003). that differentially methylated regions are not limited to

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


Alcohol toxicity and DNA methylation 773

promoters, as well as that methylation of these regions may mammalian DNA support this suggestion (Jin et al. 2010).
play a role in controlling genome function beyond promoter The formation of differential methylation patterns is very
regulation of gene expression. The presence of non-CpG important to preserve the stability of the differentiated cell
methylation in the genome suggests that DNA methylation is phenotype. Inappropriate modulations of DNA methylation
not always mitotically heritable, and can thus be maintained and associated alterations in gene expression can result in
by a balance of methylating and demethylating enzymes disease (Razin and Riggs 1980; Singh and Jaiswal 2013).
(McGowan et al. 2011). Indeed, active demethylation has There are two types of identity: the individual identity
been confirmed in this context (Lucarelli et al. 2001; encoded by DNA sequence and a cell-type identity encoded
Bruniquel and Schwartz 2003). The dynamic methylation- in the distribution of methyl groups in the same molecule of
demethylation equilibrium has been observed in postmitotic DNA. Cell type-specific, differentially methylated regions of
cells, particularly in postmitotic neurons (Weaver et al. DNA produce multiple phenotypes in a multicellular organ-
2004; Feng and Fan 2009), where conditional knockout of ism, explaining how the same genome can encode multiple
Dnmt1 resulted in DNA demethylation (Feng and Fan 2009). phenotypes. DNA methylation is cell type- and tissue-
The mechanism involved in regulation of gene expression specific. There are known cell type-specific patterns of
not associated with promoter DNA methylation includes a methylation or tissue-specific differentially methylated
recruitment of methylated DNA-binding domain proteins regions (Szyf 2011). For example, DNA methylation in
(MBD) to methylated cytosines in promoters (Nan et al. differentiated neurons is very dynamic, and Dnmt expression
1997). MBDs recruit complexes containing histone deacet- in these cells is high (Feng et al. 2010). However, increase in
ylases and histone methyltransferases to promoters which Hcy levels and thus high levels of SAH inhibit Dnmts (James
leads to shaping inactive chromatin configuration around the et al. 2002) which is especially important for brain tissue due
genes (Dobosy and Selker 2001). Several lines of evidence to the absence of betaine-homocysteine remethylation activ-
indicate that chromatin modification and DNA methylation ity (Sunden et al. 1997) and significantly reduced capacity of
are correlated (Cheng and Blumenthal 2010). Methylated the trans-sulfuration pathway (Miller 2003). Thus, alterations
DNA is enriched in regions of the genome that are packaged in DNA methylation can have tissue-specific manifestations
in inactive chromatin (Razin and Cedar 1977), suggesting a (Maegawa et al. 2010).
bilateral relationship between chromatin modification and
DNA methylation. Relationship between histone acetylation
The role of epigenetics in normal physiology and
and DNA methylation is associated with a well-known
disease
connection between chromatin compaction and DNA meth-
ylation. The commonly used histone deacetylase inhibitors, Since OCM is the main source of methyl groups, its
trichostatin A, and valproic acid induce loss of DNA disturbance by ethanol and nutrients may alter DNA and
methylation, chromatin decondensation, and transcriptional histone methylation, thereby affecting gene expression at the
activation (Cervoni and Szyf 2001; Ou et al. 2007; Dong transcriptional level. Such epigenetic changes can be inher-
et al. 2008). This may result from the facilitated access of ited during cell division (Slatkin 2009), resulting in perma-
gene regulatory regions to demethylases (Ou et al. 2007). nent maintenance of the acquired phenotype. Nutrient deficits
Since chromatin modification is dynamic, it makes altera- involved in OCM can alter OCM function by affecting a
tions in DNA methylation also dynamic, with critical methyl group provider, SAM, and an inhibitor of Dnmts,
implications in the brain (Gr€aff et al. 2012; Zentner and SAH (Choi and Friso 2010). This may lead to disturbed
Henikoff 2013). DNA and histone methylation, possibly causing various
Although the mechanisms of DNA demethylation are not disorders. Indeed, ethanol-induced liver cirrhosis is associ-
fully understood, it has been suggested that DNA demethy- ated with global hypomethylation of liver DNA (Purohit
lation may involve (i) DNA base excision repair; (ii) DNA et al. 2007). Global changes in DNA methylation are
nucleotide excision repair; (iii) oxidative or (iv) hydrolytic common in chronic diseases such as lupus (Cornacchia
removal of the methyl group (Zhu 2009). Most recently, et al. 1988; Yung and Richardson 1994) and diabetes (Choi
5-methylcytosine in mammalian DNA was found to be and Friso 2010). Aberrations in DNA methylation are also
converted to 5-hydroxymethylcytosine by methylcytosine associated with cardiovascular disease (Gluckman et al.
oxygenase TET1 (Bottiglieri and Hyland 1994; Tahiliani 2009), oncogene activation, chromosome instability, and
et al. 2009). 5-hydroxymethylcytosine might also be pro- carcinogenesis (Ji et al. 1997). An increasing body of
duced by Dnmts via addition of formaldehyde to cytosines in evidence indicates that many psychiatric and neurodegener-
DNA (Liutkeviciute et al. 2009). Unmodified cytosine is ative diseases such as Alzheimer’s, Parkinson’s, schizophre-
believed to be generated as a result of the reversible Dnmt- nia, and autism are also linked to epigenetics or at least have
catalyzed reaction from 5-hydroxymethylcytosine which may an epigenetic component (Qureshi and Mehler 2012; Fuso
therefore be an intermediate in direct DNA demethylation. 2013). In fact, only a small percentage of neurodegenerative
Significant levels of 5-hydroxymethylcytosine observed in disorders are caused by evident genetic mutations – for

