Vous êtes sur la page 1sur 8

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 a b s t r a c t

Article history: Nutrigenomics employs high-throughput genomics technologies to unravel how nutrients modulate gene
Available online 2 December 2011 and protein expression and ultimately inuence cellular and organism metabolism. The most often-
applied genomics technique so far is transcriptomics, which allows quantifying genome-wide changes
Keywords: in gene expression of thousands of genes at the same time in one sample. The performance of gene
Nutrigenomics expression quantication requires sufcient high-quality homogenous cellular material, therefore
Fatty acids research in healthy volunteers is restricted to biopsies from easy accessible tissues such as subcutaneous
Transcriptomics
adipose tissue, skeletal muscle and intestinal biopsies or even more easily accessible cells such as periph-
Gene expression
Peripheral blood mononuclear cells (PBMCs)
eral 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 regulat-
ing the activity of numerous transcription factors, including nuclear receptors such as peroxisome prolif-
erator 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 inam-
matory 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. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
2. Challenges in human nutrigenomics studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
3. Transcriptional gene regulation by (dietary) fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
4. Application of transcriptomics in human intervention studies with dietary fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
4.1. Postprandial intervention studies with dietary fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
4.2. Longer-term intervention studies with dietary fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
4.3. Overlap between long term and postprandial effects of n 3 PUFA on PBMC transcriptome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
5. Future challenges in nutrigenomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
6. Conclusions and perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68

1. Introduction inappropriate dietary patterns. The last decade has revealed an


enormous wealth of insights in the complexity of human physiology
Human nutrition research is still largely focused on issues related and in particular of pathophysiology of complex diseases. At the
to nutrient deciencies and impairment of health and disease due to same time we have to acknowledge a worldwide epidemic increase
in preventable diseases such as metabolic syndrome or diabetes
Corresponding author. Address: Nutrition, Metabolism and Genomics Group, type 2 that are linked to obesity and largely related to higher energy
Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD
intake and reduced energy expenditure [1]. On the other hand new
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
high-throughput genomics technologies for the generation, process-
(M. Mller). ing, and analysis of biological data about the composition and

0163-7827/$ - see front matter 2011 Elsevier Ltd. All rights reserved.
doi:10.1016/j.plipres.2011.11.005
64 L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370

functions of genomes has created unprecedented opportunities for areas, in patients without necessity surgery such as diabetes
increasing our understanding of how nutrients modulate gene and patients and especially in healthy volunteers, collection of samples
protein expression and ultimately inuence cellular and organ from internal organs such as liver, pancreas and visceral adipose
metabolism. To overcome the signicant gap between molecular tissue is impossible because of ethical reasons. Research in those
biology, physiology and more traditional nutrition research, nutri- volunteers, is therefore restricted to biopsies from more easily
genomics has been initiated as a new innovative strategy around accessible tissues such as subcutaneous adipose tissue, muscle
10 years ago when a working draft of the human genome was an- and intestinal biopsies without a high health risk for the volunteer.
nounced. There are several meanings of nutrigenomics as used in Although other metabolic organs such as liver and visceral fat, are
the scientic literature mostly focusing on gene-environment inter- not assessable for invasive investigations, comprehensive pheno-
actions and the relevance of genotype changes e.g. single nucleotide typing of these organs in healthy and early pathophysiological
polymorphism (SNPs) for individual variation in responses to foods, stages in the development of diseases would be extremely valu-
dietary pattern and susceptibility to develop nutrition-related dis- able. These metabolic organs fulll key physiological functions in
eases [2,3]. We have extent this denition of nutrigenomics by the efcient regulation of homeostasis related to dietary fat metab-
including innovative combinations of molecular nutrition research olism and play a key role in the early development of nutrition-re-
and genomics applications, largely performed in transgenic animal lated diseases such as metabolic syndrome and type II diabetes as
models. The diverse tissue and organ-specic effects of bioactive shown to large extent in animal studies [1016]. The development
dietary components include gene expression patterns (transcrip- of non-alcoholic fatty liver disease or non-alcoholic steatohepatitis
tome), organization and modication of the chromatin structure [15,16]; is a good example for this as a more recently frequently
(epigenome), protein expression patterns including post-transla- observed human disease phenotype that is strongly linked to met-
tional modications (proteome) as well as metabolite proles abolic syndrome and both may even be two denitions of the same
(metabolome) [46]. Nutrigenomics promotes an increased under- problem (chronic metabolic stress) with ultimately the develop-
standing on how nutrition inuences metabolic pathways and ment of diabetes type 2 and cardiovascular health problems as
homeostatic control by providing mechanistic insights into how consequences.
nutrition works (evidence-based nutrition). Furthermore it reveals To allow more efcient prevention of these diseases by evi-
how this regulation is disturbed in the early phases of diet-related dence-based nutritional and lifestyle intervention a comprehensive
disease, and the extent to which individual sensitizing genotypes understanding of the various phases (from organ dysfunction to
contribute to such diseases by allowing comprehensive and deep systemic pathophysiology) in the disease evolution is key. There-
phenotyping and the identication of early biomarkers for pre-dis- fore the study of time-dependent transitions from the healthy to
ease states [79]. Nutrigenomics is the approach that generates disease state and the role that nutrition plays in these time-depen-
the knowledge that allows development of new nutritional strate- dent changes of organ phenotypes would be essential but is cur-
gies to improve peoples health and tness according to their indi- rently not feasible because of the invasive procedure, the low
vidual needs. This necessary evolution will eventually lead to tissue and RNA yield and the presence of numerous cell types in
evidence-based dietary intervention strategies for restoring health the biopsy. A far less invasive method is the collection of blood cells
and tness and for preventing diet-related disease. out of whole blood obtained via venipuncture. Blood cells travel
through the whole body, are exposed to all physiological changes
2. Challenges in human nutrigenomics studies including changes in nutrient levels before and after meals or in
the fasting state. They are key cells in regulating organ-specic
Since the introduction of genomics techniques such as transcri- and systemic immune function and inammatory responses. Non-
ptomics many human studies have been performed to examine resolving inammation is a major driver of disease, e.g. blood cells
phenotypic differences between diseased and healthy states with can attached to and invade in the vascular wall leading to foam cells
the ultimate goal to improve early diagnostic and to increase our formation and ultimately atherosclerosis [17]. The most studied
understanding of underlying mechanisms of early disease pathol- blood cells are the peripheral blood mononuclear cells (PBMCs) that
ogy or to investigate pharmacological efcacy to modulate disease consist of monocytes and lymphocytes. Whole genome transcrip-
progression. Recently these techniques are also applied to achieve tome analyzes of PBMCs have been applied successfully in discrim-
more knowledge on metabolic, physiological or dietary changes on inating disease from healthy populations and in differentiating
alterations in transcriptional gene regulation in humans. several phases/diagnostics of disease [18,19]. It has also been
Several types of intervention studies can be performed in which shown that variation in PBMC gene expression within persons is
effects of specic nutrients (e.g. n 3 PUFA versus saturated fat) or smaller than the variation between persons [2022]. This smaller
changes in more complex dietary patterns (Mediterranean versus within person variation combined with the usual applied measure-
Western-style diets) can be examined on the levels of gene expres- ment of baseline and end samples within a person in nutritional
sion proles. The length of the study design can vary from a few intervention studies enables potential successful application of
hours in postprandial studies, to weeks and years in respectively PBMC gene expression proling in nutritional intervention studies.
