You are on page 1of 11

Review

For reprint orders, please contact: reprints@futuremedicine.com

Epigenetics and lifestyle

The concept of ‘lifestyle’ includes different factors such as nutrition, behavior, stress, physical activity,
working habits, smoking and alcohol consumption. Increasing evidence shows that environmental and
lifestyle factors may influence epigenetic mechanisms, such as DNA methylation, histone acetylation and
miRNA expression. It has been identified that several lifestyle factors such as diet, obesity, physical activity,
tobacco smoking, alcohol consumption, environmental pollutants, psychological stress and working on
night shifts might modify epigenetic patterns. Most of the studies conducted so far have been centered
on DNA methylation, whereas only a few investigations have studied lifestyle factors in relation to histone
modifications and miRNAs. This article reviews current evidence indicating that lifestyle factors might
affect human health via epigenetic mechanisms.

KEYWORDS: DNA methylation n environmental exposures n epigenetics n histone Jorge Alejandro


modifications n lifestyle Alegría-Torres1,
Andrea Baccarelli2
The term ‘lifestyle’ is broadly used to describe alterations  [5,6] . Modulation of gene methyla- & Valentina Bollati†3
the “typical way of life or manner of living char- tion has been observed in human endothelial 1
Departamento de Toxicologia
acteristic of an individual or group” [1] . This cells incubated with arachidonic acid promoting Ambiental, Facultad de Medicina,
Universidad Autonoma de San Luis
concept includes different factors such as diet, upregulation of proangiogenic mechanisms  [7] . Potosi, Mexico
behavior, stress, physical activity, working habits, Conversely, polyunsaturated fatty acids may 2
Exposure, Epidemiology & Risk
smoking and alcohol consumption. Individual have a suppressive function in tumorigenic Program, Department of
Environmental Health, Harvard School
genetic background and environmental factors processes through dampening of inflammation of Public Health, Boston, MA, USA
are intertwined with lifestyle in determining the and NF‑kB pathway [8] . Moreover, diets rich in 3
Center of Molecular & Genetic
Epidemiology, Department of
health status of individuals (Figure 1) . Increasing fruits and vegetables, which contain many nat- Environmental & Occupational Health,
evidence shows that environmental and lifestyle ural antioxidants, can yield anticancer protec- Università degli Studi di Milano
& Fondazione IRCCS Ospedale
factors may influence epigenetic mechanisms, tion [9] . Chen and Xu have extensively reviewed Maggiore Policlinico, Mangiagalli e
such as DNA methylation, histone modifications the potential epigenetic effects of several nutri- Regina Elena, Via San Barnaba 8,
and miRNA expression. Epigenetic mechanisms tional components, mostly derived from vegeta- Milan 20122, Italy

Author for correspondence:
are flexible genomic parameters that can change bles [10] . For instance, a study in healthy human Tel.: +39 250 320 127
genome function under exogenous influence but subjects fed with a single serving of broccoli Fax: +39 250 320 103
valentina.bollati@unimi.it
also provide a mechanism that allows for the sta- sprouts showed inhibition of histone deacetylase
ble propagation of gene activity states from one (HDAC) activity in circulating peripheral blood
generation of cells to the next [2] . Alterations in mononuclear cells 3–6  h after consumption,
epigenetic marks have also been associated with with concurrent induction of histone H3 and
a variety of human diseases, including cancer, H4 acetylation [11] . An in vitro study on human
cardiovascular, respiratory and neurodegenera- tumor colon cell lines revealed that high doses
tive diseases [3] . In this article we will discuss of diallyl-disulfide from garlic increased histone
examples of lifestyle factors that have been inves- H3 and H4 acetylation [12] .
tigated in relation to possible epigenetic effects,
and the implication of lifestyle-related epigenetic „„ Folate & vitamin B12 Intake
changes in disease etiology (Table 1) . Folic acid and vitamin B12 play an important role
in DNA metabolism and are required for the syn-
Nutrition thesis of methionine and S‑adenosylmethionine
As examined in multiple investigations, nutri- (SAM), the common methyl donor required for
tion might play a role in the modification of epi- the maintenance of methylation patterns in DNA
genetic mechanisms. For example, a diet rich [13] . Methylation reactions could be influenced
in polyunsaturated fatty acids could generate through the modification of the ratio between
mutagenic free radicals and oxidative stress [4] , SAM and S‑adenosylhomocysteine (SAH) [14] .
which have been directly linked to epigenetic The SAM:SAH ratio is a primary determinant

10.2217/EPI.11.22 © 2011 Future Medicine Ltd Epigenomics (2011) 3(3), 267–277 ISSN 1750-1911 267
Review Alegría-Torres, Baccarelli & Bollati

ENVIRONMENTAL TOXICANTS

Timing and concentration Source and route of exposure

Type of pollutants
Particulate matter Organic compounds

Endocrine disrupting chemicals


Metals Pesticides
In utero Age

INDIVIDUAL CHARACTERISTICS
SETTINGS

Home Environment Epigenetics Gender


Outdoor Indoor

Work Genetic
(occupational risk) make-up

Diet Exercise

Alcohol Smoking

LIFESTYLE

Figure 1. Environment–epigenetic interactions.

of the methylation capacity because SAM is con- higher-than-recommended doses (500 mg/day)


verted to SAH by methionine adeno­syltransferase. for 12 weeks [20] . In the context of the SAM
Taking into consideration that methionine is cycle, choline can donate methyl groups to SAM
regenerated by methylation of homocysteine via since it is a trimethylated molecule. Folate has
the folate and B12-dependent reactions, a folate- also been shown to reverse the dysregulation of
deficient diet could interfere with this system [15] . miRNA expression associated with hepatocel-
The SAM:SAH ratio has been related with DNA lular carcinogenesis, potentially by restoring
methylation patterns. For example, a study con- dietary methyl donors  [21] . The growing body
ducted in rats examined the maternal folate status of evidence showing that folate intake modulates
and DNA methylation in placenta. A significant epigenetic mechanisms has been actively inves-
positive correlation was found between placen- tigated in relation to potential anticarcinogenic
tal DNA methylation, hepatic and plasma folate properties suggested by epidemiological stud-
­levels, and hepatic SAM:SAH ratio [16] . ies [22–25] . Also, investigating epigenetic effects
Low folate intakes have been associated from folate might help understand paradoxical
with risk of colorectal cancer [17] . Moreover, observations, such as those linking very high
folate depletion has been shown to cause lym- folate intakes with the development of colorectal
phocyte DNA hypomethylation in healthy carcinoma from adenomatous polyps [26] .
post­menopausal women, an alteration reverted
by folate repletion [18,19] . A recent study car- „„ Polyphenols
ried out among individuals susceptible to Polyphenols are a large family of natural com-
folate deficiency showed that methylation sta- pounds widely distributed in plant foods, that
tus can be corrected with choline supply at have been shown to modify the activity of

