Sei sulla pagina 1di 9

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/51865714

Human nutrigenomics of gene regulation by


dietary fatty acids
Article in Progress in lipid research December 2011
DOI: 10.1016/j.plipres.2011.11.005 Source: PubMed

CITATIONS

READS

29

274

2 authors:
Lydia A Afman

Michael Muller

Wageningen University & Research

University of East Anglia

53 PUBLICATIONS 1,562 CITATIONS

460 PUBLICATIONS 14,830 CITATIONS

SEE PROFILE

SEE PROFILE

All content following this page was uploaded by Michael Muller on 26 December 2016.
The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document
and are linked to publications on ResearchGate, letting you access and read them immediately.

Progress in Lipid Research 51 (2012) 6370

Contents lists available at SciVerse ScienceDirect

Progress in Lipid Research


journal homepage: www.elsevier.com/locate/plipres

Review

Human nutrigenomics of gene regulation by dietary fatty acids


Lydia A. Afman , Michael Mller
Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands
Netherlands Nutrigenomics Centre, TI Food and Nutrition, Wageningen, The Netherlands

a r t i c l e

i n f o

Article history:
Available online 2 December 2011
Keywords:
Nutrigenomics
Fatty acids
Transcriptomics
Gene expression
Peripheral blood mononuclear cells (PBMCs)

a b s t r a c t
Nutrigenomics employs high-throughput genomics technologies to unravel how nutrients modulate gene
and protein expression and ultimately inuence cellular and organism metabolism. The most oftenapplied genomics technique so far is transcriptomics, which allows quantifying genome-wide changes
in gene expression of thousands of genes at the same time in one sample. The performance of gene
expression quantication requires sufcient high-quality homogenous cellular material, therefore
research in healthy volunteers is restricted to biopsies from easy accessible tissues such as subcutaneous
adipose tissue, skeletal muscle and intestinal biopsies or even more easily accessible cells such as peripheral blood mononuclear cells from blood. There is now signicant evidence that fatty acids, in particular
unsaturated fatty acids, exert many of their effects through modulation of gene transcription by regulating the activity of numerous transcription factors, including nuclear receptors such as peroxisome proliferator activated receptors, liver X receptor and sterol regulatory binding proteins. This review evaluates
the human nutrigenomics studies performed on dietary fat since the initiation of nutrigenomics research
around 10 years ago. Although the number of studies is still limited, all studies clearly suggest that
changes in dietary fatty acids intake and composition can have a signicant impact on cellular adaptive
response capacity by gene transcription changes in humans. This adds important knowledge to our
understanding of the strong effects that various fatty acids can have on numerous metabolic and inammatory pathways, signaling routes and homeostatic control in the cell and ultimately on whole body
health. It is important to use and integrate nutrigenomics in all future nutrition studies to build up the
necessary framework for evidence-based nutrition in near future.
2011 Elsevier Ltd. All rights reserved.

Contents
1.
2.
3.
4.

5.
6.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Challenges in human nutrigenomics studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Transcriptional gene regulation by (dietary) fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Application of transcriptomics in human intervention studies with dietary fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
4.1.
Postprandial intervention studies with dietary fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
4.2.
Longer-term intervention studies with dietary fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
4.3.
Overlap between long term and postprandial effects of n 3 PUFA on PBMC transcriptome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Future challenges in nutrigenomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Conclusions and perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

1. Introduction
Human nutrition research is still largely focused on issues related
to nutrient deciencies and impairment of health and disease due to
Corresponding author. Address: Nutrition, Metabolism and Genomics Group,
Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD
Wageningen, The Netherlands. Tel.: +31 (0)317 485789; fax: +31 (0)317 483342.
E-mail addresses: lydia.afman@wur.nl (L.A. Afman), michael.muller@wur.nl
(M. Mller).
0163-7827/$ - see front matter 2011 Elsevier Ltd. All rights reserved.
doi:10.1016/j.plipres.2011.11.005

63
64
64
65
65
66
66
67
68
68

inappropriate dietary patterns. The last decade has revealed an


enormous wealth of insights in the complexity of human physiology
and in particular of pathophysiology of complex diseases. At the
same time we have to acknowledge a worldwide epidemic increase
in preventable diseases such as metabolic syndrome or diabetes
type 2 that are linked to obesity and largely related to higher energy
intake and reduced energy expenditure [1]. On the other hand new
high-throughput genomics technologies for the generation, processing, and analysis of biological data about the composition and

64

L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370

functions of genomes has created unprecedented opportunities for


increasing our understanding of how nutrients modulate gene and
protein expression and ultimately inuence cellular and organ
metabolism. To overcome the signicant gap between molecular
biology, physiology and more traditional nutrition research, nutrigenomics has been initiated as a new innovative strategy around
10 years ago when a working draft of the human genome was announced. There are several meanings of nutrigenomics as used in
the scientic literature mostly focusing on gene-environment interactions and the relevance of genotype changes e.g. single nucleotide
polymorphism (SNPs) for individual variation in responses to foods,
dietary pattern and susceptibility to develop nutrition-related diseases [2,3]. We have extent this denition of nutrigenomics by
including innovative combinations of molecular nutrition research
and genomics applications, largely performed in transgenic animal
models. The diverse tissue and organ-specic effects of bioactive
dietary components include gene expression patterns (transcriptome), organization and modication of the chromatin structure
(epigenome), protein expression patterns including post-translational modications (proteome) as well as metabolite proles
(metabolome) [46]. Nutrigenomics promotes an increased understanding on how nutrition inuences metabolic pathways and
homeostatic control by providing mechanistic insights into how
nutrition works (evidence-based nutrition). Furthermore it reveals
how this regulation is disturbed in the early phases of diet-related
disease, and the extent to which individual sensitizing genotypes
contribute to such diseases by allowing comprehensive and deep
phenotyping and the identication of early biomarkers for pre-disease states [79]. Nutrigenomics is the approach that generates
the knowledge that allows development of new nutritional strategies to improve peoples health and tness according to their individual needs. This necessary evolution will eventually lead to
evidence-based dietary intervention strategies for restoring health
and tness and for preventing diet-related disease.
2. Challenges in human nutrigenomics studies
Since the introduction of genomics techniques such as transcriptomics many human studies have been performed to examine
phenotypic differences between diseased and healthy states with
the ultimate goal to improve early diagnostic and to increase our
understanding of underlying mechanisms of early disease pathology or to investigate pharmacological efcacy to modulate disease
progression. Recently these techniques are also applied to achieve
more knowledge on metabolic, physiological or dietary changes on
alterations in transcriptional gene regulation in humans.
Several types of intervention studies can be performed in which
effects of specic nutrients (e.g. n 3 PUFA versus saturated fat) or
changes in more complex dietary patterns (Mediterranean versus
Western-style diets) can be examined on the levels of gene expression proles. The length of the study design can vary from a few
hours in postprandial studies, to weeks and years in respectively
completely controlled dietary intervention studies and long-term
interventions trials. It is expected that postprandial studies mirror
the more acute effects of daily consumption whereas long-term
studies will reect the extended inuence of nutrition on whole
body health. All types of studies, if well controlled, are of pivotal
importance to increase our knowledge and understanding on
how nutrients are sensed by our organ cells and how this translates
efciently into necessary adaptive physiological response mechanism as essential for health, both on the short and long run.
The major bottleneck in studying these kind of often-subtle
changes is the limited accessibility of tissues as study populations
mostly consist of healthy volunteers. Whereas in ill subjects that
need to undergo surgery biopsies can be more easily obtained from
both pathological tissue and more normal surrounding tissue

