ArticlePDF Available

Abstract and Figures

Epigenetic modifications, especially alteration in DNA methylation, are increasingly being recognized as a key factor in the pathogenesis of complex disorders, including atherosclerosis. However, there are limited data on the epigenetic changes in the coronary artery disease (CAD) patients. In the present study we evaluated the methylation status of genomic DNA from peripheral lymphocytes in a cohort of 287 individuals: 137 angiographically confirmed CAD patients and 150 controls. The differential susceptibility of genomic DNA to methylation-sensitive restriction enzymes was utilized to assess the methylation status of the genome. We observed that the genomic DNA methylation in CAD patients is significantly higher than in controls (p < 0.05). Since elevated homocysteine levels are known to be an independent risk factor for CAD and a key modulator of macromolecular methylation, we investigated the probable correlation between plasma homocysteine levels and global DNA methylation. We observed a significant positive correlation of global DNA methylation with plasma homocysteine levels in CAD patients (p = 0.001). Further, within a higher range of serum homocysteine levels (>/=12-50 muM), global DNA methylation was significantly higher in CAD patients than in controls. The alteration in genomic DNA methylation associated with cardiovascular disease per se appears to be further accentuated by higher homocysteine levels.
Content may be subject to copyright.
Detection of Altered Global DNA Methylation in Coronary
Artery Disease Patients
Priyanka Sharma,
1,2
Jitender Kumar,
1
Gaurav Garg,
1
Arun Kumar,
1
Ashok Patowary,
1
Ganesan Karthikeyan,
3
Lakshmy Ramakrishnan,
3
Vani Brahmachari,
2
and Shantanu Sengupta
1
Epigenetic modifications, especially alteration in DNA methylation, are increasingly being recognized as a key
factor in the pathogenesis of complex disorders, including atherosclerosis. However, there are limited data on the
epigenetic changes in the coronary artery disease (CAD) patients. In the present study we evaluated the methyl-
ation status of genomic DNA from peripheral lymphocytes in a cohort of 287 individuals: 137 angiographically
confirmed CAD patients and 150 controls. The differential susceptibility of genomic DNA to methylation-sensitive
restriction enzymes was utilized to assess the methylation status of the genome. We observed that the genomic
DNA methylation in CAD patients is significantly higher than in controls ( p< 0.05). Since elevated homocysteine
levels are known to be an independent risk factor for CAD and a key modulator of macromolecular methylation,
we investigated the probable correlation between plasma homocysteine levels and global DNA methylation. We
observed a significant positive correlation of global DNA methylation with plasma homocysteine levels in CAD
patients ( p¼0.001). Further, within a higher range of serum homocysteine levels ($12–50 mM), global DNA
methylation was significantly higher in CAD patients than in controls. The alteration in genomic DNA methyl-
ation associated with cardiovascular disease per se appears to be further accentuated by higher homocysteine
levels.
Introduction
The incidence of coronary artery disease (CAD) is
on the rise worldwide and is expected to become the
main cause of death globally within the next 15 years, owing
to a rapidly increasing prevalence in developing countries
(Dodu, 1988). It is estimated that two-third of the 14 million
cardiovascular fatalities worldwide would occur in the de-
veloping countries. Mortality due to coronary heart diseases
in India increased from 1.17 million to 1.59 million from 1990
to 2000 and is expected to rise to 2.03 million by 2010 (Ah-
mad and Bhopal, 2004) and could reach epidemic propor-
tions by 2030 (Leeder et al., 2004). The early onset of CAD
along with its severity cannot be explained entirely by the
classical risk factors and genetic variations (Wilson et al.,
1998; Winkelmann and Hager, 2000). Further, the epigenetic
code for transcription regulation through DNA methylation
and chromatin modifications can add yet another level of
variability and can escape attention in the typical genetic
association studies (Turner, 2007). There is increasing evi-
dence to show that epigenetic regulation plays an important
role in the course of complex diseases in addition to its role in
X-chromosome inactivation, genomic imprinting, and main-
tenance of cellular transcriptional memory during develop-
ment (Bird, 2002; Feinberg, 2004; Moss and Mallrats, 2007;
Shames et al., 2007).
The modulators of macromolecular methylation could
be important links between the environment and epigenetic
regulation. For instance, an elevated level of homocysteine, a
thiol amino acid, is an independent risk factor for cardiovas-
cular diseases and also a modulator of macromolecular
methylation (Robinson et al., 1995; van Guldener et al., 2005).
Hyperhomocysteinemia has been reported to have a graded
effect on the risk of CAD and the extent and severity of the
disease (Chao et al., 1999). Further, a meta-analysis done by
Wald et al. (2003) along with their data clearly indicated a
significant increase in homocysteine levels with increasing
severity of CAD. It has also been found that in patients with
angiographically confirmed CAD, homocysteine levels are a
significant predictor of mortality, independent of traditional
risk factors (Yoo et al., 1999). The risk of recurrent coronary
events and death in patients with acute myocardial infarction
also was significantly associated with elevated homocysteine
levels, and this was independent of other risk factors.
1
Institute of Genomics and Integrative Biology, Delhi, India.
2
Dr. B.R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India.
3
All India Institute of Medical Sciences, New Delhi, India.
DNA AND CELL BIOLOGY
Volume 27, Number 7, 2008
ªMary Ann Liebert, Inc.
Pp. 357–365
DOI: 10.1089=dna.2007.0694
357
Although hyperhomocysteinemia has been implicated as
an independent risk factor for cardiovascular disease, the
mechanism through which it influences the disease mani-
festation is not fully understood. Several potential mecha-
nisms have been proposed to explain the role of homocysteine
in CAD; prominent among these are oxidative damage to
vascular endothelial cells, thrombotic effects of the coagula-
tion system, impairment of vasodilator properties of endo-
thelium by decreasing the bioavailability of nitric oxide,
endoplasmic reticular stress, and direct modulation through
physical interaction with proteins (Sharma et al., 2006). It
is possible that modulation of homocysteine levels could al-
ter certain fundamental intracellular processes such as the
methylation of macromolecules since homocysteine levels are
believed to dictate the fate of trans-methylation reactions that
are essential for several biological processes (Ulrey et al.,
2005).
In a recent study, using vascular smooth muscle cells in
culture, the authors observed hypomethylation of surrogate
markers Alu and LINE1 elements in spite of elevated activ-
ity of DNMT3a and DNMT3b under a high homocysteine
concentration (Yi-deng et al., 2007a). Similarly in cultured
monocytes, it has been shown that the presence of 100 mM
homocysteine elevates the level of total cholesterol and de-
creases ApoE mRNA through hypermethylation of promoter
sequences (Yi-Deng et al., 2007b).
We have undertaken a comparative analysis of global DNA
methylation at G þC-rich regions targeted by methylation-
sensitive restriction enzyme HpaII and its isoschizomer MspI,
in lymphocytes obtained from peripheral blood of CAD pa-
tients and controls. We found a significant correlation be-
tween global DNA methylation, plasma homocysteine levels,
and CAD. In addition, we observed a considerable effect of
vegetarian diet on plasma homocysteine levels and concur-
rently a correlation with global DNA methylation in CAD
patients. To the best of our knowledge, this is the first study on
the global DNA methylation along with plasma homocysteine
levels and dietary status in CAD patients.
Materials and Methods
Patient population
The study population included 287 subjects mainly from
the northern part of India. Among these, 137 were angio-
graphically confirmed CAD patients and 150 were healthy
controls who tested negative in treadmill test; that is, they
had normal heart rate and blood pressure response and ab-
sence of chest pain during exercise and had no abnormal
ST segment deviation during exercise and recovery. These
samples were randomly selected from a cohort recruited at
the All India Institute of Medical Sciences, New Delhi, India,
as a part of a project to study the genetic polymorphism
and epigenetic alteration associated with CAD. Apart from
clinical history, information on diet was also collected.
People who do not consume any animal products other than
milk and its products were classified as vegetarians (lacto-
vegetarians). The ethics committee of both the All India In-
stitute of Medical Sciences and the Institute of Genomics and
Integrative Biology approved this study. Written informed
consent was obtained from all the participants, and the study
was carried out in accordance with the principles of Helsinki
Declaration.
Blood samples for biochemical markers
Blood samples were collected from volunteers in tubes
containing anticoagulant, immediately chilled on ice, and
plasma was separated from the blood samples within an hour
of collection. Aliquots of plasma were transferred into cryostat
tubes and stored at 808C until further analysis. Genomic
DNA was isolated from blood samples using the modified
salting out method as described earlier and stored at 208C
until further analysis (Kumar et al., 2005). Genomic DNA
concentration was estimated after chelating the DNA with the
fluorescence probe picogreen (Molecular Probes, Eugene, OR).
Plasma levels of homocysteine were determined using high-
performance liquid chromatography (HPLC) equipped with a
fluorescence detector as described earlier (Ji et al., 1995). Vita-
min B12 and folate levels were determined using the Immu-
nolite Vitamin B12 (Diagnostic Product, Los Angeles, CA) and
SimulTrac-SNB Radio assay kits (MP Biomedicals, Orange-
burg, NY), respectively, as per the manufacturer’s protocol.
Assessment of global DNA methylation
The global DNA methylation status in peripheral blood
was determined using the cytosine extension assay as previ-
ously described (Ingrosso et al., 2003). Briefly, 1 mg of genomic
DNA was digested with 20 units of methylation-sensitive
enzymes HpaII and an isoschizomer MspI (New England
Biolabs, Beverly, MA) for 16–18 h in separate tubes. The di-
gested DNA was subjected to single-nucleotide extension re-
action in a 25 mL reaction mixture containing 0.5 mg of DNA, 1
Taq polymerase gold reaction buffer, 1.0 mM MgCl
2
, 0.25
units of heat-activated AmpliTaq Gold DNA polymerase
(Applied Biosystems, Weiterstadt, Germany), and 0.1 mCi of
[
3
H]dCTP (57.4 Ci=mmol; GE Healthcare Biosciences, Piscat-
away, NJ). The reaction mixture was incubated at 568C for 1 h
and chilled on ice. Ten mL aliquot in duplicate from each re-
action was applied on Whatman DE-81 ion exchange filters
with a multiscreen assay system (Millipore, Bedford, MA)
and washed three times with 0.5 M sodium phosphate buf-
fer (pH 7.0) at room temperature. Filters were dried, and the
radioactivity incorporated was counted using scintillation
counter (LS6500; Beckman, San Jose, CA).
