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

Review Obesity

EPIDEMIOLOGY/GENETICS

The Epigenomic Analysis of Human Obesity


1,2,3,4
Christopher G. Bell

Objective: Analysis of the epigenome—the chemical modifications and packaging of the genome that
can influence or indicate its activity—enables molecular insight into cell type-specific machinery. It can,
therefore, reveal the pathophysiological mechanisms at work in disease. Detected changes can also rep-
resent physiological responses to adverse environmental exposures, thus enabling the epigenetic mark of
DNA methylation to act as an epidemiological biomarker, even in surrogate tissue. This makes epige-
nomic analysis an attractive prospect to further understand the pathobiology and epidemiological
aspects of obesity. Furthermore, integrating epigenomic data with known obesity-associated common
genetic variation can aid in deciphering their molecular mechanisms.
Methods and Conclusions: This review primarily examines epidemiological or population-based stud-
ies of epigenetic modifications in relation to adiposity traits, as opposed to animal or cell models. It dis-
cusses recent work exploring the epigenome with respect to human obesity, which to date has
predominately consisted of array-based studies of DNA methylation in peripheral blood. It is of note
that highly replicated BMI DNA methylation associations are not causal, but strongly driven by
coassociations for more precisely measured intertwined outcomes and factors, such as hyperlipidemia,
hyperglycemia, and inflammation. Finally, the potential for the future exploration of the epigenome in
obesity and related disorders is considered.
Obesity (2017) 25, 1471-1481. doi:10.1002/oby.21909

Introduction The Epigenome


Epigenetic factors include the chemical modifications of DNA DNA is packaged at the strand level by wrapping around histone
and the proteins that the DNA wraps around, which influence or proteins composed of eight subunits, two of each of histones 2A,
may indicate the activity of genes. Deciphering these marks can 2B, 3, and 4. The histone proteins possess tails that can be post-
be a powerful method to understand the specialized functioning translationally modified, including the addition of methyl or acetyl
of a cell and organs (1). They may also inform how molecular groups to lysine (K) molecules on histone 3 (H3). These additions
mechanisms are impeded in disease or because of adverse can be related to active or repressed regions depending on which K
environments. is modified. Amino acid deviation in the protein structure of these
histone molecules, termed histone variants, also modifies activity
The dramatic increase of obesity rates clearly points toward and can be associated with disease processes (7).
nongenetic or environmental factors as the significant driver.
Identified genetic susceptibilities are also modulated by these Chemical modifications of DNA itself include DNA methylation, the
external influences (2,3). This has led to considerable interest in most common, stable, and well-studied process, which is the addition
studying the epigenome to potentially quantify gene activity of a methyl group onto the 50 carbon of cytosine. This robust chemi-
changes due to this environment and to possibly gain molecular cal mark is due to the strong covalent carbon-to-carbon bond that
understanding of the pathophysiological consequences of the connects cytosine to the methyl group. In differentiated cells, this
obese state. Crucial biological understanding of obesity and its occurs in the DNA sequence predominately within the context of a
consequences is still lacking (4,5) and would help focus vital CpG dinucleotide, which is where guanine follows cytosine in the 50
preventive measures to counter obesity’s substantial morbidity to 30 direction on one DNA strand. It acts canonically as a repressive
and mortality risk (6). mechanism within gene promoters. However, additional DNA

1
MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK 2 Epigenomic Medicine, Biological Sciences, Faculty of
Environmental and Natural Sciences, University of Southampton, Southampton, UK 3 Human Development and Health Academic Unit, Institute of
Developmental Sciences, University of Southampton, Southampton, UK 4 Department of Twin Research and Genetic Epidemiology, King’s College
London, London, UK. Correspondence: Christopher G. Bell (cgb@mrc.soton.ac.uk)

Disclosure: The author declared no conflicts of interest.


Additional Supporting Information may be found in the online version of this article.
Received: 28 February 2017; Accepted: 11 May 2017; Published online 28 August 2017. doi:10.1002/oby.21909

www.obesityjournal.org Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 1471


Obesity Epigenomic Analysis of Human Obesity Bell

modifications occur that are in fact progressive oxidative products of the CpG dinucleotide itself. In fact, almost one-third of single-
the active DNA demethylation process, driven by the TET enzymes, nucleotide polymorphisms (SNPs) occur at CpG locations, because
leading first to hydroxymethylcytosine (5hmC), followed by formyl- of the hypermutability of methylated cytosines. This contributes con-
cytosine (5fC), and then carboxycytosine (5caC). This final product is siderably to allelic variation in the DNA methylome (18,19).
then recognized by the base-excision pathway to enable return to an
unmodified cytosine (8). These less common modifications may also The observed epigenetic state may represent the activity or repres-
in themselves possess active roles, with 5hmC implicated functionally sion of local cis-regulatory elements (binding sites of transcription
within enhancers (9). factors that enhance or repress transcription; usually 100-1000 base
pairs [bp] in length) (20). Genetic variability within these regions
High-throughput analysis of the epigenome is now focused on DNA can lead to fixed obligatory epigenetic positions or facilitate epige-
methylation, particularly because of its stability in extracted DNA in netic variation (21). Genetic effects on the methylome via transcrip-
comparison to other epigenetic marks, such as histone modifications. tion factors (TFs) have been known for decades, including the role of
However, due to its biological plasticity, it is an attractive biomarker SP1 within CpG islands (22). Motif changes in this and other methyl-
with strong potential clinical utility (10). ation determining regions (MDR), such as those for CTCF (CCCTC-
binding factor transcription factor) and RFX (regulatory factor X fam-
ily transcription factor), give rise directly to methylation variation in
CpG-dense regions (23). TFs binding to transcription factor binding
Considerations in the Analysis of the sites can also drive down DNA methylation (24) at distal regulatory
Epigenome regions and thus genetic variation in these binding sites will also be
influential. Some TFs are explicitly sensitive to DNA methylation
To powerfully examine the epigenome, especially in a human popu-
(25), and some, in fact, require it in order to bind (26). Additionally,
lation setting (11), we must be clear on how this mechanism con-
SNPs affecting the expression of trans factors will impact the epige-
trasts from the genome. This understanding significantly impacts
design, analysis, and interpretation. The differences can be broadly netic state within their distal transcription factor binding sites or cis-
encapsulated in three principles: (i) cell-type specificity, (ii) change- regulatory element regions (27). Genetic influence on the epigenome
ability, and (iii) sequence interactive (or positional) effects. is so strong that it has been observed in enhancer variation in only 19
individuals of diverse ancestry (28).
First, as its function dictates, the epigenome is specific to tissue or cell
type. Therefore, we want, if at all possible, to examine the cell type(s) Nearby positional effects, such as polymorphic insertions of often
most relevant to the pathophysiology of the disease process being investi- heavily methylated retrotransposons, will influence the local region
gated. Any associated findings in tissue that is not the primary site of (29). The density of CpGs also has a direct relationship with the
action can be interpreted as a surrogate measure for markers of exposure, DNA methylome (30), so CpG-SNPs (SNPs creating or abrogating a
which may also act on the diseased organ. They can also be downstream CpG dinucleotide (18,31)), as well as affecting available methyl-C-
physiological changes due to this exposure or due to the disease itself. In containing motifs, can impact on the rate of change of CpG density
the analysis of obesity, this is not straightforward, as the intrinsic genetic slopes. The influence of SNPs as mQTL (methylation QTL) (32)
predisposition is governed largely within the brain (12), with central con- can capture influence on cis or trans factors, the DNA methylation
trol for energy balance directed by specific hypothalamic neurons, partic- machinery, or haplotypic effects, such as regional density or proxim-
ularly within the arcuate nucleus (13). Thus, these cells are inaccessible ity factors. Furthermore, genetically associated correlated CpGs have
except in postmortem samples. Adipose tissue is also an obvious focal been observed in clusters, termed “GeMes” (33). Finally, the epige-
interest for the dissection of obesity, given its physical and functional nome is a coordinated mechanism, so the epigenetic layers should
changes and its role as an endocrine organ (5). fit together logically, such that a DNA hypermethylated promoter
should show a lack of colocating activating chromatin marks, and
Second, the epigenome changes over time. This is most dramatic vice versa (34).
during development, when two rounds of epigenomic reprogram-
ming occur, first in gamete formation and then post fertilization.
Both rounds are characterized by global DNA demethylation (14).
However, the epigenome is not static after tissue-specific marks are
Confounding Issues
laid down. It is seen to suffer from “epigenetic drift” with age, All the above effects—tissue specificity, changeability, and sequence
whereby hypomethylated regions gain and hypermethylated regions interaction—can confound or misattribute action or effect in epige-
lose methylation stochastically over time throughout the genome nomic studies. Rigorous study designs have been proposed and
(15). This process is proposed to be involved in age-related deterio- employed to prevent this. However, because of practical, technical,
ration in function and the concurrent increased risk of chronic dis- and cost considerations, it is often not possible to perform the ideal
eases, including obesity-related conditions. On top of this are spe- study. Researchers must compromise, while acknowledging and
cific directional aging changes that can be identified at defined working within these limitations (35,36).
functional loci across the genome (16,17).
Isolating disease-relevant tissue cell types for analysis is an
Third, positional effects driven through sequence variation will be obvious first starting point, although DNA derived from peripheral
reflected in the measured epigenome. These can be direct, in cis or blood is available in many large epidemiological studies, offering
trans, and can confound analysis in human population samples, par- the benefit of large sample sizes and deep phenotyping. While
ticularly across diverse ancestries (Figure 1) (11). Direct effects with these are practical benefits, peripheral blood is also of physiologi-
respect to DNA methylation can be due to genetic polymorphism at cal interest in obesity because of known inflammatory-related