areas, in patients without necessity surgery such as diabetes


patients and especially in healthy volunteers, collection of samples
from internal organs such as liver, pancreas and visceral adipose
tissue is impossible because of ethical reasons. Research in those
volunteers, is therefore restricted to biopsies from more easily
accessible tissues such as subcutaneous adipose tissue, muscle
and intestinal biopsies without a high health risk for the volunteer.
Although other metabolic organs such as liver and visceral fat, are
not assessable for invasive investigations, comprehensive phenotyping of these organs in healthy and early pathophysiological
stages in the development of diseases would be extremely valuable. These metabolic organs fulll key physiological functions in
the efcient regulation of homeostasis related to dietary fat metabolism and play a key role in the early development of nutrition-related diseases such as metabolic syndrome and type II diabetes as
shown to large extent in animal studies [1016]. The development
of non-alcoholic fatty liver disease or non-alcoholic steatohepatitis
[15,16]; is a good example for this as a more recently frequently
observed human disease phenotype that is strongly linked to metabolic syndrome and both may even be two denitions of the same
problem (chronic metabolic stress) with ultimately the development of diabetes type 2 and cardiovascular health problems as
consequences.
To allow more efcient prevention of these diseases by evidence-based nutritional and lifestyle intervention a comprehensive
understanding of the various phases (from organ dysfunction to
systemic pathophysiology) in the disease evolution is key. Therefore the study of time-dependent transitions from the healthy to
disease state and the role that nutrition plays in these time-dependent changes of organ phenotypes would be essential but is currently not feasible because of the invasive procedure, the low
tissue and RNA yield and the presence of numerous cell types in
the biopsy. A far less invasive method is the collection of blood cells
out of whole blood obtained via venipuncture. Blood cells travel
through the whole body, are exposed to all physiological changes
including changes in nutrient levels before and after meals or in
the fasting state. They are key cells in regulating organ-specic
and systemic immune function and inammatory responses. Nonresolving inammation is a major driver of disease, e.g. blood cells
can attached to and invade in the vascular wall leading to foam cells
formation and ultimately atherosclerosis [17]. The most studied
blood cells are the peripheral blood mononuclear cells (PBMCs) that
consist of monocytes and lymphocytes. Whole genome transcriptome analyzes of PBMCs have been applied successfully in discriminating disease from healthy populations and in differentiating
several phases/diagnostics of disease [18,19]. It has also been
shown that variation in PBMC gene expression within persons is
smaller than the variation between persons [2022]. This smaller
within person variation combined with the usual applied measurement of baseline and end samples within a person in nutritional
intervention studies enables potential successful application of
PBMC gene expression proling in nutritional intervention studies.
3. Transcriptional gene regulation by (dietary) fatty acids
Dietary fatty acids inuence human health in numerous ways
and inuence several indicators of health status. In recent years
much work has focused to elucidate more precisely the mode of action how dietary fatty acids lead to short-and long-term changes in
cellular functions. There is now considerable evidence that fatty
acids, in particular unsaturated fatty acids, exert many of their biological effects through modulation of gene transcription by regulating the activity of numerous transcription factors, including
sterol regulatory binding proteins (SREBPs) or nuclear receptors
such as peroxisome proliferator activated receptors (PPARs) and
liver X receptors (LXRs).

L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370

Three different highly conserved PPAR isotypes have been identied: PPARa (NR1C1), PPARb/r (NR1C2) and PPARc (NR1C3).
They act as nutrient sensors for fatty acids, but bind also numerous
fatty acid derivatives and compounds showing structural resemblance to fatty acids and inuence the expression of specic genes
[23,24] The PPARa isotype has been shown to govern expression of
numerous genes involved in fatty acid oxidation, ketogenesis,
gluconeogenesis, cholesterol catabolism and lipoprotein metabolism as mainly studied using mouse models. More recent evidence
indicates that hepatic PPARa is not largely activated by plasma free
fatty acids as high during fasting, whereas it can be activated by
dietary fatty acids and fatty acids generated via de novo lipogenesis [25]. Recently, it was also shown that the effects of dietary
unsaturated fatty acids on hepatic gene expression are almost
exclusively mediated by PPARa and mimic the effect of synthetic
PPARa agonists [26]. Much less is known about the relevance of
these mainly mouse-based ndings for human physiology. A recent
analysis provided new insights into the mechanisms and impact of
transcriptional regulation by PPARa in human liver by performing
chromatin immunoprecipitation (ChIP)-chip in combination with
transcriptional proling on HepG2 human hepatoma cells treated
with the specic PPARa agonist GW7647. Several genes known
to be regulated by PPARa showed GW7647-induced PPARa binding to their promoter. Interestingly also certain SREBP-target genes
exhibited PPARa binding, suggesting cross-talk between PPARa
and SREBP signaling [27]. Another study compared the function
of PPARa in mouse and human hepatocytes via analysis of target
gene regulation. Minor overlap was observed between the
PPARa-regulated genes from mouse and human by a gene-by-gene
comparison, although more substantial overlap was observed at
the pathway level. This study clearly suggest that PPARa activation
has a major impact on gene regulation in human hepatocytes and
that the role of PPARa as master regulator of hepatic lipid metabolism is generally well-conserved between mouse and human [28].
More in depth studies are required to elucidate the specic role of
nutrient-sensing transcription factors in mediating the effects of
dietary fatty acids in other human cells including small intestinal
epithelial cells, macrophages or adipocytes.
LXRs, closely PPAR-related members of the nuclear receptor family, are important regulators of cholesterol, fatty acid, glucose
homeostasis and play an essential role at the interface between
metabolism and inammation [29]. Their endogenous activators
are oxysterols and other derivatives of cholesterol metabolism
(22(R)-hydroxycholesterol, 24(S)-hydroxycholesterol, 24(S), 25epoxycholesterol). LXRa is highly expressed in the liver but is also
found in kidney, intestine, adipose tissue, and macrophages. LXRb
is expressed ubiquitously[30]. Nutritional or pharmacological modulation of the activity of these ligand-activated transcription factors
have been shown to inuence the development of metabolic disorders such as hyperlipidemia and atherosclerosis [30]. Mice without
functional LXRa e.g. are healthy when fed with a low-cholesterol
diet but when fed a high-cholesterol diet these LXRa knockout mice
develop enlarged fatty livers, less functional hepatocytes, high cholesterol levels in liver, and impaired liver function [31]. LXR is a major regulator of fatty acid synthesis and the expression of other
lipogenic genes through direct interaction with their promoters as
well as inducing sterol regulatory element binding protein-1c
(SREBP-1c) gene transcription. Activation of LXR by oxysterols (related to high dietary cholesterol) or other agonists elevates nuclear
content of SREBP-1 and increases expression of the enzymes involved in de novo lipogenesis [32].
SREBPs are transcription factors of the helix-loop-helix family
and important regulators of cholesterol and fatty acid metabolism.
SREBPs are encoded by two genes, SREBP-1 and SREBP-2 resulting
in 3 proteins SREBP-1a, SREBP-1c and SREBP-2 (for more information in particular on the complex mode of activation of these