Each assay was done in duplicate, and the values were
corrected for nonspecific incorporation by measuring the in-
corporation of [
3
H] dCTP into undigested genomic DNA. The
difference in the mean incorporation of [
3
H]dCTP following
MspI and HpaII digestion at a known concentration of geno-
mic DNA was computed as the global DNA methylation level
expressed as cpm=mg DNA. The deviation between duplicates
was less than 10%. The reproducibility of the cytosine exten-
sion assay was confirmed by repeating the assay in 5% of the
samples selected randomly. Variation between the repeats
was below 10%.
ApoE promoter methylation analysis
The methylation status of ApoE gene was analyzed by direct
sequencing of sodium bisulfite–modified genomic DNA.
Genomic DNA was subjected to bisulfite modification using
the EZ DNA Gold Methylation Kit (Zymo Research, Orange,
CA) following the manufacturer’s protocol. The ApoE forward
primer 50-GGATAATTTTAGGGAGGAGTGTTTTG-30and the
reverse primer 50-CTCCAGAACAATATCATCTCTACTAC-30
358 SHARMA ET AL.
were used to amplify 574 bp representing 25 CpG of ApoE
promoter region. PCR products were purified using QIAquick
Gel Extraction Kit (Qiagen, Chatsworth, CA). These purified
PCR products were cloned using pCR-4-TOPO TA Cloning
vector (Invitrogen). Ligated PCR products were transformed
using TOP10 competent cell (Invitrogen). Plasmids were pre-
pared from selected colonies using Wizard SV 96 Plasmid
DNA Purification System (Promega, Madison, WI) and were
sequenced on ABI 3730 sequencer. For each PCR product, five
clones were sequenced. Bisulfite sequencing results were ana-
lyzed using BiQ Analyzer software tool (Bock et al., 2005).
Statistical analysis
The statistical tests were performed using GraphPad Instat
(version 3.06) software. Mann–Whitney U-test was performed
to compare the biochemical parameters and global DNA
methylation levels between patients and controls. The Spear-
man rank correlation test was used to correlate homocysteine
concentrations with DNA methylation levels in patients and
controls. In all the patients, nonparametric correlation was
employed without Gaussian assumptions as the data were
not normally distributed. Two-tailed p-value <0.05 were con-
onsidered to be significant. We also performed a binary lo-
gistic regression analysis to ascertain the factors associated
with CAD and a multiple regression analysis to check for the
association of global DNA methylation with various param-
eters in patients and controls using SPSS software (version
10.0).
Results
The samples analyzed were randomly selected from a co-
hort recruited on the basis of being angiographically positive
for the patient group and treadmill test negative for controls.
The baseline characteristics of the subjects such as age, dia-
betes, hypertension, cigarette smoking habit, BMI, and diet
were compared between the groups and are shown in Table 1.
The two groups were comparable with reference to all these
parameters ( p>0.05). Further, the number of diabetic and
hypertensive subjects was not significantly different in the
two groups ( p>0.05). To ascertain if global DNA methylation
in CAD patients was different from that in the controls, we
analyzed the global methylation in DNA from peripheral
blood in a cohort of 137 angiographically confirmed CAD
patients and 150 controls. The global DNA methylation level
was significantly higher in CAD patients as shown in Figure 1
(median 11.510
3
cpm=mg DNA) compared to the controls
(median 10.910
3
cpm=mg DNA, p<0.05), irrespective of the
levels of homocysteine, one of the independent risk factors for
CAD, thus suggesting that global methylation could be as-
sociated with CAD per se. We also performed a binary logistic
regression to ascertain the parameters that are associated with
CAD in our study and found that age ( p¼0.003) and global
DNA methylation ( p¼0.02) are significantly associated with
CAD after adjusting for other parameters. But in a back-
ground of CAD, homocysteine levels could have additional
influence as modulators of DNA methylation. Corroborating
this, we observed a significant positive correlation of global
hypermethylation with plasma homocysteine levels in CAD
patients (n¼137, r¼0.2785, p¼0.001, Fig. 2A), but not in
controls (n¼150, r¼0.01662, p>0.05, Fig. 2B). When a subset
of patients and controls with higher than normal levels of
Table 1. Baseline Characteristic of Study Population
Patients (n¼137) Controls (n¼150) p-value
Age (years) 54
a
(30–75) 52
a
(27–75) 0.06
BMI
b
24.69
a
(15.7–36.7) 24.96
a
(16.1–75.6) 0.15
Diabetic [no. (%)] 36 (26.3) 40 (26.7) 0.95
Hypertensive [no. (%)] 73 (53.3) 79 (52.7) 0.95
Vegetarians [no. (%)] 68 (49.6) 64 (42.7) 0.47
Nonvegetarians [no. (%)] 69 (50.4) 86 (57.3) 0.52
Smokers [no. (%)] 39 (28.5) 61 (40.7) 0.13
a
Median values are shown.
b
Body mass index is the weight in kilograms divided by the square of the height in meters.
FIG. 1. Global DNA methylation in CAD patients and
controls. Methylation (cpm10
3
=mg DNA) on the Y-axis
represents the level of methylation estimated in terms of the
incorporation of [
3
H] dCTP. The box represents the inter-
quartile range, which contains the 50% of values. The whis-
kers are lines that extend the box to the highest and lowest
values, excluding outliers. A line across the box indicates the
median value. The global DNA methylation in CAD patients
is significantly higher than healthy controls ( p<0.05) as
shown in the box plot.
GLOBAL DNA METHYLATION AND CORONARY ARTERY DISEASE 359
homocysteine (>12 mM) were compared, we found that the
global DNA methylation levels were significantly higher
(p¼0.0005) in CAD patients (median 12.810
3
cpm=mg
DNA) than in controls (median 10.010
3
cpm=mg DNA,
Fig. 3). This indicates a significant DNA hypermethylation in
CAD patients in the background of higher homocysteine
levels. Our results are in agreement with the recent observa-
tion that global DNA hypermethylation is associated with
inflammation and increased mortality in chronic kidney dis-
ease (CKD) patients especially due to cardiovascular disease
(Stenvinkel et al., 2007).
Aging is known to affect the global DNA methylation in
mammals, and a genome-wide decrease in methylation in
healthy individuals has been observed during aging (Fuke
et al., 2005). We analyzed the correlation of global DNA
methylation with age and observed a negative correlation in
controls as expected, while in CAD patients there was a
trend toward positive correlation, although it was not sta-
tistically significant (data not shown). However, when indi-
viduals in the higher age group (61 to 75 years) were
considered, the global DNA methylation in CAD patients
(median 14.310
3
cpm=mg DNA) was significantly higher
than that in the controls (median 10.110
3
cpm=mg DNA,
p¼0.012, Fig. 4), once again indicating an independent as-
sociation between CAD and DNA hypermethylation.
Homocysteine levels are known to be influenced by diet.
In our study population, almost 46% of the individuals
consumed a vegetarian diet. As expected, the homocysteine
levels were significantly higher in vegetarians (median
15.4 mM) than in the nonvegetarians (13.1 mM, p¼0.0015). We
utilized this parameter for stratification of the cohort and
analyzed the global DNA methylation levels. It was found
that global DNA methylation levels were significantly
higher ( p¼0.0015) in CAD patients following a vegetarian
diet (median 11.710
3
cpm=mg DNA) than in controls (me-
dian 9.210
3
cpm=mg DNA) following a similar diet (Fig. 5A).
However, in the nonvegetarian diet group, global DNA
methylation did not vary significantly between patients and
controls (Fig. 5B). The lower levels of global DNA methyla-
tion in the vegetarian control group are consistent with
higher levels of homocysteine; however, with CAD patients
who are vegetarians, in spite of higher homocysteine levels,
the global DNA methylation is significantly higher. This
again suggests an apparent independent association between
increased global DNA methylation and CAD. We also per-
formed multiple regression analysis in controls and patients
separately to check the association of global DNA methyla-
tion with various parameters, and found that in CAD pa-
tients, global DNA methylation was significantly associated
only with homocysteine levels ( p¼0.003). However, none of
the parameters were associated with global methylation of
DNA in controls.
FIG. 2. Correlation between total homocysteine levels and
global DNA methylation in (A) CAD patients (n¼137,
r¼0.2785, p¼0.001) and (B) controls (n¼150, r¼0.01662,
p>0.05). Y-axis as given in Figure 1.
FIG. 3. A comparison of global DNA methylation levels in
hyperhomocysteinemic cohorts (reference level >12 mM). The
box represents the interquartile range, which contains the 50%
of values. The whiskers are lines that extend the box to the
highest and lowest values, excluding outliers. A line across
the box indicates the median value. The box plot shows that
the global DNA methylation in hyperhomocysteinemic CAD
patients is significantly higher than that in hyperhomocys-
teinemic controls ( p¼0.0005). Y-axis as given in Figure 1.
360 SHARMA ET AL.
Folate and vitamin B12 did not show any significant cor-
relation with global DNA methylation in the study popula-
tion (data not shown).
It has recently been shown that ApoE promoter methylation
varies with homocysteine levels in monocytes in culture (Yi-
Deng et al., 2007b). We therefore analyzed the methylation
status of ApoE promoters using bisulfite sequencing in 15
patients and 15 control samples with varying homocysteine
levels. After bisulfite modification of genomic DNA, PCR
products were amplified and analyzed using bisulfite-specific
primers as mentioned in the Materials and Methods section.