completely controlled dietary intervention studies and long-term
interventions trials. It is expected that postprandial studies mirror 3. Transcriptional gene regulation by (dietary) fatty acids
the more acute effects of daily consumption whereas long-term
studies will reect the extended inuence of nutrition on whole Dietary fatty acids inuence human health in numerous ways
body health. All types of studies, if well controlled, are of pivotal and inuence several indicators of health status. In recent years
importance to increase our knowledge and understanding on much work has focused to elucidate more precisely the mode of ac-
how nutrients are sensed by our organ cells and how this translates tion how dietary fatty acids lead to short-and long-term changes in
efciently into necessary adaptive physiological response mecha- cellular functions. There is now considerable evidence that fatty
nism as essential for health, both on the short and long run. acids, in particular unsaturated fatty acids, exert many of their bio-
The major bottleneck in studying these kind of often-subtle logical effects through modulation of gene transcription by regu-
changes is the limited accessibility of tissues as study populations lating the activity of numerous transcription factors, including
mostly consist of healthy volunteers. Whereas in ill subjects that sterol regulatory binding proteins (SREBPs) or nuclear receptors
need to undergo surgery biopsies can be more easily obtained from such as peroxisome proliferator activated receptors (PPARs) and
both pathological tissue and more normal surrounding tissue liver X receptors (LXRs).
L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370 65

Three different highly conserved PPAR isotypes have been iden- transcription factors see Desvergne et al. [30,33]. Insulin induces
tied: PPARa (NR1C1), PPARb/r (NR1C2) and PPARc (NR1C3). SREBP-1 nuclear which induces lipogenesis, MUFA and triglyceride
They act as nutrient sensors for fatty acids, but bind also numerous synthesis and storage [34]. The n 3 PUFA docosahexaneoic acid
fatty acid derivatives and compounds showing structural resem- (DHA) is a very strong nutritional suppressor of nuclear SREBP-1
blance to fatty acids and inuence the expression of specic genes expression of genes involved in lipogenesis and MUFA synthesis
[23,24] The PPARa isotype has been shown to govern expression of in liver [35].
numerous genes involved in fatty acid oxidation, ketogenesis,
gluconeogenesis, cholesterol catabolism and lipoprotein metabo- 4. Application of transcriptomics in human intervention studies
lism as mainly studied using mouse models. More recent evidence with dietary fatty acids
indicates that hepatic PPARa is not largely activated by plasma free
fatty acids as high during fasting, whereas it can be activated by 4.1. Postprandial intervention studies with dietary fatty acids
dietary fatty acids and fatty acids generated via de novo lipogene-
sis [25]. Recently, it was also shown that the effects of dietary Due to an easy accessible surplus of large processed food (rich in
unsaturated fatty acids on hepatic gene expression are almost isolated sugars and saturated fats) we are in the postprandial state
exclusively mediated by PPARa and mimic the effect of synthetic the greater part of the day in particular if increased energy
PPARa agonists [26]. Much less is known about the relevance of consumption by e.g. exercise rarely allows to compensate. A better
these mainly mouse-based ndings for human physiology. A recent understanding of meal effects on body function and health are of
analysis provided new insights into the mechanisms and impact of critical importance. It is known that a meal can provoke a transient
transcriptional regulation by PPARa in human liver by performing inammatory response which is low compared to a general inam-
chromatin immunoprecipitation (ChIP)-chip in combination with matory response [36]. In addition, postprandial effects of high fat
transcriptional proling on HepG2 human hepatoma cells treated meals have been observed on vascular reactivity [37] and on blood
with the specic PPARa agonist GW7647. Several genes known cell activation [38]. The arrival of comprehensive genomics tech-
to be regulated by PPARa showed GW7647-induced PPARa bind- niques such as transcriptomics enables the unbiased measurement
ing to their promoter. Interestingly also certain SREBP-target genes of total postprandial genome-wide effects on the cellular level. Re-
exhibited PPARa binding, suggesting cross-talk between PPARa cently, we assessed postprandial transcriptional responses to high
and SREBP signaling [27]. Another study compared the function fat shakes varying in types of fatty acids [39]. Twenty healthy sub-
of PPARa in mouse and human hepatocytes via analysis of target jects consumed a butterfat-enriched saturated fatty acid (SFA)
gene regulation. Minor overlap was observed between the shake and a DHA-enriched polyunsaturated fatty acid (PUFA) shake
PPARa-regulated genes from mouse and human by a gene-by-gene in random order and whole genome PBMCs gene expression pro-
comparison, although more substantial overlap was observed at les were assessed. Postprandial gene expression responses were
the pathway level. This study clearly suggest that PPARa activation distinctly different between shakes as illustrated by the higher
has a major impact on gene regulation in human hepatocytes and number of genes changed upon PUFA shake consumption. It is ex-
that the role of PPARa as master regulator of hepatic lipid metab- pected that adaptive cellular response by means of gene expression
olism is generally well-conserved between mouse and human [28]. changes will only occur when cellular capacity is insufcient to deal
More in depth studies are required to elucidate the specic role of efciently with the high load of intracellular free fatty acids. High