65

transcription factors see Desvergne et al. [30,33]. Insulin induces


SREBP-1 nuclear which induces lipogenesis, MUFA and triglyceride
synthesis and storage [34]. The n 3 PUFA docosahexaneoic acid
(DHA) is a very strong nutritional suppressor of nuclear SREBP-1
expression of genes involved in lipogenesis and MUFA synthesis
in liver [35].
4. Application of transcriptomics in human intervention studies
with dietary fatty acids
4.1. Postprandial intervention studies with dietary fatty acids
Due to an easy accessible surplus of large processed food (rich in
isolated sugars and saturated fats) we are in the postprandial state
the greater part of the day in particular if increased energy
consumption by e.g. exercise rarely allows to compensate. A better
understanding of meal effects on body function and health are of
critical importance. It is known that a meal can provoke a transient
inammatory response which is low compared to a general inammatory response [36]. In addition, postprandial effects of high fat
meals have been observed on vascular reactivity [37] and on blood
cell activation [38]. The arrival of comprehensive genomics techniques such as transcriptomics enables the unbiased measurement
of total postprandial genome-wide effects on the cellular level. Recently, we assessed postprandial transcriptional responses to high
fat shakes varying in types of fatty acids [39]. Twenty healthy subjects consumed a butterfat-enriched saturated fatty acid (SFA)
shake and a DHA-enriched polyunsaturated fatty acid (PUFA) shake
in random order and whole genome PBMCs gene expression proles were assessed. Postprandial gene expression responses were
distinctly different between shakes as illustrated by the higher
number of genes changed upon PUFA shake consumption. It is expected that adaptive cellular response by means of gene expression
changes will only occur when cellular capacity is insufcient to deal
efciently with the high load of intracellular free fatty acids. High
concentrations of n 3 PUFA are more stress-inducing molecules
than SFA for blood cells that need to activate adaptive gene transcription program to be able to cope with the high levels of this
stressors by rapid processing of n 3 PUFA and increased cellular
protection against e.g. oxidative stress. The increased expression
of genes involved in cellular stress response upon n 3 PUFA shake
consumption points clearly in this direction. Other differences between the SFA and n 3 PUFA shake could mainly be attributed
to expression alterations of genes involved in lipid metabolism with
opposite effects for the two shakes. SFA caused a postprandial upregulation in transcription of ABCA1, ABCG1, and SREBP1, whereas
an overload of n 3 PUFA resulted in a down-regulation in expression of the same genes. Both effects were conrmed by ex vivo incubation of PBMCs with palmitic acid and the DHA, pointing to an
effect likely induced by the fatty acids itself. Interestingly, a whole
genome gene expression study in a population of around 1500 individuals showed that gene expression of ABCA1 and, ABCG1 was negatively associated with TG levels and, SCD gene expression was
found to be negatively associated with HDL-cholesterol [40]. Another study also observed a postprandial increase in expression of
LXR-related genes in PBMCs after a high fat load. In this study the
postprandial response upon a high fat load consisting of a mix of
fatty acids in 22 patients with metabolic syndrome was examined
and similar to our SFA shake the gene expression of SREBP1 and
LXRa was found to be increased. [41]. Several mice and in vitro studies have examined the effects on n 3 PUFAs on hepatic gene regulation and showed that DHA is the most potent hepatic fatty acid
suppressor of SREBP1 gene expression [35]. Less studies have
explored the effects of n 3 PUFAs on monocytes/macrophages
but in vitro studies in macrophage cell lines showed down-regulation of the LXR target genes ABCA1 and ABCG1 upon n 3 PUFA