The region included contained 25 CpG sites of which 8 CpG
sites were methylated in most of the patients and controls.
However, we did not find any significant difference in
methylation of this region in patients and controls (Fig. 6).
Even when the patients and controls were classified on the
basis of their homocysteine levels, there was no significant
difference in the methylation pattern (data not shown).
Discussion
The origin of complex diseases is attributed to a combina-
tion of heritable and environmental factors, and epigenetic
modulation could be the basis of the effects of environmental
factors on the disease outcome (van Vliet et al., 2007). DNA
methylation is one of the well-known mediators of epigenetic
effects with several documented instances of impact on dis-
ease phenotype (Tang and Ho, 2007). DNA methylation,
through the localization of gene silencing protein complexes
at the methylated CpG dinucleotides, leads to altered ex-
pression profile (Ehrlich et al., 2001; Wade, 2001). It is known
that cancer cells exhibit a global hypomethylation and CpG
island hypermethylation unlike normal cells (Boltze et al.,
2003; Futscher et al., 2004; Lund and van Lohuizen, 2004;
Yatabe et al., 2004).
The alteration in global DNA methylation along with aging
and dietary habit correlated with pathogenesis is reported in
case of cancer (Liu et al., 2003; Waggoner, 2007). However,
similar investigations in CAD are limited. We undertook the
present study to investigate if epigenetic changes are a part of
the pathogenesis in CAD. We found that the global DNA
FIG. 4. Global DNA methylation in CAD patients and con-
trols based on age groups. Data from CAD and control indi-
viduals in the age group 61 to 75 years is shown. The box
represents the interquartile range, which contains the 50% of
values. The whiskers are lines that extend the box to the
highest and lowest values, excludingoutliers. A line across the
box indicates the median value ( p¼0.0122). Y-axis: as given in
Figure 1.
FIG. 5. Global DNA methylation in CAD patients and
controls based on diet. Data from CAD and healthy con-
trols in (A) vegetarians ( p¼0.0015) and (B) nonvegetarians
(p>0.05). The box represents the interquartile range, which
contains the 50% of values. The whiskers are lines that extend
the box to the highest and lowest values, excluding outliers.
A line across the box indicates the median value. Y-axis: as
given in Figure 1.
GLOBAL DNA METHYLATION AND CORONARY ARTERY DISEASE 361
methylation in peripheral blood is significantly higher in CAD
patients than in controls. Within the cohort of CAD patients,
there is a correlation of plasma homocysteine levels with
higher global methylation. Our results are consistent with a
recent study of chromic kidney patients where an association
of hypermethylation of genomic DNA with inflammation and
increased mortality due to cardiovascular diseases has been
reported (Stenvinkel et al., 2007). The authors analyzed the
global DNA methylation by HpaII=MspI sensitivity assay on
genomic DNA from peripheral blood. We have employed a
similar approach. The CCGG sequences that are the target
sequences in our assays is suitable for genome-wide scan for
methylation in terms of their density and distribution in dif-
ferent chromosomes. HpaII=MspI sensitivity has been utilized
to scan genome-wide promoter methylation by Hatada et al.
(2006).
Aging is known to cause global DNA hypomethylation and
promoter hypermethylation (Fraga and Esteller, 2007). Our
results indicate that there is a trend (albeit not significant) of
global hypomethylation in controls and hypermethylation in
CAD patients with age (data not shown). We also observed
significant hypermethylation in CAD patients in the higher
age group (61 to 75 years) as compared to the controls. Al-
though the exact mechanism of alteration of DNA methyla-
tion is not yet known, it is believed that loss of global DNA
methylation with age may be due to the passive demethyla-
tion of heterochromatic DNA, probably as a result of a pro-
gressive loss of DNMT1 efficacy or defective targeting of the
enzyme by other cofactors (Fraga and Esteller, 2007). Apart
from the general loss of DNA methylation, several regions of
the DNA like the promoter CpG islands of estrogen receptor
(ER), myogenic differentiation antigen 1 (MYOD1), insulin-
like growth factor II (IGF2), and tumor suppressor candidate
33 (N33) become hypermethylated with age. It has been pro-
posed that these methylation changes (global hypomethyla-
tion and promoter hypermethylation) with age are mosaic in
tissues and play a triggering role in age-related diseases like
neoplasia and atherosclerosis (Issa, 2003). We believe that in
CAD patients, age-induced global DNA hypomethylation is
offset by promoter-specific hypermethylation resulting in in-
creased overall DNA methylation in CAD patients, especially
in the higher age group.
It is generally believed that an elevated homocysteine level
would lead to hypomethylation of DNA because increased
homocysteine levels are known to elevate the concentration of
S-adenosyl homocysteine (SAH), which is an inhibitor of
methyltransferases (Yi et al., 2000). In a study focusing on
global DNA methylation status in vascular patients, it was
reported that increased homocysteine levels led to hypo-
methylation of DNA (Castro et al., 2003). The authors reported
that the patients with vascular disease had significantly
higher plasma tHcy and AdoHcy concentrations, and the
DNA hypomethylation status was significantly correlated
with plasma tHcy and AdoHcy but not with plasma Ado-
Met=AdoHcy ratios. This is not consistent with the effect of
SAH as a competitive inhibitor of DNA methyltransferase,
thus indicating that the relationship between homocysteine
levels and DNA methylation in disease background is intri-
cate and not due to inhibitory action of SAH only. In addition,
the sample size in the study was small and also the clinical
attributes pertaining to vascular disease were not restricted to
CAD only (8 patients with stroke, 9 patients with myocardial
infarction, and 15 controls).
In our cohort, the range of plasma homocysteine is similar
between the patients and controls, suggesting that the plasma
level of tHcy alone does not have an effect on methylation.
However, high tHcy is significantly correlated with global
DNA hypermethylation in the background of CAD. We did
not find any significant correlation between the levels of folate
or vitamin B12 and global DNA methylation in our study.
There are reports that support the role of folate in restoring
normal homocysteine levels (Ingrosso et al., 2003), while two
recent reports published from the Norwegian Vitamin trial
(NORVIT) and Heart Outcomes Prevention Evaluation
(HOPE) trials concluded that supplements containing folic
acid, vitamin B12, or vitamin B6 did not significantly reduce
the risk of adverse events in cardiovascular disease patients
although the levels of homocysteine were substantially re-
duced (Bonaa et al., 2006; Lonn et al., 2006), thus questioning
the role of level of vitamin B12=B6 in preventing adverse ef-
fects of homocysteine in CAD. However, in these trials the
epigenetic modulation due to hyperhomocysteinemia has not
been addressed.
The data on DNA methylation from hyperlipidemic ApoE
null mice in aortae as well as peripheral blood cells indicated
both hypo- and hypermethylation of DNA (Lund et al., 2004).
In human macrophage THP-1 cell line, on stimulation of
atherogenic lipid levels, significant DNA hypermethylation
FIG. 6. Methylation pattern of 25 CpG sites in the promoter region of ApoE gene in 15 patients (A) and 15 controls (B). The
filled boxes represent methylated cytosines, while blank boxes represent unmethylated cytosines.
362 SHARMA ET AL.
was observed as compared with untreated cells (Lund et al.,
2004). Based on these results the authors observe that the
initial stages of atherogenesis are associated with alteration in
genomic DNA methylation patterns including hyper- and
hypomethylation, rather than a unidirectional change toward
global hypomethylation (Zaina et al., 2005). The same authors
have reported aberrant methylation in the peripheral blood
also with a higher level of hypomethylation (Lund et al., 2004;
Zaina et al., 2005). Recently, using monocytes in culture, it has
been demonstrated that increase in homocysteine levels leads
to hypermethylation of the ApoE promoter and decrease in its
expression. This was correlated with increase in total choles-
terol, free cholesterol, and cholesteryl ester (Yi-Deng et al.,
2007b). However, we did not find any significant differences
in the methylation pattern in the promoter region of ApoE in
peripheral blood lymphocytes of patients and controls. It
should be noted that methylation differences detected by
Yi-Deng et al. (2007b) in cultured monocytes treated with
homocysteine are significantly different from the samples
used in our study. Additionally, our samples included CAD
patients, which could contribute to further epigenetic alter-
ations.
These results suggest that additional factors, including
nutrition, affect DNA methylation patterns by mechanisms
that are likely to be independent of vitamin B12 or homo-
cysteine levels (Zaina et al., 2005). In this context, it is in-
teresting that we find a significant correlation between
methylation, CAD, and homocysteine levels in vegetarians
when the cohort is stratified with reference to diet. A major
proportion of Indian population follow a strict vegetarian
diet, which results in higher levels of plasma homocysteine as
compared to the global scenario (Refsum et al., 2001; Kumar
et al., 2005). Further, there is no significant difference in global
DNA methylation in vegetarian CAD patients and nonvege-
tarian controls in spite of lower homocysteine levels in the
latter. It remains to be seen if the sequences methylated in
vegetarian CAD patients and the nonvegetarian controls and
patients are the same.
The investigation on the effect of homocysteine on DNA
methylation in relation to cardiovascular diseases in ani-
mal models (Lund et al., 2004; Zaina et al., 2005) and cells
in culture (Yi-deng et al., 2007a, 2007b) and the recent study
on CKD patients (Stenvinkel et al., 2007) support the bidi-
rectional effect of homocysteine leading to both hypo- and
hyper-methylation of DNA and an increase in the activity
of DNMT3a and 3b (Yi-deng et al., 2007a). The promoter of
ApoE gene is hypermethylated under hyperhomocysteine-
mia, hyperlipidemia in spite of the hypomethylation of glo-
bal DNA as measured by MethyLight assay and AluIan
LINE elements as surrogate markers (Yi-Deng et al., 2007b).