nutrient-sensing transcription factors in mediating the effects of concentrations of n 3 PUFA are more stress-inducing molecules
dietary fatty acids in other human cells including small intestinal than SFA for blood cells that need to activate adaptive gene tran-
epithelial cells, macrophages or adipocytes. scription program to be able to cope with the high levels of this
LXRs, closely PPAR-related members of the nuclear receptor fam- stressors by rapid processing of n 3 PUFA and increased cellular
ily, are important regulators of cholesterol, fatty acid, glucose protection against e.g. oxidative stress. The increased expression
homeostasis and play an essential role at the interface between of genes involved in cellular stress response upon n 3 PUFA shake
metabolism and inammation [29]. Their endogenous activators consumption points clearly in this direction. Other differences be-
are oxysterols and other derivatives of cholesterol metabolism tween the SFA and n 3 PUFA shake could mainly be attributed
(22(R)-hydroxycholesterol, 24(S)-hydroxycholesterol, 24(S), 25- to expression alterations of genes involved in lipid metabolism with
epoxycholesterol). LXRa is highly expressed in the liver but is also opposite effects for the two shakes. SFA caused a postprandial up-
found in kidney, intestine, adipose tissue, and macrophages. LXRb regulation in transcription of ABCA1, ABCG1, and SREBP1, whereas
is expressed ubiquitously[30]. Nutritional or pharmacological mod- an overload of n 3 PUFA resulted in a down-regulation in expres-
ulation of the activity of these ligand-activated transcription factors sion of the same genes. Both effects were conrmed by ex vivo incu-
have been shown to inuence the development of metabolic disor- bation of PBMCs with palmitic acid and the DHA, pointing to an
ders such as hyperlipidemia and atherosclerosis [30]. Mice without effect likely induced by the fatty acids itself. Interestingly, a whole
functional LXRa e.g. are healthy when fed with a low-cholesterol genome gene expression study in a population of around 1500 indi-
diet but when fed a high-cholesterol diet these LXRa knockout mice viduals showed that gene expression of ABCA1 and, ABCG1 was neg-
develop enlarged fatty livers, less functional hepatocytes, high cho- atively associated with TG levels and, SCD gene expression was
lesterol levels in liver, and impaired liver function [31]. LXR is a ma- found to be negatively associated with HDL-cholesterol [40]. An-
jor regulator of fatty acid synthesis and the expression of other other study also observed a postprandial increase in expression of
lipogenic genes through direct interaction with their promoters as LXR-related genes in PBMCs after a high fat load. In this study the
well as inducing sterol regulatory element binding protein-1c postprandial response upon a high fat load consisting of a mix of
(SREBP-1c) gene transcription. Activation of LXR by oxysterols (re- fatty acids in 22 patients with metabolic syndrome was examined
lated to high dietary cholesterol) or other agonists elevates nuclear and similar to our SFA shake the gene expression of SREBP1 and
content of SREBP-1 and increases expression of the enzymes in- LXRa was found to be increased. [41]. Several mice and in vitro stud-
volved in de novo lipogenesis [32]. ies have examined the effects on n 3 PUFAs on hepatic gene reg-
SREBPs are transcription factors of the helix-loop-helix family ulation and showed that DHA is the most potent hepatic fatty acid
and important regulators of cholesterol and fatty acid metabolism. suppressor of SREBP1 gene expression [35]. Less studies have
SREBPs are encoded by two genes, SREBP-1 and SREBP-2 resulting explored the effects of n 3 PUFAs on monocytes/macrophages
in 3 proteins SREBP-1a, SREBP-1c and SREBP-2 (for more informa- but in vitro studies in macrophage cell lines showed down-regula-
tion in particular on the complex mode of activation of these tion 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 what the systemic implications are. In the study of Gorjao [46] be-
modulating cholesterol efux from macrophages, the n 3 PUFA- sides pooled macroarrays, stimulation assays were performed on
reduced postprandial expression of these genes in PBMCs in the lymphocytes showing an increased production of IL-10, IFN-
humans may lead to an inhibition of cellular cholesterol efux. Con- gamma and TNFa and a decreased production of IL2. However, most
versely, the increased expression of LXR and its target genes after other studies observed a decrease in production of cytokines after
SFA or mixed fatty acid consumption might result in an increased sh oil supplementation, which is more in line with the observed ef-
postprandial cholesterol efux. Interestingly, daily consumption fect on transcriptional regulation [4851]. In addition, Meydani ob-
of a lower dose of n 3 PUFAs for a longer period did not result served a higher reduction in cytokine response upon stimulation in
in a reduced expression of these genes when determined in the fast- older compared to young women [50] after n 3 PUFA supplemen-
ing state [45]. This data combined with the nding that levels of tation, underlining the potential age-dependency of responses.
LXR-related gene expression where heading back to baseline 8 h Overall, long-term sh oil supplementation reduces expression
postprandially, suggests that the effects observed in PBMCs might of leukocyte inammatory genes and decreases cytokine produc-
be temporarily and specic for the postprandial situation when tion upon leukocyte stimulation. The relevance for health outcome
blood lipid concentrations are elevated [39]. Still, the presence of of this reduced immune response has been postulated to be bene-
the postprandial state the greater part of the day may lead to an cial in progression and treatment of inammatory diseases such
continuous trigger of which the consequences on gene expression as rheumatoid arthritis [52,53].
