ArticlePDF AvailableLiterature Review

Journal Pre-proof Epigenetic modification impacting brain functions: Effects of physical activity, micronutrients, caffeine, toxins, and addictive substances Epigenetic modification impacting brain functions: Effects of physical activity, micronutrients, caffeine, toxins, and addictive substances

Authors:

Abstract

Abstract: Changes in gene expression are involved in many brain functions. Epigenetic processes modulate gene expression by histone modification and DNA methylation or RNA-mediated processes, which is important for brain function. Consequently, epigenetic changes are also a part of brain diseases such as mental illness and addiction. Understanding the role of different factors on the brain epigenome may help us understand the function of the brain. This review discussed the effects of caffeine, lipids, addictive substances, physical activity, and pollutants on the epigenetic changes in the brain and their modulatory effects on brain function.
Journal Pre-proof
Epigenetic modification impacting brain functions: Effects of physical activity,
micronutrients, caffeine, toxins, and addictive substances
Rahul Mallick, Asim K. Duttaroy
PII: S0197-0186(23)00155-9
DOI: https://doi.org/10.1016/j.neuint.2023.105627
Reference: NCI 105627
To appear in: Neurochemistry International
Received Date: 23 August 2023
Revised Date: 6 October 2023
Accepted Date: 7 October 2023
Please cite this article as: Mallick, R., Duttaroy, A.K., Epigenetic modification impacting brain functions:
Effects of physical activity, micronutrients, caffeine, toxins, and addictive substances, Neurochemistry
International (2023), doi: https://doi.org/10.1016/j.neuint.2023.105627.
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.
© 2023 Published by Elsevier Ltd.
1
Epigenetic modification impacting brain functions: Effects of
physical activity, micronutrients, caffeine, toxins, and addictive
substances.
Rahul Mallick1, and Asim K. Duttaroy2*
1A.I. Virtanen Institute for Molecular Sciences,
University of Eastern Finland, Finland
2 Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine,
University of Oslo, POB 1046 Blindern, Oslo, Norway
*Corresponding Author
Professor Asim K. Duttaroy
Department of Nutrition
Institute of Basic Medical Sciences,
Faculty of Medicine,
University of Oslo
Oslo, Norway
Email: a.k.duttaroy@medisin.uio.no
Tel: +47 22 82 15 47
Journal Pre-proof
2
Abstract:
Changes in gene expression are involved in many brain functions. Epigenetic processes
modulate gene expression by histone modification and DNA methylation or RNA-mediated
processes, which is important for brain function. Consequently, epigenetic changes are also
a part of brain diseases such as mental illness and addiction. Understanding the role of
different factors on the brain epigenome may help us understand the function of the brain.
This review discussed the effects of caffeine, lipids, addictive substances, physical activity,
and pollutants on the epigenetic changes in the brain and their modulatory effects on brain
function.
Keywords: Epigenetics; DNA methylation; brain; abusive drug; cannabis; caffeine, physical
activity
Abbreviations: CpG: CG dinucleotide; MRI: Magnetic resonance imaging; 5mC: 5-
methylcytosine; 5hmC: 5-hydroxymethylcytosine; PM: particulate matter.
Journal Pre-proof
3
Introduction
Epigenetics (the Greek word “epi” means over / around) studies chemical processes that
regulate gene expression by modulating DNA or its associated proteins without changing the
underlying DNA sequence. Various enzymes add epigenetic marks to the DNA to signal
specific genes to be active or silent, which drives cell and tissue differentiation. These
epigenetic marks vary from person to person or tissue to tissue and from cell type to cell type
within the tissue. Unlike DNA sequences, epigenetic modification is unique, changeable, and
partially heritable (Wang et al., 2022). DNA methylation, histone modifications, and the
actions of non-coding RNA molecules are epigenetic processes (Figure 1). Epigenetic
regulatory enzymes such as DNA methyltransferases, histone methyltransferases, and
histone deacetylases catalyze these alterations (Han et al., 2019). Changes in the amino acid
sequence of these enzymes have been demonstrated in studies to be closely associated with
various diseases (Han et al., 2019).
The addition of a methyl (-CH3) group to the fifth position carbon in the cytosine carbon ring
(in the context of a CG dinucleotide [CpG site]) to form 5-methylcytosine is defined as DNA
methylation (Youk et al., 2020). 70 80 % of CpG sites are methylated in humans (Youk et
al., 2020). Non-CpG methylation also occurs in the mammalian genome, specifically in the
brain (de Mendoza et al., 2021). However, the functional implication of this methylation has
yet to be made clear. Near the gene transcription starting site, CG content is enriched in
specific stretches of DNA known as CpG islands. Most gene promoters are associated with
CpG island (de Mendoza et al., 2021). Transcriptional initiation is inhibited by methylation
of promoter-associated CpG islands (Hughes et al., 2020). DNA methylation directly blocks
the binding of transcription factors to recognition sequences containing CpG sites and gene
Journal Pre-proof
4
expression (Héberlé and Bardet, 2019). Removal and reestablishment of DNA methylation
occur during gametogenesis and shortly following fertilization (Ivanova et al., 2020). DNA
methylation is carried out by a family of enzymes known as DNA (cytosine-5)-
methyltransferases, which are classified into three types: DNA (cytosine-5)-
methyltransferase 1, DNA (cytosine-5)-methyltransferase 2, and DNA (cytosine-5)-
methyltransferase 3 (Hervouet et al., 2018). DNA (cytosine-5)-methyltransferase 1 enzyme
maintains the methylation process during cell division (Hervouet et al., 2018), while de novo
methylation is maintained by DNA (cytosine-5)-methyltransferase 3a and 3b during early
development (Hervouet et al., 2018). DNA (cytosine-5)-methyltransferase 3l is
predominantly expressed during development to imprint genes and regulate DNA (cytosine-
5)-methyltransferase 3a and 3b (Hervouet et al., 2018). The functions of DNA (cytosine-5)-
methyltransferase 3l are still a mystery (Hervouet et al., 2018).
Histone determines the accessibility of stretched DNA to transcription-regulating molecules.
The tight-bound stretch of DNA to histones reduces the transcription activity. Amino acid
tails of histones can be modified post-transcriptionally to modulate the interactions among
histones, between histones and DNA, or support the recruitment of extra chromatin-
modifying proteins (Zhao and Shilatifard, 2019). Histone modifications and the enzymes that
carry them out can help in chromatin compaction, nucleosome dynamics, and transcription
(Zhao and Shilatifard, 2019). These changes can be made in response to both internal and
external stimuli. Histone acetylation, methylation, phosphorylation, and ubiquitination are
the four most prevalent modifications written by histone acetyltransferases, histone
methyltransferases, protein kinases, and ubiquitin ligases, respectively. Histone deacetylases,
histone demethylases, protein phosphatases, and deubiquitinating enzymes, on the other
Journal Pre-proof
5
hand, remove histone acetylation, methylation, phosphorylation, and ubiquitination,
respectively (Morgan and Shilatifard, 2020).
Translation-unable RNAs are non-coding RNAs that participate in epigenetic regulation by
recruiting histone- or DNA-modifying enzymes or directly modifying other RNAs or RNA-
protein complexes (Bure et al., 2022).
External factors e.g., physical activity, diet, and drugs, strongly influence epigenome (Galkin
et al., 2023; Toranõ et al., 2016). These modifications can impact any development phase and
modulate disease susceptibility (Galkin et al., 2023; Toranõ et al., 2016). Epigenetics may
influence biological changes, but nurture strongly impacts biological activities and behavior
(Figure 2). Various studies demonstrated the crucial role of histone modifications and non-
coding RNAs in memory formation in the brain and other forms of neuroplasticity (Dias et
al., 2015; Levenson and Sweatt, 2005; Saab and Mansuy, 2014; Sillivan et al., 2015). In addition,
existing variability in DNA methylation can modulate brain activities (Rasmi et al., 2023).
Albeit cell-specific nature of these dynamic epigenetic processes in human brain function
and behavior are quite unknown. Therefore, epigenetic mechanisms will help better
understand unexplained variability in neural phenotypes and precise molecular mechanisms
that may drive the emergence of inter-individual variability in brain activities. The human
genome's epigenetic modification is the subject of this article, focusing on how it links to the
effects of physical activity, micronutrients, caffeine, toxins, and addictive substances, which
addresses critical aspects of human health, genetics, and lifestyle choices. The selection
criteria of the mentioned factors can potentially inform healthcare, public policy, and
personal decision-making, ultimately contributing to improved health outcomes and a deeper
understanding of our genetic and epigenetic makeup.
Journal Pre-proof
6
Epigenetics and adult brain function
Recent studies have demonstrated that epigenetic processes are vital for brain function. The
impact of epigenetics on imaging genetics embraced the significance of environmental
factors in associations between brain function and sequence variants. Table 1 demonstrates
that psychiatric epigenetic studies targeting methylation within or near a gene's promoter
correlate with diminished gene expression and downstream neural phenotypes. These studies
indicate that DNA methylation patterns are influenced by an individual’s specific
environment, which explains the reason for variability in brain function than DNA sequence-
based variation alone (Liu et al., 2018). However, where and how these methylation patterns
start and how the mapping of methylation patterns in peripheral tissues onto the patterns in
the brain are still burgeoning fields. Despite variation in methylation, epigenetic marks are
partially heritable and modifiable in response to environmental factors (Liu et al., 2018).
Due to having similar DNA sequences in every cell (except in rare cases) of an organism
(Vijg, 2014), derived DNA from peripheral tissues should be similar to DNA in the human
brain. In contrast, epigenetic marks vary between cell types and tissues. Whether methylation
patterns measurable in DNA derived from peripheral tissues vary from methylation patterns
in brain needs to study further. Despite having varied methylation status among tissues, few
studies demonstrated blood-brain correlations in DNA methylation (Nishitani et al., 2023).
The exact mapping of entire brain and peripheral tissues methylomes remains unknown.
Several questions need to figure out to know better the epigenetics of human brain functions.
Are the mechanisms of different methylation processes in the brain and the peripheral tissues
similar or different? How does small-scale inter-individual variability in DNA methylation
affect gene expression, are the other epigenetic marks than DNA methylation correlate
Journal Pre-proof
7
between brain and peripheral tissue, and do the activities of DNA (cytosine-5)-
methyltransferase 1 through several cell divisions share the similar developmental origin of
cells or reflect experiences in adulthood, determining best proxy peripheral tissue for DNA
methylation in the brain, the correlation between the temporal stability of DNA methylation
markers in the brain and peripheral tissue methylomes demands further studies. Then
epigenetic marks can be used for diagnostic purposes accurately.
Epigenetics and brain memory function and dysfunction
Several enzymes have been found to modulate DNA or histone proteins for proper neuronal
signaling for learning and memory (Park et al., 2022). Epigenetic processes support long-
term memory formation (Feng et al., 2010; Korzus et al., 2004). Cognitive impairments can
be reversed by drugs acting on defective epigenetic components (Gräff et al., 2010; Koshibu
et al., 2011, 2009). Thus, it’s clear that epigenetic processes can modulate memory
performance. Epigenetic therapies could be potential therapeutic strategies for memory and
cognitive function disorders (Franklin and Mansuy, 2010; Gräff and Mansuy, 2009, 2008;
Urdinguio et al., 2009). For example, histone deacetylase inhibitors (histone-modifying
enzymes) could be beneficial in treating memory impairment, age-related cognitive decline,
Alzheimer’s disease, etc. (Peleg et al., 2010).
Epigenetics and brain development
As epigenetic modulations are the basis for cellular development, impetuous changes of the
nervous system during prenatal and postnatal development are influenced by environmental
conditions. Thus, innate genetic programming and sensory experiences maintain the
functional neuronal circuits and brain development. Environmental influence impacts natural
Journal Pre-proof
8
variability in the quality and quantity of interactions between mother-infant, which modulates
the infant’s response to living conditions later in life and can influence their response to stress
and aversive conditions (Table 2). Changes in these responses have been found to be
correlated to the development of anxiety and depression (Beery and Francis, 2011). In addition,
variability in maternal care stabilizes epigenetic modifications that remain beyond the period
of maternal care (Weaver et al., 2004). Severe chronic stress during early life alters a mother's
behavior in adulthood and ultimately influences children's behavior across generations.
Depression, impulsive behavior, and altered social skills are common in adults who
experienced separation from their mothers in childhood (Franklin et al., 2010; Weiss et al.,
2011). Significant brain epigenetic profile alteration (results in changes in the methylation
profile of stress-related genes) occurs in those who experience childhood abuse and commit
suicide later in life (McGowan et al., 2009). Experiences during adulthood also modulate
epigenome, even in the case of (monozygotic) twins (Fraga et al., 2005). These behavioral
changes have been found to be correlated with the alteration of epigenetic processes,
specifically in DNA methylation in various genes in the brains, which demonstrates that early
stress modulates epigenome in various cells and tissues that transmit on subsequent
generations. Epigenetic divergence, known as epigenetic drift, can happen with or without
environmental influence.
Epigenetic factors impacting brain and behavior.
1) Physical activity
Physical activity modulates brain plasticity and functions by releasing factors from
contracting muscles in children and adults into circulation (Biddle and Asare, 2011; Hillman et
al., 2008; Lees and Hopkins, 2013; Ma et al., 2017; Niederer et al., 2011; Rodriguez-Ayllon et al.,
Journal Pre-proof
9
2019; Schmidt-Kassow et al., 2014; Suwabe et al., 2017; van Praag, 2009). Regular aerobic exercise
reduces the DNA methylation of various genes (Barrès et al., 2012; King-Himmelreich et al.,
2016; Ling and Rönn, 2014; McGee et al., 2009). Studies suggested that brain capillaries regulate
positive effects on mental health and abilities in neurogenic niches that supply growth factors
(e.g., VEGF, GDF11, BDNF) to activate cellular survival pathways to induce gene
transcription responsible for neuroplasticity (Chen and Russo-Neustadt, 2009; Niederer et al.,
2011). For instance, Exercise influences BDNF chromatin regulation, DNA demethylation of
the BDNF promoter IV, and phosphorylation of MeCP2 to stimulate BDNF mRNA and
protein synthesis (Gomez-Pinilla et al., 2011). Different neurotransmitters (e.g. GABA,
glutamate, serotonin) are also secreted by neurons in the neurogenic niche (Niederer et al.,
2011). Studies found strong links among physical activity, brain health, and epigenetic
mechanisms that affect neurogenesis, brain plasticity, and function (Christiansen et al., 2016;
Fernandes et al., 2017; Horvath et al., 2015; Hunter et al., 2019; Lista and Sorrentino, 2010; Schenk
et al., 2019; van Praag, 2008; van Praag et al., 1999; Woelfel et al., 2018). It’s clear now that
inactivity is epigenetically deleterious.
2) Abusive substances
Despite adverse consequences, compulsive seeking and taking of abusive substances (e.g.
psychostimulants, opiates) is termed drug addiction (Koob and Volkow, 2016). Epigenetic
modulation plays a crucial role in the vulnerability of drug addiction. All abused or chronic
use of drugs act on mesolimbic dopamine circuitry in the fundamental cell type within the
nucleus accumbens as well as midbrain ventral tegmental area and innervation of medium
spiny neurons to induce long-lasting structural, electrophysiological, and transcriptional
changes via epigenetic maladaptations (Feng et al., 2014; Hyman et al., 2006; Kelley and Berridge,
Journal Pre-proof
10
2002). Hyperacetylation of histones (H3 and H4) in nucleus accumbens due to an imbalance
between histone acetyltransferases (e.g. cAMP response element binding protein-binding
protein) and histone deacetylases due to acute or chronic exposure to abusive drugs facilitates
rapid expression of associated genes at the specific locus to contribute to addiction (Barrett
and Wood, 2008; Botia et al., 2012; Ferguson et al., 2015, 2013; Kumar et al., 2005; Levine et al.,
2011; Malvaez et al., 2011; Pandey et al., 2008; Renthal et al., 2009, 2007; Schroeder et al., 2008;
Shen et al., 2008; Shogren-Knaak et al., 2006; Taniguchi et al., 2012). Further research demands to
know the consequences of gene-specific histone-post translational modifications within the
context of drug addiction. Alcohol addiction has been found to modulate epigenetically.
Excessive alcohol drinking not only affects individuals but also affects their offspring
throughout various stages of their development. Excessive alcohol exposure during
pregnancy leads to fetal alcohol spectrum disorder and induces various epigenetic changes
(Resendiz et al., 2013; Ungerer et al., 2012). Excessive alcohol exposure during pregnancy
induces DNA methylation at 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC)
during embryonic and brain development (Chen et al., 2013; Guo et al., 2011; Ito et al., 2011;
Kriaucionis and Heintz, 2009; Liu et al., 2009; Tahiliani et al., 2009; Zhou et al., 2011a). Alcoholism
also raises histone 3 acetylation globally and changes miRNA expression in neural stem cells
in a cell-type and stage-specific manner (Kim and Shukla, 2006; Miranda, 2012; Pal-Bhadra et al.,
2007; Shukla et al., 2007; Wang et al., 2009). Opium, an old analgesic medication, causes severe
effects on the offspring's nervous system by increased methylation at the OPRM1 promoter
region (Chorbov et al., 2011; Das et al., 2004). Prenatal exposure to methamphetamine,
another known abusive drug, led to oxidative stress in the embryonic brain and postnatal
neurodevelopment and cognitive and behavioral defects (Jeng et al., 2005; Kwiatkowski et
al., 2014). Prenatal methamphetamine exposure results in differentially methylated regions
Journal Pre-proof
11
in the hippocampal DNA of adolescent offspring and leads to abnormal behavior (Itzhak et
al., 2015). The hypermethylated and hypomethylated differential methylated regions are
enriched for “cerebral cortex GABAergic interneuron differentiation” and “embryonic
development.”
