Sie sind auf Seite 1von 14

Iranian

Iranian J Publ Health, Vol. 39, No.4, 2010 , pp.J1-Publ


14 Health, Vol. 39, No.4, 2010, pp.1-14 Review Article

Nutrigenomics and Nutrigenetics

*DD Farhud 1, M Zarif Yeganeh2, M Zarif Yeganeh3

1
School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
2
Obesity Research Center, Research Institute for Endocrine Sciences, Medical Faculty, Shahid Beheshti
University of Medical Sciences, Tehran, Iran
3
Rasoul Akaram Hospital, Iran University of Medical Sciences, Tehran, Iran

(Received 29 Mar 2010; accepted 3 Oct 2010)

Abstract
The nutrients are able to interact with molecular mechanisms and modulate the physiological functions in the body. The
Nutritional Genomics focuses on the interaction between bioactive food components and the genome, which includes
Nutrigenetics and Nutrigenomics. The influence of nutrients on f genes expression is called Nutrigenomics, while the
heterogeneous response of gene variants to nutrients, dietary components and developing nutraceticals is called Nutrigenet-
ics. Genetic variation is known to affect food tolerances among human subpopulations and may also influence dietary
requirements and raising the possibility of individualizing nutritional intake for optimal health and disease prevention on the
basis of an individual’s genome. Nutrigenomics provides a genetic understanding for how common dietary components af-
fect the balance between health and disease by altering the expression and/or structure of an individual's genetic makeup.
Nutrigenetics describes that the genetic profile have impact on the response of body to bioactive food components by
influencing their absorption, metabolism, and site of action.
In this way, considering different aspects of gene–nutrient interaction and designing appropriate diet for every specific
genotype that optimize individual health, diagnosis and nutritional treatment of genome instability, we could prevent and
control conversion of healthy phenotype to diseases.

Keywords: Nutritional genomics, Nutrigenomics, Nutrigenetics, Genetic variation

Introduction as cofactors or substrates in metabolic pathway,


With the completion of human genome sequenc- but much less is known about the impact of
ing and entering the-Omics area, the new term cofactors and/or micronutrients deficiency or ex-
"Nutritional Genomics" tends to replace the for- cess on the fidelity of DNA replication and re-
mer "nutrient-gene interactions" (1). It has been pair (3). Although the nutrients can influence the
demonstrated that numerous genetic polymor- development of a particular phenotype, the re-
phisms can influence protein structure function. sponse to a specific nutrient that determined by the
The Nutritional genomic area includes two parts: individual genotype has also to be considered
first Nutrigenomics that is the study of interaction (Fig. 1).
between dietary components and the genome, and The central role of genetic code in determining
the regulating changes in proteins and other me- genome stability and related health outcomes such
tabolism; second Nutrigenetics that identify the as developmental defects, degenerative diseases,
response to dietary components with regard to and cancer is well-established (4). The etiology
genetic differences (2). of complex chronic diseases obviously relates to
Nutrients are as environmental factors can inter- both environmental and genetic factors (5). Spe-
act with genetic material. It has been clearly dem- cifically, the "fetal basis of adult disease" or "early
onstrated that DNA metabolism and repair de- origins hypothesis" postulates that nutrition and
pend on a wide range of dietary factors that act other environmental factors during prenatal and

*Corresponding author: Tel: +98 21 88908447, Fax: +98 21 88803003, E-mail: farhud@sina.tums.ac.ir 1
DD Farhud et al: Nutrigenomics and Nutrigenetics…

early postnatal development influence gene ex- (5). Many human studies have demonstrated the
pression and cellular plasticity, which can alter evidence for interaction between SNPs in vari-
susceptibility to adult diseases (cardiovascular dis- ous genes and the metabolic response to the diet.
eases, diabetes, obesity. etc) (6). Moreover, SNPs analysis provides a potential
The concept of nutrients effects on DNA stability, molecular tool for investigating the role of nutri-
repair and on the different gene expression proc- tion in human health, diseases and identification
esses, recently became more prominent in nu- of optimal diets (9)
tritional science (7). Numerous dietary compo- Nutrients and genome interact at two levels: 1)
nents can alter genetic and epigenetic events and Nutrients can induce or repress gene expression
therefore influence health (8). thereby altering individual phenotype. 2) Conver-
SNPs (single nucleotide polymorphisms) are the sely, single nucleotide polymorphisms can alter
most common genetic variation, occur at about the bioactivity of important metabolic pathways and
500-2000 bp throughout the human genome, and mediators and influence the ability of nutrients
normally found in at least 1% of the population to interact with them (Fig. 1).

