ArticlePDF Available

Abstract and Figures

Background Omega-3 polyunsaturated fatty acids (n-3 FAs) have several beneficial effects on cardiovascular (CV) disease risk factors. These effects on CV risk profile may be mediated by several factors, including epigenetic modifications. Our objective is to investigate, using genome-wide DNA methylation analyses, methylation changes following an n-3 FA supplementation in overweight and obese subjects and to identify specific biological pathways potentially altered by the supplementation. Results Blood leukocytes genome-wide DNA methylation profiles of 36 overweight and obese subjects before and after a 6-week supplementation with 3 g of n-3 FAs were compared using GenomeStudio software. After supplementation, 308 CpG sites, assigned to 231 genes, were differentially methylated (FDR-corrected Diffscore ≥│13│~ P ≤ 0.05). Using Ingenuity Pathway Analysis system, a total of 55 pathways were significantly overrepresented following supplementation. Among these pathways, 16 were related to inflammatory and immune response, lipid metabolism, type 2 diabetes, and cardiovascular signaling. Changes in methylation levels of CpG sites within AKT3, ATF1, HDAC4, and IGFBP5 were correlated with changes in plasma triglyceride and glucose levels as well as with changes in the ratio of total cholesterol/HDL-cholesterol following the supplementation. Conclusions These data provide key differences in blood leukocytes DNA methylation profiles of subjects following an n-3 FA supplementation, which brings new, potential insights on metabolic pathways underlying the effects of n-3 FAs on CV health. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0345-3) contains supplementary material, which is available to authorized users.
Content may be subject to copyright.
R E S E A R C H Open Access
Epigenetic changes in blood leukocytes
following an omega-3 fatty acid
supplementation
Bénédicte L. Tremblay
1
, Frédéric Guénard
1
, Iwona Rudkowska
2
, Simone Lemieux
1
, Patrick Couture
1,2
and Marie-Claude Vohl
1,2*
Abstract
Background: Omega-3 polyunsaturated fatty acids (n-3 FAs) have several beneficial effects on cardiovascular (CV)
disease risk factors. These effects on CV risk profile may be mediated by several factors, including epigenetic
modifications. Our objective is to investigate, using genome-wide DNA methylation analyses, methylation changes
following an n-3 FA supplementation in overweight and obese subjects and to identify specific biological pathways
potentially altered by the supplementation.
Results: Blood leukocytes genome-wide DNA methylation profiles of 36 overweight and obese subjects before and
after a 6-week supplementation with 3 g of n-3 FAs were compared using GenomeStudio software. After
supplementation, 308 CpG sites, assigned to 231 genes, were differentially methylated (FDR-corrected Diffscore
13~P0.05). Using Ingenuity Pathway Analysis system, a total of 55 pathways were significantly
overrepresented following supplementation. Among these pathways, 16 were related to inflammatory and immune
response, lipid metabolism, type 2 diabetes, and cardiovascular signaling. Changes in methylation levels of CpG
sites within AKT3,ATF1,HDAC4, and IGFBP5 were correlated with changes in plasma triglyceride and glucose levels
as well as with changes in the ratio of total cholesterol/HDL-cholesterol following the supplementation.
Conclusions: These data provide key differences in blood leukocytes DNA methylation profiles of subjects
following an n-3 FA supplementation, which brings new, potential insights on metabolic pathways underlying the
effects of n-3 FAs on CV health.
Keywords: DNA methylation, Omega-3 fatty acids, Microarray, Metabolic pathways, Blood leukocytes
Background
Fish-oil-derived long-chain omega-3 fatty acids (n-3
FAs), including eicosapentaenoic acid (EPA, 20:5 n-3)
and docosahexaenoic acid (DHA, 22:6 n-3) have several
benefits on cardiovascular (CV) health. They exert hypo-
triglyceridemic [1, 2], anti-inflammatory [35], anti-
arrhythmic [6, 7], and anti-thrombotic effects [8, 9].
Many factors, including genetic and epigenetic factors,
may contribute to the observed effects of n-3 FAs on the
CV risk profile. Indeed, emerging evidence suggests that
n-3 FAs might influence global DNA methylation
patterns due to their role in one-carbon metabolism
[10]. A study in rats fed on a vitamin B
12
-deficient diet
demonstrated that DHA modify DNA methylation, indi-
cating that it plays a role in one-carbon metabolism [11].
DNA methylation is the best-characterized epigenetic
factor and consists of the methylation of cytosine residues,
mainly at cytosine-phosphate-guanine (CpG) dinucleo-
tides [12]. The modification of DNA methylation by the
environment may influence the regulation of CV risk fac-
tors, such as hypertension [13], atherosclerosis [14, 15],
and inflammation [16]. Moreover, the methylation of re-
petitive sequences in blood has also been associated with
CV diseases in epidemiological studies [17, 18].
Only a few studies have dealt with the impact of n-3
FAs on DNA methylation in human subjects. These
* Correspondence: marie-claude.vohl@fsaa.ulaval.ca
1
Institute of Nutrition and Functional Foods (INAF), Laval University, 2440
Hochelaga Blvd, Quebec, QC G1V 0A6, Canada
2
CHU de Québec Research Center Endocrinology and Nephrology, 2705
Laurier Blvd, Quebec, QC G1V 4G2, Canada
© The Author(s). 2017 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Tremblay et al. Clinical Epigenetics (2017) 9:43
DOI 10.1186/s13148-017-0345-3
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
studies were conducted in various populations including
children and adolescents [19, 20], adults with renal im-
pairment [21], women under caloric restriction [22], and
Yupik Alaska Native individuals [23]. Moreover, a recent
study demonstrated that a DHA supplementation during
pregnancy was associated with changes in global methy-
lation levels of inflammatory mediated genes [24].
The aim of this study was to investigate DNA methyla-
tion changes following n-3 FA supplementation in
overweight and obese subjects and identify biological
pathways potentially altered by the n-3 FA supplementa-
tion, by using whole-genome DNA methylation analyses.
To our knowledge, this is the first study to examine the
possible effect of n-3 FA supplementation on genome-
wide DNA methylation levels in blood leukocytes of
overweight and obese adults.
Results
Effects of n-3 FA supplementation
Biochemical parameters of study subjects (n= 36) before
and after the n-3 FA supplementation are presented in
Table 1. The supplementation was associated with a de-
crease in fasting plasma triglyceride (TG) concentra-
tions, similar to results reported in full cohort [25]. In
the same manner, total cholesterol (TC) and the ratio TC/
high-density lipoprotein cholesterol (HDL-C) decreased
whereas glucose concentrations slightly increased after the
supplementation (Table 1). As expected, the supplementa-
tion was associated with a decrease in percentage and ab-
solute values of linoleic, arachidonic, and total n-6 FAs
(including all cis and trans n-6 FAs) in plasma phospho-
lipids (Pvalue <0.0001 for all, data not shown). It was also
associated with an increase in percentage and absolute
values of EPA, DHA, ratio n-3/n-6, and total n-3 FAs (in-
cluding all cis and trans n-3 FAs) in plasma phospholipids
(Pvalue <0.0001 for all, data not shown).
Genome-wide DNA methylation analyses
Globally, 484,027 of the 485,577 probes (99.7%) on the
array were detected with a detection Pvalue 0.05. After
n-3 FA supplementation, 308 CpG sites, assigned to 231
genes, were differentially methylated (false discovery rate
(FDR)-corrected DiffScore 13~P0.05). A total
of 286 CpG sites were hypermethylated (93%) and 22
were hypomethylated (7%) after supplementation as
compared to levels before supplementation (Table 2). A
total of 36.4% of significant differentially methylated
CpG sites were located in gene bodies (Table 2). The
genomic localization of CpG sites is summarized in
Table 2. Detailed information about the 308 differentially
methylated CpG sites is presented in Additional file 1.
Relationship between CpG sites and surrounding SNPs
Using results from a recent GWAS done by our group
in the same cohort [26], we tested potential relationship
between pre-supplementation methylation levels and
changes in methylation levels (Δmethylation) of the 308
differentially methylated CpG sites and surrounding
single-nucleotide polymorphisms (SNPs) (±1 kb). Single-
Table 1 Biochemical parameters of subjects before and after n-3
FA supplementation (n=36)
Before n-3 suppl. After n-3 suppl. Pvalue
Gender 18 men and 18 women
Age (years) 34.7 ± 8.8
BMI (kg/m
2
) 29.2 ± 3.65 29.2 ± 3.83 0.24
Triglycerides
a
(mmol/L) 1.42 ± 0.80 1.24 ± 0.65 0.0034
Cholesterol (mmol/L)
Total 5.24 ± 0.9 5.12 ± 0.92 0.048
LDL-C 3.18 ± 0.91 3.10 ± 0.92 0.19
HDL-C 1.40 ± 0.35 1.44 ± 0.40 0.061
Ratio TC/HDL-C 4.0 ± 1.31 3.85 ± 1.35 0.018
Glucose (mmol/L) 4.86 ± 0.51 5.06 ± 0.47 0.0085
Insulin
a
(pmol/L) 108.0 ± 143.5 91.7 ± 48.4 0.70
CRP
a
(mg/L) 3.89 ± 6.57 3.24 ± 5.32 0.18
Data are shown as mean ± SD
Abbreviations:BMI body mass index, CRP C-reactive protein, HDL-C high-
density lipoprotein cholesterol, LDL-C low-density lipoprotein cholesterol, suppl
supplementation, TC total cholesterol
a
Pvalue derived from log
10
transformed data
Table 2 Summary of methylation results
n
Probes 485 577
Number of probes detected (P0.05) 484 027
Differentially methylated (FDR-corrected DiffScore ǀ13ǀ) 308
Hypermethylated after n-3 FA supplementation 286
Gene body 107
3-UTR 34
1st exon 4
5-UTR 19
Promoter region
a
50
Intergenic region 72
Hypomethylated after n-3 FA supplementation 22
Gene body 5
3-UTR 0
1st exon 1
5-UTR 5
Promoter region
a
6
Intergenic region 5
Localization according to the first annotated transcript for each CpG site and
provided for the Infinium HumanMethylation450 BeadChip
Abbreviations:FDR false discovery rate, UTR untranslated region, TSS
transcription start site
a
Promoter region includes TSS1500 and TSS200
Tremblay et al. Clinical Epigenetics (2017) 9:43 Page 2 of 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
nucleotide polymorphisms (SNPs) had an effect on pre-
supplementation methylation levels of 15 CpG sites.
