ArticlePDF Available

Primers on nutrigenetics and nutri(epi)genomics: Origins and development of precision nutrition

Authors:

Abstract and Figures

Understanding the relationship between genotype and phenotype is a central goal not just for genetics but also for medicine and biological sciences. Despite outstanding technological progresses, genetics alone is not able to completely explain phenotypes, in particular for complex diseases. Given the existence of a “missing heritability”, growing attention has been given to non-mendelian mechanisms of inheritance and to the role of the environment. The study of interaction between gene and environment represents a challenging but also a promising field with high potential for health prevention, and epigenetics has been suggested as one of the best candidate to mediate environmental effects on the genome. Among environmental factors able to interact with both genome and epigenome, nutrition is one of the most impacting. Not just our genome influences the responsiveness to food and nutrients, but vice versa, nutrition can also modify gene expression through epigenetic mechanisms. In this complex picture, nutrigenetics and nutrigenomics represent appealing disciplines aimed to define new prospectives of personalized nutrition. This review introduces to the study of gene-environment interactions and describes how nutrigenetics and nutrigenomics modulate health, promoting or affecting healthiness through life-style, thus playing a pivotal role in modulating the effect of genetic predispositions.
Content may be subject to copyright.
Review
Primers on nutrigenetics and nutri(epi)genomics: Origins and
development of precision nutrition
Laura Bordoni, Rosita Gabbianelli
*
Unit of Molecular Biology, School of Pharmacy, University of Camerino, 62032, Camerino, MC, Italy
article info
Article history:
Received 4 December 2018
Accepted 8 March 2019
Available online 13 March 2019
Keywords:
Nutrigenetics
Nutrigenomics
Epigenetics
Gene-environment interaction
Personalized nutrition
abstract
Understanding the relationship between genotype and phenotype is a central goal not just for genetics
but also for medicine and biological sciences. Despite outstanding technological progresses, genetics
alone is not able to completely explain phenotypes, in particular for complex diseases. Given the exis-
tence of a missing heritability, growing attention has been given to non-mendelian mechanisms of
inheritance and to the role of the environment. The study of interaction between gene and environment
represents a challenging but also a promising eld with high potential for health prevention, and epi-
genetics has been suggested as one of the best candidate to mediate environmental effects on the
genome.
Among environmental factors able to interact with both genome and epigenome, nutrition is one of
the most impacting. Not just our genome inuences the responsiveness to food and nutrients, but vice
versa, nutrition can also modify gene expression through epigenetic mechanisms. In this complex pic-
ture, nutrigenetics and nutrigenomics represent appealing disciplines aimed to dene new prospectives
of personalized nutrition. This review introduces to the study of gene-environment interactions and
describes how nutrigenetics and nutrigenomics modulate health, promoting or affecting healthiness
through life-style, thus playing a pivotal role in modulating the effect of genetic predispositions.
©2019 Elsevier B.V. and Société Française de Biochimie et Biologie Moléculaire (SFBBM). All rights
reserved.
Contents
1. Gene-environment interactions: from genetics to epigenetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ....................... 157
1.1. Healthy or unhealthy phenotype: is it nature or nurture? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...........................157
1.2. Genetic determinants of health . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . ..........................................157
1.3. Limits and pitfalls of the genetic approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . ......................................158
1.4. The epigenome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..................................................158
1.5. Interaction between genetics and epigenetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......................................160
1.6. Epigenetics as a bridge between the environment and the genome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...........................160
1.7. The role of environment as a strong determinant of health . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...........................161
2. Nutrigenetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ....................... 161
2.1. Introduction to nutrigenetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..........................................161
2.2. The role of genetic variants in nutrition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......................................162
2.3. Genetic determinants of responsiveness to dietary interventions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...........................163
2.4. Nutrigenetic tests for personalized nutrition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......................................163
3. Nutrigenomics . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ....................... 164
3.1. Introduction to nutrigenomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..........................................164
3.2. Nutritional factors that can influence the epigenome and the mechanisms involved . . . . . . . . . . . . . . . . . . . . . . . . . . . ......................164
3.3. Susceptible period of exposure, epigenetic reprogramming and transgenerational effects in nutrigenomics . . . . . . . . . . . . . . . . . . . ...........165
*Corresponding author.School of Pharmacy, University of Camerino, Via Gentile
III da Varano, Camerino, MC, Italy.
E-mail address: rosita.gabbianelli@unicam.it (R. Gabbianelli).
Contents lists available at ScienceDirect
Biochimie
journal homepage: www.elsevier.com/locate/biochi
https://doi.org/10.1016/j.biochi.2019.03.006
0300-9084/©2019 Elsevier B.V. and Société Française de Biochimie et Biologie Moléculaire (SFBBM). All rights reserved.
Biochimie 160 (2019) 156e171
3.4. Interactions between nutrigenetics and nutrigenomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .......................165
3.5. Personalized epigenomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .......................166
4. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ................................................ 166
Author contributions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ................................................ 166
Declaration of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ................................................ 167
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ................................................ 167
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .......................167
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .......................167
1. Gene-environment interactions: from genetics to
epigenetics
1.1. Healthy or unhealthy phenotype: is it nature or nurture?
As early as 350 BCE, trying to understand the origin of human
behavior, philosophers such as Plato and Aristotle epistemologi-
cally gave raised to the Nature vs Nurture debate. About 2000 years
after, we are nowadays almost sure that nor nature or nurture can
exist in a manner that can be considered independently quanti-
able [1]. To paraphrase Richard Lewontin [2], There are no genetic
factors that can be studied independently of the environment, and
there are no environmental factors that function independently of
the genome.
After the advent of the Human Genome Project [3], genome
scientists, medical geneticists, and science policy leaders worked to
establish the value of genomic science by dening the elds of
public health genomics and precision medicine. These elds gave
rise to a new post-genomic combination of methods and disciplines
who made a shift from a nature versus nurturedichotomy to a
more systemic vision of the gene-environment interactions,
promising to lead to more accurate and consistent explanations for
diseases and a future based on personalized prevention and treat-
ment. Nevertheless, despite the consensus about the presence of
gene-environment interactions and their inuence on health and
disease, researchers struggled to dene, analyze and quantify the
environmental effects on the genome.
The concepts explained in this rst section introduce
strengthens and weakness of genetic and epigenetic approaches to
study gene-environment interactions. These notions are propae-
deutic to clearly comprehend origins and development of nutri-
genetics and nutrigenomics, their limits and pitfalls, and the
importance of the integration between genetic and epigenetic in-
formation in precision nutrition.
1.2. Genetic determinants of health
Inheritable information given by the primary sequence of DNA
plays a key role in determining variations in the susceptibility and
severity of disease. The human genome includes about 3 10
9
base
pairs of DNA, and the amount of genetic variation in humans is such
that no two subjects (except for identical twins), have ever been
genetically identical. The amount of genetic variation between any
two humans is about 0.1%. This signies that about one base pair
out of every 1000 is different between any two individuals [4]. In
both plant and animal genomes, the predominant forms of
sequence variations is represented by single nucleotide poly-
morphisms (SNPs), which distinguished from rare variations by
having a frequency of the least abundant allele of 1% or more [5].
Copy number variants (CNVs) or copy number polymorphisms
(CNPs), including duplications, deletions, insertions and complex
multi-site variants, are again other source of variation in the
genome [6]. Genetic variants can differ between ethnicities, are
inherited from ancestors and can take place through the entire
genome. Whether functional role of various non-synonymous
variants (comprising nonsense, missense, frameshift and other
types of variations) occurring in the coding region has been hy-
pothesized, it is still matter of debate how genetic variants taking
Abbreviations
5caC 5-carboxylcytosine
5 fC 5-formylcytosine
5hmC hydroxymethylcytosine
AKU alkaptonuria
ASM alleles specic DNA methylation
BMI body mass index
CGIs CpG islands
CNP copy number polymorphisms
CNV copy number variant
DNMT DNA methyl transferases
DOHaD developmental origin of health and disease
DTC direct-to-consumer
eQTL expression quantitative trait locus
GxE gene-environment interactions
HDAC histone deacetylase
Insdel insertion/deletion
LCT lactase
LD linkage disequilibrium
LEARn latent early life associated regulation
LINE long interspersed nuclear elements
LTR long terminal repeat
MTHFD1 5,10-methylenetetrahydrofolate dehydrogenase 1
MTHFR methylenetetrahydrofolate reductase
PAR predictive adaptive response
PEMT phosphatidylethanolamine-N-methyltransferase
PGCs primordial germ cells
PKU phenylketonuria
SAH S-adenosylhomocysteine
SAM S-adenosylmethionine
SINEs short interspersed nuclear elements
SNP single nucleotide polymorphism
TET Ten-Eleven Translocation
WHO World Health Organization
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171 157
place in the non-coding genome can actually have an impact [7].
That question is particularly challenging considering that genetic
variants in the non-coding genome are the most abundant in
general, and also that most of the single-nucleotide variants
signicantly associated with an increased risk of complex diseases
have been mapped to non-coding regions [7].
However, understanding the connection between genotype and
phenotype is one of the main goals which several projects are
contributing to achieve. First of all, the reference human genome
sequence provided the basis for the study of human genetics; then
the public catalogue of variant sites (dbSNP 129) archived
approximately 11 million SNPs and 3 million short insertions and
deletions (insdels) identied in the genome; again, the Interna-
tional HapMap Project indexed both allele frequencies and the
correlation patterns between nearby variants (i.e. the linkage
disequilibrium), across several populations for 3.5 million SNPs
[3,8,9]. This knowledge leds to the genome-wide association
studies (GWAS), which analyze numerous hundred thousand of
variant sites, combining them with the information about linkage
disequilibrium structure and permitting to test the majority of
common variants (those with 5% minor allele frequency) for their
association with disease. The 1000 Genomes Project (describing the
genomes of 1092 individuals from 14 population) claried the
properties and distribution of common and rare variations,
providing insights into the processes that shape genetic diversity,
and strongly increased the knowledge about disease biology
[10,11]. As a result, GWAS and other genetic studies identied the
association of more than 15000 SNPs with numerous pathologies or
traits [12]. They have impressively extended our knowledge about
how germline genetic variations impact disease susceptibility and
outcome [13,14], and also about how somatic changes in DNA
sequence severely impair gene expression, leading to the genesis
and advancement of disease [15].
However, these increased knowledge of genotypic information
were rarely anked with downstream functional studies, that are
still needed to identify causal variants that contribute to human
phenotypes. Expression quantitative trait locus (eQTL) assays were
performed to ascertain associations between genotypes and gene
expression variations, but in most eQTLs the causal variant was
unidentied, and even when the expected causal variant could be
reliably identied, the involved regulatory mechanism was largely
challenging to be recognized [16,17]. Furthermore, whether genetic
inuence is clearly established for monogenic traits, the landscape
becomes more and more intricate for complex polygenic
characters.
1.3. Limits and pitfalls of the genetic approach
For more than a century, individual differences in human traits
have been studied; nevertheless the causes of variation in human
traits, complex traits in particular, still remain controversial
[18e23]. Actually, in the last years, research has denitely estab-
lished that GWAS ndings alone, besides large investments and
scientic efforts, does not tent to identify causal loci of complex
diseases and predict individual disease risk [13]. This has been
hypothesized to be due, among other factors, to the fact that GWAS
avoid to consider CNV and, above all, environmental factors in the
analysis. Large-scale GWAS demonstrates that many genetic vari-
ants contribute to the complex traits variation, but the effect sizes
for these traits are typically small. Furthermore, the sum of the
variance explained by the noticed variants is much smaller than the
reported heritability of the trait. This surprising and interesting
concept has been referred as missing heritability[24,25].
These observations contrast with the common diseaseecommon
variant hypothesis [13], which advocated that common variants
distributed in all populations determine phenotypic variation or
disease risk and that these variants all together are responsible for
an additive or multiplicative effect on trait variation or disease risk.
On these behalf, several explanations have been suggested to clarify
the architecture of complex traits and diseases: (A) the hypothesis
that a large number of common variants exerting a small-effect
account for disease risk and quantitative trait variation; (B) the
hypothesisthat a large number of rare variants having a large-effect
motivates the observed associations; or (C) the theory that a com-
bination of genotypic, epigenetic, and environmental interactions
can explain the observed relations [13]. This complex scenery leads
some researchers to focus on the importance of non-additive vari-
ation models in genetics [26,27]. Beside, considering that the nature
of complex diseases is multi-factorial, many researchers have sup-
ported the idea that major factors contributing to the missing her-
itability are the interactions among genetic loci, so-called epistatic
interactions. Indeed, multifaceted interactions between environ-
mental factors and genetic variants, both potentially associated to
disease risk, have been suggest to be taken into account.
For all these reasons, while numerous studies in the last decades
centered their attention to the identication of different genetic
variants that could explain a certain phenotype, nowadays concepts
such as epistasis, gene-gene interaction and gene-environment in-
teractions represent the research focus that could provide further
information about the genetic determinants of a certain phenotype
[24]. Moreover, this landscape highlights opportunities to consider
epigenetics as a functional modier of the genome and a major
contributing factor for disease etiology [28]. If heritability is classi-
cally described as the ratio of the genetic to the total phenotypic
variance, in a population [29], the more contemporary concept of
broad sense heritabilitydenotes the genetic effect including non-
additive components, such as gene-gene interactions, gene-
environment interactions (G E), and epigenetics [13].
1.4. The epigenome
Beginning over 70 years ago, the eld of epigenetics massively
grew to elucidate mechanisms through which various cellular
phenotypes originate from a single genotype throughout the
intricate process of developmental morphogenesis termed
epigenesis. The word ‘‘epigenetics’’ was rstly coined by Conrad
Waddington (1905e1975) in 1940s. He used it to dene the branch
of biology which studies the causal interactions between genes and
their products, which bring the phenotype into being[30]. After
some debates, a consensus denition was delineated and epige-
netics was dened as stably heritable phenotypes resulting from
changes in a chromosome without changes in gene sequence[31].
Epigenetic mechanisms of gene regulation, which collectively make
up the epigenome, mainly encompass enzymatic methylation of
cytosine bases (DNA methylation), post-translational modication
of tail domains of histone proteins (histone modications) and
chromatin remodeling. These modications arise all over the
developmental stages or ensue to environmental factors exposure,
providing both variability and rapid adaptability, that allow or-
ganisms to respond to external stimuli both in the short and in the
long term.
The relevance of epigenetics in the development is connected to
the ability of a single-cell zygote with a xed genomic sequence to
give rise to an organism with hundreds of cell types thanks to its
ability to control subset of genes expressed in each cell type. Spe-
cically, extensive removal and reestablishment of lineage-specic
epigenetic signatures, through a process designated as epigenetic
reprogramming, are at the basis of cellular differentiation [32].
Conservation and inheritance of these epigenetic marks during cell
division is fundamental to preserve a committed cell lineage and
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171158
cellular phenotype in descendant cells, and establish a memory of
transcriptional status. In detail, epigenetic marks are reprog-
rammed in a global scale, concomitantly with restoration of
developmental potency, at two points in the life cycle: rstly on
fertilization in the zygote, and secondly in primordial germ cells
(PGCs), that are the direct precursors of sperm or oocyte. A
distinctive set of mechanisms regulates epigenome erasure and re-
establishment [32e34]. In this picture, epigeneticmarks describe
the developmental potency of the zygote and promote differenti-
ation towards a specic cell fate in future cell generations.
Methylation of the fth carbon of the cytosine base in DNA and
post-translational histone tail modications are probably the best-
studied epigenetic modications in mammals [35]. DNA methyl-
ation is the covalent addition of a methyl group at the 5-carbon of a
cytosine ring, resulting in 5-methylcytosine (5 mC), likewise
informally dened as the fth baseof DNA. This reaction is
catalyzed by DNA methyltransferases (DNMTs) enzymes [36]. There
are three main DNMTs: DNMT1 copies methylation marks from the
parental strand of DNA to the newly synthesized strand during the
process of DNA replication (thus it is dened as the maintenance
DNMT, which allows transmission of DNA methylation patterns
from cell to cell), while DNMT3A and DNMT3B establish a de novo
DNA methylation [36,37]. DNA methylation is the most chemically
stable epigenetic modication and it is unambiguously stably
transmitted during cell division. As a consequence of its biologic
interest, it is the most well characterized epigenetic mark and the
most extensively measured in epidemiologic research.
In mammals, DNA methylation occurs primarily on cytosines
within a CpG dinucleotide, of whom approximately 70e80% are
methylated [38]. Besides, stretches of CpG-rich sequences with low
levels of DNA methylation, also called CpG islands (CGIs), exist
[11,39]. CpG islands are dened as sequences with a G þC content
above 60% and a ratio of CpG to GpC of at least 0.6 [40]. They
frequently are highly enriched at gene promoters (about 60% of all
mammalian gene promoters are CpG-rich). Unmethylated CpG
islands are usually open regions of DNA with low nucleosome oc-
cupancy (euchromatin), promoting relaxed chromatin structure
that facilitates accessibility to the transcription start site of RNA
polymerase II and other components of the basal transcription
machinery [41,42]. On the other hand, DNA methylation is
frequently related to gene repression [43,44]. Many targets of de
novo DNA methylation are promoters of stem cell- and germline-
specic genes during differentiation, repetitive DNA sequences,
such as those within the chromosomes centromeric and pericen-
tromeric regions or in the endogenous transposable elements (i.e.
long interspersed nuclear elements (LINEs), short interspersed
nuclear elements (SINEs) and long terminal repeat (LTR)-contain-
ing endogenous retroviruses) [43,45]. Moreover, DNA methylation
recruits methyl-CpG-binding proteins which interacts with pro-
teins that can play a role in the repression of genes with CpG islands
(i.e. Methyl CpG binding protein 1, MeCP1) or, on the other hand,
can add silencing modications to neighboring histones (i.e.
MeCP2) [46]. This harmonization between DNA methylation and
silencing histone marks determines the compaction of chromatin
and gene repression. However, DNA methylation is also found
within the bodies of genes, where higher levels of intragenic
methylation correlate with higher levels of gene expression. Thus,
the functional signicance of gene body methylation is less clear,
and regulation of alternative transcription initiation sites or regu-
lation of splicing are two potential role hypothesized to explain this
phenomenon [47e50].