Polymorphic and Mutant Genes


(Changed enzymatic and hormonal activities)

Macronutrients (proteins, lipids, etc)


Micronutrient (minerals, vitamins, etc)

Fig.1: Nutrigenomics and Nutrigenetics are resulted from genes and nutrients interaction.

Genetic responses involve: effect on genome evolution, mutation, selection, programming, viability, gene expression,
chromosome stability, signal transduction and metabolic pathways, protein synthesis and structure, epigenetic events,
chronic diseases.
Nutritional responses involve: effect on nutrients absorption, nutrients utilization and requirement, food/nutrient tolerance,
and food atopies

2
Iranian J Publ Health, Vol. 39, No.4, 2010, pp.1-14

Nutritional Genomics chromatosis), which can be reasonably well man-


The interaction between the nutrients and cellu- aged with dietary restrictions (15).
lar/genetic processes is being referred to as As mentioned earlier SNPs study can be catego-
"nutritional Genomics" (10). This term describes rized in the field of Nutrigenetics. Some specific
the interface of biochemistry genomics, human examples of the association between SNPs and
nutrition, understanding of reactions and interac- specific food components such as enzymes defi-
tions at the molecular genomic levels (11). The ciency are reviewed in this article. For example,
conceptual basis for this genomic research can different mutations in galactose-1-phosphate uridyl-
be summarized with the following five princi- transferase (GALT) gene (14-18), phenylalanine
ples: 1) Common dietary chemicals act on the hydroxylase gene (19, 20), and Glucose-6-phos-
human genome, either directly or indirectly, to phate dehydrogenize (G6PD) gene (21-24) resulted
alter gene expression and/or structure. 2) Under in Galactosemia, Phenylketonuria (PKU), and
certain circumstances and in some individuals, Favism diseases, respectively. Other examples of
diet can be a serious risk factor for a number of enzymes polymorphisms include Lactase-phlorizin
diseases. 3) Some diet-regulated genes (and their hydrolase gene (LPH) polymorphisms that show
normal, common variants) are likely to play a how SNPs alter gene expression. This polymor-
role in the onset, incidence, progression, and/or phism is in the upstream of the lactase-phlorizin
severity of chronic diseases. hydrolase gene (LPH) associated with hypolactasia
4) The degree to which diet influences the bal- and changes tolerance to dietary lactose (milk
ance between healthy and disease states may de- sugar, LPH hydrolyzes lactose into glucose and
pend on an individuals genetic background. 5) galactose) and allows different expression of the
Dietary intervention based on knowledge of indi- LPH (25, 26).
vidual nutritional requirement, nutritional status, Glutathione peroxide gene polymorphism is an-
and genotype (i.e., “individualized nutrition") can be other example. The association between selenium
used to prevent, relieve, or cure chronic disease. supplementation and reduced incidence of liver,
colon, prostate, and lung cancer in human has been
Nutrigenetics shown. However, no individuals may respond
Nutrigenetics term was used first time by Dr equally. Glutathione peroxide is a selenium-de-
R.O Brennan in 1975 in his book Nutrigenetics pendent enzyme that acts as an antioxidant en-
(12). Nutrigenetics points to understanding how zyme. Polymorphism at codon 198 of human
the genetic background of an individual impact glutathione peroxides results in a subsituation of
to the diet (13). proline to leucine amino acid, and has been
The study of gene-nutrient interaction is a devel- associated with an increase risk of lung cancer.
oping area of science. This idea that adverse Investigators shown that persons with (Pro/Lue)
diet/genome interaction can cause disease is not genotype were at 80% greater risk for lung cancer
new and the unsuitable diet for any individual and (Lue/Lue) genotypes were at 130% greater
genotype could be a risk factor for monogenetic risk compared risk those with the (Pro/Pro)
and polygenetic disease (10, 14). Genetic polymor- genotype. The leucine-coding allele was less re-
phisms can influence response to environmental sponsive to increased activity because of sele-
elements, such as enzymatic activities changes that nium supplementation as compared with the prolin-
affect circulating concentrations and ultimately the containing allele (8).
effectiveness of chemicals and their metabolites Manganese super oxide dismutase (MnSOD) is
(5). Furthermore, metabolic disorders are other a mitochondrial enzyme that plays a key role in
examples of influence of the genetic variations to detoxification of reactive oxygen species. A poly-
diet such as PKU, defects associated with long morphism valine to alanin subsituation in in this
chain fatty acid oxidation, iron absorption (haemo- enzyme alters its transport into mitochondria,