Moreover, SNPs only affected Δmethylation of two CpG
sites. Indeed, rs41286653 and rs899388 affected Δmethy-
lation of cg02296904 located in the body of the gene
GAK (chromosome 4). The SNP rs114329043 also
affected Δmethylation of cg27270362 located in an inter-
genic region on chromosome 10. We can conclude that
the potential effect of the n-3 FA supplementation on
Δmethylation of the 308 CpG sites is due in very small
proportion to surrounding SNPs.
Pathway analyses
From the 231 differentially methylated genes following
supplementation, Ingenuity Pathway Analysis (IPA)
mapped 227 genes. IPA revealed 55 pathways that were
significantly overrepresented (P0.05) (detailed path-
ways are presented in the Additional file 2), but we fo-
cused on the 16 pathways related to CV health. Selected
CV health-related pathways, associated Pvalues, and dif-
ferentially methylated genes identified in pathways are
presented in Table 3. These selected pathways revealed
relevant genes known to be related to inflammatory and
immune response (AKT3,ATF1,BAX,CASP6,DHRS9,
FAS,PRKAG2,PRKCZ,PRKD3,PTEN,TRIM24), lipid
metabolism (AKT3,IGFBP5,KLK6,NUDT3,PLCH1,
PPP2RE5,PTEN,PTPN12,PRKAG2,PRKCZ,PRKD3,
SLCO1B3), type 2 diabetes (T2D) (AKT3,PRKAG2,
PRKCZ,PRKD3), and CV signaling (AKT3,HDAC4,
PRKAG2,PRKCZ,PRKD3).
DNA methylation and biochemical parameters
We further investigated the possible relationship be-
tween Δmethylation of these 19 genes related to CV
health and changes in the four biochemical parameters
modified by the n-3 FA supplementation (ΔTG, ΔTC,
ΔTC/HDL-C, and ΔGlucose). As shown in Table 4,
Δmethylation of cg00011856 (IGFBP5) was positively
correlated with ΔTG (r= 0.39, p= 0.023) while Δmethy-
lation of cg05655647 (ATF1) was negatively correlated
with ΔTG (r=0.35, p= 0.047). The Δmethylation of
cg00011856 (IGFBP5) and cg24455383 (AKT3) were
positively correlated with ΔTC/HDL-C (r= 0.36, p=
0.042 and r= 0.42, p= 0.016, respectively). The Δmethy-
lation of cg15656521 (HDAC4) was positively correlated
with ΔGlucose (r= 0.35, p= 0.043). There was no signifi-
cant correlation with ΔTC. After adjustments for age,
sex, and body mass index (BMI), the correlation between
Δmethylation of cg00011856 and ΔTG remained signifi-
cant (r= 0.42, p= 0.020) as well as the one between
Δmethylation of cg24455383 and ΔTC/HDL-C (r= 0.40,
p= 0.031).
Using previous data on gene expression in the same
cohort [27], we performed further analyses to test
whether correlations between Δmethylation and changes
in biochemical parameters may potentially be attribut-
able to changes in expression levels (Δexpression). We
first correlated Δmethylation and Δexpression for these
four genes (ATF1,AKT3,HDAC4, and IGFBP5). ATF1
and HDAC4 showed significant correlation between
Δmethylation and Δexpression after adjustments for the
Table 3 CV health-related overrepresented pathways identified from differential methylation analysis following an n-3 FA
supplementation
IPA canonical pathways Pvalue Differentially methylated genes
Tumoricidal function of hepatic natural killer cells
a
0.0017 BAX,CASP6,FAS
RAR activation
a
0.0035 AKT3,DHRS9,PRKAG2,PRKCZ,PRKD3,PTEN,TRIM24
VDR/RXR activation
b
0.0078 IGFBP5,KLK6,PRKCZ,PRKD3
Fcγreceptor-mediated phagocytosis in macrophages and monocytes
a
0.017 AKT3,PRKCZ,PRKD3,PTEN
D-myo-inositol-5-phosphate metabolism
b
0.018 NUDT3,PLCH1,PPP2R5E,PTEN,PTPN12
Nitric oxide signaling in the cardiovascular system
c
0.026 AKT3,PRKAG2,PRKCZ,PRKD3
IL-3 signaling
a
0.033 AKT3,PRKCZ,PRKD3
PXR/RXR activation
b
0.035 AKT3,SLCO1B3,PRKAG2
LPS-stimulated MAPK signaling
a
0.035 ATF1,PRKCZ,PRKD3
NF-кB activation by viruses
a
0.037 AKT3,PRKCZ,PRKD3
CCR5 signaling in nacrophages
a
0.037 FAS,PRKCZ,PRKD3
Role of NFAT in cardiac hypertrophy
c
0.038 AKT3,HDAC4,PRKAG2,PRKCZ,PRKD3
P2Y purigenic receptor signaling pathway
c
0.040 AKT3,PRKAG2,PRKCZ,PRKD3
Cytotoxic T lymphocyte-mediated apoptosis of target cells
a
0.040 FAS,CASP6
PI3K signaling in B lymphocytes
a
0.044 AKT3,ATF1,PRKCZ,PTEN
Type II diabetes mellitus signaling
d
0.049 AKT3,PRKAG2,PRKCZ,PRKD3
Pathways related to the following:
a
Inflammatory and immune response (n= 9);
b
Lipid metabolism (n= 3);
c
Cardiovascular signaling (n= 3);
d
Diabetes (n=1)
Tremblay et al. Clinical Epigenetics (2017) 9:43 Page 3 of 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
effects of age, sex, and BMI (r=0.44, p= 0.02 and r=
0.47, p= 0.01). We then correlated Δexpression of ATF1
and HDAC4 with ΔTG and ΔGlucose, respectively.
However, we found no significant association.
Discussion
The aim of this study was to investigate DNA methyla-
tion changes following n-3 FA supplementation and
identify potentially altered biological pathways. We first
observed that a 6-week supplementation with 3 g of n-3
FAs per day was associated with a decrease in n-6 FAs
and an increase in n-3 FAs in plasma phospholipids of
36 overweight and obese adults. The n-3 FA supplemen-
tation also decreased plasma TG, TC, and the ratio TC/
HDL-C, while increasing plasma glucose concentrations.
The slight increase in glucose concentrations was similar
to the one reported in the entire cohort [25]. Studies
have reported conflicting results on the effect of n-3 FAs
on blood glucose concentrations [28]. There is a large
variability in plasma glucose response to n-3 FAs with
effects varying from 1.61 mmol/L net reduction to
1.4 mmol/L net increase [29].
Using a genome-wide methylation analysis, we identi-
fied differences in CpG sites methylation levels following
an n-3 FA supplementation. As previously mentioned,
only few studies were conducted on the impact of n-3
FAs on DNA methylation in humans [1924, 30]. Our
results are in agreement with these studies suggesting
that EPA and DHA can modulate DNA methylation
levels. Moreover, we identified overrepresented pathways
from differentially methylated genes. More precisely,
pathway analysis revealed 16 overrepresented pathways
related to CV health. These results are in line with pre-
viously reported effects of n-3 FAs and DNA methyla-
tion on CV health. Indeed, n-3 FAs have beneficial
effects on CV risk factors [19], even if they are not
associated with CV disease events [31]. Moreover, epi-
demiological studies have reported the association be-
tween global DNA methylation levels and prevalence of
CV diseases [17, 18].
Among the 16 pathways related to CV health, nine
were related to inflammatory and immune response
which may be in line with potential anti-inflammatory
effects of n-3 FAs, more particularly DHA [5, 32]. Stud-
ies have also reported the link between DNA methyla-
tion and inflammation. A recent study in the GOLDN
study and the ENCODE consortium reported that higher
erythrocyte total n-3 FAs was associated with lower
cg01770232 methylation (IL-6) and lower plasma IL-6
concentration [30]. The hypomethylation of long-
interspersed element-1 was also associated with higher
serum vascular cell adhesion molecule-1 in elderly men
[33]. Moreover, three pathways were related to lipid me-
tabolism, which is in accordance with known hypotrigly-
ceridemic effects of n-3 FAs [1]. Methylation levels of
one CpG site in APOE and one in ABCA1 were nega-
tively associated with plasma TG and HDL-C levels, re-
spectively, in the GOLDN study [34, 35]. DNA
methylation is also implicated in the regulation of ath-
erosclerosis. Indeed, genome-wide DNA methylation
changes (mainly hypermethylation) occur during the on-
set and progression of atherosclerotic lesions in humans
[36, 37]. A total of three pathways were related to car-
diovascular signaling. Indeed, reported antiplatelet ef-
fects of n-3 FAs [38] are consistent with the
overrepresentation of the nitric oxide signaling in the
cardiovascular system pathway identified herein. Finally,
one pathway was related to T2D, which may be in line
with potential, but yet controversial, effects of n-3 FAs
on glucose homeostasis [39]. Data mining analysis sug-
gests a role of epigenetic factors in the pathogenesis of
T2D [40]. Differential methylation profiles in pancreatic
islets from T2D and non-diabetic subjects were also
identified thus suggesting a role of DNA methylation in
pathogenesis of T2D [41]. Moreover, blood DNA methy-
lation of some CpG sites have been associated with
blood glucose concentrations in an epigenome-wide as-
sociation study [42]. All together, these finding suggest a
possible link between n-3 FAs, DNA methylation, and
CV risk factors.