Despite it was originally retained that DNA methylation was a
stabile mark which once established was then maintained
throughout the life course of the organism (because of its ther-
modynamic stability and the initial incertitude of a biochemical
mechanism that could directly remove the methyl group from
5 mC), it is now clear that DNA methylation can be dynamically
regulated [49]. Recent discoveries showed that, together with
DNMTs-mediated methylation processes, passive or enzymatically-
directed DNA demethylation also occur. Several DNA demethylases
such as Ten-Eleven Translocation (TET) proteins, Methyl Binding
Domain protein, DNA repair endonucleases XPG and a G/T
mismatch repair DNA glycosylase has been identied. They do not
act by directly removing the methyl group, but through a multistep
process linked either to DNA repair mechanisms or through further
modication of 5 mC such as 5-hydroxymethylcytosine (5hmC), 5-
formylcytosine (5 fC) and 5-carboxylcytosine (5caC). TET proteins
can oxidize 5 mC to 5hmC but also to 5 fC and/or 5caC, which are
subsequently excised by thymine DNA glycosylase, or deaminated
by activation-induced deaminase, whose deamination product (5-
hydroxymethyluracil), activates base-excision repair pathway
leading to demethylation [51e56]. Among these other DNA modi-
cations, 5hmC acquired growing importance, specically in
certain cell types, not just as an intermediate of demethylation
processes, but as an epigenetic mark itself. High levels of 5hmC are
found in embryonic stem cells, in multipotent adult stem cells and
progenitor cells. During differentiation, levels decrease in most of
cells, except that in Purkinje neurons and other neural subtypes,
where high levels can be still measured [57]. Like 5 mC, 5hmC is not
uniformly distributed though the genome. 5hmC are enriched
within gene bodies and at transcription start sites and promoters
associated with gene expression, supporting the premise that
5hmC is associated with gene activation [58e60]. Interestingly,
DNA hydroxymethylation has been demonstrated to be affected in
response to environmental stress through redox system alterations
and, in particular, TET proteins activation [61,62]. Which are the
connections between 5 mC, 5hmC and gene expression regulation
is still to be completely elucidated.
Together with DNA modications, epigenetic gene regulation
also includes modications in histones that make up the nucleo-
somes. Nucleosomes are the basic unit of chromatin in eukaryotic
organisms, composed by the DNA wrapped around a core of eight
histone proteins (H2A, H2B, H3 and H4), essential to reduce its
size. Beside this fundamental function, it is now clear that histones
are not only important for DNA packaging, but also exert pivotal
roles in gene expression regulation, in conjunction with DNA
methylation [63]. Histone proteins contain a globular domain and
an amino tail domain. The amino tail domains protrude out of the
nucleosomes and are rich in positively charged amino acids, that
interact with the negatively charged DNA. These tails are subject to
a large number of post-translational modications, among which
the most frequent are acetylation, methylation, ubiquitination,
sumoylation, and phosphorylation, raising up to thousands of
potential combinations of modications within a single nucleo-
some [64]. Thus, whereas DNA can primarily be methylated, his-
tones are capable of carrying a wide array of post-translational
modications, with different role in gene expression regulation
processes [65,66]. They are dynamic, and several enzymes
involved in their modulations have been identied [67]. Recur-
rently, specic histone variants are found at denite locations
within the chromatin or are used to demarcate heterochromatic
and euchromatic regions. Histone modications can directly in-
uence interactions between histone and DNA or between
different histones, or they can be targeted by protein effectors also
called histone-binding domains. To dene proteins that deposit,
remove and recognize histones post-translational modications,
respectively, the terms writer,eraserand readerwere coined
[68,69]. They can act in cooperation, and it is the peculiar
arrangement of histone modications at a specic site that
habitually denes which protein complexes are recruited to
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171 159
activate or repress transcription, catalyze further histone modi-
cations or recruit other histone-modifying proteins [70], regu-
lating various DNA-dependent processes, including DNA
replication, transcription and repair. The collection of the post
translational modications of specic amino acid residues within
the histones that leads to the binding of effector proteins that, in
turn, bring about specic cellular processesis dened as histone
code [71,72]. While mechanisms of transmission of DNA
methylation has been recognized, it is still debated how histone
modications are transmitted during cell replication [73,74].
Furthermore, more recent evidence suggests that local and three-
dimensional chromatin architecture provide additional levels of
gene regulation in pluripotent stem cells. Local chromatin archi-
tecture denes the position and density of nucleosomes as well as
the presence of histone variants [75,76]. However, its roles in
cellular reprogramming has not been completely elucidated yet.
Ongoing projects are producing cell-specic reference data sets
that offer a basis for dening the complex interaction between
epigenomic processes and the transcriptome: ENCODE (Encyclo-
pedia of DNA Elements) project and the International Human
Epigenome Consortium [77] intended to classify the regulatory
elements in human cells and to investigate the epigenomic sig-
natures of cell cultures; the Roadmap Epigenomics Project (from
US National Institutes of Health) extends the ENCODE project and
is devoted to clarify in what way epigenetics contribute to human
biology and disease [78,79]. Providing reference epigenomes for
numerous human tissues and cell-types, research provided the
basis to understand how epigenomic are linked to the corre-
sponding genetic information. The nal goal would be to clarify
the complete landscape of epigenomic elements which controls
gene expression in the human body [80].
1.5. Interaction between genetics and epigenetics
Epigenetic mechanisms may be considered complementary to
genetic functions in the regulation of gene expression and can be
saw as the way by which a specic cell or tissue interprets the
genome information [81]. At the same time, primary DNA sequence
is a strong determinant of the epigenetic state. This can be
evidently inferred by noting that the distribution of epigenetic
marks across the genome is, at least in part, determined by CpG
density and G:C content in the sequence [82,83]. Additionally,
proximity to repetitive elements such as Alu and LINE, nuclear ar-
chitecture and binding sequences for transacting proteins repre-
sent further genetic inuences. Furthermore, some evidences
suggested that genetic polymorphisms can affect epigenetic state
[34]. In fact, mutations in genes encoding epigenetic modiers
(such as DNMTs, chromatin remodeling proteins or histone modi-
fying enzymes) can contribute to epigenetic changes, and have
been well documented in several diseases [34]. Aberrant epigenetic
modications can directly modulate regulation of target genes or
can interact with specic genetic variants predisposing to them
[34]. Furthermore, studies that investigated both genetic variations
and DNA methylation demonstrated that alleles specicDNA
methylation, related to polymorphic nucleotides situated nearby
the DNA methylation site, can extensively occur through the
genome [84].
Given the complexity of the genome and the notable intricacyof
epigenetic changes, that take account of dozens of different post-
translational histone modications and more than 50 million
sites of potential DNA methylation in a diploid human genome, it
appears that no two human cells would have identical epigenomes,
which, additionally, change over time in response to develop-
mental and pathological progressions, as well as consequentially to
environmental exposures and random drift [34].
1.6. Epigenetics as a bridge between the environment and the
genome
In accordance with the World Health Organization (WHO), more
than 13 million deceases per annum are caused by environmental
issues and so far as 24% of disease is due to exposures which could
be prevented [85]. A conspicuous amount of lifestyle and envi-
ronmental factors have been revealed to affect numerous diseases;
however, not all of them have been characterized as genotoxic
agents, able to promote DNA sequence mutation [86]. Thus,
genome-environment interactions have been discussed exten-
sively, and the role of epigenetics has been progressively more
acknowledged as a mechanism of interface between them [87].
Indeed, multiple differences in gene expression have been recog-
nized in numerous tissues already from newborn identical twins
(presumably reecting intrauterine epigenetic differences), sug-
gesting that not just differences in the genome, but also different
exposure to environment, can affect the epigenome [88]. Consid-
ering that several epigenetic events have been identied as tissue-
specic and reversible, epigenetics is particularly compelling to
explain differential susceptibilities in the exposed population and
why exposures affect precise organs.
Since a lot of epigenetic modications can be modulated by both
external and internal factors and can change gene expressions,
epigenetics is considered a key mechanism through which ge-
nomes interact with environmental exposures, providing a novel
approach in the exploration of etiological factors in numerous
environment related pathologies [89]. As these epigenetic marks
are potentially cumulative and could take place over time, to
identify the cause-effect associations among epigenetic changes,
environmental factors and diseases represents a big goal. However,
even if mechanisms of action of some of these agents remains to be
completely elucidated, some others has been well characterized
[34,89,90].
The epigenome appears more susceptible to environmental
factors during periods of extensive epigenetic reprogramming in
early life, particularly during the prenatal, neonatal and pubertal
periods, when the epigenome is being established and environ-
mental insults may interfere with processes that regulates its
reprogramming. However, somatic changes to epigenetic marks
might also ensue from environmental exposures in adults, as it has
been observed in aging and numerous disease processes (i.e. can-
cer, neurodegenerative and metabolic diseases among others)
[34,89,90].
Furthermore, numerous environmental factors, from nutrition
to toxicants, have been shown to induce an epigenetic trans-
generational inheritance [91], which is described as the germline
transmission of epigenetic information between generations
without direct exposure [92]. Several different model of epigenetic
inheritance of disease and phenotypic variation has been proposed
to be linked to environmental exposure. Drake and Lui [93e95]
outline three possible mechanisms that could be responsible of
multigenerational observations: a) persistent environmental ex-
posures (i.e. generation after generation) during early develop-
ment; b) a single maternal environmentexposure that can yet
induce a multigenerational phenotype; and c) epigenetic effects
which can be transmitted across the germline. Transgenerational
epigenetic inheritance induced by environmental agents has been
mainly studied performing exposure to environmental insult dur-
ing pregnancy, which can affect a mother (F0 generation), the
developing fetus (F1 generation) but also the fetus germ cells which
will go on to form the F2 generation. Nutrition [96], temperature
[97], stress [91], and toxicants [91] can all induce epigenetic
transgenerational inheritance of phenotypic variation [98]. This
evidence has been demonstrated in plants, sh, insects, pigs,
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171160
rodents, and humans [91]. The altered transgenerational pheno-
types have been observed for generations in mammals [91], and for
hundreds of generations in plants [99]. The capacity of environ-
ment to modify phenotype and phenotypic variation through
epigenetic mechanism is suggested to be important for evolution.
Environmentally induced epigenetic inheritance can bring a pop-
ulation closer to an increased tness in a faster way than genetic
changes; then, genetic variations may ultimately follow (if the new
environment is stable), likewise the genetic xation of initially
induced phenotypes occurs. Furthermore, the selection can act
upon randomly induced metastable epialleles, thus contributing to
adaptation in a similar way than genetics [10 0].
Indeed, environmental epigenetics and epigenetic trans-
generational inheritance represent the molecular mechanism able
to support the neo-Lamarckian theory, which assert that environ-
mental factors directly alter phenotypes. Although aspects of the
original Lamarckian evolution theory, such as having directed
phenotypes within a generation, were not accurate [101e103], the
notion that environment can impact phenotype is reinforced by
environmental and transgenerational epigenetic studies. These
ndings do not contrast the Darwinian theories, but rather overlap
with them, suggesting that a new integrated theory should be hy-
pothesized. Specically, the well-established aspect of Darwinian
evolution is the ability of environment through natural selection to
act on phenotypic variation, with genetic mutations and variation
considered the main molecular mechanism involved in generating
the phenotypic variation. Nevertheless, being the environment able
to impact epigenetic programming through generations, environ-
mentally induced epigenetic changes can be considered as another
source of phenotypic variation. Guerrero-Bosagna and colleagues
reports an example of how developmental effects of environmental
exposures can inuence adult characters in mammals also poten-
tially having evolutionary consequences [104]. They demonstrated
that a high consumption of isoavones can alter both epigenetic
and morphometric characters or sexual maturation, which are
characters that might play relevant roles from an evolutionary
perspective. All in all, the unied evolutionary theory sustains that
both environmental epigenetics (impacting on phenotypic varia-
tion) and the capacity of environment to intercede in natural se-
lection will be equally important for evolution [105]. Another
relevant aspect is the ability of epigenetic processes to endorse
genetic mutations [106], in particular CG to TG transitions [107];
thus, environmental epigenetics might not merely provide
increased phenotypic variation, but could drive genetic change and
directly increase genotypic variation likewise.
Being both genetic and environment pivotal phenotypic de-
terminants, genetic X epigenetic X environmental interactions has
to be taken into account [108], in order to carefully dene intricate
biological interactions and, as ultimate goal, ascertain susceptible
subpopulations. Beyond implication of evolutionary prospectives, it
is intuitive that epigenetics has considerable potential for identi-
fying new biomarkers to predict which exposures would increase
the risk in exposed subjects and which individuals are particularly
vulnerable to develop disease.
1.7. The role of environment as a strong determinant of health
There has been a growing awareness of environmental effects on
human health, and that neither purely environmental factors, nor
purely genetic factors can entirelyexplain the observed estimates of
disease incidence and progression. Furthermore, the balance be-
tween genetic and epigenetic contributions in the development of
pathologies appears to change during life. While, for instance, the
majority of childhood tumors are connected to an inherited genetic
or epigenetic (for example, imprinted) problem, this equilibrium
shifts in favor of acquired epigenetic and genetic burden in tumors in
adult or elderly age [109]. Many epidemiologic studies investigated
the effects of exposure to chemical, social or physical factors in
relation to several pathologies, such as cardiovascular disease, dia-
betes and canceramong others. These kind of studies are starting to
incorporate gene-environment interactions and epigenetic modi-
cations to better investigate the multidisciplinary nature of indi-
vidual, in order to have a better estimate of the complexity of
exposure biology and the smalleffects that are easily disturbed[110].
Numerous environmental factors have been suggested to be able
to inuence the epigenome, resulting in long-term changes in gene
expression and metabolism: air pollution, tobacco smoke, oxidative
stress, organic chemicals, endocrine disruptors, metals and, last but
not least, nutrient intake and social environments [34,109]. The
totality of our exposures from conception onward has been dened
by Christopher Wild with the new coined term exposome. Expo-
sures come from our external environment and lifestyle, but are also
the outcome of our internal biological processes and metabolism;
given this more complete view of the exposome, the concept was
redened by Miller and Jones as the cumulative measure of envi-
ronmental inuences and associated biological responses
throughout the lifespan.Current scientic opinion sustains that the
study of the exposome could be helpful to clarify the interaction
between genetic and environmental factors that contribute to dis-
ease, with the potential to revolutionize biomedical science, espe-
cially in term of prevention of late onset chronic disease that
represent the main burden in the modern society [111].
Several different models have been hypothesized to explain the
role of epigenetics on the late onset disease. Barker hypothesized
that adult diseases are consequences of fetal adverse conditions
due to the fetus adaptation to a certain environment to which it was
exposed in early life [112 ]. Adaptive responses, which can be either
in the form of metabolic changes or sensitivity of the target organs
to hormones, will not induce immediate consequences in the
newborn but could lead to physiologic and metabolic disturbances
in later life. Gluckman and Hanson suggested that fetal exposure to
adverse conditions makes immediate changes which are reversible,
except in the case that stress conditions persist [113 ]; in that case
fetus undergoes to irreversible changes that will persist throughout
life, inuencing (positively or negatively) the adulthood [114]. They
coined term predictive adaptive response (PAR) for the phenome-
non. Another hypothesis is represented by the DOHaD (Develop-
mental origin of health and disease) model, which postulate that
not merely embryonic development but also the period of devel-
opment during infancy is responsible for late life risk of diseases.
Another theory, which represents the evolution of the previously
listed, is the LEARn (Latent early life associated regulation) model.
This concept sustains that environmental agents such as nutrition,
metal exposure, head trauma and lifestyle are hitsthat are related
to the cause and progression of common late onset diseases. LEARn
is based on the idea that latent epigenetic changes induced in early
life do not result in any disease symptom immediately, but create a
perturbation in the genome. It is just later in life, after a latency
period (which nish when a second triggering agent manifest), that
the epigenetic perturbation will result in manifested consequences.
Genes that respond late in relation to early life responses are called
LEARned genes, while others which don't are called unLEARNed.
The responses to the early life environmental triggers after the la-
tency period is dened as LEARning [115 ].
2. Nutrigenetics
2.1. Introduction to nutrigenetics
The notion that interactions between genetics and nutrition are
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171 161
responsible for the nal phenotype was recognized by Archibald E.
Garrod in 1902, when he published in The Lancet a milestone paper
in which he depicted his observations of people with black urine or
black bone disease, also known as alkaptonuria (AKU) [116]. AKU is
a rare disorder of autosomal inheritance. It is caused by a mutation
in the homogentisate 1,2 dioxygenase gene, resulting in the accu-
mulation of homogentisic acid. It was one of the rst disorders
found to conform with the principles of Mendelian recessive in-
heritance in humans, and was primarily described as an example of
genetic disruption of food metabolism. The increasing in
biochemical knowledge gradually started to support fruitful
nutritional intervention for handling some of these metabolic pa-
thologies. In 1934, Asbjørn Følling discovered that another defec-
tive metabolism of a dietary amino acid (phenylalanine) could
induce severe mental deciency in subjects affected by a metabolic
defect called phenylketonuria (PKU). Later, in 1953, Horst Bickel
demonstrated that nutritional treatment can be effective in treating
this condition, helping to prevent devastating consequences just
starting a specic nutritional treatment few days after birth. The
same happened with other untreatable inherited diseases (maple
syrup urine disease, biotinidase deciency and others), for which
early nutritional intervention resulted to be effective [117].
In 1960, Dr JA Roper explained the links between genetics and
nutrition with a paper entitled Genetic determination of nutri-
tional requirements[118]. There was quite slight progress in un-
derstanding interactions between genotypes and nutrition in
humans until the Human Genome Project was completed; few time
later, nutrigenomics, i.e. the study of the gene-nutrients in-
teractions, was predicted would be the future of nutrition [119 ,120].
Nutritional genomics (or nutrigenomics) has been described as the
branch of science investigating all types of interactions between
nutrition and the genome by high-throughput genomic tools [121].
Nutritional genetics (or nutrigenetics) is described as a sub-set of
nutrigenomics, which aims to understand how genomic variants
interact with dietary factors and which implications derive from
such interactions (Fig. 1 A).
2.2. The role of genetic variants in nutrition
Nutrigenetics examines inherited differences in nutrient meta-
bolism and investigates how to use individual genetic information
to tailor better nutrition plans [117 ]. Several different types of
genomic structural variation emerged from the investigation of
human genome [122]. SNP, inverted gene sequences, gene deletion,
segmental duplication and CNV has been almost all associated to
some nutritional-related phenotype or showed to be able to modify
individual's response to diet.
While the simpleMendelian genetics is responsible for inborn
errors of metabolism such as alkaptonuria or phenylketonuria,
multifactorial diseases, such as diet-related diseases and obesity,
are seldom due to single genetic variants. For example, at least
ninety-seven variants resulted to be associated with body fatness,
and together these explain <3% of the variance in BMI [123,124].
Numerous pathways affecting the central nervous system (i.e.
satiety regulations or food intake) or metabolic features, such as
lipid metabolism and adipogenesis, are regulated by the involved
genes. Additionally, genetic variants involved in various cell
biology, cell signaling and in RNA binding processing has been
related with adiposity risk as well [123]. This is just an example
intended to underline the complexity of nutrigenetics, which aim
to study complex polygenic traits, related to numerous different
physiological pathways.