66

L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370

incubation[4244]. As these LXR-target genes play a major role in


modulating cholesterol efux from macrophages, the n 3 PUFAreduced postprandial expression of these genes in PBMCs in
humans may lead to an inhibition of cellular cholesterol efux. Conversely, the increased expression of LXR and its target genes after
SFA or mixed fatty acid consumption might result in an increased
postprandial cholesterol efux. Interestingly, daily consumption
of a lower dose of n 3 PUFAs for a longer period did not result
in a reduced expression of these genes when determined in the fasting state [45]. This data combined with the nding that levels of
LXR-related gene expression where heading back to baseline 8 h
postprandially, suggests that the effects observed in PBMCs might
be temporarily and specic for the postprandial situation when
blood lipid concentrations are elevated [39]. Still, the presence of
the postprandial state the greater part of the day may lead to an
continuous trigger of which the consequences on gene expression
and cholesterol efux are yet unknown.
4.2. Longer-term intervention studies with dietary fatty acids
Daily changes in postprandial exposure will ultimately lead to
longer-term adaptations in the whole body. Daily exposure to specic fatty acids will activate adaptive transcriptional responses frequently, ultimately leading to functional alterations in activity of
pathways and networks in organs such as the small intestine, liver,
heart, skeletal muscle and adipose tissue. Longer-term consumption will in addition lead to incorporation of fatty acids into membranes of practical every cell in the body, which will have their
consequences as well.
To determine longer-term effects of fatty acids on the cellular
transcriptome we assessed changes in whole genome transcriptional proles in PBMCs and adipose tissue after long-term consumption of different fatty acids. The study comprised a 26 weeks
intervention with daily supplementation of 0.4 g EPA/DHA, 1.8 g
EPA/DHA or a control oil in an elderly population. Daily consumption
of 1.8 g n 3 PUFA resulted in changes in expression of 1000 genes
while expression of only 300 genes was changed in the control
group that consumed high oleic sunower oil. The larger part of
the changes in the n 3 PUFA could be attributed to a down-regulation in expression of immune related genes and genes known to be
involved in development of atherosclerosis. Additional changes
were related to an increased expression of cell-cycle genes and of
genes involved in transcriptional and translational regulation [45].
Another nutrigenomics study [46] explored the effects of two
months supplementation with 3 g EPA/DHA daily in 10 male volunteers on gene expression using pooled RNA of lymphocytes on a
macroarray with a selected gene set. Parallel to our results they observed a decrease in expression of cytokine-related genes, but contrary to our data a decrease in expression of cell cycle and
transcriptional regulation genes was observed. However, it must
be noted that this study used pooled samples for macroarray analyzes in which signicant inter-individual differences are impossible to determine and one must rely on fold change cut offs.
Furthermore, PBMCs consist of a combination of lymphocytes and
monocytes which likely will have inuenced the effects on transcription as well. Besides the use of arrays to study whole genome
gene transcription or a large set of genes simultaneously one study
examined the expression of a selection of genes in PBMCs in 27
healthy volunteers and found a reduction in gene expression of some
cytokines after 4 weeks supplementation with 775 mg EPA and borage oil (831 mg GLA) daily [47]. Although no control group consuming placebo oil with the same energy content was included in the
study and the changes in gene expression cannot solely be attributed
to EPA, the results point in the same direction as the previous two
studies. The question remains what a down-regulation in transcription of inammatory genes means for cellular functionality and

what the systemic implications are. In the study of Gorjao [46] besides pooled macroarrays, stimulation assays were performed on
the lymphocytes showing an increased production of IL-10, IFNgamma and TNFa and a decreased production of IL2. However, most
other studies observed a decrease in production of cytokines after
sh oil supplementation, which is more in line with the observed effect on transcriptional regulation [4851]. In addition, Meydani observed a higher reduction in cytokine response upon stimulation in
older compared to young women [50] after n 3 PUFA supplementation, underlining the potential age-dependency of responses.
Overall, long-term sh oil supplementation reduces expression
of leukocyte inammatory genes and decreases cytokine production upon leukocyte stimulation. The relevance for health outcome
of this reduced immune response has been postulated to be benecial in progression and treatment of inammatory diseases such
as rheumatoid arthritis [52,53].
Besides effects of consumption of fatty acids on transcriptional
activity in blood cells dietary fatty acid effects have also been determined on whole genome gene expression of adipose tissue. Within a
completely controlled randomized dietary intervention trial of
8 weeks the effects of different types of fatty acids was studied on
adipose tissue transcriptome. Participants either consumed a butter-derived saturated fatty acids (SFA)-rich diet or a rened olive
oilderived monounsaturated fatty acids (MUFA)-rich diet and adipose tissue biopsies were taken before and after intervention. Whole
genome microarray analyses of these samples revealed that the SFAdiet resulted in the most distinct changes in gene expression. Where
SFA-diet effected expression of around 1500 genes, MUFA consumption effected expression of 600 genes. The most remarkable observation was the increased transcription of genes involved in immune
function after consumption of the SFA-rich diet, which was
distinctly different from the effects on the MUFA diet, after which
a much smaller and more or less opposite effect was observed on
expression of immune-related genes. Interestingly, the SFA-induced
changes in gene expression were similar to differences observed in
gene expression in adipose tissue of obese versus lean subject
[54,55] and opposite to the changes observed after weight loss in obese subjects [56]. Keeping in mind that no weight changes were
present in the participants, consumption of the more healthy type
of dietary fat (unsaturated) may be of great relevance for healthy
functional adipose tissue in obesity. Although no changes in macrophage inltration were observed, prolonged consumption of a SFArich diet may result in inltration of macrophages as frequently perceived in obesity [57,58]. Some studies have shown that SFA may
activate immune related genes by binding and activation of Toll-like
receptor 4 [59,60]. In addition, saturated FA are poor ligands for
PPARy [61], and as PPARy activation result in a reduced inammation, a decreased activation of PPARy by SFA might lead to increased
inammation. PPARy gene expression in adipose tissue and expression of some target genes was reduced upon SFA consumption.
PPARc also plays an important role in alternative M2 macrophage
activation that is of key importance for adipose tissue remodeling,
inammatory status and health [62] as shown in numerous mouse
studies [63] and increasingly also in human studies [62]. Interestingly, in a recently performed mouse study we found that adipose
tissue function is essential for the prevention of non-alcoholic fatty
liver disease [12]. This again highlights the importance of the comprehensive understanding of nutrition-relevant inter-organ dependency and crosstalk as essential basis for a healthy body function.
Future nutrigenomics research should be leading in this effort.
4.3. Overlap between long term and postprandial effects of n
on PBMC transcriptome

3 PUFA

The above described studies show that fatty acids can not only
affect gene transcription directly after consumption but will also