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


774 I. I. Kruman and A.-K. Fowler

instance, Huntington’s disease and the familial forms of syndrome which is also characterized by hypomethylation,
Parkinson’s and Alzheimer’s diseases (Lill and Bertram results from mutations in genes encoding proteins that
2011). The pathogenesis of sporadic onset versus Mendelian regulate DNA methylation homeostasis (Berdasco and
inheritance of most cases of neurodegenerative diseases Esteller 2013). As epigenetic information can be inherited,
remains elusive. Nevertheless, it has been suggested that the and changes in DNA methylation pattern could be main-
sporadic forms may be associated with environmental factors tained through DNA replication cycles, transient exposure to
mediating epigenetic changes, particularly evidenced in an environmental agent such as diet, nutrients, exposure to
Alzheimer’s disease (Migliore and Coppede 2009; Fuso chemicals, and alcohol could have a lasting impact on DNA
and Scarpa 2011; Mastroeni et al. 2011). This suggestion is methylation and thereby, phenotype. The honeybee model
supported by observation that the treatment of a transgenic clearly demonstrates the epigenetic effects of diet on the
mouse model of Alzheimer’s disease with a diet deficient in phenotype. Honeybees grow into either long-lived and very
vitamin B12, B6, and folate, fundamental for OCM homeo- reproductive queens or short-lived functionally sterile work-
stasis and therefore methylation potential, resulted in aberrant ers, depending on whether they are fed royal jelly or
methylation, increased amyloid processing, and cognitive beebread. The difference in phenotype has been suggested to
impairment. At the same time, supplementation with SAM be caused by epigenetic modifications in DNA methylation
attenuated these effects (Fuso and Scarpa 2011; Fuso et al. patterns induced by the different types of diet (Kucharski
2012). Altered levels of SAM were observed in neurological et al. 2008).
disorders, including Alzheimer’s disease (Bottiglieri and Social factors (e.g., maternal care) may also be responsible
Hyland 1994; Tchantchou et al. 2006). Together, these for these epigenetic changes. Early childhood is known to be
findings demonstrate the involvement of epigenetic altera- extremely critical in shaping these behavioral patterns in
tions in sporadic diseases and a potential role for OCM mammals (Champagne 2010). Maternal behavior defines
impairment in such alterations. This, in turn, suggests that life-long changes in the offspring’s responsiveness to stress
nutritional deficiency, excessive alcohol consumption, and (Liu et al. 1997; Francis et al. 1999). For illustration, it is
other environmental factors are able to modulate methylation known that exposure of infant rats to stressed caretakers with
metabolism. This also suggests that environmental factors abusive behavior produced persisting changes in methylation
can have both therapeutic and toxic implications. pattern of the adult prefrontal cortex (Roth et al. 2009);
Since embryonic development is characterized by rapid early-life stress in mice caused sustained DNA hypomethy-
cell replication and formation of DNA methylation patterns, lation of an important regulatory region of the arginine
the impairment of OCM or other elements of DNA vasopressin gene (Murgatroyd et al. 2009). It has been well
methylation machinery such as Dnmts may cause alterations documented that maternal care differences modifies epige-
in DNA methylation (Dolinoy et al. 2007). Deficiency in netic status of the glucocorticoid receptor gene in the
methylated DNA-binding domain proteins Mbd1 in mutant hippocampus and has a global effect on the hippocampal
(Mbd1/) mice was characterized by several deficits transcriptome. This impacts the stress response, learning, and
associated with autism such as social interaction difficulties, memory of the offspring. As early-life experience has long-
learning deficits, anxiety, and depression (Allan et al. 2008). term consequences on behavior, it is potentially reversible in
Folic acid deficiency during gestation resulted in multiple adulthood (Weaver et al. 2004, 2006) and may also be
teratogenic effects, which might be explained by reduced reversed by certain interventions such as changes in meth-
methyl availability due to OCM dysfunction and consequent ylation pattern (Cervoni and Szyf 2001; Cervoni et al. 2002;
epigenetic aberrations (Pogribny et al. 1997; Brunaud et al. Detich et al. 2003). Thus, both adverse chemical and social
2003). If these aberrations affect the expression of genes exposures at this early period of life could have a long-lasting
involved in immune programming, the offspring develops impact on behavior and memory through changes in DNA
allergic disorders (Bach 2002). Agents interfering with OCM methylation (Lahiri et al. 2009). These findings support the
and therefore DNA methylation during certain time when idea that although epigenetic programs established early in
these patterns are generated could disrupt the DNA methyl- life are sustained throughout life, they are maintained by a
ation pattern, cellular differentiation, and organogenesis, dynamic balance between DNA methylation and demethy-
given the importance of epigenetic programing for cell lation and can be potentially changed.
differentiation (Huang and Fan 2010). Aberrations in DNA
methylation patterns can target different regions of the
Alcohol-induced aberrant DNA methylation in the
genome and cause teratogenesis or manifest later in life,
adult organism
transforming the adult health-related phenotype (Sinclair
et al. 2007). For example, immunodeficiency and facial Alcohol metabolites have been shown to directly affect the
anomalies syndrome (ICF) - a rare chromosome breakage epigenome (Knezovich and Ramsay 2012). There is increas-
disease - are characterized by global hypomethylation ing evidence that alcohol-induced epigenetic changes may
(Hansen et al. 1999; Okano et al. 1999; Ehrlich 2003). Rett result from OCM dysfunction lowering the availability of