3) Cannabis
Chronic cannabinoid exposure maintains protracted effects. The epigenome provides the
cellular context for cannabinoid exposure to modulate the functionality of genes and related
behavior (Szutorisz et al., 2016; Szutorisz and Hurd, 2018, 2016). Epigenetics contribute to
regulating the endocannabinoid system, which is critical in controlling different synaptic
communication and plasticity in healthy brain and different neuropsychiatric disorders over
short and long period (Batool et al., 2019; Bayraktar and Kreutz, 2018; D’Addario et al., 2013;
Dambacher et al., 2013; Dillon, 2012; Meccariello et al., 2020; Weaver, 2014). Table 3
demonstrates the epigenetic mechanisms of developmental cannabis exposure. These studies
suggest that cannabis exposure during multiple stages of development modulates epigenetic
mechanisms to change neural and behavioral phenotypes. Cannabis acts through the germ
line to modulate synaptic development and behavior across generations (Szutorisz et al.,
2014; Watson et al., 2015).
4) Micronutrients:
Research focusing on dietary impact on gene expression via epigenetic mechanisms on brain
development and neuropsychiatric diseases/disorders is evolving (Canani et al., 2011; Levi and
Sanderson, 2004; Prado and Dewey, 2014; Roseboom et al., 2006). Studies found that
neuropsychiatric diseases during adulthood were linked with prenatal exposure to inadequate
Journal Pre-proof
12
nutrition (Roseboom et al., 2006; St Clair et al., 2005; Susser et al., 1996; Susser and Lin, 1992).
Early malnutrition induces lasting epigenetic changes in the brain, leading to behavioral
consequences and diseases/disorders in later life (Canani et al., 2011; Kundakovic and Jaric,
2017). For instance, studies found an association between maternal iron deficiency and the
risk of autism spectrum disorders among offspring due to epigenetic modulation (Insel et al.,
2008; Schmidt et al., 2014). The effects of folic acid on epigenetics through the methionine
pathway to generate methyl donors for DNA and histone methylation might support fetal
neural tube development (Akchiche et al., 2012; Berry et al., 1999; Guéant et al., 2013; “Use
of Folic Acid for Prevention of Spina Bifida and Other Neural Tube Defects1983-1991,”
1991). N-3 polyunsaturated fatty acids (PUFAs) are also known to control DNA methylation
state globally and via gene-specific methylation of promoter sequences during development
(Heberden and Maximin, 2019). Higher intake of n-3 PUFAs during pregnancy supports fetal
brain development (Basak and Duttaroy, 2022). Despite some studies on epigenetic changes in
neurodevelopmental-related genes (Kundakovic et al., 2013; Toledo-Rodriguez et al., 2010),
there is no established epigenetic mechanism for how the environment does have
confounding effects on neurodevelopment disorders. Therefore, more studies need to be done
to deepen the knowledge about the relationship between nutrition, epigenetics, and
neurodevelopment.
5) Caffeine
As an adenosine receptor blocker, caffeine is a widely used stimulant worldwide. Caffeine
accumulation aggravates stress response (Yeomans et al., 2007). Chronic caffeine ingestion
activates the maternal and placental reninangiotensin system (RAS) and induces p53-
dependent placental apoptosis, which leads to fetal intrauterine growth retardation (Huang et
Journal Pre-proof
13
al., 2012). Soellner and colleagues have demonstrated that chronic prenatal caffeine exposure
interrupts novel object recognition and radial arm maze behaviors in adult rats (Soellner et
al., 2009). Caffeine exposure during pregnancy inhibits the development and function of the
fetal hypothalamic-pituitary-adrenal axis-associated neuroendocrine metabolism (Liu et al.,
2012; Xia et al., 2014; D. Xu et al., 2012b, 2012a; Xu et al., 2011). Prenatal caffeine exposure
inhibits fetal adrenal steroidogenesis by blocking the enzymes (StAR/P450scc, 3β-HSD,
P450c21, and P450c11) due to altered epigenetic modifications (DNA methylation and
histone acetylation) of the promoter region for the transcriptional activator SF-1 (Yan et al.,
2014). However, further studies might clarify the role of epigenetic modification by caffeine.
6) Pollutants
Different types of pollution are increasing daily due to industrialization, which is contributing
to causing various diseases or disorders. For example, tobacco smoking is the most common
pollutant that modulates early neurobehavioral development. Prenatal smoking increases
children's risk of attention deficit hyperactivity disorder due to DAT1, DRD4, and CHRNA4
gene variations (Becker et al., 2008; Kahn et al., 2003; Todd and Neuman, 2007). The epigenetic
effects transmitted intergenerationally due to smoking predict family dysfunction and poor
health (Miles and Weden, 2012; Seeman et al., 2010; Taylor et al., 2006). However, the necessity
of a better understanding of underlying microprocesses doesn’t preclude policies, sanctions,
and universal public health campaigns against childhood exposure to tobacco smoke in
domestic settings.
Traffic-related air pollution is also a significant source of air pollution in urban areas,
particularly for particulate matters (PMs) [according to size, categorized as "coarse" (PM10),
"fine" (PM2.5) μm, and "ultrafine" (PM0.1), having an aerodynamic diameter less than 10 μm,
Journal Pre-proof
14
less than 2.5 μm and less than 0.1 μm, respectively], which is composed of gases like nitrogen
oxides (e.g., NO2, NOx) and sulfur dioxide (SO2) as well as black carbon, absorbed metals,
and polyaromatic hydrocarbons of various size fractions (Johnson et al., 2021; Rider and
Carlsten, 2019). Traffic-related air pollution modulates brain development and function
through DNA methylation (Rider and Carlsten, 2019). Studies found that prenatal exposure
to PM2.5 results in thinning of the cortex in many regions of the brain and impaired inhibitory
control, which is related to neurobehavioral dysfunctions such as addictive behavior and
attention deficit hyperactivity disorder due to altered DNA methylation, including global
hypomethylation, gene-specific changes in methylation process as well as downregulated
expresión of miR-21, miR-146a, and miR-222 (Johnson et al., 2021).
Lead toxicity is also quite common. Lead exposure is commonly caused by food, water,
tobacco smoke, air, dust, and soil. The fetus can be exposed via placental transfer (“Scientific
Opinion on Lead in Food,” 2010; World Health Organization, 2010). Surprisingly,
bioavailable lead is absorbed better in infants than adults and developmental neurotoxicity is
a significant health effect of lead exposure (“Scientific Opinion on Lead in Food,” 2010; Tarragó
and Brown, 2017). Lead can interrupt epigenetic modulation (Khalid and Abdollahi, 2019). In
newborns, prenatal lead exposure results in genomic DNA methylation (CLEC11A, DNHD1,
LINE1) (Pilsner et al., 2009; Wu et al., 2017). Epigenetic modulations, including DNA
methylation, influence BDNF expression across tissues, including the brain and blood
(Ikegame et al., 2013; Kundakovic et al., 2015; Stenz et al., 2015). Therefore, BDNF can be
used as a peripheral biomarker of psychiatric disorders (Kundakovic et al., 2015; Stenz et al.,
2015). However, further research might explain lead exposure-mediated psychiatric diseases.
Journal Pre-proof
15
Another known pollutant is bisphenol A. Due to industrialization, the endocrine-disrupting
chemical bisphenol A induces neurotoxicity through ingesting contaminated foods and drinks
or inhalation (Chianese et al., 2017). Bisphenol A interrupts androgenic activities via binding
with steroid receptors, e.g., estrogen receptor α, estrogen receptor β, estrogen-related
receptor γ, androgen receptor, GPER30, etc (Chianese et al., 2017; Murata and Kang, 2018;
Tavares et al., 2016; Vandenberg et al., 2013). Bisphenol A and its analogs change methylation
of CpG islands in the promoter regions of specific genes or the genome-wide methylation in
fetal and adult brain through DNA methyltransferases modulation, while is transmitted across
the generations (Doshi et al., 2011; Drobná et al., 2018; Wolstenholme et al., 2011; Yaoi et
al., 2008). Bisphenol A and its analogs modulate histone methylation and acetylation to affect
chromatin remodeling by NAD+-dependent deacetylase sirtuin 1 (Chen et al., 2017; Doherty
et al., 2010; Eichenlaub-Ritter and Pacchierotti, 2015; Viré et al., 2006). Even Bisphenol A
mediated post-transcriptional modification of other RNA species by non-coding RNAs (e.g.,
microRNA, long non-coding RNA, circRNA) affects brain physiology in health and disease
(Godlewski et al., 2019; Leighton and Bredy, 2018; Noack and Calegari, 2018; Sekar and Liang, 2019;
Shi et al., 2017). Bisphenol A-induced impaired hippocampal neurogenesis correlates with
upregulated DNA methylation of the CREB-regulated transcription coactivator 1 (Jang et al.,
2012). Bisphenol A also increases histone H3 acetylation in the cerebral cortex and
hippocampus to promote memory and cognitive dysfunction (Bale, 2015; Keverne, 2014; Kumar
and Thakur, 2017). Not only gestating mothers but also paternal exposure to bisphenol A
influences fetus development [as spermatozoa use non-coding RNAs to carry paternal
hereditary information] (Dobrzyńska et al., 2015; Guerrero-Bosagna et al., 2013; Kuruto-
Niwa et al., 2007; Mendonca et al., 2014). Bisphenol A exposure causes sex-specific, dose-
Journal Pre-proof
16
dependent (linear and curvilinear), and brain region-specific changes in the expression of
epigenetic regulators (DNMT1 and DNMT3A) as well as genes encoding estrogen receptors
and estrogen-related receptor-γ (Kundakovic et al., 2013). Bisphenol A increases DNA
methylation levels in the promoter region of the GRIN2B gene (Alavian-Ghavanini et al.,
2018). Bisphenol A has been shown to induce hypermethylation of the 5-prime end promoter
region of the BDNF gene in female offspring but enhances DNA methylation of the
transcriptional regulators of the glucocorticoid receptors FKBP5 was found within the
hippocampus of male rats to influence spatial learning and memory capabilities (Alavian-
Ghavanini et al., 2018; Cheong et al., 2018; Kitraki et al., 2015). Interestingly, bisphenol A
exposure in the fetal stage did not significantly affect hippocampal DNA methylation (Aiba
et al., 2018). Overall, bisphenol A induces behavior-related and sex-specific epigenetic
modifications predominantly targeting the expression pattern of sexually dimorphic genes.
However, further studies are needed to determine the exact dose range and exposure time
during development by which bisphenol A can induce epigenetic modifications.
7) Hypoxia
Maternal and fetal hypoxia in pregnancy disorders influences normal fetal development and
pathological processes (Pouyssegur and López-Barneo, 2016). Various reviews accredit
maternal-fetal hypoxia affects organogenesis and brain functions (Faa et al., 2016; Newby et
al., 2015; Schlotz and Phillips, 2009). Abnormal levels of fetal hypoxia provoke epigenetic
modulation that modifies target gene expression (Cerda and Weitzman, 1997; Luo et al., 2006).
However, more mechanistic studies are necessary to study hypoxia-mediated direct and
indirect effects on fetal development, gene expression, epigenetic changes in specific genes,
and consequences later in life.
Journal Pre-proof
17
Conclusions
This thorough review sheds light on the complex interplay between epigenetic modifications
and their substantial impact on brain functions. The critical impacts of the micronutrients,
physical activity, caffeine, toxins, and harmful substances in the brain's epigenetic landscape
are now evident. The intricate links between epigenetic changes and behavioral outcomes
emphasize the possibility of targeted therapies that could harness the power of epigenetic
control to improve cognitive function and attenuate the detrimental consequences of
substance addiction and environmental pollutants. Despite strong evidence of the different
roles of epigenetic alterations in gene expression and phenotypic outcomes, translating the
findings from animal studies to the health effects of environmental exposure to humans needs
to be improvised. To untangle the intricacies of epigenetic modifications and their long-term
impact, there is still a need for interdisciplinary collaboration, advanced technical
breakthroughs, and longitudinal investigations.
Declarations
Funding: No funding was available
Conflicts of interest/Competing interests: Authors express no conflicts of interest
Authors Contributions: Conceptualisation RM writingoriginal draft preparation, RM
review and editing, RM and A.D.R. Both authors have read and agreed to the published
version of the manuscript.
Ethics approval: Not applicable
Availability of data and material (data transparency): Not Applicable
References:
Aiba, T., Saito, T., Hayashi, A., Sato, S., Yunokawa, H., Maruyama, T., Fujibuchi, W., Ohsako, S.,
2018. Does the prenatal bisphenol A exposure alter DNA methylation levels in the mouse
hippocampus?: An analysis using a high-sensitivity methylome technique. Genes Environ 40,
12. https://doi.org/10.1186/s41021-018-0099-y
Journal Pre-proof
18
Akchiche, N., Bossenmeyer-Pourié, C., Kerek, R., Martin, N., Pourié, G., Koziel, V., Helle, D.,
Alberto, J.M., Ortiou, S., Camadro, J.M., Léger, T., Guéant, J.L., Daval, J.L., 2012.
Homocysteinylation of neuronal proteins contributes to folate deficiency-associated
alterations of differentiation, vesicular transport, and plasticity in hippocampal neuronal
cells. FASEB Journal 26. https://doi.org/10.1096/fj.12-205757
Alavian-Ghavanini, A., Lin, P.I., Lind, P.M., Risén Rimfors, S., Halin Lejonklou, M., Dunder, L., Tang,
M., Lindh, C., Bornehag, C.G., Rüegg, J., 2018. Prenatal Bisphenol A Exposure is Linked to
Epigenetic Changes in Glutamate Receptor Subunit Gene Grin2b in Female Rats and Humans.
Sci Rep 8. https://doi.org/10.1038/s41598-018-29732-9
Bai, M., Zhu, X., Zhang, Y., Zhang, S., Zhang, L., Xue, L., Yi, J., Yao, S., Zhang, X., 2012. Abnormal
Hippocampal BDNF and miR-16 Expression Is Associated with Depression-Like Behaviors
Induced by Stress during Early Life. PLoS One 7.
https://doi.org/10.1371/journal.pone.0046921
Bale, T.L., 2015. Epigenetic and transgenerational reprogramming of brain development. Nat Rev
Neurosci. https://doi.org/10.1038/nrn3818
Balmer, N. v., Weng, M.K., Zimmer, B., Ivanova, V.N., Chambers, S.M., Nikolaeva, E., Jagtap, S.,
Sachinidis, A., Hescheler, J., Waldmann, T., Leist, M., 2012. Epigenetic changes and disturbed
neural development in a human embryonic stem cell-based model relating to the fetal
valproate syndrome. Hum Mol Genet 21. https://doi.org/10.1093/hmg/dds239
Barrès, R., Yan, J., Egan, B., Treebak, J.T., Rasmussen, M., Fritz, T., Caidahl, K., Krook, A., O’Gorman,
D.J., Zierath, J.R., 2012. Acute exercise remodels promoter methylation in human skeletal
muscle. Cell Metab 15. https://doi.org/10.1016/j.cmet.2012.01.001
Barrett, R.M., Wood, M.A., 2008. Beyond transcription factors: The role of chromatin modifying
enzymes in regulating transcription required for memory. Learning and Memory.
https://doi.org/10.1101/lm.917508
Basak, S., Duttaroy, A.K., 2022. Maternal PUFAs, Placental Epigenetics, and Their Relevance to
Fetal Growth and Brain Development. Reproductive Sciences.
https://doi.org/10.1007/s43032-022-00989-w
Batool, S., Raza, H., Zaidi, J., Riaz, S., Hasan, S., Syed, N.I., 2019. Synapse formation: From cellular
and molecular mechanisms to neurodevelopmental and neurodegenerative disorders. J
Neurophysiol 121. https://doi.org/10.1152/jn.00833.2018
Bayraktar, G., Kreutz, M.R., 2018. Neuronal DNA Methyltransferases: Epigenetic Mediators
between Synaptic Activity and Gene Expression? Neuroscientist.
https://doi.org/10.1177/1073858417707457
Becker, K., El-Faddagh, M., Schmidt, M.H., Esser, G., Laucht, M., 2008. Interaction of dopamine
transporter genotype with prenatal smoke exposure on ADHD symptoms. J Pediatr 152, 263
9. https://doi.org/10.1016/j.jpeds.2007.07.004
Journal Pre-proof
19
Beery, A.K., Francis, D.D., 2011. Adaptive significance of natural variations in maternal care in rats:
A translational perspective. Neurosci Biobehav Rev.
https://doi.org/10.1016/j.neubiorev.2011.03.012
Berry, R.J., Li, Z., Erickson, J.D., Li, S., Moore, C.A., Wang, H., Mulinare, J., Zhao, P., Wong, L.-Y.C.,
Gindler, J., Hong, S., Correa, A., 1999. Prevention of neural-tube defects with folic acid in
China. China-U.S. Collaborative Project for Neural Tube Defect Prevention. English Journal
341.