L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370

induce long-term changes. Based on the pathway analyses it


appears that depending on the length of the intervention expression of other genes and pathways are affected. To reveal whether
overlapping genes in PBMCs were changed between postprandial
and long-term n 3 PUFA consumption we compared datasets of
the 26 weeks intervention and the postprandial study. The overlap
was relatively low; of the 900 genes specically changed by n 3
PUFA in the long-term study and the 291 genes in the postprandial
study, 29 genes were changed upon n 3 PUFA in both studies
(Fig. 1). The expression of the larger part of the genes i.e. 19 genes
are changed in the same direction and may reect the overall effects of sh oil consumption. The expression of the remaining 10
genes is changed in the opposite direction. One of the genes upregulated postprandial and down-regulated after long-term intervention was ADFP, a known PPARa target gene [64]. Other PPARa
target genes were down-regulated in the long-term intervention.
We hypothesize that daily intake of higher amounts of n 3 PUFA
as mentioned above induces whole body adaptation in particular in
metabolic organs such as the small intestine, liver or adipose tissue
with the consequence of lower FFA levels and consequently different changes in gene expression levels in PBMCs compared to acute
studies. The same may hold for JUNB, a stressor/inammatory gene
and the other genes in the list. The function of the overlap in genes
varies, but several genes are immune-related such as CLEC4E and
Il13RA1 or lipid-related such as SCD. G-protein coupled receptor
18 (GPR18) is one of the genes with the highest signicantly average increase upon both long-term and acute consumption. GPR18
is know to be highly expressed in lymphocytes and N-arachidonylglycine has been suggested to be an endogenous ligand of GPR18
[65], but further research concerning its function and the relationship to n 3 PUFA is needed. The analysis also reveals that transcriptional changes of a large number of genes are very specic
for either long-term or acute consumption of n 3 PUFA.
In conclusion, acute postprandial studies lead to short term direct effects of fatty acids on transcription whereas long-term intervention with daily consumption will lead to more systemic effects
that still can be monitored in PBMCs.

67

5. Future challenges in nutrigenomics


It is evident from the rst 10 years of nutrigenomics research and
applying comprehensive genomics techniques such as transcriptomics that researchers have to acknowledge relatively small changes
in gene expression induced by nutrients as compared to drugs
[26,66] with large inter-individual variations. This is not unexpected
considering the different ways drugs and nutrients work [4]. Where
drugs are designed to specically and constantly activate certain
receptors with the goal to strongly activate specic pathways, nutrients will induce a much milder activation of specic receptors and
consequently of specic pathways. Instead nutrients will lead to a
more balanced cellular response by activation of multiple control
systems with the main purpose to metabolize the nutrient and to
regulate metabolism to rapidly reach homeostatic balance. If studies
are performed under physiological conditions it consequently
means that rather low amounts of bioactive compounds are applied
and low numbers of signicantly changed genes and an lower induction in changes in gene expression can be expected as there is no
need for a stronger transcriptional response. On average fatty acids
induce effect sizes ranging from a few percent till 100% with individual outliers to 300% or 400% often for immune-related genes
[39,45,67]. Both points, the low fold changes and low proportion
of genes signicantly changed require a rigorous high-quality control system coupled to an array analysis platform with high quality
arrays, strict quality control criteria and the need for integrated bioinformatics and statistical procedures optimized for studies with
small effect sizes [68]. Moreover, it emphasizes the importance of
use of whole genome data and not only a biased selection and the
application of most recent well annotated pathway analyzes tools,
enabling the uncovering or enrichment of certain pathways or
signaling networks [69,70].
Another challenge is the great individual variations in gene
expression response to nutritional interventions largely due to
age, sex and genotype differences. As with other metabolites in plasma responders and non-responders to a dietary intervention can be
identied [64]. The challenge lies in the identication of subgroups

Fig. 1. Average gene expression changes of genes signicantly changed upon long term and acute consumption of omega3-FA. Each row represents a single gene; the rst
column represents the long-term study, the second column represents the acute study. The expression scale is log2-based and ranges from 6 0.3 (green) to P0.3 (red).

68

L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370

of responder-genes which is still complicated in the small populations studied, so far. This variance in response also emphasizes the
need for within person measurements and the relevance of crossover interventions or large intervention groups when using parallel
designs. The underlying reason for this response variance can be several and may vary from genetic and epigenetic variation to nutritional and life style factors. For example, imprinting during
pregnancy or during early years in life may affect phenotypic
responses at older age with the consequence of increased risk to develop nutrition-related diseases [71]. Nutritional habits may also be
a reason why persons will or will not respond to a change in a diet.
Nutrigenomics can be used to asses this. For instance, people that
regularly consume sh rich in n 3 PUFA will likely exhibit a less
pronounced transcriptional response upon a sh oil challenge than
subjects that do not eat sh at all. This also may be the scientic basis
for the nutritional advice to enjoy a healthy diet consisting of a varied food pattern as this will keep the homeostatic balance of our
organs most exible.
Many diseases are a result of disturbance of homeostasis or
homeostatic imbalance. As a hallmark of ageing every organism
will lose its efciency in its sensing and regulatory systems.
Diseases with a strong link to nutrition result from a homeostatic
imbalance such as obesity, metabolic syndrome or diabetes type
2. We increasingly understand the complex regulation of homeostatic control mechanisms on cellular, organ and system level that
should prevent this imbalance from occurring. However, in some
people, these mechanisms do not work efciently enough because
of signicant genetic susceptibility [1] and or epigenetic mechanism [71]. These people are much more vulnerable to develop a
disease in particular if the homeostatic control mechanisms are
chronically challenged by surplus of calories, non-ideal dietary patterns and lifestyles [72]. Because we are still not able to detect the
early warning signals of such homeostatic imbalance efciently
most often medical intervention is necessary at a very late stage
of the disease development that is often unable however to restore
the homeostatic balance to regain normal health with irreversible
damage of organs as a consequence.
To change this inefcient concept in treatment of lifestyle
diseases we have to use interventions that allow the quantication
of the homeostatic balance or resilience by assessing it after challenging or perturbing a homeostatic situation. A well known example for such as challenge is the oral glucose tolerance test (OGTT). A
standard 2 h OGTT is sufcient to diagnose or exclude forms of
diabetes mellitus at the various stages of development. Recently,
plasma metabolic proling combined with a glucose challenge
was used to differentiate between healthy individuals and individuals with an impaired glucose tolerance [73]. Application of metabolic perturbation with comprehensive nutrigenomics-based
metabolic proling may improve the diagnostic sensitivity to help
discriminating between healthy, less-healthy and unhealthy people.
Furthermore, it may allow to predict differences in the responses between treatments [74]. In addition, combination of dietary interventions with metabolic challenges magnies the difcult measurable
and relatively mild effects of nutritional alterations by testing the
cellular adaptive response capacity (for a more in depth discussion
[75,76]). This rather new concept of applying various challenges
on homeostatic control mechanisms in nutritional interventions is
still in its infancies and it is therefore necessary to carefully study
its value and limitations. One may expect important sex-dependent
differences in applying various homeostatic challenge-test as demonstrated recently for OGTT [77]. But ultimately it will be the most
promising strategy to combine nutrigenomics-based comprehensive phenotyping with systems perturbation interventions to allow
the precise assessment of individual health and nutritional needs.