Among differentially methylated genes, some were also
of particular interest in the field of CV health. For ex-
ample, a genetic variation in the FAS gene was associ-
ated with an increased occurrence of myocardial
Table 4 Significant correlations between changes in biochemical parameters and changes in methylation levels following an n-3 FA
supplementation
CpG site ID (gene, position
a
)ΔTriglyceride ΔTC/HDL-C ΔGlucose
Δcg00011856 (IGFBP5, Chr2:217560946) 0.39 (0.023)
b
0.36 (0.042)
Δcg05655647 (ATF1, Chr12:51157023) -0.35 (0.047) ––
Δcg24455383 (AKT3, Chr1:243736307) 0.42 (0.016)
b
Δcg15656521 (HDAC4, Chr2:239970617) ––0.35 (0.043)
Results are R(Pvalue)
Abbreviations:HDL-C high-density lipoprotein cholesterol, TC total cholesterol
a
All positions are from the Genome Build 37
b
Correlation remains significant after adjustments for age, sex, and body mass index
Tremblay et al. Clinical Epigenetics (2017) 9:43 Page 4 of 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
infarction in Japanese subjects [43]. Interestingly, FAS
methylation levels were significantly associated with n-3
FA intakes in Yupik Alaska Native individuals [23].
Moreover, a total of four CpG sites showed correlations
between changes in their methylation levels and changes
in biochemical parameters following the supplementa-
tion. First, Δmethylation of cg15656521 in HDAC4 gene
was positively correlated with Δglucose. HDAC4 gene
encodes for a histone deacetylase 4 that is a signal-
dependent modulator of transcription with role in
muscle differentiation and neuronal survival [44].
HDAC4 methylation was inversely associated with n-3
FAs in whole blood of men [45]. HDAC4 also downregu-
lates GLUT4 transcription in cultured adipocytes and
fasting mice [46]. Interestingly, Benton et al. reported a
robust inverse correlation between changes in fasting
glucose and changes in methylation of HDAC4
(cg26078407) in subcutaneous fat [47]. However, they
did not test for association with cg15656521 since it was
not differentially methylated in their cohort after gastric
bypass surgery [47]. Second, Δmethylation of cg00011856
in IGFBP5 gene was positively correlated with ΔTG (even
after adjustments for age, sex, and BMI) and with ΔTC/
HDL-C. IGFBP5 gene encodes for insulin-like growth fac-
tor binding protein 5 [48]. Young patients with coronary
heart disease have significantly higher serum IGFBP5 than
age- and BMI-matched controls [49]. A study in arthritic
rats demonstrated that EPA increases IGFBP5 mRNA in
the gastrocnemius muscle [50]. Another study found
higher IGFBP5 expression in the liver of beef fed with n-3
FAs compared to control diet [51]. Unfortunately, we can-
not verify if the n-3 FA supplementation increases IGFBP5
expression in our sample since it is not expressed in blood
[52], but it would be interesting to look at its expression
in the liver after an n-3 FA supplementation. Third,
Δmethylation of cg24455383 in AKT3 was positively cor-
related with ΔTC-HDL-C, even after adjustments. AKT3
gene encodes for an AKTserine/threonine kinase 3 stimu-
lated by insulin and growth factors [53, 54]. It is likely
involved in insulin-stimulated glucose transport in the hu-
man skeletal muscles [55]. AKT3 also has anti-atherogenic
properties due to its capacity to inhibit macrophages foam
cells formation by reducing lipoprotein uptake and pro-
moting ACAT-1 degradation [56, 57]. AKT signaling has
also been shown to regulate lipid metabolism through
phosphorylation and inhibition of GSK3 [58]. The associ-
ation between changes in AKT3 methylation and changes
in TC/HDL-C ratio reported herein may be plausible since
AKT3 seems to be involved in lipid metabolism. Finally,
Δmethylation of cg05655647 in ATF1 gene was negatively
correlated with ΔTG. The ATF 1 gene encodes for the acti-
vating transcription factor 1 that leads to the production
of atheroprotective macrophages [59, 60]. SNPs within
this gene were also associated with an increased risk of
essential hypertension in a case-control study [61]. Data
are insufficient to propose a mechanism of action between
methylation of ATF1 and plasma TG levels. Nevertheless,
we could hypothesize that methylation of these genes may
play a role in the effects of n-3 FAs on CV health since
they are modulated by n-3 FAs, they have been associated
with CV health in pathway analysis, and they are corre-
lated with changes in biochemical parameters. However,
we acknowledge that our study design does not allow us
to prove or investigate causality between n-3 FA supple-
mentation, DNA methylation, and CV disease risk factors.
Moreover, we were not able to investigate the potential
link between Δmethylation of AT F 1 ,AKT3,IGFBP5,and
HDAC4 and changes in biochemical parameters using
Δexpression. At this time, we cannot rule out the pos-
sibility that the small sample size or low Δexpression
values of these genes limit our statistical power to de-
tect significant associations. This possible link does
not seem to be due either to surrounding SNPs since
they affected Δmethylation of only two CpG sites
among the 308 CpG sites.
The present study has some limitations. DNA methy-
lation levels are specific to the type of cell and tissue
[62]. These methylation profiles in blood leukocytes
might not represent DNA methylation in other tissues
even though these patterns are globally conserved across
tissues [6365]. Pathway analysis has certain methodo-
logical considerations. As part of Genetic Analysis
Workshop 18, seven research groups raised the fact that
annotation of genetic variants is inconsistent across da-
tabases, incomplete and biased toward known genes
[66]. Moreover, insufficient statistical power is an issue
in pathway analyses [66]. Thus, these results need to be
validated in larger and independent studies considering
that replication remains the gold standard to establish
validity of the findings. Finally, the sample size is rela-
tively small so these results need to be validated in lar-
ger, independent studies.
Conclusions
In conclusion, the present data provide key differences
in blood leukocytes DNA methylation levels of subjects
following n-3 FA supplementation, which provides new,
potential insights on novel genes and metabolic path-
ways underlying the effects of n-3 FAs on the CV risk
profile. Further studies in larger, independent samples
are required to unveil potential functional mechanisms
underlying metabolic improvements with n-3 FA
supplementation.
Methods
Study population and study design
The present study is based on a subsample of a larger
intervention study that aimed at studying the inter-
Tremblay et al. Clinical Epigenetics (2017) 9:43 Page 5 of 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
individual variability in TG response to an n-3 FA sup-
plementation as previously described [2]. A total of 254
subjects from the greater Quebec City metropolitan area
(Canada) were recruited between September 2009 and
December 2011. In total, 210 subjects completed the
intervention. Non-smoker subjects, aged between 18 and
50, with a BMI between 25 and 40 kg/m
2
, and not taking
any lipid-lowering medication were included. Subjects
were excluded if they had taken n-3 FA supplementation
at least 6 months prior to the intervention or had been
diagnosed with any metabolic disorder. A subset of 36
subjects who first completed the study (equal proportion
of men and women) was selected for the purpose of this
genome-wide DNA methylation analysis.
First, subjects received dietary instructions by a trained
registered dietician to achieve recommendations from
Canadas Food Guide during a 2-week run-in period
[67]. After this period, subjects receive a bottle contain-
ing fish oil capsules needed for the following 6 weeks.
They were invited to take five capsules per day providing
a total of 3 g of n-3 FAs (including 1.92.2 g of EPA and
1.1 g of DHA) per day. Compliance was assessed by the
count of remaining capsules and by measuring EPA and
DHA in plasma phospholipids. Details on the study de-
sign have been described elsewhere [2]. The experimen-
tal protocol was approved by the Ethics Committees of
Laval University Hospital Research Center and Laval
University. This trial was registered at clinicaltrials.gov
as NCT01343342.
Anthropometric and metabolic measurements
Body weight, waist girth, and height were measured ac-
cording to the procedures recommended by the Airlie
Conference [68] and were taken before the run-in period
as well as before and after the supplementation period.
Blood samples were collected from an antecubital vein
into vacutainer tubes containing EDTA after 12-h over-
night fast and 48-h alcohol abstinence before the run-in
period to identify and exclude participants with meta-
bolic disorders. Afterwards, the selected participants had
blood samples taken before and after the supplementa-
tion period. Plasma was separated by centrifugation
(2500 g for 10 min at 4 °C), and samples were aliquoted
and frozen (80 °C) for subsequent analyses. Enzymatic
assays were used to measure plasma TC and TG concen-
trations [69, 70]. Precipitation of very-low density lipo-
protein (VLDL) and low-density lipoprotein (LDL)
particles in the infranatant with heparin manganese
chloride generated the HDL-C fraction [71]. LDL choles-
terol (LDL-C) was calculated with the Friedewald for-
mula [72]. Using a sensitive assay, plasma C-reactive
protein (CRP) was measured by nephelometry (Prospec
equipment Behring) [73].
DNA extraction and DNA methylation analysis
We extracted genomic DNA from blood leukocytes
using the GenElute Blood Genomic DNA kit (Sigma-Al-
drich, St. Louis, MO, USA) for the 72 samples: 36 sam-
ples before and 36 samples after the supplementation.
DNA was quantified using both NanoDrop Spectropho-
tometer (Thermo Scientific, Wilmington, DE, USA) and
PicoGreen DNA methods. McGill University and Gen-
ome Quebec Innovation Center (Montreal, QC, Canada)
conducted the bisulfite conversion and quantitative
DNA methylation analysis using Infinium Human-
Methylation450 array (Illumina, San Diego, CA, USA).
Three samples (one sample before and two samples after
supplementation) were excluded from microarray ana-
lysis following quality control steps (bisulfite conversion,
extension, staining, hybridization, target removal, nega-
tive and nonpolymorphic control probes).