268 Epigenomics (2011) 3(3) future science group


Epigenetics & lifestyle Review
DNA methyltransferases, histone acetylases Obesity & physical activity
and HDACs [27,28] . In particular, studies on Overweight, obesity and sedentary lifestyle are
cancer cells have shown that polyphenols can established and prevalent risk factors for several
reverse in in vitro models some of the epigenetic diseases, including cancer and cardiovascular
aberrations associated with malignant transfor- disease [48–50] . Because body weight is regu-
mation [29] . Inhibitory effects on DNA methyl­ lated by genes controlling energy homeostasis,
transferases have been observed both in  vitro it has been hypothesized that dietary macro­
and in vivo using different dietary sources of nutrients that affect DNA methylation could
polyphenols [28] . A retrospective ana­lysis asso- contribute to develop obesity through epigenetic
ciated CDX2 and BMP‑2 gene hypermethyla- mechanisms [51] . Epigenetic biomarkers of obes-
tion with past low intake of polyphenol sources ity, including genes involved in adipogenesis
such as cruciferous vegetables and green tea in (SOCS1/SOCS3), methylation patterns of obes-
patients with primary gastrocarcinoma [30,31] . ity-related genes (FGF2, PTEN, CDKN1A and
Green tea contains (-)-epigallocatechin-3-gallate ESR1), inflammation genes as well as intermedi-
(EGCG), which is an inhibitor of DNA methyl­ ary metabolism and insulin signaling pathway
transferases activity. EGCG has been shown to genes, could help to predict susceptibility and
reactivate methylation-silenced genes in cancer prevent obesity [52] .
cell lines [32,33] . In in  vitro experiments with Emerging evidence indicates that epigenetic
esophageal, prostate, colon and breast cancer mechanisms may be involved in mediating effects
cells lines, several CpG islands in various loci of physical activity. In a recent work, physical
were efficiently demethylated by EGCG, thus activity was associated with higher methylation
leading to the expression of previously silenced in peripheral blood lymphocytes of long inter-
genes [32–35] . spersed nucleotide element (LINE)‑1 elements,
Soy beans are also extremely rich in poly­ a class of repeated sequences highly repeated
phenols [28] . Soy polyphenols include phytoes- in the human genome [53] . Low methylation
trogens such as genistein, biochanin A and daid-
zein [36] . These compounds have also been shown Table 1. Lifestyle factors with epigenetic effects.
to inhibit DNA methyltransferases and HDACs Factor Example Studies on Ref.
in cancer cell lines and to revert aberrant CpG
Nutrition Folate Humans [18,19]
island methylation [37] . Li et al. demonstrated in Phytoestrogen Breast cancer benign human cells [38]
benign (MCF‑10AT) and cancer (MCF7) breast Polyphenols Human cancer cells [37]
cells that all three main DNA methyltransferases Selenium Human cancer cells [29,32–35]
(DNMTs; DNMT1, DNMT3a and DNMT3b) Humans [30,31]
were downregulated by genistein [38] . These Human cancer cells [40]

results might help explain epidemiology data Physical activity Exercise Humans [53,57]
indicating that soy consumption is associated Human muscle biopsy tissues [56]

with reduced risk of hormone-related cancers [39] . Tobacco smoke Cigarette smoke Humans [66]
Cigarette smoke Lung cancer patients [63]
condensate Placentas [68]
„„ Selenium Respiratory epithelia [62]
Selenium can epigenetically modulate DNA Rats and mice [69]
and histones to activate methylation-silenced Alcohol High alcohol intake Humans [73]
genes [40] . Increasing data suggest that selenium Chronic exposure Mouse fetal neurons [74,75]
may have anticarcinogenic properties through Acute exposure Neural stem cells [76]
modifications of epigenetic processes in the Mouse [77,78]
cell [41–43] . Selenium has been shown to directly Pollutants Arsenic Humans [81–83]
inhibit DNMT expression and activity  [44,45] . PM10 Humans [89,95]

Selenium can also restore the expression of Black carbon Humans [94]
Benzene Humans [97]
hypermethylated genes, such as GSTP1, APC
PAHs Humans [103]
and CSR1, in human prostate cancer cells by Human lymphoblastoid cells [100]
downregulating DNMTs and inhibiting HDAC POPs Human umbilical cord blood [102]
activity [40] . These genes are known to have anti- Humans [103]
cancer activity by protecting against oxidative Emotional Stressful experiences Rats [105]
damage, detoxification of carcinogenetic chemi- Mice [108]
cals or tumor suppression [40] . Moreover, in ani- Suicide victims [109]

mal models, a selenium-deficient diet has been Shiftwork Working at night Humans [115]

shown to induce DNA hypomethylation [46,47] . PAH: Polycyclic aromatic hydrocarbon; PM: Particulate matter; POP: Persistent organic pollutant.

future science group www.futuremedicine.com 269


Review Alegría-Torres, Baccarelli & Bollati

in LINE‑1 repetitive elements has been associ- A study that evaluated global DNA meth-
ated with inflammatory responses, as well as ylation from buccal cells of children exposed
with chromosomal instability [54] . Interestingly, to prenatal maternal smoking demonstrated
elderly individuals with high LINE‑1 meth- hypomethylation of LINE‑1 repetitive ele-
ylation in peripheral blood lymphocytes have ments. In the same study, a microarray ana­
been recently shown to have lower incidence lysis of 1536 CpG sites identified differential
and mortality from ischemic heart disease and methylation of CpG loci in eight genes. Two
stroke [55] . Whether the decreased cardiovascu- of them, AXL and PTPRO, were validated
lar risks associated with LINE‑1 methylation by pyrosequencing and showed significant
reflect beneficial effects from physical activity increases in methylation [66] . Following find-
remains to be determined. In human muscle ings indicating that miRNAs in human placen-
biopsies following exercise, a global increase in tas are differentially expressed in association
H3K36 acetylation has also been observed [56] . with adverse pregnancy outcomes [67] , a recent
Moreover, a brief exercise has been shown to study found that candidate miRNAs impli-
alter miRNA profiles in circulating neutrophils cated in growth and developmental processes
in humans, including 38 miRNAs involved in (i.e., miR‑16, miR‑21 and miR‑146a) were sig-
inflammatory pathways [57] . nificantly downregulated in cigarette smoke-
exposed placentas compared with controls [68] .
Tobacco smoke Moreover, downregulation of miRNA expres-
Tobacco smoke contains a complex mixture of sion was also observed in animal experiments
organic and inorganic chemicals, many of which when the lungs of mice and rats were exposed
have carcinogenic, proinflammatory and proath- to cigarette smoke. In this study, mir‑34b,
erogenic properties. Individual effects of these mir‑345, mir‑421, mir‑450b, mir‑466 and
components have been examined through dif- mir‑469 were downregulated at high doses
ferent epigenetic studies, but the results are still of exposure; however, expression was restored
inconclusive. For example, an in  vitro chronic 1 week after smoking cessation [69] .
toxicity study of normal human fibroblast on
benzo[a]pyrene  –  a prominent carcinogenic Alcohol consumption
polycyclic aromatic hydrocarbon (PAH) found In contrast to PAHs and other carcinogenic mol-
in cigarette smoke – found no aberrant patterns of ecules found in tobacco smoke and tar, ethyl
DNA methylation in genomic regions of relevance alcohol is not mutagenic per se, but rather acts
for lung cancer [58] . mainly as a cocarcinogen [70] . A Netherlands
Conversely, cigarette smoke condensate has cohort study on diet and cancer correlated the
been shown to decrease the nuclear levels of intake of folate and alcohol with changes in
certain histone modifications such as H4K16 methylation of tumor suppressor and DNA repair
acetylation and H4K20 trimethylation in res- genes (APC‑1A, p14ARF, p16INK4A, hMLH1,
piratory epithelial cells [59] . These alterations O6‑MGMT and RASSF1A) in paraffin-embed-
were similar to changes in histone modifications ded colorectal cancer tissues [71] . Also, this work
that can be found in lung cancer tissues and suggested the association between the intake of
commonly precede aberrant DNA methyla- other methyl donors such as methionine, vita-
tion [60,61] . Moreover, histone demethylation in mins B6 and B12 with an increased frequency
H19 and IGF2 occurred before the DNA hyper- of promoter hypermethylation of genes involved
methylation-mediated silencing of some tumor in colorectal carcinogenesis [71] . However, a sec-
suppressor genes such as p16, MGMT, DAPK, ond cohort study did not find any association of
E‑cadherin and Cdh13 in lung carcinogen- folate intake, methionine or alcohol with MLH1
esis induced by tobacco smoke [62] . Therefore, hypermethylation, a frequent and well-charac-
histone modifications might anticipate DNA terized early event in the development of color-
methylation changes [60–62] . ectal cancer [72] . A positive association between
p53 hypomethylation has been reported in vitamin B6 intake and tumors showing MLH1
peripheral blood lymphocytes of smoking lung hypermethylation was found, suggesting vita-
cancer patients [63] . Despite the lack of consist- min B6 may enhance colorectal cancer risk [72] .
ent evidence for p53 methylation data in human Alcohol consumption has also been suggested to
cancer, p53 hypomethylation has been proposed modify the association between blood markers of
to be associated with early events in carcino- DNA methylation and disease. In a population-
genesis such as DNA double-strand breaks and based case–control study on a Polish population,
chromosomal instability [64,65] . Hou et al. demonstrated that repetitive-element