Fig. 1. Graphical representation of interactions between diet and the genome. (A) Nutrigenetics: genetic polymorphisms can induce differential gene expression. As a result, different
metabotypes, which show different responses to diet, different nutrient requirement and potential food intolerance, exist. Of note, the location of the SNPs can also affect epigenetic
modications. (B-C-D) Nutrigenomics: methyl donors availability, bioactivity of dietary compound and xenobiotics (B) can affect the one-carbon cycle and other pathways thus,
consequentially, affect DNA methylation and histone modications (C). Not just parental molecules (B) but also derived compounds and metabolic products of microbial activity (D)
can affect these pathways (C).
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171162
Genetic variants able to modulate the effects of certain dietary
factors or to affect food preferences can be investigated through
different experimental approaches. The candidate gene approach is
based on the selection of a gene because of its putative function or
for other specic knowledges about it. In dependence on the
number of SNPs in the gene and their potential functional effects,
assessments can be conducted using single SNPs or combinations of
them, such as haplotypes. More recently, genome-wide approach
started to be applied in modern studies. This method is based on
the identication of previously unknown genetic variants which
can modify response to diet scanning the entire genome. Whether a
candidate gene approach is preferential if few genetic variance
(selected a priori following a certain hypothesis) want to be tested
with a high power, GWAS has the advantage to be hypothesis-free
and can be useful for exploratory analysis, testing large number of
variants at the same time. At the same time, very large populations
are required for GWA studies to have a good power. Other strate-
gies, such as meta-analysis of GWAS or calculation of genetic risk
score, can be helpful to increase the analyzed population or take
into account different genetic model, respectively.
Recognizing relevant diet-gene interactions will not only be
useful for individual personalized dietary advices, but will improve
public health recommendations (supported by scientic evidences
connecting specic dietary compounds to different health out-
comes) and scientic research as well. In fact, studying people re-
sponses to a nutrient assuming that they are all metabolically
similar, often results in the identication responders and non-
responders to the intervention, and this observed variation in the
outcome is frequently attributed to weaknesses in the scientic
design of the study, making obtained results difcult to publish ad
difcult to use to improve human health. Thus, the usage in study
projecting of modern genetic methods, through which is possible to
predict who are the responders, could strongly help to improve
results and optimize resources [117].
2.3. Genetic determinants of responsiveness to dietary interventions
Diet represents a key modiable risk factor for human health.
However, benets we currently gain are signicantly reduced
comparing to the full potential of its protective effects. Several
reasons can be at the basis of this phenomenon, including indi-
vidual variability in response to certain nutritional regimen. Taking
into account that genetic variants can create metabolic in-
efciencies, it is reasonable to hypostesize that such SNPs can in-
uence dietary requirements. Thus, it is almost evident that taking
into account individual responses is essential to gain the full benet
of dietary regimes [125].
There are considerable evidences that inter-individual variation
in response to dietary interventions can inuence benecial effect
that certain individuals or population subgroups can obtain, more
than others, from a diet, in dependence on their genotype,
phenotype, and environment [126]. Despite that, dietary reference
values, which are designed for the general population assuming a
unique Gaussian distribution, are not optimized for genetic sub-
groups, which may signicantly differ for noteworthy metabolic
aspects (such as for example the activity of metabolic enzyme
requiring micronutrients as cofactors and/or micronutrient trans-
port proteins) because of their genetics [126]. Considering that the
one-size-ts-allapproach applied in nutrition until now resulted
to be unsuccessful, the identication of genetic variants, that
recognize responsive and non-responsive individuals for specic
dietetic intervention, represents one of the most challenging and
potentially useful goal of nutrition research. If the issue is relatively
easy for monogenic characters (such as the genetic determinant of
lactose intolerance), the landscape becomes more intricate for
complex polygenic traits, such as predisposition to hypertension or
diabetes. Despite consistent efforts, it is still to be cleared how to
modulate disease development through consumption of a complex
diet based on different genotypes [125].
2.4. Nutrigenetic tests for personalized nutrition
Precision nutrition can occur at three levels: (1) conventional
nutrition, following general guidelines for population groups by
age, gender and social determinants; (2) individualized nutrition,
that take into account also phenotypic information about current
nutritional status of the subject (such as, among others,
biochemical and metabolic analysis, anthropometry and physical
activity), and (3) genotype-directed nutrition, taking into consid-
eration rare or common gene variation which determine different
responses to certain nutritional plans [127]. The use of genotypic
information in tailoring personalized dietary advice has been a
major objective since the beginning of the modern nutrigenomics
era [128]. Several benecial effects of providing personalized
nutritional advices, such as supporting disease prevention,
reducing health care costs and improving motivation to change,
has been observed [129,130]. Besides, recent randomized control
trials showed that genotype-based personalized dietary advices
were better understood and increased the adherence to the
nutritional plan than general dietary advice [130,131]. This result is
not irrelevant, considering that compliance and diet adherence has
been identied as one of the most effective parameters in nutri-
tional intervention success [132].
Relevant ndings concerning this aspect come from the EU-
funded Food4Me project [133]. It is a multi-center study aimed
to investigate if fully internet delivered personalized nutrition
advice (according to individual phenotype and genotype) could
affect people's lifestyle. Promising data from the Food4Me Euro-
pean randomized controlled trial involving 683 participants shows
greater body weight and weight circumference reductions in risk
carriers than in non-risk carriers of the fat mass and obesity-
associated (FTO) gene, when participants were informed to be
carriers of the FTO risk allele [134]. Further investigations from the
Food4Me study showed that adherence to specic healthy regi-
mens, such as the Mediterranean diet, can have benecial effects
on anthropometric parameters overcoming an adverse genetic
load [135 ]. Nevertheless, San-Cristobal and colleagues demon-
strated that a higher genetic risk score (calculated by several ge-
netic variants related to metabolic risk features) may reduce
benets on total cholesterol levels and inuences the levels of
plasma carotenoids, indeed suggesting that gene nutrient in-
teractions might contribute to the implementation of practical
accurate nutrigenetic advice. However, despite promising evi-
dences, no univocal demonstration that including phenotypic plus
genotypic information can improve the effectiveness of the
personalized nutritional advice can be inferred from this big study
[136,137].
All in all, nutrigenetics is still involved in an extensive discus-
sion about personal genetics, which started in 2001 with the
presentation of Sciona Ltd. (in the United Kingdom), and persisted
with the subsequent launch of popular companies such as
23andMe, Navigenics and deCode in the following years [13 8]. The
principal query, indeed, concerns the clinical utility of nutritional
genetic tests: can the evidences coming from nutrigenetic studies
be translated into helpful dietary recommendation which would
not be accessible without the use of genetic information? There is
emerging consensus on the idea that each subject's health is
established by interactions between his or her xed genotype and
nutrition (among other environmental exposures), in addition to
the effects of stochastic events, as hypothesized in the ‘‘health
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171 163
pendulum’’ theory. Nevertheless, current knowledge in this area is
fragmentary, and a limited number of dietegeneehealth associa-
tions have been tested for causality in intervention studies on
humans [128]. Filling these gaps will require larger, even better-
designed randomized controlled trials. At the same time, it is
also true that most of nutritional recommendation come from
observational and epidemiological studies. Thus, it is debated why
the level of evidence for genetically inuenced nutritional advice
are not evaluated according to the same standards used for
traditional nutritional recommendations [138]. Gorman and col-
laborators also discuss risks and benets of precautionary princi-
ple, thus ignoring nutrigenetics. For certain nutrigenetic
evidences, such as those regarding methylenetetrahydrofolate
reductase (MTHFR) gene C677T polymorphism, folic acid, and
homocysteine, it is to choose that chronically high homocysteine is
potentially less dangerous than increasing daily folic acid intake, in
individuals who don't benet of the standard recommended
intake because carriers of the TT genotype. Whether ignore new
scientic evidences can sometimes be synonymous of applying the
precautionary approach, it is probably not always the case of
nutrigenetics. Wildavsky [13 9] claims that in the case of lack of
knowledge, small-risk taking, followed by stepwise evaluation, is a
safer course than avoiding risk. Cautious estimation of the balance
between benets and risks, together with a step by steps approach
able to progressively increase the benets and diminish the risks,
would be probably the best way to approach nutrigenetics. This
means that more and more research on this promising eld is
required. The inconsistent results produced by the candidate-gene
association studies are actually not associated to the research
quality in general, but rather to the complexity of nutritional ef-
fects in the long term. Technological improvements and the
increasing usage of genotype analysis in randomized control trials
suggest a promising increase of knowledge in this eld in the next
years.
Thus, it is clear that nutrigenetics is not a science with easy
answers and it doesn't rely on a standard dietary recommendation
to each genotype. Nutritional factors interactions with the genome
are very complex and need competent nutrition professionals who
can guide patients successfully through this complex landscape
[117]. Furthermore, from a dietetic point of view, there is often not
just one simply solution for a specic problem. On the contrary
many solutions can be designed by dieticians, physician and
nutritionist, depending by other characteristics of each analyzed
subject beyond its genetics. In fact, personalized nutrition is based,
per denition, on the knowledge and integration of the genetic
background with biological and cultural variations, such as food
preferences, intolerances and allergies. This means that the genetic
prole alone is usually not sufcient to provide a personalized di-
etetic plan, while it has to be integrated by expert professionals
with patient anamnesis, anthropometry, food preferences and life-
style. For this reason, another open debate is currently centered on
the legitimacy of direct-to-consumer (DTC) tests [140], which, in a
certain way, bypasses this multifaceted approach, avoiding the
mediation of professionals able to provide a correct interpretation
and usage of genetic data.
Concluding, to understand what can be legitimately used, a
deep knowledge of the topic is essential. In addition, personalized
nutrition needs to be kept in its proper context, that not overlaps
with clinical genetics, disease treatment, or disease prediction. In
fact, nutrigenetics uses genetic information in a different way
than classical genetics; it does not estimate disease risk based on
association studies but provides exact information based on
specic interactions between gene and diet, to identify subgroups
which could maximize the benet of different nutritional
interventions.
3. Nutrigenomics
3.1. Introduction to nutrigenomics
Nutrition research has gone through a relevant shift in the past
decade, from focusing on physiology and epidemiology to
biochemistry, genetics and molecular biology. Micronutrients and
macronutrients have been clearly recognized as powerful dietary
signals able to affect metabolic programming of cells, with a central
role in the control of body homeostasis [141]. These evidences
make the scientic community to realize that it is not possible to
really understand the impact of nutrition on health and disease
without a deep knowledge of molecular effects of nutrients.
Nutrigenomics, which also includes the study of genes that inu-
ence different predisposition to nutrition-related impairment
(hence nutrigenetics), attempts to study in a broad way the
genome-wide inuences of nutrition, with the major goal to apply
this knowledge to prevent diet-related diseases.
3.2. Nutritional factors that can inuence the epigenome and the
mechanisms involved
In some ways, nutrigenomics can resemble to pharmacoge-
nomics [142]. However, an important difference between these two
disciplines is that pharmacogenomics concerns with the effects on
the genome of drugs, which are pure compounds, given in exact
doses, while nutrigenomics has to take into account the complexity
and variability of nutrition. This concept is just a tip to have an idea
of the complexity of this research eld.
The study of gene expression patterns, protein expression and
production of metabolites in response to certain nutrients have
been the main object of nutrigenomic studies at the beginning of
this new science. From the point of view of nutrigenomics, nutri-
ents are dietary signals which are perceived by the cellular sensor
systems, and which are able to affect gene and protein expression
and, consequently, metabolite production [143].
Recently, among the wide spectrum of activities for which many
nutrients are known in their role on prevention and mitigation of
various diseases, epigenetic effects acquired an emerging impor-
tance. This specic research area, which describes effects of nutri-
ents on human health through epigenetic modications, has been
referred as nutritional epigenomics, or nutriepigenomics[144]
(Fig. 1B and C).
Whether several studies demonstrated that numerous nutrients
and bioactive compounds inuence different pathways through
which epigenetics affects gene expression, there are still relatively
few information about the precise mechanisms through which
nutrients modulate epigenetics. Different ways through which
what we eat can inuence the expression of our gene through
epigenetics have been suggested. These multiple mechanisms are
mutually compatible and may operate together in time, enriching
the complexity of this regulative pathway [144 ,145]. They can be
clustered in three main groups: 1) food provides substrates
necessary for proper methylation of DNA and histones, cofactors
that modulate enzymatic activity of DNA methyltransferases and
can regulate activity of the enzymes involved in the one-carbon
cycle; 2) bioactive molecules contained in food can directly or
indirectly interact with the epigenome, as well as 3) toxicant con-
tained in food also can (Fig. 1 B).
Most of the understanding concerning the ability of nutritional
factors to modulate gene expression by epigenetic mechanisms
refers to the one-carbon metabolism, a complex network of inter-
related biochemical reactions in which methyl donor nutrients
provide one-carbon units to different biochemical and molecular
reactions. This step is essential for several molecular pathways
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171164
including DNA synthesis, purine synthesis, methylation of DNA,
RNA, protein, phospholipids and small molecules [146]. Nutrients
are processed through the folate cycle and the methionine cycle,
serving as methyl sources for the universal methyl donor, S-ade-
nosylmethionine (SAM). A methyl group from SAM can be enzy-
matically transferred to other molecules (i.e. specic cytosines in
the DNA), thus generating S-adenosylhomocysteine (SAH) (which
acts as an inhibitor of methyltransferases themselves) as an end
product. For this reason, nutrients affecting one of the two main
metabolites of the one-carbon metabolism (i.e. SAM or SAH) can
potentially alter the methylation of DNA and histones. DNA
methylation can be affected by four different type of nutrients: 1)
dietary methyl donor nutrients (methionine, choline, betaine,
serine); 2) B vitamins (B12, B6, B2, B9) as coenzymes of one-carbon
metabolism (with folate acting as acceptor or donor of methyl
groups), 3) micronutrients which can affect one-carbon meta-
bolism (zinc, retinoic acid, selenium) and 4) bioactive food com-
pounds that can modulate DNA methyltransferasesactivity [81].
Not just specic nutrients play a role in nutrigenomics, but also
bioactive molecules, such as secondary plant metabolites, can
modulate gene expression. Epigallocatechin-3-gallate, genistein,
equol, myricetin, but also butyrate, sulforaphane and curcumin
have been showed to be epigenetically active [144,147], not just
regulating DNMTs functions, but also acting as chromatin remod-
elers through modulation of histone deacetylases (HDAC).
Interestingly, it must be noticed that epigenetic mechanisms are
strongly associated to cellular oxidative stress homeostasis [14 8],
and, nally, they are not just involved in nuclear gene expression
regulation, but also strictly involved in mitochondrial functions
regulation too [149]. These observations further increase the
number of potential indirect effects exerted by nutrigenomics on
health.
Another aspect to consider is that molecules contained in the
food we eat can affect and be affected by the gut microbiome. The
production of metabolites acting as allosteric regulators and critical
cofactors of epigenetic processes, is one of the major mechanisms
linking gut microbiota and control of gene expression. Indeed, the
gut microbiota produce numerous low weight bioactive molecules
which can play a role in epigenetic processes, i.e. folate, butyrate,
biotin, and acetate. In addition, the absorption and excretion of
minerals such as zinc, selenium, iodine, cobalt (indeed, cofactors of
enzymes participating in epigenetic processes) is inuenced by the
microbiota, which can also metabolize bioactive compounds con-
tained in food (i.e. ellagic acid and ellagitannins are metabolized in
urolithins) inuencing their bioavailability [150,151](Fig. 1 D).
Moreover not just natural food components but also several
classes of pesticides (including persistent organic pollutants,
arsenic, endocrine disruptors, several herbicides and insecticides)
have been shown to modify epigenetic marks [152e154].
Numerous investigations studied the effects of environmental ex-
posures on epigenetic markers, identifying many toxicants able to
modify epigenetic states (in particular in terms of DNA methylation
and histone modications) similarly to what happens in some
pathological conditions [152,155e157]. Additional investigations
are necessary to clarify if epigenetics can act as a causal link be-
tween exposure to pesticide and health outcomes, or rather be a
sensitive early biomarker of exposure.
3.3. Susceptible period of exposure, epigenetic reprogramming and
transgenerational effects in nutrigenomics
The mechanisms previously described provide convincing evi-
dence that epigenetic marks serve as a memory of exposure to
environmental factors and, among others, inadequate or inappro-
priate nutritional factors. These environmental stimuli can have
different impact depending on the period of life of the exposed
organisms. Considering the epigenetic plasticity of growing and
developing tissue, exposures during early life represent a critical
period [158e160 ]. Not just pre-natal and intrauterine periods, but
also post-natal early life and periods of epigenetic remodeling
characterized by rapid physiological changes (such as puberty and
aging) represent susceptible period of exposure [147,161].
Several examples of late onset disease have been found to take
their origins in early life period, or at least to be inuenced by
episodes occurring in the rst stages of life. Furthermore, in addi-
tion to prenatal and postnatal nutritional effects, which can result
in stable changes and predispose individuals to disease later in life
(which is referred as early life programming), transgenerational
mechanisms must be considered. Transgenerational epigenetic
inheritance can result from several different environmental expo-
sures, even though little is known about the mechanism undergone
to the maintenance of the epigenetic marks suggested to be
involved in this phenomenon. It has been established that factors
like maternal diabetes, behavioral programming (maternal care),
nutritional interventions (carbohydrate-rich or fat-rich diet or
caloric restriction), glucocorticoids and exercise, endocrine dis-
ruptors, stress during gestation and lactation may all cause
imprinting in the following generations [144].
One of the most cited and studied example that clearly shows
the role of nutrigenomics is the Agouti mouse, where coat color
variation and healthy/unhealthy phenotype is established early in
development according to maternal diet [162,163]. Another
example is represented by protein malnutrition in pregnant mice
which resulted to determine signicant gene expression changes,
miRNA changes, and different DNA methylation patterns in brains
of the offspring [164]. Studies on humans that corroborate trans-
generational inheritance also exists. One of the rst example is the
the Dutch famine study, which showed that starvation in one
generation affects the risk for glucose intolerance and metabolic
disorders in its offspring [165]. Another pivotal study has been
conducted on the
Overkalix population, where overeating by
paternal grandfather or father induced increased risk for cardio-
vascular diseases or diabetes in grandsons, while a reduced food
availability during father adolescence exerted an opposite effect in
the offspring [166 ].
Despite better controlled studies in humans are needed, a hy-
pothesis which could powerfully impact our lives is emerging:
what do we eat, is not just important for us, but may affect future
generationshealth as well. Moreover, given that thousands of
nutrients and other compounds are contained in food, but only few
of these have been tested for transgenerational epigenetic effects,
further research in this eld is essential in order to promote public
health and set sensible public policy.
3.4. Interactions between nutrigenetics and nutrigenomics
Even if based on different scientic approach, nutrigenetics and
nutrigenomics cannot be considered separately. In fact, if it is true
that certain dietary molecules can potentially modify cellular ho-
meostasis, it is also true that the alteration of the homeostatic
mechanisms especially occurs in individuals with susceptible ge-
notypes [143]. Indeed, not only nutrients, but also the genetic
make-up can surely impact one-carbon metabolism (Fig. 1 A).