We used Illumina GenomeStudio software version
2011.1 and the Methylation Module to analyze methyla-
tion data on 485,577 CpG sites. Methylation levels (vary-
ing from 0 to 1) were estimated as the proportion of
total signal intensity from methylated-specific probe. To
avoid false positives, probes with a detection Pvalue
>0.05 in more than 10% of samples were removed.
Probes on the X and Y chromosomes were also removed
to eliminate gender bias. Thus, 472,715 probes were
considered in differential methylation analysis. Differ-
ences in methylation levels before and after n-3 FA sup-
plementation were tested using the Illumina Custom
model in GenomeStudio software. FDR-corrected Diff-
Scores were computed to account for multiple testing
and limit false positives. We established significant dif-
ferences following n-3 FA supplementation with a FDR-
corrected DiffScore 13~P0.05.
Pathway analyses
We used the IPA system (Ingenuity® Systems, www.in-
genuity.com) to analyze potentially modified pathways
from differentially methylated genes following the sup-
plementation. Using a right-tailed Fishers exact test, IPA
measured the likelihood that pathways were overrepre-
sented among the list of significant differentially methyl-
ated genes.
Statistical analyses
We tested potential relationship between pre-
supplementation methylation levels and Δmethylation
of the 308 CpG sites and surrounding SNPs (±1 kb).
We used results from a recent GWAS done by our
group in the same cohort (n= 141) [26]. We consid-
ered only SNPs located at ±1 kb from CpG sites and
with a minor allele frequency 1%. We tested 652 as-
sociations using analysis of variance (general linear
model, type III sum of squares) and adjusted for the
Tremblay et al. Clinical Epigenetics (2017) 9:43 Page 6 of 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
effects of age, sex, and BMI. We used a Bonferroni
correction for multiple testing; thus, associations with
aPvalue 7.67 × 10
5
(0.05/652) were considered
significant.
Biochemical parameters are expressed as means ± SD.
We used a paired Studentsttest to test differences in bio-
chemical parameters before and after the supplementa-
tion. Variables not normally distributed were log
10
transformed before analyses. Δmethylation of CpG sites
(n= 20) within genes in the 16 pathways related to CV
health were defined as post-supplementation methylation
levels minus pre-supplementation methylation levels.
Changes in the four significantly modulated biochemical
parameters (ΔTG, ΔTC, ΔTC/HDL-C, and ΔGlucose)
were defined as (post-supplementation values minus pre-
supplementation values)/ pre-supplementation values to
account for baseline values. Potential relationships
between Δmethylation, Δexpression, and changes in bio-
chemical parameters (ΔTG, ΔTC, ΔTC/HDL-C, and
ΔGlucose) were investigated using Pearsons correlation.
We also accounted for potential confounding effects of
age, sex, and BMI in correlations. Statistical analyses were
conducted using SAS software version 9.3 (SAS Institute,
Cary, NC, USA).
Additional files
Additional file 1: Differentially methylated probes after the n-3 FA
supplementation. Description of data: List of all 308 differentially
methylated probes (FDR-corrected DiffScore 13~P0.05).
(XLSX 42 kb)
Additional file 2: Overrepresented pathways identified from differential
methylation analysis following an n-3 FA supplementation. Description of
data: Table describing all 55 significant overrepresented pathways identified
from methylation analysis (IPA canonical pathways, associated Pvalue, and
list of differentially methylated genes). (DOCX 21 kb)
Abbreviations
BMI: Body mass index; CpG: Cytosine-phosphate-guanine; CV: Cardiovascular;
DHA: Docosahexaenoic acid; EPA: Eicosapentaenoic acid; FDR: False
discovery rate; HDL-C: High-density lipoprotein cholesterol; IPA: Ingenuity
Pathway Analysis; n-3 FAs: Omega-3 fatty acids; SNPs: Single-nucleotide
polymorphisms; T2D: Type 2 diabetes; TC: Total cholesterol; TG: Triglyceride
Acknowledgements
We would like to thank Véronique Garneau, Ann-Marie Paradis, Élisabeth Thifault,
Karelle Dugas-Bourdage, Catherine Ouellette, and Annie Bouchard-Mercier, who
contributed to the success of this study. We also thank Catherine Raymond for
the laboratory work.
Funding
This work was supported by a grant from Canadian Institutes of Health
Research (CIHR)-(MOP-110975). IR holds a Junior 1 Research Scholar from the
Fonds de Recherche du QuébecSanté (FRQS). BLT is a recipient of a
scholarship from FRQS. MCV is Tier 1 Canada Research Chair in Genomics
Applied to Nutrition and Health.
Availability of data and materials
Methylation datasets supporting the conclusion of this article are available in
the Gene Expression Omnibus (GEO) database, www.ncbi.nlm.nih.gov/geo
(GSE98056).
Authorscontributions
Each author contribution to work: IR, SL, and MCV designed the research; PC
was responsible for the medical follow-up; BLT and FG conducted the re-
search and performed the statistical analyses; BLT wrote the paper; and BLT
and MCV have primary responsibility for the final content. All authors read
and approved the final manuscript.
Competing interests
The authors declare that they have no competing interests.
Consent for publication
Not applicable.
Ethics approval and consent to participate
All participants signed an informed consent document. This trial was
registered at clinicaltrials.gov as NCT01343342.
PublishersNote
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Received: 15 December 2016 Accepted: 14 April 2017
References
1. Caslake MJ, Miles EA, Kofler BM, Lietz G, Curtis P, Armah CK, Kimber AC,
Grew JP, Farrell L, Stannard J, et al. Effect of sex and genotype on
cardiovascular biomarker response to fish oils: the FINGEN Study. Am J Clin
Nutr. 2008;88:61829.
2. Cormier H, Rudkowska I, Paradis AM, Thifault E, Garneau V, Lemieux S,
Couture P, Vohl MC. Association between polymorphisms in the fatty acid
desaturase gene cluster and the plasma triacylglycerol response to an n-3
PUFA supplementation. Nutrients. 2012;4:102641.
3. Farzaneh-Far R, Harris WS, Garg S, Na B, Whooley MA. Inverse association of
erythrocyte n-3 fatty acid levels with inflammatory biomarkers in patients
with stable coronary artery disease: The Heart and Soul Study.
Atherosclerosis. 2009;205:53843.
4. Madsen T, Christensen JH, Blom M, Schmidt EB. The effect of dietary n-3
fatty acids on serum concentrations of C-reactive protein: a dose-response
study. Br J Nutr. 2003;89:51722.
5. Allaire J, Couture P, Leclerc M, Charest A, Marin J, Lepine MC, Talbot D,
Tchernof A, Lamarche B. A randomized, crossover, head-to-head
comparison of EPA and DHA supplementation to reduce inflammation
markers in men and women: Comparing EPA to DHA (ComparED) Study.
Am J Clin Nutr. 2016;104(2):280-7.
6. Harris WS, Miller M, Tighe AP, Davidson MH, Schaefer EJ. Omega-3 fatty
acids and coronary heart disease risk: clinical and mechanistic perspectives.
Atherosclerosis. 2008;197:1224.
7. Xin W, Wei W, Li XY. Short-term effects of fish-oil supplementation on heart
rate variability in humans: a meta-analysis of randomized controlled trials.
Am J Clin Nutr. 2013;97:92635.
8. von Schacky C, Fischer S, Weber PC. Long-term effects of dietary marine
omega-3 fatty acids upon plasma and cellular lipids, platelet function, and
eicosanoid formation in humans. J Clin Invest. 1985;76:162631.
9. Thies F, Garry JM, Yaqoob P, Rerkasem K, Williams J, Shearman CP, Gallagher
PJ, Calder PC, Grimble RF. Association of n-3 polyunsaturated fatty acids
with stability of atherosclerotic plaques: a randomised controlled trial.
Lancet. 2003;361:47785.
10. Burdge GC, Lillycrop KA. Fatty acids and epigenetics. Curr Opin Clin Nutr
Metab Care. 2014;17:15661.
11. Kulkarni A, Dangat K, Kale A, Sable P, Chavan-Gautam P, Joshi S. Effects of altered
maternal folic acid, vitamin B12 and docosahexaenoic acid on placental global
DNA methylation patterns in Wistar rats. PLoS One. 2011;6:e17706.
12. Daxinger L, Whitelaw E. Understanding transgenerational epigenetic
inheritance via the gametes in mammals. Nat Rev Genet. 2012;13:15362.
13. Friso S, Pizzolo F, Choi SW, Guarini P, Castagna A, Ravagnani V, Carletto A,
Pattini P, Corrocher R, Olivieri O. Epigenetic control of 11 beta-
hydroxysteroid dehydrogenase 2 gene promoter is related to human
hypertension. Atherosclerosis. 2008;199:3237.
14. Lund G, Andersson L, Lauria M, Lindholm M, Fraga MF, Villar-Garea A,
Ballestar E, Esteller M, Zaina S. DNA methylation polymorphisms precede
Tremblay et al. Clinical Epigenetics (2017) 9:43 Page 7 of 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
any histological sign of atherosclerosis in mice lacking apolipoprotein E. J
Biol Chem. 2004;279:2914754.
15. Turunen MP, Aavik E, Yla-Herttuala S. Epigenetics and atherosclerosis.
Biochim Biophys Acta. 1790;2009:88691.
16. Makar KW, Wilson CB. DNA methylation is a nonredundant repressor of the
Th2 effector program. J Immunol. 2004;173:44026.
17. Baccarelli A, Wright R, Bollati V, Litonjua A, Zanobetti A, Tarantini L, Sparrow
D, Vokonas P, Schwartz J. Ischemic heart disease and stroke in relation to
blood DNA methylation. Epidemiology. 2010;21:81928.
18. Kim M, Long TI, Arakawa K, Wang R, Yu MC, Laird PW. DNA methylation as a
biomarker for cardiovascular disease risk. PLoS One. 2010;5:e9692.