Therefore, it can be inferred that the effect is gene specific
and could be influenced by additional factors. However, the
significant difference in global DNA methylation we ob-
served is consistent with the increased activity of DNMT3a
and 3b under hyperhomocysteimic conditions in vascular
cells in culture (Yi-Deng et al., 2007b).
Others and we have shown that elevated levels of homo-
cysteine induce endoplasmic reticulum stress in models such
as yeast and human cell lines (Werstuck et al., 2001; Kumar
et al., 2006). Further, homocysteine-induced ER stress acti-
vates sterol regulatory binding proteins (SREBP), which is
associated with the increased expression of genes involved in
cholesterol transport and metabolism. This has been sug-
gested as a possible mechanism for homocysteine-induced
development and progression of hepatic steatosis and ath-
erosclerotic lesions. Hepatic steatosis is also a hallmark of
chronic alcoholism, and Bonsch et al. (2004) have shown that
in patients with chronic alcoholism, homocysteine levels are
positively correlated with global DNA methylation and hy-
permethylation of homocysteine-induced endoplasmic retic-
ulum protein (HERP) promoter region. This resulted in the
decreased expression of HERP gene, which is an endoplasmic
reticulum stress response gene (Bleich et al., 2006). Recently,
Azfer et al. (2006) have shown the activation of ER stress re-
sponse in the development of ischemic heart disease in mice.
These studies suggest that in CAD patients there are complex
interactions, where elevated levels of homocysteine lead to
the counter intuitive consequence, namely hypermethylation.
The genes coding for ER and HERP may be examples of genes
contributing to the pool of global DNA hypermethylation.
Further studies are required to elucidate the functional con-
sequences of these interactions in CAD.
In summary, our results strongly suggest that an alteration
in DNA methylation profile could be associated with CAD
condition itself. A comparative analysis of the methylation
profile at individual genes in CAD patients is underway to
understand the effect of environment through modulation of
homocysteine levels superimposed on the disease pheno-
type.
Acknowledgments
We are grateful to Dr. Rita Castro, Dr. Dwaipayan Bhar-
adwaj, Dr. Sridhar Sivasubbu, and Dr. Beena Pillai for pro-
viding critical inputs and Dr. Saurabh Ghosh (Indian
Statistical Institute, Kolkata) for inputs in statistical analysis.
We also acknowledge the help of Dr. Elayanambi Sundar-
amoorthy in preparation of the manuscript and Lijo John
for help in data analysis. P.S. and J.K. thank the University
Grants Commission for financial support through a research
fellowship. A.K. is thankful to the Council of Scientific and
Industrial Research for research fellowship. This study was
supported by funds provided by the Department of Bio-
technology, Govt. of India (S.S., G.K., and V.B.), under pro-
ject BT=PR4525=Med=14=533=2003.
References
Ahmad, N., and Bhopal, R. (2004). Burden of non-communicable
diseases in South Asia. Evidence for epidemic of coronary
heart disease in India is weak. BMJ 328, 1499.
Azfer, A., Niu, J., Rogers, L.M., Adamski, F.M., and Kolattuku-
dy, P.E. (2006). Activation of endoplasmic reticulum stress
response during the development of ischemic heart disease.
J Physiol Heart Circ Physiol 291, H1411–H1420.
Bird, A. (2002). DNA methylation patterns and epigenetic
memory. Genes Dev 16, 6–21.
Bleich, S., Lenz, B., Ziegenbein, M., Beutler, S., Frieling, H.,
Kornhuber, J., and Bonsch, D. (2006). Epigenetic DNA hy-
permethylation of the HERP gene promoter induces down-
regulation of its mRNA expression in patients with alcohol
dependence. Alcohol Clin Exp Res 4, 587–591.
Bock, C., Reither, S., Mikeska, T., Paulsen, M., Walter, J., and
Lengauer, T. (2005). BiQ analyzer: visualization and quality
GLOBAL DNA METHYLATION AND CORONARY ARTERY DISEASE 363
control for DNA methylation data from bisulfite sequencing.
Bioinformatics 21, 4067–4068.
Boltze, C., Schneider-Stockm, R., Quednow, C., Hinze, R., Ma-
wrin, C., Hribaschek, A., Roessner, A., and Hoang-Vu, C.
(2003). Silencing of the maspin gene by promoter hyper-
methylation in thyroid cancer. Int J Mol Med 12, 479–484.
Bonaa, K.H., Njolstad, I., Ueland, P.M., Schirmer, H., Tverdal,
A., Steigen, T., Wang, H., Nordrehaug, J.E., Arnesen, E., Ras-
mussen, K., and NORVIT Trial Investigators. (2006). Homo-
cysteine lowering and cardiovascular events after acute
myocardial infarction. N Engl J Med 13, 1629–1632.
Bonsch, D., Lenz, B., Reulbach, U., Kornhuber, J., and Bleich, S.
(2004). Homocysteine associated genomic DNA hypermethy-
lation in patients with chronic alcoholism. J Neural Transm
111, 1611–1616.
Castro, R., Rivera, I., Struys, E.A., Jansen, E.E., Ravasco, P., Ca-
milo, M.E., Blom, H.J., Jakobs, C., and Tavares de Almeida, I.
(2003). Increased homocysteine and S-adenosylhomocysteine
concentrations and DNA hypomethylation in vascular disease.
Clin Chem 49, 1292–1296.
Chao, C.L., Tsai, H.H., Lee, C.M., Hsu, S.M., Kao, J.T., Chien,
K.L., Sung, F.C., and Lee, Y.T. (1999). The graded effect of
hyperhomocysteinemia on the severity and extent of coronary
atherosclerosis. Atherosclerosis 147, 379–386.
Dodu, S.R. (1988). Emergence of cardiovascular disease in de-
veloping countries. Cardiolog 75, 56–64.
Ehrlich, M., Buchanan, K.L., Tsien, F., Jiang, G., Sun, B., Uicker,
W., Weemaes, C.M., Smeets, D., Sperling, K., Belohradsky,
B.H., Tommerup, N., Misek, D.E., Rouillard, J.M., Kuick, R.,
and Hanash, S.M. (2001). DNA methyltransferase 3B muta-
tions linked to the ICF syndrome cause dysregulation of
lymphogenesis genes. Hum Mol Genet 10, 2917–2931.
Feinberg, A.P. (2004). The epigenetics of cancer etiology. Semin
Cancer Biol 14, 427–432.
Fraga, M.F., and Esteller, M. (2007). Epigenetics and aging: the
targets and the marks. Trends Genet 23, 413–418.
Fuke, C., Shimabukuro, M., Petronis, A., Sugimoto, J., Oda, T.,
Miura, K., Miyazaki, T., Ogura, C., Okazaki, Y., and Jinno, Y.
(2005). Age related changes in 5-methylcytosine content in
human peripheral leukocytes and placentas: an HPLC-based
study. Ann Hum Genet 68, 196–204.
Futscher, B.W., O’Meara, M.M., Kim, C.J., Rennels, M.A., Lu, D.,
Gruman, L.M., Seftor, R.E., Hendrix, M.J., and Domann, F.E.
(2004). Aberrant methylation of the maspin promoter is an
early event in human breast cancer. Neoplasia 6, 380–389.
Hatada, I., Fukasawa, M., Kimura, M., Morita, S., Yamada, K.,
Yoshikawa, T., Yamanaka, S., Endo, C., Sakurada, A., Sato, M.,
Kondo, T., Horii, A., Ushijima, T., and Sasaki, H. (2006).
Genome-wide profiling of promoter methylation in human.
Oncogene 25, 3059–3064.
Ingrosso, D., Cimmino, A., Perna, A.F., Masella, L., de Santo,
N.G., de Bonis, M.L., Vacca, M., D’Esposito, M., D’Urso, M.,
Galletti, P., and Zappia, V. (2003). Folate treatment and un-
balanced methylation and changes of allelic expression induced
by hyperhomocysteinaemia in patients with uraemia. Lancet
361, 1693–1699.
Issa, J.P. (2003). Age-related epigenetic changes and the immune
system. Clin Immunol 109, 103–108.
Ji, A.J., Savon, S.R., and Jacobsen, D.W. (1995). Determination of
total serum sulfite by HPLC with fluorescence detection. Clin
Chem 41, 897–903.
Kumar, A., John, L., Alam, M.M., Gupta, A., Sharma, G., Pillai,
B., and Sengupta, S. (2006). Homocysteine- and cysteine-
mediated growth defect is not associated with induction of
oxidative stress response genes in yeast. Biochem J 396, 61–69.
Kumar, J., Das, S.K., Sharma, P., Karthikeyan, G., Ramakrishnan,
L., and Sengupta, S. (2005). Homocysteine levels are associ-
ated with MTHFR A1298C polymorphism in Indian popula-
tion. J Hum Genet 50, 655–663.
Leeder, S., Raymond, S., Greenberg, H., Liu, H., and Esson, K.
(2004). A Race Against Time. The Challenge of Cardiovascular
Disease in Developing Economies (Columbia University, NY).
Liu, L., Wylie, R.C., Andrews, L.G., and Tollefsbol, T.O. (2003).
Aging, cancer and nutrition: the DNA methylation connection.
Mech Ageing Dev 124, 989–998.
Lonn, E., Yusuf, S., Arnold, M.J., Sheridan, P.,Pogue, J., Micks, M.,
McQueen, M.J., Probstfield, J., Fodor, G., Held, C., Genest, J., Jr.,
and Heart Outcomes Prevention Evaluation (HOPE) 2 In-
vestigators. (2006). Homocysteine lowering with folic acid and B
vitamins in vascular disease. N Engl J Med 354, 1567–1577.
Lund, A.H., and van Lohuizen, M. (2004). Epigenetics and
cancer. Genes Dev 18, 2315–2335.
Lund, G., Andersson, L., Lauria, M., Lindholm, M., Fraga, M.F.,
Villar-Garea, A., Ballestar, E., Esteller, M., and Zaina, S. (2004).