1472 Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 www.obesityjournal.org


Review Obesity
EPIDEMIOLOGY/GENETICS

Figure 1 Sequence factors influencing DNA methylation array analysis. (A) Direct CpG-SNPs. (B) SNPs under probe sequences. (C) Cis and trans SNP effects
with increased (orange) or decreased (grey) transcription factor (TF) expression influencing the methylation state of binding site. (D) Cis effects can include
regional effects from hypermethylated repeats or methylation determining region (MDR) motifs within CpG-dense regions. (E) Density change within intermedi-
ate CpG density regions such as CpG island shores also influence methylation state.

changes (37) and its usefulness for detecting passive exposure bio- samples or tissue biopsies are required to gain further insight into
markers. Furthermore, the potential clinical utility of findings in underlying mechanisms.
blood is significant because of its ease of access compared to adi-
pose tissue biopsy. However, these DNA methylation differences Peripheral blood analysis also contains the various leukocyte cell
are unlikely to correlate with other target tissues. Postmortem types and, as such, represents a meta-epigenome of these subsets

www.obesityjournal.org Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 1473


Obesity Epigenomic Analysis of Human Obesity Bell

(38). This mixture needs to be quantified and, even in normal condi- Cell Type-Specific Signals in Peripheral
tions, may vary between individuals because of many factors,
including sex and genetic background. Thus, DNA methylation Blood
profiling in blood will strongly identify signals driven by subcell Dissecting the strong DNA methylation signals that can be identified
composition changes due to the disease state or immune responses with tobacco smoking in peripheral blood starts to reveal the biolog-
associated with it. While isolation of the most disease-relevant ical intricacies of epigenetic changes in this heterogeneous tissue.
cell type is preferable (39), deconvolution algorithms have been Understanding these robust results is very useful in interpreting what
devised using differentially methylated positions (DMPs) that esti- may be possible in DNA methylation studies of obesity and other
mate leukocyte cell-type proportions (40). In fact, because of the diseases. There are distinct results depending on which cellular sub-
cell-type specificity of epigenomes, once accurately delineated, set is examined (58). The strongest tobacco-associated signal in the
this deconvolution is set to become one of the most powerful epi- gene AHRR appears so conspicuously because the epigenetic change
genomic tools. The mapping of these cell and tissue types has occurs specifically within granulocytes and monocytes; this is then
begun through efforts such as the Epigenomics Roadmap (41), amplified by the fact that smoking itself increases the proportion of
Blueprint (42), and future cell-type defining surveys (43). Kim granulocytes in peripheral blood. However, tobacco has little impact
et al. recently showed that an expanded set of reference leuko- on AHRR T cell DNA methylation. In comparison, lymphoid-
cytes, with pathologically important cell fractions including imma- specific inflammatory changes occur, such as those seen in GPR15,
ture memory plasma B cells, activated natural killer (NK) cells, where the DNA methylation changes are only seen in T and B cells
and na€ıve T cells, can explain significantly more variability in (58,59). Biologically, these changes are likely inflammatory
peripheral blood methylomes (44). Additionally, these data responses driven by smoking-caused injury to tissues. On the mye-
improved case and control discrimination in immune-related disor- loid side, monocyte to macrophage differentiation may be influenced
ders, including the metabolic syndrome. by tobacco, as is a proposed carbon monoxide environmental selec-
tion on bone marrow progenitor cells (58). Thus, the identification
Changeability in developed tissues encapsulates two often- of these subtype-specific signals allows for the proposition and
intersecting mechanisms: (1) significant directional changes with exploration of novel hypotheses and mechanisms.
time in proportions of a heterogeneous collection of cells, and
(2) the activation or representation of certain pathways. Relative
change is seen clearly, as mentioned, in peripheral blood, with
responses to acute infection or even chronic low-grade inflamma- Obesity-Induced Changes in Adipose and
tion (45). Other significant proportional changes occur because of
aging, with a skew toward a greater myeloid cell fraction in
Blood
blood (46), but also a reduced ratio of neurons to glia in the Adipose tissue is not a homogenous organ. The adipocytes of subcu-
brain (47). In many studies, however, the phenotype of interest taneous fat are mostly white because of the storage of triglycerides,
may alter the proportional composition of the cell types within which secrete leptin and adipokine. There are also small but discern-
the analyzed target tissue (48). In fact, in some cases, correction ible levels of brown and beige fat (5). The role of brown fat in
for cell type may impede the identification of cell type-specific humans has garnered significant interest because of its energy
processes (49). dissipation properties and fat burning actions via uncoupling protein
1-containing mitochondria. Stimulation by the sympathetic nervous
Finally, the influence of genetic factors on the epigenome is system after cold exposure leads to heat production. Beige, or
strong. Therefore, considerable power comes from longitudinal brown-to-white, thermogenic adipocytes have a progenitor cell ori-
studies (35) and monozygotic, or identical twin, discordant analy- gin, and conversion can be induced by cold temperatures, exercise,
ses, to eliminate this significant influence (50,51). In high- and endocrine factors (5).
throughput DNA methylation array analysis, the influence of
genetic effects can be driven by SNPs at the investigated CpGs In obesity, adipose tissue operates as a large immunologically active
themselves, under the probes, or nearby (Figure 1). In a recent endocrine organ (60). Obesity-related apoptosis of adipose cells leads to
Illumina Infinium HumanMethylation450 BeadChip (450k array) infiltration by macrophages and other inflammatory immune cells (60).
analysis, drastic effects leading to discrete genotypic grouping, or This, in turn, leads to chronic adipose tissue inflammation (61), with the
“gap signals” were identified at 11,007 of the interrogated CpGs secreted proinflammatory cytokines contributing to insulin-resistance
(52). Previous 450k array analysis found population genetic varia- (62). Visceral fat deposits, including omentum and mesenteric fat, are
tion attributable in 66,877 of the probes (13.8%) (53). Chen et al. more detrimental than subcutaneous fat (63) and are associated with
recently observed this strong genetic influence in a study of many of the systemic metabolic consequences of obesity (5).
genetic drivers of epigenetic variation in human immune cells,
and it was noted to be a concern for epigenome-wide association Obesity induces changes in innate immune cells, increases CD41 and
study (EWAS) interpretation (54). More complex haplotypic CD81 T cells, and reduces tolerance-promoting regulatory B cells, as
effects may also exist, with strong consequences captured by gap well as inducing abnormal B cell function (64). Lymphoblast-derived
signals, though more subtle outcomes will converge. We should NK cells, which have been shown to be significant regulators of mac-
bear in mind not only how these genetic influences can confound rophage polarization and insulin resistance (65), have a proposed role
studies, but also how they may facilitate functional variation. in obesity, and their methylome is remodeled in conversion from naive
CpG-SNPs are proposed to play a significant facilitative epige- to activated cells (66). Thus, a chronic low-grade inflammation is
netic role, and these effects were recently seen be to enriched associated with obesity, and specific DNA methylation changes,
within genome-wide association study (GWAS) regions in several assessed via C-reactive protein levels within peripheral blood (45),
diseases, including type 2 diabetes (55-57). have been observed because of this.