19. Lind MV, Martino D, Harslof LB, Kyjovska ZO, Kristensen M, Lauritzen L.
Genome-wide identification of mononuclear cell DNA methylation sites
potentially affected by fish oil supplementation in young infants: a pilot
study. Prostaglandins Leukot Essent Fatty Acids. 2015;101:17.
20. Voisin S, Almen MS, Moschonis G, Chrousos GP, Manios Y, Schioth HB. Dietary
fat quality impacts genome-wide DNA methylation patterns in a cross-
sectional study of Greek preadolescents. Eur J Hum Genet. 2015;23:65462.
21. Hoile SP, Clarke-Harris R, Huang RC, Calder PC, Mori TA, Beilin LJ, Lillycrop
KA, Burdge GC. Supplementation with N-3 long-chain polyunsaturated fatty
acids or olive oil in men and women with renal disease induces differential
changes in the DNA methylation of FADS2 and ELOVL5 in peripheral blood
mononuclear cells. PLoS One. 2014;9:e109896.
22. DoAmaral CL, Milagro FI, Curi R, Martinez JA. DNA methylation pattern in
overweight women under an energy-restricted diet supplemented with fish
oil. Biomed Res Int. 2014;2014:675021.
23. Aslibekyan S, Wiener HW, Havel PJ, Stanhope KL, OBrien DM, Hopkins SE,
Absher DM, Tiwari HK, Boyer BB. DNA methylation patterns are associated
with n-3 fatty acid intake in Yupik people. J Nutr. 2014;144:42530.
24. Lee HS, Barraza-Villarreal A, Hernandez-Vargas H, Sly PD, Biessy C,
Ramakrishnan U, Romieu I, Herceg Z. Modulation of DNA methylation states
and infant immune system by dietary supplementation with omega-3 PUFA
during pregnancy in an intervention study. Am J Clin Nutr. 2013;98:4807.
25. Thifault E, Cormier H, Bouchard-Mercier A, Rudkowska I, Paradis AM,
Garneau V, Ouellette C, Lemieux S, Couture P, Vohl MC. Effects of age, sex,
body mass index and APOE genotype on cardiovascular biomarker
response to an n-3 polyunsaturated fatty acid supplementation. J
Nutrigenet Nutrigenomics. 2013;6:7382.
26. Rudkowska I, Guenard F, Julien P, Couture P, Lemieux S, Barbier O, Calder
PC, Minihane AM, Vohl MC. Genome-wide association study of the plasma
triglyceride response to an n-3 polyunsaturated fatty acid supplementation.
J Lipid Res. 2014;55:124553.
27. Rudkowska I, Paradis AM, Thifault E, Julien P, Barbier O, Couture P, Lemieux
S, Vohl MC. Differences in metabolomic and transcriptomic profiles
between responders and non-responders to an n-3 polyunsaturated fatty
acids (PUFAs) supplementation. Genes Nutr. 2013;8:41123.
28. Clark LF, Thivierge MC, Kidd CA, McGeoch SC, Abraham P, Pearson DW,
Horgan GW, Holtrop G, Thies F, Lobley GE. Fish oil supplemented for
9 months does not improve glycaemic control or insulin sensitivity in
subjects with impaired glucose regulation: a parallel randomised controlled
trial. Br J Nutr. 2016;115:7586.
29. Balk EM, Lichtenstein AH, Chung M, Kupelnick B, Chew P, Lau J. Effects of
omega-3 fatty acids on serum markers of cardiovascular disease risk: a
systematic review. Atherosclerosis. 2006;189:1930.
30. Ma Y, Smith CE, Lai CQ, Irvin MR, Parnell LD, Lee YC, Pham LD, Aslibekyan S,
Claas SA, Tsai MY, et al. The effects of omega-3 polyunsaturated fatty acids
and genetic variants on methylation levels of the interleukin-6 gene
promoter. Mol Nutr Food Res. 2016;60:4109.
31. Rizos EC, Ntzani EE, Bika E, Kostapanos MS, Elisaf MS. Association between
omega-3 fatty acid supplementation and risk of major cardiovascular
disease events: a systematic review and meta-analysis. JAMA.
2012;308:102433.
32. Li K, Huang T, Zheng J, Wu K, Li D. Effect of marine-derived n-3
polyunsaturated fatty acids on C-reactive protein, interleukin 6 and tumor
necrosis factor alpha: a meta-analysis. PLoS One. 2014;9:e88103.
33. Baccarelli A, Tarantini L, Wright RO, Bollati V, Litonjua AA, Zanobetti A,
Sparrow D, Vokonas PS, Schwartz J. Repetitive element DNA methylation
and circulating endothelial and inflammation markers in the VA normative
aging study. Epigenetics. 2010;5:2228.
34. Naj AC, Jun G, Reitz C, Kunkle BW, Perry W, Park YS, Beecham GW,
Rajbhandary RA, Hamilton-Nelson KL, Wang LS, et al. Effects of multiple
genetic loci on age at onset in late-onset Alzheimer disease: a genome-
wide association study. JAMA Neurol. 2014;71:1394404.
35. Ma Y, Follis JL, Smith CE, Tanaka T, Manichaikul AW, Chu AY, Samieri C,
Zhou X, Guan W, Wang L, et al. Interaction of methylation-related genetic
variants with circulating fatty acids on plasma lipids: a meta-analysis of 7
studies and methylation analysis of 3 studies in the Cohorts for Heart and
Aging Research in Genomic Epidemiology consortium. Am J Clin Nutr.
2016;103:56778.
36. Zaina S, Heyn H, Carmona FJ, Varol N, Sayols S, Condom E, Ramirez-Ruz J,
Gomez A, Goncalves I, Moran S, Esteller M. DNA methylation map of human
atherosclerosis. Circ Cardiovasc Genet. 2014;7:692700.
37. Valencia-Morales Mdel P, Zaina S, Heyn H, Carmona FJ, Varol N, Sayols S,
Condom E, Ramirez-Ruz J, Gomez A, Moran S, et al. The DNA methylation
drift of the atherosclerotic aorta increases with lesion progression. BMC
Med Genomics. 2015;8:7.
38. Larson MK, Shearer GC, Ashmore JH, Anderson-Daniels JM, Graslie EL,
Tholen JT, Vogelaar JL, Korth AJ, Nareddy V, Sprehe M, Harris WS. Omega-3
fatty acids modulate collagen signaling in human platelets. Prostaglandins
Leukot Essent Fatty Acids. 2011;84:938.
39. Akinkuolie AO, Ngwa JS, Meigs JB, Djousse L. Omega-3 polyunsaturated
fatty acid and insulin sensitivity: a meta-analysis of randomized controlled
trials. Clin Nutr. 2011;30:7027.
40. Wren JD, Garner HR. Data-mining analysis suggests an epigenetic
pathogenesis for type 2 diabetes. J Biomed Biotechnol. 2005;2005:10412.
41. Volkmar M, Dedeurwaerder S, Cunha DA, Ndlovu MN, Defrance M, Deplus R,
Calonne E, Volkmar U, Igoillo-Esteve M, Naamane N, et al. DNA methylation
profiling identifies epigenetic dysregulation in pancreatic islets from type 2
diabetic patients. EMBO J. 2012;31:140526.
42. Kriebel J, Herder C, Rathmann W, Wahl S, Kunze S, Molnos S, Volkova N,
Schramm K, Carstensen-Kirberg M, Waldenberger M, et al. Association
between DNA methylation in whole blood and measures of glucose
metabolism: KORA F4 Study. PLoS One. 2016;11:e0152314.
43. Hanasaki H, Takemura Y, Fukuo K, Ohishi M, Onishi M, Yasuda O, Katsuya T,
Awata N, Kato N, Ogihara T, Rakugi H. Fas promoter region gene
polymorphism is associated with an increased risk for myocardial infarction.
Hypertens Res. 2009;32:2614.
44. Mihaylova MM, Vasquez DS, Ravnskjaer K, Denechaud PD, Yu RT, Alvarez JG,
Downes M, Evans RM, Montminy M, Shaw RJ. Class IIa histone deacetylases
are hormone-activated regulators of FOXO and mammalian glucose
homeostasis. Cell. 2011;145:60721.
45. de la Rocha C, Perez-Mojica JE, Leon SZ, Cervantes-Paz B, Tristan-Flores FE,
Rodriguez-Rios D, Molina-Torres J, Ramirez-Chavez E, Alvarado-Caudillo Y,
Carmona FJ, et al. Associations between whole peripheral blood fatty acids
and DNA methylation in humans. Sci Rep. 2016;6:25867.
46. Weems JC, Griesel BA, Olson AL. Class II histone deacetylases downregulate
GLUT4 transcription in response to increased cAMP signaling in cultured
adipocytes and fasting mice. Diabetes. 2012;61:140414.
47. Benton MC, Johnstone A, Eccles D, Harmon B, Hayes MT, Lea RA, Griffiths L,
Hoffman EP, Stubbs RS, Macartney-Coxson D. An analysis of DNA methylation
in human adipose tissue reveals differential modification of obesity genes
before and after gastric bypass and weight loss. Genome Biol. 2015;16:8.
48. Beattie J, Allan GJ, Lochrie JD, Flint DJ. Insulin-like growth factor-binding
protein-5 (IGFBP-5): a critical member of the IGF axis. Biochem J. 2006;395:119.
49. Fischer F, Schulte H, Mohan S, Tataru MC, Kohler E, Assmann G, von
Eckardstein A. Associations of insulin-like growth factors, insulin-like growth
factor binding proteins and acid-labile subunit with coronary heart disease.
Clin Endocrinol (Oxf). 2004;61:595602.