and cholesterol efux are yet unknown. Besides effects of consumption of fatty acids on transcriptional
activity in blood cells dietary fatty acid effects have also been deter-
4.2. Longer-term intervention studies with dietary fatty acids mined on whole genome gene expression of adipose tissue. Within a
completely controlled randomized dietary intervention trial of
Daily changes in postprandial exposure will ultimately lead to 8 weeks the effects of different types of fatty acids was studied on
longer-term adaptations in the whole body. Daily exposure to spe- adipose tissue transcriptome. Participants either consumed a but-
cic fatty acids will activate adaptive transcriptional responses fre- ter-derived saturated fatty acids (SFA)-rich diet or a rened olive
quently, ultimately leading to functional alterations in activity of oilderived monounsaturated fatty acids (MUFA)-rich diet and adi-
pathways and networks in organs such as the small intestine, liver, pose tissue biopsies were taken before and after intervention. Whole
heart, skeletal muscle and adipose tissue. Longer-term consump- genome microarray analyses of these samples revealed that the SFA-
tion will in addition lead to incorporation of fatty acids into mem- diet resulted in the most distinct changes in gene expression. Where
branes of practical every cell in the body, which will have their SFA-diet effected expression of around 1500 genes, MUFA consump-
consequences as well. tion effected expression of 600 genes. The most remarkable obser-
To determine longer-term effects of fatty acids on the cellular vation was the increased transcription of genes involved in immune
transcriptome we assessed changes in whole genome transcrip- function after consumption of the SFA-rich diet, which was
tional proles in PBMCs and adipose tissue after long-term con- distinctly different from the effects on the MUFA diet, after which
sumption of different fatty acids. The study comprised a 26 weeks a much smaller and more or less opposite effect was observed on
intervention with daily supplementation of 0.4 g EPA/DHA, 1.8 g expression of immune-related genes. Interestingly, the SFA-induced
EPA/DHA or a control oil in an elderly population. Daily consumption changes in gene expression were similar to differences observed in
of 1.8 g n 3 PUFA resulted in changes in expression of 1000 genes gene expression in adipose tissue of obese versus lean subject
while expression of only 300 genes was changed in the control [54,55] and opposite to the changes observed after weight loss in ob-
group that consumed high oleic sunower oil. The larger part of ese subjects [56]. Keeping in mind that no weight changes were
the changes in the n 3 PUFA could be attributed to a down-regula- present in the participants, consumption of the more healthy type
tion in expression of immune related genes and genes known to be of dietary fat (unsaturated) may be of great relevance for healthy
involved in development of atherosclerosis. Additional changes functional adipose tissue in obesity. Although no changes in macro-
were related to an increased expression of cell-cycle genes and of phage inltration were observed, prolonged consumption of a SFA-
genes involved in transcriptional and translational regulation [45]. rich diet may result in inltration of macrophages as frequently per-
Another nutrigenomics study [46] explored the effects of two ceived in obesity [57,58]. Some studies have shown that SFA may
months supplementation with 3 g EPA/DHA daily in 10 male volun- activate immune related genes by binding and activation of Toll-like
teers on gene expression using pooled RNA of lymphocytes on a receptor 4 [59,60]. In addition, saturated FA are poor ligands for
macroarray with a selected gene set. Parallel to our results they ob- PPARy [61], and as PPARy activation result in a reduced inamma-
served a decrease in expression of cytokine-related genes, but con- tion, a decreased activation of PPARy by SFA might lead to increased
trary to our data a decrease in expression of cell cycle and inammation. PPARy gene expression in adipose tissue and expres-
transcriptional regulation genes was observed. However, it must sion of some target genes was reduced upon SFA consumption.
be noted that this study used pooled samples for macroarray ana- PPARc also plays an important role in alternative M2 macrophage
lyzes in which signicant inter-individual differences are impossi- activation that is of key importance for adipose tissue remodeling,
ble to determine and one must rely on fold change cut offs. inammatory status and health [62] as shown in numerous mouse
Furthermore, PBMCs consist of a combination of lymphocytes and studies [63] and increasingly also in human studies [62]. Interest-
monocytes which likely will have inuenced the effects on tran- ingly, in a recently performed mouse study we found that adipose
scription as well. Besides the use of arrays to study whole genome tissue function is essential for the prevention of non-alcoholic fatty
gene transcription or a large set of genes simultaneously one study liver disease [12]. This again highlights the importance of the com-
examined the expression of a selection of genes in PBMCs in 27 prehensive understanding of nutrition-relevant inter-organ depen-
healthy volunteers and found a reduction in gene expression of some dency and crosstalk as essential basis for a healthy body function.
cytokines after 4 weeks supplementation with 775 mg EPA and bor- Future nutrigenomics research should be leading in this effort.