6. Conclusions and perspective


In summary, well-controlled nutrigenomics studies on the regulation of gene transcription by dietary fatty acids in humans are
still limited partly because nutrigenomics is still a relative new
eld and not yet fully integrated in nutrition research and partly
because of still signicant costs of the genomics applications that
do not allow comprehensive phenotyping in large cohorts. The
studies that have been performed were largely of explorative nature with the purpose to identify whether transcriptional changes
can be detected after changes in dietary fatty acids consumption.
Although the number of studies is still limited, all studies clearly
suggest that changes in dietary fatty acids intake and composition
can have a signicant impact on cellular adaptive response capacity by gene transcription changes in humans. This adds important
knowledge to our understanding of the strong effects that various
fatty acids can have on numerous metabolic and inammatory
pathways, signaling routes and homeostatic control in the cell
and ultimately on whole body health.
Nutritional science is entering a new era with a shift away from
little science towards big science. The major nutritional problems facing the world today (e.g. obesity, diabetes and malnutrition) with huge impact on public and private parties cannot be
solved by little science, characterized by individualistic research,
lacking coordination and integration. Big science is needed, i.e.
multiple groups with complementary knowledge and expertise
working together in large national and international consortia similar to consortia as currently working on the human microbiome
project [78] or genome-wide association studies [79]. This new
era takes full advantage of modern, high resolution, comprehensive
and unbiased Genomics technologies, bio-banking, sophisticated
and partly web-based database management, providing a new
platform for scientic dialogue and exchange [80]. We dene this
new Nutritional Science as Nutritional Science 2.0.
References
[1] McCarthy MI. Genomics, type 2 diabetes, and obesity. N Engl J Med
2010;363:233950.
[2] Ordovas JM, Corella D. Nutritional genomics. Annu Rev Genomics Hum Genet
2004;5:71118.
[3] Kaput J, Rodriguez RL. Nutritional genomics: the next frontier in the
postgenomic era. Physiol Genomics 2004;16:16677.
[4] Muller M, Kersten S. Nutrigenomics: goals and strategies. Nat Rev Genet
2003;4:31522.
[5] Grayson M. Nutrigenomics. Nature 2010;468:S1.
[6] Masotti A, Da Sacco L, Bottazzo GF, Alisi A. Microarray technology: a promising
tool in nutrigenomics. Crit Rev Food Sci Nutr 2010;50:6938.
[7] Afman L, Muller M. Nutrigenomics: from molecular nutrition to prevention of
disease. J Am Diet Assoc 2006;106:56976.
[8] Kussmann M, Krause L, Siffert W. Nutrigenomics: where are we with genetic
and epigenetic markers for disposition and susceptibility? Nutr Rev
2010;68(Suppl. 1):S3847.
[9] Kussmann M, Panchaud A, Affolter M. Proteomics in nutrition: status quo and
outlook for biomarkers and bioactives. J Proteome Res 2010;9:487687.
[10] Kleemann R, van Erk M, Verschuren L, van den Hoek AM, Koek M, Wielinga PY,
et al. Time-resolved and tissue-specic systems analysis of the pathogenesis of
insulin resistance. PLoS One 2010;5:e8817.
[11] Radonjic M, de Haan JR, van Erk MJ, van Dijk KW, van den Berg SA, de Groot PJ,
et al. Genome-wide mRNA expression analysis of hepatic adaptation to highfat diets reveals switch from an inammatory to steatotic transcriptional
program. PLoS One 2009;4:e6646.
[12] Duval C, Thissen U, Keshtkar S, Accart B, Stienstra R, Boekschoten MV, et al.
Adipose tissue dysfunction signals progression of hepatic steatosis towards
nonalcoholic steatohepatitis in C57BL/6 mice. Diabetes 2010;59:318191.
[13] Lichtenstein L, Mattijssen F, de Wit NJ, Georgiadi A, Hooiveld GJ, van der Meer
R, et al. Angptl4 protects against severe proinammatory effects of saturated
fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages.
Cell Metab 2010;12:58092.
[14] de Wit NJ, Bosch-Vermeulen H, de Groot PJ, Hooiveld GJ, Bromhaar MM, Jansen
J, et al. The role of the small intestine in the development of dietary fatinduced obesity and insulin resistance in C57BL/6J mice. BMC Med Genomics
2008;1:14.