Among different combinations of nutrients and genes, folate and
the MTHFR 677 C to T SNP represents a peculiar example of
nutrient gene interactions affecting DNA methylation. In partic-
ular, carriers of the MTHFR 677TT genotype display a reduced
availability of 5-methyl tetrahydrofolate and a consequent higher
folate requirement for the regulation of plasma homocysteine
concentrations. Interestingly, researchers showed that not all
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171 165
MTHFR 677 TT carriers had impaired global DNA methylation
levels, but just those who were decient for folate, suggesting that
MTHFR C677TT SNP affects genomic DNA methylation status
through an interaction with folate status [167].
Similar conditions can occur for other metabolites of the one-
carbon cycle, such as for choline, a methyl donor necessary for
the conversion of homocysteine to methionine [168 ]. Excluding
diet, the only other source of choline is the de novo biosynthesis of
phosphatidylcholine (can be converted to choline) catalyzed by
phosphatidylethanolamine-N-methyltransferase (PEMT) in liver.
Considering that the consume of foods containing choline are often
discouraged because rich in fat and cholesterol (e.g. eggs and liver),
it has been measured that only a small percent of the population
achieves the recommended adequate intake for choline, in partic-
ular man, post-menopausal women and the 44% of pre-
menopausal women. The risk of choline deciency is reduced in
young woman because PEMT is estrogen-inducible; however, it is
interesting that those women that are more prone to choline
deciency (even at youngest age) have a SNP in PEMT
(rs12325817); similarly women with the rs2236225 SNP in the
gene MTHFD1 (5,10-methylenetetrahydrofolate dehydrogenase 1)
are 15 times more predisposed to develop signs of choline de-
ciency in case of low-choline diet respect to wild types [169]. This
example shows how dietetic intake of choline can be particularly
important for a certain subpopulation (pre-menopausal women
carriers of the susceptible gene alleles), not just simply counter-
acting a specic metabolic deciency, but also protecting from
impairment of epigenetic regulation processes.
Another different interesting example of interaction between
genetic and epigenetics is represented by the lactase persistence. To
date, inter-individual differences in lactase expression in human
adults have been ascribed merely to DNA sequence variation up-
stream of lactase (LCT) gene. Specically, the rs4988235 SNP (C/T-
13910) has been related to the phenotypes of lactase persistence
and non-persistence in European populations [17 0]. Nevertheless,
it is interesting to notice that in non-Europeans, this SNP does not
fully explain lactase persistence, with certain African individuals
exhibiting lactase in the absence of LCT-associated variants.
Furthermore, the molecular mechanism able to explain the age-
dependent changes of LCT expression (that varies from very high
levels in infancy to signicant downregulation in most of adults) is
also unclear. Considering that DNA sequence is steady, more dy-
namic regulatory systems must be dragged in the temporal varia-
tion of lactase non-persistence. Interestingly, recent studies
showed that lactase non-persistence derives from accumulation of
transcriptionally suppressive epigenetic changes on the SNP C-
13910 carriers, while T-13910 carriers escape from epigenetic
inactivation facilitating lactase persistence [171 ].
3.5. Personalized epigenomics
Improvement in personalized epigenetics for the therapy and
management of several specic pathologies is quickly conducting
to an important increase of the tools accessible to clinicians in
preventing and controlling diseases that have an epigenetic base in
their etiology and pathogenesis. An case in point is represented by
chronic pain management, for which an important role of epige-
netics has been highlighted. In this case, assessing the epigenomic
marks in subjects suffering from chronic pain could have sub-
stantial utility in the selection of adequate analgesics which can
give relief to patients affected by chronic pain [172]. Several genes
which are under epigenetic control can also affect the onset of
obesity and related metabolic diseases (for instance diabetes) [173].
Dietary factors are well known to produce epigenetic changes, with
a certain inter-individual variability of the effects. Concerning
obesity, both the quality and the quantity of diet have been
demonstrated to modulate the epigenetic signature of individuals
inducing epigenetic irregularities that could be managed by
personalized therapy [174]. Thus, taking into account personalized
epigenetic approaches would represent a greatimprovement in the
efcacy of obesity management. Furthermore, personalized epi-
genetics can be useful also in prevention, considering that envi-
ronmental factors have central roles in the development of obesity,
and epigenetic modications can be reversible through changes in
environmental factors (life-style in particular) that lead to such a
disorder [174].
Even if this prospective is quite far from immediate practical
application, interesting evidences about therapeutic applications of
epigenetically active nutrients are available [147]. In fact, as
epigenetic modications are reversible and tissue-specic, a regu-
lation of these processes through diet or specic nutrients could
also help diseases prevention and health maintenance. Some of the
natural products which showed positive outcomes on particular
human diseases are also being studied in clinical trials. Surrogate
endpoints associated with metabolic syndrome resulted to be
improved by genistein indirectly reducing the risk of developing
diabetes and cardiovascular disease [175 ]; similarly, a reduction of
type II diabetes onset has been observed in pre-diabetic individuals
supplemented with curcumin [176]. Genistein, curcumin, epi-
gallocatechin-3-gallate and resveratrol are some of the phyto-
chemicals that have been demonstrated to trigger the anti-
inammatory machinery and improve some of the symptoms
associated with metabolic syndrome [177]. These are just few ex-
amples of potential epigenetically active nutrients and their
benecial effect that has been hypothesized to be exerted through
epigenetic processes [147].
Furthermore, nutrigenomics strongly improves current knowl-
edge in nutrition providing, through the usage of metabolomic and
epigenetic approaches, novel biomarkers of food intake and dietary
patterns which will lead to more objective and robust measures of
dietary exposure [121]. For all these reasons, it is clear that appli-
cation of nutrigenomics research can offer considerable potential to
improve public health [178 ].
4. Conclusions
Nutrition is one of the most important life-long environmental
factors able to impact human wellbeing. Among the mechanisms
involved, genome nutrient interactions have been denitely
demonstrated to play an important role in health maintenance and
disease prevention. The disciplines of nutrigenetics and nutrige-
nomics aim to address how genetics and epigenetics can explain
individual dietary susceptibility and to understand how human
variability in preferences, requirements and responses to diet can
be implemented in a personalized nutrition. Despite a growing
interest of the scientic community for these topics, the body of
research is still to be enlarged to make the actual knowledge able to
provide personalized advices tailored by nutrigenetics and nutri-
genomics [179 ]. Given the high potential of these disciplines,
research on nutrigenetics and nutrigenomics should be promoted
and divulged to a wide-reaching audience [180,181 ], in order to
make both professionals and the general population aware of the
profound effects of nutrition on our health.
Author contributions
L.B. wrote the article, R.G. supervised, revised and proofread the
paper. All the authors approved the nal version of the manuscript.
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171166
Declaration of interest
Material submitted is original, all authors are in agreement to
have the article published.
Conict of interest
Authors have neither founding sources nor competing interests
to declare.
Acknowledgements
We thank the artist Irene Saluzzi who give an important
contribution in the drawing of the Fig. 1, helping us to make the
complexity of nutrigenetics and nutrigenomics easier to be un-
derstood and communicated.
This research did not receive any specic grant from funding
agencies in the public, commercial, or not-for-prot sectors.
References
[1] M.J. Meaney, Nature, nurture, and the disunity of knowledge, Ann. N. Y. Acad.
Sci. 935 (2001) 50e61, https://doi.org/10.1111/j.1749-6632.2001.tb03470.x.
[2] R. Lewontin, The Genetics of Human Diversity, Free. Press, New York, 1980.
[3] R. Sachidanandam, D. Weissman, S.C. Schmidt, J.M. Kakol, L.D. Stein,
G. Marth, S. Sherry, J.C. Mullikin, B.J. Mortimore, D.L. Willey, S.E. Hunt,
C.G. Cole, P.C. Coggill, C.M. Rice, Z. Ning, J. Rogers, D.R. Bentley, P.Y. Kwok,
E.R. Mardis, R.T. Yeh, B. Schultz, L. Cook, R. Davenport, M. Dante, L. Fulton,
L. Hillier, R.H. Waterston, J.D. McPherson, B. Gilman, S. Schaffner, W.J. Van
Etten, D. Reich, J. Higgins, M.J. Daly, B. Blumenstiel, J. Baldwin, N. Stange-
Thomann, M.C. Zody, L. Linton, E.S. Lander, D. Altshuler, A map of human
genome sequence variation containing 1.42 million single nucleotide poly-
morphisms, Nature 409 (2001) 928e933, https://doi.org/10.1038/35057149.
[4] R. Mourad, Probabilistic Graphical Models for Genetics, Genomics, and
Postgenomics, 2014.
[5] J.G.R. Cardoso, M.R. Andersen, M.J. Herrgard, N. Sonnenschein, Analysis of
genetic variation and potential applications in genome-scale metabolic
modeling, Front. Bioeng. Biotechnol. 3 (2015) 13, https://doi.org/10.3389/
fbioe.2015.00013.
[6] L. Feuk, A.R. Carson, S.W. Scherer, Structural variation in the human genome,
Nat. Rev. Genet. 7 (2006) 85e97, https://doi.org/10.1038/nrg1767.
[7] F. Zhang, J.R. Lupski, Non-coding genetic variants in human disease, Hum.
Mol. Genet. 24 (2015) R102eR110, https://doi.org/10.1093/hmg/ddv259.
[8] The International HapMap Consortium, A haplotype map of the human
genome, Nature 437 (2005) 1299e1320. https://doi.org/10.1038/
nature04226.
[9] The International HapMap Consortium, A second generation human haplo-
type map of over 3.1 million SNPs, Nature 449 (2007) 851e861. https://doi.
org/10.1038/nature06258.
[10] The 1000 Genomes Project Consortium, A map of human genome variation
from population-scale sequencing, Nature 467 (2010) 1061e1073. https://
doi.org/10.1038/nature09534.
[11] The 1000 Genomes Project Consortium, an integrated map of genetic vari-
ation from 1,092 human genomes, Nature 491 (2012) 56e65, https://doi.org/
10.1038/nature11632.
[12] D. Welter, J. MacArthur, J. Morales, T. Burdett, P. Hall, H. Junkins, A. Klemm,
P. Flicek, T. Manolio, L. Hindorff, H. Parkinson, The NHGRI GWAS Catalog, a
curated resource of SNP-trait associations, Nucleic Acids Res. 42 (2014)
D1001eD1006, https://doi.org/10.1093/nar/gkt1229.
[13] G. Gibson, Rare and common variants: twenty arguments, Nat. Rev. Genet. 13
(2011) 135e145, https://doi.org/10.1038/nrg3118.
[14] S. Sivakumaran, F. Agakov, E. Theodoratou, J.G. Prendergast, L. Zgaga,
T. Manolio, I. Rudan, P. McKeigue, J.F. Wilson, H. Campbell, Abundant plei-
otropy in human complex diseases and traits, Am. J. Hum. Genet. 89 (2011)
607e618, https://doi.org/10.1016/j.ajhg.2011.10.004.
[15] M. Hartman, E.Y. Loy, C.S. Ku, K.S. Chia, Molecular epidemiology and its
current clinical use in cancer management, Lancet Oncol. 11 (2010)
383e390, https://doi.org/10.1016/S1470-2045(10)70005-X.
[16] R.B. Brem, G. Yvert, R. Clinton, L. Kruglyak, Genetic dissection of transcrip-
tional regulation in budding yeast, Science 296 (2002) 752e755, https://
doi.org/10.1126/science.1069516.
[17] J.-B. Veyrieras, S. Kudaravalli, S.Y. Kim, E.T. Dermitzakis, Y. Gilad,
M. Stephens, J.K. Pritchard, High-resolution mapping of expression-QTLs
yields insight into human gene regulation, PLoS Genet. 4 (2008),
e1000214, https://doi.org/10.1371/journal.pgen.1000214.
[18] J.H. Moore, Analysis of gene-gene interactions, Curr. Protoc. Hum. Genet.
(2004), https://doi.org/10.1002/0471142905.hg0114s39 (Chapter 1) Unit
1.14.
[19] W.G. Hill, M.E. Goddard, P.M. Visscher, Data and theory point to mainly
additive genetic variance for complex traits, PLoS Genet. 4 (2008), e1000008,
https://doi.org/10.1371/journal.pgen.1000008.
[20] B.J. Traynor, A.B. Singleton, Nature versus nurture: death of a dogma, and the
road ahead, Neuron 68 (2010) 196e200, https://doi.org/10.1016/
j.neuron.2010.10.002.
[21] O. Zuk, E. Hechter, S.R. Sunyaev, E.S. Lander, The mystery of missing heri-
tability: genetic interactions create phantom heritability, Proc. Natl. Acad.
Sci. U. S. A 109 (2012) 1193e1198, https://doi.org/10.1073/
pnas.1119675109.
[22] B.E. Stranger, E.A. Stahl, T. Raj, Progress and promise of genome-wide as-
sociation studies for human complex trait genetics, Genetics 187 (2011)
367e383, https://doi.org/10.1534/genetics.110.120907.
[23] B. Maher, Personal genomes: the case of the missing heritability, Nature 456
(2008) 18e21, https://doi.org/10.1038/456018a.
[24] T.J.C. Polderman, B. Benyamin, C.A. de Leeuw, P.F. Sullivan, A. van Bochoven,
P.M. Visscher, D. Posthuma, Meta-analysis of the heritability of human traits
based on fty years of twin studies, Nat. Genet. 47 (2015) 702e709, https://
doi.org/10.1038/ng.3285.
[25] S.E. Antonarakis, A. Chakravarti, J.C. Cohen, J. Hardy, Mendelian disorders and
multifactorial traits: the big divide or one for all? Nat. Rev. Genet. 11 (2010)
380e384, https://doi.org/10.1038/nrg2793.
[26] T.F.C. Mackay, Epistasis and quantitative traits: using model organisms to
study gene-gene interactions, Nat. Rev. Genet. 15 (2014) 22e33, https://
doi.org/10.1038/nrg3627.
[27] W.-H. Wei, G. Hemani, C.S. Haley, Detecting epistasis in human complex
traits, Nat. Rev. Genet. 15 (2014) 722e733, https://doi.org/10.1038/nrg3747.
[28] S.-M. Ho, A. Johnson, P. Tarapore, V. Janakiram, X. Zhang, Y.-K. Leung,
Environmental epigenetics and its implication on disease risk and health
outcomes, ILAR J. 53 (2012) 289e305, https://doi.org/10.1093/ilar.53.3-
4.289.
[29] P.M. Visscher, W.G. Hill, N.R. Wray, Heritability in the genomics eraconcepts
and misconceptions, Nat. Rev. Genet. 9 (2008) 255e266, https://doi.org/
10.1038/nrg2322.
[30] C. Waddington, Canalization of development and the inheritance of acquired
characters, Nature 150 (1942) 563e565.
[31] S.L. Berger, T. Kouzarides, R. Shiekhattar, A. Shilatifard, An operational de-
nition of epigenetics, Genes Dev. 23 (2009) 781e783, https://doi.org/
10.1101/gad.1787609.
[32] I. Cantone, A.G. Fisher, Epigenetic programming and reprogramming during
development, Nat. Struct. Mol. Biol. 20 (2013) 282e289, https://doi.org/
10.1038/nsmb.2489.
[33] S. Seisenberger, J.R. Peat, T.A. Hore, F. Santos, W. Dean, W. Reik, Reprog-
ramming DNA methylation in the mammalian life cycle: building and
breaking epigenetic barriers, Philos. Trans. R. Soc. B Biol. Sci. 368 (2012),
https://doi.org/10.1098/rstb.2011.0330, 20110330e20110330.
[34] V.K. Cortessis, D.C. Thomas, A. Joan Levine, C.V. Breton, T.M. Mack,
K.D. Siegmund, R.W. Haile, P.W. Laird, Environmental epigenetics: prospects
for studying epigenetic mediation of exposure-response relationships, Hum.
Genet. 131 (2012) 1565e1589, https://doi.org/10.1007/s00439-012-1189-8.
[35] R.S. Dwivedi, J.G. Herman, T.A. McCaffrey, D.S.C. Raj, Beyond genetics:
epigenetic code in chronic kidney disease, Kidney Int. 79 (2011) 23e32,
https://doi.org/10.1038/ki.2010.335.
[36] N. Carey, C.J. Marques, W. Reik, DNA demethylases: a new epigenetic frontier
in drug discovery, Drug Discov. Today 16 (2011) 683e690, https://doi.org/
10.1016/j.drudis.2011.05.004.
[37] J.P. Jost, J. Hofsteenge, The repressor MDBP-2 is a member of the histone H1
family that binds preferentially in vitro and in vivo to methylated nonspe-
cic DNA sequences, Proc. Natl. Acad. Sci. Unit. States Am. 89 (1992)
9499e9503, https://doi.org/10.1073/pnas.89.20.9499.
[38] A. Bird, DNA methylation patterns and epigenetic memory, Genes Dev. 16
(2002) 6e21, https://doi.org/10.1101/gad.947102.
[39] A.M. Deaton, A. Bird, CpG islands and the regulation of transcription, Genes
Dev. 25 (2011) 1010e1022, https://doi.org/10.1101/gad.2037511.
[40] P.A. Jones, Functions of DNA methylation: islands, start sites, gene bodies and
beyond, Nat. Rev. Genet. 13 (2012) 484e492, https://doi.org/10.1038/
nrg3230.
[41] A.P. Bird, DNA methylation and the frequency of CpG in animal DNA, Nucleic
Acids Res. 8 (1980) 1499e1504.
[42] B.E. Bernstein, A. Meissner, E.S. Lander, The mammalian epigenome, Cell 128
(2007) 669e681, https://doi.org/10.1016/j.cell.2007.01.033.
[43] F. Mohn, M. Weber, M. Rebhan, T.C. Roloff, J. Richter, M.B. Stadler, M. Bibel,
D. Schubeler, Lineage-specic polycomb targets and de novo DNA methyl-
ation dene restriction and potential of neuronal progenitors, Mol. Cell. 30
(2008) 755e766, https://doi.org/10.1016/j.molcel.2008.05.007.
[44] A. Meissner, T.S. Mikkelsen, H. Gu, M. Wernig, J. Hanna, A. Sivachenko,
X. Zhang, B.E. Bernstein, C. Nusbaum, D.B. Jaffe, A. Gnirke, R. Jaenisch,
E.S. Lander, Genome-scale DNA methylation maps of pluripotent and
differentiated cells, Nature 454 (2008) 766e770, https://doi.org/10.1038/
nature07107.
[45] M. Weber, I. Hellmann, M.B. Stadler, L. Ramos, S. Paabo, M. Rebhan,
D. Schubeler, Distribution, silencing potential and evolutionary impact of
promoter DNA methylation in the human genome, Nat. Genet. 39 (2007)
457e466, https://doi.org/10.1038/ng1990.