age oil (831 mg GLA) daily [47]. Although no control group consum-
ing placebo oil with the same energy content was included in the 4.3. Overlap between long term and postprandial effects of n 3 PUFA
study and the changes in gene expression cannot solely be attributed on PBMC transcriptome
to EPA, the results point in the same direction as the previous two
studies. The question remains what a down-regulation in transcrip- The above described studies show that fatty acids can not only
tion of inammatory genes means for cellular functionality and affect gene transcription directly after consumption but will also
L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370 67

induce long-term changes. Based on the pathway analyses it 5. Future challenges in nutrigenomics
appears that depending on the length of the intervention expres-
sion of other genes and pathways are affected. To reveal whether It is evident from the rst 10 years of nutrigenomics research and
overlapping genes in PBMCs were changed between postprandial applying comprehensive genomics techniques such as transcripto-
and long-term n 3 PUFA consumption we compared datasets of mics that researchers have to acknowledge relatively small changes
the 26 weeks intervention and the postprandial study. The overlap in gene expression induced by nutrients as compared to drugs
was relatively low; of the 900 genes specically changed by n 3 [26,66] with large inter-individual variations. This is not unexpected
PUFA in the long-term study and the 291 genes in the postprandial considering the different ways drugs and nutrients work [4]. Where
study, 29 genes were changed upon n 3 PUFA in both studies drugs are designed to specically and constantly activate certain
(Fig. 1). The expression of the larger part of the genes i.e. 19 genes receptors with the goal to strongly activate specic pathways, nutri-
are changed in the same direction and may reect the overall ef- ents will induce a much milder activation of specic receptors and
fects of sh oil consumption. The expression of the remaining 10 consequently of specic pathways. Instead nutrients will lead to a
genes is changed in the opposite direction. One of the genes up- more balanced cellular response by activation of multiple control
regulated postprandial and down-regulated after long-term inter- systems with the main purpose to metabolize the nutrient and to
vention was ADFP, a known PPARa target gene [64]. Other PPARa regulate metabolism to rapidly reach homeostatic balance. If studies
target genes were down-regulated in the long-term intervention. are performed under physiological conditions it consequently
We hypothesize that daily intake of higher amounts of n 3 PUFA means that rather low amounts of bioactive compounds are applied
as mentioned above induces whole body adaptation in particular in and low numbers of signicantly changed genes and an lower induc-
metabolic organs such as the small intestine, liver or adipose tissue tion in changes in gene expression can be expected as there is no
with the consequence of lower FFA levels and consequently differ- need for a stronger transcriptional response. On average fatty acids
ent changes in gene expression levels in PBMCs compared to acute induce effect sizes ranging from a few percent till 100% with individ-
studies. The same may hold for JUNB, a stressor/inammatory gene ual outliers to 300% or 400% often for immune-related genes
and the other genes in the list. The function of the overlap in genes [39,45,67]. Both points, the low fold changes and low proportion
varies, but several genes are immune-related such as CLEC4E and of genes signicantly changed require a rigorous high-quality con-
Il13RA1 or lipid-related such as SCD. G-protein coupled receptor trol system coupled to an array analysis platform with high quality
18 (GPR18) is one of the genes with the highest signicantly aver- arrays, strict quality control criteria and the need for integrated bio-
age increase upon both long-term and acute consumption. GPR18 informatics and statistical procedures optimized for studies with
is know to be highly expressed in lymphocytes and N-arachidonyl- small effect sizes [68]. Moreover, it emphasizes the importance of
glycine has been suggested to be an endogenous ligand of GPR18 use of whole genome data and not only a biased selection and the
[65], but further research concerning its function and the relation- application of most recent well annotated pathway analyzes tools,
ship to n 3 PUFA is needed. The analysis also reveals that tran- enabling the uncovering or enrichment of certain pathways or
scriptional changes of a large number of genes are very specic signaling networks [69,70].
for either long-term or acute consumption of n 3 PUFA. Another challenge is the great individual variations in gene
In conclusion, acute postprandial studies lead to short term di- expression response to nutritional interventions largely due to
rect effects of fatty acids on transcription whereas long-term inter- age, sex and genotype differences. As with other metabolites in plas-
vention with daily consumption will lead to more systemic effects ma responders and non-responders to a dietary intervention can be
that still can be monitored in PBMCs. 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 popula- 6. Conclusions and perspective
tions studied, so far. This variance in response also emphasizes the
need for within person measurements and the relevance of cross- In summary, well-controlled nutrigenomics studies on the reg-
over interventions or large intervention groups when using parallel ulation of gene transcription by dietary fatty acids in humans are
designs. The underlying reason for this response variance can be sev- still limited partly because nutrigenomics is still a relative new
eral and may vary from genetic and epigenetic variation to nutri- eld and not yet fully integrated in nutrition research and partly
tional and life style factors. For example, imprinting during because of still signicant costs of the genomics applications that
pregnancy or during early years in life may affect phenotypic do not allow comprehensive phenotyping in large cohorts. The
responses at older age with the consequence of increased risk to de- studies that have been performed were largely of explorative nat-
velop nutrition-related diseases [71]. Nutritional habits may also be ure with the purpose to identify whether transcriptional changes
a reason why persons will or will not respond to a change in a diet. can be detected after changes in dietary fatty acids consumption.
Nutrigenomics can be used to asses this. For instance, people that Although the number of studies is still limited, all studies clearly
regularly consume sh rich in n 3 PUFA will likely exhibit a less suggest that changes in dietary fatty acids intake and composition
pronounced transcriptional response upon a sh oil challenge than can have a signicant impact on cellular adaptive response capac-
subjects that do not eat sh at all. This also may be the scientic basis ity by gene transcription changes in humans. This adds important
for the nutritional advice to enjoy a healthy diet consisting of a var- knowledge to our understanding of the strong effects that various
ied food pattern as this will keep the homeostatic balance of our fatty acids can have on numerous metabolic and inammatory
organs most exible. pathways, signaling routes and homeostatic control in the cell
Many diseases are a result of disturbance of homeostasis or and ultimately on whole body health.
homeostatic imbalance. As a hallmark of ageing every organism Nutritional science is entering a new era with a shift away from
will lose its efciency in its sensing and regulatory systems. little science towards big science. The major nutritional prob-
Diseases with a strong link to nutrition result from a homeostatic lems facing the world today (e.g. obesity, diabetes and malnutri-
imbalance such as obesity, metabolic syndrome or diabetes type tion) with huge impact on public and private parties cannot be
2. We increasingly understand the complex regulation of homeo- solved by little science, characterized by individualistic research,
static control mechanisms on cellular, organ and system level that lacking coordination and integration. Big science is needed, i.e.
should prevent this imbalance from occurring. However, in some multiple groups with complementary knowledge and expertise
people, these mechanisms do not work efciently enough because working together in large national and international consortia sim-
of signicant genetic susceptibility [1] and or epigenetic mecha- ilar to consortia as currently working on the human microbiome
nism [71]. These people are much more vulnerable to develop a project [78] or genome-wide association studies [79]. This new
disease in particular if the homeostatic control mechanisms are era takes full advantage of modern, high resolution, comprehensive
chronically challenged by surplus of calories, non-ideal dietary pat- and unbiased Genomics technologies, bio-banking, sophisticated
terns and lifestyles [72]. Because we are still not able to detect the and partly web-based database management, providing a new
early warning signals of such homeostatic imbalance efciently platform for scientic dialogue and exchange [80]. We dene this
most often medical intervention is necessary at a very late stage new Nutritional Science as Nutritional Science 2.0.
of the disease development that is often unable however to restore
the homeostatic balance to regain normal health with irreversible References
damage of organs as a consequence.