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


Alcohol toxicity and DNA methylation 775

SAM which compromises DNA and histone methylation suggesting a causal role for OCM impairment in alcohol
(Hamid et al. 2009). Alcoholic patients often have reduced neurotoxicity. Alcohol has been found to modify DNA
blood folate and elevated plasma Hcy (Cravo et al. 1996), methylation (and expression) of specific genes. Bonsch and
both indicative of OCM impairment. However, toxic ethanol associates reported significant increase of DNA methylation
metabolites such as acetaldehyde, may directly interact with at a synuclein (Bonsch et al. 2005), which is also observed
different components of OCM (e.g. MS) thereby disturbing in Alzheimer’s disease (Alim et al. 2004). Alcohol epige-
methylation reactions (Barak et al. 1993; Villanueva and netically increased expression of the glutamate N-methyl D-
Halsted 2004). Indeed, ethanol exposure leads to reduced aspartate receptor subtype 2B by demethylation of corre-
levels of the major methyl donor, SAM, increased levels of sponding gene (Qiang et al. 2010). Increased N-methyl D-
SAH, lower SAM⁄SAH ratio and as a consequence, global aspartate receptor subtype 2B expression has been implicated
hypomethylation (Stickel et al. 2000; Murillo-Fuentes et al. in ethanol-associated phenotypes (Krystal et al. 2003). Such
2005). A significant decrease of mRNA expression of epigenetic alterations induced by alcohol may play a role in
Dnmt3a Dnmt3b in alcoholics (Bonsch et al. 2006) has also the pathophysiology of psychiatric disorders including
been demonstrated. Ethanol has been found to cause site- depression (Frieling et al. 2007; Hillemacher 2011) and
specific methylation and acetylation of histones. For exam- schizophrenia (Bleich et al. 2006).
ple, by activating histone acetyltransferase, ethanol exposure
induced increased histone acetylation (H3K9) in a promoter
Developmental aspect of alcohol-induced aberrant
region of the gene encoding the alcohol-metabolizing
methylation
enzyme ADH 1 in rat hepatocytes (Park et al. 2005). In
addition, exposure of hepatocytes to ethanol resulted in Alcohol exposure during development is associated with a
distinct histone H3 methylation patterns at lysine 9 and lysine multifactorial developmental deficit known as fetal alcohol
4, which correlated with increases and decreases in gene spectrum disorders (FASD) wherein its mechanism is not
expression, respectively (Pal-Bhadra et al. 2007). Further- clear. However, such alcohol effects may be caused by its
more, epidemiological observations implicate excess ethanol interference with OCM and induction of aberrant methylation
ingestion, as well as low dietary folate (the latter being one of (Liu et al. 2009; Zhou et al. 2011). Recent animal study
the most studied aspects of the toxic role of OCM performed on a model of gestational alcohol exposure
impairment; Fuso and Scarpa 2011) as risk factors for demonstrated changes in the expression of an epigenetically
several cancers of the oral cavity, esophagus, liver, colon, sensitive allele, Agouti viable yellow (Avy), in the offspring
rectum, and breast (Giovannucci 2002; Dumitrescu and after maternal ethanol consumption (Kaminen-Ahola et al.
Shields 2005; Boffetta et al. 2006). Alcohol drinking was 2010). As a result, the probability of transcriptional silencing
associated with hypermethylation at DNA repair genes such at this locus increased, enhancing the number of offspring
as the O6-methyl guanine methyl transferase involved in the with an agouti-colored coat. This transcriptional silencing
removal of mutagenic methyl adducts from guanine (Puri correlated with hypermethylation at Avy. This reveals that
et al. 2005) which may impact DNA repair leading to ethanol can affect adult phenotype epigenetically changing
genomic instability which plays a causative role in carcino- the early embryo. Alcohol-induced changes in DNA meth-
genesis. These findings suggest the involvement of OCM ylation which are highly regulated during development, and
impairment and consequent aberrations in DNA methylation which disrupts key developmental events, such as differen-
in the mechanisms underlying cancer. The role of abnormal tiation and the fate of early developing precursor cells
hepatic OCM in alcohol liver disease is well documented (Sachdev 2005; Liu et al. 2009; Wu and Zhang 2011; Zhou
(Medici and Halsted 2013) and supported by the protective et al. 2011), can underlie FASD, analogous to developmen-
effects of supplementation with the universal methyl donor in tal defects associated with aberrant DNA methylation under
eukaryotes, SAM (Song et al. 2003; Bailey et al. 2006; conditions of vitamin B12 or folic acid deficiency (Cravo and
Sykora et al. 2009; Andringa et al. 2010). Ethanol has been Camilo 2000; Mason and Choi 2005). Indeed, chronic
found to alter DNA methylation in the brain (Pignataro et al. ethanol consumption in pregnant mice resulted in demethy-
2009). Under conditions of chronic alcohol exposure, lation of fetal DNA (Garro et al. 1991). Such epigenetic
hyperhomocysteinemia - a hallmark of OCM dysfunction is alterations have been found to disrupt migration, neural cell
correlated with loss of brain volume, as well as cognitive and lineage differentiation during early brain development (Feng
memory decline and withdrawal seizures (Vogiatzoglou et al. 2007; MacDonald and Roskams 2009). In vitro
et al. 2008; Bleich and Hillemacher 2009; Coppede 2010; experiments confirmed that ethanol effects associated with
Tangney et al. 2011). Recently, we demonstrated that compromised DNA methylation patterns also inhibited
chronic ethanol exposure results in global DNA hypomethy- neural stem cell differentiation, similar to treatment with
lation of brain tissue and neurotoxicity, and this alcohol the methylation inhibitor 5-aza-cytidine (Zhou et al. 2011).
neurotoxicity was exaggerated by the additional OCM Furthermore, it has been demonstrated that the alcohol-
impairment in mice deficient in MTHFR (Fowler et al. 2012), induced changes in gene expression were linked to changes