Biddle, S.J.H., Asare, M., 2011. Physical activity and mental health in children and adolescents: A
review of reviews. Br J Sports Med. https://doi.org/10.1136/bjsports-2011-090185
Bihaqi, S.W., Huang, H., Wu, J., Zawia, N.H., 2011. Infant exposure to lead (Pb) and epigenetic
modifications in the aging primate brain: Implications for Alzheimer’s disease. Journal of
Alzheimer’s Disease 27. https://doi.org/10.3233/JAD-2011-111013
Booij, L., Szyf, M., Carballedo, A., Frey, E.M., Morris, D., Dymov, S., Vaisheva, F., Ly, V., Fahey, C.,
Meaney, J., Gill, M., Frodl, T., 2015. DNA methylation of the serotonin transporter gene in
peripheral cells and stress-related changes in hippocampal volume: A study in depressed
patients and healthy controls. PLoS One 10. https://doi.org/10.1371/journal.pone.0119061
Bose, R., Onishchenko, N., Edoff, K., Lang, A.M.J., Ceccatelli, S., 2012. Inherited effects of low-dose
exposure to methylmercury in neural stem cells. Toxicological Sciences 130.
https://doi.org/10.1093/toxsci/kfs257
Botia, B., Legastelois, R., Alaux-Cantin, S., Naassila, M., 2012. Expression of Ethanol-Induced
Behavioral Sensitization Is Associated with Alteration of Chromatin Remodeling in Mice. PLoS
One 7. https://doi.org/10.1371/journal.pone.0047527
Breton, C. v., Byun, H.M., Wenten, M., Pan, F., Yang, A., Gilliland, F.D., 2009. Prenatal tobacco
smoke exposure affects global and gene-specific DNA methylation. Am J Respir Crit Care Med
180. https://doi.org/10.1164/rccm.200901-0135OC
Bure, I. V., Nemtsova, M. V., Kuznetsova, E.B., 2022. Histone Modifications and Non-Coding RNAs:
Mutual Epigenetic Regulation and Role in Pathogenesis. Int J Mol Sci 23, 5801.
https://doi.org/10.3390/ijms23105801
Buscariollo, D.L., Fang, X., Greenwood, V., Xue, H., Rivkees, S.A., Wendler, C.C., 2014. Embryonic
caffeine exposure acts via A1 adenosine receptors to alter adult cardiac function and DNA
methylation in mice. PLoS One 9. https://doi.org/10.1371/journal.pone.0087547
Canani, R.B., di Costanzo, M., Leone, L., Bedogni, G., Brambilla, P., Cianfarani, S., Nobili, V.,
Pietrobelli, A., Agostoni, C., 2011. Epigenetic mechanisms elicited by nutrition in early life.
Nutr Res Rev. https://doi.org/10.1017/S0954422411000102
Castillo, P., Ibáñez, F., Guajardo, A., Llanos, M.N., Ronco, A.M., 2012. Impact of Cadmium Exposure
during Pregnancy on Hepatic Glucocorticoid Receptor Methylation and Expression in Rat
Fetus. PLoS One 7. https://doi.org/10.1371/journal.pone.0044139
Journal Pre-proof
20
Cerda, S., Weitzman, S.A., 1997. Influence of oxygen radical injury on DNA methylation. Mutat Res
Rev Mutat Res. https://doi.org/10.1016/S1383-5742(96)00050-6
Champagne, F.A., Curley, J.P., 2009. Epigenetic mechanisms mediating the long-term effects of
maternal care on development. Neurosci Biobehav Rev.
https://doi.org/10.1016/j.neubiorev.2007.10.009
Chen, M.J., Russo-Neustadt, A.A., 2009. Running exercise-induced up-regulation of hippocampal
brain-derived neurotrophic factor is CREB-dependent. Hippocampus 19.
https://doi.org/10.1002/hipo.20579
Chen, Y., Ozturk, N.C., Zhou, F.C., 2013. DNA Methylation Program in Developing Hippocampus
and Its Alteration by Alcohol. PLoS One 8. https://doi.org/10.1371/journal.pone.0060503
Chen, Z., Zuo, X., He, D., Ding, S., Xu, F., Yang, H., Jin, X., Fan, Y., Ying, L., Tian, C., Ying, C., 2017.
Long-term exposure to a a€safe’ dose of bisphenol A reduced protein acetylation in adult rat
testes. Sci Rep 7. https://doi.org/10.1038/srep40337
Cheong, A., Johnson, S.A., Howald, E.C., Ellersieck, M.R., Camacho, L., Lewis, S.M., Vanlandingham,
M.M., Ying, J., Ho, S.M., Rosenfeld, C.S., 2018. Gene expression and DNA methylation
changes in the hypothalamus and hippocampus of adult rats developmentally exposed to
bisphenol A or ethinyl estradiol: a CLARITY-BPA consortium study. Epigenetics 13.
https://doi.org/10.1080/15592294.2018.1497388
Chianese, R., Troisi, J., Richards, S., Scafuro, M., Fasano, S., Guida, M., Pierantoni, R., Meccariello,
R., 2017. Bisphenol A in Reproduction: Epigenetic Effects. Curr Med Chem 25.
https://doi.org/10.2174/0929867324666171009121001
Choi, S., Han, K.M., Won, E., Yoon, B.J., Lee, M.S., Ham, B.J., 2015. Association of brain-derived
neurotrophic factor DNA methylation and reduced white matter integrity in the anterior
corona radiata in major depression. J Affect Disord 172.
https://doi.org/10.1016/j.jad.2014.09.042
Chorbov, V.M., Todorov, A.A., Lynskey, M.T., Cicero, T.J., 2011. Elevated levels of DNA methylation
at the OPRM1 promoter in blood and sperm from male opioid addicts. J Opioid Manag 7.
https://doi.org/10.5055/jom.2011.0067
Christiansen, L., Lenart, A., Tan, Q., Vaupel, J.W., Aviv, A., Mcgue, M., Christensen, K., 2016. DNA
methylation age is associated with mortality in a longitudinal Danish twin study. Aging Cell
15. https://doi.org/10.1111/acel.12421
Cronican, A.A., Fitz, N.F., Carter, A., Saleem, M., Shiva, S., Barchowsky, A., Koldamova, R., Schug, J.,
Lefterov, I., 2013. Genome-Wide Alteration of Histone H3K9 Acetylation Pattern in Mouse
Offspring Prenatally Exposed to Arsenic. PLoS One 8.
https://doi.org/10.1371/journal.pone.0053478
Cuccurazzu, B., Zamberletti, E., Nazzaro, C., Prini, P., Trusel, M., Grilli, M., Parolaro, D., Tonini, R.,
Rubino, T., 2018. Adult Cellular Neuroadaptations Induced by Adolescent THC Exposure in
Journal Pre-proof
21
Female Rats Are Rescued by Enhancing Anandamide Signaling. International Journal of
Neuropsychopharmacology 21. https://doi.org/10.1093/ijnp/pyy057
D’Addario, C., di Francesco, A., Pucci, M., Finazzi Agrò, A., MacCarrone, M., 2013. Epigenetic
mechanisms and endocannabinoid signalling. FEBS Journal.
https://doi.org/10.1111/febs.12125
Dalton, V.S., Kolshus, E., McLoughlin, D.M., 2014. Epigenetics and depression: Return of the
repressed. J Affect Disord. https://doi.org/10.1016/j.jad.2013.10.028
Dambacher, S., de Almeida, G.P., Schotta, G., 2013. Dynamic changes of the epigenetic landscape
during cellular differentiation. Epigenomics. https://doi.org/10.2217/epi.13.67
Dannlowski, U., Kugel, H., Redlich, R., Halik, A., Schneider, I., Opel, N., Grotegerd, D., Schwarte, K.,
Schettler, C., Ambrée, O., Rust, S., Domschke, K., Arolt, V., Heindel, W., Baune, B.T., Suslow,
T., Zhang, W., Hohoff, C., 2014. Serotonin transporter gene methylation is associated with
hippocampal gray matter volume. Hum Brain Mapp 35. https://doi.org/10.1002/hbm.22555
Darnaudéry, M., Maccari, S., 2008. Epigenetic programming of the stress response in male and
female rats by prenatal restraint stress. Brain Res Rev.
https://doi.org/10.1016/j.brainresrev.2007.11.004
Das, A., Poole, W.K., Bada, H.S., 2004. A repeated measures approach for simultaneous modeling
of multiple neurobehavioral outcomes in newborns exposed to cocaine in utero. Am J
Epidemiol 159. https://doi.org/10.1093/aje/kwh114
de Mendoza, A., Poppe, D., Buckberry, S., Pflueger, J., Albertin, C.B., Daish, T., Bertrand, S., de la
Calle-Mustienes, E., Gómez-Skarmeta, J.L., Nery, J.R., Ecker, J.R., Baer, B., Ragsdale, C.W.,
Grützner, F., Escriva, H., Venkatesh, B., Bogdanovic, O., Lister, R., 2021. The emergence of the
brain non-CpG methylation system in vertebrates. Nat Ecol Evol 5.
https://doi.org/10.1038/s41559-020-01371-2
Dias, B.G., Maddox, S.A., Klengel, T., Ressler, K.J., 2015. Epigenetic mechanisms underlying learning
and the inheritance of learned behaviors. Trends Neurosci.
https://doi.org/10.1016/j.tins.2014.12.003
Dillon, N., 2012. Factor mediated gene priming in pluripotent stem cells sets the stage for lineage
specification. BioEssays. https://doi.org/10.1002/bies.201100137
Dinieri, J.A., Wang, X., Szutorisz, H., Spano, S.M., Kaur, J., Casaccia, P., Dow-Edwards, D., Hurd, Y.L.,
2011. Maternal cannabis use alters ventral striatal dopamine D2 gene regulation in the
offspring. Biol Psychiatry 70. https://doi.org/10.1016/j.biopsych.2011.06.027
Dobrzyńska, M.M., Gajowik, A., Radzikowska, J., Tyrkiel, E.J., Jankowska-Steifer, E.A., 2015. Male-
mediated F1 effects in mice exposed to bisphenol A, either alone or in combination with X-
irradiation. Mutat Res Genet Toxicol Environ Mutagen 789790.
https://doi.org/10.1016/j.mrgentox.2015.06.015
Doherty, L.F., Bromer, J.G., Zhou, Y., Aldad, T.S., Taylor, H.S., 2010. In utero exposure to
diethylstilbestrol (DES) or bisphenol-A (BPA) increases EZH2 expression in the mammary
Journal Pre-proof
22
gland: An epigenetic mechanism linking endocrine disruptors to breast cancer. Horm Cancer
1. https://doi.org/10.1007/s12672-010-0015-9
Doshi, T., Mehta, S.S., Dighe, V., Balasinor, N., Vanage, G., 2011. Hypermethylation of estrogen
receptor promoter region in adult testis of rats exposed neonatally to bisphenol A.
Toxicology 289. https://doi.org/10.1016/j.tox.2011.07.011
Drobná, Z., Henriksen, A.D., Wolstenholme, J.T., Montiel, C., Lambeth, P.S., Shang, S., Harris, E.P.,
Zhou, C., Flaws, J.A., Adli, M., Rissman, E.F., 2018. Transgenerational effects of bisphenol a on
gene expression and DNA methylation of imprinted genes in brain. Endocrinology 159.
https://doi.org/10.1210/en.2017-00730
Eichenlaub-Ritter, U., Pacchierotti, F., 2015. Bisphenol a effects on mammalian oogenesis and
epigenetic integrity of oocytes: A case study exploring risks of endocrine disrupting
chemicals. Biomed Res Int. https://doi.org/10.1155/2015/698795
Faa, G., Manchia, M., Pintus, R., Gerosa, C., Marcialis, M.A., Fanos, V., 2016. Fetal programming of
neuropsychiatric disorders. Birth Defects Res C Embryo Today.
https://doi.org/10.1002/bdrc.21139
Feng, J., Wilkinson, M., Liu, X., Purushothaman, I., Ferguson, D., Vialou, V., Maze, I., Shao, N.,
Kennedy, P., Koo, J.W., Dias, C., Laitman, B., Stockman, V., LaPlant, Q., Cahill, M.E., Nestler,
E.J., Shen, L., 2014. Chronic cocaine-regulated epigenomic changes in mouse nucleus
accumbens. Genome Biol 15. https://doi.org/10.1186/gb-2014-15-4-r65
Feng, J., Zhou, Y., Campbell, S.L., Le, T., Li, E., Sweatt, J.D., Silva, A.J., Fan, G., 2010. Dnmt1 and
Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain
neurons. Nat Neurosci 13. https://doi.org/10.1038/nn.2514
Ferguson, D., Koo, J.W., Feng, J., Heller, E., Rabkin, J., Heshmati, M., Renthal, W., Neve, R., Liu, X.,
Shao, N., Sartorelli, V., Shen, L., Nestler, E.J., 2013. Essential role of SIRT1 signaling in the
nucleus accumbens in cocaine and morphine action. Journal of Neuroscience 33.
https://doi.org/10.1523/JNEUROSCI.1284-13.2013
Ferguson, D., Shao, N., Heller, E., Feng, J., Neve, R., Kim, H.D., Call, T., Magazu, S., Shen, L., Nestler,
E.J., 2015. SIRT1-FOXO3a regulate cocaine actions in the nucleus accumbens. Journal of
Neuroscience 35. https://doi.org/10.1523/JNEUROSCI.4012-14.2015
Fernandes, J., Arida, R.M., Gomez-Pinilla, F., 2017. Physical exercise as an epigenetic modulator of
brain plasticity and cognition. Neurosci Biobehav Rev.
https://doi.org/10.1016/j.neubiorev.2017.06.012
Fraga, M.F., Ballestar, E., Paz, M.F., Ropero, S., Setien, F., Ballestar, M.L., Heine-Suñer, D.,
Cigudosa, J.C., Urioste, M., Benitez, J., Boix-Chornet, M., Sanchez-Aguilera, A., Ling, C.,
Carlsson, E., Poulsen, P., Vaag, A., Stephan, Z., Spector, T.D., Wu, Y.Z., Plass, C., Esteller, M.,
2005. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad
Sci U S A 102. https://doi.org/10.1073/pnas.0500398102
Journal Pre-proof
23
Franklin, T.B., Mansuy, I.M., 2010. The prevalence of epigenetic mechanisms in the regulation of
cognitive functions and behaviour. Curr Opin Neurobiol.
https://doi.org/10.1016/j.conb.2010.04.007
Franklin, T.B., Russig, H., Weiss, I.C., Grff, J., Linder, N., Michalon, A., Vizi, S., Mansuy, I.M., 2010.
Epigenetic transmission of the impact of early stress across generations. Biol Psychiatry 68.
https://doi.org/10.1016/j.biopsych.2010.05.036
Frodl, T., Szyf, M., Carballedo, A., Ly, V., Dymov, S., Vaisheva, F., Morris, D., Fahey, C., Meaney, J.,
Gill, M., Booij, L., 2015. DNA methylation of the serotonin transporter gene (SLC6A4) is
associated with brain function involved in processing emotional stimuli. Journal of Psychiatry
and Neuroscience 40. https://doi.org/10.1503/jpn.140180
Galkin, F., Kovalchuk, O., Koldasbayeva, D., Zhavoronkov, A., Bischof, E., 2023. Stress, diet,
exercise: Common environmental factors and their impact on epigenetic age. Ageing Res Rev.
https://doi.org/10.1016/j.arr.2023.101956
Garro, A.J., McBeth, D.L., Lima, V., Lieber, C.S., 1991. Ethanol Consumption Inhibits Fetal DNA
Methylation in Mice: Implications for the Fetal Alcohol Syndrome. Alcohol Clin Exp Res 15.
https://doi.org/10.1111/j.1530-0277.1991.tb00536.x
Godlewski, J., Lenart, J., Salinska, E., 2019. MicroRNA in brain pathology: Neurodegeneration the
other side of the brain cancer. Noncoding RNA. https://doi.org/10.3390/ncrna5010020
Gomez-Pinilla, F., Zhuang, Y., Feng, J., Ying, Z., Fan, G., 2011. Exercise impacts brain-derived
neurotrophic factor plasticity by engaging mechanisms of epigenetic regulation. European
Journal of Neuroscience 33. https://doi.org/10.1111/j.1460-9568.2010.07508.x
Gräff, J., Koshibu, K., Jouvenceau, A., Dutar, P., Mansuy, I.M., 2010. Protein phosphatase 1-
dependent transcriptional programs for long-term memory and plasticity. Learning and
Memory 17. https://doi.org/10.1101/lm.1766510
Gräff, J., Mansuy, I.M., 2009. Epigenetic dysregulation in cognitive disorders. European Journal of
Neuroscience. https://doi.org/10.1111/j.1460-9568.2009.06787.x
Gräff, J., Mansuy, I.M., 2008. Epigenetic codes in cognition and behaviour. Behavioural Brain
Research. https://doi.org/10.1016/j.bbr.2008.01.021
Guéant, J.L., Namour, F., Guéant-Rodriguez, R.M., Daval, J.L., 2013. Folate and fetal programming:
A play in epigenomics? Trends in Endocrinology and Metabolism.
https://doi.org/10.1016/j.tem.2013.01.010
Guerrero-Bosagna, C., Savenkova, M., Haque, M.M., Nilsson, E., Skinner, M.K., 2013.