To change this inefcient concept in treatment of lifestyle [1] McCarthy MI. Genomics, type 2 diabetes, and obesity. N Engl J Med
2010;363:233950.
diseases we have to use interventions that allow the quantication [2] Ordovas JM, Corella D. Nutritional genomics. Annu Rev Genomics Hum Genet
of the homeostatic balance or resilience by assessing it after chal- 2004;5:71118.
lenging or perturbing a homeostatic situation. A well known exam- [3] Kaput J, Rodriguez RL. Nutritional genomics: the next frontier in the
postgenomic era. Physiol Genomics 2004;16:16677.
ple for such as challenge is the oral glucose tolerance test (OGTT). A [4] Muller M, Kersten S. Nutrigenomics: goals and strategies. Nat Rev Genet
standard 2 h OGTT is sufcient to diagnose or exclude forms of 2003;4:31522.
diabetes mellitus at the various stages of development. Recently, [5] Grayson M. Nutrigenomics. Nature 2010;468:S1.
[6] Masotti A, Da Sacco L, Bottazzo GF, Alisi A. Microarray technology: a promising
plasma metabolic proling combined with a glucose challenge tool in nutrigenomics. Crit Rev Food Sci Nutr 2010;50:6938.
was used to differentiate between healthy individuals and individu- [7] Afman L, Muller M. Nutrigenomics: from molecular nutrition to prevention of
als with an impaired glucose tolerance [73]. Application of meta- disease. J Am Diet Assoc 2006;106:56976.
[8] Kussmann M, Krause L, Siffert W. Nutrigenomics: where are we with genetic
bolic perturbation with comprehensive nutrigenomics-based
and epigenetic markers for disposition and susceptibility? Nutr Rev
metabolic proling may improve the diagnostic sensitivity to help 2010;68(Suppl. 1):S3847.
discriminating between healthy, less-healthy and unhealthy people. [9] Kussmann M, Panchaud A, Affolter M. Proteomics in nutrition: status quo and
outlook for biomarkers and bioactives. J Proteome Res 2010;9:487687.
Furthermore, it may allow to predict differences in the responses be-
[10] Kleemann R, van Erk M, Verschuren L, van den Hoek AM, Koek M, Wielinga PY,
tween treatments [74]. In addition, combination of dietary interven- et al. Time-resolved and tissue-specic systems analysis of the pathogenesis of
tions with metabolic challenges magnies the difcult measurable insulin resistance. PLoS One 2010;5:e8817.
and relatively mild effects of nutritional alterations by testing the [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 high-
cellular adaptive response capacity (for a more in depth discussion fat diets reveals switch from an inammatory to steatotic transcriptional
[75,76]). This rather new concept of applying various challenges program. PLoS One 2009;4:e6646.
on homeostatic control mechanisms in nutritional interventions is [12] Duval C, Thissen U, Keshtkar S, Accart B, Stienstra R, Boekschoten MV, et al.
Adipose tissue dysfunction signals progression of hepatic steatosis towards
still in its infancies and it is therefore necessary to carefully study nonalcoholic steatohepatitis in C57BL/6 mice. Diabetes 2010;59:318191.
its value and limitations. One may expect important sex-dependent [13] Lichtenstein L, Mattijssen F, de Wit NJ, Georgiadi A, Hooiveld GJ, van der Meer
differences in applying various homeostatic challenge-test as dem- R, et al. Angptl4 protects against severe proinammatory effects of saturated
fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages.
onstrated recently for OGTT [77]. But ultimately it will be the most Cell Metab 2010;12:58092.
promising strategy to combine nutrigenomics-based comprehen- [14] de Wit NJ, Bosch-Vermeulen H, de Groot PJ, Hooiveld GJ, Bromhaar MM, Jansen
sive phenotyping with systems perturbation interventions to allow J, et al. The role of the small intestine in the development of dietary fat-
induced obesity and insulin resistance in C57BL/6J mice. BMC Med Genomics
the precise assessment of individual health and nutritional needs.
2008;1:14.
L.A. Afman, M. Mller / Progress in Lipid Research 51 (2012) 6370 69

[15] Centis E, Marzocchi R, Di Domizio S, Ciaravella MF, Marchesini G. The effect of [46] Gorjao R, Verlengia R, Lima TM, Soriano FG, Boaventura MF, Kanunfre CC, et al.
lifestyle changes in non-alcoholic fatty liver disease. Dig Dis 2010;28:26773. Effect of docosahexaenoic acid-rich sh oil supplementation on human
[16] Yki-Jarvinen H. Liver fat in the pathogenesis of insulin resistance and type 2 leukocyte function. Clin Nutr 2006;25:92338.
diabetes. Dig Dis 2010;28:2039. [47] Weaver KL, Ivester P, Seeds M, Case LD, Arm JP, Chilton FH. Effect of dietary
[17] Nathan C, Ding A. Nonresolving inammation. Cell 2010;140:87182. fatty acids on inammatory gene expression in healthy humans. J Biol Chem
[18] Martin KJ, Graner E, Li Y, Price LM, Kritzman BM, Fournier MV, et al. High- 2009;284:154007.
sensitivity array analysis of gene expression for the early detection of [48] Grimble RF, Howell WM, OReilly G, Turner SJ, Markovic O, Hirrell S, et al. The
disseminated breast tumor cells in peripheral blood. Proc Natl Acad Sci USA ability of sh oil to suppress tumor necrosis factor alpha production by
2001;98:264651. peripheral blood mononuclear cells in healthy men is associated with
[19] Valk PJ, Verhaak RG, Beijen MA, Erpelinck CA, Barjesteh van Waalwijk van polymorphisms in genes that inuence tumor necrosis factor alpha
Doorn-Khosrovani S, Boer JM, et al. Prognostically useful gene-expression production. Am J Clin Nutr 2002;76:4549.
proles in acute myeloid leukemia. N Engl J Med 2004;350:161728. [49] Caughey GE, Mantzioris E, Gibson RA, Cleland LG, James MJ. The effect on
[20] Radich JP, Mao M, Stepaniants S, Biery M, Castle J, Ward T, et al. Individual- human tumor necrosis factor alpha and interleukin 1 beta production of diets
specic variation of gene expression in peripheral blood leukocytes. Genomics enriched in n 3 fatty acids from vegetable oil or sh oil. Am J Clin Nutr
2004;83:9808. 1996;63:11622.