1474 Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 www.obesityjournal.org


Review Obesity
EPIDEMIOLOGY/GENETICS

Analysis of the Epigenome in Obesity sequential data, rather than causal. This was also supported by
Mendelian randomization (MR) analysis. A further analysis by Main
Up to the beginning of this decade, obesity epigenetic studies pri- et al. revealed a relatively high level of familiarity (h2 51%-64%)
marily focused on candidate genes or on total measures of the epige- for HIF3A DNA methylation in blood (76), which was close to
nome, such as global DNA methylation (67). The latter can be use- the heritability level of obesity itself. Epidemiological evidence has
ful in gross abnormalities, such as cancer, in which significant also pointed at vitamin B2 and B12 levels as influencing HIF3A epi-
hypomethylation is strong enough to drive global variation. How- genetic levels (73).
ever, it is far less useful in more nuanced phenotypes and has not
led to consistent findings (68). The study of candidate genes, as Another BMI EWAS analysis with the 450k array in peripheral
with complex trait genetics, has not been particularly fruitful, with blood DNA was performed in 2,097 African American adults in the
weak and inconsistent results. In the exploration of imprinting genes Atherosclerosis Risk in Communities (ARIC) study (72). A similarly
or those involved in metabolism, few (if any) have been supported sized replication set of 2,377 individuals of European ancestry from
by subsequent genome-wide, more adequately powered, and the Framingham Heart Study was employed, along with isolated
confounding-aware studies (68). Many early studies therefore need CD41 T cell DNA from 991 individuals of European ancestry from
reassessing in the light of current findings and knowledge. the Genetics of Lipid Lowering Drugs and Diet Network Study
(GOLDN). This enabled 37 DMPs to be robustly identified with
All DNA methylation studies prior to at least 2012 are likely to BMI, and an additional DMP was specifically associated with waist
have not taken the confounding effects of cell-type proportions into circumference only. Sixteen DMPs were also seen to be consistent
account. Furthermore, many of these studies have involved small in 648 adipose tissue samples. Novel results included LGALS3BP,
sample sizes, with added potential issues due to genetic heterogene- KDM2B, PBX1, BBS2, and genes implicated in lipid metabolism,
ity. While a stronger effect size can be found for phenotypic traits cytokine signaling, and immune response.
or complex diseases in epigenetic associations than genetic associa-
tions (69), lack of subsequent replication points to their weakness In a 2013 study, Wahl et al. analyzed a collection of large GWAS
(68). set with the 450k array in peripheral blood (77). The discovery
set contained a mix of individuals of Southeast Asian (n 5 2,680)
and European (n 5 2,707) ancestry and identified 278 BMI-
associated CpGs residing in 207 genetic loci while correcting for
EWAS for Obesity (BMI) in Peripheral leukocyte cell-type proportions. Taking forward the strongest indi-
Blood vidual CpG associations within these regions to an additional 4,874
samples, 187 out of 207 replicated at a significance level of
In 2014, Dick et al. published one of the first large EWAS for BMI
P < 0.05. The results did not vary significantly across differing
using the 450k array (70). This was performed in whole blood-
ancestry groupings, except for seven DMPs in which very strong
derived DNA from individuals of European ancestry. The initial dis- population variation between Asian and European was seen (hetero-
covery set was 479 individuals, followed by two rounds of replica- geneity P < 1 3 10-7), hinting at population-specific or uncaptured
tion, first in 339 samples and then in 1,789 samples. The discovery genetic effects. All the changes were identified as the potential con-
set identified five CpGs across three genetic loci associated with sequence of obesity rather than causative, except for one CpG
BMI. Three CpGs were replicated, all of which resided in the first (cg26663590) in NFATC2IP. These results were strongly enriched
intron of HIF3A, the hypoxia inducible factor 3 alpha subunit gene, for methylation scores within an intermediate level of 20% to 80%.
which is involved in regulating hypoxia-inducible gene expression This could point toward variation between certain cell subsets driv-
(cg22891070, cg27146050, and cg16672562). Potential lymphocyte ing the signal, although analysis of four subfractions (monocytes,
cell-type effects on cg22891070 were tested for after the initial anal- neutrophils, CD41 T cells, and CD81 T cells) indicated that these
ysis. While a small association with leukocyte number was seen, results could in fact be seen across all cell types. Thus, they differ
adjustment for subtypes did not substantially reduce the BMI from the biologically defined tobacco smoking results in which cell
association. type-specific divergent myeloid versus lymphoid signals could be
found (58). A longitudinal sample set of 1,435 participants was eval-
Then, to investigate HIF3A across different tissues, DNA from adi- uated over a period of 7 years, showing a consistent relationship
pose tissue (n 5 635) and skin (n 5 395) was assessed, with signifi- between change in methylation and change in BMI over time in 178
cant methylation changes identified in fat only. One driver of this of 187 CpGs.
may be the high level of inflammatory blood cell invasion into adi-
pose tissue. Genetic effects were explored, and two SNPs Another large-scale BMI EWAS was recently reported by Mendel-
(rs8102595 and rs3826795) were both independently associated with son et al., again using the 450k array in peripheral blood (78). The
cg22891070’s DNA methylation state in all data sets, although they discovery set was in those of predominately European ancestry,
were not significantly associated with BMI themselves. including individuals from the Lothian Birth Cohort in Scotland and
the US Framingham Heart Study. One hundred thirty-five CpGs
In addition to being replicated in several studies (71-73), including were initially identified as BMI DMPs. Eighty-three DMPs repli-
in adipose tissue (74), this result was also explored for causality in cated in at least one of the three additional replication cohorts,
the ALSPAC cohort from Bristol in the United Kingdom (75). ARIC (n 5 2,096), GOLDN (n 5 992), and Prospective Investigation
Genetic and multiple time point DNA methylation data were avail- of the Vasculature in Uppsala Seniors (PIVUS) (n 5 967).
able in 1,000 mother-offspring pairs. The DNA methylation
changes identified were proposed to be secondary to differences in Gene ontology enrichment was identified for lipid metabolism in
BMI through examination of the temporal relationship of changes in those DNA methylation variation-associated genes that also had

www.obesityjournal.org Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 1475


Obesity Epigenomic Analysis of Human Obesity Bell

TABLE 1 Consistent BMI DMPs from Demerath et al. (72), Wahl et al. (77), and Mendelson et al. (78)

Methyl Additional Other


chr Start Stop CpG Gene BMI CGI Location BMI phenotype(s)

chr11 68607621 68607622 cg00574958 CPT1A 2 Shore Intronic BMI and WCa (82) TG (49,86,87); VLDL-C (87);
Lipoprotein subfractions (79);
HTG waist (91);
MetS in Eur (83)
chr15 52554170 52554171 cg06192883 MYO5C 1 2 Intronic BMI in Arab (84) CRP (45)
chr21 43656586 43656587 cg06500161 ABCG1 1 Shore Intronic TG, HDL-C (49,88,89);
Glucose-related (90);
HTG waist (91)
chr16 11422408 11422409 cg06946797 RMI2 2 2 Intronic BMI in Arab (84) Glucose-related (90)
chr17 40927698 40927699 cg08857797 VPS25 1 2 Intronic BMI in Arab (84) TG (49)
chr22 50327985 50327986 cg09349128 CRELD2 2 Shore Intergenic Glucose-related (90);
IBD (92)
chr17 2612405 2612406 cg09664445 CLUH 1 Shore Intronic BMI in Arab (84)
and in Eur (70)
chr17 17730093 17730094 cg11024682 SREBF1 1 Shelf Intronic BMI in Eur (85) TG (49,89);
Glucose-related (90)
chr6 31681881 31681882 cg13123009 LY6G6F 1 2 Intronic BMI in Arab (84)
chr5 158634084 158634085 cg26403843 RNF145 1 Shelf Intronic

a
This study was performed earlier with many of the same cohorts as Wahl et al.
Methyl BMI, (2) hypomethylation; (1) hypermethylation with increasing BMI; WC, waist circumference; TG, triglycerides; HDL-C, low density lipoprotein cholesterol; VLDL-
C, very low-density lipoprotein cholesterol; HTG, hypertriglyceridemic; MetS., metabolic syndrome; CRP, C-reactive protein; IBD, inflammatory bowel disease.