DNA methylation polymorphisms precede any histological
sign of atherosclerosis in mice lacking apolipoprotein E. J Biol
Chem 279, 29147–29154.
Moss, T.J., and Mallrats, L.L. (2007). Connections between epi-
genetic gene silencing and human disease. Mutation Res 618,
163–174.
Refsum, H., Yajnik, C.S., Gadkari, M., Schneede, J., Vollset, S.E.,
Orning, L., Guttormsen, A.B., Joglekar, A., Sayyad, M.G., Ul-
vik, A., and Ueland, P.M. (2001). Hyperhomocysteinemia and
elevated methylmalonic acid indicate a high prevalence of
cobalamin deficiency in Asian Indians. Am J Clin Nutr 74,
233–241.
Robinson, K., Mayer, E.L., Dave, P., Miller, M.S., Green, R., van
Lente, F., Gupta, A., Marchant, K.K., Savon, S.R., Selhub, J.,
Nissen, S.E., Kutner, M., Topol, E.J., and Jacobsen, D.W.
(1995). Hyperhomocysteinemia and low pyridoxal phosphate
common and independent reversible risk factors for coronary
artery disease. Circulation 92, 2825–2830.
Shames, D.S., Minna, J.D., and Gazdar, A.F. (2007). DNA Me-
thylation in health, disease and cancer. Curr Mol Med 1, 85–102.
Sharma, P., Senthilkumar, R.D., Brahmachari, V., Sundar-
amoorthy, E., Mahajan, A., Sharma, A., and Sengupta, S. (2006).
Mining literature for a comprehensive pathway analysis a case
study for retrieval of homocysteine related genes for genetic
and epigenetic studies. Lipids Health Dis 5, 1.
Stenvinkel, P., Karimi, M., Johansson, S., Axelsson, J., Suliman,
M., Lindholm, B., Heimbu
¨rger, O., Barany, P., Alvestrand,
A., Nordfors, L., Qureshi, A.R., Ekstro
¨m, T.J., and Schalling,
M. (2007). Impact of inflammation on epigenetic DNA
methylation—a novel risk factor for cardiovascular disease?
J Intern Med 261, 488–499.
Tang, W.Y., and Ho, S.M. (2007). Epigenetic reprogramming and
imprinting in origins of disease. Rev Endocr Metab Disord 8,
173–182.
Turner, B.M. (2007). Defining an epigenetic code. Nature Cell
Biol 9, 2–6.
Ulrey, C.L., Liu, L., Andrews, L.G., and Tollefsbol, T.O. (2005).
The impact of metabolism on DNA methylation. Hum Mol
Genet 14, R139–R147.
van Guldener, C., Stam, F., and Stehouwer, C.D. (2005). Hy-
perhomocysteinaemia in chronic kidney disease: focus on
transmethylation. Clin Chem Lab Med 43, 1026–1031.
364 SHARMA ET AL.
van Vliet, J., Oates, N.A., and Whitelaw, E. (2007). Epigenetic
mechanisms in the context of complex diseases. Cell Mol Life
Sci 64, 1531–1538.
Wade, P.A. (2001). Methyl CpG-binding proteins and tran-
scriptional repression. Bioessays 23, 1131–1137.
Waggoner, D. (2007). Mechanism of disease: epigenetics. Semin
Pediatr Neurol 1, 7–14.
Wald, D.S., Law, M., and Morris, J. (2003). Serum homocysteine
and the severity of coronary artery disease. Thromb Res 111,
55–57.
Werstuck, G.H., Lentz, S.R., Dayal, S., Hossain, G.S., Sood, S.K.,
Shi, Y.Y., Zhou, J., Maeda, N., Krisans, S.K., Malinow, M.R.,
and Austin, R.C. (2001). Homocysteine-induced endoplasmic
reticulum stress causes dysregulation of the cholesterol and
triglyceride biosynthetic pathways. J Clin Invest 107, 1263–
1273.
Wilson, P.W., D’Agostino, R.B., Levy, D., Belanger, A.M., Sil-
bershatz, H., and Kannel, B. (1998). Prediction of coronary heart
disease using risk factor categories. Circulation 97, 1837–1847.
Winkelmann, B.R., and Hager, J. (2000). Genetic variation in cor-
onary heart disease and myocardial infarction: methodological
overview and clinical evidence. Pharmacogenomics 1, 73–94.
Yatabe, Y., Mitsudomi, T., and Takahashi, T. (2004). Maspin
expression in normal lung and non-small-cell lung cancers:
cellular property-associated expression under the control of
promoter DNA methylation. Oncogene 23, 4041–4049.
Yi, P., Melnyk, S., Pogribna, M., Pogribny, I.P., Hine, R.J., and
James, S.J. (2000). Increase in plasma homocysteine associated
with parallel increases in plasma S-adenosylhomocysteine and
lymphocyte DNA hypomethylation. J Biol Chem 275, 29318–
29323.
Yi-deng, J., Jianzhong, Z., Ying, H., Juan, S., Jinge, Z., Shenglan,
W., Xiaoqun, H., and Shuren, W. (2007a). Homocysteine-
mediated expression of SAHH, DNMTs, MBD2, and DNA
hypomethylation potential pathogenic mechanism in VSMCs.
DNA Cell Biol 26, 603–611.
Yi-Deng, J., Tao, S., Ping, Z.H., Tuan, X.J., Jun, C., Zhong, L.G.,
and Ren, W.S. (2007b). Folate and ApoE DNA methylation
induced by homocysteine in human monocytes. DNA Cell
Biol 26, 1–8.
Yoo, J.H., Park, J.E., Hong, K.P., Lee, S.H., Kim, D.K., Lee, W.R.,
and Park, S.C. (1999). Moderate hyperhomocyst(e)inemia is
associated with the presence of coronary artery disease and
the severity of coronary atherosclerosis in Koreans. Thromb
Res 94, 45–52.
Zaina, S., Lindholm, M.W., and Lund, G. (2005). Nutrition and
aberrant DNA methylation patterns in atherosclerosis: more
than just hyperhomocysteinemia? J Nutr 135, 5–8.
Address reprint requests to:
Shantanu Sengupta, Ph.D.
Institute of Genomics and Integrative Biology
Mall Road
Delhi 110007
India
E-mail: shantanus@igib.res.in
Vani Brahmachari, Ph.D.
Dr. B.R. Ambedkar Centre for Biomedical Research
University of Delhi
Delhi 110007
India
E-mail: vbrahmachari@acbr.du.ac.in
Received for publication October 22, 2007; received in re-
vised form March 17, 2008; accepted March 25, 2008.
GLOBAL DNA METHYLATION AND CORONARY ARTERY DISEASE 365
This article has been cited by:
1. M. Talikka, N. Sierro, N. V. Ivanov, N. Chaudhary, M. J. Peck, J. Hoeng, C. R. E. Coggins, M. C. Peitsch. 2012. Genomic
impact of cigarette smoke, with application to three smoking-related diseases. Critical Reviews in Toxicology 1-13. [CrossRef]
2. Roosmarijn Luttmer, Annemieke M. Spijkerman, Robert M. Kok, Carel Jakobs, Henk J. Blom, Erik H. Serne, Jacqueline
M. Dekker, Yvo M. Smulders. 2012. Metabolic syndrome components are associated with DNA hypomethylation. Obesity
Research & Clinical Practice . [CrossRef]
3. F.I. Milagro, M.L. Mansego, C. De Miguel, J.A. Martínez. 2012. Dietary Factors, Epigenetic Modifications And Obesity
Outcomes: Progresses And Perspectives. Molecular Aspects of Medicine . [CrossRef]
4. Bram P. Prins, Vasiliki Lagou, Folkert W. Asselbergs, Harold Snieder, Jingyuan Fu. 2012. Genetics of coronary artery disease:
Genome-wide association studies and beyond. Atherosclerosis . [CrossRef]
5. M. S. Pearce, J. C. McConnell, C. Potter, L. M. Barrett, L. Parker, J. C. Mathers, C. L. Relton. 2012. Global LINE-1 DNA
methylation is associated with blood glycaemic and lipid profiles. International Journal of Epidemiology 41:1, 210-217.
[CrossRef]
6. Susan E Puumala, Heather H Nelson, Julie A Ross, Ruby HN Nguyen, Mark A Damario, Logan G Spector. 2012. Similar
DNA methylation levels in specific imprinting control regions in children conceived with and without assisted reproductive
technology: a cross-sectional study. BMC Pediatrics 12:1, 33. [CrossRef]
7. Boda Zhou, Andriana Margariti, Qingbo XuThe Role of Epigenetics in Cardiovascular Disease 395-414. [CrossRef]
8. Jan Bressler, Lawrence C. Shimmin, Eric Boerwinkle, James E. Hixson. 2011. Global DNA methylation and risk of subclinical
atherosclerosis in young adults: The Pathobiological Determinants of Atherosclerosis in Youth (PDAY) study. Atherosclerosis
. [CrossRef]
9. Anthony R. Cyr , Frederick E. Domann . 2011. The Redox Basis of Epigenetic Modifications: From Mechanisms to Functional
Consequences. Antioxidants & Redox Signaling 15:2, 551-589. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF
with Links]
10. Christopher A. Hamm, Fabricio F. Costa. 2011. The impact of epigenomics on future drug design and new therapies. Drug
Discovery Today 16:13-14, 626-635. [CrossRef]
11. Erin L. Kinnally, Caroline Feinberg, David Kim, Kerel Ferguson, Rudolph Leibel, Jeremy D. Coplan, J. John Mann. 2011.