[21] Whitney AR, Diehn M, Popper SJ, Alizadeh AA, Boldrick JC, Relman DA, et al. [50] Meydani SN, Endres S, Woods MM, Goldin BR, Soo C, Morrill-Labrode A, et al.
Individuality and variation in gene expression patterns in human blood. Proc Oral (n 3) fatty acid supplementation suppresses cytokine production and
Natl Acad Sci USA 2003;100:1896901. lymphocyte proliferation: comparison between young and older women. J
[22] Cobb JP, Mindrinos MN, Miller-Graziano C, Calvano SE, Baker HV, Xiao W, et al. Nutr 1991;121:54755.
Application of genome-wide expression analysis to human health and disease. [51] Vedin I, Cederholm T, Freund-Levi Y, Basun H, Hjorth E, Irving GF, et al.
Proc Natl Acad Sci USA 2005;102:48016. Reduced prostaglandin F2 alpha release from blood mononuclear leukocytes
[23] Mandard S, Muller M, Kersten S. Peroxisome proliferator-activated receptor after oral supplementation of omega3 fatty acids: the OmegAD study. J Lipid
alpha target genes. Cell Mol Life Sci 2004;61:393416. Res 2010;51:117985.
[24] Rakhshandehroo M, Knoch B, Muller M, Kersten S. Peroxisome proliferator- [52] Ruggiero C, Lattanzio F, Lauretani F, Gasperini B, Andres-Lacueva C, Cherubini
activated receptor alpha target genes. PPAR Res. 2010;2010. A. Omega-3 polyunsaturated fatty acids and immune-mediated diseases:
[25] Sanderson LM, Degenhardt T, Koppen A, Kalkhoven E, Desvergne B, Muller M, inammatory bowel disease and rheumatoid arthritis. Curr Pharm Des
et al. Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) 2009;15:413548.
but not PPARalpha serves as a plasma free fatty acid sensor in liver. Mol Cell [53] Galli C, Calder PC. Effects of fat and fatty acid intake on inammatory and
Biol 2009;29:625767. immune responses: a critical review. Ann Nutr Metab 2009;55:12339.
[26] Sanderson LM, de Groot PJ, Hooiveld GJ, Koppen A, Kalkhoven E, Muller M, [54] Nair S, Lee YH, Rousseau E, Cam M, Tataranni PA, Baier LJ, et al. Increased
et al. Effect of synthetic dietary triglycerides: a novel research paradigm for expression of inammation-related genes in cultured preadipocytes/stromal
nutrigenomics. PLoS One 2008;3:e1681. vascular cells from obese compared with non-obese Pima Indians.
[27] van der Meer DL, Degenhardt T, Vaisanen S, de Groot PJ, Heinaniemi M, de Diabetologia 2005;48:17848.
Vries SC, et al. Proling of promoter occupancy by PPARalpha in human [55] Lee YH, Nair S, Rousseau E, Allison DB, Page GP, Tataranni PA, et al. Microarray
hepatoma cells via ChIP-chip analysis. Nucleic Acids Res 2010;38:283950. proling of isolated abdominal subcutaneous adipocytes from obese vs. non-
[28] Rakhshandehroo M, Hooiveld G, Muller M, Kersten S. Comparative analysis of obese Pima Indians: increased expression of inammation-related genes.
gene regulation by the transcription factor PPARalpha between mouse and Diabetologia 2005;48:177683.
human. PLoS One 2009;4:e6796. [56] Clement K, Viguerie N, Poitou C, Carette C, Pelloux V, Curat CA, et al. Weight
[29] Im SS, Osborne TF. Liver X receptors in atherosclerosis and inammation. Circ loss regulates inammation-related genes in white adipose tissue of obese
Res 2011;108:9961001. subjects. FASEB J 2004;18:165769.
[30] Desvergne B, Michalik L, Wahli W. Transcriptional regulation of metabolism. [57] Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante Jr AW.
Physiol Rev 2006;86:465514. Obesity is associated with macrophage accumulation in adipose tissue. J Clin
[31] Peet DJ, Turley SD, Ma W, Janowski BA, Lobaccaro JM, Hammer RE, et al. Invest 2003;112:1796808.
Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear [58] Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inammation in
oxysterol receptor LXR alpha. Cell 1998;93:693704. fat plays a crucial role in the development of obesity-related insulin resistance.
[32] Jump DB. Fatty acid regulation of hepatic lipid metabolism. Curr Opin Clin J Clin Invest 2003;112:182130.
Nutrit Metab Care 2011;14:11520. [59] Lee JY, Hwang DH. The modulation of inammatory gene expression by lipids:
[33] Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete mediation through Toll-like receptors. Mol Cells 2006;21:17485.
program of cholesterol and fatty acid synthesis in the liver. J Clin Invest [60] Kennedy A, Martinez K, Chuang CC, LaPoint K, McIntosh M. Saturated fatty
2002;109:112531. acid-mediated inammation and insulin resistance in adipose tissue:
[34] Unger RH, Scherer PE. Gluttony, sloth and the metabolic syndrome: a roadmap mechanisms of action and implications. J Nutr 2009;139:14.
to lipotoxicity. Trends Endocrinol Metab 2010;21:34552. [61] Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard SG, Brown PJ, et al.
[35] Jump DB. N 3 polyunsaturated fatty acid regulation of hepatic gene Molecular recognition of fatty acids by peroxisome proliferator-activated
transcription. Curr Opin Lipidol 2008;19:2427. receptors. Mol Cell 1999;3:397403.
[36] Margioris AN. Fatty acids and postprandial inammation. Curr Opin Clin Nutr [62] Olefsky JM, Glass CK. Macrophages, inammation, and insulin resistance. Annu
Metab Care 2009;12:12937. Rev Physiol 2010;72:21946.