270 Epigenomics (2011) 3(3) future science group


Epigenetics & lifestyle Review
hypomethylation in blood leukocyte DNA was A large body of in vitro and animal studies have
associated with gastric cancer and that the asso- shown that arsenic subtracts methyl donors from
ciation between LINE‑1 hypomethylation and DNA methylation reactions and induces global
gastric cancer was stronger among individuals DNA hypomethylation [82] . An unexpected
who were current alcohol drinkers [73] . finding was recently reported in vivo, as a glo-
Growing evidence indicates that alcohol may bal dose-dependent hypermethylation of blood
have effects on growth and neuronal development DNA was observed in Bangladeshi adults with
through epigenetic marks. Mouse fetal cortical chronic arsenic exposure. This effect was modi-
neurons chronically exposed to ethanol in vitro fied by folate, suggesting that arsenic-induced
experienced NR2B gene demethylation, which increases in DNA methylation were dependent
encodes an ionotropic glutamate receptor possibly on methyl availability [82] . The same group,
involved in certain memory and learning proc- however, subsequently reported that lower blood
esses [74,75] . Instead, acute exposure to ethanol DNA methylation was a strongly associated
induced hypermethylation of specific cell cycle with arsenic-induced skin lesions in a related
genes inhibiting the growth factor-regulated cell Bangladeshi population [83] .
cycle progression in monolayer cultures of neural
stem cells. Lengthening the time between G1 and „„ Air pollution
S phase was observed when cells were exposed Exposure to air pollution, particularly to par-
for 48 h [76] . In the mouse strain C57BL/6, alco- ticulate matter (PM), has been associated with
hol exposure at the early embryo stage altered increased morbidity and mortality from cardi-
DNA methylation in embryos with a neural tube orespiratory disease, as well as with increased lung
defect phenotype [77] . This mouse model showed cancer risk [84–88] . In a human study, Tarantini
changes in the expression of genes involved in et al. recently demonstrated that iNOS promoter
metabolism and development, such as NLGN3, methylation decreased in blood samples of foun-
ELAV12, SOX21 and SIM1. These alterations dry workers with well-characterized exposure to
may contribute to malformations and abnormal PM10 in samples taken at the end of a 4-day work
fetal development [77] . Subsequently, Zhou et al. week compared with baseline samples [89] . iNOS
found a reduction in expression of neurogenin, demethylation is expected to increase expression
Sox5, Bhlhe22, Igf1, Efemp1, Tieg and Edil3 and activity of the iNOS protein, an established
in mouse embryo cultures [78] . In this study, the key player in inflammation and oxidative stress
changes in the expression of the genes involved generation, two primary mechanisms that have
in neural tube development were modulated by been suggested to link inhalation of air pollutants
changes in DNA methylation patterns [78] . to their acute health effects [90–92] . In the same
study, long-term exposure to PM10 was nega-
Environmental pollutants tively associated with methylation in both Alu
In environmental studies, the flexibility of epi- and LINE‑1 [89] . Decreased LINE‑1 methylation
genetic states has generated growing interest in was also observed in association with exposure
evaluating whether environmental exposures to black carbon, a marker of traffic particles, on
can modify epigenetic states, including DNA 1097 blood DNA samples from the Normative
methyl­ation and histone modifications [79] . Aging Study (NAS), a repeated-measure inves-
Studies of DNA methylation and histone modi- tigation of elderly men in the Boston area (MA,
fication in relation to environmental exposures USA). As blood LINE‑1 hypomethylation has
to potentially toxic chemicals have been exam- been found in patients with cancer [93] and
ined in detail in a recent review article [80] . In the cardiovascular disease [94] , such changes may
following sections, we briefly review the main reproduce epi­genetic processes related to disease
classes of environmental exposures that are most development and represent mechanisms by which
frequently considered epigenetic toxicants. particulate air pollution affects human health [94] .
A recent occupational study has recently exam-
„„ Arsenic ined the effects of exposure to PM and metal
In a human study from India, a significant DNA components on miRNA expression in 63 work-
hypermethylation of p53 and p16 promoter ers at an electric-furnace steel plant. miR‑222
regions was observed in blood DNA of subjects and miR‑21 – two candidate miRNAs related to
exposed to toxic arsenic levels compared with oxidative stress and inflammation – were overex-
controls [81] . In this study, p53 and p16 hyper- pressed and positively correlated with the levels
methylation showed a dose–response relation- of lead exposure and oxidative DNA damage,
ship with arsenic measured in drinking water. respectively [95] .