L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370


[15] Centis E, Marzocchi R, Di Domizio S, Ciaravella MF, Marchesini G. The effect of
lifestyle changes in non-alcoholic fatty liver disease. Dig Dis 2010;28:26773.
[16] Yki-Jarvinen H. Liver fat in the pathogenesis of insulin resistance and type 2
diabetes. Dig Dis 2010;28:2039.
[17] Nathan C, Ding A. Nonresolving inammation. Cell 2010;140:87182.
[18] Martin KJ, Graner E, Li Y, Price LM, Kritzman BM, Fournier MV, et al. Highsensitivity array analysis of gene expression for the early detection of
disseminated breast tumor cells in peripheral blood. Proc Natl Acad Sci USA
2001;98:264651.
[19] Valk PJ, Verhaak RG, Beijen MA, Erpelinck CA, Barjesteh van Waalwijk van
Doorn-Khosrovani S, Boer JM, et al. Prognostically useful gene-expression
proles in acute myeloid leukemia. N Engl J Med 2004;350:161728.
[20] Radich JP, Mao M, Stepaniants S, Biery M, Castle J, Ward T, et al. Individualspecic variation of gene expression in peripheral blood leukocytes. Genomics
2004;83:9808.
[21] Whitney AR, Diehn M, Popper SJ, Alizadeh AA, Boldrick JC, Relman DA, et al.
Individuality and variation in gene expression patterns in human blood. Proc
Natl Acad Sci USA 2003;100:1896901.
[22] Cobb JP, Mindrinos MN, Miller-Graziano C, Calvano SE, Baker HV, Xiao W, et al.
Application of genome-wide expression analysis to human health and disease.
Proc Natl Acad Sci USA 2005;102:48016.
[23] Mandard S, Muller M, Kersten S. Peroxisome proliferator-activated receptor
alpha target genes. Cell Mol Life Sci 2004;61:393416.
[24] Rakhshandehroo M, Knoch B, Muller M, Kersten S. Peroxisome proliferatoractivated receptor alpha target genes. PPAR Res. 2010;2010.
[25] Sanderson LM, Degenhardt T, Koppen A, Kalkhoven E, Desvergne B, Muller M,
et al. Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta)
but not PPARalpha serves as a plasma free fatty acid sensor in liver. Mol Cell
Biol 2009;29:625767.
[26] Sanderson LM, de Groot PJ, Hooiveld GJ, Koppen A, Kalkhoven E, Muller M,
et al. Effect of synthetic dietary triglycerides: a novel research paradigm for
nutrigenomics. PLoS One 2008;3:e1681.
[27] van der Meer DL, Degenhardt T, Vaisanen S, de Groot PJ, Heinaniemi M, de
Vries SC, et al. Proling of promoter occupancy by PPARalpha in human
hepatoma cells via ChIP-chip analysis. Nucleic Acids Res 2010;38:283950.
[28] Rakhshandehroo M, Hooiveld G, Muller M, Kersten S. Comparative analysis of
gene regulation by the transcription factor PPARalpha between mouse and
human. PLoS One 2009;4:e6796.
[29] Im SS, Osborne TF. Liver X receptors in atherosclerosis and inammation. Circ
Res 2011;108:9961001.
[30] Desvergne B, Michalik L, Wahli W. Transcriptional regulation of metabolism.
Physiol Rev 2006;86:465514.
[31] Peet DJ, Turley SD, Ma W, Janowski BA, Lobaccaro JM, Hammer RE, et al.
Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear
oxysterol receptor LXR alpha. Cell 1998;93:693704.
[32] Jump DB. Fatty acid regulation of hepatic lipid metabolism. Curr Opin Clin
Nutrit Metab Care 2011;14:11520.
[33] Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete
program of cholesterol and fatty acid synthesis in the liver. J Clin Invest
2002;109:112531.
[34] Unger RH, Scherer PE. Gluttony, sloth and the metabolic syndrome: a roadmap
to lipotoxicity. Trends Endocrinol Metab 2010;21:34552.
[35] Jump DB. N 3 polyunsaturated fatty acid regulation of hepatic gene
transcription. Curr Opin Lipidol 2008;19:2427.
[36] Margioris AN. Fatty acids and postprandial inammation. Curr Opin Clin Nutr
Metab Care 2009;12:12937.
[37] Jackson KG, Armah CK, Minihane AM. Meal fatty acids and postprandial
vascular reactivity. Biochem Soc Trans 2007;35:4513.
[38] van Oostrom AJ, Rabelink TJ, Verseyden C, Sijmonsma TP, Plokker HW, De
Jaegere PP, et al. Activation of leukocytes by postprandial lipemia in healthy
volunteers. Atherosclerosis 2004;177:17582.
[39] Bouwens M, Grootte Bromhaar M, Jansen J, Muller M, Afman LA. Postprandial
dietary lipid-specic effects on human peripheral blood mononuclear cell gene
expression proles. Am J Clin Nutr 2010;91:20817.
[40] Zeller T, Wild P, Szymczak S, Rotival M, Schillert A, Castagne R, et al. Genetics
and beyond the transcriptome of human monocytes and disease
susceptibility. PLoS One 2010;5:e10693.
[41] Dolores Mayas M, Isabel Queipo-Ortuno M, Clemente-Postigo M, Macias M, El
Bekay R, Tinahones FJ, et al. Inuence of a fat overload on lipogenic regulators
in metabolic syndrome patients. Br J Nutr 2010:17.
[42] Uehara Y, Miura S, von Eckardstein A, Abe S, Fujii A, Matsuo Y, et al.
Unsaturated fatty acids suppress the expression of the ATP-binding cassette
transporter G1 (ABCG1) and ABCA1 genes via an LXR/RXR responsive element.
Atherosclerosis 2007;191:1121.
[43] Yoshikawa T, Shimano H, Yahagi N, Ide T, Amemiya-Kudo M, Matsuzaka T,
et al. Polyunsaturated fatty acids suppress sterol regulatory element-binding
protein 1c promoter activity by inhibition of liver X receptor (LXR) binding to
LXR response elements. J Biol Chem 2002;277:170511.
[44] Takeuchi Y, Yahagi N, Izumida Y, Nishi M, Kubota M, Teraoka Y, et al.
Polyunsaturated fatty acids selectively suppress sterol regulatory elementbinding protein-1 through proteolytic processing and autoloop regulatory
circuit. J Biol Chem 2010;285:1168191.
[45] Bouwens M, van de Rest O, Dellschaft N, Bromhaar MG, de Groot LC,
Geleijnse JM, et al. Fish-oil supplementation induces antiinammatory
gene expression proles in human blood mononuclear cells. Am J Clin
Nutr 2009;90:41524.