[37] Jackson KG, Armah CK, Minihane AM. Meal fatty acids and postprandial [63] Stienstra R, Duval C, Keshtkar S, van der Laak J, Kersten S, Muller M.
vascular reactivity. Biochem Soc Trans 2007;35:4513. Peroxisome proliferator-activated receptor gamma activation promotes
[38] van Oostrom AJ, Rabelink TJ, Verseyden C, Sijmonsma TP, Plokker HW, De inltration of alternatively activated macrophages into adipose tissue. J Biol
Jaegere PP, et al. Activation of leukocytes by postprandial lipemia in healthy Chem 2008;283:226207.
volunteers. Atherosclerosis 2004;177:17582. [64] Bouwens M, Afman LA, Muller M. Activation of peroxisome proliferator-
[39] Bouwens M, Grootte Bromhaar M, Jansen J, Muller M, Afman LA. Postprandial activated receptor alpha in human peripheral blood mononuclear cells reveals
dietary lipid-specic effects on human peripheral blood mononuclear cell gene an individual gene expression prole response. BMC Genomics 2008;9:262.
expression proles. Am J Clin Nutr 2010;91:20817. [65] Kohno M, Hasegawa H, Inoue A, Muraoka M, Miyazaki T, Oka K, et al.
[40] Zeller T, Wild P, Szymczak S, Rotival M, Schillert A, Castagne R, et al. Genetics Identication of N-arachidonylglycine as the endogenous ligand for orphan G-
and beyond the transcriptome of human monocytes and disease protein-coupled receptor GPR18. Biochem Biophys Res Commun
susceptibility. PLoS One 2010;5:e10693. 2006;347:82732.
[41] Dolores Mayas M, Isabel Queipo-Ortuno M, Clemente-Postigo M, Macias M, El [66] Patsouris D, Reddy JK, Muller M, Kersten S. Peroxisome proliferator-activated
Bekay R, Tinahones FJ, et al. Inuence of a fat overload on lipogenic regulators receptor alpha mediates the effects of high-fat diet on hepatic gene expression.
in metabolic syndrome patients. Br J Nutr 2010:17. Endocrinology 2006;147:150816.
[42] Uehara Y, Miura S, von Eckardstein A, Abe S, Fujii A, Matsuo Y, et al. [67] van Dijk SJ, Feskens EJ, Bos MB, Hoelen DW, Heijligenberg R, Bromhaar MG,
Unsaturated fatty acids suppress the expression of the ATP-binding cassette et al. A saturated fatty acid-rich diet induces an obesity-linked
transporter G1 (ABCG1) and ABCA1 genes via an LXR/RXR responsive element. proinammatory gene expression prole in adipose tissue of subjects at risk
Atherosclerosis 2007;191:1121. of metabolic syndrome. Am J Clin Nutr 2009;90:165664.
[43] Yoshikawa T, Shimano H, Yahagi N, Ide T, Amemiya-Kudo M, Matsuzaka T, [68] Pawitan Y, Murthy KR, Michiels S, Ploner A. Bias in the estimation of false
et al. Polyunsaturated fatty acids suppress sterol regulatory element-binding discovery rate in microarray studies. Bioinformatics 2005;21:386572.
protein 1c promoter activity by inhibition of liver X receptor (LXR) binding to [69] Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al.
LXR response elements. J Biol Chem 2002;277:170511. PGC-1alpha-responsive genes involved in oxidative phosphorylation are
[44] Takeuchi Y, Yahagi N, Izumida Y, Nishi M, Kubota M, Teraoka Y, et al. coordinately downregulated in human diabetes. Nat Genet 2003;34:26773.
Polyunsaturated fatty acids selectively suppress sterol regulatory element- [70] Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA,
binding protein-1 through proteolytic processing and autoloop regulatory et al. Gene set enrichment analysis: a knowledge-based approach for
circuit. J Biol Chem 2010;285:1168191. interpreting genome-wide expression proles. Proc Natl Acad Sci USA
[45] Bouwens M, van de Rest O, Dellschaft N, Bromhaar MG, de Groot LC, 2005;102:1554550.
Geleijnse JM, et al. Fish-oil supplementation induces antiinammatory [71] Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, et al. Persistent
gene expression proles in human blood mononuclear cells. Am J Clin epigenetic differences associated with prenatal exposure to famine in humans.
Nutr 2009;90:41524. 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 [77] Anderwald C, Gastaldelli A, Tura A, Krebs M, Promintzer-Schifferl M, Kautzky-
to lipotoxicity. Trends Endocrinol Metab 2010;21:34552. Willer A, et al. Mechanism and effects of glucose absorption during an oral
[73] Shaham O, Wei R, Wang TJ, Ricciardi C, Lewis GD, Vasan RS, et al. Metabolic glucose tolerance test among females and males. J Clin Endocrinol Metab 2010.
proling of the human response to a glucose challenge reveals distinct axes of [78] Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut
insulin sensitivity. Mol Syst Biol 2008;4:214. microbial gene catalogue established by metagenomic sequencing. Nature
[74] Wopereis S, Rubingh CM, van Erk MJ, Verheij ER, van Vliet T, Cnubben NH, et al. 2010;464:5965.
Metabolic proling of the response to an oral glucose tolerance test detects [79] Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, Koseki M,
subtle metabolic changes. PLoS One 2009;4:e4525. et al. Biological, clinical and population relevance of 95 loci for blood lipids.
[75] van Ommen B, Keijer J, Heil SG, Kaput J. Challenging homeostasis to dene Nature 2010;466:70713.
biomarkers for nutrition related health. Mol Nutr Food Res 2009;53:795804. [80] van Ommen B, Bouwman J, Dragsted LO, Drevon CA, Elliott R, de Groot P, et al.
[76] van Ommen B, Keijer J, Kleemann R, Elliott R, Drevon CA, McArdle H, et al. The Challenges of molecular nutrition research 6: the nutritional phenotype
challenges for molecular nutrition research 2: quantication of the nutritional database to store, share and evaluate nutritional systems biology studies.
phenotype. Genes Nutr 2008;3:519. Genes Nutr 2010;5:189203.

Vous aimerez peut-être aussi