future science group www.futuremedicine.com 271


Review Alegría-Torres, Baccarelli & Bollati

„„ Aromatic hydrocarbons & other for 48 h with hydroquinone, one of the active
organic pollutants benzene metabolites [100] . In a study of Polish
High-level exposure to benzene has been associ- male nonsmoking coke-oven workers, chronic
ated with increased risk of acute myelogenous exposure to PAHs has been shown to modify
leukemia [96] , which is characterized by aber- the methylation status of specific gene promot-
rant global hypomethylation and gene-specific ers (p53, p16, HIC1 and IL-6 ), as well as of Alu
hypermethylation/hypomethylation. In a and LINE‑1 repetitive elements [101] . Perera
study of gasoline station attendants and traf- et al. published an exploratory study that used
fic police officers, airborne benzene exposure methylation-sensitive restriction fingerprinting
was shown to be associated with a significant to analyze umbilical cord white blood cell DNA
reduction in LINE‑1 and Alu methylation in of 20 children exposed to PAHs. Over 30 DNA
peripheral blood DNA [97] . Airborne benzene sequences were identified whose methylation
was also associated with hypermethylation status was dependent on the level of maternal
in p15 and hypomethyl­ation of the MAGE‑1 PAH exposure [102] . Rusiecki et al. evaluated the
cancer-antigen gene [97] . These findings show relationship between plasma concentrations of
that benzene exposure at relatively low levels persistent organic pollutants and blood global
may induce altered DNA methylation, repro- DNA methylation, estimated in Alu repeated
ducing the aberrant epi­genetic patterns found elements, in 70 Greenlandic Inuit, a population
in malignant cells. Also, benzene-associated presenting some of the highest reported levels
demethylation of repetitive elements may help of persistent organic pollutants worldwide. In
explain the epidemiological data linking ben- this article, a significant inverse linear relation-
zene exposure with increases risk of multiple ship was found between trichlorodichloro­
myeloma [98,99] , which also exhibits reduced phenylethane (DDT), dichlorodiphenyldichlo-
methylation in Alu and LINE‑1 repetitive ele- roethylene (DDE), b‑benzene hexachloride,
ments [97] . These human data were recently con- oxychlordane, a‑chlordane, mirex, several
firmed by the finding of global hypomethyla- polychlorinated biphenyls and the sum of all
tion in human TK6 lymphoblastoid cells treated persistent organic pollutants [103] .

Food DNA methylation Food


• Folate • Polyphenols from
• EGCG from green tea vegetables
• Selenium Gene expression regulation • Selenium

Physical activity Physical activity


Hyper Hypo
Tobacco smoke
Silencing Expression Covalent modifications
Intrauterine life of histones
• Maternal diet
• Tobacco smoke Me

Alcohol Relaxed/compact chromatin structure


• High intake associated with differential
transcriptional activity
Pollutants
• Arsenic Me
• Chromate
• PM Regulation of miRNA expression (by DNA methylation
• Benzene of miRNAs loci)
Me
• PAHs
• POPs
Physical activity
Aging
Translational repression Cigarette smoke
Stress conditions of transcript degradation
Intrauterine life
Shiftwork • Tobacco smoke

Figure 2. Lifestyle factors participating in environment–epigenetic interactions.


EGCG: (-)-epigallocatechin-3-gallate; PAH: Polycyclic aromatic hydrocarbon; PM: Particulate matter; POP: Persistent organic pollutant.

272 Epigenomics (2011) 3(3) future science group


Epigenetics & lifestyle Review
Psychological stress Shiftwork
Earlier studies have indicated that DNA methyla- Recent advances have revealed that chronobio-
tion is sensitive to stressful environmental expo- logical regulators may induce chromatin remod-
sures in early development and later in life [104–109] . eling [112] . The CLOCK gene has been suggested
The glucocorticoid receptor gene promoter was to regulate circadian rhythms through a his-
studied in the hippocampus of human suicide vic- tone acetyltransferase activity which promotes
tims and controls [109] . Hypermethylation of the chromatin-remodeling events implicated in cir-
glucocorticoid receptor gene was found among cadian control of gene expression [113,114] . The
suicide victims with a history of abuse in child- circadian adjustment may be affected by dif-
hood, but not among controls or suicide victims ferent factors such as shiftwork. According to
with a negative history of childhood abuse [109] . several epidemiological studies shiftwork that
On the contrary, positive early social experiences requires working at night can have a negative
might have a mitigating effect on stress responses impact on the health and wellbeing of workers
later in life via epigenetic mechanisms, suggest- owing to a mismatch between the endogenous
ing a protective role for positive early parental circadian timing system and the environmen-
care [110,111] . This is shown in animal studies that tal synchronizers (e.g., light/dark cycle) [115] .
have demonstrated that higher maternal care, Epigenetic reprogramming events in circadian
as reflected in higher licking and grooming of genes have been proposed as potential regula-
the pups, induces hypomethylation of the gluco- tory mechanisms of circadian rhythms [116,117] .
corticoid receptor gene in the hippocampus and A recent study on a population of night-shift-
reduces responses to stress [110] . workers has shown alterations in blood DNA

Executive summary
ƒƒ Lifestyle includes different factors such as nutrition, behavior, stress, physical activity, working habits, smoking and alcohol consumption.
ƒƒ Environmental and lifestyle factors may influence epigenetic mechanisms.
Nutrition
ƒƒ Folate and vitamin B12 intake
– Epidemiological data support the anticarcinogenic property of folate.
– A protective effect of low folate status against colorectal cancer was reported.
– Contrasting results suggest that folic acid supplementation could exert a negative effect on already existing lesions.
ƒƒ Polyphenols
– Polyphenols can impact DNA methyltransferases, histone acetylases and histone deacetylases inducing reversibility of
epigenetic dysregulation.
ƒƒ Selenium
– Selenium can impact the DNA methylation status interacting directly with DNA methyltransferases.
Obesity & physical activity
ƒƒ Macronutrient composition of the diet could help to develop obesity through epigenetic mechanisms.
ƒƒ Epigenetic mechanisms may be implicated in mediating the effects of physical activity.
Tobacco smoke
ƒƒ Tobacco smoke effects have been examined through different epigenetic studies, but the results are still under debate.
ƒƒ Smoking during pregnancy has been associated with an increased risk of developing diseases in fetal or later life, through
epigenetic mechanisms.
Alcohol consumption
ƒƒ Alcohol is an antagonist of folate metabolism and may have effects on DNA methylation.
Environmental pollutants
ƒƒ Arsenic
– Hypo/hypermethylation was observed in DNA of blood samples from subjects exposed to toxic level of arsenic.
ƒƒ Air pollution
– Particulate air pollution may affect human health through DNA methylation alterations.
ƒƒ Aromatic hydrocarbons and other organic compounds
– Repetitive element hypomethylation as well as either hyper- or hypo-methylation of specific genes has been reported for benzene
and polycyclic aromatic hydrocarbon exposures.
Psychological stress
ƒƒ DNA methylation is sensitive to environmental stressful exposures early in development and later in life.
Shiftwork
ƒƒ An epigenetic reprogramming of circadian genes, changes in Alu repetitive elements methylation and gene-specific methylation of IFN‑g
and TNF‑a promoters have been observed.

future science group www.futuremedicine.com 273


Review Alegría-Torres, Baccarelli & Bollati

methylation, including changes in Alu repeti- epigenetic mechanisms and lifestyle are modifi-
tive element methylation and gene-specific able, epigeneticists have largely untapped oppor-
methylation of inflammatory genes such as tunities to determine how epigenetic markers are
IFN‑g and TNF‑a [118] . dependent on lifestyle factors and whether and
how much epigenetic mechanisms can be modi-
Conclusion & future perspective fied after positive or negative lifestyle changes are
In the last few years, several investigations have acquired and sustained (Figure 2) . Several of the
examined the relationship between epigenetic results linking lifestyle differences to epigenetics
marks and lifestyle factors, including nutrition, are derived from observational studies. Many of
behavior, stress, physical activity, working hab- the studies cited here should therefore be taken
its, smoking and alcohol consumption. Although as presumptive until results are confirmed and
epigenetic modifications are influenced by the replicated in different settings.
environment, most of these changes tend to be re-
established at each generation; however, this reset- Financial & competing interests disclosure
ting of epigenetic marks might not happen at some This work was funded by New Investigator funding from
loci in the human genome [119,120] . This leaves the HSPH-NIEHS Center for Environmental Health
open the possibility that environmentally induced (ES000002) and Lombardy Region Founding (Effetti sulla
epigenetic changes might impact successive gen- salute degli inquinanti aerodispersi in regione Lombardia
erations, a concept referred to as transgenerational [ESSIA]). The authors have no other relevant affiliations
epigenetic inheritance [121–124] . Epigenetics is or financial involvement with any organization or entity
expected to help explain how gene expression is with a financial interest in or financial conflict with the
modulated by lifestyle and environmental fac- subject matter or materials discussed in the manuscript
tors, and to bring a more complete understand- apart from those disclosed.
ing of individual responses to environmental cues No writing assistance was utilized in the production of
and acquired risk factors (Figure 1) . Because both this manuscript.