Environmentally Induced Epigenetic Transgenerational Inheritance of Altered Sertoli Cell
Transcriptome and Epigenome: Molecular Etiology of Male Infertility. PLoS One 8.
https://doi.org/10.1371/journal.pone.0059922
Guo, J.U., Su, Y., Zhong, C., Ming, G.L., Song, H., 2011. Hydroxylation of 5-methylcytosine by TET1
promotes active DNA demethylation in the adult brain. Cell 145.
https://doi.org/10.1016/j.cell.2011.03.022
Journal Pre-proof
24
Han, M., Jia, L., Lv, W., Wang, L., Cui, W., 2019. Epigenetic enzyme mutations: Role in
tumorigenesis and molecular inhibitors. Front Oncol.
https://doi.org/10.3389/fonc.2019.00194
Heberden, C., Maximin, E., 2019. Epigenetic effects of N-3 polyunsaturated fatty acids, in:
Handbook of Nutrition, Diet, and Epigenetics. https://doi.org/10.1007/978-3-319-55530-
0_45
Héberlé, É., Bardet, A.F., 2019. Sensitivity of transcription factors to DNA methylation. Essays
Biochem. https://doi.org/10.1042/EBC20190033
Heim, C., Binder, E.B., 2012. Current research trends in early life stress and depression: Review of
human studies on sensitive periods, gene-environment interactions, and epigenetics. Exp
Neurol. https://doi.org/10.1016/j.expneurol.2011.10.032
Hervouet, E., Peixoto, P., Delage-Mourroux, R., Boyer-Guittaut, M., Cartron, P.F., 2018. Specific or
not specific recruitment of DNMTs for DNA methylation, an epigenetic dilemma. Clin
Epigenetics. https://doi.org/10.1186/s13148-018-0450-y
Hillman, C.H., Erickson, K.I., Kramer, A.F., 2008. Be smart, exercise your heart: Exercise effects on
brain and cognition. Nat Rev Neurosci. https://doi.org/10.1038/nrn2298
Hollins, S.L., Zavitsanou, K., Walker, F.R., Cairns, M.J., 2014. Alteration of imprinted Dlk1-Dio3
miRNA cluster expression in the entorhinal cortex induced by maternal immune activation
and adolescent cannabinoid exposure. Transl Psychiatry 4.
https://doi.org/10.1038/tp.2014.99
Horvath, S., Pirazzini, C., Bacalini, M.G., Gentilini, D., di Blasio, A.M., Delledonne, M., Mari, D.,
Arosio, B., Monti, D., Passarino, G., de Rango, F., D’Aquila, P., Giuliani, C., Marasco, E.,
Collino, S., Descombes, P., Garagnani, P., Franceschi, C., 2015. Decreased epigenetic age of
PBMCs from Italian semi-supercentenarians and their offspring. Aging 7.
https://doi.org/10.18632/aging.100861
Huang, J., Zhou, S., Ping, J., Pan, X., Liang, G., Xu, D., Kou, H., Bao, C., Wang, H., 2012. Role of p53-
dependent placental apoptosis in the reproductive and developmental toxicities of caffeine
in rodents. Clin Exp Pharmacol Physiol 39. https://doi.org/10.1111/j.1440-1681.2012.05676.x
Hughes, A.L., Kelley, J.R., Klose, R.J., 2020. Understanding the interplay between CpG island-
associated gene promoters and H3K4 methylation. Biochim Biophys Acta Gene Regul Mech.
https://doi.org/10.1016/j.bbagrm.2020.194567
Hunter, D.J., James, L., Hussey, B., Wadley, A.J., Lindley, M.R., Mastana, S.S., 2019. Impact of
aerobic exercise and fatty acid supplementation on global and gene-specific DNA
methylation. Epigenetics 14. https://doi.org/10.1080/15592294.2019.1582276
Hyman, S.E., Malenka, R.C., Nestler, E.J., 2006. Neural mechanisms of addiction: The role of
reward-related learning and memory. Annu Rev Neurosci.
https://doi.org/10.1146/annurev.neuro.29.051605.113009
Journal Pre-proof
25
Ibn Lahmar Andaloussi, Z., Taghzouti, K., Abboussi, O., 2019. Behavioural and epigenetic effects of
paternal exposure to cannabinoids during adolescence on offspring vulnerability to stress.
International Journal of Developmental Neuroscience 72.
https://doi.org/10.1016/j.ijdevneu.2018.11.007
Ikegame, T., Bundo, M., Murata, Y., Kasai, K., Kato, T., Iwamoto, K., 2013. DNA methylation of the
BDNF gene and its relevance to psychiatric disorders. J Hum Genet.
https://doi.org/10.1038/jhg.2013.65
Insel, B.J., Schaefer, C.A., McKeague, I.W., Susser, E.S., Brown, A.S., 2008. Maternal iron deficiency
and the risk of schizophrenia in offspring. Arch Gen Psychiatry 65.
https://doi.org/10.1001/archpsyc.65.10.1136
Intarasunanont, P., Navasumrit, P., Waraprasit, S., Chaisatra, K., Suk, W.A., Mahidol, C.,
Ruchirawat, M., 2012. Effects of arsenic exposure on DNA methylation in cord blood samples
from newborn babies and in a human lymphoblast cell line. Environ Health 11.
https://doi.org/10.1186/1476-069X-11-31
Ito, S., Shen, L., Dai, Q., Wu, S.C., Collins, L.B., Swenberg, J.A., He, C., Zhang, Y., 2011. Tet proteins
can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science (1979)
333. https://doi.org/10.1126/science.1210597
Itzhak, Y., Ergui, I., Young, J.I., 2015. Long-term parental methamphetamine exposure of mice
influences behavior and hippocampal DNA methylation of the offspring. Mol Psychiatry 20.
https://doi.org/10.1038/mp.2014.7
Ivanova, E., Canovas, S., Garcia-Martínez, S., Romar, R., Lopes, J.S., Rizos, D., Sanchez-Calabuig,
M.J., Krueger, F., Andrews, S., Perez-Sanz, F., Kelsey, G., Coy, P., 2020. DNA methylation
changes during preimplantation development reveal inter-species differences and
reprogramming events at imprinted genes. Clin Epigenetics 12.
https://doi.org/10.1186/s13148-020-00857-x
Jack, A., Connelly, J.J., Morris, J.P., 2012. DNA methylation of the oxytocin receptor gene predicts
neural response to ambiguous social stimuli. Front Hum Neurosci.
https://doi.org/10.3389/fnhum.2012.00280
Jang, Y.J., Park, H.R., Kim, T.H., Yang, W.J., Lee, J.J., Choi, S.Y., Oh, S.B., Lee, E., Park, J.H., Kim, H.P.,
Kim, H.S., Lee, J., 2012. High dose bisphenol A impairs hippocampal neurogenesis in female
mice across generations. Toxicology 296. https://doi.org/10.1016/j.tox.2012.03.007
Jeng, W., Wong, A.W., Ting-A-Kee, R., Wells, P.G., 2005. Methamphetamine-enhanced embryonic
oxidative DNA damage and neurodevelopmental deficits, in: Free Radical Biology and
Medicine. https://doi.org/10.1016/j.freeradbiomed.2005.03.015
Johnson, N.M., Hoffmann, A.R., Behlen, J.C., Lau, C., Pendleton, D., Harvey, N., Shore, R., Li, Y.,
Chen, J., Tian, Y., Zhang, R., 2021. Air pollution and children’s health—a review of adverse
effects associated with prenatal exposure from fine to ultrafine particulate matter. Environ
Health Prev Med. https://doi.org/10.1186/s12199-021-00995-5
Journal Pre-proof
26
Kahn, R.S., Khoury, J., Nichols, W.C., Lanphear, B.P., 2003. Role of dopamine transporter genotype
and maternal prenatal smoking in childhood hyperactive-impulsive, inattentive, and
oppositional behaviors. Journal of Pediatrics 143. https://doi.org/10.1016/S0022-
3476(03)00208-7
Kelley, A.E., Berridge, K.C., 2002. The Neuroscience of Natural Rewards: Relevance to Addictive
Drugs. Journal of Neuroscience. https://doi.org/10.1523/jneurosci.22-09-03306.2002
Kerek, R., Geoffroy, A., Bison, A., Martin, N., Akchiche, N., Pourié, G., Helle, D., Guéant, J.L.,
Bossenmeyer-Pourié, C., Daval, J.L., 2013. Early methyl donor deficiency may induce
persistent brain defects by reducing Stat3 signaling targeted by miR-124. Cell Death Dis 4.
https://doi.org/10.1038/cddis.2013.278
Keverne, E.B., 2014. Significance of epigenetics for understanding brain development, brain
evolution and behaviour. Neuroscience. https://doi.org/10.1016/j.neuroscience.2012.11.030
Khalid, M., Abdollahi, M., 2019. Epigenetic modifications associated with pathophysiological
effects of lead exposure. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 37.
https://doi.org/10.1080/10590501.2019.1640581
Kile, M.L., Baccarelli, A., Hoffman, E., Tarantini, L., Quamruzzaman, Q., Rahman, M., Mahiuddin, G.,
Mostofa, G., Hsueh, Y.M., Wright, R.O., Christiani, D.C., 2012. Prenatal arsenic exposure and
DNA methylation in maternal and umbilical cord blood leukocytes. Environ Health Perspect
120. https://doi.org/10.1289/ehp.1104173
Kim, J.S., Shukla, S.D., 2006. Acute in vivo effect of ethanol (binge drinking) on histone H3
modifications in rat tissues. Alcohol and Alcoholism 41.
https://doi.org/10.1093/alcalc/agh248
Kim, K.C., Kim, P., Go, H.S., Choi, C.S., Yang, S. il, Cheong, J.H., Shin, C.Y., Ko, K.H., 2011. The critical
period of valproate exposure to induce autistic symptoms in Sprague-Dawley rats. Toxicol
Lett 201. https://doi.org/10.1016/j.toxlet.2010.12.018
King-Himmelreich, T.S., Schramm, S., Wolters, M.C., Schmetzer, J., Möser, C. v., Knothe, C., Resch,
E., Peil, J., Geisslinger, G., Niederberger, E., 2016. The impact of endurance exercise on global
and AMPK gene-specific DNA methylation. Biochem Biophys Res Commun 474.
https://doi.org/10.1016/j.bbrc.2016.04.078
Kippler, M., Engström, K., Jurkovic Mlakar, S., Bottai, M., Ahmed, S., Hossain, M.B., Raqib, R.,
Vahter, M., Broberg, K., 2013. Sex-specific effects of early life cadmium exposure on DNA
methylation and implications for birth weight. Epigenetics 8.
https://doi.org/10.4161/epi.24401
Kitraki, E., Nalvarte, I., Alavian-Ghavanini, A., Rüegg, J., 2015. Developmental exposure to
bisphenol A alters expression and DNA methylation of Fkbp5, an important regulator of the
stress response. Mol Cell Endocrinol 417. https://doi.org/10.1016/j.mce.2015.09.028
Journal Pre-proof
27
Klein, C., Kemmel, V., Taleb, O., Aunis, D., Maitre, M., 2009. Pharmacological doses of gamma-
hydroxybutyrate (GHB) potentiate histone acetylation in the rat brain by histone deacetylase
inhibition. Neuropharmacology 57. https://doi.org/10.1016/j.neuropharm.2009.04.013
Klengel, T., Mehta, D., Anacker, C., Rex-Haffner, M., Pruessner, J.C., Pariante, C.M., Pace, T.W.W.,
Mercer, K.B., Mayberg, H.S., Bradley, B., Nemeroff, C.B., Holsboer, F., Heim, C.M., Ressler,
K.J., Rein, T., Binder, E.B., 2013. Allele-specific FKBP5 DNA demethylation mediates gene-
childhood trauma interactions. Nat Neurosci 16. https://doi.org/10.1038/nn.3275
Koob, G.F., Volkow, N.D., 2016. Neurobiology of addiction: a neurocircuitry analysis. Lancet
Psychiatry. https://doi.org/10.1016/S2215-0366(16)00104-8
Korzus, E., Rosenfeld, M.G., Mayford, M., 2004. CBP histone acetyltransferase activity is a critical
component of memory consolidation. Neuron 42.
https://doi.org/10.1016/j.neuron.2004.06.002
Koshibu, K., Gräff, J., Beullens, M., Heitz, F.D., Berchtold, D., Russig, H., Farinelli, M., Bollen, M.,
Mansuy, I.M., 2009. Protein phosphatase 1 regulates the histone code for long-term
memory. Journal of Neuroscience 29. https://doi.org/10.1523/JNEUROSCI.3610-09.2009
Koshibu, K., Gräff, J., Mansuy, I.M., 2011. Nuclear protein phosphatase-1: An epigenetic regulator
of fear memory and amygdala long-term potentiation. Neuroscience 173.
https://doi.org/10.1016/j.neuroscience.2010.11.023
Kriaucionis, S., Heintz, N., 2009. The nuclear DNA base 5-hydroxymethylcytosine is present in
purkinje neurons and the brain. Science (1979) 324.
https://doi.org/10.1126/science.1169786
Kumar, A., Choi, K.H., Renthal, W., Tsankova, N.M., Theobald, D.E.H., Truong, H.T., Russo, S.J.,
LaPlant, Q., Sasaki, T.S., Whistler, K.N., Neve, R.L., Self, D.W., Nestler, E.J., 2005. Chromatin
remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron 48.
https://doi.org/10.1016/j.neuron.2005.09.023
Kumar, D., Thakur, M.K., 2017. Effect of perinatal exposure to bisphenol-A on DNA methylation
and histone acetylation in cerebral cortex and hippocampus of postnatal male mice. Journal
of Toxicological Sciences 42. https://doi.org/10.2131/jts.42.281
Kundakovic, M., Gudsnuk, K., Franks, B., Madrid, J., Miller, R.L., Perera, F.P., Champagne, F.A.,
2013. Sex-specific epigenetic disruption and behavioral changes following low-dose in utero
bisphenol a exposure. Proc Natl Acad Sci U S A 110.
https://doi.org/10.1073/pnas.1214056110
Kundakovic, M., Gudsnuk, K., Herbstman, J.B., Tang, D., Perera, F.P., Champagne, F.A., 2015. DNA
methylation of BDNF as a biomarker of early-life adversity. Proc Natl Acad Sci U S A 112.
https://doi.org/10.1073/pnas.1408355111
Kundakovic, M., Jaric, I., 2017. The epigenetic link between prenatal adverse environments and
neurodevelopmental disorders. Genes (Basel). https://doi.org/10.3390/genes8030104
Journal Pre-proof
28
Kuruto-Niwa, R., Tateoka, Y., Usuki, Y., Nozawa, R., 2007. Measurement of bisphenol A
concentrations in human colostrum. Chemosphere 66.
https://doi.org/10.1016/j.chemosphere.2006.06.073
Kutay, H., Klepper, C., Wang, B., Hsu, S. hao, Datta, J., Yu, L., Zhang, X., Majumder, S., Motiwala, T.,
Khan, N., Belury, M., McClain, C., Jacob, S., Ghoshal, K., 2012. Reduced susceptibility of DNA
methyltransferase 1 hypomorphic (Dnmt1N/+) mice to hepatic steatosis upon feeding liquid
alcohol diet. PLoS One 7. https://doi.org/10.1371/journal.pone.0041949
Kwiatkowski, M.A., Roos, A., Stein, D.J., Thomas, K.G.F., Donald, K., 2014. Effects of prenatal
methamphetamine exposure: A review of cognitive and neuroimaging studies. Metab Brain
Dis. https://doi.org/10.1007/s11011-013-9470-7
Lees, C., Hopkins, J., 2013. Effect of aerobic exercise on cognition, academic achievement, and
psychosocial function in children: A systematic review of randomized control trials. Prev
Chronic Dis. https://doi.org/10.5888/pcd10.130010
Leighton, L.J., Bredy, T.W., 2018. Functional interplay between small non-coding RNAs and RNA
modification in the brain. Noncoding RNA. https://doi.org/10.3390/NCRNA4020015
Levenson, J.M., Sweatt, J.D., 2005. Epigenetic mechanisms in memory formation. Nat Rev
Neurosci. https://doi.org/10.1038/nrn1604
Levi, R.S., Sanderson, I.R., 2004. Dietary regulation of gene expression. Curr Opin Gastroenterol.
https://doi.org/10.1097/00001574-200403000-00015
Levine, A., Huang, Y.Y., Drisaldi, B., Griffin, E.A., Pollak, D.D., Xu, S., Yin, D., Schaffran, C., Kandel,
D.B., Kandel, E.R., 2011. Molecular mechanism for a gateway drug: Epigenetic changes
initiated by nicotine prime gene expression by cocaine. Sci Transl Med 3.
https://doi.org/10.1126/scitranslmed.3003062
Liang, Y., Li, Y., Li, Z., Liu, Z., Zhang, Z., Chang, S., Wu, J., 2012. Mechanism of folate deficiency-
induced apoptosis in mouse embryonic stem cells: Cell cycle arrest/apoptosis in G1/G0
mediated by microRNA-302a and tumor suppressor gene Lats2. International Journal of
Biochemistry and Cell Biology 44. https://doi.org/10.1016/j.biocel.2012.07.014
Ling, C., Rönn, T., 2014. Epigenetic adaptation to regular exercise in humans. Drug Discov Today.
https://doi.org/10.1016/j.drudis.2014.03.006
Lista, I., Sorrentino, G., 2010. Biological mechanisms of physical activity in preventing cognitive
decline. Cell Mol Neurobiol. https://doi.org/10.1007/s10571-009-9488-x
Liu, C., Jiao, C., Wang, K., Yuan, N., 2018. DNA Methylation and Psychiatric Disorders. pp. 175232.
https://doi.org/10.1016/bs.pmbts.2018.01.006
Liu, Y., Balaraman, Y., Wang, G., Nephew, K.P., Zhou, F.C., 2009. Alcohol exposure alters DNA
methylation profiles in mouse embryos at early neurulation. Epigenetics 4.
https://doi.org/10.4161/epi.4.7.9925
Journal Pre-proof
29
Liu, Y., Xu, D., Feng, J., Kou, H., Liang, G., Yu, H., He, X., Zhang, B., Chen, L., Magdalou, J., Wang, H.,
2012. Fetal rat metabonome alteration by prenatal caffeine ingestion probably due to the
increased circulatory glucocorticoid level and altered peripheral glucose and lipid metabolic
pathways. Toxicol Appl Pharmacol 262. https://doi.org/10.1016/j.taap.2012.05.002
Luo, Z.C., Fraser, W.D., Julien, P., Deal, C.L., Audibert, F., Smith, G.N., Xiong, X., Walker, M., 2006.