concordant expression changes. Seventy-seven of the nonredundant (Genomic Regions Enrichment of Annotations Tool) for these 10
CpGs (i.e., correlation jrj < 0.7) captured 18% of the interindividual CpGs (compared with the 450k array probes as background) identi-
variation in BMI. The previous HIF3A result was replicated with the fied nominal significance for biological processes, such as regulation
strongest association seen in the youngest subset (cg22891070, of cholesterol and lipids, human phenotypes including hyperlipid-
P 5 0.003). Just 11 CpGs revealed a recognized three-way association, emia, and diseases of the hepatobiliary system (Supporting Informa-
whereby the DNA methylation was associated with BMI and expres- tion Table S1).
sion, as well as with BMI-associated expression changes. These
included ABCG1, CACNA2D3, CPT1A (79), DHCR24, SARS, SLC1A5, In fact, many of these same CpGs are associated with lipid-related
and SREBF1. No enrichment for blood or adipose-specific regulatory traits from specific EWAS performed for these measures, presum-
features was seen using eFORGE (80) to detect tissue specificity of the ably because of their relationship to the development of adiposity or
DMPs through colocalization with tissue-specific DNase-I hypersensi- downstream-induced epigenetic changes from BMI-associated
tivity sites. DMPs were more enriched in enhancer regions (H3K4me1) altered blood lipid profiles. Four of the ten DMPs were significantly
than promoter regions, although this may indicate the less dynamic related to triglyceride levels in a recent study by Dekkers et al. (49),
nature of these promoter CpGs (81). The top cis-mQTL for the repli- as well as in earlier and contemporary lipid studies (79,86-89). Fur-
cated BMI DMPs, selected by lowest P value 6 500 kilobases [kb] thermore, a study by Kriebel et al. found that four of these CpGs
from the CpG, was not strongly attenuated by BMI association in 81 of are related to glucose-related phenotypes (90). This clearly shows
the 83 CpGs. One gene, SREBF1 (a lipid metabolism transcription fac- the power of EWAS to identify more precise biochemical pheno-
tor), was proposed as possibly causal; it is known to induce conversion types (38) and the benefit of closely examining the distinct biologi-
of acetyl-CoA to triglycerides, thus promoting glycolysis, lipogenesis, cal changes associated with broad epidemiological measures, such
and adipogenesis. A role for this gene in adiposity, insulin resistance, as BMI. A number of these exact CpGs also are supported by addi-
coronary artery disease, and obesity-related dyslipidemia is also sup- tional studies in or related to BMI, including analyses involving
ported in model organism and human studies (78). Arabs (84), metabolic syndrome (83), and hypertriglyceridemic
waist in Mexican Americans (91). Of interest is that one of the
A comparison of the results from these three recent large EWAS CpGs, cg06192883, in MYO5C was recently identified in an EWAS
studies (72,77,78) identified 10 CpGs that were robust and replicated for the inflammatory marker C-reactive protein (45). Another,
within each as well as being common to all (Table 1). These 10 cg09349128, in CRELD2, was associated with inflammatory bowel
CpGs are located in 10 unique genes, predominately within intra- disease (92), which is known itself to be connected to lipid abnor-
genic intronic loci and in almost half within CpG island shores. malities (93). Two additional genes, LGALS3BP and SBNO2, were
They are also generally supported in other BMI-related studies consistently identified across all three studies but were represented
(70,82-85). Gene ontology enrichment analysis via GREAT by differing CpGs.

1476 Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 www.obesityjournal.org


Review Obesity
EPIDEMIOLOGY/GENETICS

EWAS Analysis in Adipose Tissue evidence that DNA methylation plays a role in this process in
humans, the possibility for some repetitive elements such as SINE-
Agha et al. analyzed adipose tissue with the 450k array and associ-
VNTR-Alu (SVAs) to escape the global demethylation and subse-
ated measures of adiposity obtained by dual-energy x-ray
quent reprogramming and then be vulnerable to environmental influ-
absorptiometry-assessed android fat mass, android:gynoid fat ratio,
ence cannot be excluded at this stage (14).
and trunk:limb fat ratio, as well as BMI (71). This was in 106 indi-
viduals (64% women, 68% white) after adjusting for smoking, race, Epidemiological data have proposed that in utero exposure to
and sex as well as reference-free adipose cell-mixture effects. A under- and overnutrition affects obesity risk in later life, potentially
gene-focused analysis identified association with these adiposity through epigenetic modifications (100). Several studies have
traits including ANGPT2, AOC3, AQP7, CETP, LIPE, and SOD3. explored this through the unique model of contrasting children
HIF3A was also explored and found to be positively associated with born before and after maternal gastric bypass surgery. Guenard
BMI at the same three CpGs as identified by Dick et al. (70) et al. compared DNA methylation from siblings born from 25
mothers, adjusting for age, sex, and puberty (101). Children born
Ronn et al. investigated age, BMI, and hemoglobin A1c (HbA1c) prior to the procedure possessed significantly higher BMI z scores
levels, all risk factors for disease, in both adipose tissue and blood as well as poorer insulin resistance values. Temporal DNA methyl-
DNA methylation (74). The DNA methylation aging signals seen ation variation was identified, although in blood and not adjusted
previously in ELOVL2, FHL2, KLF14, and GLRA1 in blood were for cell-type effects, but gene enrichment for glucose homeostasis
identified in adipose. This study supported the HIF3A finding in adi- and immune function was seen. However, more recent and power-
pose within females only and found HbA1c associations with 711 ful studies have not borne out these earlier findings. Willmer et al.
sites in adipose tissue. There was minimal overlap in signal between conducted a direct obesity analysis in a Scandinavian study of 164
HbA1c and BMI or age. children born before and 176 born after maternal gastric bypass
surgery; at 4 years of age, the children did not show any improving
A study of post-gastric bypass DNA methylation variation in adi- effect of bariatric surgery on weight development (77). Sharp et al.
pose tissue identified changes within genes associated with obesity examined 1,018 neonatal cord blood-derived DNA methylomes
by pre- versus post-surgery analysis (94). Both omental and subcuta- with the 450k array for the relationship between maternal adiposity
neous adipose tissue was compared, providing further insight into and later childhood adiposity. Both high and low maternal weight
the dramatic physiological changes that occur with rapid postopera- extremes were seen to lead to significant neonatal epigenetic
tive weight loss. changes, although weight gain during pregnancy was not influential
(102). A large Danish birth cohort of 30,655 trio families examined
children at 7 years to also directly explore the influence of parental
BMI through pregnancy. This study supported a role for the intrau-
Obesity- and Aging-Associated Changes terine environment in fetal and child growth trajectories but found
Horvath et al. observed that obesity accelerated detected epigenetic that later parent-child associations in weight were likely to be
aging of the liver when assessed by the Horvath “Epigenetic Clock,” but influenced by shared known genetic or environmental factors with
this acceleration did not occur in the other tissue types tested, including both parents (103).
blood, adipose tissue, and muscle (95). Obesity-related nonalcoholic
fatty liver disease did not drive these hepatic changes, although it was The genetic component of these maternal adiposity epidemiological
postulated to be associated with obesity-driven liver comorbidities, observations was recently dissected through a large-scale GWAS
including insulin resistance and hepatocellular carcinoma. The lack of and was seen to be a significant, but not complete, contributor
changes in other tissues was hypothesized as due to liver-specific oxida- (104). In addition, Richmond et al. performed a direct MR analysis,
tive stress and also perhaps the suboptimal measure of obesity by BMI. using a weighted genetic score from variants for BMI, for maternal
obesity influences in children from ages 7 to 18 years. In more than
A study by Simpkin et al. found that an accelerated epigenetic age 2,000 samples in both discovery and replication sets, they found lit-
measure at birth was also associated with a more rapid increase in tle support for a strong causal intrauterine effect of increasing mater-
BMI in childhood (96). Obesity was further proposed to contribute nal BMI with increased childhood adiposity (105). The complexity
to the “exposome” that influences epigenetic aging, with increased of the MR analysis in this scenario was recently discussed by Law-
BMI from young adulthood to middle age contributing to greater lor et al. (106).
age acceleration (97). In an analysis integrating blood-derived
aging-related differentially methylated regions (DMRs) within
GWAS-associated regions, three were identified within loci for fast-
ing glucose-related traits that interact with BMI (17). Limitations and Future Directions
Current large-scale high-throughput analysis is limited to DNA
methylation array-based techniques, commonly the 450k array and
the more recent 850k array. These provide only a partial story with
Developmental and In Utero Influences regard to DNA methylome, as 28 million CpGs reside within the
Critical windows may exist for environmental or intrauterine factors genome, with the additional potential for non-CG changes. An
to influence the epigenome as it being defined during early develop- obvious area of underexploration by this approach is repetitive ele-
ment (98). These early changes are proposed to have the ability to ments, which are proposed to conceal significant functionality, such
act as “metastable epialleles” that then propagate through the germ as the strong overlap of chromatin enhancer evidence within long
layers to affect all cell types (99). Although there is not strong terminal repeat (LTR) class member LTR12C (107). Examples also