50. Castillero E, Lopez-Menduina M, Martin AI, Villanua MA, Lopez-Calderon A.
Comparison of the effects of the n-3 polyunsaturated fatty acid
eicosapentaenoic and fenofibrate on the inhibitory effect of arthritis on
IGF1. J Endocrinol. 2011;210:3618.
51. Coyne GS, Kenny DA, Waters SM. Effect of dietary n-3 polyunsaturated fatty acid
supplementationonbovineuterineendometrial and hepatic gene expression of
the insulin-like growth factor system. Theriogenology. 2011;75:50012.
52. Liu X, Yu X, Zack DJ, Zhu H, Qian J. TiGER: a database for tissue-specific
gene expression and regulation. BMC Bioinformatics. 2008;9:271.
53. Gai D, Haan E, Scholar M, Nicholl J, Yu S. Phenotypes of AKT3 deletion: a
case report and literature review. Am J Med Genet A. 2015;167A:1749.
54. Sandirasegarane L, Kester M. Enhanced stimulation of Akt-3/protein kinase
B-gamma in human aortic smooth muscle cells. Biochem Biophys Res
Commun. 2001;283:15863.
Tremblay et al. Clinical Epigenetics (2017) 9:43 Page 8 of 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
55. Brozinick Jr JT, Roberts BR, Dohm GL. Defective signaling through Akt-2 and
-3 but not Akt-1 in insulin-resistant human skeletal muscle: potential role in
insulin resistance. Diabetes. 2003;52:93541.
56. Yu H, Littlewood T, Bennett M. Akt isoforms in vascular disease. Vascul
Pharmacol. 2015;71:5764.
57. Ding L, Biswas S, Morton RE, Smith JD, Hay N, Byzova TV, Febbraio M,
Podrez EA. Akt3 deficiency in macrophages promotes foam cell formation
and atherosclerosis in mice. Cell Metab. 2012;15:86172.
58. Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell.
2007;129:126174.
59. Boyle JJ, Johns M, Kampfer T, Nguyen AT, Game L, Schaer DJ, Mason JC,
Haskard DO. Activating transcription factor 1 directs Mhem atheroprotective
macrophages through coordinated iron handling and foam cell protection.
Circ Res. 2012;110:2033.
60. Wan X, Huo Y, Johns M, Piper E, Mason JC, Carling D, Haskard DO, Boyle JJ.
5-AMP-activated protein kinase-activating transcription factor 1 cascade
modulates human monocyte-derived macrophages to atheroprotective
functions in response to heme or metformin. Arterioscler Thromb Vasc Biol.
2013;33:247080.
61. Yang S, Gao Y, Liu G, Li J, Shi K, Du B, Si D, Yang P. The human ATF1
rs11169571 polymorphism increases essential hypertension risk through
modifying miRNA binding. FEBS Lett. 2015;589:208793.
62. Bell JT, Tsai PC, Yang TP, Pidsley R, Nisbet J, Glass D, Mangino M, Zhai G,
Zhang F, Valdes A, et al. Epigenome-wide scans identify differentially
methylated regions for age and age-related phenotypes in a healthy ageing
population. PLoS Genet. 2012;8:e1002629.
63. Ma B, Wilker EH, Willis-Owen SA, Byun HM, Wong KC, Motta V, Baccarelli AA,
Schwartz J, Cookson WO, Khabbaz K, et al. Predicting DNA methylation level
across human tissues. Nucleic Acids Res. 2014;42:351528.
64. Ronn T, Volkov P, Gillberg L, Kokosar M, Perfilyev A, Jacobsen AL, Jorgensen
SW, Brons C, Jansson PA, Eriksson KF, et al. Impact of age, BMI and HbA1c
levels on the genome-wide DNA methylation and mRNA expression
patterns in human adipose tissue and identification of epigenetic
biomarkers in blood. Hum Mol Genet. 2015;24:3792813.
65. Guénard F DY, Hould FS, Lebel S, Tchernof A, Marceau P and Vohl MC. Use
of Blood as a Surrogate Model for the Assessment of Visceral Adipose
Tissue Methylation Profiles Associated with the Metabolic Syndrome in Men.
J Mol Genet Med. 2016;10:1
66. Aslibekyan S, Almeida M, Tintle N. Pathway analysis approaches for rare and
common variants: insights from Genetic Analysis Workshop 18. Genet
Epidemiol. 2014;38 Suppl 1:S8691.
67. Health Canada. Eating Well with the Canada's Food Guide. Ottawa:
Publications Health Canada: 2011.
68. Callaway C, Chumlea W, Bouchard C, Himes J, Lohman T, Martin A, Mitchell
C, Mueller W, Roche A, Seefeldt V. Standardization of anthropomeric
measurements : The Airlie (VA) Consensus Conference. (TG L, AF R, R M
editors). Champaign: Human Kinetics; 1988:39-80. pp. 39-80.
69. McNamara JR, Schaefer EJ. Automated enzymatic standardized lipid analyses
for plasma and lipoprotein fractions. Clin Chim Acta. 1987;166:18.
70. BURSTEIN M, SAMAILLE J. On a rapid determination of the cholesterol bound
to the serum alpha- and beta-lipoproteins. Clin Chim Acta. 1960;5:609.
71. Albers JJ, Warnick GR, Wiebe D, King P, Steiner P, Smith L, Breckenridge C,
Chow A, Kuba K, Weidman S, et al. Multi-laboratory comparison of three
heparin-Mn2+ precipitation procedures for estimating cholesterol in high-
density lipoprotein. Clin Chem. 1978;24:8536.
72. Friedewald WT, Levy RI, Fredrickson DS. Estimation of the concentration of
low-density lipoprotein cholesterol in plasma, without use of the
preparative ultracentrifuge. Clin Chem. 1972;18:499502.
73. Pirro M, Bergeron J, Dagenais GR, Bernard PM, Cantin B, Despres JP,
Lamarche B. Age and duration of follow-up as modulators of the risk for
ischemic heart disease associated with high plasma C-reactive protein levels
in men. Arch Intern Med. 2001;161:247480.
We accept pre-submission inquiries
Our selector tool helps you to find the most relevant journal
We provide round the clock customer support
Convenient online submission
Thorough peer review
Inclusion in PubMed and all major indexing services
Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
Tremblay et al. Clinical Epigenetics (2017) 9:43 Page 9 of 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Likewise, the Mediterranean diet has been associated with changes in the epigenome through DNA methylation in genes related to immunocompetence and inflammation [54]. In this sense, N3-PUFA supplementation has been associated with differentially methylated CpG sites related to inflammatory and immune response [55]. For example, a study conduceted by Arpon et al. [56] showed that a Mediterranean diet supplemented with extra virgin olive oil was associated with differential methylation of inflammation-related genes in humans. ...
Article
Full-text available
Background: The pathoetiology of systemic lupus erythematosus (SLE) involves a multifactorial interaction consisting of various genetic, epigenetic and environmental factors. Considering epigenetic characteristics, notable alterations in DNA methylation, particularly hypomethylation in immune-related pathways, such as T cell receptor have been observed. In turn, these alterations are associated with the overexpression of genes related to autoimmunity and a loss of immunological self-tolerance. Furthermore, DNA hypomethylation levels in SLE may contribute to disease progression and also impact disease activity and clinical manifestations. Summary: It is well-established that nutritional epigenetics elucidates the role of nutrition and dietary factors on the interactions of metabolic systems with the molecules that bind to DNA, regulating gene expression. Specific nutritional interventions may reverse initial epigenetic patterns, thereby significantly impacting the chronic disease´s treatment and prognostic. In fact, dietary nutrients and bioactive food compounds may influence DNA methylation patterns by inhibiting enzymes related to DNA methylation reactions or by altering the availability of different substrates involved in DNA methylation process (e.g methyl-donor nutrients). Key Messages: The knowledge of how diet plays a role in changing DNA methylation patterns in SLE is in the early stages. While a few studies in the literature have assessed the effects of nutrient intake, supplementation, or treatment on DNA methylation levels and have demonstrated their relevance, further research is imperative to deepen our comprehension of the interactions between epigenetics and nutrients, which is vital for the development of novel precision nutrition approaches.
... Many studies have shown that the effect of PUFAs on non-communicable chronic diseases might be mediated by DNAm [14]. For n-3 PUFA, Tremblay and collaborators found methylation changes following eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) intake in genes related to inflammatory and immune responses, lipid metabolism, cardiovascular signaling and type 2 diabetes in 36 overweight and obese individuals [15], while Amaral and collaborators found changes related to lipid metabolism, inflammatory response and phagocytosis in 12 overweight and obese young women [16]. n-6 PUFA intake was found to be associated with methylation in the tumor necrosis factor-α (TNF-α) pathway and inflammation processes in a group of 40 normal-weight young women [17]. ...