Tracing the origins of “fetal origins” of adult diseases: Programming by oxidative stress? Med
Hypotheses 66. https://doi.org/10.1016/j.mehy.2005.08.020
Ma, C.L., Ma, X.T., Wang, J.J., Liu, H., Chen, Y.F., Yang, Y., 2017. Physical exercise induces
hippocampal neurogenesis and prevents cognitive decline. Behavioural Brain Research.
https://doi.org/10.1016/j.bbr.2016.09.067
Maccani, J.Z.J., Koestler, D.C., Houseman, E.A., Marsit, C.J., Kelsey, K.T., 2013. Placental DNA
methylation alterations associated with maternal tobacco smoking at the RUNX3 gene are
also associated with gestational age. Epigenomics 5. https://doi.org/10.2217/epi.13.63
Maccani, M.A., Avissar-Whiting, M., Banister, C.E., McGonnigal, B., Padbury, J.F., Marsit, C.J., 2010.
Maternal cigarette smoking during pregnancy is associated with downregulation of miR-16,
miR-21 and miR-146a in the placenta. Epigenetics 5. https://doi.org/10.4161/epi.5.7.12762
Malvaez, M., Mhillaj, E., Matheos, D.P., Palmery, M., Wood, M.A., 2011. CBP in the nucleus
accumbens regulates cocaine-induced histone acetylation and is critical for cocaine-
associated behaviors. Journal of Neuroscience 31. https://doi.org/10.1523/JNEUROSCI.2747-
11.2011
McGee, S.L., Fairlie, E., Garnham, A.P., Hargreaves, M., 2009. Exercise-induced histone
modifications in human skeletal muscle. Journal of Physiology 587.
https://doi.org/10.1113/jphysiol.2009.181065
McGowan, P.O., Sasaki, A., D’Alessio, A.C., Dymov, S., Labonté, B., Szyf, M., Turecki, G., Meaney,
M.J., 2009. Epigenetic regulation of the glucocorticoid receptor in human brain associates
with childhood abuse. Nat Neurosci 12. https://doi.org/10.1038/nn.2270
Meccariello, R., Santoro, A., D’Angelo, S., Morrone, R., Fasano, S., Viggiano, A., Pierantoni, R.,
2020. The epigenetics of the endocannabinoid system. Int J Mol Sci.
https://doi.org/10.3390/ijms21031113
Mendonca, K., Hauser, R., Calafat, A.M., Arbuckle, T.E., Duty, S.M., 2014. Bisphenol A
concentrations in maternal breast milk and infant urine. Int Arch Occup Environ Health 87.
https://doi.org/10.1007/s00420-012-0834-9
Miles, J.N.V., Weden, M.M., 2012. Is the intergenerational transmission of smoking from mother
to child mediated by children’s behavior problems? Nicotine and Tobacco Research 14.
https://doi.org/10.1093/ntr/ntr328
Miranda, R.C., 2012. MicroRNAs and fetal brain development: Implications for ethanol teratology
during the second trimester period of neurogenesis. Front Genet.
https://doi.org/10.3389/fgene.2012.00077
Journal Pre-proof
30
Monti, B., Polazzi, E., Contestabile, A., 2010. Biochemical, Molecular and Epigenetic Mechanisms
of Valproic Acid Neuroprotection. Curr Mol Pharmacol 2.
https://doi.org/10.2174/1874467210902010095
Morgan, M.A.J., Shilatifard, A., 2020. Reevaluating the roles of histone-modifying enzymes and
their associated chromatin modifications in transcriptional regulation. Nat Genet 52.
https://doi.org/10.1038/s41588-020-00736-4
Murata, M., Kang, J.H., 2018. Bisphenol A (BPA) and cell signaling pathways. Biotechnol Adv.
https://doi.org/10.1016/j.biotechadv.2017.12.002
Newby, E.A., Myers, D.A., Ducsay, C.A., 2015. Fetal endocrine and metabolic adaptations to
hypoxia: the role of the hypothalamic-pituitary-adrenal axis. Am J Physiol Endocrinol Metab
309, E429-39. https://doi.org/10.1152/ajpendo.00126.2015
Niederer, I., Kriemler, S., Gut, J., Hartmann, T., Schindler, C., Barral, J., Puder, J.J., 2011.
Relationship of aerobic fitness and motor skills with memory and attention in preschoolers
(Ballabeina): A cross-sectional and longitudinal study. BMC Pediatr 11.
https://doi.org/10.1186/1471-2431-11-34
Nikolova, Y.S., Koenen, K.C., Galea, S., Wang, C.M., Seney, M.L., Sibille, E., Williamson, D.E., Hariri,
A.R., 2014. Beyond genotype: Serotonin transporter epigenetic modification predicts human
brain function. Nat Neurosci 17. https://doi.org/10.1038/nn.3778
Nishitani, S., Isozaki, M., Yao, A., Higashino, Y., Yamauchi, T., Kidoguchi, M., Kawajiri, S.,
Tsunetoshi, K., Neish, H., Imoto, H., Arishima, H., Kodera, T., Fujisawa, T.X., Nomura, S.,
Kikuta, K., Shinozaki, G., Tomoda, A., 2023. Cross-tissue correlations of genome-wide DNA
methylation in Japanese live human brain and blood, saliva, and buccal epithelial tissues.
Transl Psychiatry 13. https://doi.org/10.1038/s41398-023-02370-0
Noack, F., Calegari, F., 2018. Epitranscriptomics: A new regulatory mechanism of brain
development and function. Front Neurosci. https://doi.org/10.3389/fnins.2018.00085
Onishchenko, N., Karpova, N., Sabri, F., Castrén, E., Ceccatelli, S., 2008. Long-lasting depression-
like behavior and epigenetic changes of BDNF gene expression induced by perinatal exposure
to methylmercury. J Neurochem 106. https://doi.org/10.1111/j.1471-4159.2008.05484.x
Otero, N.K.H., Thomas, J.D., Saski, C.A., Xia, X., Kelly, S.J., 2012. Choline Supplementation and DNA
Methylation in the Hippocampus and Prefrontal Cortex of Rats Exposed to Alcohol During
Development. Alcohol Clin Exp Res 36. https://doi.org/10.1111/j.1530-0277.2012.01784.x
Ouko, L.A., Shantikumar, K., Knezovich, J., Haycock, P., Schnugh, D.J., Ramsay, M., 2009. Effect of
alcohol consumption on CpG methylation in the differentially methylated regions of H19 and
IG-DMR in male gametes - Implications for fetal alcohol spectrum disorders. Alcohol Clin Exp
Res 33. https://doi.org/10.1111/j.1530-0277.2009.00993.x
Pal-Bhadra, M., Bhadra, U., Jackson, D.E., Mamatha, L., Park, P.H., Shukla, S.D., 2007. Distinct
methylation patterns in histone H3 at Lys-4 and Lys-9 correlate with up- & down-regulation
of genes by ethanol in hepatocytes. Life Sci 81. https://doi.org/10.1016/j.lfs.2007.07.030
Journal Pre-proof
31
Pandey, S.C., Ugale, R., Zhang, H., Tang, L., Prakash, A., 2008. Brain chromatin remodeling: A novel
mechanism of alcoholism. Journal of Neuroscience 28.
https://doi.org/10.1523/JNEUROSCI.5731-07.2008
Park, J., Lee, K., Kim, K., Yi, S.J., 2022. The role of histone modifications: from neurodevelopment
to neurodiseases. Signal Transduct Target Ther. https://doi.org/10.1038/s41392-022-01078-9
Peleg, S., Sananbenesi, F., Zovoilis, A., Burkhardt, S., Bahari-Javan, S., Agis-Balboa, R.C., Cota, P.,
Wittnam, J.L., Gogol-Doering, A., Opitz, L., Salinas-Riester, G., Dettenhofer, M., Kang, H.,
Farinelli, L., Chen, W., Fischer, A., 2010. Altered histone acetylation is associated with age-
dependent memory impairment in mice. Science (1979) 328.
https://doi.org/10.1126/science.1186088
Pilsner, J.R., Hu, H., Ettinger, A., Sánchez, B.N., Wright, R.O., Cantonwine, D., Lazarus, A., Lamadrid-
Figueroa, H., Mercado García, A., Téllez-Rojo, M.M., Hernández-Avila, M., 2009. Influence of
prenatal lead exposure on genomic methylation of cord blood DNA. Environ Health Perspect
117. https://doi.org/10.1289/ehp.0800497
Pouyssegur, J., López-Barneo, J., 2016. Hypoxia in health and disease. Mol Aspects Med.
https://doi.org/10.1016/j.mam.2016.02.001
Prado, E.L., Dewey, K.G., 2014. Nutrition and brain development in early life. Nutr Rev 72.
https://doi.org/10.1111/nure.12102
Prini, P., Penna, F., Sciuccati, E., Alberio, T., Rubino, T., 2017. Chronic Δ8-THC Exposure Differently
Affects Histone Modifications in the Adolescent and Adult Rat Brain. Int J Mol Sci 18.
https://doi.org/10.3390/ijms18102094
Puglia, M.H., Lillard, T.S., Morris, J.P., Connelly, J.J., 2015. Epigenetic modification of the oxytocin
receptor gene influences the perception of anger and fear in the human brain. Proc Natl Acad
Sci U S A 112. https://doi.org/10.1073/pnas.1422096112
Rager, J.E., Bailey, K.A., Smeester, L., Miller, S.K., Parker, J.S., Laine, J.E., Drobná, Z., Currier, J.,
Douillet, C., Olshan, A.F., Rubio-Andrade, M., Stýblo, M., García-Vargas, G., Fry, R.C., 2014.
Prenatal arsenic exposure and the epigenome: Altered microRNAs associated with innate and
adaptive immune signaling in newborn cord blood. Environ Mol Mutagen 55.
https://doi.org/10.1002/em.21842
Rasmi, Y., Shokati, A., Hassan, A., Aziz, S.G.-G., Bastani, S., Jalali, L., Moradi, F., Alipour, S., 2023.
The role of DNA methylation in progression of neurological disorders and neurodegenerative
diseases as well as the prospect of using DNA methylation inhibitors as therapeutic agents
for such disorders. IBRO Neurosci Rep 14, 2837.
https://doi.org/10.1016/j.ibneur.2022.12.002
Renthal, W., Kumar, A., Xiao, G., Wilkinson, M., Covington, H.E., Maze, I., Sikder, D., Robison, A.J.,
LaPlant, Q., Dietz, D.M., Russo, S.J., Vialou, V., Chakravarty, S., Kodadek, T.J., Stack, A.,
Kabbaj, M., Nestler, E.J., 2009. Genome-wide Analysis of Chromatin Regulation by Cocaine
Reveals a Role for Sirtuins. Neuron 62. https://doi.org/10.1016/j.neuron.2009.03.026
Journal Pre-proof
32
Renthal, W., Maze, I., Krishnan, V., Covington, H.E., Xiao, G., Kumar, A., Russo, S.J., Graham, A.,
Tsankova, N., Kippin, T.E., Kerstetter, K.A., Neve, R.L., Haggarty, S.J., McKinsey, T.A., Bassel-
Duby, R., Olson, E.N., Nestler, E.J., 2007. Histone Deacetylase 5 Epigenetically Controls
Behavioral Adaptations to Chronic Emotional Stimuli. Neuron 56.
https://doi.org/10.1016/j.neuron.2007.09.032
Resendiz, M., Chen, Y., Öztürk, N.C., Zhou, F.C., 2013. Epigenetic medicine and fetal alcohol
spectrum disorders. Epigenomics. https://doi.org/10.2217/epi.12.80
Réus, G.Z., Abelaira, H.M., dos Santos, M.A.B., Carlessi, A.S., Tomaz, D.B., Neotti, M. v., Liranço,
J.L.G., Gubert, C., Barth, M., Kapczinski, F., Quevedo, J., 2013. Ketamine and imipramine in
the nucleus accumbens regulate histone deacetylation induced by maternal deprivation and
are critical for associated behaviors. Behavioural Brain Research 256.
https://doi.org/10.1016/j.bbr.2013.08.041
Rider, C.F., Carlsten, C., 2019. Air pollution and DNA methylation: Effects of exposure in humans.
Clin Epigenetics. https://doi.org/10.1186/s13148-019-0713-2
Rodriguez-Ayllon, M., Cadenas-Sánchez, C., Estévez-López, F., Muñoz, N.E., Mora-Gonzalez, J.,
Migueles, J.H., Molina-García, P., Henriksson, H., Mena-Molina, A., Martínez-Vizcaíno, V.,
Catena, A., Löf, M., Erickson, K.I., Lubans, D.R., Ortega, F.B., Esteban-Cornejo, I., 2019. Role of
Physical Activity and Sedentary Behavior in the Mental Health of Preschoolers, Children and
Adolescents: A Systematic Review and Meta-Analysis. Sports Medicine.
https://doi.org/10.1007/s40279-019-01099-5
Roseboom, T., de Rooij, S., Painter, R., 2006. The Dutch famine and its long-term consequences for
adult health. Early Hum Dev 82, 485491. https://doi.org/10.1016/j.earlhumdev.2006.07.001
Saab, B.J., Mansuy, I.M., 2014. Neuroepigenetics of memory formation and impairment: The role
of microRNAs. Neuropharmacology. https://doi.org/10.1016/j.neuropharm.2014.01.026
Sanders, A.P., Smeester, L., Rojas, D., DeBussycher, T., Wu, M.C., Wright, F.A., Zhou, Y.H., Laine,
J.E., Rager, J.E., Swamy, G.K., Ashley-Koch, A., Miranda, M.L., Fry, R.C., 2014. Cadmium
exposure and the epigenome: Exposure-associated patterns of DNA methylation in
leukocytes from mother-baby pairs. Epigenetics 9. https://doi.org/10.4161/epi.26798
Sathyan, P., Golden, H.B., Miranda, R.C., 2007. Competing interactions between micro-RNAs
determine neural progenitor survival and proliferation after ethanol exposure: Evidence from
an ex vivo model of the fetal cerebral cortical neuroepithelium. Journal of Neuroscience 27.
https://doi.org/10.1523/JNEUROSCI.1269-07.2007
Schenk, A., Koliamitra, C., Bauer, C.J., Schier, R., Schweiger, M.R., Bloch, W., Zimmer, P., 2019.
Impact of acute aerobic exercise on genome-wide DNA-methylation in natural killer cells-a
pilot study. Genes (Basel) 10. https://doi.org/10.3390/genes10050380
Scherma, M., Qvist, J.S., Asok, A., Huang, S.S.C., Masia, P., Deidda, M., Wei, Y.B., Soni, R.K., Fratta,
W., Fadda, P., Kandel, E.R., Kandel, D.B., Melas, P.A., 2020. Cannabinoid exposure in rat
adolescence reprograms the initial behavioral, molecular, and epigenetic response to
cocaine. Proc Natl Acad Sci U S A 117. https://doi.org/10.1073/pnas.1920866117
Journal Pre-proof
33
Schlotz, W., Phillips, D.I.W., 2009. Fetal origins of mental health: Evidence and mechanisms. Brain
Behav Immun 23. https://doi.org/10.1016/j.bbi.2009.02.001
Schmidt, R.J., Tancredi, D.J., Krakowiak, P., Hansen, R.L., Ozonoff, S., 2014. Maternal intake of
supplemental iron and risk of autism spectrum disorder. Am J Epidemiol 180.
https://doi.org/10.1093/aje/kwu208
Schmidt-Kassow, M., Zink, N., Mock, J., Thiel, C., Vogt, L., Abel, C., Kaiser, J., 2014. Treadmill
walking during vocabulary encoding improves verbal long-term memory. Behavioral and
Brain Functions 10. https://doi.org/10.1186/1744-9081-10-24
Schneider, J.S., Kidd, S.K., Anderson, D.W., 2013. Influence of developmental lead exposure on
expression of DNA methyltransferases and methyl cytosine-binding proteins in hippocampus.
Toxicol Lett 217. https://doi.org/10.1016/j.toxlet.2012.12.004
Schroeder, F.A., Penta, K.L., Matevossian, A., Jones, S.R., Konradi, C., Tapper, A.R., Akbarian, S.,
2008. Drug-induced activation of dopamine D1 receptor signaling and inhibition of class I/II
histone deacetylase induce chromatin remodeling in reward circuitry and modulate cocaine-
related behaviors. Neuropsychopharmacology 33. https://doi.org/10.1038/npp.2008.15
Scientific Opinion on Lead in Food, 2010. . EFSA Journal 8.
https://doi.org/10.2903/j.efsa.2010.1570
Seeman, T., Epel, E., Gruenewald, T., Karlamangla, A., Mcewen, B.S., 2010. Socio-economic
differentials in peripheral biology: Cumulative allostatic load. Ann N Y Acad Sci.
https://doi.org/10.1111/j.1749-6632.2009.05341.x
Sekar, S., Liang, W.S., 2019. Circular RNA expression and function in the brain. Noncoding RNA Res.
https://doi.org/10.1016/j.ncrna.2019.01.001
Shen, H.-Y., Kalda, A., Yu, L., Ferrara, J., Zhu, J., Chen, J.-F., 2008. Additive effects of histone
deacetylase inhibitors and amphetamine on histone H4 acetylation, cAMP responsive
element binding protein phosphorylation and ΔFosB expression in the striatum and
locomotor sensitization in mice. Neuroscience 157, 644655.
https://doi.org/10.1016/j.neuroscience.2008.09.019
Shi, C., Zhang, L., Qin, C., 2017. Long non-coding RNAs in brain development, synaptic biology, and
Alzheimer’s disease. Brain Res Bull. https://doi.org/10.1016/j.brainresbull.2017.03.010
Shogren-Knaak, M., Ishii, H., Sun, J.M., Pazin, M.J., Davie, J.R., Peterson, C.L., 2006. Histone H4-K16
acetylation controls chromatin structure and protein interactions. Science (1979) 311.
https://doi.org/10.1126/science.1124000
Shukla, S.D., Lee, Y.J., Park, P.H., Aroor, A.R., 2007. Acetaldehyde alters MAP kinase signalling and
epigenetic histone modifications in hepatocytes, in: Novartis Foundation Symposium.
https://doi.org/10.1002/9780470511848.ch16
Shumay, E., Logan, J., Volkow, N.D., Fowler, J.S., 2012. Evidence that the methylation state of the
monoamine oxidase A (MAOA) gene predicts brain activity of MAO A enzyme in healthy men.