www.obesityjournal.org Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 1477


Obesity Epigenomic Analysis of Human Obesity Bell

include a potential epigenetic role for an Alu repetitive element in the use of histone deacetylase inhibitors in inflammation-related dis-
POMC in childhood obesity (108). eases, as well the potential of targeted epigenetic drugs via CRISPR,
points to future epigenomic therapeutic possibilities (119).
Furthermore, coordinated variation across a functional region, such
as an enhancer, by the delineation of a significant DMR can increase
statistical and biological confidence of the result (109). However,
sparse array data are not the most powerful for robust DMR calling Conclusion
(110). Enrichment for disease ontology-based results is stronger Epigenomic analysis is a significant tool in the hunt to improve risk
using sequencing-derived DMRs rather than isolated DMPs (58). prediction, as well as prognosis, beyond the rudimentary measure of
Thus, larger and more in-depth sequencing-focused analyses could BMI for obesity-related diseases. By observing at a molecular level
identify further novel findings. the biologically perturbed mechanisms associated with this disorder,
we may understand more precisely, for instance, the significant path-
External confounding factors in EWAS analysis include tobacco,
ogenic influence of visceral fat. On the epidemiological side, while
alcohol, diet, medications, and other chemicals (68), though these
population-based strategies have borne the greatest improvements in
signatures are now beginning to be precisely defined. Strong and
human health in the past, there is now evidence that this may not be
replicated effects from prenatal exposures with smoking that persist
the case for obesity (120). More focused policies may be required
into childhood exist, although these are not yet robust for diet. How-
because of the widening distribution of BMI over time.
ever, evidence exists in several murine and other animal models that
have explored prenatal dietary exposures (111). DNA methylation
Large-scale DNA methylation array analyses in blood have revealed
associations with blood serum metabolites have been identified in
the precise sequelae of the obese state, such as epigenetic changes
humans through EWAS (112).
driven by dyslipidemia, hyperglycemia, and chronic inflammation.
This points to the use of more precisely defined biochemical pheno-
Major areas for future exploration are the influence of rare cell types
and an issue not yet resolved in biology: at what point does a types in EWAS analysis. Nevertheless, further study may implicate
change in the epigenome of a cell indicate a new cell type? Epige- additional novel disease-associated outcomes, exposures, or even
netic variability occurs even in assumed homogenous cell types, causes. These could include or exclude unproven but intriguingly
such as neutrophils (113). It is important to note that when robustly proposed factors, such as air pollution (121), metal exposure (122),
controlled experiments have reduced genetic and cell-type variability or other “obesogens” in the environment and food chain (123) that
by the use of isolated cell types in monozygotic twin discordant could play a role in obesity or other diseases. The powerful potential
models, minimal significant directional disease-associated DMPs of robustly identified DNA methylation biomarkers, even if not
have been identified (39). Although increased variability at distinct causal, can be seen in tobacco-associated AHRR cg05575921, which
positions can be found, it is not attributable and could therefore be a captures future smoking-related morbidity and mortality (124).
technical artifact or unaccounted environmental effects. Underlining Regarding obesity, the potential to predict future cardiovascular dis-
these cell type-specific effects, the BMI EWAS by Mendelson et al. ease exists in the lipid-related DNA methylation changes that influ-
only saw 22 of 135 initial DMPs replicated in isolated CD41 T cells ence ABCG1 expression (125).
(78). Validation and, most crucially, independent replication are
required for robust findings to be reported, with subsequent func- The analysis of the epigenome is constantly improving and being
tional investigation in appropriate biological models. refined, such as in a recent discussion by van Iterson et al. on con-
trolling genomic bias in DNA methylation EWAS (69). This trajec-
The integration of genetic and epigenetic data is another area in which tory will enable us to more accurately define the epigenome and
powerful insights can be made. Analysis within GWAS loci to deter- interpret these findings. Increased high-throughput access to distal
mine allelic or haplotype epigenetic differences was first shown with regulatory regions via the 850k array, larger sample-sized sequenc-
DNA methylation in the obesity-related FTO locus (31). This amalga- ing-based DNA methylome studies, and the analysis of additional
mation of epigenetic and haplotypic data has now become a valuable modifications, including population chromatin data, will drive this
tool in the dissection of GWAS traits (114). Incorporating chromatin progress forward. Also, the potential of third generation sequencing
segmentation data across multiple tissue types, including adipose tis- to directly assess DNA modifications will be a significant step for
sue, has enabled the localization of enhancer variation in adipocytes epigenomics (126). We will be able to more accurately define cur-
influencing IRX3 and IRX5 expression, within the large linkage dise- rently abstractly lumped together “environmental” changes by more
quilibrium block region of FTO. These regulatory genes were shown precisely excluding or integrating genetic effects, as well as defining
to be involved with the browning of fat and thermogenesis (115). cell type-specific and cell proportion changes, including increasingly
rare cell types. With this will come improved biological interpreta-
Few studies to date have analyzed histone variants or post- tion and understanding of critical pathological changes within
translational modifications with respect to human obesity, and they defined cell types and genetic pathways. This knowledge will hope-
have been limited to extremely small numbers and/or only global fully help reduce the chronic burden of obesity worldwide.O
measures (116). Murine studies have proposed potential biological
insights that may merit human exploration, such as histone variant C 2017 The Obesity Society
V
MacroH2A1, which may have an antiadipogenic role in differentiat-
ing adipose tissue (117). Large-scale human studies are on the hori- References
1. Zhou VW, Goren A, Bernstein BE. Charting histone modifications and the
zon, however, as can be seen in the example of a recent successful functional organization of mammalian genomes. Nat Rev Genet 2011;12:7-18.
postmortem histone acetylome-wide association study in the active 2. Reddon H, Gueant JL, Meyre D. The importance of gene-environment interactions
chromatin mark H3K27ac for autism spectrum disorder (118). Also, in human obesity. Clin Sci (Lond) 2016;130:1571-1597.