plays a regulatory role in the pro-angiogenic 14 Gonzalez S, Huerta JM, Alvarez-Uria J,


Bibliography VEGF-receptor (KDR) gene expression in Fernandez S, Patterson AM, Lasheras C: Serum
Papers of special note have been highlighted as: endothelial cells. J. Physiol. Pharmacol. 60(4), selenium is associated with plasma
n of interest
49–53 (2009). homocysteine concentrations in elderly
nn of considerable interest
8 Nowak J, Weylandt KH, Habbel P et al.: humans. J. Nutr. 134(7), 1736–1740 (2004).
1 APA: Thesaurus of Psychological Index Terms Colitis associated colon tumorigenesis is 15 Johnson IT, Belshaw NJ: Environment, diet
(7th Edition). Gallagher L (Ed.). (2007). suppressed in transgenic mice rich in and CpG island methylation: epigenetic signals
2 Probst AV, Dunleavy E, Almouzni G: endogenous n‑3 fatty acids. Carcinogenesis in gastrointestinal neoplasia. Food Chem.
Epigenetic inheritance during the cell cycle. 28(9), 1991–1995 (2007). Toxicol. 46(4), 1346–1359 (2008).
Nat. Rev. Mol. Cell. Biol. 10(3), 192–206 9 Borek C: Dietary antioxidants and human 16 Kim JM, Hong K, Lee JH, Lee S, Chang N:
(2009). cancer. Integr. Cancer Ther. 3(4), 333–341 Effect of folate deficiency on placental DNA
3 Santos-Reboucas CB, Pimentel MM: (2004). methylation in hyperhomocysteinemic rats.
Implication of abnormal epigenetic patterns 10 Chen J, Xu X: Diet, epigenetic, and cancer J. Nutr. Biochem. 20(3), 172–176 (2009).
for human diseases. Eur. J. Hum. Genet. prevention. Adv. Genet. 71, 237–255 (2010). 17 Giovannucci E: Epidemiologic studies of folate
15(1), 10–17 (2007). and colorectal neoplasia: a review. J. Nutr.
nn Describes epigenetic reprogramming as a
4 Bartsch H, Nair J: Oxidative stress and lipid 132(Suppl. 8), 2350S–2355S (2002).
method of gene regulation through
peroxidation-derived DNA-lesions in 18 Jacob RA, Gretz DM, Taylor PC et al.:
chemoprevention or lifestyle intervention.
inflammation driven carcinogenesis. Cancer Moderate folate depletion increases plasma
Detect. Prev. 28(6), 385–391 (2004). 11 Dashwood RH, Ho E: Dietary histone
homocysteine and decreases lymphocyte DNA
deacetylase inhibitors: from cells to mice to
5 Lawless MW, O’Byrne KJ, Gray SG: methylation in postmenopausal women.
man. Semin. Cancer Biol. 17(5), 363–369
Oxidative stress induced lung cancer and J. Nutr. 128(7), 1204–1212 (1998).
(2007).
COPD: opportunities for epigenetic therapy. 19 Rampersaud GC, Kauwell GP, Hutson AD,
J. Cell. Mol. Med. 13(9A), 2800–2821 12 Druesne N, Pagniez A, Mayeur C et al.:
Cerda JJ, Bailey LB: Genomic DNA
(2009). Diallyl disulfide (DADS) increases histone
methylation decreases in response to moderate
acetylation and p21(waf1/cip1) expression in
6 Arsova-Sarafinovska Z, Eken A, Matevska N folate depletion in elderly women. Am. J. Clin.
human colon tumor cell lines. Carcinogenesis
et al.: Increased oxidative/nitrosative stress Nutr. 72(4), 998–1003 (2000).
25(7), 1227–1236 (2004).
and decreased antioxidant enzyme activities 20 Shin W, Yan J, Abratte CM, Vermeylen F,
in prostate cancer. Clin. Biochem. 42(12), 13 Zingg JM, Jones PA: Genetic and epigenetic
Caudill MA: Choline intake exceeding current
1228–1235 (2009). aspects of DNA methylation on genome
dietary recommendations preserves markers of
expression, evolution, mutation and
7 Kiec-Wilk B, Razny U, Mathers JC, cellular methylation in a genetic subgroup of
carcinogenesis. Carcinogenesis 18(5), 869–882
Dembinska-Kiec A: DNA methylation, folate-compromised men. J. Nutr. 140(5),
(1997).
induced by b­‑carotene and arachidonic acid, 975–980 (2010).