Epigenetics 7, 115160. https://doi.org/10.4161/epi.21976
Journal Pre-proof
34
Sillivan, S.E., Vaissière, T., Miller, C.A., 2015. Neuroepigenetic regulation of pathogenic memories.
Neuroepigenetics. https://doi.org/10.1016/j.nepig.2014.10.003
Soellner, D.E., Grandys, T., Nuñez, J.L., 2009. Chronic prenatal caffeine exposure impairs novel
object recognition and radial arm maze behaviors in adult rats. Behavioural Brain Research
205. https://doi.org/10.1016/j.bbr.2009.08.012
St Clair, D., Xu, M., Wang, P., Yu, Y., Fang, Y., Zhang, F., Zheng, X., Gu, N., Feng, G., Sham, P., He, L.,
2005. Rates of adult schizophrenia following prenatal exposure to the Chinese famine of
1959-1961. JAMA 294. https://doi.org/10.1001/jama.294.5.557
Stenz, L., Zewdie, S., Laforge-Escarra, T., Prados, J., la Harpe, R., Dayer, A., Paoloni-Giacobino, A.,
Perroud, N., Aubry, J.M., 2015. BDNF promoter I methylation correlates between post-
mortem human peripheral and brain tissues. Neurosci Res 91.
https://doi.org/10.1016/j.neures.2014.10.003
Subbanna, S., Shivakumar, M., Umapathy, N.S., Saito, M., Mohan, P.S., Kumar, A., Nixon, R.A.,
Verin, A.D., Psychoyos, D., Basavarajappa, B.S., 2013. G9a-mediated histone methylation
regulates ethanol-induced neurodegeneration in the neonatal mouse brain. Neurobiol Dis 54.
https://doi.org/10.1016/j.nbd.2013.01.022
Susser, E., Neugebauer, R., Hoek, H.W., Brown, A.S., Lin, S., Labovitz, D., Gorman, J.M., 1996.
Schizophrenia after prenatal famine further evidence. Arch Gen Psychiatry 53.
https://doi.org/10.1001/archpsyc.1996.01830010027005
Susser, E.S., Lin, S.P., 1992. Schizophrenia After Prenatal Exposure to the Dutch Hunger Winter of
1944-1945. Arch Gen Psychiatry 49. https://doi.org/10.1001/archpsyc.1992.01820120071010
Suter, M., Abramovici, A., Showalter, L., Hu, M., Shope, C. do, Varner, M., Aagaard-Tillery, K., 2010.
In utero tobacco exposure epigenetically modifies placental CYP1A1 expression. Metabolism
59. https://doi.org/10.1016/j.metabol.2010.01.013
Suter, M., Ma, J., Harris, A., Patterson, L., Brown, K.A., Shope, C., Showalter, L., Abramovici, A.,
Aagaard-Tillery, K.M., 2011. Maternal tobacco use modestly alters correlated epigenome-
wide placental DNA methylation and gene expression. Epigenetics 6.
https://doi.org/10.4161/epi.6.11.17819
Suwabe, K., Hyodo, K., Byun, K., Ochi, G., Yassa, M.A., Soya, H., 2017. Acute moderate exercise
improves mnemonic discrimination in young adults. Hippocampus 27.
https://doi.org/10.1002/hipo.22695
Szutorisz, H., DiNieri, J.A., Sweet, E., Egervari, G., Michaelides, M., Carter, J.M., Ren, Y., Miller,
M.L., Blitzer, R.D., Hurd, Y.L., 2014. Parental THC exposure leads to compulsive heroin-
seeking and altered striatal synaptic plasticity in the subsequent generation.
Neuropsychopharmacology 39. https://doi.org/10.1038/npp.2013.352
Szutorisz, H., Egervári, G., Sperry, J., Carter, J.M., Hurd, Y.L., 2016. Cross-generational THC
exposure alters the developmental sensitivity of ventral and dorsal striatal gene expression in
male and female offspring. Neurotoxicol Teratol. https://doi.org/10.1016/j.ntt.2016.05.005
Journal Pre-proof
35
Szutorisz, H., Hurd, Y.L., 2018. High times for cannabis: Epigenetic imprint and its legacy on brain
and behavior. Neurosci Biobehav Rev. https://doi.org/10.1016/j.neubiorev.2017.05.011
Szutorisz, H., Hurd, Y.L., 2016. Epigenetic effects of cannabis exposure. Biol Psychiatry.
https://doi.org/10.1016/j.biopsych.2015.09.014
Szyf, M., 2013. The genome- and system-wide response of DNA methylation to early life adversity
and its implication on mental health. Canadian Journal of Psychiatry.
https://doi.org/10.1177/070674371305801208
Tahiliani, M., Koh, K.P., Shen, Y., Pastor, W.A., Bandukwala, H., Brudno, Y., Agarwal, S., Iyer, L.M.,
Liu, D.R., Aravind, L., Rao, A., 2009. Conversion of 5-methylcytosine to 5-
hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science (1979) 324.
https://doi.org/10.1126/science.1170116
Taniguchi, M., Carreira, M.B., Smith, L.N., Zirlin, B.C., Neve, R.L., Cowan, C.W., 2012. Histone
Deacetylase 5 Limits Cocaine Reward through cAMP-Induced Nuclear Import. Neuron 73.
https://doi.org/10.1016/j.neuron.2011.10.032
Tarragó, O., Brown, M.J., 2017. Case Studies in Environmental Medicine (Csem)-Lead Toxicity.
Agency for Toxic Substances and Disease Registry (ATSDR).
Tavares, R.S., Escada-Rebelo, S., Correia, M., Mota, P.C., Ramalho-Santos, J., 2016. The non-
genomic effects of endocrine-disrupting chemicals on mammalian sperm. Reproduction.
https://doi.org/10.1530/REP-15-0355
Taylor, S.E., Eisenberger, N.I., Saxbe, D., Lehman, B.J., Lieberman, M.D., 2006. Neural Responses to
Emotional Stimuli Are Associated with Childhood Family Stress. Biol Psychiatry 60.
https://doi.org/10.1016/j.biopsych.2005.09.027
Todd, R.D., Neuman, R.J., 2007. Gene-environment interactions in the development of combined
type ADHD: Evidence for a synapse-based model. American Journal of Medical Genetics, Part
B: Neuropsychiatric Genetics 144. https://doi.org/10.1002/ajmg.b.30640
Toledo-Rodriguez, M., Lotfipour, S., Leonard, G., Perron, M., Richer, L., Veillette, S., Pausova, Z.,
Paus, T., 2010. Maternal smoking during pregnancy is associated with epigenetic
modifications of the brain-derived neurotrophic factor-6 exon in adolescent offspring.
American Journal of Medical Genetics, Part B: Neuropsychiatric Genetics 153.
https://doi.org/10.1002/ajmg.b.31109
Tomasiewicz, H.C., Jacobs, M.M., Wilkinson, M.B., Wilson, S.P., Nestler, E.J., Hurd, Y.L., 2012.
Proenkephalin mediates the enduring effects of adolescent cannabis exposure associated
with adult opiate vulnerability. Biol Psychiatry 72.
https://doi.org/10.1016/j.biopsych.2012.04.026
Tomas-Roig, J., Benito, E., Agis-Balboa, R.C., Piscitelli, F., Hoyer-Fender, S., di Marzo, V.,
Havemann-Reinecke, U., 2017. Chronic exposure to cannabinoids during adolescence causes
long-lasting behavioral deficits in adult mice. Addiction Biology 22.
https://doi.org/10.1111/adb.12446
Journal Pre-proof
36
Toranõ, E.G., Garciá, M.G., Fernández-Morera, J.L., Ninõ-Garciá, P., Fernández, A.F., 2016. The
Impact of External Factors on the Epigenome: In Utero and over Lifetime. Biomed Res Int.
https://doi.org/10.1155/2016/2568635
Uchida, S., Hara, K., Kobayashi, A., Otsuki, K., Yamagata, H., Hobara, T., Suzuki, T., Miyata, N.,
Watanabe, Y., 2011. Epigenetic status of Gdnf in the ventral striatum determines
susceptibility and adaptation to daily stressful events. Neuron 69.
https://doi.org/10.1016/j.neuron.2010.12.023
Ungerer, M., Knezovich, J., Ramsay, M., 2012. In utero alcohol exposure, epigenetic changes, and
their consequences. Alcohol Res.
Urdinguio, R.G., Sanchez-Mut, J. v., Esteller, M., 2009. Epigenetic mechanisms in neurological
diseases: genes, syndromes, and therapies. Lancet Neurol. https://doi.org/10.1016/S1474-
4422(09)70262-5
Ursini, G., Bollati, V., Fazio, L., Porcelli, A., Iacovelli, L., Catalani, A., Sinibaldi, L., Gelao, B., Romano,
R., Rampino, A., Taurisano, P., Mancini, M., di Giorgio, A., Popolizio, T., Baccarelli, A., de Blasi,
A., Blasi, G., Bertolino, A., 2011. Stress-related methylation of the catechol-O-
methyltransferase Val 158 allele predicts human prefrontal cognition and activity. J Neurosci
31, 66928. https://doi.org/10.1523/JNEUROSCI.6631-10.2011
Use of Folic Acid for Prevention of Spina Bifida and Other Neural Tube Defects1983-1991, 1991. .
JAMA: The Journal of the American Medical Association 266.
https://doi.org/10.1001/jama.1991.03470090024009
van Praag, H., 2009. Exercise and the brain: something to chew on. Trends Neurosci.
https://doi.org/10.1016/j.tins.2008.12.007
van Praag, H., 2008. Neurogenesis and exercise: Past and future directions. Neuromolecular Med.
https://doi.org/10.1007/s12017-008-8028-z
van Praag, H., Kempermann, G., Gage, F.H., 1999. Running increases cell proliferation and
neurogenesis in the adult mouse dentate gyrus. Nat Neurosci 2.
https://doi.org/10.1038/6368
Vandenberg, L.N., Ehrlich, S., Belcher, S.M., Ben-Jonathan, N., Dolinoy, D.C., Hugo, E.R., Hunt, P. a.,
Newbold, R.R., Rubin, B.S., Saili, K.S., Soto, A.M., Wang, H.-S., Saal, F.S. vom, 2013. Low dose
effects of bisphenol A: An integrated review of in vitro, laboratory animal, and epidemiology
studies. Endocrine Disruptors 1.
Vijg, J., 2014. Somatic mutations, genome mosaicism, cancer and aging. Curr Opin Genet Dev.
https://doi.org/10.1016/j.gde.2014.04.002
Viré, E., Brenner, C., Deplus, R., Blanchon, L., Fraga, M., Didelot, C., Morey, L., van Eynde, A.,
Bernard, D., Vanderwinden, J.M., Bollen, M., Esteller, M., di Croce, L., de Launoit, Y., Fuks, F.,
2006. The Polycomb group protein EZH2 directly controls DNA methylation. Nature 439.
https://doi.org/10.1038/nature04431
Journal Pre-proof
37
Vukojevic, V., Kolassa, I.T., Fastenrath, M., Gschwind, L., Spalek, K., Milnik, A., Heck, A., Vogler, C.,
Wilker, S., Demougin, P., Peter, F., Atucha, E., Stetak, A., Roozendaal, B., Elbert, T.,
Papassotiropoulos, A., de Quervain, J.F.D., 2014. Epigenetic modification of the
glucocorticoid receptor gene is linked to traumatic memory and post-traumatic stress
disorder risk in genocide survivors. Journal of Neuroscience 34.
https://doi.org/10.1523/JNEUROSCI.1526-14.2014
Walton, E., Liu, J., Hass, J., White, T., Scholz, M., Roessner, V., Gollub, R., Calhoun, V.D., Ehrlich, S.,
2014. MB-COMT promoter DNA methylation is associated with working-memory processing
in schizophrenia patients and healthy controls. Epigenetics 9.
https://doi.org/10.4161/epi.29223
Wang, D., Szyf, M., Benkelfat, C., Provençal, N., Turecki, G., Caramaschi, D., Côté, S.M., Vitaro, F.,
Tremblay, R.E., Booij, L., 2012. Peripheral SLC6A4 DNA methylation is associated with in vivo
measures of human brain serotonin synthesis and childhood physical aggression. PLoS One 7.
https://doi.org/10.1371/journal.pone.0039501
Wang, K., Liu, H., Hu, Q., Wang, L., Liu, J., Zheng, Z., Zhang, W., Ren, J., Zhu, F., Liu, G.H., 2022.
Epigenetic regulation of aging: implications for interventions of aging and diseases. Signal
Transduct Target Ther. https://doi.org/10.1038/s41392-022-01211-8
Wang, L.L., Zhang, Z., Li, Q., Yang, R., Pei, X., Xu, Y., Wang, J., Zhou, S.F., Li, Y., 2009. Ethanol
exposure induces differential microRNA and target gene expression and teratogenic effects
which can be suppressed by folic acid supplementation. Human Reproduction 24.
https://doi.org/10.1093/humrep/den439
Watson, C.T., Szutorisz, H., Garg, P., Martin, Q., Landry, J.A., Sharp, A.J., Hurd, Y.L., 2015. Genome-
Wide DNA Methylation Profiling Reveals Epigenetic Changes in the Rat Nucleus Accumbens
Associated with Cross-Generational Effects of Adolescent THC Exposure.
Neuropsychopharmacology 40. https://doi.org/10.1038/npp.2015.155
Weaver, I.C.G., 2014. Integrating Early Life Experience, Gene Expression, Brain Development, and
Emergent Phenotypes. pp. 277307. https://doi.org/10.1016/B978-0-12-800222-3.00011-5
Weaver, I.C.G., Cervoni, N., Champagne, F.A., D’Alessio, A.C., Sharma, S., Seckl, J.R., Dymov, S.,
Szyf, M., Meaney, M.J., 2004. Epigenetic programming by maternal behavior. Nat Neurosci 7.
https://doi.org/10.1038/nn1276
Weiss, I.C., Franklin, T.B., Vizi, S., Mansuy, I.M., 2011. Inheritable effect of unpredictable maternal
separation on behavioral responses in mice. Front Behav Neurosci.
https://doi.org/10.3389/fnbeh.2011.00003
Woelfel, J.R., Dudley-Javoroski, S., Shields, R.K., 2018. Precision physical therapy: Exercise, the
epigenome, and the heritability of environmentally modified traits. Phys Ther 98.
https://doi.org/10.1093/ptj/pzy092
Wolff, G.L., Kodell, R.L., Moore, S.R., Cooney, C.A., 1998. Maternal epigenetics and methyl
supplements affect agouti gene expression in A vy /a mice . The FASEB Journal 12.
https://doi.org/10.1096/fasebj.12.11.949
Journal Pre-proof
38
Wolstenholme, J.T., Rissman, E.F., Connelly, J.J., 2011. The role of Bisphenol A in shaping the brain,
epigenome and behavior. Horm Behav. https://doi.org/10.1016/j.yhbeh.2010.10.001
World Health Organization, 2010. Exposure to Lead: A major public health concern. World Healh
Organization.
Wu, S., Hivert, M.F., Cardenas, A., Zhong, J., Rifas-Shiman, S.L., Agha, G., Colicino, E., Just, A.C.,
Amarasiriwardena, C., Lin, X., Litonjua, A.A., Demeo, D.L., Gillman, M.W., Wright, R.O., Oken,
E., Baccarelli, A.A., 2017. Exposure to low levels of lead in utero and umbilical cord blood
DNA methylation in project viva: An epigenome-wide association study. Environ Health
Perspect 125. https://doi.org/10.1289/EHP1246
Xia, L.P., Shen, L., Kou, H., Zhang, B.J., Zhang, L., Wu, Y., Li, X.J., Xiong, J., Yu, Y., Wang, H., 2014.
Prenatal ethanol exposure enhances the susceptibility to metabolic syndrome in offspring
rats by HPA axis-associated neuroendocrine metabolic programming. Toxicol Lett 226.
https://doi.org/10.1016/j.toxlet.2014.01.023
Xie, Y., Liu, J., Benbrahim-Tallaa, L., Ward, J.M., Logsdon, D., Diwan, B.A., Waalkes, M.P., 2007.