DNA methylation as a risk factor in the effects of early life stress. Brain, Behavior, and Immunity . [CrossRef]
12. Claudio Napoli. 2011. Developmental Mechanisms Involved in the Primary Prevention of Atherosclerosis and Cardiovascular
Disease. Current Atherosclerosis Reports 13:2, 170-175. [CrossRef]
13. Asmita Kulkarni , Preeti Chavan-Gautam , Savita Mehendale , Hemlata Yadav , Sadhana Joshi . 2011. Global DNA
Methylation Patterns in Placenta and Its Association with Maternal Hypertension in Pre-Eclampsia. DNA and Cell Biology
30:2, 79-84. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
14. L. Badimon, R. F. Storey, G. Vilahur. 2011. Update on lipids, inflammation and atherothrombosis. Thrombosis and
Haemostasis 105:Suppl. 1, S34-S42. [CrossRef]
15. Elissa H Wilker, Stacey E Alexeeff, Helen Suh, Pantel S Vokonas, Andrea Baccarelli, Joel Schwartz. 2011. Ambient
pollutants, polymorphisms associated with microRNA processing and adhesion molecules: the Normative Aging Study.
Environmental Health 10:1, 45. [CrossRef]
16. Magdalena Kowalewska, Radoslawa Nowak, Magdalena Chechlinska. 2010. Implications of cancer-associated systemic
inflammation for biomarker studies. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer 1806:2, 163-171. [CrossRef]
17. D. Brinza, M. Schultz, G. Tesler, V. Bafna. 2010. RAPID detection of gene-gene interactions in genome-wide association
studies. Bioinformatics 26:22, 2856-2862. [CrossRef]
18. José M. Ordovás, Caren E. Smith. 2010. Epigenetics and cardiovascular disease. Nature Reviews Cardiology 7:9, 510-519.
[CrossRef]
19. Smriti M. Krishna, Anthony E. Dear, Paul E. Norman, Jonathan Golledge. 2010. Genetic and epigenetic mechanisms and
their possible role in abdominal aortic aneurysm. Atherosclerosis 212:1, 16-29. [CrossRef]
20. Jaroslav A. Hubacek, Vladimír Stan#k, Marie Gebauerová, Alexandra Pilip#incová, Dana Dlouhá, Rudolf Poledne, Michal
Aschermann, Hana Skalická, Jana Matoušková, Andreas Kruger. 2010. A FTO variant and risk of acute coronary syndrome.
Clinica Chimica Acta 411:15-16, 1069-1072. [CrossRef]
21. Rutger J. Wierda, Sacha B. Geutskens, J. Wouter Jukema, Paul H.A. Quax, Peter J. Van Den Elsen. 2010. Epigenetics in
atherosclerosis and inflammation. Journal of Cellular and Molecular Medicine 14:6a, 1225-1240. [CrossRef]
22. Magdalena Chechlinska, Magdalena Kowalewska, Radoslawa Nowak. 2010. Systemic inflammation as a confounding factor
in cancer biomarker discovery and validation. Nature Reviews Cancer 10:1, 2-3. [CrossRef]
23. Mikko P. Turunen, Einari Aavik, Seppo Ylä-Herttuala. 2009. Epigenetics and atherosclerosis. Biochimica et Biophysica Acta
(BBA) - General Subjects 1790:9, 886-891. [CrossRef]
... Study carried out in 2017, in Chinese population with Acute Coronary Syndrome (ACS) revealed some DNA methylation-based biomarkers which are relevant to several pathways [17] Interestingly, in French-Canadian population, DNA hypomethylation was reported that in the promoter region of TNNT1 gene which was associated with dyslipidemia and the risk for CAD [18]. In India, global DNA methylation is much higher in CAD patients and have been associated with hyperhomocysteinemia, which is an independent risk factor for CAD [19,20]. Our previously reported epigenome wide association study (EWAS) [21],using a comprehensive microarray analysis with a high coverage BeadChip, revealed significant top CpG sites which are involved in the pathogenesis of CAD. ...
... A study conducted in Delhi, India in 2008 and subsequently in 2014 [19,20] associated global methylation levels to the development of CAD. However, they included CAD negative controls based on stress test as opposed to our study which included only angiographically negative controls. ...
Article
Full-text available
Background DNA methylation, one of the most stable forms of epigenetic modification is associated with the development and progression of coronary artery disease (CAD). Our previously reported study on epigenome-wide microarray analysis showed significantly methylated CpG sites. Top 5 significant CpGs from HLA gene were selected and analysed by Pyrosequencing (PSQ) to determine their association with severity of CAD. Methods Blood samples of 50-age matched angiographically CAD positive male cases with 50 angiographically CAD negative male controls were subjected to lipid profile estimation and PSQ for methylation level analysis. Findings and subgroup analysis were evaluated by Mann–Whitney U; Kruskal–Wallis’ rank test and two-way ANOVA by MedCalc (v19.6). Results Methylation levels in HLA-DQA1 for cg10217052 was 78.5 (37–85) and 76.5 (24–84); cg09411910 was 81 (72.0 to 93.0) and 81.5 (50.0 to 89.0) in cases and controls respectively. Levels in HLA-DQB1-cg03344051, were 28.88 + 9.41 for cases and 30.36 + 9.37 in controls. For HLA-DRB1-cg07889003, levels in cases and controls were 15.5 (5.00–39.00) and 10.5 (5.00–29.0); while in cg08269402 were 52 (16–65) and 42.5 (17–61) respectively. No association was observed between methylation levels and lipid profile. cg03344051, cg07889003 and cg08269402 were significantly differentiated in double or triple vessel disease (DVD or TVD) as compared to single vessel disease (SVD) suggesting an increase in the extent of methylation with the increase in CAD severity. Conclusion The present study shows significant increase in the extent of methylation in 3 CpG sites in DVD/TVD cases as compared to SVD cases. Additionally, a novel site, cg07889003 identified in our discovery phase has shown association with the severity of CAD.
... To date, multiple publications supported the importance of DNA methylation in a broad spectrum of cardiovascular diseases, including atherogenesis (47), coronary artery diseases (48,49), dilated cardiomyopathy (50) and heart failure (51)(52)(53). In our analysis, we identified three pathways that could be potentially associated with different methylation (Figure 4). ...
Article
Full-text available
Background The mode of delivery represents an epigenetic factor with potential to affect further development of the individual by multiple mechanisms. DNA methylation may be one of them, representing a major epigenetic mechanism involving direct chemical modification of the individual’s DNA. This pilot study aims to examine whether a specific mode of delivery induces changes of DNA methylation by comparing the umbilical cord blood and peripheral blood of the newborns. Methods Blood samples from infants born by vaginal delivery and caesarean section were analysed to prepare the Methylseq library according to NEBNext enzymatic Methyl-seq Methylation Library Preparation Kit with further generation of target-enriched DNA libraries using the Twist Human Methylome Panel. DNA methylation status was determined using Illumina next-generation sequencing (NGS). Results We identified 168 differentially methylated regions in umbilical cord blood samples and 157 regions in peripheral blood samples. These were associated with 59 common biological, metabolic and signalling pathways for umbilical cord and peripheral blood samples. Conclusion Caesarean section is likely to represent an important epigenetic factor with the potential to induce changes in the genome that could play an important role in development of a broad spectrum of disorders. Our results could contribute to the elucidation of how epigenetic factors, such as a specific mode of delivery, could have adverse impact on health of an individual later in their life.
... We found a total of 4264 and 703 unique hypermethylated and 290 and 127 hypomethylated DMRs in the promoter region of male and female liver, respectively. This corroborates with our previous study that showed a significant association of DNA hypermethylation with hyperhomocysteinemia due to vitamin B12 deficiency [15,40]. Methylation levels at~75% to 90% of DMRs were reversed upon vitamin B12 RC, suggesting a causal inference (Supplementary Table 4). ...
Article
Background: Maternal vitamin B12 (B12) deficiency plays a vital role in fetal programming, as corroborated by previous studies on murine models and longitudinal human cohorts. Objective: This study assessed the effects of diet-induced maternal B12 deficiency on F1 offspring in terms of cardiometabolic health and normalization of these effects by maternal peri-conceptional B12 supplementation. Methods: A diet-induced maternal B12 deficient Wistar rat model was generated where female rats were either fed a control AIN-76A diet (with 0.01 g/Kg B12) or same diet with B12 removed. Females from B12 deficient group were mated with males on control diet. A subset of B12 deficient females was repleted with B12 on day 1 of conception. The offspring in F1 generation were assessed for changes in body composition, plasma biochemical and molecular changes in the liver. A multi-omics approach was used to get a mechanistic insight into the changes in the offspring liver. Results: We show that a 36% reduction in plasma B12 levels during pregnancy in F0 females can lead to continued B12 deficiency (60-70% compared to control) in the F1 offspring and program them for cardio-metabolic adversities. These adversities like high triglycerides and low HDL were seen only among F1 males but not females. DNA methylome analysis in the liver of F1 3-month-old offspring highlights sexual dimorphism in the alteration of methylation status of genes critical to signaling processes. Proteomics and targeted metabolomics analysis confirm that this sex-specific alterations occur through modulations in PPAR signaling and steroid hormone biosynthesis pathway. Repletion of deficient mothers with B12 at conception normalizes most of the molecular and biochemical changes. Conclusions: Maternal B12 deficiency has a programming effect on the next generation and increases the risk for cardio-metabolic syndrome in a sex-specific manner. Normalization of the molecular risk markers on B12 supplementation indicates a causal role.
... DNA hypermethylation can directly inhibit transcription, or gene expression transcriptional silencing (Qureshi and Mehler 2010). Changes in DNA methylation have been associated with cardiovascular and cerebrovascular diseases (Sharma et al. 2008). RNA sequencing is a powerful transcriptome analysis method that provides a global unbiased transcriptomic analysis with high sensitivity and specificity. ...