274 Epigenomics (2011) 3(3) future science group


Epigenetics & lifestyle Review
21 Ross SA, Dwyer J, Umar A et al.: 33 Lee WJ, Shim JY, Zhu BT: Mechanisms for 45 Fiala ES, Staretz ME, Pandya GA,
Introduction: diet, epigenetic events and the inhibition of DNA methyltransferases by El-Bayoumy K, Hamilton SR: Inhibition of
cancer prevention. Nutr. Rev. 66(Suppl. 1), tea catechins and bioflavonoids. Mol. DNA cytosine methyltransferase by
S1–S6 (2008). Pharmacol. 68(4), 1018–1030 (2005). chemopreventive selenium compounds,
n Compilation of the symposium on 34 Fang M, Chen D, Yang CS: Dietary determined by an improved assay for DNA
polyphenols may affect DNA methylation. cytosine methyltransferase and DNA
epigenetics, diet and cancer prevention.
J. Nutr. 137(Suppl. 1), 223S–228S (2007). cytosine methylation. Carcinogenesis 19(4),
22 Bravi F, Polesel J, Bosetti C et al.: Dietary 597–604 (1998).
intake of selected micronutrients and the risk 35 Lambert JD, Lee MJ, Diamond L et al.:
Dose-dependent levels of epigallocatechin-3- 46 Davis CD, Uthus EO: Dietary selenite and
of pancreatic cancer: an Italian case–control
gallate in human colon cancer cells and azadeoxycytidine treatments affect
study. Ann. Oncol. 22(1), 202–206 (2011).
mouse plasma and tissues. Drug Metab. dimethylhydrazine-induced aberrant crypt
23 Gonzalez CA, Travier N, Lujan-Barroso L formation in rat colon and DNA
Dispos. 34(1), 8–11 (2006).
et al.: Dietary factors and in situ and invasive methylation in HT‑29 cells. J. Nutr. 132(2),
cervical cancer risk in the European 36 Adlercreutz H, Mazur W: Phyto-oestrogens
292–297 (2002).
prospective investigation into cancer and and Western diseases. Ann. Med. 29(2),
95–120 (1997). 47 Davis CD, Uthus EO, Finley JW: Dietary
nutrition study. Int. J. Cancer (2010) (Epub
selenium and arsenic affect DNA
ahead of print). 37 Qin W, Zhu W, Shi H et al.: Soy isoflavones
methylation in vitro in Caco‑2 cells and
24 Shanmugham JR, Zavras AI, Rosner BA, have an antiestrogenic effect and alter
in vivo in rat liver and colon. J. Nutr.
Giovannucci EL: Alcohol–folate interactions mammary promoter hypermethylation in
130(12), 2903–2909 (2000).
in the risk of oral cancer in women: healthy premenopausal women. Nutr. Cancer
61(2), 238–244 (2009). 48 Gastaldelli A, Basta G: Ectopic fat and
a prospective cohort study. Cancer Epidemiol.
cardiovascular disease: what is the link?
Biomarkers Prev. 19(10), 2516–2524 (2010). 38 Li Y, Liu L, Andrews LG, Tollefsbol TO:
Nutr. Metab. Cardiovasc. Dis. 20(7),
25 Shitara K, Muro K, Ito S et al.: Folate intake Genistein depletes telomerase activity
481–490 (2010).
along with genetic polymorphisms in through cross-talk between genetic and
epigenetic mechanisms. Int. J. Cancer 125(2), 49 Allender S, Rayner M: The burden of
methylenetetrahydrofolate reductase and
286–296 (2009). overweight and obesity-related ill health in
thymidylate synthase in patients with
the UK. Obes. Rev. 8(5), 467–473 (2007).
advanced gastric cancer. Cancer Epidemiol. 39 Cross HS, Kallay E, Lechner D, Gerdenitsch
Biomarkers Prev. 19(5), 1311–1319 (2010). W, Adlercreutz H, Armbrecht HJ: 50 Klein S, Allison DB, Heymsfield SB et al.:
Phytoestrogens and vitamin D metabolism: Waist circumference and cardiometabolic
26 Kim YI: Folate: a magic bullet or a double
a new concept for the prevention and therapy risk: a consensus statement from Shaping
edged sword for colorectal cancer prevention?
of colorectal, prostate, and mammary America’s Health: Association for Weight
Gut 55(10), 1387–1389 (2006).
carcinomas. J. Nutr. 134(5), 1207S–1212S Management and Obesity Prevention;
27 Fini L, Selgrad M, Fogliano V et al.: Annurca NAASO, the Obesity Society; the American
(2004).
apple polyphenols have potent demethylating Society for Nutrition; and the American
activity and can reactivate silenced tumor 40 Xiang N, Zhao R, Song G, Zhong W:
Diabetes Association. Obesity (Silver Spring)
suppressor genes in colorectal cancer cells. Selenite reactivates silenced genes by
15(5), 1061–1067 (2007).
J. Nutr. 137(12), 2622–2628 (2007). modifying DNA methylation and histones in
prostate cancer cells. Carcinogenesis 29(11), 51 Lomba A, Milagro FI, Garcia-Diaz DF,
28 Link A, Balaguer F, Goel A: Cancer Marti A, Campion J, Martinez JA: Obesity
2175–2181 (2008).
chemoprevention by dietary polyphenols: induced by a pair-fed high fat sucrose diet:
promising role for epigenetics. Biochem. 41 Davis CD, Milner J: Frontiers in
methylation and expression pattern of genes
Pharmacol. 80(12), 1771–1792 (2010). nutrigenomics, proteomics, metabolomics
related to energy homeostasis. Lipids Health
and cancer prevention. Mutat. Res. 551(1–2),
29 Paluszczak J, Krajka-Kuzniak V, Malecka Z Dis. 9, 60 (2010).
51–64 (2004).
et al.: Frequent gene hypermethylation in 52 Campion J, Milagro FI, Martinez JA:
laryngeal cancer cell lines and the resistance 42 Huang S: Histone methyltransferases, diet
Individuality and epigenetics in obesity.
to demethylation induction by plant nutrients and tumour suppressors. Nat. Rev.
Obes. Rev. 10(4), 383–392 (2009).
polyphenols. Toxicol. In Vitro 25(1), 213–221 Cancer 2(6), 469–476 (2002).
53 Zhang FF, Cardarelli R, Carroll J et al.:
(2011). n Describes important implications for cancer Physical activity and global genomic DNA
30 Yuasa Y, Nagasaki H, Akiyama Y et al.: DNA treatment with an emphasis on gene-therapy methylation in a cancer-free population.
methylation status is inversely correlated with delivery of methyltransferase genes to Epigenetics 6(3), 293–299 (2011).
green tea intake and physical activity in inhibit tumor growth.
54 Schulz WA, Steinhoff C, Florl AR:
gastric cancer patients. Int. J. Cancer 124(11),
43 Davis CD, Uthus EO: DNA methylation, Methylation of endogenous human
2677–2682 (2009).
cancer susceptibility, and nutrient retroelements in health and disease. Curr.
31 Yuasa Y, Nagasaki H, Akiyama Y et al.: interactions. Exp. Biol. Med. (Maywood) Top. Microbiol. Immunol. 310, 211–250
Relationship between CDX2 gene 229(10), 988–995 (2004). (2006).
methylation and dietary factors in gastric n Review on studies related to cancer- nn Highlights the importance of
cancer patients. Carcinogenesis 26(1),
protective effects associated with bioactive retroelements in cancer and various
193–200 (2005).
food components and DNA methylation. autoimmune diseases.
32 Fang Mz, Wang Y, Ai N et al.: Tea polyphenol
44 Cox R, Goorha S: A study of the mechanism 55 Baccarelli A, Wright R, Bollati V et al.:
(-)-epigallocatechin-3-gallate inhibits DNA
of selenite-induced hypomethylated DNA Ischemic heart disease and stroke in relation
methyltransferase and reactivates
and differentiation of Friend erythroleukemic to blood DNA methylation. Epidemiology
methylation-silenced genes in cancer cell
cells. Carcinogenesis 7(12), 2015–2018 21(6), 819–828 (2010).
lines. Cancer Res. 63(22), 7563–7570 (2003).
(1986).