Aberrant DNA methylation and gene expression in livers of newborn mice transplacentally
exposed to a hepatocarcinogenic dose of inorganic arsenic. Toxicology 236.
https://doi.org/10.1016/j.tox.2007.03.021
Xu, D., Chen, M., Pan, X. liang, Xia, L. ping, Wang, H., 2011. Dexamethasone induces fetal
developmental toxicity through affecting the placental glucocorticoid barrier and depressing
fetal adrenal function. Environ Toxicol Pharmacol 32.
https://doi.org/10.1016/j.etap.2011.08.003
Xu, D., Liang, G., Yan, Y.E., He, W.W., Liu, Y.S., Chen, L.B., Magdalou, J., Wang, H., 2012a. Nicotine-
induced over-exposure to maternal glucocorticoid and activated glucocorticoid metabolism
causes hypothalamic-pituitary-adrenal axis-associated neuroendocrine metabolic alterations
in fetal rats. Toxicol Lett 209. https://doi.org/10.1016/j.toxlet.2012.01.006
Xu, D., Wu, Y., Liu, F., Liu, Y.S., Shen, L., Lei, Y.Y., Liu, J., Ping, J., Qin, J., Zhang, C., Chen, L.B.,
Magdalou, J., Wang, H., 2012b. A hypothalamic-pituitary-adrenal axis-associated
neuroendocrine metabolic programmed alteration in offspring rats of IUGR induced by
prenatal caffeine ingestion. Toxicol Appl Pharmacol 264.
https://doi.org/10.1016/j.taap.2012.08.016
Xu, Dan, Zhang, B., Liang, G., Ping, J., Kou, H., Li, X., Xiong, J., Hu, D., Chen, L., Magdalou, J., Wang,
H., 2012. Caffeine-Induced Activated Glucocorticoid Metabolism in the Hippocampus Causes
Hypothalamic-Pituitary-Adrenal Axis Inhibition in Fetal Rats. PLoS One 7.
https://doi.org/10.1371/journal.pone.0044497
Yan, Y. e., Liu, L., Wang, J. fei, Liu, F., Li, X. hai, Qin, H. quan, Wang, H., 2014. Prenatal nicotinic
exposure suppresses fetal adrenal steroidogenesis via steroidogenic factor 1 (SF-1)
deacetylation. Toxicol Appl Pharmacol 277. https://doi.org/10.1016/j.taap.2014.03.019
Journal Pre-proof
39
Yaoi, T., Itoh, K., Nakamura, K., Ogi, H., Fujiwara, Y., Fushiki, S., 2008. Genome-wide analysis of
epigenomic alterations in fetal mouse forebrain after exposure to low doses of bisphenol A.
Biochem Biophys Res Commun 376. https://doi.org/10.1016/j.bbrc.2008.09.028
Yeo, M., Berglund, K., Hanna, M., Guo, J.U., Kittur, J., Torres, M.D., Abramowitz, J., Busciglio, J.,
Gao, Y., Birnbaumer, L., Liedtke, W.B., 2013. Bisphenol A delays the perinatal chloride shift in
cortical neurons by epigenetic effects on the Kcc2 promoter. Proc Natl Acad Sci U S A 110.
https://doi.org/10.1073/pnas.1300959110
Yeomans, M.R., Mobini, S., Chambers, L., 2007. Additive effects of flavour-caffeine and flavour-
flavour pairings on liking for the smell and flavour of a novel drink. Physiol Behav 92.
https://doi.org/10.1016/j.physbeh.2007.06.006
Yochum, C., Doherty-Lyon, S., Hoffman, C., Hossain, M.M., Zelikoff, J.T., Richardson, J.R., 2014.
Prenatal cigarette smoke exposure causes hyperactivity and aggressive behavior: Role of
altered catecholamines and BDNF. Exp Neurol 254.
https://doi.org/10.1016/j.expneurol.2014.01.016
Youk, J., An, Y., Park, S., Lee, J.K., Ju, Y.S., 2020. The genome-wide landscape of C:G > T:A
polymorphism at the CpG contexts in the human population. BMC Genomics 21.
https://doi.org/10.1186/s12864-020-6674-1
Zhao, Z., Shilatifard, A., 2019. Epigenetic modifications of histones in cancer. Genome Biol.
https://doi.org/10.1186/s13059-019-1870-5
Zhou, F.C., Balaraman, Y., Teng, M., Liu, Y., Singh, R.P., Nephew, K.P., 2011a. Alcohol Alters DNA
Methylation Patterns and Inhibits Neural Stem Cell Differentiation. Alcohol Clin Exp Res 35.
https://doi.org/10.1111/j.1530-0277.2010.01391.x
Zhou, F.C., Chen, Y., Love, A., 2011b. Cellular DNA methylation program during neurulation and its
alteration by alcohol exposure. Birth Defects Res A Clin Mol Teratol 91.
https://doi.org/10.1002/bdra.20820
Ziegler, C., Dannlowski, U., Bräuer, D., Stevens, S., Laeger, I., Wittmann, H., Kugel, H., Dobel, C.,
Hurlemann, R., Reif, A., Lesch, K.P., Heindel, W., Kirschbaum, C., Arolt, V., Gerlach, A.L.,
Hoyer, J., Deckert, J., Zwanzger, P., Domschke, K., 2015. Oxytocin Receptor Gene
Methylation: Converging Multilevel Evidence for a Role in Social Anxiety.
Neuropsychopharmacology 40. https://doi.org/10.1038/npp.2015.2
Journal Pre-proof
40
Figures Legend
Figure 1: Epigenetic processes
Most common epigenetic processes: (1) DNA methylation, which inhibits the transcription
process (2) Histone modification, such as methylation and acetylation (histone acetylation
results in relaxation of the chromatin and ultimately greater transcription) (3) non-coding
RNAs block transcription and/or translation process.
Figure 2: Influence on brain development and functions by aversive environmental
exposure
The continuous increment of different types of environmental factors influences epigenetic
marks, including DNA methylation (5mC, 5hmC) and histone modification (H3ac, H3K4me,
etc.) to affect synaptogenesis and neurogenesis together affect the brain function and
neuropsychiatric disease at a given time of the life.
Journal Pre-proof
41
Table 1: Summary of imaging epigenetics study
Methods
Year
Gene
Findings
Structural
magnetic
resonance
imaging (MRI)
2013
FKBP5
Upregulated FKBP5 methylation reduces volume of the
right hippocampal head (Klengel et al., 2013).
2014
BDNF
Upregulated BDNF promoter methylation reduces white
matter integrity in patients with major depressive
disorder (Choi et al., 2015).
SLC6A4
Increased methylation in a functional element of the
SLC6A4 promoter upregulates hippocampal volume
(Dannlowski et al., 2014).
2015
SLC6A4
Increased SLC6A4 methylation was associated with
childhood trauma and decreased hippocampal volume
(Booij et al., 2015).
Functional MRI
2011
COMT
Reduced methylation of the Val158 COMT allele
diminishes cortical efficiency in a working memory task,
particularly in the context of stress (Ursini et al., 2011).
2012
OXTR
Raised methylation of the OXTR gene promoter increases
the activity in the temporal-parietal junction and dorsal
anterior cingulate cortex during a social perception task
(Jack et al., 2012).
2014
SLC6A4
Increased SLC6A4 promoter methylation increases
amygdala reactivity to threatening faces (Nikolova et al.,
2014).
COMT
Increased COMT promoter methylation upregulates left
dorsolateral prefrontal cortex activity during a working-
memory task across patients with schizophrenia and
controls (Walton et al., 2014).
NR3C1
Increased NR3C1 promoter methylation upregulates the
activity in the right ventrolateral prefrontal cortex and
cuneus, as well as reduced performance, in a memory
task in healthy men (Vukojevic et al., 2014).
2015
SLC6A4
SLC6A4 methylation modulates the activity differently in
the insula, operculum, hippocampus and amygdala in an
emotional attention-shifting task (Frodl et al., 2015).
OXTR
Upregulated methylation of the OXTR gene promoter
increases the activity in the amygdala, insula and
fusiform gyrus, as well as decreases the amygdala
connectivity with regulatory regions during threatening
face processing (Puglia et al., 2015).
Decreased OXTR methylation upregulates amygdala
activity during social-phobia related word processing in
individuals with social anxiety disorder (Ziegler et al.,
2015).
Positron emission
tomography
2012
SLC6A4
Increased SLC6A4 promoter methylation reduces 5-HT
synthesis in the orbitofrontal cortex (Wang et al., 2012).
MAOA
Increased methylation near the MAOA promoter was
associated with lower MAOA activity in healthy men
(Shumay et al., 2012).
Journal Pre-proof
42
Table 2: Studies on prenatal exposure to environmental agents related with epigenetics changes
Histone
modification
DNA
methylation
miRNAs
Outcomes
H3Kac,
H3K4me,
H3K9me,
H3K27me (Pal-
Bhadra et al.,
2007; Shukla et
al., 2007;
Subbanna et al.,
2013)
5mC, 5hmC
(Chen et al.,
2013; Garro et
al., 1991; Otero
et al., 2012;
Ouko et al.,
2009; Wolff et
al., 1998)
miR148,
miR152,
miR21,
miR153,
miR335
(Kutay et al.,
2012;
Sathyan et al.,
2007)
Delayed
formation in
the neural
tube,
forebrain,
hindbrain
(Zhou et al.,
2011b)
Delayed
maturation
and
diminished
size of
hippocampus
(Chen et al.,
2013)
Declined
neuron cell
number,
cortical plate,
thickness
(Zhou et al.,
2011b)
H3ac (Cronican
et al., 2013)
5mC
(Intarasunanont
et al., 2012; Kile
et al., 2012; Xie
et al., 2007)
let 7a, miR16,
miR17,
miR20a,
miR20b,
miR26b,
miR96,
miR98,
miR107,
miR126,
miR195, and
miR-454
(Rager et al.,
2014)
Interrupted
spatial and
episodic memory,
as well as fear
conditioning
performance
(Cronican et al.,
2013)
H3ac (Yaoi et
al., 2008)
5mC (Yaoi et al.,
2008)
N/A
Delay the
perinatal chloride
shift in cortical
neurons (Yeo et
al., 2013)
N/A
5mC
(Buscariollo et
al., 2014; Dan
Xu et al., 2012)
N/A
Growth
retardation (Dan
Xu et al., 2012)
H3K4me,
H3K9me
(Dinieri et al.,
2011)
N/A
N/A
Upregulated
opiate reward
sensitivity in adult
(Dinieri et al.,
2011)
Journal Pre-proof
43
N/A
5mC (Castillo et
al., 2012;
Kippler et al.,
2013; Sanders et
al., 2014)
N/A
Decreased birth
weight and height
(Castillo et al.,
2012)
H3ac (Akchiche
et al., 2012)
5mC (Guéant et
al., 2013)
miR124,
miR302a
(Kerek et al.,
2013; Liang
et al., 2012)
Brain size
reduction, growth
retardation (Kerek
et al., 2013)
H3ac, H3K4me
(Bihaqi et al.,
2011)
5mC (Bihaqi et
al., 2011;
Schneider et al.,
2013)
N/A
Increased
neurodegeneration
in primate (Bihaqi
et al., 2011)
H3ac, H3K27me
(Onishchenko et
al., 2008)
5mC (Bose et
al., 2012;
Onishchenko et
al., 2008)
N/A
Depression like
behaviour
(Onishchenko et
al., 2008)
N/A
5mC (Itzhak et
al., 2015)
N/A
Increased cocaine
reward and hyper-
locomotion as well
as diminished
conditional fear
(Itzhak et al.,
2015)
H3ac (Levine et
al., 2011)
5mC (Breton et
al., 2009;
Maccani et al.,
2013; Suter et
al., 2011, 2010)
miR16,
miR21,
miR146a
(Maccani et
al., 2010)
Increased
aggression,
locomotion in
adult male
(Yochum et
al., 2014)
Birth weight
reduction
(Suter et al.,
2011)
N/A
5mC (Chorbov
et al., 2011)
N/A
N/A
H3ac, H3K9me,
H3K27me
(Dalton et al.,
2014; Réus et
al., 2013)
5mC
(Champagne
and Curley,
2009;
Darnaudéry and
Maccari, 2008;
Heim and
Binder, 2012;
Szyf, 2013)
miR16, miR9,
miR29a,
miR124,
miR132,
miR212 (Bai
et al., 2012;
Uchida et al.,
2011)
Induces
depressive-like
behaviors, altered
response to
aversive
environments
(Champagne and
Curley, 2009;
Franklin et al.,
2010)
H3ac (Balmer et
al., 2012; Monti
et al., 2010)
N/A
N/A
Diminished birth
rate, reduced
sociability, and
social preference
(Kim et al., 2011)
H3ac (Klein et
al., 2009)
N/A
N/A
N/A
Journal Pre-proof
44
Table 3: Studies on cannabinoid exposure related with epigenetics dysregulation
Cannabinoid types
Epigenetic
modifications
Exposure
period and
models
Studied brain region and effects
Δ9-
tetrahydrocannabinol
H3K4me2,
H3K9me3
Promoter, gene
body
Prenatal
male rats
Drd2 mRNA levels in nucleus
accumbens of adult brain (Dinieri et
al., 2011)
Adolescent
male rats
Penk mRNA levels in nucleus
accumbens shell of adult brain
(Tomasiewicz et al., 2012)
Global H3K9me3
levels,
promoters
Adolescent
female rats
↓ mRNA expression of genes
related to endocannabinoid system
and synaptic plasticity in prefrontal
cortex of adult brain (Cuccurazzu et
al., 2018)
H3K4me2,
H3K9me3,
Global H3K14ac
levels
Adolescent
and adult
female rats
Brain region-specific and age-
specific alterations of histone
modifications at different times
after exposure in hippocampus,
nucleus accumbens and amygdala
of adolescent and adult brains (Prini
et al., 2017)
CpG DNA
methylation at
promoters,
intergenic
regions,
especially in
gene bodies
Adolescent
female and
male rats
Altered methylation at loci
implicated in synaptic plasticity,
including the Dlg4 gene network of
nucleus accumbens in adult brains
(Watson et al., 2015)
DNA
methylation at
promoter
Adult male
rats
↓ DNA methylation of synaptic
Dlgap2 in nucleus accumbens of
adult brains (Watson et al., 2015)
WIN-55,2122
cannabinoid agonist
Intragenic DNA
methylation
Adolescent
male mice
↑ DNA methylation and ↓ mRNA
expression of Rgs7 in hippocampus
of adult brains (Tomas-Roig et al.,
2017)
Chromatin
accessibility
(ATAC-seq) at
promoter, gene
body
↑ Accessibility at Npas2 and
splicing at prefrontal cortex of adult
brain (Scherma et al., 2020).
Global DNA
methylation
levels
↑ DNA methylation and ↑ DNMT
expression at prefrontal cortex of
adult brain (Ibn Lahmar Andaloussi
et al., 2019).
Journal Pre-proof
45
HU-210 cannabinoid
agonist
microRNAs
Adolescent
male mice
Expression of various microRNAs
altered at entorhinal cortex of
adolescent male rat (Hollins et al.,
2014)
.
Journal Pre-proof
46
Figure-1
Journal Pre-proof
47
Figure-2
Journal Pre-proof
Highlights
A complex interplay between epigenetic modifications and their substantial impact on brain
functions.
Micronutrients, caffeine, toxins, and harmful substances affect the brain's epigenetic
landscape.
The intricate links between epigenetic changes and behavioral outcomes emphasize the
possibility of targeted therapies in brain disorders
Journal Pre-proof
Conflicts of interest/Competing interests: Authors express no conflicts of interest
Journal Pre-proof
... Regardless of microbiota in the uterus, maternal gut microbial metabolites play a vital role in supplying energy, nutrients, and essential vitamins like B complex, folate, choline, and betaine. Environmental variables, such as early life stress, can have long-lasting effects on the brain and behavior via the epigenome, which regulates gene expression [148,149]. The metabolic and immunological alterations observed in pregnant women due to these factors induce changes in maternal gut microbiota that initiate in the second trimester and increase throughout the third trimester [35]. ...
Article
Full-text available
Evidence is emerging on the role of maternal diet, gut microbiota, and other lifestyle factors in establishing lifelong health and disease, which are determined by transgenerationally inherited epigenetic modifications. Understanding epigenetic mechanisms may help identify novel biomarkers for gestation-related exposure, burden, or disease risk. Such biomarkers are essential for developing tools for the early detection of risk factors and exposure levels. It is necessary to establish an exposure threshold due to nutrient deficiencies or other environmental factors that can result in clinically relevant epigenetic alterations that modulate disease risks in the fetus. This narrative review summarizes the latest updates on the roles of maternal nutrients (n-3 fatty acids, polyphenols, vitamins) and gut microbiota on the placental epigenome and its impacts on fetal brain development. This review unravels the potential roles of the functional epigenome for targeted intervention to ensure optimal fetal brain development and its performance in later life.
... This orchestrated interplay of neurotrophic, anti-inflammatory, and metabolic processes work hand in hand with the gut microbiome in conferring neuroprotection associated with physical activity. Epigenetic changes from physical activity also contribute to its neuroprotective role (106,112,(128)(129)(130). Given that yoga is both exercise and a mind-body therapy, both processes are likely contributing to the beneficial effects of yoga on the microbiota-gut-brain-immune interface and therefore reduced risk of neurodegenerative disease. ...
Article
Full-text available
Importance Research is beginning to elucidate the sophisticated mechanisms underlying the microbiota-gut-brain-immune interface, moving from primarily animal models to human studies. Findings support the dynamic relationships between the gut microbiota as an ecosystem (microbiome) within an ecosystem (host) and its intersection with the host immune and nervous systems. Adding this to the effects on epigenetic regulation of gene expression further complicates and strengthens the response. At the heart is inflammation, which manifests in a variety of pathologies including neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and Multiple Sclerosis (MS). Observations Generally, the research to date is limited and has focused on bacteria, likely due to the simplicity and cost-effectiveness of 16s rRNA sequencing, despite its lower resolution and inability to determine functional ability/alterations. However, this omits all other microbiota including fungi, viruses, and phages, which are emerging as key members of the human microbiome. Much of the research has been done in pre-clinical models and/or in small human studies in more developed parts of the world. The relationships observed are promising but cannot be considered reliable or generalizable at this time. Specifically, causal relationships cannot be determined currently. More research has been done in Alzheimer’s disease, followed by Parkinson’s disease, and then little in MS. The data for MS is encouraging despite this. Conclusions and relevance While the research is still nascent, the microbiota-gut-brain-immune interface may be a missing link, which has hampered our progress on understanding, let alone preventing, managing, or putting into remission neurodegenerative diseases. Relationships must first be established in humans, as animal models have been shown to poorly translate to complex human physiology and environments, especially when investigating the human gut microbiome and its relationships where animal models are often overly simplistic. Only then can robust research be conducted in humans and using mechanistic model systems.