3
DD Farhud et al: Nutrigenomics and Nutrigenetics…

which has been associated with increased risk of the negative health effect of these SNPs with de-
breast cancer (8). crease in plasma homocysteine levels (2, 27, 28).
Methylen tetrahydrofulate redoctase (MTHFR) Enzymes that utilize and metabolize vitamin B12
enzyme catalyzes the reaction that produces 5- have been associated with NTDs, increased risk
methyl tetrahydrofolate. The one-carbon units of Down syndrome and colon cancer. For ex-
are carried on N-5 or N10 of tetrahydrofolate. ample, a common polymorphism in the HFE gene
One-carbone metabolism is needed for the de- (Cys282Tyr) is associated with iron storage disease
novo synthesis of purine nucleotides and thymidi- hereditary haemochromatosis, leading to an iron
late and for the remethylation of homocyisteine to accumulation in the liver, heart and endocrine
methionine. With methionine adenylation S-adeno- glands. This protein is an important regulator of
sylmethionine (SAM) is formed, which is a co- cellular iron homeostasis and has role in intestinal
factor for numerous methylation reactions such iron absorption by regulating the interaction of
as DNA methylation that affect gene regulation the transferrin receptor with transferrin (27).
(27). For the MTHFR gene tow important SNPs Cytochrome P450s (CYPs) enzymes play a cen-
has been well recognized: C677T (cytosine-to- tral role in the oxidative biotransformation of ster-
thymidine subsituation resulting in the conversion oids, prostaglandins, nutrients, drugs, chemicals
of an alanine to valine) and A1298C (adenine- and carcinogens. Several dietary factors can alter
to-cytosine subsituation resulting in the conversion the expression of CYP isoforms. CYP1A2 plays
of an alanine to glotamic acid). The C677T poly- an essential role in the metabolism of wide range of
morphism is the most common variant that occurs drug and chemical substances. For example,
as homozygous T/T in 5-10% of the and as het- CYP1A2 activates dietary carcinogens such as
erozygous C/T genotypes up to 40% general po- aromatic amines, but also detoxifies compounds
pulation (28). The presence of C677T or A1298C such as caffeine. Low-activity CYP1A2 genotype
mutations is associated with reduction in MTHFR with an increased risk of myocardial infarction
enzyme activity and impairs folate accumulation, suggests that this enzyme detoxify a substance,
which may cause increases homocysteine concen- which may be an important risk factor in the po-
tration in plasma, a risk factor for venous throm- pulation. Indeed, individuals with a low-activity
boembolic and ischemic arterial diseases (2). CYP1A2 genotype are at a greater risk of cof-
Another polymorphism of MTHFR gene is fee-associated heart disease. As caffeine is the
Ala222Val that affects folate metabolism. It in- main substance in coffee and is detoxified by
creases the conversion of dUMP to dTMP and CYP1A2, it may be an important risk factor for
leads to more folate-dependent thymidine biosyn- heart disease in certain population (5).
thesis and folate deficiency (27). This polymor- Glutathione S transferase (GST) enzyme is a su-
phism is a risk factor for spontaneous abortions perfamily of enzymes that play an important role
and decreased fetal viability, thus maternal folate in the detoxification of several dietary compounds.
supplementation can be useful for individuals with GSTM1, GSTT1 and GSTP1 are isoforms of this
this polymorphism (29). enzyme. The GSTM1 and GSTT1 null genotype
MTHFR is also involved in maintenance genomic have been associated with both an increased and
CpG methylation patterns and prevention of DNA a decreased risk of some types of cancers such
strand breaks, these mutations are associated with as breast cancer (5, 30). Some components such
increased risk of neural tube defects and some as dietary isothiocyanates that are found in cruci-
types of cancer (27). ferous vegetables are eliminated with GSTs en-
Changes in the concentration of folate (the MTHFR zymes. Indeed, protective effect of the GSTM1
substrate) and riboflavin (the MTHFR cofactor) null genotype on colon and lung cancer has been
can modulate the activity of MTHFR gene (28). related to lower urinary excretion of glutathione-
Generally, folic acid supplementation can help conjugated phytochemicals indicating they are not