[46] F. Fuks, P.J. Hurd, D. Wolf, X. Nan, A.P. Bird, T. Kouzarides, The methyl-CpG-
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171 167
binding protein MeCP2 links DNA methylation to histone methylation, J. Biol.
Chem. 278 (2003) 4035e4040, https://doi.org/10.1074/jbc.M210256200.
[47] L. Laurent, E. Wong, G. Li, T. Huynh, A. Tsirigos, C.T. Ong, H.M. Low, K.W. Kin
Sung, I. Rigoutsos, J. Loring, C.-L. Wei, Dynamic changes in the human
methylome during differentiation, Genome Res. 20 (2010) 320e331, https://
doi.org/10.1101/gr.101907.109.
[48] A. Hellman, A. Chess, Gene body-specic methylation on the active X chro-
mosome, Science 315 (2007) 1141e1143, https://doi.org/10.1126/
science.1136352.
[49] A.K. Maunakea, R.P. Nagarajan, M. Bilenky, T.J. Ballinger, C. D'Souza,
S.D. Fouse, B.E. Johnson, C. Hong, C. Nielsen, Y. Zhao, G. Turecki, A. Delaney,
R. Varhol, N. Thiessen, K. Shchors, V.M. Heine, D.H. Rowitch, X. Xing, C. Fiore,
M. Schillebeeckx, S.J.M. Jones, D. Haussler, M.A. Marra, M. Hirst, T. Wang,
J.F. Costello, Conserved role of intragenic DNA methylation in regulating
alternative promoters, Nature 466 (2010) 253e257. https://doi.org/10.1038/
nature09165.
[50] R. Tirado-Magallanes, K. Rebbani, R. Lim, S. Pradhan, T. Benoukraf, Whole
genome DNA methylation: beyond genes silencing, Oncotarget 8 (2017)
5629e5637, https://doi.org/10.18632/oncotarget.13562.
[51] Y.-F. He, B.-Z. Li, Z. Li, P. Liu, Y. Wang, Q. Tang, J. Ding, Y. Jia, Z. Chen, L. Li,
Y. Sun, X. Li, Q. Dai, C.-X. Song, K. Zhang, C. He, G.-L. Xu, Tet-mediated for-
mation of 5-carboxylcytosine and its excision by TDG in mammalian DNA,
Science 333 (2011) 1303e1307, https://doi.org/10.1126/science.1210944.
[52] S. Ito, L. Shen, Q. Dai, S.C. Wu, L.B. Collins, J.A. Swenberg, C. He, Y. Zhang, Tet
proteins can convert 5-methylcytosine to 5-formylcytosine and 5-
carboxylcytosine, Science 333 (2011) 1300e1303, https://doi.org/10.1126/
science.1210597.
[53] T. Pfaffeneder, B. Hackner, M. Truss, M. Munzel, M. Muller, C.A. Deiml,
C. Hagemeier, T. Carell, The discovery of 5-formylcytosine in embryonic stem
cell DNA, Angew Chem. Int. Ed. Engl. 50 (2011) 7008e7012, https://doi.org/
10.1002/anie.201103899.
[54] L. Zhang, X. Lu, J. Lu, H. Liang, Q. Dai, G.-L. Xu, C. Luo, H. Jiang, C. He, Thymine
DNA glycosylase specically recognizes 5-carboxylcytosine-modied DNA,
Nat. Chem. Biol. 8 (2012) 328e330, https://doi.org/10.1038/nchembio.914.
[55] J.U. Guo, Y. Su, C. Zhong, G. Ming, H. Song, Hydroxylation of 5-methylcytosine
by TET1 promotes active DNA demethylation in the adult brain, Cell 145
(2011) 423e434, https://doi.org/10.1016/j.cell.2011.03.022.
[56] Y. Huang, A. Rao, New functions for DNA modications by TET-JBP, Nat.
Struct. Mol. Biol. 19 (2012) 1061e1064, https://doi.org/10.1038/nsmb.2437.
[57] S. Kriaucionis, N. Heintz, The nuclear DNA base, 5-hydroxymethylcytosine is
present in brain and enriched in Purkinje neurons, Science 324 (2009)
929e930, https://doi.org/10.1126/science.1169786.
[58] W.A. Pastor, U.J. Pape, Y. Huang, H.R. Henderson, R. Lister, M. Ko,
E.M. McLoughlin, Y. Brudno, S. Mahapatra, P. Kapranov, M. Tahiliani,
G.Q. Daley, X.S. Liu, J.R. Ecker, P.M. Milos, S. Agarwal, A. Rao, Genome-wide
mapping of 5-hydroxymethylcytosine in embryonic stem cells, Nature 473
(2011) 394e397, https://doi.org/10.1038/nature10102.
[59] H. Wu, A.C. D'Alessio, S. Ito, Z. Wang, K. Cui, K. Zhao, Y.E. Sun, Y. Zhang,
Genome-wide analysis of 5-hydroxymethylcytosine distribution reveals its
dual function in transcriptional regulation in mouse embryonic stem cells,
Genes Dev. 25 (2011) 679e684, https://doi.org/10.1101/gad.2036011.
[60] Y. Xu, F. Wu, L. Tan, L. Kong, L. Xiong, J. Deng, A.J. Barbera, L. Zheng, H. Zhang,
S. Huang, J. Min, T. Nicholson, T. Chen, G. Xu, Y. Shi, K. Zhang, Y.G. Shi,
Genome-wide regulation of 5hmC, 5mC, and gene expression by Tet1 hy-
droxylase in mouse embryonic stem cells, Mol. Cell. 42 (2011) 451e464,
https://doi.org/10.1016/j.molcel.2011.04.005.
[61] N. Chia, L. Wang, X. Lu, M.C. Senut, C. Brenner, D.M. Ruden, Hypothesis:
environmental regulation of 5-hydroxymethyl-cytosine by oxidative stress,
Epigenetics 6 (2011) 853e856, https://doi.org/10.4161/epi.6.7.16461.
[62] J.L. García-Gim
enez, J.S. Iba~
nez-Cabellos, M. Seco-Cervera, F.V. Pallard
o,
Glutathione and cellular redox control in epigenetic regulation, Free Radic.
Biol. Med. 75 (Suppl 1:S3) (2014), https://doi.org/10.1016/
j.freeradbiomed.2014.10.828.
[63] S.B. Rothbart, B.D. Strahl, Interpreting the language of histone and DNA
modications, Biochim. Biophys. Acta 1839 (2014) 627e643, https://doi.org/
10.1016/j.bbagrm.2014.03.001.
[64] K. Sarma, D. Reinberg, Histone variants meet their match, Nat. Rev. Mol. Cell
Biol. 6 (2005) 139e149, https://doi.org/10.1038/nrm1567.
[65] H. Huang, B.R. Sabari, B.A. Garcia, C.D. Allis, Y. Zhao, SnapShot: histone
modications, Cell 159 (2014) 458e458.e1. https://doi.org/10.1016/j.cell.
2014.09.037.
[66] A. Portela, M. Esteller, Epigenetic modications and human disease, Nat.
Biotechnol. 28 (2010) 1057e1068, https://doi.org/10.1038/nbt.1685.
[67] T. Kouzarides, Chromatin modications and their function, Cell 128 (2007)
693e705, https://doi.org/10.1016/j.cell.2007.02.005.
[68] C.A. Musselman, M.-E. Lalonde, J. Cote, T.G. Kutateladze, Perceiving the
epigenetic landscape through histone readers, Nat. Struct. Mol. Biol. 19
(2012) 1218e1227, https://doi.org/10.1038/nsmb.2436.
[69] N. Ahuja, H. Easwaran, S.B. Baylin, Harnessing the potential of epigenetic
therapy to target solid tumors, J. Clin. Investig. 124 (2014) 56e63, https://
doi.org/10.1172/JCI69736.
[70] T. Zhang, S. Cooper, N. Brockdorff, The interplay of histone modications -
writers that read, EMBO Rep. 16 (2015) 1467e1481, https://doi.org/
10.15252/embr.201540945.
[71] B.M. Turner, Histone acetylation and an epigenetic code, Bioessays 22 (2000)
836e845, https://doi.org/10.1002/1521-1878(200009)22:9<836::AID-
BIES9>3.0.CO;2-X.
[72] A. Manuscript, L.-M.P. Britton, M. Gonzales-Cope, B.M. Zee, B.A. Garcia,
Breaking the histone code with quantitative mass spectrometry, Expert Rev.
Proteomics 8 (2012) 631e643, https://doi.org/10.1586/epr.11.47 (Breaking).
[73] B. Zhu, D. Reinberg, Epigenetic inheritance: uncontested? Cell Res. 21 (2011)
435e441, https://doi.org/10.1038/cr.2011.26.
[74] V.N. Budhavarapu, M. Chavez, J.K. Tyler, How is epigenetic information
maintained through DNA replication? Epigenet. Chromatin 6 (2013) 32,
https://doi.org/10.1186/1756-8935-6-32.
[75] P.J. Skene, S. Henikoff, Histone variants in pluripotency and disease, Devel-
opment 140 (2013) 2513e2524, https://doi.org/10.1242/dev.091439.
[76] I. Houston, C.J. Peter, A. Mitchell, J. Straubhaar, E. Rogaev, S. Akbarian, Epi-
genetics in the human brain, Neuropsychopharmacology 38 (2013)
183e197, https://doi.org/10.1038/npp.2012.78.
[77] J.-B. Bae, Perspectives of international human epigenome Consortium, Ge-
nomics Inform 11 (2013) 7e14, https://doi.org/10.5808/GI.2013.11.1.7.
[78] Roadmap Epigenomics Consortium, A. Kundaje, W. Meuleman, J. Ernst,
M. Bilenky, A. Yen, A. Heravi-Moussavi, P. Kheradpour, Z. Zhang, J. Wang,
M.J. Ziller, V. Amin, J.W. Whitaker, M.D. Schultz, L.D. Ward, A. Sarkar,
G. Quon, R.S. Sandstrom, M.L. Eaton, Y.-C. Wu, A.R. Pfenning, X. Wang,
M. Claussnitzer, Y. Liu, C. Coarfa, R.A. Harris, N. Shoresh, C.B. Epstein,
E. Gjoneska, D. Leung, W. Xie, R.D. Hawkins, R. Lister, C. Hong, P. Gascard,
A.J. Mungall, R. Moore, E. Chuah, A. Tam, T.K. Caneld, R.S. Hansen, R. Kaul,
P.J. Sabo, M.S. Bansal, A. Carles, J.R. Dixon, K.-H. Farh, S. Feizi, R. Karlic, A.-
R. Kim, A. Kulkarni, D. Li, R. Lowdon, G. Elliott, T.R. Mercer, S.J. Neph,
V. Onuchic, P. Polak, N. Rajagopal, P. Ray, R.C. Sallari, K.T. Siebenthall,
N.A. Sinnott-Armstrong, M. Stevens, R.E. Thurman, J. Wu, B. Zhang, X. Zhou,
A.E. Beaudet, L.A. Boyer, P.L. De Jager, P.J. Farnham, S.J. Fisher, D. Haussler,
S.J.M. Jones, W. Li, M.A. Marra, M.T. McManus, S. Sunyaev, J.A. Thomson,
T.D. Tlsty, L.-H. Tsai, W. Wang, R.A. Waterland, M.Q. Zhang, L.H. Chadwick,
B.E. Bernstein, J.F. Costello, J.R. Ecker, M. Hirst, A. Meissner, A. Milosavljevic,
B. Ren, J.A. Stamatoyannopoulos, T. Wang, M. Kellis, R.E. Consortium, Inte-
grative analysis of 111 reference human epigenomes, Nature 518 (2015)
317e330. https://doi.org/10.1038/nature14248.
[79] The ENCODE (ENCyclopedia of DNA elements) project, Science 306 (2004)
636e640, https://doi.org/10.1126/science.1105136.
[80] C.E. Romanoski, C.K. Glass, H.G. Stunnenberg, L. Wilson, G. Almouzni, Epi-
genomics: Roadmap for regulation, Nature 518 (2015) 314e316, https://
doi.org/10.1038/518314a.
[81] S. Choi, S. Friso, Nutrients and Epigenetics, CRC Press, 2009.
[82] J.P. Thomson, P.J. Skene, J. Selfridge, T. Clouaire, J. Guy, S. Webb, A.R.W. Kerr,
A. Deaton, R. Andrews, K.D. James, D.J. Turner, R. Illingworth, A. Bird, CpG
islands inuence chromatin structure via the CpG-binding protein Cfp1,
Nature 464 (2010) 1082e1086, https://doi.org/10.1038/nature08924.
[83] A. Tanay, A.H. O'Donnell, M. Damelin, T.H. Bestor, Hyperconserved CpG do-
mains underlie Polycomb-binding sites, Proc. Natl. Acad. Sci. U. S. A 104
(2007) 5521e5526, https://doi.org/10.1073/pnas.0609746104.
[84] J. Gertz, K.E. Varley, T.E. Reddy, K.M. Bowling, F. Pauli, S.L. Parker, K.S. Kucera,
H.F. Willard, R.M. Myers, Analysis of DNA methylation in a three-generation
family reveals widespread genetic inuence on epigenetic regulation, PLoS
Genet. 7 (2011) e1002228, https://doi.org/10.1371/journal.pgen.1002228.
[85] A. Corval
an, C. Prüss-Üstün, Preventing disease through healthy environ-
ments. Towards an estimate of the environmental burden of disease, Gen-
evaWorld Heal. Organ (2006) 104 p. ISBN 9241593822, http://www.who.int/
iris/handle/10665/43457.
[86] M.K. Skinner, M.D. Anway, M.I. Savenkova, A.C. Gore, D. Crews, Trans-
generational epigenetic programming of the brain transcriptome and anxi-
ety behavior, PLoS One 3 (2008) e3745. https://doi.org/10.1371/journal.pone.
0003745.
[87] C.J. Steves, T.D. Spector, S.H.D. Jackson, Ageing, genes, environment and
epigenetics: what twin studies tell us now, and in the future, Age Ageing 41
(2012) 581e586, https://doi.org/10.1093/ageing/afs097.
[88] M.F. Fraga, E. Ballestar, M.F. Paz, S. Ropero, F. Setien, M.L. Ballestar, D. Heine-
Suner, J.C. Cigudosa, M. Urioste, J. Benitez, M. Boix-Chornet, A. Sanchez-
Aguilera, C. Ling, E. Carlsson, P. Poulsen, A. Vaag, Z. Stephan, T.D. Spector, Y.-
Z. Wu, C. Plass, M. Esteller, Epigenetic differences arise during the lifetime of
monozygotic twins, Proc. Natl. Acad. Sci. U. S. A 102 (2005) 10604e10609,
https://doi.org/10.1073/pnas.0500398102.
[89] V. Bollati, A. Baccarelli, Environmental epigenetics (2010), https://doi.org/
10.1038/hdy.2010.2.
[90] L. Hou, X. Zhang, D. Wang, A. Baccarelli, Environmental chemical exposures
and human epigenetics, Int. J. Epidemiol. 41 (2012) 79e105, https://doi.org/
10.1093/ije/dyr154.
[91] M.K. Skinner, Endocrine disruptor induction of epigenetic transgenerational
inheritance of disease, Mol. Cell. Endocrinol. 398 (2014) 4e12, https://
doi.org/10.1016/j.mce.2014.07.019.
[92] M.K. Skinner, Environmental epigenetic transgenerational inheritance and
somatic epigenetic mitotic stability, Epigenetics 6 (2011) 838e842.
[93] A.J. Drake, L. Liu, Intergenerational transmission of programmed effects:
public health consequences, Trends Endocrinol. Metabol. 21 (2010)
206e213, https://doi.org/10.1016/j.tem.2009.11.006.
[94] D.C. Benyshek, The early lifeorigins of obesity-related health disorders:
new discoveries regarding the intergenerational transmission of develop-
mentally programmed traits in the global cardiometabolic health crisis, Am.
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171168
J. Phys. Anthropol. 93 (2013) 79e93, https://doi.org/10.1002/ajpa.22393.
[95] H.J. Clarke, K.F. Vieux, Epigenetic inheritance through the female germ-line:
the known, the unknown, and the possible, Semin. Cell Dev. Biol. 43 (2015)
106e116, https://doi.org/10.1016/j.semcdb.2015.07.003.
[96] G.C. Burdge, S.P. Hoile, T. Uller, N.A. Thomas, P.D. Gluckman, M.A. Hanson,
K.A. Lillycrop, Progressive, transgenerational changes in offspring phenotype
and epigenotype following nutritional transition, PLoS One 6 (2011), e28282.
https://doi.org/10.1371/journal.pone.0028282.
[97] J. Song, J. Irwin, C. Dean, Remembering the prolonged cold of winter, Curr.
Biol. 23 (2013) R807eR811, https://doi.org/10.1016/j.cub.2013.07.027.
[98] A.E. Peaston, E. Whitelaw, Epigenetics and phenotypic variation in mammals,
Mamm. Genome 17 (2006) 365e374, https://doi.org/10.1007/s00335-005-
0180-2.
[99] P. Cubas, C. Vincent, E. Coen, An epigenetic mutation responsible for natural
variation in oral symmetry, Nature 401 (1999) 157e161, https://doi.org/
10.1038/43657.
[100] M.I. Lind, F. Spagopoulou, Evolutionary consequences of epigenetic inheri-
tance, Heredity 121 (2018) 205e209, https://doi.org/10.1038/s41437-018-
0113-y.
[101] E.V. Koonin, Y.I. Wolf, Is evolution darwinian or/and lamarckian? Biol. Direct
4 (2009) 42, https://doi.org/10.1186/1745-6150-4-42.
[102] E. V Koonin, Calorie restriction a lamarck, Cell 158 (2014) 237e238, https://
doi.org/10.1016/j.cell.2014.07.004.
[103] J. Lamarck, Recherches sur lorganisation des corps vivans., Paris Chez Lau-
teur, Mail, 1802.
[104] C.M. Guerrero-Bosagna, P. Sabat, F.S. Valdovinos, L.E. Valladares, S.J. Clark,
Epigenetic and phenotypic changes result from a continuous pre and post
natal dietary exposure to phytoestrogens in an experimental population of
mice, BMC Physiol. 8 (2008) 17, https://doi.org/10.1186/1472-6793-8-17.
[105] M.K. Skinner, Environmental epigenetics and a unied theory of the mo-
lecular aspects of evolution: a neo-lamarckian concept that facilitates neo-
darwinian evolution, Genome Biol. Evol. 7 (2015) 1296e1302, https://
doi.org/10.1093/gbe/evv073.
[106] A.P. Feinberg, The epigenetics of cancer etiology, Semin. Canc. Biol. 14 (2004)
427e432, https://doi.org/10.1016/j.semcancer.2004.06.005.