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


776 I. I. Kruman and A.-K. Fowler

in DNA methylation in the hippocampus and prefrontal chromatin methylation. DNA methylation is an important
cortex of animal FASD models (Otero et al. 2012; Resendiz epigenetic determinant in gene expression (Mason 2003). As
et al. 2013). Ethanol-induced alterations in methylation of epigenetic information can be inherited, and maintained
particular genes were also demonstrated. For example, fetal through DNA replication cycles, it can also be modified by
alcohol exposure induced long-lasting hypermethylation of exposure to agents that interfere with OCM or other elements of
Pomc gene involved in stress control, metabolic, and immune DNA methylation machinery (e.g., DNMTs) such as diet,
functions (Govorko et al. 2012). These effects were found nutrients, or chemicals and alcohol (Dolinoy et al. 2007; Choi
during early gastrulation periods (Kaminen-Ahola et al. and Friso 2010). Aberrant OCM therefore could result in a
2010). Gastrulation is generally considered the most sensitive lasting impact on gene expression at the transcriptional level
to teratogenic insults, because differentiating cells appear to associated with various abnormalities (Anway et al. 2005).
be particularly vulnerable to the teratogenic effects of alcohol Indeed, both folate deficiency and ethanol exposure - which
(Kobor 2011). For instance, ethanol exposure of pregnant lead to OCM dysfunction - are associated with liver (Guerrerio
mice from the 9th through the 11th day of gestation resulted et al. 2012; Medici and Halsted 2013) and heart (Verhaar et al.
in global hypomethylation of fetal DNA (Garro et al. 1991). 2002; Djousse and Gaziano 2008) diseases, cancer (Gio-
Accumulating evidence suggests that alteration in DNA vannucci 2002; Dumitrescu and Shields 2005; Boffetta et al.
methylation may affect not only one generation but produce 2006), and neurodegeneration (Harper 2009; Fuso 2013).
effects across generations (Anway et al. 2005). This concept Since epigenetic alterations are especially important during
of transgenerational transmission of altered DNA methyla- differentiation (Wu and Zhang 2011), agents that interfere with
tion is supported by work showing specific exposure-induced OCM or other factors involved in methylation, may disturb
sperm methylation patterns reproduced between generations cellular differentiation and organogenesis during embryonic
(Guerrero-Bosagna et al. 2010). Indeed, the offspring sired development (Sinclair et al. 2007). Indeed, both folate defi-
by male mice chronically exposed to ethanol prior to ciency and alcohol are teratogenic (Pogribny et al. 1997;
conception inherited aberrant epigenetic marks and revealed Brunaud et al. 2003; Downing et al. 2011; Kobor 2011).
growth retardation phenotype in the immediate post weaning These observations strongly support the role of OCM impair-
period (Knezovich and Ramsay 2012). Importantly, male ment and consequent epigenetic alterations in alcohol toxicity.
gametes in both mice and humans chronically exposed to
alcohol underwent site-specific hypomethylation (Ouko
et al. 2009; Stouder et al. 2011), consistent with a significant Acknowledgments and conflict of interest
decrease in postnatal growth of children of alcoholic men disclosure
(Little and Sing 1987). Consistent with these profound This work was supported by the Foundation for Alcohol Research,
epigenetic effects of alcohol, are the beneficial effects of the South Plains Alcohol and Addiction Research Center
supplementation of animals with choline, betaine or folic (SPAARC).
acid before mating, throughout prenatal or neonatal periods. All experiments were conducted in compliance with the ARRIVE
Indeed, the supplementation has been shown to reverse guidelines. The authors have no conflict of interest to declare.
alcohol-induced changes in methylation and partially ame-
liorated ethanol teratogenesis (Thomas et al. 2010; Downing
References
et al. 2011; Otero et al. 2012). Together, these findings
suggest that alcohol has a substantial impact on the Alim M. A., Ma Q. L., Takeda K. et al. (2004) Demonstration of a role
epigenome via OCM disturbance, and these epigenetic for alpha-synuclein as a functional microtubule-associated protein.
J. Alzheimers Dis. 6, 435–442.
aberrations may play an important role in alcohol-induced
Allan A. M., Liang X., Luo Y., Pak C., Li X., Szulwach K. E., Chen D.,
developmental defects. Jin P. and Zhao X. (2008) The loss of methyl-CpG binding protein
1 leads to autism-like behavioral deficits. Hum. Mol. Genet. 17,
2047–2057.
Conclusion Andringa K. K., King A. L., Eccleston H. B., Mantena S. K., Landar A.,
Jhala N. C., Dickinson D. A., Squadrito G. L. and Bailey S. M.
Excessive alcohol consumption has adverse effects on many
(2010) Analysis of the liver mitochondrial proteome in response to
tissues and organ systems (Harper 2009). As alcohol’s harmful ethanol and S-adenosylmethionine treatments: novel molecular
health effects are well known, the underlying mechanisms are targets of disease and hepatoprotection. Am. J. Physiol. Gastrointest.
not fully understood. One of the mechanisms underlying Liver Physiol. 298, G732–G745.
alcohol toxicity is OCM impairment induced by ethanol or its Anway M. D., Cupp A. S., Uzumcu M. and Skinner M. K. (2005)
Epigenetic transgenerational actions of endocrine disruptors and
metabolites (Cravo et al. 1996; Giovannucci 2004; Fowler
male fertility. Science 308, 1466–1469.
et al. 2012). OCM is essential for cellular homeostasis, Bach J. F. (2002) The effect of infections on susceptibility to
providing cells with DNA precursors, necessary for cell autoimmune and allergic diseases. N. Engl. J. Med. 347, 911–920.
proliferation and DNA repair. It is likewise the main source of Bailey S. M., Robinson G., Pinner A. et al. (2006) S-adenosyl-
methyl groups for methylation reactions, including DNA and methionine prevents chronic alcohol-induced mitochondrial