1478 Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 www.obesityjournal.org


Review Obesity
EPIDEMIOLOGY/GENETICS

3. Young AI, Wauthier F, Donnelly P. Multiple novel gene-by-environment 34. Wilson GA, Butcher LM, Foster HR, et al. Human-specific epigenetic variation in
interactions modify the effect of FTO variants on body mass index. Nat Commun the immunological Leukotriene B4 Receptor (LTB4R/BLT1) implicated in
2016;7:12724. doi:10.1038/ncomms12724 common inflammatory diseases. Genome Med 2014;6:19. doi:10.1186/gm536
4. Bray GA, Fruhbeck G, Ryan DH, Wilding JP. Management of obesity. Lancet 35. Heijmans BT, Mill J. Commentary: the seven plagues of epigenetic epidemiology.
2016;387:1947-1956. Int J Epidemiol 2012;41:74-78.
5. Heymsfield SB, Wadden TA. Mechanisms, pathophysiology, and management of 36. Birney E, Smith GD, Greally JM. Epigenome-wide association studies and the
obesity. N Engl J Med 2017;376:254-266. interpretation of disease -omics. PLoS Genet 2016;12:e1006105. doi:10.1371/
6. Kitahara CM, Flint AJ, Berrington de Gonzalez A, et al. Association between class journal.pgen.1006105
III obesity (BMI of 40–59 kg/m2) and mortality: a pooled analysis of 20 37. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev
prospective studies. PLoS Med 2014;11:e1001673. doi:10.1371/journal. Immunol 2011;29:415-445
pmed.1001673 38. Houseman EA, Kim S, Kelsey KT, Wiencke JK. DNA methylation in whole
7. Maze I, Noh KM, Soshnev AA, Allis CD. Every amino acid matters: essential blood: uses and challenges. Curr Environ Health Rep 2015;2:145-154.
contributions of histone variants to mammalian development and disease. Nat Rev 39. Paul DS, Teschendorff AE, Dang MA, et al. Increased DNA methylation
Genet 2014;15:259-271. variability in type 1 diabetes across three immune effector cell types. Nat Commun
8. Branco MR, Ficz G, Reik W. Uncovering the role of 5-hydroxymethylcytosine in 2016;7:13555. doi:10.1038/ncomms13555
the epigenome. Nat Rev Genet 2012;13:7-13. 40. McGregor K, Bernatsky S, Colmegna I, et al. An evaluation of methods correcting
9. Hon Gary C, Song C-X, Du T, et al. 5mC oxidation by Tet2 modulates enhancer for cell-type heterogeneity in DNA methylation studies. Genome Biol 2016;17:84.
activity and timing of transcriptome reprogramming during differentiation. Mol doi:10.1186/s13059-016-0935-y
Cell 2014;56:286-297.
41. Roadmap Epigenomics C, Kundaje A, Meuleman W, et al. Integrative analysis of
10. BLUEPRINT consortium. Quantitative comparison of DNA methylation assays for 111 reference human epigenomes. Nature 2015;518:317-330.
biomarker development and clinical applications. Nat Biotechnol 2016;34:726-737.
42. BLUEPRINT Consortium. Quantitative comparison of DNA methylation assays for
11. Greally JM. Population epigenetics. Curr Opin Syst Biol 2017;1:84-89. biomarker development and clinical applications. Nat Biotech 2016;34:726-737.
12. O’Rahilly S. Human genetics illuminates the paths to metabolic disease. Nature 43. Tanay A, Regev A. Scaling single-cell genomics from phenomenology to
2009;462:307-314. mechanism. Nature 2017;541:331-338.
13. Bell CG, Walley AJ, Froguel P. The genetics of human obesity. Nat Rev Genet 44. Kim S, Eliot M, Koestler DC, et al. Enlarged leukocyte referent libraries can
2005;6:221-234. explain additional variance in blood-based epigenome-wide association studies.
14. Tang WW, Kobayashi T, Irie N, Dietmann S, Surani MA. Specification and Epigenomics 2016;8:1185-1192.
epigenetic programming of the human germ line. Nat Rev Genet 2016;17:585-600. 45. Ligthart S, Marzi C, Aslibekyan S, et al. DNA methylation signatures of chronic
15. Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and low-grade inflammation are associated with complex diseases. Genome Biol 2016;
implications. Nat Rev Genet 2011;13:97-109. 17:255. doi:10.1186/s13059-016-1119-5
16. Teschendorff AE, Menon U, Gentry-Maharaj A, et al. Age-dependent DNA 46. Rimmele P, Bigarella Carolina L, Liang R, et al. Aging-like phenotype and
methylation of genes that are suppressed in stem cells is a hallmark of cancer. defective lineage specification in SIRT1-deleted hematopoietic stem and
Genome Res 2010;20:440-446. progenitor cells. Stem Cell Reports 2014;3:44-59.
17. Bell CG, Xia Y, Yuan W, et al. Novel regional age-associated DNA methylation 47. Hernandez DG, Nalls MA, Gibbs JR, et al. Distinct DNA methylation changes
changes within human common disease-associated loci. Genome Biol 2016;17:193. highly correlated with chronological age in the human brain. Hum Mol Genet
doi:10.1186/s13059-016-1051-8 2011;20:1164-1172.
18. Schalkwyk LC, Meaburn EL, Smith R, et al. Allelic skewing of DNA methylation 48. Houseman EA. DNA methylation and cell-type distribution. In: Teschendorff AE,
is widespread across the genome. Am J Hum Genet 2010;86:196-212. ed. Computational and Statistical Epigenomics. Translational Bioinformatics 7.
19. Shoemaker R, Deng J, Wang W, Zhang K. Allele-specific methylation is prevalent and Dordrecht, Netherlands: Springer; 2015:35-50.
is contributed by CpG-SNPs in the human genome. Genome Res 2010;20:883-889. 49. Dekkers KF, van Iterson M, Slieker RC, et al. Blood lipids influence DNA
20. Heintzman ND, Ren B. Finding distal regulatory elements in the human genome. methylation in circulating cells. Genome Biol 2016;17:138. doi:10.1186/s13059-
Curr Opin Genet Dev 2009;19:541-549. 016-1000-6
21. Richards EJ. Inherited epigenetic variation--revisiting soft inheritance. Nat Rev 50. Rakyan VK, Beyan H, Down TA, et al. Identification of type 1 diabetes-associated
Genet 2006;7:395-401. DNA methylation variable positions that precede disease diagnosis. PLoS Genet
22. Brandeis M, Frank D, Keshet I, et al. Sp1 elements protect a CpG island from de 2011;7:e1002300. doi:10.1371/journal.pgen.1002300
novo methylation. Nature 1994;371:435-438. 51. Roos L, Spector TD, Bell CG. Using epigenomic studies in monozygotic twins to
23. Lienert F, Wirbelauer C, Som I, Dean A, Mohn F, Schubeler D. Identification of improve our understanding of cancer. Epigenomics 2014;6:299-309.
genetic elements that autonomously determine DNA methylation states. Nat Genet 52. Andrews SV, Ladd-Acosta C, Feinberg AP, Hansen KD, Fallin MD. “Gap
2011;43:1091-1097. hunting” to characterize clustered probe signals in Illumina methylation array data.
24. Stadler MB, Murr R, Burger L, et al. DNA-binding factors shape the mouse Epigenetics Chromatin 2016;9:56. doi:10.1186/s13072-016-0107-z
methylome at distal regulatory regions. Nature 2011;480:490-495. 53. Chen Y-a, Lemire M, Choufani S, et al. Discovery of cross-reactive probes and
25. Domcke S, Bardet AF, Adrian Ginno P, Hartl D, Burger L, Schubeler D. polymorphic CpGs in the Illumina Infinium HumanMethylation450 microarray.
Competition between DNA methylation and transcription factors determines Epigenetics 2013;8:203-209.
binding of NRF1. Nature 2015;528:575-579. 54. Chen L, Ge B, Casale Francesco P, Vasquez L, et al. Genetic drivers of
26. Yin Y, Morgunova E, Jolma A, et al. Impact of cytosine methylation on DNA epigenetic and transcriptional variation in human immune cells. Cell 2016;167:
binding specificities of human transcription factors. Science 2017;356. doi: 1398-414.e24.
10.1126/science.aaj2239 55. Dayeh T, Volkov P, Salo S, et al. Genome-wide DNA methylation analysis of
27. Bonder MJ, Luijk R, Zhernakova DV, et al. Disease variants alter transcription human pancreatic islets from type 2 diabetic and non-diabetic donors identifies
factor levels and methylation of their binding sites. Nat Genet 2017;49:131-138. candidate genes that influence insulin secretion. PLoS Genet 2014;10:e1004160.
28. Kasowski M, Kyriazopoulou-Panagiotopoulou S, Grubert F, et al. Extensive doi:10.1371/journal.pgen.1004160
variation in chromatin states across humans. Science 2013;342:750-752. 56. Olsson AH, Volkov P, Bacos K, et al. Genome-wide associations between
29. Grandi FC, Rosser JM, Newkirk SJ, et al. Retrotransposition creates sloping genetic and epigenetic variation influence mRNA expression and insulin secretion
shores: a graded influence of hypomethylated CpG islands on flanking CpG sites. in human pancreatic islets. PLoS Genet 2014;10:e1004735. doi:10.1371/
Genome Res 2015;25:1135-1146. journal.pgen.1004735
30. Schubeler D. Function and information content of DNA methylation. Nature 2015; 57. McClay JL, Shabalin AA, Dozmorov MG, et al. High density methylation QTL
517:321-326. analysis in human blood via next-generation sequencing of the methylated genomic
31. Bell CG, Finer S, Lindgren CM, et al. Integrated genetic and epigenetic analysis DNA fraction. Genome Biol 2015;16:291. doi:10.1186/s13059-015-0842-7
identifies haplotype-specific methylation in the FTO type 2 diabetes and obesity 58. Su D, Wang X, Campbell MR, et al. Distinct epigenetic effects of tobacco
susceptibility locus. PLoS One 2010;5:e14040. doi:10.1371/journal.pone.0014040 smoking in whole blood and among leukocyte subtypes. PLoS One 2016;11:
32. Gibbs JR, van der Brug MP, Hernandez DG, et al. Abundant quantitative trait loci e0166486. doi:10.1371/journal.pone.0166486
exist for DNA methylation and gene expression in human brain. PLoS Genet 59. Bauer M, Linsel G, Fink B, et al. A varying T cell subtype explains apparent
2010;6:e1000952. doi:10.1371/journal.pgen.1000952 tobacco smoking induced single CpG hypomethylation in whole blood. Clin
33. Liu Y, Li X, Aryee Martin J, et al. GeMes, clusters of DNA methylation under Epigenetics 2015;7:81. doi:10.1186/s13148-015-0113-1
genetic control, can inform genetic and epigenetic analysis of disease. Am J Hum 60. Grant RW, Dixit VD. Adipose tissue as an immunological organ. Obesity (Silver
Genet 2014;94:485-495. Spring) 2015;23:512-518.