... Caffeine finds extensive use in both the food and pharmaceutical industries (Mallick & Duttaroy, 2023;Rohweder et al., 2024). Its proven benefits include enhancing alertness, concentration, and expediting central nervous system processes, effectively combating fatigue (Deljou et al., 2024;Shaddel & Rajabi-Moghaddam, 2024). ...
Article
Full-text available
Micropollutants, such as caffeine (M-CF), pose a significant threat to ecosystems and human health through water and food sources. The utilization of metal oxide-based photocatalysts has proven to be an effective treatment method for the removal of organic pollutants. This study explores the efficacy of Ag-doped ZnO (Ag/ZnO) for removing M-CF from wastewater. The characterization of Ag/ZnO underscores the crucial role of band gap energy in the photocatalytic degradation process. This parameter influences the separation of electrons and holes (e⁻/h⁺) and the generation of reactive radicals. Under solar light, Ag/ZnO demonstrated markedly superior photocatalytic activity, achieving an impressive degradation efficiency of approximately 93.4%, in stark contrast to the 53.2% occurred by ZnO. Moreover, Ag/ZnO exhibited a remarkable degradation efficiency of M-CF in wastewater, reaching 83.5%. A key advantage of Ag/ZnO lies in its potential for recovery and reuse in subsequent treatments, contributing to a reduction in operational costs for industrial wastewater treatment. Impressively, even after five cycles, Ag/ZnO maintained a noteworthy photodegradation rate of M-CF at 78.6%. These results strongly suggest that Ag/ZnO presents a promising solution for the removal of micropollutants in wastewater, with potential scalability for industrial and large-scale applications.
... It is worth to cite the potential impact of epigenetics to the pathogenesis of human diseases [57][58][59]. Currently, the role of control of gene expression by non-coding RNAs is a hot topic of research including cardiovascular diseases [60][61][62]. ...
Article
Full-text available
Atherosclerotic Cardiovascular Disease (ASCVD) represents the leading cause of death worldwide, and individual screening should be based on behavioral, metabolic, and genetic profile derived from data collected in large population-based studies. Due to the polygenic nature of ASCVD, we aimed to assess the association of genomics with ASCVD risk and its impact on the occurrence of acute myocardial infarction, stroke, or peripheral artery thrombotic-ischemic events at population level. CardioVascular Genes (CV-GENES) is a nationwide, multicenter, 1:1 case-control study of 3,734 patients in Brazil. Inclusion criterion for cases is the first occurrence of one of the ASCVD events. Individuals without known ASCVD will be eligible as controls. A core lab will perform the genetic analyses through low-pass whole genome sequencing and whole exome sequencing. In order to estimate the independent association between genetic polymorphisms and ASCVD, a polygenic risk score (PRS) will be built through a hybrid approach including effect size of each Single Nucleotide Polymorphism (SNP), number of effect alleles observed, sample ploidy, total number of SNPs included in the PRS, and number of non-missing SNPs in the sample. In addition, the presence of pathogenic or likely pathogenic variants will be screened in 8 genes ( ABCG5 , ABCG8 , APOB , APOE , LDLR , LDLRAP1 , LIPA , PCSK9 ) associated with atherosclerosis. Multiple logistic regression will be applied to estimate adjusted odds ratios (OR) and 95% confidence intervals (CI), and population attributable risks will be calculated. Clinical trial registration: This study is registered in clinicaltrials.gov ( NCT05515653 ).
... Toxicants directly or indirectly affect epigenetic processes like DNA methylation, histone modifications and noncoding RNA expression. High-throughput sequencing defines epigenetic alterations and illuminates new toxicity pathways missed by purely genetic approaches [58]. Notable examples linking toxic exposures and epigenetic changes include air pollution-induced respiratory effects, arsenic carcinogenesis mechanisms and transgenerational impacts of certain pesticides. ...
Article
Full-text available
Traditional toxicity testing emphasizes animal models with growing concerns regarding predictive capacity, throughput and ethics. Rapid innovation surrounding human cell platforms, bioengineered tissues, omics techniques and computational tools offers more modern alternatives aligned with expanding knowledge of chemical biological pathways. These disruptive approaches promise immense potential to transform next-generation chemical safety assessment and drug development pipelines. This review provides clinical researchers an updated, comprehensive perspective across evolving areas of focus in new toxicity testing methods with analysis of latest advances and translational context. We survey progress in two- and three-dimensional human cell cultures recapitulating tissue/organ complexity impossible in conventional assays. Complementing this, computational modeling integrates structure-activity relationships, physicochemical properties and physiological interactions to predict pharmacokinetics and toxicity in silico. Expanding model organisms add further dimensionality and demographic relevance. High-throughput omics and imaging technologies unravel mechanisms and illuminate biomarkers undetectable by standard measures. Specialized techniques show high promise addressing toxicodynamic intricacies within disease contexts like diabetes and NAFLD. Evaluating traditional medicines and expanding phytochemicals likewise represents an area of growth well-suited for contemporary platforms. Future outlook weighs remarkable potential advantages in reducing animal testing demands, enabling precision toxicology links to clinical medicine and overhauling core chemical risk assessment frameworks. This review intends to catalyze discourse on strategic optimization priorities and roadmaps towards fully unlocking the immense yet still emerging public health potential of these disruptive techniques poising transformation in toxicity sciences centered on human-focused models.
Article
Purpose of review Perinatal mental health research provides an important perspective on neurobehavioral development. Here, we aim to review the association of maternal perinatal health with offspring neurodevelopment, providing an update on (self-)regulation problems, hypothesized mechanistic pathways, progress and challenges, and implications for mental health. Recent findings (1) Meta-analyses confirm that maternal perinatal mental distress is associated with (self-)regulation problems which constitute cognitive, behavioral, and affective social-emotional problems, while exposure to positive parental mental health has a positive impact. However, effect sizes are small. (2) Hypothesized mechanistic pathways underlying this association are complex. Interactive and compensatory mechanisms across developmental time are neglected topics. (3) Progress has been made in multiexposure studies. However, challenges remain and these are shared by clinical, translational and public health sciences. (4) From a mental healthcare perspective, a multidisciplinary and system level approach employing developmentally-sensitive measures and timely treatment of (self-)regulation and coregulation problems in a dyadic caregiver-child and family level approach seems needed. The existing evidence-base is sparse. Summary During the perinatal period, addressing vulnerable contexts and building resilient systems may promote neurobehavioral development. A pluralistic approach to research, taking a multidisciplinary approach to theoretical models and empirical research needs to be fostered.
Article
Full-text available
Epigenetic aging clocks have gained significant attention as a tool for predicting age-related health conditions in clinical and research settings. They have enabled geroscientists to study the underlying mechanisms of aging and assess the effectiveness of anti-aging therapies, including diet, exercise and environmental exposures. This review explores the effects of modifiable lifestyle factors' on the global DNA methylation landscape, as seen by aging clocks. We also discuss the underlying mechanisms through which these factors contribute to biological aging and provide comments on what these findings mean for people willing to build an evidence-based pro-longevity lifestyle.
Article
Full-text available
Neuroepigenetics considers genetic sequences and the interplay with environmental influences to elucidate vulnerability risk for various neurological and psychiatric disorders. However, evaluating DNA methylation of brain tissue is challenging owing to the issue of tissue specificity. Consequently, peripheral surrogate tissues were used, resulting in limited progress compared with other epigenetic studies, such as cancer research. Therefore, we developed databases to establish correlations between the brain and peripheral tissues in the same individuals. Four tissues, resected brain tissue, blood, saliva, and buccal mucosa (buccal), were collected from 19 patients (aged 13-73 years) who underwent neurosurgery. Moreover, their genome-wide DNA methylation was assessed using the Infinium HumanMethylationEPIC BeadChip arrays to determine the cross-tissue correlation of each combination. These correlation analyses were conducted with all methylation sites and with variable CpGs, and with when these were adjusted for cellular proportions. For the averaged data for each CpG across individuals, the saliva-brain correlation (r = 0.90) was higher than that for blood-brain (r = 0.87) and buccal-brain (r = 0.88) comparisons. Among individual CpGs, blood had the highest proportion of CpGs correlated to the brain at nominally significant levels (19.0%), followed by saliva (14.4%) and buccal (9.8%). These results were similar to the previous IMAGE-CpG results; however, cross-database correlations of the correlation coefficients revealed a relatively low (brain vs. blood: r = 0.27, saliva: r = 0.18, and buccal: r = 0.24). To the best of our knowledge, this is the fifth study in the literature initiating the development of databases for correlations between the brain and peripheral tissues in the same individuals. We present the first database developed from an Asian population, specifically Japanese samples (AMAZE-CpG), which would contribute to interpreting individual epigenetic study results from various Asian populations.
Article
Full-text available
Genome-wide studies related to neurological disorders and neurodegenerative diseases have pointed to the role of epigenetic changes such as DNA methylation, histone modification, and noncoding RNAs. DNA methylation machinery controls the dynamic regulation of methylation patterns in discrete brain regions. Objective This review aims to describe the role of DNA methylation in inhibiting and progressing neurological and neurodegenerative disorders and therapeutic approaches. Methods A Systematic search of PubMed, Web of Science, and Cochrane Library was conducted for all qualified studies from 2000 to 2022. Results For the current need of time, we have focused on the DNA methylation role in neurological and neurodegenerative diseases and the expression of genes involved in neurodegeneration such as Alzheimer's, Depression, and Rett Syndrome. Finally, it appears that the various epigenetic changes do not occur separately and that DNA methylation and histone modification changes occur side by side and affect each other. We focused on the role of modification of DNA methylation in several genes associated with depression (NR3C1, NR3C2, CRHR1, SLC6A4, BDNF, and FKBP5), Rett syndrome (MECP2), Alzheimer's, depression (APP, BACE1, BIN1 or ANK1) and Parkinson's disease (SNCA), as well as the co-occurring modifications to histones and expression of non-coding RNAs. Understanding these epigenetic changes and their interactions will lead to better treatment strategies. Conclusion This review captures the state of understanding of the epigenetics of neurological and neurodegenerative diseases. With new epigenetic mechanisms and targets undoubtedly on the horizon, pharmacological modulation and regulation of epigenetic processes in the brain holds great promise for therapy.
Article
Full-text available
Aging is accompanied by the decline of organismal functions and a series of prominent hallmarks, including genetic and epigenetic alterations. These aging-associated epigenetic changes include DNA methylation, histone modification, chromatin remodeling, non-coding RNA (ncRNA) regulation, and RNA modification, all of which participate in the regulation of the aging process, and hence contribute to aging-related diseases. Therefore, understanding the epigenetic mechanisms in aging will provide new avenues to develop strategies to delay aging. Indeed, aging interventions based on manipulating epigenetic mechanisms have led to the alleviation of aging or the extension of the lifespan in animal models. Small molecule-based therapies and reprogramming strategies that enable epigenetic rejuvenation have been developed for ameliorating or reversing aging-related conditions. In addition, adopting health-promoting activities, such as caloric restriction, exercise, and calibrating circadian rhythm, has been demonstrated to delay aging. Furthermore, various clinical trials for aging intervention are ongoing, providing more evidence of the safety and efficacy of these therapies. Here, we review recent work on the epigenetic regulation of aging and outline the advances in intervention strategies for aging and age-associated diseases. A better understanding of the critical roles of epigenetics in the aging process will lead to more clinical advances in the prevention of human aging and therapy of aging-related diseases.
Article
Full-text available
Epigenetic regulatory mechanisms, including DNA methylation, histone modification, chromatin remodeling, and microRNA expression, play critical roles in cell differentiation and organ development through spatial and temporal gene regulation. Neurogenesis is a sophisticated and complex process by which neural stem cells differentiate into specialized brain cell types at specific times and regions of the brain. A growing body of evidence suggests that epigenetic mechanisms, such as histone modifications, allow the fine-tuning and coordination of spatiotemporal gene expressions during neurogenesis. Aberrant histone modifications contribute to the development of neurodegenerative and neuropsychiatric diseases. Herein, recent progress in understanding histone modifications in regulating embryonic and adult neurogenesis is comprehensively reviewed. The histone modifications implicated in neurodegenerative and neuropsychiatric diseases are also covered, and future directions in this area are provided.
Article
Full-text available
Dietary polyunsaturated fatty acids (PUFAs), especially omega-3 (n-3) and n-6 long-chain (LC) PUFAs, are indispensable for the fetus' brain supplied by the placenta. Despite being highly unsaturated, n-3 LCPUFA-docosahexaenoic acid (DHA) plays a protective role as an antioxidant in the brain. Deficiency of DHA during fetal development may cause irreversible damages in neurodevelopment programming. Dietary PUFAs can impact placental structure and functions by regulating early placentation processes, such as angiogenesis. They promote remodeling of uteroplacental architecture to facilitate increased blood flow and surface area for nutrient exchange. The placenta's fatty acid transfer depends on the uteroplacental vascular development, ensuring adequate maternal circulatory fatty acids transport to fulfill the fetus' rapid growth and development requirements. Maternal n-3 PUFA deficiency predominantly leads to placental epigenetic changes than other fetal developing organs. A global shift in DNA methylation possibly transmits epigenetic instability in developing fetuses due to n-3 PUFA deficiency. Thus, an optimal level of maternal omega-3 (n-3) PUFAs may protect the placenta's structural and functional integrity and allow fetal growth by controlling the aberrant placental epigenetic changes. This narrative review summarizes the recent advances and underpins the roles of maternal PUFAs on the structure and functions of the placenta and their relevance to fetal growth and brain development.
Article
Full-text available
In the last few years, more and more scientists have suggested and confirmed that epigenetic regulators are tightly connected and form a comprehensive network of regulatory pathways and feedback loops. This is particularly interesting for a better understanding of processes that occur in the development and progression of various diseases. Appearing on the preclinical stages of diseases, epigenetic aberrations may be prominent biomarkers. Being dynamic and reversible, epigenetic modifications could become targets for a novel option for therapy. Therefore, in this review, we are focusing on histone modifications and ncRNAs, their mutual regulation, role in cellular processes and potential clinical application.
Article
Full-text available
Background Particulate matter (PM), a major component of ambient air pollution, accounts for a substantial burden of diseases and fatality worldwide. Maternal exposure to PM during pregnancy is particularly harmful to children’s health since this is a phase of rapid human growth and development. Method In this review, we synthesize the scientific evidence on adverse health outcomes in children following prenatal exposure to the smallest toxic components, fine (PM 2.5 ) and ultrafine (PM 0.1 ) PM. We highlight the established and emerging findings from epidemiologic studies and experimental models. Results Maternal exposure to fine and ultrafine PM directly and indirectly yields numerous adverse birth outcomes and impacts on children’s respiratory systems, immune status, brain development, and cardiometabolic health. The biological mechanisms underlying adverse effects include direct placental translocation of ultrafine particles, placental and systemic maternal oxidative stress and inflammation elicited by both fine and ultrafine PM, epigenetic changes, and potential endocrine effects that influence long-term health. Conclusion Policies to reduce maternal exposure and health consequences in children should be a high priority. PM 2.5 levels are regulated, yet it is recognized that minority and low socioeconomic status groups experience disproportionate exposures. Moreover, PM 0.1 levels are not routinely measured or currently regulated. Consequently, preventive strategies that inform neighborhood/regional planning and clinical/nutritional recommendations are needed to mitigate maternal exposure and ultimately protect children’s health.
Article
Full-text available
Mammalian brains feature exceptionally high levels of non-CpG DNA methylation alongside the canonical form of CpG methylation. Non-CpG methylation plays a critical regulatory role in cognitive function, which is mediated by the binding of MeCP2, the transcriptional regulator that when mutated causes Rett syndrome. However, it is unclear whether the non-CpG neural methylation system is restricted to mammalian species with complex cognitive abilities or has deeper evolutionary origins. To test this, we investigated brain DNA methylation across 12 distantly related animal lineages, revealing that non-CpG methylation is restricted to vertebrates. We discovered that in vertebrates, non-CpG methylation is enriched within a highly conserved set of developmental genes transcriptionally repressed in adult brains, indicating that it demarcates a deeply conserved regulatory program. We also found that the writer of non-CpG methylation, DNMT3A, and the reader, MeCP2, originated at the onset of vertebrates as a result of the ancestral vertebrate whole-genome duplication. Together, we demonstrate how this novel layer of epigenetic information assembled at the root of vertebrates and gained new regulatory roles independent of the ancestral form of the canonical CpG methylation. This suggests that the emergence of non-CpG methylation may have fostered the evolution of sophisticated cognitive abilities found in the vertebrate lineage. By studying brain DNA methylation across 13 distantly related animals, the authors show that non-CpG DNA methylation, which plays a regulatory role in cognition, is restricted to vertebrates and was assembled at the origin of the vertebrate lineage as a result of the ancestral vertebrate whole-genome duplication.
Article
Histone-modifying enzymes are implicated in the control of diverse DNA-templated processes including gene expression. Here, we outline historical and current thinking regarding the functions of histone modifications and their associated enzymes. One current viewpoint, based largely on correlative evidence, posits that histone modifications are instructive for transcriptional regulation and represent an epigenetic ‘code’. Recent studies have challenged this model and suggest that histone marks previously associated with active genes do not directly cause transcriptional activation. Additionally, many histone-modifying proteins possess non-catalytic functions that overshadow their enzymatic activities. Given that much remains unknown regarding the functions of these proteins, the field should be cautious in interpreting loss-of-function phenotypes and must consider both cellular and developmental context. In this Perspective, we focus on recent progress relating to the catalytic and non-catalytic functions of the Trithorax–COMPASS complexes, Polycomb repressive complexes and Clr4/Suv39 histone-modifying machineries. Recent progress relating to the catalytic and non-catalytic functions of histone modifying complexes warrants a fresh look at the role of histone modifications and the “histone code” model.