[107] J. Sved, A. Bird, The expected equilibrium of the CpG dinucleotide in verte-
brate genomes under a mutation model, Proc. Natl. Acad. Sci. U.S.A. 87
(1990), https://doi.org/10.1073/pnas.87.12.4692.
[108] M.T. Salam, H.-M. Byun, F. Lurmann, C. V Breton, X. Wang, S.P. Eckel,
F.D. Gilliland, Genetic and epigenetic variations in inducible nitric oxide
synthase promoter, particulate pollution, and exhaled nitric oxide levels in
children, J. Allergy Clin. Immunol. 129 (2012) 232e237, https://doi.org/
10.1016/j.jaci.2011.09.037.
[109] G. Burdge, K. Lillycrop, Nutrition, Epigenetics and Health, 2016. https://doi.
org/10.1142/10123.
[110] T. Su, L. Joseph, Chiang, Environmental Epigenetics, Springer-Verlag London,
2015, https://doi.org/10.1007/978-1-4471-6678-8.
[111] K.K. Dennis, S.S. Auerbach, D.M. Balshaw, Y. Cui, M.D. Fallin, M.T. Smith,
A. Spira, S. Sumner, G.W. Miller, The importance of the biological impact of
exposure to the concept of the exposome, Environ. Health Perspect. 124
(2016) 1504e1510, https://doi.org/10.1289/EHP140.
[112] D.J. Barker, P.D. Gluckman, K.M. Godfrey, J.E. Harding, J.A. Owens,
J.S. Robinson, Fetal nutrition and cardiovascular disease in adult life, Lancet
(N. Am. Ed.) 341 (1993) 938e941.
[113] P.D. Gluckman, M.A. Hanson, H.G. Spencer, Predictive adaptive responses
and human evolution, Trends Ecol. Evol. 20 (2005) 527e533, https://doi.org/
10.1016/j.tree.2005.08.001.
[114] P.D. Gluckman, M.A. Hanson, C. Cooper, K.L. Thornburg, Effect of in utero and
early-life conditions on adult health and disease, N. Engl. J. Med. 359 (2008)
61e73, https://doi.org/10.1056/NEJMra0708473.
[115] D.K. Lahiri, B. Maloney, N.H. Zawia, The LEARn model: an epigenetic expla-
nation for idiopathic neurobiological diseases, Mol. Psychiatr. 14 (2009)
992e1003, https://doi.org/10.1038/mp.2009.82.
[116] A. Garrod, The incidence of alkaptonuria: a study in chemical individuality,
Lancet 2 (1902) 1616e1620.
[117] M. Kohlmeier, Nutrigenetics: Applying the Science of Personal Nutrition,
2012.
[118] J. Roper, Genetic determination of nutritional requirements, Proc. Nutr. Soc.
19 (1960) 39e45.
[119] T. Peregrin, The new frontier of nutrition science: nutrigenomics, J. Am. Diet.
Assoc. 101 (2001) 1306, https://doi.org/10.1016/S0002-8223(01)00309-1.
[120] S.B. Astley, An introduction to nutrigenomics developments and trends,
Genes Nutr 2 (2007) 11e13, https://doi.org/10.1007/s12263-007-0011-z.
[121] J.C. Mathers, Nutrigenomics in the modern era, Proc. Nutr. Soc. 44 (2016)
1e11, https://doi.org/10.1017/S002966511600080X.
[122] A. Mullally, J. Ritz, Beyond HLA: the signicance of genomic variation for
allogeneic hematopoietic stem cell transplantation, Blood 109 (2007)
1355e1362, https://doi.org/10.1182/blood-2006-06-030858.
[123] J. Fu, M. Hofker, C. Wijmenga, Apple or pear: size and shape matter, Cell
Metabol. 21 (2015) 507e508, https://doi.org/10.1016/j.cmet.2015.03.016.
[124] D. Shungin, T.W. Winkler, D.C. Croteau-Chonka, T. Ferreira, A.E. Locke,
R. Magi, R.J. Strawbridge, T.H. Pers, K. Fischer, A.E. Justice, T. Workalemahu,
J.M.W. Wu, M.L. Buchkovich, N.L. Heard-Costa, T.S. Roman, A.W. Drong,
C. Song, S. Gustafsson, F.R. Day, T. Esko, T. Fall, Z. Kutalik, J. Luan, J.C. Randall,
A. Scherag, S. Vedantam, A.R. Wood, J. Chen, R. Fehrmann, J. Karjalainen,
B. Kahali, C.-T. Liu, E.M. Schmidt, D. Absher, N. Amin, D. Anderson,
M. Beekman, J.L. Bragg-Gresham, S. Buyske, A. Demirkan, G.B. Ehret,
M.F. Feitosa, A. Goel, A.U. Jackson, T. Johnson, M.E. Kleber, K. Kristiansson,
M. Mangino, I.M. Leach, C. Medina-Gomez, C.D. Palmer, D. Pasko,
S. Pechlivanis, M.J. Peters, I. Prokopenko, A. Stancakova, Y.J. Sung, T. Tanaka,
A. Teumer, J.V. Van Vliet-Ostaptchouk, L. Yengo, W. Zhang, E. Albrecht,
J. Arnlov, G.M. Arscott, S. Bandinelli, A. Barrett, C. Bellis, A.J. Bennett, C. Berne,
M. Bluher, S. Bohringer, F. Bonnet, Y. Bottcher, M. Bruinenberg, D.B. Carba,
I.H. Caspersen, R. Clarke, E.W. Daw, J. Deelen, E. Deelman, G. Delgado,
A.S. Doney, N. Eklund, M.R. Erdos, K. Estrada, E. Eury, N. Friedrich,
M.E. Garcia, V. Giedraitis, B. Gigante, A.S. Go, A. Golay, H. Grallert,
T.B. Grammer, J. Grassler, J. Grewal, C.J. Groves, T. Haller, G. Hallmans,
C.A. Hartman, M. Hassinen, C. Hayward, K. Heikkila, K.-H. Herzig, Q. Helmer,
H.L. Hillege, O. Holmen, S.C. Hunt, A. Isaacs, T. Ittermann, A.L. James,
I. Johansson, T. Juliusdottir, I.-P. Kalafati, L. Kinnunen, W. Koenig, I.K. Kooner,
W. Kratzer, C. Lamina, K. Leander, N.R. Lee, P. Lichtner, L. Lind, J. Lindstrom,
S. Lobbens, M. Lorentzon, F. Mach, P.K. Magnusson, A. Mahajan,
W.L. McArdle, C. Menni, S. Merger, E. Mihailov, L. Milani, R. Mills,
A. Moayyeri, K.L. Monda, S.P. Mooijaart, T.W. Muhleisen, A. Mulas, G. Muller,
M. Muller-Nurasyid, R. Nagaraja, M.A. Nalls, N. Narisu, N. Glorioso, I.M. Nolte,
M. Olden, N.W. Rayner, F. Renstrom, J.S. Ried, N.R. Robertson, L.M. Rose,
S. Sanna, H. Scharnagl, S. Scholtens, B. Sennblad, T. Seufferlein, C.M. Sitlani,
A.V. Smith, K. Stirrups, H.M. Stringham, J. Sundstrom, M.A. Swertz, A.J. Swift,
A.-C. Syvanen, B.O. Tayo, B. Thorand, G. Thorleifsson, A. Tomaschitz, C. Troffa,
F.V. van Oort, N. Verweij, J.M. Vonk, L.L. Waite, R. Wennauer, T. Wilsgaard,
M.K. Wojczynski, A. Wong, Q. Zhang, J.H. Zhao, E.P. Brennan, M. Choi,
P. Eriksson, L. Folkersen, A. Franco-Cereceda, A.G. Gharavi, A.K. Hedman, M.-
F. Hivert, J. Huang, S. Kanoni, F. Karpe, S. Keildson, K. Kiryluk, L. Liang,
R.P. Lifton, B. Ma, A.J. McKnight, R. McPherson, A. Metspalu, J.L. Min,
M.F. Moffatt, G.W. Montgomery, J.M. Murabito, G. Nicholson, D.R. Nyholt,
C. Olsson, J.R. Perry, E. Reinmaa, R.M. Salem, N. Sandholm, E.E. Schadt,
R.A. Scott, L. Stolk, E.E. Vallejo, H.-J. Westra, K.T. Zondervan, P. Amouyel,
D. Arveiler, S.J. Bakker, J. Beilby, R.N. Bergman, J. Blangero, M.J. Brown,
M. Burnier, H. Campbell, A. Chakravarti, P.S. Chines, S. Claudi-Boehm,
F.S. Collins, D.C. Crawford, J. Danesh, U. de Faire, E.J. de Geus, M. Dorr,
R. Erbel, J.G. Eriksson, M. Farrall, E. Ferrannini, J. Ferrieres, N.G. Forouhi,
T. Forrester, O.H. Franco, R.T. Gansevoort, C. Gieger, V. Gudnason,
C.A. Haiman, T.B. Harris, A.T. Hattersley, M. Heliovaara, A.A. Hicks,
A.D. Hingorani, W. Hoffmann, A. Hofman, G. Homuth, S.E. Humphries,
E. Hypponen, T. Illig, M.-R. Jarvelin, B. Johansen, P. Jousilahti, A.M. Jula,
J. Kaprio, F. Kee, S.M. Keinanen-Kiukaanniemi, J.S. Kooner, C. Kooperberg,
P. Kovacs, A.T. Kraja, M. Kumari, K. Kuulasmaa, J. Kuusisto, T.A. Lakka,
C. Langenberg, L. Le Marchand, T. Lehtimaki, V. Lyssenko, S. Mannisto,
A. Marette, T.C. Matise, C.A. McKenzie, B. McKnight, A.W. Musk,
S. Mohlenkamp, A.D. Morris, M. Nelis, C. Ohlsson, A.J. Oldehinkel, K.K. Ong,
L.J. Palmer, B.W. Penninx, A. Peters, P.P. Pramstaller, O.T. Raitakari,
T. Rankinen, D.C. Rao, T.K. Rice, P.M. Ridker, M.D. Ritchie, I. Rudan,
V. Salomaa, N.J. Samani, J. Saramies, M.A. Sarzynski, P.E. Schwarz,
A.R. Shuldiner, J.A. Staessen, V. Steinthorsdottir, R.P. Stolk, K. Strauch,
A. Tonjes, A. Tremblay, E. Tremoli, M.-C. Vohl, U. Volker, P. Vollenweider,
J.F. Wilson, J.C. Witteman, L.S. Adair, M. Bochud, B.O. Boehm, S.R. Bornstein,
C. Bouchard, S. Cauchi, M.J. Cauleld, J.C. Chambers, D.I. Chasman,
R.S. Cooper, G. Dedoussis, L. Ferrucci, P. Froguel, H.-J. Grabe, A. Hamsten,
J. Hui, K. Hveem, K.-H. Jockel, M. Kivimaki, D. Kuh, M. Laakso, Y. Liu, W. Marz,
P.B. Munroe, I. Njolstad, B.A. Oostra, C.N. Palmer, N.L. Pedersen, M. Perola,
L. Perusse, U. Peters, C. Power, T. Quertermous, R. Rauramaa, F. Rivadeneira,
T.E. Saaristo, D. Saleheen, J. Sinisalo, P.E. Slagboom, H. Snieder, T.D. Spector,
K. Stefansson, M. Stumvoll, J. Tuomilehto, A.G. Uitterlinden, M. Uusitupa,
P. van der Harst, G. Veronesi, M. Walker, N.J. Wareham, H. Watkins, H.-
E. Wichmann, G.R. Abecasis, T.L. Assimes, S.I. Berndt, M. Boehnke,
I.B. Borecki, P. Deloukas, L. Franke, T.M. Frayling, L.C. Groop, D.J. Hunter,
R.C. Kaplan, J.R. O'Connell, L. Qi, D. Schlessinger, D.P. Strachan,
U. Thorsteinsdottir, C.M. van Duijn, C.J. Willer, P.M. Visscher, J. Yang,
J.N. Hirschhorn, M.C. Zillikens, M.I. McCarthy, E.K. Speliotes, K.E. North,
C.S. Fox, I. Barroso, P.W. Franks, E. Ingelsson, I.M. Heid, R.J. Loos, L.A. Cupples,
A.P. Morris, C.M. Lindgren, K.L. Mohlke, New genetic loci link adipose and
insulin biology to body fat distribution, Nature 518 (2015) 187e196, https://
doi.org/10.1038/nature14132.
[125] B. de Roos, Personalised nutrition: ready for practice? Proc. Nutr. Soc. 72
(2013) 48e52, https://doi.org/10.1017/S0029665112002844.
[126] M. Fenech, A. El-Sohemy, L. Cahill, L.R. Ferguson, T.A.C. French, E.S. Tai,
J. Milner, W.P. Koh, L. Xie, M. Zucker, M. Buckley, L. Cosgrove, T. Lockett,
K.Y.C. Fung, R. Head, Nutrigenetics and nutrigenomics: viewpoints on the
current status and applications in nutrition research and practice,
J. Nutrigenetics Nutrigenomics 4 (2011) 69e89, https://doi.org/10.1159/
000327772.
[127] L.R. Ferguson, R. De Caterina, U. G
orman, H. Allayee, M. Kohlmeier, C. Prasad,
M.S. Choi, R. Curi, D.A. De Luis,
A. Gil, J.X. Kang, R.L. Martin, F.I. Milagro,
C.F. Nicoletti, C.B. Nonino, J.M. Ordovas, V.R. Parslow, M.P. Portillo, J.L. Santos,
C.N. Serhan, A.P. Simopoulos, A. Vel
azquez-Arellano, M.A. Zulet,
J.A. Martinez, Guide and position of the international society of nutrige-
netics/nutrigenomics on personalised nutrition: Part 1 - elds of precision
nutrition, J. Nutrigenetics Nutrigenomics 9 (2016) 12e27, https://doi.org/
10.1159/000445350.
[128] H.-G. Joost, M.J. Gibney, K.D. Cashman, U. Gorman, J.E. Hesketh, M. Mueller,
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171 169
B. van Ommen, C.M. Williams, J.C. Mathers, Personalised nutrition: status
and perspectives, Br. J. Nutr. 98 (2007) 26e31, https://doi.org/10.1017/
S0007114507685195.
[129] R. Fallaize, A.L. Macready, L.T. Butler, J.A. Ellis, J.A. Lovegrove, An insight into
the public acceptance of nutrigenomic-based personalised nutrition, Nutr.
Res. Rev. 26 (2013) 39e48, https://doi.org/10.1017/S0954422413000024.
[130] J. Horne, J. Madill, C. O'Connor, J. Shelley, J. Gilliland, A systematic review of
genetic testing and lifestyle behaviour change: are we using high-quality
genetic interventions and considering behaviour change theory? Lifestyle
Genomics (2018) https://doi.org/10.1159/000488086.
[131] D.E. Nielsen, A. El-Sohemy, A randomized trial of genetic information for
personalized nutrition, Genes Nutr 7 (2012) 559e566, https://doi.org/
10.1007/s12263-012-0290-x.
[132] S. Alhassan, S. Kim, A. Bersamin, A.C. King, C.D. Gardner, Dietary adherence
and weight loss success among overweight women: results from the A TO Z
weight loss study, Int. J. Obes. 32 (2008) 985e991, https://doi.org/10.1038/
ijo.2008.8.
[133] C. Celis-Morales, K.M. Livingstone, C.F.M. Marsaux, H. Forster,
C.B. O'Donovan, C. Woolhead, A.L. Macready, R. Fallaize, S. Navas-Carretero,
R. San-Cristobal, S. Kolossa, K. Hartwig, L. Tsirigoti, C.P. Lambrinou,
G. Moschonis, M. Godlewska, A. Surwillo, K. Grimaldi, J. Bouwman, E.J. Daly,
V. Akujobi, R. O'Riordan, J. Hoonhout, A. Claassen, U. Hoeller, T.E. Gundersen,
S.E. Kaland, J.N.S. Matthews, Y. Manios, I. Traczyk, C.A. Drevon, E.R. Gibney,
L. Brennan, M.C. Walsh, J.A. Lovegrove, J. Alfredo Martinez, W.H.M. Saris,
H. Daniel, M. Gibney, J.C. Mathers, Design and baseline characteristics of the
Food4Me study: a web-based randomised controlled trial of personalised
nutrition in seven European countries, Genes Nutr 10 (2015) 450, https://
doi.org/10.1007/s12263-014-0450-2.
[134] C. Celis-Morales, C.F. Marsaux, K.M. Livingstone, S. Navas-Carretero, R. San-
Cristobal, R. Fallaize, A.L. Macready, C. O'Donovan, C. Woolhead, H. Forster,
S. Kolossa, H. Daniel, G. Moschonis, C. Mavrogianni, Y. Manios, A. Surwillo,
I. Traczyk, C.A. Drevon, K. Grimaldi, J. Bouwman, M.J. Gibney, M.C. Walsh,
E.R. Gibney, L. Brennan, J.A. Lovegrove, J.A. Martinez, W.H. Saris, J.C. Mathers,
Can genetic-based advice help you lose weight? Findings from the Food4Me
European randomized controlled trial, Am. J. Clin. Nutr. 105 (2017)
1204e1213, https://doi.org/10.3945/ajcn.116.145680.
[135] R. San-Cristobal, S. Navas-Carretero, K.M. Livingstone, C. Celis-Morales,
A.L. Macready, R. Fallaize, C.B. O'Donovan, C.P. Lambrinou, G. Moschonis,
C.F.M. Marsaux, Y. Manios, M. Jarosz, H. Daniel, E.R. Gibney, L. Brennan,
C.A. Drevon, T.E. Gundersen, M. Gibney, W.H.M. Saris, J.A. Lovegrove,
K. Grimaldi, L.D. Parnell, J. Bouwman, B. Van Ommen, J.C. Mathers,
J.A. Martinez, Mediterranean diet adherence and genetic background roles
within a web-based nutritional intervention: the Food4Me study, Nutrients
9 (2017), https://doi.org/10.3390/nu9101107.
[136] C. Celis-Morales, K.M. Livingstone, C.F. Marsaux, A.L. Macready, R. Fallaize,
C.B. O'Donovan, C. Woolhead, H. Forster, M.C. Walsh, S. Navas-Carretero,
R. San-Cristobal, L. Tsirigoti, C.P. Lambrinou, C. Mavrogianni, G. Moschonis,
S. Kolossa, J. Hallmann, M. Godlewska, A. Surwillo, I. Traczyk, C.A. Drevon,
J. Bouwman, B. van Ommen, K. Grimaldi, L.D. Parnell, J.N. Matthews,
Y. Manios, H. Daniel, J.A. Martinez, J.A. Lovegrove, E.R. Gibney, L. Brennan,
W.H. Saris, M. Gibney, J.C. Mathers, Effect of personalized nutrition on
health-related behaviour change: evidence from the Food4Me European
randomized controlled trial, Int. J. Epidemiol. 46 (2017) 578e588, https://
doi.org/10.1093/ije/dyw186.