future science group www.futuremedicine.com 275


Review Alegría-Torres, Baccarelli & Bollati

56 Mcgee SL, Fairlie E, Garnham AP, 68 Maccani MA, Avissar-Whiting M, 80 Baccarelli A, Bollati V: Epigenetics and
Hargreaves M: Exercise-induced histone Banister CE, McGonnigal B, Padbury JF, environmental chemicals. Curr. Opin.
modifications in human skeletal muscle. Marsit CJ: Maternal cigarette smoking during Pediatr. 21(2), 243–251 (2009).
J. Physiol. 587(Pt 24), 5951–5958 (2009). pregnancy is associated with downregulation 81 Chanda S, Dasgupta UB, Guhamazumder
57 Radom-Aizik S, Zaldivar F Jr, Oliver S, of miR‑16, miR‑21 and miR‑146a in the D et al.: DNA hypermethylation of
Galassetti P, Cooper DM: Evidence for placenta. Epigenetics 5(7), 583–589 (2010). promoter of gene p53 and p16 in arsenic-
microRNA involvement in exercise-associated 69 Izzotti A, Larghero P, Longobardi M et al.: exposed people with and without
neutrophil gene expression changes. J. Appl. Dose-responsiveness and persistence of malignancy. Toxicol. Sci. 89(2), 431–437
Physiol. 109(1), 252–261 (2010). microRNA expression alterations induced by (2006).
58 Tommasi S, Kim SI, Zhong X, Wu X, cigarette smoke in mouse lung. Mutat Res. 82 Pilsner JR, Liu X, Ahsan H et al.: Genomic
Pfeifer GP, Besaratinia A: Investigating the (2010) (Epub ahead of print). methylation of peripheral blood leukocyte
epigenetic effects of a prototype smoke-derived 70 Irigaray P, Newby JA, Clapp R et al.: DNA: influences of arsenic and folate in
carcinogen in human cells. PLoS ONE 5(5), Lifestyle-related factors and environmental Bangladeshi adults. Am. J. Clin. Nutr.
e10594 (2010). agents causing cancer: an overview. Biomed. 86(4), 1179–1186 (2007).
59 Marwick JA, Kirkham PA, Stevenson CS Pharmacother. 61(10), 640–658 (2007). 83 Pilsner JR, Liu X, Ahsan H et al.: Folate
et al.: Cigarette smoke alters chromatin nn Provides an analysis of lifestyle and deficiency, hyperhomocysteinemia, low
remodeling and induces proinflammatory environmental factors involved in urinary creatinine, and hypomethylation of
genes in rat lungs. Am. J. Respir. Cell Mol. Biol. carcinogenic processes. leukocyte DNA are risk factors for
31(6), 633–642 (2004). arsenic-induced skin lesions. Environ.
71 Van Engeland M, Weijenberg MP,
60 Van Den Broeck A, Brambilla E, Moro-Sibilot Health Perspect. 117(2), 254–260 (2009).
Roemen GM et al.: Effects of dietary folate
D et al.: Loss of histone H4K20 trimethylation and alcohol intake on promoter methylation 84 Baccarelli A, Cassano PA, Litonjua A et al.:
occurs in preneoplasia and influences in sporadic colorectal cancer: the Netherlands Cardiac autonomic dysfunction: effects
prognosis of non-small cell lung cancer. Clin. cohort study on diet and cancer. Cancer Res. from particulate air pollution and protection
Cancer Res. 14(22), 7237–7245 (2008). 63(12), 3133–3137 (2003). by dietary methyl nutrients and metabolic
61 Toyooka S, Tokumo M, Shigematsu H et al.: polymorphisms. Circulation 117(14),
72 De Vogel S, Bongaerts BW, Wouters KA
Mutational and epigenetic evidence for 1802–1809 (2008).
et al.: Associations of dietary methyl donor
independent pathways for lung intake with MLH1 promoter 85 Brook RD, Franklin B, Cascio W et al.:
adenocarcinomas arising in smokers and never hypermethylation and related molecular Air pollution and cardiovascular disease: a
smokers. Cancer Res. 66(3), 1371–1375 phenotypes in sporadic colorectal cancer. statement for healthcare professionals from
(2006). Carcinogenesis 29(9), 1765–1773 (2008). the Expert Panel on Population and
62 Liu F, Killian JK, Yang M et al.: Epigenomic Prevention Science of the American Heart
73 Hou L, Wang H, Sartori S et al.: Blood
alterations and gene expression profiles in Association. Circulation 109(21), 2655–
leukocyte DNA hypomethylation and gastric
respiratory epithelia exposed to cigarette 2671 (2004).
cancer risk in a high-risk Polish population.
smoke condensate. Oncogene 29(25), Int. J. Cancer 127(8), 1866–1874 (2010). 86 Peters A: Particulate matter and heart
3650–3664 (2010). disease: evidence from epidemiological
74 Marutha Ravindran CR, Ticku MK: Changes
63 Woodson K, Mason J, Choi SW et al.: studies. Toxicol. Appl. Pharmacol.
in methylation pattern of NMDA receptor
Hypomethylation of p53 in peripheral blood 207(Suppl. 2), 477–482 (2005).
NR2B gene in cortical neurons after chronic
DNA is associated with the development of ethanol treatment in mice. Brain Res. Mol. 87 Samet JM, Dominici F, Curriero FC,
lung cancer. Cancer Epidemiol. Biomarkers Brain Res. 121(1–2), 19–27 (2004). Coursac I, Zeger SL: Fine particulate air
Prev. 10(1), 69–74 (2001). pollution and mortality in 20 U.S. cities,
75 Marutha Ravindran CR, Ticku MK: Role of
64 Pogribny IP, Basnakian AG, Miller BJ, 1987–1994. N. Engl. J. Med. 343(24),
CpG islands in the up-regulation of NMDA
Lopatina NG, Poirier LA, James SJ: Breaks in 1742–1749 (2000).
receptor NR2B gene expression following
genomic DNA and within the p53 gene are chronic ethanol treatment of cultured cortical 88 Vineis P, Husgafvel-Pursiainen K: Air
associated with hypomethylation in livers of neurons of mice. Neurochem. Int. 46(4), pollution and cancer: biomarker studies in
folate/methyl-deficient rats. Cancer Res. 55(9), 313–327 (2005). human populations. Carcinogenesis 26(11),
1894–1901 (1995). 1846–1855 (2005).
76 Hicks SD, Middleton FA, Miller MW:
65 Kim YI, Pogribny IP, Basnakian AG et al.: Ethanol-induced methylation of cell cycle 89 Tarantini L, Bonzini M, Apostoli P et al.:
Folate deficiency in rats induces DNA strand genes in neural stem cells. J. Neurochem. Effects of particulate matter on genomic
breaks and hypomethylation within the p53 114(6), 1767–1780 (2010). DNA methylation content and iNOS
tumor suppressor gene. Am. J. Clin. Nutr. promoter methylation. Environ. Health
77 Liu Y, Balaraman Y, Wang G, Nephew KP,
65(1), 46–52 (1997). Perspect. 117(2), 217–222 (2009).
Zhou FC: Alcohol exposure alters DNA
66 Breton CV, Byun HM, Wenten M, Pan F, methylation profiles in mouse embryos at 90 Alexeeff SE, Litonjua AA, Wright RO et al.:
Yang A, Gilliland FD: Prenatal tobacco smoke early neurulation. Epigenetics 4(7), 500–511 Ozone exposure, antioxidant genes, and
exposure affects global and gene-specific DNA (2009). lung function in an elderly cohort: VA
methylation. Am. J. Respir. Crit. Care Med. normative aging study. Occup. Environ.
78 Zhou FC, Zhao Q, Liu Y et al.: Alteration of
180(5), 462–467 (2009). Med. 65(11), 736–742 (2008).
gene expression by alcohol exposure at early
67 Pineles BL, Romero R, Montenegro D et al.: neurulation. BMC Genomics 12, 124–140 91 Baccarelli A, Zanobetti A, Martinelli I
Distinct subsets of microRNAs are expressed (2001). et al.: Air pollution, smoking, and plasma
differentially in the human placentas of homocysteine. Environ. Health Perspect.
79 Bollati V, Baccarelli A: Environmental
patients with preeclampsia. Am. J. Obstet. 115(2), 176–181 (2007).
epigenetics. Heredity 105(1), 105–112 (2010).
Gynecol. 196(3), e261–e266 (2007).