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


Alcohol toxicity and DNA methylation 777

dysfunction in the rat liver. Am. J. Physiol. Gastrointest. Liver Choi S. W. and Friso S. (2010) Epigenetics: a new bridge between
Physiol. 291, G857–G867. nutrition and health. Adv. Nutr. 1, 8–16.
Barak A. J., Beckenhauer H. C., Junnila M. and Tuma D. J. (1993) Coppede F. (2010) One-carbon metabolism and Alzheimer’s disease:
Dietary betaine promotes generation of hepatic S-adenosyl- focus on epigenetics. Curr. Genomics 11, 246–260.
methionine and protects the liver from ethanol-induced fatty Cornacchia E., Golbus J., Maybaum J., Strahler J., Hanash S. and
infiltration. Alcohol. Clin. Exp. Res. 17, 552–555. Richardson B. (1988) Hydralazine and procainamide inhibit T cell
Berdasco M. and Esteller M. (2013) Genetic syndromes caused by DNA methylation and induce autoreactivity. J. Immunol. 140,
mutations in epigenetic genes. Hum. Genet. 132, 359–383. 2197–2200.
Bleich S. and Hillemacher T. (2009) Homocysteine, alcoholism and its Cravo M. L. and Camilo M. E. (2000) Hyperhomocysteinemia in chronic
molecular networks. Pharmacopsychiatry 42(Suppl 1), S102–S109. alcoholism: relations to folic acid and vitamins B(6) and B(12)
Bleich S., Lenz B., Ziegenbein M., Beutler S., Frieling H., Kornhuber J. status. Nutrition 16, 296–302.
and B€ onsch D. (2006) EpigeneticDNA hypermethylation of the Cravo M. L., Gloria L. M., Selhub J., Nadeau M. R., Camilo M. E.,
HERP gene promoterinduces down-regulation of its mRNA Resende M. P., Cardoso J. N., Leit~ao C. N. and Mira F. C. (1996)
expression in patients with alcohol dependence. Alcohol. Clin. Hyperhomocysteinemia in chronic alcoholism: correlation with
Exp. Res. 30, 587–591. folate, vitamin B-12, and vitamin B-6 status. Am. J. Clin. Nutr. 63,
Blom H. J., Shaw G. M., den Heijer M. and Finnell R. H. (2006) Neural 220–224.
tube defects and folate: case far from closed. Nat. Rev. Neurosci. 7, D’Anci K. E. and Rosenberg I. H. (2004) Folate and brain function in
724–731. elderly. Curr. Opin. Clin. Nutr. Metab. Care 7, 659–664.
Boffetta P., Hashibe M., Vecchia C. L., Zatonski W. and Rehm J. (2006) Deitrich R. (2011) Ethanol as aprodrug: brain metabolism of ethanol
The burden of cancer attributable to alcohol drinking. Int. J. mediates its reinforcing effects–a commentary. Alcohol. Clin. Exp.
Cancer 119, 884–887. Res. 35, 581–583.
Bohacek J., Gapp K., Saab B. J. and Mansuy I. M. (2013) Deng X. S. and Deitrich R. A. (2008) Putative role of brain acetaldehyde
Transgenerational epigenetic effects on brain functions. Biol. in ethanol addiction. Curr. Drug Abuse Rev. 1, 3–8.
Psychiatry 73, 313–320. Detich N., Bovenzi V. and Szyf M. (2003) Valproate induces
Bonsch D., Lenz B., Kornhuber J. and Bleich S. (2005) DNA replication-independent active DNA demethylation. J. Biol.
hypermethylation of the alpha synuclein promoter in patients Chem. 278, 27586–27592.
with alcoholism. NeuroReport 16, 167–170. Djousse L. and Gaziano M. J. (2008) Alcohol consumption and heart
Bonsch D., Lenz B., Fiszer R., Frieling H., Kornhuber J. and Bleich S. failure. Curr. Atheroscler. Rep. 10, 117–120.
(2006) Lowered DNA methyltransferase (DNMT-3b) mRNA Dobosy J. R. and Selker E. U. (2001) Emerging connections between
expression is associated with genomic DNA hypermethylation DNA methylation and histone acetylation. Cell. Mol. Life Sci. 58,
in patients with chronic alcoholism. J. Neural. Transm. 113, 721–727.
1299–1304. Dolinoy D. C., Weidman J. R. and Jirtle R. L. (2007) Epigenetic gene
Bottiglieri T. and Hyland K. (1994) S-adenosylmethionine levels in regulation: linking early developmental environment to adult
psychiatric and neurological disorders: a review. Acta Neurol. disease. Reprod. Toxicol. 23, 297–307.
Scand. Suppl. 154, 19–26. Dong E., Nelson M., Grayson D. R., Costa E. and Guidotti A. (2008)
Brunaud L., Alberto J. M., Ayav A., Gerard P., Namour F., Antunes L., Clozapine and sulpiride but not haloperidol or olanzapine activate
Braun M., Bronowicki J. P., Bresler L. and Gueant J. L. (2003) brain DNA demethylation. Proc. Natl Acad. Sci. USA 105,
Effects of vitamin B12 and folate deficiencies on DNA methylation 13614–13619.
and carcinogenesis in rat liver. Clin. Chem. Lab. Med. 41, Downing C., Johnson T. E., Larson C., Leakey T. I., Siegfried R. N.,
1012–1019. Rafferty T. M. and Cooney C. A. (2011) Subtle decreases in DNA
Bruniquel D. and Schwartz R. H. (2003) Selective, stable demethylation methylation and gene expression at the mouse Igf2 locus following
of the interleukin-2 gene enhances transcription by an active prenatal alcohol exposure: effects of a methyl-supplemented diet.
process. Nat. Immunol. 4, 235–240. Alcohol 45, 65–71.
Carpenter S. P., Lasker J. M. and Raucy J. L. (1996) Expression, Dumitrescu R. G. and Shields P. G. (2005) The etiology of alcohol-
induction, and catalytic activity of the ethanol-inducible induced breast cancer. Alcohol 35, 213–225.
cytochrome P450 (CYP2E1) in human fetal liver and Ehrlich M. (2003) The ICF syndrome, a DNA methyltransferase 3B
hepatocytes. Mol. Pharmacol. 49, 260–268. deficiency and immunodeficiency disease. Clin. Immunol. 109,
Cederbaum A. I. (2012) Alcohol metabolism. Clin. Liver Dis. 16, 17–28.
667–685. Feng J. and Fan G. (2009) The role of DNA methylation in the central
Cervoni N. and Szyf M. (2001) Demethylase activity is directed by nervous system and neuropsychiatric disorders. Int. Rev.
histone acetylation. J. Biol. Chem. 276, 40778–40787. Neurobiol. 89, 67–84.
Cervoni N., Detich N., Seo S.B., Chakravarti D. and Szyf M. (2002) The Feng J., Fouse S. and Fan G. (2007) Epigenetic regulation of neural gene
oncoprotein Set/TAF-1beta, an inhibitor of histone expression and neuronal function. Pediatr. Res. 61, 58R–63R.
acetyltransferase, inhibits active demethylation of DNA, Feng J., Zhou Y., Campbell S. L., Le T., Li E., Sweatt J. D., Silva A. J.
integrating DNA methylation and transcriptional silencing. J. Biol. and Fan G. (2010) Dnmt1 and Dnmt3a maintain DNA methylation
Chem. 277, 25026–25031. and regulate synaptic function in adult forebrain neurons. Nat.
Champagne F. A. (2010) Epigenetic perspectives on development: Neurosci. 13, 423–430.
evolving insights on the origins of variation. Dev. Psychobiol. 52, e1. Fowler A. K., Hewetson A., Agrawal R. G. et al. (2012) Alcohol-
Cheng X. and Blumenthal R. M. (2010) Coordinated chromatin control: induced one-carbon metabolism impairment promotes dysfunction
structural and functional linkage of DNA and histone methylation. of DNA base excision repair in adult brain. J. Biol. Chem. 287,
Biochemistry 49, 2999–3008. 43533–43542.
Chiang P. K., Gordon R. K., Tal J., Zeng G. C., Doctor B. P., Francis D., Diorio J., Liu D. and Meaney M. J. (1999) Nongenomic
Pardhasaradhi K. and McCann P. P. (1996) S-Adenosylmethionine transmission across generations of maternal behavior and stress
and methylation. FASEB J. 10, 471–480. responses in the rat. Science 286, 1155–1158.