www.obesityjournal.org Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 1479


Obesity Epigenomic Analysis of Human Obesity Bell

61. Huh JY, Park YJ, Ham M, Kim JB. Crosstalk between adipocytes and immune 89. Braun KVE, Dhana K, de Vries PS, et al. Epigenome-wide association study
cells in adipose tissue inflammation and metabolic dysregulation in obesity. Mol (EWAS) on lipids: the Rotterdam Study. Clin Epigenetics 2017;9:15. doi:10.1186/
Cells 2014;37:365-371. s13148-016-0304-4
62. Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell 90. Kriebel J, Herder C, Rathmann W, et al. Association between DNA methylation in
2014;156:20-44. whole blood and measures of glucose metabolism: KORA F4 study. PLoS One
63. Emdin CA, Khera AV, Natarajan P, et al. Genetic association of waist-to-hip ratio 2016;11:e0152314. doi:10.1371/journal.pone.0152314
with cardiometabolic traits, type 2 diabetes, and coronary heart disease. JAMA 91. Mamtani M, Kulkarni H, Dyer TD, et al. Genome- and epigenome-wide
2017;317:626-634. association study of hypertriglyceridemic waist in Mexican American families.
64. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Clin Epigenetics 2016;8:6. doi:10.1186/s13148-016-0173-x
Nature 2017;542:177-185. 92. Ventham NT, Kennedy NA, Adams AT, et al. Integrative epigenome-wide
65. Wensveen FM, Jelencic V, Valentic S, et al. NK cells link obesity-induced adipose analysis demonstrates that DNA methylation may mediate genetic risk in
stress to inflammation and insulin resistance. Nat Immunol 2015;16:376-385. inflammatory bowel disease. Nat Commun 2016;7:13507. doi:10.1038/
ncomms13507
66. Wiencke JK, Butler R, Hsuang G, et al. The DNA methylation profile of activated
human natural killer cells. Epigenetics 2016;11:363-380. 93. Agouridis AP, Elisaf M, Milionis HJ. An overview of lipid abnormalities in
patients with inflammatory bowel disease. Ann Gastroenterol 2011;24:181-187.
67. Bell CG. Epigenetic factors in human obesity. In: Tollefsbol TO, ed. Epigenetics
in Human Disease. London, UK: Elsevier Academic Press; 2012:273-296. 94. Benton MC, Johnstone A, Eccles D, et al. An analysis of DNA methylation in human
adipose tissue reveals differential modification of obesity genes before and after gastric
68. van Dijk SJ, Molloy PL, Varinli H, Morrison JL, Muhlhausler BS. Epigenetics and bypass and weight loss. Genome Biol 2015;16:8. doi:10.1186/s13059-014-0569-x
human obesity. Int J Obes (Lond) 2015;39:85-97.
95. Horvath S, Erhart W, Brosch M, et al. Obesity accelerates epigenetic aging of
69. van Iterson M, van Zwet EW, Consortium B, Heijmans BT. Controlling bias and human liver. Proc Natl Acad Sci U S A 2014;111:15538-15543.
inflation in epigenome- and transcriptome-wide association studies using the
empirical null distribution. Genome Biol 2017;18:19. doi:10.1186/s13059-016- 96. Simpkin AJ, Howe LD, Tilling K, et al. The epigenetic clock and physical development
1131-9 during childhood and adolescence: longitudinal analysis from a UK birth cohort. Int J
Epidemiol 2017;46:549–558.
70. Dick KJ, Nelson CP, Tsaprouni L, et al. DNA methylation and body-mass index: a
genome-wide analysis. Lancet 2014;383:1990-1998. 97. Nevalainen T, Kananen L, Marttila S, et al. Obesity accelerates epigenetic aging in
middle-aged but not in elderly individuals. Clin Epigenetics 2017;9:20. doi:
71. Agha G, Houseman EA, Kelsey KT, Eaton CB, Buka SL, Loucks EB. Adiposity is 10.1186/s13148-016-0301-7
associated with DNA methylation profile in adipose tissue. Int J Epidemiol 2015;
44:1277-1287. 98. Faulk C, Dolinoy DC. Timing is everything: The when and how of
environmentally induced changes in the epigenome of animals. Epigenetics 2011;
72. Demerath EW, Guan W, Grove ML, et al. Epigenome-wide association study 6:791-797.
(EWAS) of BMI, BMI change and waist circumference in African American
99. Dominguez-Salas P, Moore SE, Baker MS, et al. Maternal nutrition at conception
adults identifies multiple replicated loci. Hum Mol Genet 2015;24:4464-4479.
modulates DNA methylation of human metastable epialleles. Nat Commun 2014;5:
73. Huang T, Zheng Y, Qi Q, et al. DNA methylation variants at HIF3A locus, B- 3746. doi:10.1038/ncomms4746
vitamin intake, and long-term weight change: gene-diet interactions in two U.S.
100. Gluckman PD, Hanson MA, Cooper C, Thornburg KL. Effect of in utero and
cohorts. Diabetes 2015;64:3146-3154.
early-life conditions on adult health and disease. N Engl J Med 2008;359:61-73.
74. R€onn T, Volkov P, Gillberg L, et al. Impact of age, BMI and HbA1c levels on the
101. Guenard F, Deshaies Y, Cianflone K, Kral JG, Marceau P, Vohl M-C. Differential
genome-wide DNA methylation and mRNA expression patterns in human adipose tissue
methylation in glucoregulatory genes of offspring born before vs. after maternal
and identification of epigenetic biomarkers in blood. Hum Mol Genet 2015;24:3792-
gastrointestinal bypass surgery. Proc Natl Acad Sci USA 2013;110:11439-11444.
3813.
102. Sharp GC, Lawlor DA, Richmond RC, et al. Maternal pre-pregnancy BMI and
75. Richmond RC, Sharp GC, Ward ME, et al. DNA methylation and BMI:
gestational weight gain, offspring DNA methylation and later offspring adiposity:
investigating identified methylation sites at HIF3A in a causal framework.
findings from the Avon Longitudinal Study of Parents and Children. Int J
Diabetes 2016;65:1231-1244.
Epidemiol 2015;44:1288-1304.
76. Main AM, Gillberg L, Jacobsen AL, et al. DNA methylation and gene expression
103. Sorensen T, Ajslev TA, Angquist L, Morgen CS, Ciuchi IG, Davey Smith G.
of HIF3A: cross-tissue validation and associations with BMI and insulin
Comparison of associations of maternal peri-pregnancy and paternal
resistance. Clin Epigenetics 2016;8:89. doi:10.1186/s13148-016-0258-6
anthropometrics with child anthropometrics from birth through age 7 y assessed in
77. Willmer M, Berglind D, Sørensen TIA, N€aslund E, Tynelius P, Rasmussen F. the Danish National Birth Cohort. Am J Clin Nutr 2016;104:389-396.
Surgically induced interpregnancy weight loss and prevalence of overweight and
104. Horikoshi M, Beaumont RN, Day FR, et al. Genome-wide associations for birth
obesity in offspring. PLoS One 2013;8:e82247. doi:10.1371/journal.pone.0082247
weight and correlations with adult disease. Nature 2016;538:248-252.
78. Mendelson MM, Marioni RE, Joehanes R, et al. Association of body mass index
105. Richmond RC, Timpson NJ, Felix JF, et al. Using genetic variation to explore the
with DNA methylation and gene expression in blood cells and relations to
causal effect of maternal pregnancy adiposity on future offspring adiposity: a
cardiometabolic disease: a Mendelian randomization approach. PLoS Med 2017; mendelian randomisation study. PLoS Med 2017;14:e1002221. doi:10.1371/
14:e1002215. doi:10.1371/journal.pmed.1002215 journal.pmed.1002221
79. Frazier-Wood AC, Aslibekyan S, Absher DM, et al. Methylation at CPT1A locus 106. Lawlor D, Richmond R, Warrington N, et al. Using Mendelian randomization to
is associated with lipoprotein subfraction profiles. J Lipid Res 2014;55:1324-1330. determine causal effects of maternal pregnancy (intrauterine) exposures on
80. Breeze CE, Paul DS, van Dongen J, et al. eFORGE: a tool for identifying cell offspring outcomes: Sources of bias and methods for assessing them. Wellcome
type-specific signal in epigenomic data. Cell Rep 2016;17:2137-2150. Open Res 2017;2:11. doi:10.12688/wellcomeopenres.10567.1
81. Ziller MJ, Gu H, Muller F, et al. Charting a dynamic DNA methylation landscape 107. Leung D, Jung I, Rajagopal N, et al. Integrative analysis of haplotype-resolved
of the human genome. Nature 2013;500:477-481. epigenomes across human tissues. Nature 2015;518:350-354.
82. Aslibekyan S, Demerath EW, Mendelson M, et al. Epigenome-wide study 108. Kuehnen P, Mischke M, Wiegand S, et al. An Alu element-associated
identifies novel methylation loci associated with body mass index and waist hypermethylation variant of the POMC gene is associated with childhood obesity.
circumference. Obesity (Silver Spring) 2015;23:1493-1501. PLoS Genet 2012;8:e1002543. doi:10.1371/journal.pgen.1002543
83. Das M, Sha J, Hidalgo B, et al. Association of DNA methylation at CPT1A locus 109. Jaffe AE, Murakami P, Lee H, et al. Bump hunting to identify differentially
with metabolic syndrome in the Genetics of Lipid Lowering Drugs and Diet methylated regions in epigenetic epidemiology studies. Int J Epidemiol 2012;41:
Network (GOLDN) Study. PLoS One 2016;11:e0145789. doi:10.1371/ 200-209.
journal.pone.0145789 110. Liu Y, Aryee MJ, Padyukov L, et al. Epigenome-wide association data implicate
84. Al Muftah WA, Al-Shafai M, Zaghlool SB, et al. Epigenetic associations of type 2 DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat
diabetes and BMI in an Arab population. Clin Epigen 2016;8:13. doi:10.1186/ Biotech 2013;31:142-147.
s13148-016-0177-6 111. Lillycrop KA, Burdge GC. Epigenetic mechanisms linking early nutrition to long
85. Shah S, Bonder MJ, Marioni RE, et al. Improving phenotypic prediction by term health. Best Pract Res Clin Endocrinol Metab 2012;26:667-676.
combining genetic and epigenetic associations. Am J Hum Genet 2015;97:75-85. 112. Petersen AK, Zeilinger S, Kastenmuller G, et al. Epigenetics meets metabolomics:
86. Gagnon F, Aissi D, Carrie A, Morange PE, Tregouet DA. Robust validation of an epigenome-wide association study with blood serum metabolic traits. Hum Mol
methylation levels association at CPT1A locus with lipid plasma levels. J Lipid Genet 2014;23:534-545.
Res 2014;55:1189-1191. 113. Ecker S, Chen L, Pancaldi V, et al. Genome-wide analysis of differential
87. Irvin MR, Zhi D, Joehanes R, et al. Epigenome-wide association study of fasting transcriptional and epigenetic variability across human immune cell types. Genome
blood lipids in the Genetics of Lipid-lowering Drugs and Diet Network study. Biol 2017;18:18. doi:10.1186/s13059-017-1156-8
Circulation 2014;130:565-572. 114. Ward LD, Kellis M. HaploReg v4: systematic mining of putative causal variants,
88. Pfeiffer L, Wahl S, Pilling LC, et al. DNA methylation of lipid-related genes cell types, regulators and target genes for human complex traits and disease.
affects blood lipid levels. Circ Cardiovasc Genet 2015;8:334-342. Nucleic Acids Res 2016;44:D877-D881.