276 Epigenomics (2011) 3(3) future science group


Epigenetics & lifestyle Review
92 Chahine T, Baccarelli A, Litonjua A et al.: 102 Perera F, Tang WY, Herbstman J et al.: 113 Hirayama J, Sahar S, Grimaldi B et al.:
Particulate air pollution, oxidative stress Relation of DNA methylation of 5´-CpG CLOCK-mediated acetylation of BMAL1
genes, and heart rate variability in an elderly island of ACSL3 to transplacental exposure to controls circadian function. Nature 450
cohort. Environ. Health Perspect. 115(11), airborne polycyclic aromatic hydrocarbons and (7172), 1086–1090 (2007).
1617–1622 (2007). childhood asthma. PLoS ONE 4(2), e4488 114 Grimaldi B, Nakahata Y, Kaluzova M,
93 Ehrlich M: DNA hypomethylation in cancer (2009). Masubuchi S, Sassone-Corsi P: Chromatin
cells. Epigenomics 1(2), 239–259 (2009). 103 Rusiecki JA, Baccarelli A, Bollati V, remodeling, metabolism and circadian clocks:
n Review that discusses the active Tarantini L, Moore LE, Bonefeld-Jorgensen the interplay of CLOCK and SIRT1. Int.
EC: Global DNA hypomethylation is J. Biochem. Cell Biol. 41(1), 81–86 (2009).
demethylation of DNA and the relationship
associated with high serum-persistent organic 115 Costa G: The problem: shiftwork.
of cancer-associated DNA hypomethylation.
pollutants in Greenlandic Inuit. Environ. Chronobiol. Int. 14(2), 89–98 (1997).
94 Castro R, Rivera I, Struys EA et al.: Increased Health Perspect. 116(11), 1547–1552 (2008).
homocysteine and S‑adenosylhomocysteine 116 Zhu Y, Zheng T, Stevens RG, Zhang Y,
104 Jirtle RL, Skinner MK: Environmental Boyle P: Does ‘clock’ matter in prostate
concentrations and DNA hypomethylation in
epigenomics and disease susceptibility. Nat. cancer? Cancer Epidemiol. Biomarkers Prev.
vascular disease. Clin. Chem. 49(8),
Rev. Genet. 8(4), 253–262 (2007). 15(1), 3–5 (2006).
1292–1296 (2003).
nn Review about heritable epigenetic 117 Sahar S, Sassone-Corsi P: Circadian clock and
n Proposes epigenetic markers of
modifications triggered by the environment. breast cancer: a molecular link. Cell Cycle
vascular disease.
105 Miller CA, Sweatt JD: Covalent modification 6(11), 1329–1331 (2007).
95 Bollati V, Marinelli B, Apostoli P et al.:
of DNA regulates memory formation. Neuron 118 Bollati V, Baccarelli A, Sartori S et al.:
Exposure to metal-rich particulate matter
53(6), 857–869 (2007). Epigenetic effects of shiftwork on blood DNA
modifies the expression of candidate
microRNAs in peripheral blood leukocytes. 106 Szyf M, McGowan P, Meaney MJ: The social methylation. Chronobiol. Int. 27(5),
Environ. Health Perspect. 118(6), 763–768 environment and the epigenome. Environ. Mol. 1093–1104 (2010).
(2010). Mutagen. 49(1), 46–60 (2008). 119 Morgan DK, Whitelaw E: The case for
96 Snyder R: Benzene and leukemia. Crit. Rev. nn Suggests a hypothesis that links the social transgenerational epigenetic inheritance in
Toxicol. 32(3), 155–210 (2002). environment early in life and long-term humans. Mamm. Genome 19(6), 394–397
epigenetic programming of behavior and (2008).
97 Bollati V, Baccarelli A, Hou L et al.: Changes
in DNA methylation patterns in subjects responsiveness to stress and health status later 120 Chao MJ, Ramagopalan SV, Herrera BM
exposed to low-dose benzene. Cancer Res. in life. et al.: Epigenetics in multiple sclerosis
67(3), 876–880 (2007). susceptibility: difference in transgenerational
107 Miller CA, Gavin CF, White JA et al.: Cortical
risk localizes to the major histocompatibility
98 Costantini AS, Benvenuti A, Vineis P et al.: DNA methylation maintains remote memory.
complex. Hum. Mol. Genet. 18(2), 261–266
Risk of leukemia and multiple myeloma Nat. Neurosci. 13(6), 664–666 (2010).
(2009).
associated with exposure to benzene and other 108 Murgatroyd C, Patchev AV, Wu Y et al.:
organic solvents: evidence from the Italian 121 Daxinger L, Whitelaw E: Transgenerational
Dynamic DNA methylation programs
multicenter case–control study. Am. J. Ind. epigenetic inheritance: more questions than
persistent adverse effects of early-life stress.
Med. 51(11), 803–811 (2008). answers. Genome Res. 20(12), 1623–1628
Nat. Neurosci. 12(12), 1559–1566 (2009).
(2010).
99 Kirkeleit J, Riise T, Bratveit M, Moen BE: 109 McGowan PO, Sasaki A, D’Alessio AC et al.:
Increased risk of acute myelogenous leukemia 122 Nelson VR, Nadeau JH: Transgenerational
Epigenetic regulation of the glucocorticoid
and multiple myeloma in a historical cohort genetic effects. Epigenomics 2(6), 797–806
receptor in human brain associates with
of upstream petroleum workers exposed to (2010).
childhood abuse. Nat. Neurosci. 12(3),
crude oil. Cancer Causes Control 19(1), 13–23 342–348 (2009). 123 Skinner MK, Guerrero-Bosagna C:
(2008). Environmental signals and transgenerational
110 Weaver IC: Epigenetic programming by
100 Ji Z, Zhang L, Peng V, Ren X, McHale CM, epigenetics. Epigenomics 1(1), 111–117
maternal behavior and pharmacological
Smith MT: A comparison of the cytogenetic (2009).
intervention. Nature versus nurture: let’s call
alterations and global DNA hypomethylation the whole thing off. Epigenetics 2(1), 22–28 n Review of how environmental and toxicant
induced by the benzene metabolite, (2007). factors can promote epigenetic
hydroquinone, with those induced by transgenerational phenotypes.
n Description of epigenetic plasticity.
melphalan and etoposide. Leukemia 24(5),
111 Weaver IC, Cervoni N, Champagne FA et al.: 124 Skinner MK, Manikkam M, Guerrero-
986–991 (2010).
Epigenetic programming by maternal behavior. Bosagna C: Epigenetic transgenerational
101 Pavanello S, Bollati V, Pesatori AC et al.: actions of environmental factors in disease
Nat. Neurosci. 7(8), 847–854 (2004).
Global and gene-specific promoter etiology. Trends Endocrinol. Metab. 21(4),
methylation changes are related to anti-B[a] 112 Nakahata Y, Grimaldi B, Sahar S, Hirayama J,
214–222 (2010).
PDE-DNA adduct levels and influence Sassone-Corsi P: Signaling to the circadian
micronuclei levels in polycyclic aromatic clock: plasticity by chromatin remodeling.
hydrocarbon-exposed individuals. Int. Curr. Opin. Cell Biol. 19 (2), 230–237
J. Cancer 125(7), 1692–1697 (2009). (2007).

future science group www.futuremedicine.com 277

View publication stats

You might also like