[137] R. Fallaize, C. Celis-Morales, A.L. Macready, C.F. Marsaux, H. Forster,
C. O'Donovan, C. Woolhead, R. San-Cristobal, S. Kolossa, J. Hallmann,
C. Mavrogianni, A. Surwillo, K.M. Livingstone, G. Moschonis, S. Navas-Car-
retero, M.C. Walsh, E.R. Gibney, L. Brennan, J. Bouwman, K. Grimaldi,
Y. Manios, I. Traczyk, C.A. Drevon, J.A. Martinez, H. Daniel, W.H. Saris,
M.J. Gibney, J.C. Mathers, J.A. Lovegrove, The effect of the apolipoprotein E
genotype on response to personalized dietary advice intervention: ndings
from the Food4Me randomized controlled trial, Am. J. Clin. Nutr. 104 (2016)
827e836, https://doi.org/10.3945/ajcn.116.135012.
[138] U. G
orman, J.C. Mathers, K.A. Grimaldi, J. Ahlgren, K. Nordstr
om, Do we know
enough? A scientic and ethical analysis of the basis for genetic-based
personalized nutrition, Genes Nutr 8 (2013) 373e381, https://doi.org/
10.1007/s12263-013-0338-6.
[139] A.B. Wildavsky, Searching for safety, Trans. Publ. 10 (1988).
[140] M. Guasch-Ferre, H.S. Dashti, J. Merino, Nutritional genomics and direct-to-
consumer genetic testing: an overview, Adv. Nutr. 9 (2018) 128e135,
https://doi.org/10.1093/advances/nmy001.
[141] G.A. Francis, E. Fayard, F. Picard, J. Auwerx, Nuclear receptors and the control
of metabolism, Annu. Rev. Physiol. 65 (2003) 261e311, https://doi.org/
10.1146/annurev.physiol.65.092101.142528.
[142] W.E. Evans, H.L. McLeod, Pharmacogenomics-drug disposition, drug targets,
and side effects, N. Engl. J. Med. 348 (2003) 538e549, https://doi.org/
10.1056/NEJMra020526.
[143] M. Müller, S. Kersten, Nutrigenomics: goals and strategies, Nat. Rev. Genet. 4
(2003) 315e322, https://doi.org/10.1038/nrg1047.
[144] M. Remely, B. Stefanska, L. Lovrecic, U. Magnet, A.G. Haslberger, Nutriepi-
genomics: the role of nutrition in epigenetic control of human diseases, Curr.
Opin. Clin. Nutr. Metab. Care 18 (2015) 328e333, https://doi.org/10.1097/
MCO.0000000000000180.
[145] N. Zhang, Epigenetic modulation of DNA methylation by nutrition and its
mechanisms in animals, Anim. Nutr. 1 (2015) 144e151. https://doi.org/10.
1016/j.aninu.2015.09.002.
[146] K.S. Crider, T.P. Yang, R.J. Berry, L.B. Bailey, Folate and DNA Methylation : a
review of molecular mechanisms and the evidence for folate s role, Am. Soc.
Nutr. 3 (2012) 21e38, https://doi.org/10.3945/an.111.000992.Figure.
[147] M. Remely, L. Lovrecic, A.L. de la Garza, L. Migliore, B. Peterlin, F.I. Milagro,
A.J. Martinez, A.G. Haslberger, Therapeutic perspectives of epigenetically
active nutrients, Br. J. Pharmacol. 172 (2015) 2756e2768, https://doi.org/
10.1111/bph.12854.
[148] A. Guillaumet-Adkins, Y. Ya~
nez, M.D. Peris-Diaz, I. Calabria, C. Palanca-Bal-
lester, J. Sandoval, Epigenetics and oxidative stress in aging, Oxid. Med. Cell.
Longev. 2017 (2017), https://doi.org/10.1155/2017/9175806.
[149] H. Manev, S. Dzitoyeva, Progress in mitochondrial epigenetics, Biomol.
Concepts 4 (2013) 381e389, https://doi.org/10.1515/bmc-2013-0005.
[150] B. Paul, S. Barnes, W. Demark-Wahnefried, C. Morrow, C. Salvador, C. Skibola,
T.O. Tollefsbol, Inuences of diet and the gut microbiome on epigenetic
modulation in cancer and other diseases, Clin. Epigenet. 7 (2015), https://
doi.org/10.1186/s13148-015-0144-7.
[151] F.A. Tomas-Barberan, A. Gonzalez-Sarrias, R. Garcia-Villalba, M.A. Nunez-
Sanchez, M. V Selma, M.T. Garcia-Conesa, J.C. Espin, Urolithins, the rescue of
oldmetabolites to understand a newconcept: metabotypes as a nexus
among phenolic metabolism, microbiota dysbiosis, and host health status,
Mol. Nutr. Food Res. 61 (2017), https://doi.org/10.1002/mnfr.201500901.
[152] M. Collotta, P.A. Bertazzi, V. Bollati, Epigenetics and pesticides, Toxicology
307 (2013) 35e41, https://doi.org/10.1016/j.tox.2013.01.017.
[153] M.H. Lee, E.R. Cho, J. Lim, S.H. Jee, Association between serum persistent
organic pollutants and DNA methylation in Korean adults, Environ. Res. 158
(2017) 333e341. https://doi.org/10.1016/j.envres.2017.06.017.
[154] L. Cantone, S. Iodice, L. Tarantini, B. Albetti, I. Restelli, L. Vigna, M. Bonzini,
A.C. Pesatori, V. Bollati, Particulate matter exposure is associated with in-
ammatory gene methylation in obese subjects, Environ. Res. 152 (2017)
478e484. https://doi.org/10.1016/j.envres.2016.11.002.
[155] S. Modgil, D.K. Lahiri, V.L. Sharma, A. Anand, Role of early life exposure and
environment on neurodegeneration: implications on brain disorders, Transl.
Neurodegener. 3 (2014) 9, https://doi.org/10.1186/2047-9158-3-9.
[156] C. Song, A. Kanthasamy, V. Anantharam, F. Sun, a G. Kanthasamy, Environ-
mental neurotoxic pesticide increases histone acetylation to promote
apoptosis in dopaminergic neuronal cells: relevance to epigenetic mecha-
nisms of neurodegeneration, Mol. Pharmacol. 77 (2010) 621e632, https://
doi.org/10.1124/mol.109.062174.
[157] D. Fedeli, M. Montani, L. Bordoni, R. Galeazzi, C. Nasuti, L. Correia-S
a,
V.F. Domingues, M. Jayant, V. Brahmachari, L. Massaccesi, E. Laudadio,
R. Gabbianelli, In vivo and in silico studies to identify mechanisms associated
with Nurr1 modulation following early life exposure to permethrin in rats,
Neuroscience 340 (2017), https://doi.org/10.1016/j.neuroscience.2016.
10.071.
[158] H. Miyaso, K. Sakurai, S. Takase, A. Eguchi, M. Watanabe, H. Fukuoka, C. Mori,
The methylation levels of the H19 differentially methylated region in human
umbilical cords reect newborn parameters and changes by maternal
environmental factors during early pregnancy, Environ. Res. 157 (2017) 1e8.
https://doi.org/10.1016/j.envres.2017.05.006.
[159] M.H. Vickers, Early life nutrition, epigenetics and programming of later life
disease, Nutrients 6 (2014) 2165e2178, https://doi.org/10.3390/nu6062165.
[160] C.-H. Chen, S.S. Jiang, I.-S. Chang, H.-J. Wen, C.-W. Sun, S.-L. Wang, Associa-
tion between fetal exposure to phthalate endocrine disruptor and genome-
wide DNA methylation at birth, Environ. Res. 162 (2018) 261e270. https://
doi.org/10.1016/j.envres.2018.01.009.
[161] L. Bordoni, C. Nasuti, M. Mirto, F. Caradonna, R. Gabbianelli, Intergenerational
effect of early life exposure to permethrin: changes in global DNA methyl-
ation and in Nurr1 gene expression, Toxics 3 (2015) 451e461, https://
doi.org/10.3390/toxics3040451.
[162] H.D. Morgan, H.G. Sutherland, D.I. Martin, E. Whitelaw, Epigenetic inheri-
tance at the agouti locus in the mouse, Nat. Genet. 23 (1999) 314e318,
https://doi.org/10.1038/15490.
[163] G.L. Wolff, R.L. Kodell, S.R. Moore, C.A. Cooney, Maternal epigenetics and
methyl supplements affect agouti gene expression in Avy/a mice, FASEB J.
Off. Publ. Fed. Am. Soc. Exp. Biol. 12 (1998) 949e957.
[164] R. Goyal, D. Goyal, A. Leitzke, C.P. Gheorghe, L.D. Longo, Brain renin-
angiotensin system: fetal epigenetic programming by maternal protein re-
striction during pregnancy, Reprod. Sci. 17 (2010) 227e238, https://doi.org/
10.1177/1933719109351935.
[165] L.H. Lumey, Decreased birthweights in infants after maternal in utero
exposure to the Dutch famine of 1944-1945, Paediatr. Perinat. Epidemiol. 6
(1992) 240e253.
[166] G. Kaati, L.O. Bygren, S. Edvinsson, Cardiovascular and diabetes mortality
determined by nutrition during parents' and grandparents' slow growth
period, Eur. J. Hum. Genet. 10 (2002) 682e688, https://doi.org/10.1038/
sj.ejhg.5200859.
[167] S. Friso, S.-W. Choi, D. Girelli, J.B. Mason, G.G. Dolnikowski, P.J. Bagley,
O. Olivieri, P.F. Jacques, I.H. Rosenberg, R. Corrocher, J. Selhub, A common
mutation in the 5,10-methylenetetrahydrofolate reductase gene affects
genomic DNA methylation through an interaction with folate status, Proc.
Natl. Acad. Sci. U. S. A 99 (2002) 5606e5611, https://doi.org/10.1073/
pnas.062066299.
[168] S.H. Zeisel, Choline: critical role during fetal development and dietary re-
quirements in adults, Annu. Rev. Nutr. 26 (2006) 229e250, https://doi.org/
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171170
10.1146/annurev.nutr.26.061505.111156.
[169] S.H. Zeisel, Nutritional genomics: dening the dietary requirement and ef-
fects of choline, J. Nutr. 141 (2011) 531e534, https://doi.org/10.3945/
jn.110.130369.
[170] N.S. Enattah, T. Sahi, E. Savilahti, J.D. Terwilliger, L. Peltonen, I. Jarvela,
Identication of a variant associated with adult-type hypolactasia, Nat.
Genet. 30 (2002) 233e237, https://doi.org/10.1038/ng826.
[171] V. Labrie, O.J. Buske, E. Oh, R. Jeremian, C. Ptak, G. Gasi
unas, A. Maleckas,
R. Petereit, A.
Zvirbliene, K. Adamonis, E. Kriukien _
e, K. Koncevi
cius,
J. Gordevi
cius, A. Nair, A. Zhang, S. Ebrahimi, G. Oh, V.
Sik
snys, L. Kup
cinskas,
M. Brudno, A. Petronis, Lactase non-persistence is directed by DNA variation-
dependent epigenetic aging HHS Public Access, Nat. Struct. Mol. Biol. 23
(2016) 566e573, https://doi.org/10.1038/nsmb.3227.
[172] A. Doehring, G. Geisslinger, J. Lotsch, Epigenetics in pain and analgesia: an
imminent research eld, Eur. J. Pain 15 (2011) 11e16, https://doi.org/
10.1016/j.ejpain.2010.06.004.
[173] R. St
oger, Epigenetics and obesity, Pharmacogenomics 9 (2008) 1851e1860,
https://doi.org/10.2217/14622416.9.12.1851.
[174] T. Tollefsbol, Personalized Epigenetics, 2015.
[175] F. Squadrito, H. Marini, A. Bitto, D. Altavilla, F. Polito, E.B. Adamo, R. D'Anna,
V. Arcoraci, B.P. Burnett, L. Minutoli, A. Di Benedetto, G. Di Vieste,
D. Cucinotta, C. de Gregorio, S. Russo, F. Corrado, A. Saitta, C. Irace, S. Corrao,
G. Licata, Genistein in the metabolic syndrome: results of a randomized
clinical trial, J. Clin. Endocrinol. Metab. 98 (2013) 3366e3374, https://
doi.org/10.1210/jc.2013-1180.
[176] S. Chuengsamarn, S. Rattanamongkolgul, R. Luechapudiporn,
C. Phisalaphong, S. Jirawatnotai, Curcumin extract for prevention of type 2
diabetes, Diabetes Care 35 (2012) 2121e2127, https://doi.org/10.2337/dc12-
0116.
[177] F.I. Milagro, M.L. Mansego, C. De Miguel, J.A. Martinez, Dietary factors,
epigenetic modications and obesity outcomes: progresses and perspectives,
Mol. Aspect. Med. 34 (2013) 782e812, https://doi.org/10.1016/
j.mam.2012.06.010.
[178] A. Scalbert, L. Brennan, C. Manach, C. Andres-Lacueva, L.O. Dragsted,
J. Draper, S.M. Rappaport, J.J.J. van der Hooft, D.S. Wishart, The food
metabolome: a window over dietary exposure, Am. J. Clin. Nutr. 99 (2014)
1286e1308, https://doi.org/10.3945/ajcn.113.076133.
[179] C. Murgia, M.M. Adamski, Translation of nutritional genomics into nutrition
practice: the next step, Nutrients 9 (2017), https://doi.org/10.3390/
nu9040366.
[180] J. Collins, M.M. Adamski, C. Twohig, C. Murgia, Opportunities for training for
nutritional professionals in nutritional genomics: what is out there? Nutr.
Diet. 75 (2018) 206e218, https://doi.org/10.1111/1747-0080.12398.
[181] T. Fournier, J.-P. Poulain, Eating according to one's genes? Exploring the
French public's understanding of and reactions to personalized nutrition,
Qual. Health Res. 28 (2018) 2195e2207, https://doi.org/10.1177/
1049732318793417.
L. Bordoni, R. Gabbianelli / Biochimie 160 (2019) 156e171 171
... Braz Nutrigenomics is a new and emerging field of medicine that uses complementary molecular tools, including biochemistry, physiology, proteomics, metabolomics, transcriptomics, and epigenomics, to identify and explain how bioactive compounds in a specific diet can influence gene expression. It is the area that analyzes and reflects on the interaction between nutrients and the genome at the molecular level, to explain how specific nutrients or dietary regimes can affect human health (Gentile et al., 2015;Bordoni;Gabbianelli, 2019;Madonna;De Caterina, 2020). ...
... addition,Bordoni et al. (2019) found beneficial results with the use of EVOO and Nigella sativa (NG), as they induced antioxidant and epigenetic mechanisms that would be important in the prevention and control of inflammatory processes in diseases such as diabetes, obesity, and cardiovascular diseases. The model used was performed in cell cultures to investigate the properties of two high-quality oils, EVOO and NG, in an in vitro model of low-grade inflammation of human macrophages (THP-1 cells). ...
Article
Introduction: Cardiovascular diseases (CVD) are responsible for 31% of global mortality, most in the form of coronary heart disease (CHD) and stroke. Nutrigenomics and nutrigenetics have been tools in the prevention and treatment of cardiovascular diseases. Objective: to review the main polymorphisms related to food metabolism that can be used in the diagnosis and monitoring of cardiovascular diseases. Material and Methods: This study is qualitative and descriptive through a literature review that consulted the PubMed, Virtual Health Library, Scielo, Capes Portal and Google Scholar databases with original articles published in Portuguese and English in the period between January 2010 and September 2021. Results: 26 articles were found as potentially in accordance with the search criteria. After analyzing the abstracts, 17 articles (65.38%) were excluded and 10 (38.46%) articles were selected. Discussion: Benefits of changing the dietary pattern were found that can promote prevention and reduce the risk of cardiovascular events. Conclusion: The use of bioactive substances can contribute positively to modulating the expression of genes associated with increased cardiovascular risk.
... Traditional dietary guidelines often follow a generalized approach, which may not consider the genetic differences that affect how people respond to food and exercise. Genetic testing allows for more individualized dietary interventions, taking into account specific genetic factors that influence metabolism, nutrient absorption, and energy expenditure [16][17][18]. In the context of sports, genetic testing can help athletes understand how their bodies react to different diets and training programs [19]. ...
Article
Full-text available
Genetic testing has emerged as a tool for personalizing nutrition and athletic training by identifying individual genetic variations that influence metabolism, nutrient absorption, and physical performance. However, its effectiveness in optimizing athletic outcomes remains debated. This study aims to assess the impact of genetic testing on athletes’ dietary habits, digestion, body composition, and performance, exploring the perceived benefits of personalized nutrition in sports. A cross-sectional survey was conducted among 448 athletes in Saudi Arabia, spanning various sports disciplines and competitive levels. The survey measured athletes’ perceptions of genetic testing’s impact on diet and performance using Likert-scale questions. The results indicate that the athletes moderately agreed that genetic testing influenced their dietary adjustments (mean = 3.69), with improved digestion and nutrient absorption being the most positively perceived outcome (mean = 4.01). However, the perceived impact on tailored diets was lower (mean = 2.95), and recovery time showed minimalimprovement (mean = 2.92). Significant gender differences were observed in digestion (p = .0086) and body composition changes (p < .0001), with males reporting greater improvements. Athletes in artistic sports reported the most significant changes in body composition (mean = 4.36, p = .0005), while professionals had the highest perceived benefit from personalized diets (mean = 4.40, p < .0001). In conclusion, genetic testing shows promise in enhancing athletes’ nutrition and performance, particularly in digestion and body composition. However, its impact remains limited in areas like recovery, highlighting the need for a holistic approach integrating genetic data with broader training and lifestyle strategies.
... Nutrition not only interacts with our genetic sensitivity to food and nutrients but can also alter gene expression through epigenetic mechanisms. The epigenetic effects of nutrients have gained significant importance in preventing, alleviating, and treating many diseases [5]. Epigenetics regulates gene expression without altering the coding sequence of DNA (deoxyribonucleic acid), determining how and when specific genes will be turned on or off [6]. ...