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


778 I. I. Kruman and A.-K. Fowler

Frieling H., Hillemacher T., Demling J. H., Kornhuber J. and Bleich S. Hirano T. (2011) Alcohol consumption and oxidative DNA damage. Int.
(2007) Newoptionsinthe treatment of depression. Fortschr. Neurol. J. Environ. Res. Public Health 8, 2895–2906.
Psychiatrie 75, 641–652. Huang K. and Fan G. (2010) DNA methylation in cell differentiation and
Fuso A. (2013) The ‘golden age’ of DNA methylation in neu reprogramming: an emerging systematic view. Regen. Med. 5,
rodegenerative diseases. Clin. Chem. Lab. Med. 51, 523–534. 531–544.
Fuso A. and Scarpa S. (2011) One-carbon metabolism and Alzheimer’s Irizarry M. C., Gurol M. E., Raju S., Diaz-Arrastia R., Locascio J. J.,
disease: is it all a methylation matter? Neurobiol. Aging 32, Tennis M., Hyman B. T., Growdon J. H., Greenberg S. M. and
1192–1195. Bottiglieri T. (2005) Association of homocysteine with plasma
Fuso A., Ferraguti G., Grandoni F., Ruggeri R., Scarpa S., Strom R. and amyloid beta protein in aging and neurodegenerative disease.
Lucarelli M. (2010) Early demethylation of non-CpG, CpC-rich, Neurology 65, 1402–1408.
elements in the myogenin 50 -flanking region: a priming effect on James S. J., Melnyk S., Pogribna M. and Caudill M. A. (2002) Elevation
the spreading of active demethylation. Cell Cycle 9, 3965–3976. of S adenosylhomocysteine and DNA hypomethylation: potential
Fuso A., Nicolia V., Ricceri L., Cavallaro R. A., Isopi E., Mangia F. and epigenetic mechanism for homocysteine-related pathology. J. Nutr.
Scarpa S. (2012) S-adenosylmethionine reduces the progress of the 132, 2361S–2366S.
Alzheimer-like features induced by B-vitamin deficiency in mice. Ji W., Hernandez R., Zhang X. Y., Qu G. Z., Frady A., Varela
Neurobiol. Aging 33(1482), e1. M. and Ehrlich M. (1997) DNA demethylation and
Garro A. J., McBeth D. L., Lima V. and Lieber C. S. (1991) Ethanol pericentromeric rearrangements of chromosome 1. Mutat. Res. 379,
consumption inhibits fetal DNA methylation in mice: implications 33–41.
for the fetal alcohol syndrome. Alcohol. Clin. Exp. Res. 15, 395–398. Jin S. G., Kadam S. and Pfeifer G. P. (2010) Examination of the
Giovannucci E. (2002) Epidemiologic studies of folate and colorectal specificity of DNA methylation profiling techniques towards
neoplasia. J. Nutr. 132, 2350–2355. 5-methylcytosine and 5-hydroxymethylcytosine. Nucleic Acids
Giovannucci E. (2004) Alcohol, one-carbon metabolism, and colorectal Res. 38, e125.
cancer: recent insights from molecular studies. J. Nutr. 134, Kaminen-Ahola N., Ahola A., Maga M., Mallitt K. A., Fahey P., Cox T.
2475S–2481S. C., Whitelaw E. and Chong S. (2010) Maternal ethanol
Gluckman P. D., Hanson M. A., Buklijas T., Low F. M. and Beedle A. S. consumption alters the epigenotyp eand the phenotype of
(2009) Epigenetic mechanisms that underpin metabolic and offspring in a mouse model. PLoS Genet. 6, e1000811.
cardiovascular diseases. Nat. Rev. Endocrinol. 5, 401–408. Kharbanda K. K. (2009) Alcoholic liver disease and methionine
Govorko D., Bekdash R. A., Zhang C. and Sarkar D. K. (2012) Male metabolism. Semin. Liver Dis. 29, 155–165.
germline transmits fetal alcohol adverse effect on hypothalamic Knezovich J. C. and Ramsay M. (2012) The effect of preconception
proopiomelanocortin gene across generations. Biol. Psychiatry 72, paternal alcohol exposure on epigenetic remodeling of the h19 and
378–388. rasgrf1 imprinting control regions in mouse offspring. Front.
Gr€aff J., Woldemichael B. T., Berchtold D., Dewarrat G. and Mansuy I. Genet. 3, 10–20.
M. (2012) Dynamic histone marks in the hippocampus and cortex Kobor W. J. (2011) Epigenetics and fetal alcohol spectrum disorders.
facilitate memory consolidation. Nat. Commun. 3, 991. Alcohol Res. Health 34, 29–37.
Gruenbaum Y., Stein R., Cedar H. and Razin A. (1981) Methylation of Kruman I. I., Kumaravel T. S., Lohani A., Pedersen W. A., Cutler R. G.,
CpG sequences in eukaryotic DNA. FEBS Lett. 124, 67–71. Kruman Y., Haughey N., Lee J., Evans M. and Mattson M. P.
Gruenbaum Y., Cedar H. and Razin A. (1982) Substrate and sequence (2002) Folic acid deficiency and homocysteine impair DNA repair
specificity of a eukaryotic DNA methylase. Nature 295, 620–622. in hippocampal neurons and sensitize them to amyloid toxicity in
Guerrerio A. L., Colvin R. M., Schwartz A. K. et al. (2012) Choline experimental models of Alzheimer’s disease. J. Neurosci. 22,
intake in a large cohort of patients with nonalcoholic fatty liver 1752–1762.
disease. Am. J. Clin. Nutr. 95, 892–900. Krystal J. H., Petrakis I. L., Mason G., Trevisan L. and D’Souza D. C.
Guerrero-Bosagna C., Settles M., Lucker B. and Skinner M. K. (2010) (2003) N-methyl-D aspartate glutamate receptors and alcoholism:
Epigenetic transgenerational actions of vinclozolin on promoter reward, dependence, treatment and vulnerability. Pharmacol. Ther.
regions of the sperm epigenome. PLoS ONE 5, e13100–e13117. 99, 79–94.
Halsted C. H. and Medici V. (2011) Vitamin-dependent methionine Kucharski R., Maleszka J., Foret S. and Maleszka R. (2008) Nutritional
metabolism and alcoholic liver disease. Adv. Nutr. 2, 421–427. control of reproductivestatus in honeybees via DNA methylation.
Hamid A., Wani N. A. and Kaur J. (2009) New perspectives on folate Science 319, 1827–1830.
transport in relation to alcoholism-induced folate malabsorption- Lahiri D. K., Maloney B. and Zawia N. H. (2009) The LEARn model: an
association with epigenome stability and cancer development. epigenetic explanation for idiopathic neurobiological diseases.
FEBS J. 276, 2175–2191. Mol. Psychiatry 14, 992–1003.
Hansen R. S., Wijmenga C., Luo P., Stanek A. M., Canfield T. K., Lamberti P., Zoccolella S., Armenise E., Lamberti S. V., Fraddosio A.,
Weemaes C. M. and Gartler S. M. (1999) The DNMT3B DNA de Mari M., Iliceto G. and Livrea P. (2005) Hyperhomo-
methyltransferase gene is mutated in the ICF immunodeficiency cysteinemia in L-dopa treated Parkinson’s disease patients: effect
syndrome. Proc. Natl Acad. Sci. USA 96, 14412–14417. of cobalamin and folate administration. Eur. J. Neurol. 12, 365–
Haorah J., Ramirez S. H., Floreani N., Gorantla S., Morsey B. and 368.
Persidsky Y. (2008) Mechanism of alcohol-induced oxidative Laurence K. M., James N., Miller M. H., Tennant G. B. and Campbell H.
stress and neuronal injury. Free Radic. Biol. Med. 45, 1542–1550. (1981) Double-blind randomised controlled trial of folate treatment
Harper C. (2009) The neuropathology of alcohol-related brain damage. before conception to prevent recurrence of neural-tube defects. Br.
Alcohol Alcohol. 44, 136–140. Med. J. (Clin. Res. Ed.) 282, 1509–1511.
He K., Merchant A., Rimm E. B., Rosner B. A., Stampfer M. J., Willett Li Y., Liu Y., Strickland F. M. and Richardson B. (2010) Age-dependent
W. C. and Ascherio A. (2004) Folate, vitamin B6, and B12 intakes decreases in DNA methyltransferase levels and low transmethylation
in relation to risk of stroke among men. Stroke 35, 169–174. micronutrient levels synergize to promote overexpression of genes
Hillemacher T. (2011) Biological mechanisms in alcohol dependence– implicated in autoimmunity and acute coronary syndromes. Exp.
new perspectives. Alcohol Alcohol. 46, 224–230. Gerontol. 45, 312–322.