Article
Full-text available
Background Dietary intake of n-3 polyunsaturated fatty acids (PUFA) may have a protective effect on the development of cardiovascular diseases, diabetes, depression and cancer, while a high intake of n-6 PUFA was often reported to be associated with inflammation-related traits. The effect of PUFAs on health outcomes might be mediated by DNA methylation (DNAm). The aim of our study is to identify the impact of PUFA intake on DNAm in the Cooperative Health Research in the Region of Augsburg (KORA) FF4 cohort and the Leiden Longevity Study (LLS). Results DNA methylation levels were measured in whole blood from the population-based KORA FF4 study (N = 1354) and LLS (N = 448), using the Illumina MethylationEPIC BeadChip and Illumina HumanMethylation450 array, respectively. We assessed associations between DNAm and intake of eight and four PUFAs in KORA and LLS, respectively. Where possible, results were meta-analyzed. Below the Bonferroni correction threshold (p < 7.17 × 10–8), we identified two differentially methylated positions (DMPs) associated with PUFA intake in the KORA study. The DMP cg19937480, annotated to gene PRDX1, was positively associated with docosahexaenoic acid (DHA) in model 1 (beta: 2.00 × 10–5, 95%CI: 1.28 × 10–5-2.73 × 10–5, P value: 6.98 × 10–8), while cg05041783, annotated to gene MARK2, was positively associated with docosapentaenoic acid (DPA) in our fully adjusted model (beta: 9.80 × 10–5, 95%CI: 6.25 × 10–5-1.33 × 10–4, P value: 6.75 × 10–8). In the meta-analysis, we identified the CpG site (cg15951061), annotated to gene CDCA7L below Bonferroni correction (1.23 × 10–7) associated with eicosapentaenoic acid (EPA) intake in model 1 (beta: 2.00 × 10–5, 95% CI: 1.27 × 10–5–2.73 × 10–5, P value = 5.99 × 10–8) and we confirmed the association of cg19937480 with DHA in both models 1 and 2 (beta: 2.07 × 10–5, 95% CI: 1.31 × 10–5–2.83 × 10–5, P value = 1.00 × 10–7 and beta: 2.19 × 10–5, 95% CI: 1.41 × 10–5–2.97 × 10–5, P value = 5.91 × 10–8 respectively). Conclusions Our study identified three CpG sites associated with PUFA intake. The mechanisms of these sites remain largely unexplored, highlighting the novelty of our findings. Further research is essential to understand the links between CpG site methylation and PUFA outcomes.
... For other PUFA such as arachidonic acid (AA) and EPA, epigenetic modification such as DNA methylation profiling was also changed in E18.5 fetal male brains after supplementation with linoleic acid and AA for 2 weeks prior to conception in mice 34 and in blood leukocytes of overweight and obese adults following omega-3 fatty acid (EPA and DHA) supplementation. 35 The combination of EPA and DHA supplementation has similar changes in modifying global H3K9ac epigenetic programming in a rat colon cancer progression model. 36 Furthermore, feeding energy-restricted diets supplemented with DHA-enriched fish oil in obese subjects led to increased CPG site methylation levels at PDK4 (−222 and −50) and FADS1 (−25, −22, and −20). ...
Article
Goat milk is increasingly recognized by consumers due to its high nutritional value, richness in short- and medium-chain fatty acids, and richness in polyunsaturated fatty acids (PUFA). Exogenous supplementation of docosahexaenoic acid (DHA) is an important approach to increasing the content of PUFA in goat milk. Several studies have reported benefits of dietary DHA in terms of human health, including potential against chronic diseases and tumors. However, the mechanisms whereby an increased supply of DHA regulates mammary cell function is unknown. In this study, we investigated the effect of DHA on lipid metabolism processes in goat mammary epithelial cells (GMEC) and the function of H3K9ac epigenetic modifications in this process. Supplementation of DHA promoted lipid droplet accumulation increased the DHA content and altered fatty acid composition in GMEC. Lipid metabolism processes were altered by DHA supplementation through transcriptional programs in GMEC. ChIP-seq analysis revealed that DHA induced genome-wide H3K9ac epigenetic changes in GMEC. Multiomics analyses (H3K9ac genome-wide screening and RNA-seq) revealed that DHA-induced expression of lipid metabolism genes (FASN, SCD1, FADS1, FADS2, LPIN1, DGAT1, MBOAT2), which were closely related with changes in lipid metabolism processes and fatty acid profiles, were regulated by modification of H3K9ac. In particular, DHA increased the enrichment of H3K9ac in the promoter region of PDK4 and promoted its transcription, while PDK4 inhibited lipid synthesis and activated AMPK signaling in GMEC. The activation of the expression of fatty acid metabolism-related genes FASN, FADS2, and SCD1 and their upstream transcription factor SREBP1 by the AMPK inhibitor was attenuated in PDK4-overexpressing GMEC. In conclusion, DHA alters lipid metabolism processes via H3K9ac modifications and the PDK4-AMPK-SREBP1 signaling axis in goat mammary epithelial cells, providing new insights into the mechanism through which DHA affects mammary cell function and regulates milk fat metabolism.
Article
Full-text available
Obesity and chronic low-grade inflammation, often occurring together, significantly contribute to severe metabolic and inflammatory conditions like type 2 diabetes (T2D), cardiovascular disease (CVD), and cancer. A key player is elevated levels of gut dysbiosis-associated lipopolysaccharide (LPS), which disrupts metabolic and immune signaling leading to metabolic endotoxemia, while short-chain fatty acids (SCFAs) beneficially regulate these processes during homeostasis. SCFAs not only safeguard the gut barrier but also exert metabolic and immunomodulatory effects via G protein-coupled receptor binding and epigenetic regulation. SCFAs are emerging as potential agents to counteract dysbiosis-induced epigenetic changes, specifically targeting metabolic and inflammatory genes through DNA methylation, histone acetylation, microRNAs (miRNAs), and long non-coding RNAs (lncRNAs). To assess whether SCFAs can effectively interrupt the detrimental cascade of obesity and inflammation, this review aims to provide a comprehensive overview of the current evidence for their clinical application. The review emphasizes factors influencing SCFA production, the intricate connections between metabolism, the immune system, and the gut microbiome, and the epigenetic mechanisms regulated by SCFAs that impact metabolism and the immune system.
Article
Full-text available
Machine learning models based on DNA methylation data can predict biological age but often lack causal insights. By harnessing large-scale genetic data through epigenome-wide Mendelian randomization, we identified CpG sites potentially causal for aging-related traits. Neither the existing epigenetic clocks nor age-related differential DNA methylation are enriched in these sites. These CpGs include sites that contribute to aging and protect against it, yet their combined contribution negatively affects age-related traits. We established a new framework to introduce causal information into epigenetic clocks, resulting in DamAge and AdaptAge—clocks that track detrimental and adaptive methylation changes, respectively. DamAge correlates with adverse outcomes, including mortality, while AdaptAge is associated with beneficial adaptations. These causality-enriched clocks exhibit sensitivity to short-term interventions. Our findings provide a detailed landscape of CpG sites with putative causal links to lifespan and healthspan, facilitating the development of aging biomarkers, assessing interventions, and studying reversibility of age-associated changes.
Article
Research investigating the effects of the food matrix on health is needed to untangle many unresolved questions in nutritional science. Emulsion structure plays a fundamental role in this inquiry; however, the effects of oil-in-water emulsion structure on broad metabolic, physiological, and health-related outcomes have not been comprehensively reviewed. This systematic scoping review targets this gap and examines methodological considerations for the field of relating food structure and health. MEDLINE, Web of Science, and CAB Direct were searched from inception to December 2022, returning 3106 articles, 52 of which were eligible for inclusion. Many investigated emulsion lipid droplet size and/or gastric colloidal stability and their relation to postprandial weight-loss-related outcomes. The present review also identifies numerous novel relationships between emulsion structures and health-related outcomes. "Omics" endpoints present an exciting avenue for more comprehensive analysis in this area, yet interpretation remains difficult. Identifying valid surrogate biomarkers for long-term outcomes and disease risk will be a turning point for food structure research, leading to breakthroughs in the pace and utility of research that generates advancements in health. The review's findings and recommendations aim to support new hypotheses, future trial design, and evidence-based emulsion design for improved health and well-being.
Article
Full-text available
Background Epigenetic studies have reported relationships between dietary nutrient intake and methylation levels. However, genetic variants that may affect DNA methylation (DNAm) pattern, called methylation quantitative loci (mQTL), are usually overlooked in these analyses. We investigated whether mQTL change the relationship between dietary nutrient intake and leukocyte DNAm levels with an example of estimated fatty acid intake and ATP-binding cassette transporter A1 (ABCA1). Methods A cross-sectional study on 231 participants (108 men, mean age: 62.7 y) without clinical history of cancer and no prescriptions for dyslipidemia. We measured leukocyte DNAm levels of 8 CpG sites within ABCA1 gene by pyrosequencing method and used mean methylation levels for statistical analysis. TaqMan assay was used for genotyping a genetic variant of ABCA1 (rs1800976). Dietary fatty acid intake was estimated with a validated food frequency questionnaire and adjusted for total energy intake by using residual methods. Results Mean ABCA1 DNAm levels were 5% lower with the number of minor alleles in rs1800976 (CC, 40.6%; CG, 35.9%; GG, 30.6%). Higher dietary n-3 PUFA intake was associated with lower ABCA1 DNAm levels (1st (ref) vs. 4th, β [95% CI]: –2.52 [–4.77, –0.28]). After controlling for rs180076, the association between dietary n-3 PUFA intake and ABCA1 DNAm levels was attenuated, but still showed an independent association (1st (ref) vs. 4th, β [95% CI]: –2.00 [–3.84, –0.18]). The interaction of mQTL and dietary n-3 PUFA intake on DNAm levels was not significant. Conclusions This result suggested that dietary n-3 PUFA intake would be an independent predictor of DNAm levels in ABCA1 gene after adjusting for individual genetic background. Considering mQTL need to broaden into other genes and nutrients for deeper understanding of DNA methylation, which can contribute to personalized nutritional intervention.
Article
Cardiovascular disease (CVD) is a leading cause of death worldwide. Supplementation with the marine omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) is associated with lower CVD risk. However, results from randomized controlled trials that examine the effect of omega-3 supplementation on CVD risk are inconsistent. This risk-reducing effect may be mediated by reducing inflammation, oxidative stress and serum triglyceride (TG) levels. However, not all individuals respond by reducing TG levels after omega-3 supplementation. This inter-individual variability in TG response to omega-3 supplementation is not fully understood. Hence, we aim to review the evidence for how interactions between omega-3 fatty acid supplementation and genetic variants, epigenetic and gene expression profiling, gut microbiota and habitual intake of omega-3 fatty acids can explain why the TG response differs between individuals. This may contribute to understanding the current controversies and play a role in defining future personalized guidelines to prevent CVD.