Article
Full-text available
Background: Nutrigenetics explores how genetic variations influence an individual's responses to nutrients, enabling personalized nutrition. As dietary supplements gain popularity, understanding genetic factors in their metabolism and effectiveness is crucial for optimal health outcomes. This study examines the role of genetic differences in the metabolism and effects of nutraceuticals, underscoring the significance of personalized nutrition within precision health. It aims to reveal how individual genetic profiles influence responses to dietary supplements, highlighting the value of nutrigenetics in optimizing health interventions. The study explores how genetic variations affect the absorption and effects of nutraceuticals, focusing on personalized supplement choices based on nutrigenetics. Methods: Sixteen patients from an Epigenetic Coaching clinic who were using supplements such as quercetin, curcumin, green tea, and sulforaphane and reporting side effects were studied. Their clinical outcomes were analyzed in relation to their supplement choices and genetic backgrounds. The study involved five women and 11 men, including eight with autism and others with conditions like Hashimoto's thyroiditis (HT) disease and joint pain. Results: In the study, it was observed that removing sulforaphane and sulfur-rich supplements from the diet of five patients reduced agitation. Removing sulforaphane and sulfur-rich supplements from the diet of four patients reduced clinical symptoms. Green tea caused discomfort in two patients. Responses to quercetin showed clinical differences in two patients. Anxiety and hyperactivity increased in three patients who took curcumin. Conclusion This study highlights the importance of considering individual genetic profiles when recommending dietary supplements. The findings suggest that personalized nutrition, guided by nutrigenetic insights, can enhance the efficacy and safety of nutraceutical interventions. Tailoring supplement choices based on genetic information can lead to better health outcomes and reduced adverse effects, emphasizing the need for integrating genetic testing into nutritional planning and healthcare practices.
... The precise identification and analysis of metabolites using mass spectrometry (MS), infra-red (IR), and Raman spectroscopy, as well as nuclear magnetic resonance (NMR) in combination with modern computational approaches, can further advance the progress in the discipline of metabolomics (19,20). Additionally, different approaches including nutrigenetics, epigenetics, and nutriepigenetics (21,22), proteomics (23), gut microbiota (24), dietary interventions (25), nutraceuticals (26), lifestyle (27), and machine learning tools (28) have shown promise in the field of precision nutrition focused toward better human health ( Figure 1). ...
Article
Full-text available
Currently, the treatment of various human ailments is based on different therapeutic approaches including traditional and modern medicine systems. Precision nutrition has come into existence as an emerging approach considering the diverse aspects such as age, sex, genetic and epigenetic makeup, apart from the pathophysiological conditions. The continuously and gradually evolving disciplines of genomics about nutrition have elucidated the importance of genetic variations, epigenetic information, and expression of myriads of genes in disease progression apart from the involvement in modulating therapeutic responses. Further, the investigations have presented the considerable role of gut microbiota comprising of commensal and symbionts performing innumerable activities such as release of bioactive molecules, defense against pathogenic microbes, and regulation of immunity. Noteworthy, the characteristics of the microbiome change depending on host attributes, environmental factors, and habitat, in addition to diet, and therefore can be employed as a biomarker to unravel the response to given food. The specific diet and the components thereof can be suggested for supporting the enrichment of the desired microbial community to some extent as an important part of precision nutrition to achieve not only the goal of human health but also of healthy aging.
... Moreover, another mechanism of action of microbiota is represented by the capability of GM metabolites to interact with epigenomic mechanisms [28]. Dietary fibers undergo saccharolytic fermentation with subsequent production of short/chain fatty acids (SCFAs), such as butyrate, propionate, and acetate. ...
Article
Full-text available
The exposome encompasses all endogenous and exogenous exposure individuals encounter throughout their lives, including biological, chemical, physical, psychological, relational, and socioeconomic factors. It examines the duration and intensity of these types of exposure and their complex interactions over time. This interdisciplinary approach involves various scientific disciplines, particularly toxicology, to understand the long-term effects of toxic exposure on health. Factors like air pollution, racial background, and socioeconomic status significantly contribute to diseases such as metabolic, cardiovascular, neurodegenerative diseases, infertility, and cancer. Advanced analytical methods measure contaminants in biofluids, food, air, water, and soil, but often overlook the cumulative risk of multiple chemicals. An exposome analysis necessitates sophisticated tools and methodologies to understand health interactions and integrate findings into precision medicine for better disease diagnosis and treatment. Chronic exposure to environmental and biological stimuli can lead to persistent low-grade inflammation, which is a key factor in chronic non-communicable diseases (NCDs), such as obesity, cardiometabolic disorders, cancer, respiratory diseases, autoimmune conditions, and depression. These NCDs are influenced by smoking, unhealthy diets, physical inactivity , and alcohol abuse, all shaped by genetic, environmental, and social factors. Dietary patterns, especially ultra-processed foods, can exacerbate inflammation and alter gut microbiota. This study investigates the exposome's role in the prevention, development, and progression of NCDs, focusing on endogenous and exogenous factors.
... Se ha demostrado que el asesoramiento basado en la nutrigenómica tiene un impacto positivo en las actitudes de los pacientes. Un proyecto fundado por la Unión Europea denominado "Food4Me", es el primer ensayo controlado aleatorizado multicéntrico realizado a gran escala, cuyo enfoque se basa en la nutrición personalizada a través de la web, su objetivo fue determinar si el asesoramiento dietético personalizado (basado en el genotipo y fenotipo individual) tiene mayores beneficios en la alimentación y salud en comparación con el asesoramiento convencional basado en recomendaciones generales a la población (36,37); un estudio ECA con 683 participantes mostró una mayor reducción del peso corporal y de la circunferencia de la cintura en los portadores del gen de riesgo asociado a la masa grasa y la obesidad (FTO) cuando se informó a los participantes que eran portadores del alelo de riesgo FTO comparado con los no portadores del gen de riesgo (37). ...
Article
Full-text available
Introducción. Esta revisión bibliográfica explora los avances en genómica nutricional y su aplicación en la nutrición personalizada para abordar la obesidad. Considerando la complejidad del genoma humano y las susceptibilidades genéticas a enfermedades como la obesidad, la nutrición personalizada emerge como una solución prometedora para esta enfermedad multifactorial que impacta la salud pública global. Objetivo. El objetivo del estudio es identificar los nutracéuticos más efectivos en la nutrición personalizada para mitigar o tratar la obesidad, mediante ajustes dietéticos específicos basados en la genética individual. Metodología. Se llevó a cabo una revisión no sistemática de la literatura científica publicada entre 2019 y 2023, se seleccionó estudios según criterios de relevancia, accesibilidad y actualidad. De 82 artículos evaluados, 40 cumplieron con los criterios de inclusión. Resultados. Los estudios seleccionados resaltan la relevancia de compuestos como quercetina, curcumina, resveratrol y antocianinas en la modulación de la respuesta genética y metabólica ante la obesidad. Estos nutracéuticos han probado ser efectivos en reducir la adiposidad y las inflamaciones relacionadas. Discusión. La nutrigenética y nutrigenómica son esenciales para personalizar la nutrición, ya que la variabilidad genética individual afecta significativamente la respuesta a diversas dietas y tratamientos, lo que justifica un enfoque personalizado que considere el genotipo específico del individuo. Conclusión. Integrar nutracéuticos en la dieta basándose en el perfil genético puede ser clave para el manejo y tratamiento efectivo de la obesidad. La investigación apunta hacia un cambio a la nutrición personalizada, subrayando la necesidad de más estudios y su implementación en prácticas clínicas y dietéticas.
Book
Full-text available
Genetic research in the field of sports sciences offers new perspectives on various crucial aspects, including the determination of individual performance, predisposition to sports injuries, and the personalization of nutrition strategies. Sports Paradigms VIII (Genetic Research in Sports) comprehensively examines the impact of genetics on sports performance, presenting scientific findings aimed at maximizing athletes' physical and mental capacities. This book provides an in-depth exploration of key topics such as the importance of genetics in sports performance, genetic predisposition to sports injuries, muscle fiber types and genetics, genetics and nutrition, psychological resilience and genetics, as well as the effects of genetics on doping in sports. The chapters, prepared by expert researchers in the field, serve as a valuable resource for both the academic community and sports professionals. We hope that this book, which presents the latest scientific advancements in sports genetics, will contribute to research aimed at enhancing athlete health and performance.
Article
Full-text available
In this article, we analyze qualitatively the understanding of and reactions to personalized nutrition (PN) among the French public. Focus groups were conducted to identify the opinions and discourses about two applications of knowledge from nutritional (epi)genomics: a biotechnology (nutrigenetic testing) and a public awareness campaign (the “first thousand days of life” initiative). Our objective was to understand to what extent PN could lead to changes in eating practices as well as in the representations of food–health relationships within France, a country characterized by a strong commitment to commensality and a certain “nutritional relativism.” Although discourses on nutritional genomics testify to a resistance to food medicalization, nutritional epigenomics appears as more performative because it introduces the question of transgenerational transmission, thus parental responsibility.
Article
Full-text available
The increasing prevalence in polygenic diseases, such as obesity, cardiovascular disease, and type 2 diabetes, observed over the past few decades is more likely linked to a rapid transition in lifestyle rather than to changes in the sequence of the nuclear genome. In the new era of precision medicine, nutritional genomics holds the promise to be translated into tailored nutritional strategies to prevent and manage polygenic diseases more effectively. Nutritional genomics aims to prevent, treat, and manage polygenic diseases through targeted therapies formulated from individuals' genetic makeup and dietary intake. Direct-to-consumer genetic testing (DTC-GT) has become commercially available to equip individuals with information on their genetic vulnerability to different diseases. This information may potentially prompt behavioral changes against adverse factors. However, scientific evidence behind the clinical recommendations is a matter of continuous debate, and behavioral modifications after disclosing genetic information remain inconclusive. In this review, we provide an overview of nutritional genomics and related nutritional DTC-GT services and discuss whether available data are sufficient to be translated into clinical recommendations and public health initiatives. Overall, the scientific evidence supporting the dissemination of genomic information for nutrigenomic purposes remains sparse. Therefore, additional knowledge needs to be generated, particularly for polygenic traits.
Article
Full-text available
Background: Studying the impact of genetic testing interventions on lifestyle behaviour change has been a priority area of research in recent years. Substantial heterogeneity exists in the results and conclusions of this literature, which has yet to be explained using validated behaviour change theory and an assessment of the quality of genetic interventions. The theory of planned behaviour (TPB) helps to explain key contributors to behaviour change. It has been hypothesized that personalization could be added to this theory to help predict changes in health behaviours. Purpose: This systematic review provides a detailed, comprehensive identification, assessment, and summary of primary research articles pertaining to lifestyle behaviour change (nutrition, physical activity, sleep, and smoking) resulting from genetic testing interventions. The present review further aims to provide in-depth analyses of studies conducted to date within the context of the TPB and the quality of genetic interventions provided to participants while aiming to determine whether or not genetic testing facilitates changes in lifestyle habits. This review is timely in light of a recently published "call-to-action" paper, highlighting the need to incorporate the TPB into personalized healthcare behaviour change research. Methods: Three bibliographic databases, one key website, and article reference lists were searched for relevant primary research articles. The PRISMA Flow Diagram and PRISMA Checklist were used to guide the search strategy and manuscript preparation. Out of 32,783 titles retrieved, 26 studies met the inclusion criteria. Three quality assessments were conducted and included: (1) risk of bias, (2) quality of genetic interventions, and (3) consideration of theoretical underpinnings - primarily the TPB. Results: Risk of bias in studies was overall rated to be "fair." Consideration of the TPB was "poor," with no study making reference to this validated theory. While some studies (n = 11; 42%) made reference to other behaviour change theories, these theories were generally mentioned briefly, and were not thoroughly incorporated into the study design or analyses. The genetic interventions provided to participants were overall of "poor" quality. However, a separate analysis of studies using controlled intervention research methods demonstrated the use of higher-quality genetic interventions (overall rated to be "fair"). The provision of actionable recommendations informed by genetic testing was more likely to facilitate behaviour change than the provision of genetic information without actionable lifestyle recommendations. Several studies of good quality demonstrated changes in lifestyle habits arising from the provision of genetic interventions. The most promising lifestyle changes were changes in nutrition. Conclusions: It is possible to facilitate behaviour change using genetic testing as the catalyst. Future research should ensure that high-quality genetic interventions are provided to participants, and should consider validated theories such as the TPB in their study design and analyses. Further recommendations for future research are provided.
Article
Full-text available
Mediterranean Diet (MedDiet) adherence has been proven to produce numerous health benefits. In addition, nutrigenetic studies have explained some individual variations in the response to specific dietary patterns. The present research aimed to explore associations and potential interactions between MedDiet adherence and genetic background throughout the Food4Me web-based nutritional intervention. Dietary, anthropometrical and biochemical data from volunteers of the Food4Me study were collected at baseline and after 6 months. Several genetic variants related to metabolic risk features were also analysed. A Genetic Risk Score (GRS) was derived from risk alleles and a Mediterranean Diet Score (MDS), based on validated food intake data, was estimated. At baseline, there were no interactions between GRS and MDS categories for metabolic traits. Linear mixed model repeated measures analyses showed a significantly greater decrease in total cholesterol in participants with a low GRS after a 6-month period, compared to those with a high GRS. Meanwhile, a high baseline MDS was associated with greater decreases in Body Mass Index (BMI), waist circumference and glucose. There also was a significant interaction between GRS and the MedDiet after the follow-up period. Among subjects with a high GRS, those with a high MDS evidenced a highly significant reduction in total carotenoids, while among those with a low GRS, there was no difference associated with MDS levels. These results suggest that a higher MedDiet adherence induces beneficial effects on metabolic outcomes, which can be affected by the genetic background in some specific markers.
Article
Full-text available
Aging is a multifactorial process characterized by the progressive loss of physiological functions, leading to an increased vulnerability to age-associated diseases and finally to death. Several theories have been proposed to explain the nature of aging. One of the most known identifies the free radicals produced by the mitochondrial metabolism as the cause of cellular and DNA damage. However, there are also several evidences supporting that epigenetic modifications, such as DNA methylation, noncoding RNAs, and histone modifications, play a critical role in the molecular mechanism of aging. In this review, we explore the significance of these findings and argue how the interlinked effects of oxidative stress and epigenetics can explain the cause of age-related declines.
Article
Background: Phthalic acid esters are ubiquitous and antiandrogenic, and may cause systemic effects in humans, particularly with in utero exposure. Epigenetic modification, such as DNA methylation, has been hypothesized to be an important mechanism that mediates certain biological processes and pathogenic effects of in utero phthalate exposure. Objective: The aim of this study was to examine the association between genome-wide DNA methylation at birth and prenatal exposure to phthalate. Methods: We studied 64 infant-mother pairs included in TMICS (Taiwan Maternal and Infant Cohort Study), a long-term follow-up birth cohort from the general population. DNA methylation levels at more than 450,000 CpG sites were measured in cord blood samples using Illumina Infinium HumanMethylation450 BeadChips. The concentrations of three metabolites of di-(2-ethylhexyl) phthalate (DEHP) were measured using liquid chromatography tandem-mass spectrometry (LC-MS/MS) in urine samples collected from the pregnant women during 28-36 weeks gestation. Results: We identified 25 CpG sites whose methylation levels in cord blood were significantly correlated with prenatal DEHP exposure using a false discovery rate (FDR) of 5% (q-value < 0.05). Via gene-set enrichment analysis (GSEA), we also found that there was significant enrichment of genes involved in the androgen response, estrogen response, and spermatogenesis within those genes showing DNA methylation changes in response to exposure. Specifically, PA2G4, HMGCR, and XRCC6 genes were involved in genes in response to androgen. Conclusions: Phthalate exposure in utero may cause significant alterations in the DNA methylation in cord blood. These changes in DNA methylation might serve as biomarkers of maternal exposure to phthalate in infancy and potential candidates for studying mechanisms via which phthalate may impact on health in later life. Future investigations are warranted.
Article
Aim: To identify and profile training courses available to dietitians and nutritionists in the area of nutritional genomics. Genetic technology is progressing quickly, leading to increased public interest and requests from the public for personalised nutrition advice based on genetic background. Tertiary courses often lack specific curriculum in nutritional genomics, preventing graduates from discussing confidently with their clients the relationships between genetics, nutrition and health. This has increased the demand for professional development in this field. Methods: The search strategy was intended to replicate real-life practice. Google and snowball searches were conducted using terms related to education and nutritional genomics. Results included online or face-to-face courses in any country providing content on nutritional genomics. One-off courses and those courses no longer accessible were excluded. A descriptive analysis of characteristics of courses was undertaken, reporting on mode of delivery, cost, duration, content, qualification awarded, target audience and affiliations. Results: In total, 37 courses varying in duration, content and cost were identified: 4 postgraduate university degrees, 5 university course units, 4 recurring face-to-face workshops, 15 online short courses, 8 pre-recorded presentations and 1 service offering regular live webinars. Affiliations with food and pharmaceutical industry (e.g. genetic testing companies), professional organisations and research/education institutes were observed. Conclusions: Training courses identified were predominantly delivered online, enabling nutrition professionals worldwide to upskill in nutritional genomics and personalised nutrition. Additional courses exist. Those seeking training should scrutinise and compare cost, duration, mode, content and affiliations of course providers to ensure learning needs are met.
Book
This book examines the toxicological and health implications of environmental epigenetics and provides knowledge through an interdisciplinary approach. Included in this volume are chapters outlining various environmental risk factors such as phthalates and dietary components, life states such as pregnancy and ageing, hormonal and metabolic considerations, and specific disease risks such as cancer cardiovascular diseases, and other non-communicable diseases. Environmental Epigenetics imparts integrative knowledge of the science of epigenetics and the issues raised in environmental epidemiology. This book is intended to serve both as a reference compendium on environmental epigenetics for scientists in academia, industry, and laboratories and as a textbook for graduate level environmental health courses.
Article
Background: Exposure to persistent organic pollutants (POPs) has been associated with epigenetic changes such as DNA methylation, which can influence human health. However, the association between POPs and DNA methylation by sex was not shown in previous studies. Objectives: We investigated the association between POPs and DNA methylation in men and women using a larger population. Methods: A cross-sectional study was conducted using the data of 444 Koreans (253 men and 191 women). Measurements for sixteen different POPs, including six organochlorine pesticides (OCPs) and ten polychlorinated biphenyls (PCBs) were taken in serum. DNA methylation via Alu and LINE-1 in serum was measured by pyrosequencing. To evaluate the association between POPs and DNA methylation, the Pearson's correlation and multiple linear regression analyses were performed. Results: Except for PCB52 and PCB101, we found significant inverse associations between p,p'-DDE, cis-Heptachlor epoxide, and PCBs and Alu assay in men after adjusting for age, BMI, smoking status, and alcohol consumption (β = -0.67 for p,p'-DDE; -0.28 for cis-Heptachlor epoxide; in the range -0.43 to -1.60 for PCBs). In women, PCB153 and PCB180 showed statistically significant inverse association with Alu assay (β = -0.22 for PCB153; -0.22 for PCB180). Except for PCB101, p,p'-DDE and PCBs were positively associated with LINE-1 assay in women (β = 0.48 for p,p'-DDE; in the range 0.40-0.89 for PCBs) while p,p'-DDE, PCB153, and PCB180 showed positive associations with LINE-1 assay in men (β = 0.55 for p,p'-DDE; 0.65 for PCB153; 1.02 for PCB180). Conclusions: We found that several POPs were associated with global DNA hypomethylation in the Alu assay for men and global DNA hypermethylation in the LINE-1 assay for women.