Article
Full-text available
Inflammation and DNA methylation have been reported to play key roles in intracerebral hemorrhage (ICH). This study aimed to investigate new diagnostic biomarkers associated with inflammation and DNA methylation using a comprehensive bioinformatics approaches. GSE179759 and GSE125512 were collected from the Gene Expression Omnibus database, and 3222 inflammation-related genes (IFRGs) were downloaded from the Molecular Signatures Database. Key differentially expressed methylation-regulated and inflammation-related genes (DE-MIRGs) were identified by overlapping methylation-regulated differentially expressed genes (MeDEGs) between patients with ICH and control samples, module genes from weighted correlation network analysis, and IFRGs. Functional annotation of DE-MIRGs was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). A protein–protein interaction (PPI) network was constructed to clarify the interrelationships between different DE-MIRGs. The key genes were categorized by least absolute shrinkage selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE), and gene set enrichment analysis (GSEA). A total of 22 DE-MIRGs were acquired from 451 MeDEGs, 3222 IFRGs, and 302 module genes, and were mainly enriched in the GO terms of wound healing, blood coagulation, and hemostasis; and the KEGG pathways of PI3K/Akt signaling, focal adhesion, and regulation of actin cytoskeleton. A PPI network with 22 nodes and 87 edges was constructed based on the 22 DE-MIRGs, 11 of which were selected for key gene selection. Two 2 key genes (SELP and S100A4) were identified using LASSO and SVM-RFE. Finally, SELP was mainly enriched in cell morphogenesis involved in differentiation, cytoplasmic translation, and actin binding of GO terms, and the KEGG pathway including endocytosis, focal adhesion, and platelet activation. S100A4 was mainly enriched in GO terms including mitochondrial inner membrane; mitochondrial respirasome and lysosomal membrane; and the KEGG pathway of oxidative phosphorylation, regulation of actin cytoskeleton, and chemical carcinogenesis-reactive oxygen species. Twenty-two DE-MIRGs-associated inflammation and DNA methylation were identified between patients with ICH and normal controls, and two key genes (SELP and S100A4) were identified and regarded as biomarkers for ICH, which could provide the research foundation for further investigation of the pathological mechanism of ICH.
... To date multiple publications supported importance of DNA methylation in broad spectrum of cardiovascular diseases, including atherogenesis(Ying, Hassanain et al. 2000), coronary artery diseases(Sharma, Kumar et al. 2008, Sharma, Garg et al. 2014), dilated cardiomyopythy (Haas, Frese et al. 2013) and heart failure (Movassagh, Choy et al. 2010, Movassagh, Choy et al. 2011,Koczor, Lee et al. 2013). In our analysis were identi ed three pathways that could be potentially associated with different methylation(Fig. ...
Preprint
Full-text available
Background: A type of delivery represents an epigenetic factor with potential to affect further development by multiple mechanisms. One of them can be DNA methylation, which represents major epigenetic mechanism inolving direct chemical modification to the DNA. This study aims to examine wheather type of delivery induces changes of DNA methylation comparing the umbilical cord blood and periphery blood of the newborns. Methods: The blood samples from infants, born by vaginal delivery and cesarean section, were used to preparation of the Methylseq library according to NEBNext enzymatic Methyl-seq Methylation Library Preparation Kit with further generation of target-enriched DNA libraries by Twist Human Methylome Panel. DNA methylation status was measured using Illumina next-generation sequencing. Results: We identified 168 differentially methylated regions in umbilical cord blood samples and 157 regions in peripheral blood samples. These were associated with 59 common biological, metabolic and signaling pathways for umbilical cord and peripheral blood samples. Conclusions: Cesarean section probably represents important epigenetic factor with potential to induce changes in genome, which could play an important role in development of broad spectrum of disorders. Our results could contribute to elucidate how epigenetic factors, like type of delivery, could have an adverse impact on health later in the life.
... The global methylation of DNA was investigated and reported by N = 9 studies. Six of them reported a moderate to large increase in global methylation level associated with the disease of interest: In a case-control study of coronary artery disease (N = 137 cases and N = 150 controls), global DNA methylation was quantified using radiolabeling with incorporation of [ 3 H] dCTP (Deoxycytidine [3H] triphosphate tetra-sodium salt) [23]. Another study investigated global methylation levels in patients with and without acute coronary syndrome (n = 190) using an Enzyme-Linked Immunosorbent Assay (ELISA) [48]. ...
Article
Full-text available
Background: Cardiovascular disease (CVD) is the leading cause of death worldwide and considered one of the most environmentally driven diseases. The role of DNA methylation in response to the individual exposure for the development and progression of CVD is still poorly understood and a synthesis of the evidence is lacking. Results: A systematic review of articles examining measurements of DNA cytosine methylation in CVD was conducted in accordance with PRISMA (preferred reporting items for systematic reviews and meta-analyses) guidelines. The search yielded 5,563 articles from PubMed and CENTRAL databases. From 99 studies with a total of 87,827 individuals eligible for analysis, a database was created combining all CpG-, gene- and study-related information. It contains 74,580 unique CpG sites, of which 1452 CpG sites were mentioned in ≥ 2, and 441 CpG sites in ≥ 3 publications. Two sites were referenced in ≥ 6 publications: cg01656216 (near ZNF438) related to vascular disease and epigenetic age, and cg03636183 (near F2RL3) related to coronary heart disease, myocardial infarction, smoking and air pollution. Of 19,127 mapped genes, 5,807 were reported in ≥ 2 studies. Most frequently reported were TEAD1 (TEA Domain Transcription Factor 1) and PTPRN2 (Protein Tyrosine Phosphatase Receptor Type N2) in association with outcomes ranging from vascular to cardiac disease. Gene set enrichment analysis of 4,532 overlapping genes revealed enrichment for Gene Ontology molecular function "DNA-binding transcription activator activity" (q = 1.65 × 10-11) and biological processes "skeletal system development" (q = 1.89 × 10-23). Gene enrichment demonstrated that general CVD-related terms are shared, while "heart" and "vasculature" specific genes have more disease-specific terms as PR interval for "heart" or platelet distribution width for "vasculature." STRING analysis revealed significant protein-protein interactions between the products of the differentially methylated genes (p = 0.003) suggesting that dysregulation of the protein interaction network could contribute to CVD. Overlaps with curated gene sets from the Molecular Signatures Database showed enrichment of genes in hemostasis (p = 2.9 × 10-6) and atherosclerosis (p = 4.9 × 10-4). Conclusion: This review highlights the current state of knowledge on significant relationship between DNA methylation and CVD in humans. An open-access database has been compiled of reported CpG methylation sites, genes and pathways that may play an important role in this relationship.
... Turunen et al. (2009) showed that the genomic DNA isolated from human atherosclerotic lesions was hypomethylated. Despite that the data from the patients of the coronary disease was inconsistent with decreases and increases in methylation status, genes associated with atherosclerosis, such as extracellular superoxide dismutase, estrogen receptor a, endothelial nitric oxide synthase, and 15lipoxygenase, have been found to cause specific methylation changes, mostly hypermethylation (Lee and Wang, 1999;Castro et al., 2006;Sharma et al., 2008). ...
Article
Full-text available
Fetal Origins of Adult Diseases" (FOAD) or "Barker hypothesis," also known as the developmental origins of health and diseases (DoHaD), was initially proposed by David Barker in the 1980s. Progress in past 2 to 3 decades demonstrated that many adult disorders, including hypertension, diabetes, obesity, cancer, and others, could be linked to poor development resulting from in utero insults. Utero-environments play a critical role in fetal development. Because the placenta and umbilical cord are the only important connections between the fetus and mother in the uterus, this review pays special attention to recent research and progress in the study of the relationship between those tissues and FOAD. We discuss the conception and possible underlying mechanisms of FOAD, and focus on cardiovascular diseases and epigenetic mechanisms. This review also summarizes physiology, pathology, and the important roles of fetoplacental vasculature, which might contribute to FOAD as initiators. We proposed the "Three hits" hypothesis that highlights the importance of intrauterine and early postnatal factors as contributors to FOAD, which could be significant for early prevention and treatments of FOAD.
Article
Full-text available
Cardiovascular diseases, such as atherosclerosis and heart failure, are major public health problems and leading causes of mortality in the world. Many risk factors for the disease, e.g. smoking, hyperlipidemia, and hypertension, have been identified, but the mechanism of the disease development remains clarified. Recently, studies on epigenetic modification in cardiovascular system have discovered the crucial role in the pathogenesis of cardiovascular disease. For instance, DNA methylation and histone accetylation were identified in the vessel wall during arteriosclerotic formation in animal models and human samples. Epigenetic modification can directly or indirectly influence vascular cell growth, migration, and apoptosis. In this chapter, we will update the progress on epigenetic modification in cardiovascular disease, discuss the molecular mechanisms on how histone acetylation results in alterations in cell functions, and highlight the challenging issues in the research field.
Article
Full-text available
There is considerable evidence that inhaled toxicants such as cigarette smoke can cause both irreversible changes to the genetic material (DNA mutations) and putatively reversible changes to the epigenetic landscape (changes in the DNA methylation and chromatin modification state). The diseases that are believed to involve genetic and epigenetic perturbations include lung cancer, chronic obstructive pulmonary disease (COPD), and cardiovascular disease (CVD), all of which are strongly linked epidemiologically to cigarette smoking. In this review, we highlight the significance of genomics and epigenomics in these major smoking-related diseases. We also summarize the in vitro and in vivo findings on the specific perturbations that smoke and its constituent compounds can inflict upon the genome, particularly on the pulmonary system. Finally, we review state-of-the-art genomics and new techniques such as high-throughput sequencing and genome-wide chromatin assays, rapidly evolving techniques which have allowed epigenetic changes to be characterized at the genome level. These techniques have the potential to significantly improve our understanding of the specific mechanisms by which exposure to environmental chemicals causes disease. Such mechanistic knowledge provides a variety of opportunities for enhanced product safety assessment and the discovery of novel therapeutic interventions.