69

[46] Gorjao R, Verlengia R, Lima TM, Soriano FG, Boaventura MF, Kanunfre CC, et al.
Effect of docosahexaenoic acid-rich sh oil supplementation on human
leukocyte function. Clin Nutr 2006;25:92338.
[47] Weaver KL, Ivester P, Seeds M, Case LD, Arm JP, Chilton FH. Effect of dietary
fatty acids on inammatory gene expression in healthy humans. J Biol Chem
2009;284:154007.
[48] Grimble RF, Howell WM, OReilly G, Turner SJ, Markovic O, Hirrell S, et al. The
ability of sh oil to suppress tumor necrosis factor alpha production by
peripheral blood mononuclear cells in healthy men is associated with
polymorphisms in genes that inuence tumor necrosis factor alpha
production. Am J Clin Nutr 2002;76:4549.
[49] Caughey GE, Mantzioris E, Gibson RA, Cleland LG, James MJ. The effect on
human tumor necrosis factor alpha and interleukin 1 beta production of diets
enriched in n 3 fatty acids from vegetable oil or sh oil. Am J Clin Nutr
1996;63:11622.
[50] Meydani SN, Endres S, Woods MM, Goldin BR, Soo C, Morrill-Labrode A, et al.
Oral (n 3) fatty acid supplementation suppresses cytokine production and
lymphocyte proliferation: comparison between young and older women. J
Nutr 1991;121:54755.
[51] Vedin I, Cederholm T, Freund-Levi Y, Basun H, Hjorth E, Irving GF, et al.
Reduced prostaglandin F2 alpha release from blood mononuclear leukocytes
after oral supplementation of omega3 fatty acids: the OmegAD study. J Lipid
Res 2010;51:117985.
[52] Ruggiero C, Lattanzio F, Lauretani F, Gasperini B, Andres-Lacueva C, Cherubini
A. Omega-3 polyunsaturated fatty acids and immune-mediated diseases:
inammatory bowel disease and rheumatoid arthritis. Curr Pharm Des
2009;15:413548.
[53] Galli C, Calder PC. Effects of fat and fatty acid intake on inammatory and
immune responses: a critical review. Ann Nutr Metab 2009;55:12339.
[54] Nair S, Lee YH, Rousseau E, Cam M, Tataranni PA, Baier LJ, et al. Increased
expression of inammation-related genes in cultured preadipocytes/stromal
vascular cells from obese compared with non-obese Pima Indians.
Diabetologia 2005;48:17848.
[55] Lee YH, Nair S, Rousseau E, Allison DB, Page GP, Tataranni PA, et al. Microarray
proling of isolated abdominal subcutaneous adipocytes from obese vs. nonobese Pima Indians: increased expression of inammation-related genes.
Diabetologia 2005;48:177683.
[56] Clement K, Viguerie N, Poitou C, Carette C, Pelloux V, Curat CA, et al. Weight
loss regulates inammation-related genes in white adipose tissue of obese
subjects. FASEB J 2004;18:165769.
[57] Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante Jr AW.
Obesity is associated with macrophage accumulation in adipose tissue. J Clin
Invest 2003;112:1796808.
[58] Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inammation in
fat plays a crucial role in the development of obesity-related insulin resistance.
J Clin Invest 2003;112:182130.
[59] Lee JY, Hwang DH. The modulation of inammatory gene expression by lipids:
mediation through Toll-like receptors. Mol Cells 2006;21:17485.
[60] Kennedy A, Martinez K, Chuang CC, LaPoint K, McIntosh M. Saturated fatty
acid-mediated inammation and insulin resistance in adipose tissue:
mechanisms of action and implications. J Nutr 2009;139:14.
[61] Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard SG, Brown PJ, et al.
Molecular recognition of fatty acids by peroxisome proliferator-activated
receptors. Mol Cell 1999;3:397403.
[62] Olefsky JM, Glass CK. Macrophages, inammation, and insulin resistance. Annu
Rev Physiol 2010;72:21946.
[63] Stienstra R, Duval C, Keshtkar S, van der Laak J, Kersten S, Muller M.
Peroxisome proliferator-activated receptor gamma activation promotes
inltration of alternatively activated macrophages into adipose tissue. J Biol
Chem 2008;283:226207.
[64] Bouwens M, Afman LA, Muller M. Activation of peroxisome proliferatoractivated receptor alpha in human peripheral blood mononuclear cells reveals
an individual gene expression prole response. BMC Genomics 2008;9:262.
[65] Kohno M, Hasegawa H, Inoue A, Muraoka M, Miyazaki T, Oka K, et al.
Identication of N-arachidonylglycine as the endogenous ligand for orphan Gprotein-coupled receptor GPR18. Biochem Biophys Res Commun
2006;347:82732.
[66] Patsouris D, Reddy JK, Muller M, Kersten S. Peroxisome proliferator-activated
receptor alpha mediates the effects of high-fat diet on hepatic gene expression.
Endocrinology 2006;147:150816.
[67] van Dijk SJ, Feskens EJ, Bos MB, Hoelen DW, Heijligenberg R, Bromhaar MG,
et al. A saturated fatty acid-rich diet induces an obesity-linked
proinammatory gene expression prole in adipose tissue of subjects at risk
of metabolic syndrome. Am J Clin Nutr 2009;90:165664.
[68] Pawitan Y, Murthy KR, Michiels S, Ploner A. Bias in the estimation of false
discovery rate in microarray studies. Bioinformatics 2005;21:386572.
[69] Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al.
PGC-1alpha-responsive genes involved in oxidative phosphorylation are
coordinately downregulated in human diabetes. Nat Genet 2003;34:26773.
[70] Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA,
et al. Gene set enrichment analysis: a knowledge-based approach for
interpreting genome-wide expression proles. Proc Natl Acad Sci USA
2005;102:1554550.
[71] Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, et al. Persistent
epigenetic differences associated with prenatal exposure to famine in humans.
Proc Natl Acad Sci USA 2008;105:170469.

70

L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370

[72] Unger RH, Scherer PE. Gluttony, sloth and the metabolic syndrome: a roadmap
to lipotoxicity. Trends Endocrinol Metab 2010;21:34552.
[73] Shaham O, Wei R, Wang TJ, Ricciardi C, Lewis GD, Vasan RS, et al. Metabolic
proling of the human response to a glucose challenge reveals distinct axes of
insulin sensitivity. Mol Syst Biol 2008;4:214.
[74] Wopereis S, Rubingh CM, van Erk MJ, Verheij ER, van Vliet T, Cnubben NH, et al.
Metabolic proling of the response to an oral glucose tolerance test detects
subtle metabolic changes. PLoS One 2009;4:e4525.
[75] van Ommen B, Keijer J, Heil SG, Kaput J. Challenging homeostasis to dene
biomarkers for nutrition related health. Mol Nutr Food Res 2009;53:795804.
[76] van Ommen B, Keijer J, Kleemann R, Elliott R, Drevon CA, McArdle H, et al. The
challenges for molecular nutrition research 2: quantication of the nutritional
phenotype. Genes Nutr 2008;3:519.

View publication stats

[77] Anderwald C, Gastaldelli A, Tura A, Krebs M, Promintzer-Schifferl M, KautzkyWiller A, et al. Mechanism and effects of glucose absorption during an oral
glucose tolerance test among females and males. J Clin Endocrinol Metab 2010.
[78] Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut
microbial gene catalogue established by metagenomic sequencing. Nature
2010;464:5965.
[79] Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, Koseki M,
et al. Biological, clinical and population relevance of 95 loci for blood lipids.
Nature 2010;466:70713.
[80] van Ommen B, Bouwman J, Dragsted LO, Drevon CA, Elliott R, de Groot P, et al.
Challenges of molecular nutrition research 6: the nutritional phenotype
database to store, share and evaluate nutritional systems biology studies.
Genes Nutr 2010;5:189203.

Potrebbero piacerti anche