Article
Full-text available
Fatty acids (FA) modify DNA methylation in vitro, but limited information is available on whether corresponding associations exist in vivo and reflect any short-term effect of the diet. Associations between global DNA methylation and FAs were sought in blood from lactating infants (LI; n = 49) and adult males (AMM; n = 12) equally distributed across the three conventional BMI classes. AMM provided multiple samples at 2-hour intervals during 8 hours after either a single Western diet-representative meal (post-prandial samples) or no meal (fasting samples). Lipid/glucose profile, HDAC4 promoter and PDK4 5’UTR methylation were determined in AMM. Multiple regression analysis revealed that global (in LI) and both global and PDK4-specific DNA methylation (in AMM) were positively associated with eicosapentaenoic and arachidonic acid. HDAC4 methylation was inversely associated with arachidonic acid post-prandially in AMM. Global DNA methylation did not show any defined within-day pattern that would suggest a short-term response to the diet. Nonetheless, global DNA methylation was higher in normal weight subjects both post-prandially and in fasting and coincided with higher polyunsaturated relative to monounsaturated and saturated FAs. We show for the first time strong associations of DNA methylation with specific FAs in two human cohorts of distinct age, diet and postnatal development stage.
Article
Full-text available
Epigenetic regulation has been postulated to affect glucose metabolism, insulin sensitivity and the risk of type 2 diabetes. Therefore, we performed an epigenome-wide association study for measures of glucose metabolism in whole blood samples of the population-based Cooperative Health Research in the Region of Augsburg F4 study using the Illumina HumanMethylation 450 BeadChip. We identified a total of 31 CpG sites where methylation level was associated with measures of glucose metabolism after adjustment for age, sex, smoking, and estimated white blood cell proportions and correction for multiple testing using the Benjamini-Hochberg (B-H) method (four for fasting glucose, seven for fasting insulin, 25 for homeostasis model assessment-insulin resistance [HOMA-IR]; B-H-adjusted p-values between 9.2x10-5 and 0.047). In addition, DNA methylation at cg06500161 (annotated to ABCG1) was associated with all the aforementioned phenotypes and 2-hour glucose (B-H-adjusted p-values between 9.2x10-5 and 3.0x10-3). Methylation status of additional three CpG sites showed an association with fasting insulin only after additional adjustment for body mass index (BMI) (B-H-adjusted p-values = 0.047). Overall, effect strengths were reduced by around 30% after additional adjustment for BMI, suggesting that this variable has an influence on the investigated phenotypes. Furthermore, we found significant associations between methylation status of 21 of the aforementioned CpG sites and 2-hour insulin in a subset of samples with seven significant associations persisting after additional adjustment for BMI. In a subset of 533 participants, methylation of the CpG site cg06500161 (ABCG1) was inversely associated with ABCG1 gene expression (B-H-adjusted p-value = 1.5x10-9). Additionally, we observed an enrichment of the top 1,000 CpG sites for diabetes-related canonical pathways using Ingenuity Pathway Analysis. In conclusion, our study indicates that DNA methylation and diabetes-related traits are associated and that these associations are partially BMI-dependent. Furthermore, the interaction of ABCG1 with glucose metabolism is modulated by epigenetic processes.
Article
Full-text available
Epigenetic mechanisms are known to be involved in tissue-specific differentiation. DNA methylation patterns have been shown to be largely conserved across tissues but with variation for specific genes. However, it is unclear whether the variability observed in the methylation profile of a metabolically active tissue is reflected in other sources such as hematopoietic tissue. This study aimed to test blood genome-wide CpG site methylation levels as a surrogate model for visceral adipose tissue (VAT) methylation and to verify whether it appropriately reflects differences in methylation levels found in VAT between men discordant for the metabolic syndrome (MetS). Tissue specimens (VAT and blood samples) were obtained from 16 severely obese individuals discordant for the MetS. CpG sites methylation levels were measured with the Infinium HumanMethylation450 BeadChip and correlations of methylation levels between VAT and blood were computed. Differences in methylation levels between individuals with and without MetS were tested in both tissues. Pathway analysis was conducted for differentially methylated CpG sites common to both tissues. High cross-tissue correlations were observed for VAT and blood (0.952±0.014) while some CpG sites had significantly different methylation levels in VAT versus blood. Differential methylation analysis between individuals with and without MetS demonstrated a higher number of differentially methylated CpG sites in VAT than in blood (11,778 vs. 881, respectively) with nearly 4% of differentially methylated sites found in VAT being also represented in blood. Common differentially methylated sites were involved in inflammatory-, lipid- and diabetes-related pathways. These results suggest that blood methylation levels of specific CpG sites may adequately reflect VAT methylation levels for some of the MetS-related genes, specifically for inflammatory, lipid and glucose metabolism genes.
Article
Full-text available
Background: DNA methylation is influenced by diet and single nucleotide polymorphisms (SNPs), and methylation modulates gene expression. Objective: We aimed to explore whether the gene-by-diet interactions on blood lipids act through DNA methylation. Design: We selected 7 SNPs on the basis of predicted relations in fatty acids, methylation, and lipids. We conducted a meta-analysis and a methylation and mediation analysis with the use of data from the CHARGE (Cohorts for Heart and Aging Research in Genomic Epidemiology) consortium and the ENCODE (Encyclopedia of DNA Elements) consortium. Results: On the basis of the meta-analysis of 7 cohorts in the CHARGE consortium, higher plasma HDL cholesterol was associated with fewer C alleles at ATP-binding cassette subfamily A member 1 (ABCA1) rs2246293 (β = -0.6 mg/dL, P = 0.015) and higher circulating eicosapentaenoic acid (EPA) (β = 3.87 mg/dL, P = 5.62 × 10(21)). The difference in HDL cholesterol associated with higher circulating EPA was dependent on genotypes at rs2246293, and it was greater for each additional C allele (β = 1.69 mg/dL, P = 0.006). In the GOLDN (Genetics of Lipid Lowering Drugs and Diet Network) study, higher ABCA1 promoter cg14019050 methylation was associated with more C alleles at rs2246293 (β = 8.84%, P = 3.51 × 10(18)) and lower circulating EPA (β = -1.46%, P = 0.009), and the mean difference in methylation of cg14019050 that was associated with higher EPA was smaller with each additional C allele of rs2246293 (β = -2.83%, P = 0.007). Higher ABCA1 cg14019050 methylation was correlated with lower ABCA1 expression (r = -0.61, P = 0.009) in the ENCODE consortium and lower plasma HDL cholesterol in the GOLDN study (r = -0.12, P = 0.0002). An additional mediation analysis was meta-analyzed across the GOLDN study, Cardiovascular Health Study, and the Multi-Ethnic Study of Atherosclerosis. Compared with the model without the adjustment of cg14019050 methylation, the model with such adjustment provided smaller estimates of the mean plasma HDL cholesterol concentration in association with both the rs2246293 C allele and EPA and a smaller difference by rs2246293 genotypes in the EPA-associated HDL cholesterol. However, the differences between 2 nested models were NS (P > 0.05). Conclusion: We obtained little evidence that the gene-by-fatty acid interactions on blood lipids act through DNA methylation.
Article
Background: To date, most studies on the anti-inflammatory effects of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in humans have used a mixture of the 2 fatty acids in various forms and proportions. Objectives: We compared the effects of EPA supplementation with those of DHA supplementation (re-esterified triacylglycerol; 90% pure) on inflammation markers (primary outcome) and blood lipids (secondary outcome) in men and women at risk of cardiovascular disease. Design: In a double-blind, randomized, crossover, controlled study, healthy men (n = 48) and women (n = 106) with abdominal obesity and low-grade systemic inflammation consumed 3 g/d of the following supplements for periods of 10 wk: 1) EPA (2.7 g/d), 2) DHA (2.7 g/d), and 3) corn oil as a control with each supplementation separated by a 9-wk washout period. Primary analyses assessed the difference in cardiometabolic outcomes between EPA and DHA. Results: Supplementation with DHA compared with supplementation with EPA led to a greater reduction in interleukin-18 (IL-18) (-7.0% ± 2.8% compared with -0.5% ± 3.0%, respectively; P = 0.01) and a greater increase in adiponectin (3.1% ± 1.6% compared with -1.2% ± 1.7%, respectively; P < 0.001). Between DHA and EPA, changes in CRP (-7.9% ± 5.0% compared with -1.8% ± 6.5%, respectively; P = 0.25), IL-6 (-12.0% ± 7.0% compared with -13.4% ± 7.0%, respectively; P = 0.86), and tumor necrosis factor-α (-14.8% ± 5.1% compared with -7.6% ± 10.2%, respectively; P = 0.63) were NS. DHA compared with EPA led to more pronounced reductions in triglycerides (-13.3% ± 2.3% compared with -11.9% ± 2.2%, respectively; P = 0.005) and the cholesterol:HDL-cholesterol ratio (-2.5% ± 1.3% compared with 0.3% ± 1.1%, respectively; P = 0.006) and greater increases in HDL cholesterol (7.6% ± 1.4% compared with -0.7% ± 1.1%, respectively; P < 0.0001) and LDL cholesterol (6.9% ± 1.8% compared with 2.2% ± 1.6%, respectively; P = 0.04). The increase in LDL-cholesterol concentrations for DHA compared with EPA was significant in men but not in women (P-treatment × sex interaction = 0.046). Conclusions: DHA is more effective than EPA in modulating specific markers of inflammation as well as blood lipids. Additional studies are needed to determine the effect of a long-term DHA supplementation per se on cardiovascular disease risk. This trial was registered at clinicaltrials.gov as NCT01810003.
Article
Atherosclerotic risk factors can be divided into two main categories-genetical and environmental issues. The latter ones include habitual factors since human habits manifest as environmental factors at the cellular level. Environmental issues can govern human health via epigenetic modification of chromatin structure. This review discusses the recent findings linking general epigenetic mechanisms of chromatin modifications to atherosclerosis development. © 2012 Springer Science+Business Media New York. All rights are reserved.