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


Alcohol toxicity and DNA methylation 779

Lieber C. S. (2004) The discovery of the microsomal ethanol oxidizing after maternal alcohol exposure on offspring rats. Addict. Biol. 10,
system and its physiologic and pathologic role. Drug Metab. Rev. 139–144.
36, 511–529. Nan X., Campoy F. J. and Bird A. (1997) MeCP2 is a transcriptional
Lill C. M. and Bertram L. (2011) Towards unveiling the genetics of repressor with abundant binding sites in genomic chromatin. Cell
neurodegenerative diseases. Semin. Neurol. 31, 531–541. 88, 471–481.
Lim U. and Song M. A. (2012) Dietary and lifestyle factors of DNA Okano M., Bell D. W., Haber D. A. and Li E. (1999) DNA
methylation. Methods Mol. Biol. 863, 359–376. methyltransferases Dnmt3a and Dnmt3b are essential for de novo
Lister R., Pelizzola M., Dowen R. H. et al. (2009) Human DNA methylation and mammalian development. Cell 99, 247–257.
methylomes at base resolution show widespread epigenomic Otero N. K., Thomas J. D., Saski C. A., Xia X. and Kelly S. J. (2012)
differences. Nature 462, 315–322. Choline supplementation and DNA methylation in the
Little R. E. and Sing C. F. (1987) Father’s drinking and infant birth hippocampus and prefrontal cortex of rats exposed to alcohol
weight: report of an association. Teratology 36, 59–65. during development. Alcohol. Clin. Exp. Res. 36, 1701–1709.
Liu D., Diorio J., Tannenbaum B., Caldji C., Francis D., Freedman A., Ou J. N., Torrisani J., Unterberger A., Provencal N., Shikimi K., Karimi
Sharma S., Pearson D., Plotsky P. M. and Meaney M. J. (1997) M., Ekstrom T. J. and Szyf M. (2007) Histone deacetylase inhibitor
Maternal care, hippocampal glucocorticoid receptors, and Trichostatin A induces global and gene-specific DNA
hypothalamic-pituitaryadrenal responses to stress. Science 277, demethylation in human cancer cell lines. Biochem. Pharmacol.
1659–1662. 73, 1297–1307.
Liu Y., Balaraman Y., Wang G., Nephew K. P. and Zhou F. C. (2009) Ouko L. A., Shantikumar K., Knezovich J., Haycock P., Schnugh D. J.
Alcohol exposure alters DNA methylation profiles in mouse and Ramsay M. (2009) Effect of alcohol consumption on CpG
embryos at early neurulation. Epigenetics 4, 500–511. methylation in the differentially methylated regions of H19 and
Liutkeviciute Z., Lukinavicius G., Masevicius V., Daujotyte D. and IG-DMR in male gametes: implications for fetal alcohol spectrum
Klimasauskas S. (2009) Cytosine-5-methyltransferases add disorders. Alcohol. Clin. Exp. Res. 33, 1615–1627.
aldehydes to DNA. Nat. Chem. Biol. 5, 400–402. Pal-Bhadra M., Bhadra U., Jackson D. J., Mamatha L., Park P. and
Lu S. C. (2000) S-Adenosylmethionine. Int. J. Biochem. Cell Biol. 32, Shukla S. D. (2007) Distinct methylation patterns in histone H3 at
391–3955. lys-4 and lys-9 correlated with up- and down-regulation of genes
Lucarelli M., Fuso A., Strom R. and Scarpa S. (2001) The dynamics of by ethanol in hepatocytes. Life Sci. 81, 979–987.
myogenin site-specific demethylation is strongly correlated with its Park P. H., Lim R. W. and Shukla S. D. (2005) Involvement of
expression and with muscle differentiation. J. Biol. Chem. 276, histone acetyltransferase (HAT) in ethanol-induced acetylation of
7500–7506. histone H3 in hepatocytes: potential mechanism for gene
MacDonald J. L. and Roskams A. J. (2009) Epigenetic regulation of expression. Am. J. Physiol. Gastrointest. Liver Physiol. 289,
nervous system development by DNA methylation and histone G1124–G1136.
deacetylation. Prog. Neurobiol. 88, 170–183. Pignataro L., Varodayan F. P., Tannenholz L. E. and Harrison N. L.
Maegawa S., Hinkal G., Kim H. S., Shen L., Zhang L., Zhang J., Zhang (2009) The regulation of neuronal gene expression by alcohol.
N., Liang S., Donehower L. A. and Issa J. P. (2010) Widespread Pharmacol. Ther. 124, 324–335.
and tissue specific age-related DNA methylation changes in mice. Pogribny I. P., Miller B. J. and James S. J. (1997) Alterations in hepatic
Genome Res. 20, 332–340. p53 gene methylation patterns during tumor progression with
Mason J. B. (2003) Biomarkers of nutrient exposure and status in one- folate/methyl deficiency in the rat. Cancer Lett. 115, 31–38.
carbon (methyl) metaboism. J. Nutr. 133, 941S–947S. Puri S. K., Si L., Fan C. Y. and Hanna E. (2005) Aberrant promoter
Mason J. B. and Choi S. W. (2005) Effects of alcohol on folate hypermethylation of multiple genes in head squamous cell
metabolism: implications for carcinogenesis. Alcohol 35, 235–241. carcinoma. Am. J. Otolaryngol. 26, 12–17.
Mastroeni D., Grover A., Delvaux E., Whiteside C., Coleman P. D. and Purohit V., Abdelmalek M. F., Barve S. et al. (2007) Role of S-
Rogers J. (2011) Epigenetic mechanisms in Alzheimer’s disease. adenosylmethionine, folate, and betaine in the treatment of
Neurobiol. Aging 32, 1161–1180. alcoholic liver disease: summary of a symposium. Am. J. Clin.
McGowan P. O., Suderman M., Sasaki A., Huang T. C., Hallett M., Nutr. 86, 14–24.
Meaney M. J. and Szyf M. (2011) Broad epigenetic signature of Qiang M., Denny A., Chen J., Ticku M. K., Yan B. and Henderson G.
maternal care in the brain of adult rats. PLoS ONE 6, e14739. (2010) The site specific demethylation in the 50 -regulatory area of
Medici V. and Halsted C. H. (2013) Folate, alcohol, and liver disease. NMDA receptor 2B subunit gene associated with CIE-induced up-
Mol. Nutr. Food Res. 57, 596–606. regulation of transcription. PLoS ONE 5, e8798.
Migliore L. and Coppede F. (2009) Genetics, environmental factors and Qureshi I. A. and Mehler M. F. (2012) Epigenetic mechanisms
the emerging role of epigenetics in neurodegenerative diseases. governing the process of neurodegeneration. Mol. Aspects Med.
Mutat. Res. 667, 82–97. 34, 875–882.
Miller A. L. (2003) The methionine-homocysteine cycle and its effects Razin A. and Cedar H. (1977) Distribution of 5-methylcytosine in
on cognitive diseases. Altern. Med. Rev. 8, 7–19. chromatin. Proc. Natl Acad. Sci. USA 74, 2725–2728.
Montoliu C., Valles S., Reneu-Piqueras J. and Guerri C. (1994) Ethanol Razin A. and Riggs A. D. (1980) DNA methylation and gene function.
induced oxygen radical formation and lipid peroxidation in rat Science 210, 604–610.
brain: effect of chronic alcohol consumption. J. Neurochem. 63, Razin A., Webb C., Szyf M., Yisraeli J., Rosenthal A., Naveh-Many T.,
1855–1862. Sciaky-Gallili N. and Cedar H. (1984) Variations in DNA
Murgatroyd C., Patchev A. V., Wu Y., Micale V., Bockm€uhl Y., Fischer methylation during mouse cell differentiation in vivo and in
D., Holsboer F., Wotjak C. T., Almeida O. F. and Spengler D. vitro. Proc. Natl Acad. Sci. USA 81, 2275–2279.
(2009) Dynamic DNA methylation programs persistent adverse Resendiz M., Chen Y., Ozt€urk N. C. and Zhou F. C. (2013) Epigenetic
effects of early-life stress. Nat. Neurosci. 12, 1559–1566. medicineand fetal alcohol spectrumdisorders.Epigenomics 5, 73–86.
Murillo-Fuentes M. L., Artillo R., Ubeda N., Varela-Moreiras G., Reynolds E. H. (2002) Folic acid, ageing, depression, and dementia.
Murillo M. L. and Carreras O. (2005) Hepatic S-adenosylmethionine BMJ 324, 1512–1515.