Article
Full-text available
Nutritional factors play a life-long role in human health. Indeed, there is growing evidence that one of the mechanisms by which nutrients and bioactive compounds affect metabolic traits is epigenetics. Complex interactions among food components and histone modifications, DNA methylation, non-coding RNA expression and chromatin remodeling factors lead to a dynamic regulation of gene expression that controls the cellular phenotype. Although perinatal period is the time of highest phenotypic plasticity, contributing largely to developmental programming, also during adulthood there is evidence about a nutritional influence on epigenetic regulation. Similarly to type 2 diabetes, hypertension, atherosclerosis and other metabolic disorders, obesity predisposition and weight loss outcomes have been repeatedly associated to changes in epigenetic patterns. Different non-nutritional risk factors that usually accompany obesity seem also to be involved in these epigenetic modifications, especially hyperglycemia, inflammation, hypoxia and oxidative stress. There are currently three major objectives in epigenetic research in relation to obesity: to search for epigenetic biomarkers to predict future health problems or detect the individuals at most risk, to understand the obesity-related environmental factors that could modulate gene expression by affecting epigenetic mechanisms, and to study novel therapeutic strategies based on nutritional or pharmacological agents that can modify epigenetic marks. At this level, the major tasks are: development of robust epigenetic biomarkers of weight regulation, description of those epigenetic marks more susceptible to be modified by dietary exposures, identification of the active ingredients (and the doses) that alter the epigenome, assessment of the real importance of other obesity-related factors on epigenetic regulation, determination of the period of life in which best results are obtained, and understanding the importance of the inheritance of these epigenetic marks.
Article
Full-text available
Hepatic steatosis is common in patients having severe hyperhomocysteinemia due to deficiency for cystathionine beta-synthase. However, the mechanism by which homocysteine promotes the development and progression of hepatic steatosis is unknown. We report here that homocysteine-induced endoplasmic reticulum (ER) stress activates both the unfolded protein response and the sterol regulatory element-binding proteins (SREBPs) in cultured human hepatocytes as well as vascular endothelial and aortic smooth muscle cells. Activation of the SREBPs is associated with increased expression of genes responsible for cholesterol/triglyceride biosynthesis and uptake and with intracellular accumulation of cholesterol. Homocysteine-induced gene expression was inhibited by overexpression of the ER chaperone, GRP78/BiP, thus demonstrating a direct role of ER stress in the activation of cholesterol/triglyceride biosynthesis. Consistent with these in vitro findings, cholesterol and triglycerides were significantly elevated in the livers, but not plasmas, of mice having diet-induced hyperhomocysteinemia. This effect was not due to impaired hepatic export of lipids because secretion of VLDL-triglyceride was increased in hyperhomocysteinemic mice. These findings suggest a mechanism by which homocysteine-induced ER stress causes dysregulation of the endogenous sterol response pathway, leading to increased hepatic biosynthesis and uptake of cholesterol and triglycerides. Furthermore, this mechanism likely explains the development and progression of hepatic steatosis and possibly atherosclerotic lesions observed in hyperhomocysteinemia.
Article
Full-text available
While a possible link between assisted reproductive technology (ART) and rare imprinting disorders has been found, it is not clear if this is indicative of subtler disruptions of epigenetic mechanisms. Results from previous studies have been mixed, but some methylation differences have been observed. Children conceived through ART and children conceived spontaneously were recruited for this cross-sectional study. Information about reproductive history, demographic factors, birth characteristics, and infertility treatment was obtained from maternal interview and medical records. Peripheral blood lymphocytes and buccal cell samples were collected from participating children. Methylation analysis was performed on five loci using pyrosequencing. Statistical analysis of methylation differences was performed using linear regression with generalized estimating equations. Results are reported as differences with 95% confidence intervals (CI). A total of 67 ART children and 31 spontaneously conceived (SC) children participated. No significant difference in methylation in lymphocyte samples was observed between groups for any loci. Possible differences were found in buccal cell samples for IGF2 DMR0 (Difference: 2.07; 95% confidence interval (CI): -0.28, 4.42; p = 0.08) and IGF2R (Difference: -2.79; 95% CI: -5.74, 0.16; p = 0.06). Subgroup analysis indicated potential lower methylation in those whose parents used ART for unexplained infertility. Observed differences in methylation between the ART and SC groups were small for all loci in the two sample types examined and no statistical differences were observed. It is still unclear whether or not small differences observed in several studies represent a real difference between groups and if this difference is biologically meaningful. Larger studies with long term follow-up are needed to fully answer these questions.
Article
Full-text available
Patterns of DNA methylation change with age and these changes are believed to be associated with the development of common complex diseases. The hypothesis that Long Interspersed Nucleotide Element 1 (LINE-1) DNA methylation (an index of global DNA methylation) is associated with biomarkers of metabolic health was investigated in this study. Global LINE-1 DNA methylation was quantified by pyrosequencing in blood-derived DNA samples from 228 individuals, aged 49-51 years, from the Newcastle Thousand Families Study (NTFS). Associations between log-transformed LINE-1 DNA methylation levels and anthropometric and blood biochemical measurements, including triglycerides, total cholesterol, low-density lipoprotein (LDL) and high-density lipoprotein (HDL) cholesterol, fasting glucose and insulin secretion and resistance were examined. Linear regression, after adjustment for sex, demonstrated positive associations between log-transformed LINE-1 DNA methylation and fasting glucose {coefficient 2.80 [95% confidence interval (CI) 0.39-5.22]}, total cholesterol [4.76 (95% CI 1.43-8.10)], triglycerides [3.83 (95% CI 1.30-6.37)] and LDL-cholesterol [5.38 (95% CI 2.12-8.64)] concentrations. A negative association was observed between log-transformed LINE-1 methylation and both HDL cholesterol concentration [-1.43 (95% CI -2.38 to -0.48)] and HDL:LDL ratio [-1.06 (95% CI -1.76 to -0.36)]. These coefficients reflect the millimoles per litre change in biochemical measurements per unit increase in log-transformed LINE-1 methylation. These novel associations between global LINE-1 DNA methylation and blood glycaemic and lipid profiles highlight a potential role for epigenetic biomarkers as predictors of metabolic disease and may be relevant to future diagnosis, prevention and treatment of this group of disorders. Further work is required to establish the role of confounding and reverse causation in the observed associations.
Article
Background: Disturbances of DNA methylation have been associated with multiple diseases, including cardiovascular disease, cancer and, as some have suggested, glucometabolic disturbances. Our aim was to assess the association of the metabolic syndrome and its individual components with DNA methylation in a population-based study. Materials and methods: In a human population (n = 738) stratified by age, sex and glucose metabolism, we explored associations of the metabolic syndrome according to National Cholesterol Education Program/Adult Treatment Panel-III criteria and its individual components (fasting glucose, high-density lipoprotein cholesterol, triglycerides, blood pressure, waist circumference) with global leukocyte DNA methylation. DNA methylation was measured as the methylcytosine/cytosine ratio in peripheral leukocytes using liquid chromatography-tandem mass spectrometry. Results: Individuals with the metabolic syndrome had relative DNA hypomethylation compared to participants without the syndrome (β = -0.05; p = 0.01). This association was mainly attributable to linear associations of two metabolic syndrome components with DNA methylation: fasting plasma glucose (β = -0.02; p = 0.004) and high-density lipoprotein cholesterol (β = 0.07; p = 0.004). People with type 2 diabetes or impaired glucose metabolism had DNA hypomethylation compared to normoglycemic individuals (β = -0.05; p = 0.004). Conclusions: DNA hypomethylation is independently associated with hyperglycemia and low high-density lipoprotein cholesterol, both essential components of the metabolic syndrome. The potential implications and direction of possible causality require further study.
Article
Genome-wide association (GWA) studies on coronary artery disease (CAD) have been very successful, identifying a total of 32 susceptibility loci so far. Although these loci have provided valuable insights into the etiology of CAD, their cumulative effect explains surprisingly little of the total CAD heritability. In this review, we first highlight and describe the type of genetic variants potentially underlying the missing heritability of CAD: single nucleotide polymorphisms (SNPs) or structural variants, each of which may either be common or rare. Although finding missing heritability is important, we further argue in this review that it constitutes only a first step towards a fuller understanding of the etiology of CAD development. To close the gap between the genotype and phenotype, we propose a systems genetics approach in the post-GWA study era. This approach that integrates genetic, epigenetic, transcriptomic, proteomic, metabolic and intermediate outcome variables has potential to significantly aid the understanding of CAD etiology.
Article
To examine whether moderate hyperhomocyst(e)inemia is associated with coronary artery disease and the number of diseased coronary vessels in Koreans, we investigated 201 patients with angiographically documented coronary artery disease and 187 healthy subjects without evidence of stroke and coronary artery disease. The mean plasma total homocysteine in patients was higher than in controls (10.3 μmol/L; 95% confidence interval, 7.0–13.6 vs. 8.9 μmol/L; 6.0–11.8) (p=0.005). The prevalence of moderate hyperhomocyst(e)inemia, defined as the top 90th percentile in controls (13.2 μmol/L), was higher in patients than in controls (23.9% vs. 10.2%, p=0.001). Plasma homocyst(e)ine levels were not correlated to age, body mass index, levels of serum cholesterol, creatinine, and uric acid. Based on multiple logistic regression analyses with adjustment for total cholesterol, hypertension, smoking status, diabetes, age, and body mass index, the adjusted odds ratio of moderate hyperhomocyst(e)inemia for coronary artery disease was 1.53 (95% confidence interval: 1.39–1.65, p=0.0001). Moderate hyperhomocyst(e)inemia, diabetes mellitus, and old age were more prevalent in patients with triple-vessel disease than in single- or double-vessel disease (p=0.02). Multiple logistic regression analysis revealed that moderate hyperhomocyst(e)inemia was a significant predictor of triple-vessel disease with odds ratio of 2.78 (95% confidence interval: 1.08–7.10, p=0.02). We conclude that moderate hyperhomocyst(e)inemia is an independent risk factor for coronary artery disease, and also related significantly to the presence of triple-vessel disease.