1480 Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 www.obesityjournal.org


Review Obesity
EPIDEMIOLOGY/GENETICS

115. Claussnitzer M, Dankel SN, Kim K-H, et al. FTO obesity variant circuitry and 121. Bolton JL, Smith SH, Huff NC, et al. Prenatal air pollution exposure induces
adipocyte browning in humans. N Engl J Med 2015;373:895-907. neuroinflammation and predisposes offspring to weight gain in adulthood in a sex-
116. Jufvas A, Sjodin S, Lundqvist K, Amin R, Vener AV, Stralfors P. Global specific manner. FASEB J 2012;26:4743-4754.
differences in specific histone H3 methylation are associated with overweight and 122. Park S, Skaar D, Jirtle RL, Hoyo C. Epigenetics, obesity and early-life cadmium
type 2 diabetes. Clin Epigenetics 2013;5:15. doi:10.1186/1868-7083-5-15 or lead exposure. Epigenomics 2016;9:57-75.
117. Pazienza V, Panebianco C, Rappa F, et al. Histone macroH2A1.2 promotes 123. Burgio E, Lopomo A, Migliore L. Obesity and diabetes: from genetics to
metabolic health and leanness by inhibiting adipogenesis. Epigenetics Chromatin epigenetics. Mol Biol Rep 2015;42:799-818.
2016;9:45. doi:10.1186/s13072-016-0098-9 124. Bojesen SE, Timpson N, Relton C, Davey Smith G, Nordestgaard BG. AHRR
118. Sun W, Poschmann J, Cruz-Herrera Del Rosario R, et al. Histone acetylome-wide (cg05575921) hypomethylation marks smoking behaviour, morbidity and mortality.
association study of autism spectrum disorder. Cell 2016;167:1385-1397.e11. Thorax 2017;72:646-653.
119. Bell CG. The emerging potential for epigenetic therapeutics in noncancer 125. Hedman ÅK, Mendelson MM, Marioni RE, et al. Epigenetic patterns in blood
disorders. In: Egger G, Arimondo P, eds. Drug Discovery in Cancer Epigenetics. associated with lipid traits predict incident coronary heart disease events and are
Oxford, UK: Academic Press; 2016:437-456. enriched for results from genome-wide association studies. Circ Cardiovasc Genet
120. Razak F, Davey Smith G, Subramanian SV. The idea of uniform change: is it time 2017;10:e001487. doi:10.1161/CIRCGENETICS.116.001487
to revisit a central tenet of Rose’s “Strategy of Preventive Medicine”? Am J Clin 126. Simpson JT, Workman RE, Zuzarte PC, David M, Dursi LJ, Timp W. Detecting DNA
Nutr 2016;104:1497-1507. cytosine methylation using nanopore sequencing. Nat Methods 2017;14:407-410.

www.obesityjournal.org Obesity | VOLUME 25 | NUMBER 9 | SEPTEMBER 2017 1481

You might also like