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780


780 I. I. Kruman and A.-K. Fowler

Roth T. L., Lubin F. D., Funk A. J. and Sweatt J. D. (2009) Lasting Thomas J. D., Idrus N. M., Monk B. R. and Dominguez H. D. (2010)
epigenetic influence of early-life adversity on the BDNF gene. Prenatal choline supplementation mitigates behavioral alterations
Biol. Psychiatry 65, 760–769. associated with prenatal alcohol exposure in rats. Birth Defects Res.
Sachdev P. S. (2005) Homocysteine and brain atrophy. Prog. Part A Clin. Mol. Teratol. 88, 827–837.
Neuropsychopharmacol. Biol. Psychiatry 29, 1152–1161. Verhaar M. C., Stroes E. and Rabelink T. J. (2002) Folates and
Seitz H. K. and Stickel F. (2007) Molecular mechanisms of alcohol- Cardiovascular disease. Arterioscler. Thromb. Vasc. Biol. 22,
mediated carcinogenesis. Nat. Rev. Cancer 7, 599–612. 6–13.
Sinclair K. D., Allegrucci C., Singh R. et al. (2007) DNA methylation, Villanueva J. A. and Halsted C. H. (2004) Hepatic transmethylation
insulin resistance, and blood pressure in offspring determined by reactions in micropigs with alcoholic liver disease. Hepatology 39,
maternal periconceptional B vitamin and methionine status. Proc. 1303–1310.
Natl Acad. Sci. USA 104, 19351–19356. Villanueva J. A., Esfandiari F., White M. E., Devaraj S., French S. W.
Singh K. and Jaiswal D. (2013) One-carbon metabolism, and Halsted C. H. (2007) S-adenosylmethionine attenuates
spermatogenesis, and male infertility. Reprod. Sci. 20, 622–630. oxidative liver injury in micropigs fed ethanol with a folate-
Slatkin M. (2009) Epigenetic inheritance and the missing heritability deficient diet. Alcohol. Clin. Exp. Res. 31, 1934–1943.
problem. Genetics 182, 845–850. Vogiatzoglou A., Refsum H., Johnston C., Smith S. M., Bradley K. M.,
Song Z., Zhou Z., Chen T., Hill D., Kang J., Barve S. and McClain C. de Jager C., Budge M. M. and Smith A. D. (2008) Vitamin B12
(2003) S-adenosylmethionine (SAMe) protects against acute alcohol status and rate of brain volume loss in community-dwelling elderly.
induced hepatotoxicity in mice. J. Nutr. Biochem. 14, 591–597. Neurology 71, 826–832.
Stanger O., Fowler B., Piertzik K., Huemer M., Haschke-Becher E., Warner M. and Gustafsson J. A. (1994) Effect of ethanol on
Semmler A., Lorenzl S. and Linnebank M. (2009) Homocysteine, cytochrome P450 in the rat brain. Proc. Natl Acad. Sci. USA 91,
folate and vitamin B12 in neuropsychiatric diseases: review and 1019–1023.
treatment recommendations. Expert Rev. Neurother. 9, 1393–1412. Weaver I. C., Cervoni N., Champagne F. A., D’Alessio A. C., Sharma
Stickel F., Choi S. W., Kim Y. I., Bagley P. J., Seitz H. K., Russell R. S., Seckl J. R., Dymov S., Szyf M. and Meaney M. J. (2004)
M., Selhub J. and Mason J. B. (2000) Effect of chronic alcohol Epigenetic programming by maternal behavior. Nat. Neurosci. 7,
consumption on total plasma homocysteine level in rats. Alcohol. 847–854.
Clin. Exp. Res. 24, 259–264. Weaver I. C., Michael J., Meaney M. J. and Moshe Szyf M. (2006)
Stouder C., Somm E. and Paoloni-Giacobino A. (2011) Prenatal Maternal care effects on the hipocampal transcriptome and anxiety-
exposure to ethanol: a specific effect on the H19 gene in sperm. mediated behaviors in the offspring that are reversible in
Reprod. Toxicol. 31, 507–512. adulthood. Proc. Natl Acad. Sci. USA 103, 3480–3485.
Stover P. J. (2004) Physiology of folate and vitamin B12 in health and Wu H. and Zhang Y. (2011) Mechanisms and functions of Tet
disease. Nutr. Rev. 62, S3–S12. protein-mediated 5-methylcytosine oxidation. Genes Dev. 25,
Stover P. J. (2009) One-carbon metabolism-genome interactions in 2436–2452.
folate-associated pathologies. J. Nutr. 139, 2402–2405. Wu D., Wang X., Zhou R. and Cederbaum A. (2010) CYP2E1
Sunden S. L., Renduchintala M. S., Park E. I., Miklasz S. D. and Garrow enhances ethanol-induced lipid accumulation but impairs
T. A. (1997) Betaine-homocysteine methyltransferase expression autophagy in HepG2 E47 cells. Biochem. Biophys. Res.
in porcine and human tissues and chromosomal localization of the Commun. 402, 116–122.
human gene. Arch. Biochem. Biophys. 345, 171–174. Yung R. L. and Richardson B. C. (1994) Role of T cell DNA
Sykora P., Kharbanda K. K., Crumm S. E. and Cahill A. (2009) S- methylation in lupus syndromes. Lupus 3, 487–491.
adenosyl-L-methionine co-administration prevents the ethanol- Zentner G. E. and Henikoff S. (2013) Regulation of nucleosome
elicited dissociation of hepatic mitochondrial ribosomes in male dynamics by histone modifications. Nat. Struct. Mol. Biol. 20,
rats. Alcohol. Clin. Exp. Res. 33, 1–9. 259–266.
Szyf M. (2011) The implications of DNA methylation for toxicology: Zerilli A., Lucas D., Amet Y., Beauge F., Volant A., Folch H. H.,
toward toxicomethylomics, the toxicology of DNA methylation. Berthou F. and Menez J. F. (1995) Cytochrome P-450 2E1 in rat
Toxicol. Sci. 120, 235–255. liver, kidney and lung microsomes after chronic administration of
Tahiliani M., Koh K. P., Shen Y. et al. (2009) Conversion of 5- ethanol either orally or by inhalation. Alcohol Alcohol. 30,
methylcytosine to 5-hydroxymethylcytosine in mammalian DNA 357–365.
by MLL partner TET1. Science 324, 930–935. Zhou F. C., Balaraman Y., Teng M. X., Yunlong Liu Y., Singh R.,
Tangney C. C., Aggarwal N. T., Li H., Wilson R. S., Decarli C., Evans Kenneth P. and Nephew K. P. (2011) Alcohol alters DNA
D. A. and Morris M. C. (2011) Vitamin B12, cognition, and brain Methylation patterns and inhibits neural stem cell differentiation.
MRI measures: a crosssectional examination. Neurology 77, Alcohol. Clin. Exp. Res. 35, 735–746.
1276–1282. Zhu J. K. (2009) Active DNA demethylation mediated by DNA
Tchantchou F., Graves M., Ortiz D., Chan A., Rogers E. and Shea T. B. glycosylases. Annu. Rev. Genet. 43, 143–166.
(2006) S-adenosyl methionine: a connection between nutritional Zimatkin S. M., Pronko S. P., Vasiliou V., Gonzalez F. J. and Deitrich R.
and genetic risk factors for neurodegeneration in Alzheimer’s A. (2006) Enzymatic mechanisms of ethanol oxidation in the brain.
disease. J. Nutr. Health Aging 10, 541–544. Alcohol. Clin. Exp. Res. 30, 1500–1505.

© 2014 International Society for Neurochemistry, J. Neurochem. (2014) 129, 770--780

You might also like