4
Iranian J Publ Health, Vol. 39, No.4, 2010, pp.1-14

rapidly excreted. GSTT1 plays a similar role to some 11q and highly polymorph and has a
GSTM1 in eliminating beneficial phytochemicals specific SNP in its promoter region (19, 20). An
found in cruciferous vegetables. Moreover, in ve- Adenin/Guanin subsituation in the promoter region
getables rich in phytochemicals such as isothio- (-75bp) of the ApoA1 gene is common in differ-
cyanates the expression of GSTs is increased ent populations. The presence of A allele (A/A
conjugating them to more water-soluble forms and A/G) has been associated with incresed HDL-
that are easily excreted (5). cholesterol. Moreover, mild increase in APOA-1
Endothelial nitric oxide synthase (eNOS) is syn- concentrations in subjects with the G/G geno-
thesized from the amino acid L-argenine by NO type was observed (28, 30). APOA-5 gene is
synthase (NOS). The eNOS is expressed in the also an important regulator of triglyceride (TG)-
endothelium and produces NO that diffuses to rich lipoprotein (TRL) metabolism (30).
vascular smooth muscle cell, where it increases One of the Vitamin D receptor (VDR) polymor-
the concentration of cGMP, leading to vascular phism is Fok1. Individuals with F allele have three
relaxation. NO has central role in the pathogenesis amino acids more than those without F allele in
of coronary spasm and atherogenesis. Several their VDR. The Ff or ff genotype is associated
polymorphisms of eNOS may be associated with with 51% and 84% greater risk of colorectal
specific phenotype. For example, a Glu298Asp cancer, respectively. Individuals that consumed
polymorphism in the eNOS gene has been associ- low calcium and fat diet have more than double
ated with ischemic heart disease, myocardial in- risk of colorectal cancer, specifically in persons
farction, and coronary spasm (30). with ff genotype rather than Ff genotype (8).
Genetic polymorphisms in catechol-O-methyltrans- VDR polymorphisms have been also associated
ferase, sulfotransferase, and UDP-glucuronosyl- with childhood and adult's asthma (27).
transferase result in differences in enzymatic ac- Peroxisome proliferator-activated receptors (PPARs)
tivity. These enzymes metabolize some of dietary are nuclear receptor supper family that plays an
compounds. For example, green tea was associ- essential role in fatty acid oxidation, glucose, and
ated with a lower risk of breast cancer only in extracellular lipid metabolism. PPARs are the
women with the low-activity allele for catechol- best-known fatty-acid-regulated nuclear receptors.
O-methyltransferase. This enzyme catalyzes the me- One of the three members of the PPARs family
thylation of catechins (a polyphenolic antioxidant regulates many genes involved in fatty acid me-
plant secondary metabolite) in green tea making tabolism. PPR-α (PPARA) plays a central role in
them more quickly eliminated (5). lipid oxidation and inflammation, whereas PPAR-γ
Apolipoprotein E (ApoE) gene has three differ- is involved in adipocytes differentiation, glucose
ent alleles (ε2, ε3, ε4). Persons with ε4 variant and lipid storage, and inflammation. PPAR-δ
respond to a high-fat diet negatively with an in- (also known as PPAR-β), may has a crucial role
creased risk for coronary heart disease (CHD). in development, lipid metabolism, and inflam-
In these individuals, low-fat diet should be use- mation. These receptors bind to fatty acid and
ful (2). Moreover, there is an important relation- regulate the expression of genes involved in fatty
ship between allelic variants in the ApoA1/C3/ acid transport and metabolism. PPARs family also
A4/A5 genes and the effect of dietary fats on involve in activation of about 300 genes (31).
lipoprotein metabolism and CVD (cardio vascular The PPAR-α gene has a polymorphism at codon
diseases) risk. Linkage disequilibrium within Apo 162 (Lue162Val) that has been associated with
A1/C3/A4/A5 cluster has been represented to changes in total cholesterol, LDL-associated cho-
affect plasma lipid concentration and CVD risk. lesterol, and Apo B concentrations. The less
Apolipoprptein A-1 is and is a key component common V162 allele is associated with signifi-
of high-density lipoprotein particles (HDL). The cantly higher serum concentration of total cho-
locus of gene encoding APOA-1 is on chromo- lesterol, LDL cholesterol, Apo B, and Apo C-III

5
DD Farhud et al: Nutrigenomics and Nutrigenetics…

than in carriers of L162 allele, especially in men may act as ligands for transcription factor re-
(20). For individuals with the common L162 ceptors (36, 37) or be metabolized by primary or
allele, increased intake of polyunsaturated fatty secondary metabolic pathways, thereby altering
acids (PUFAs) had little effect on fasting tria- concentrations of substrates or intermediates, and
cylglycerol concentrations. In those with the less finally positively or negatively affect signal path-
common V162 allele, however, fasting triacyl- ways (38-40).
glycerol concentrations fell abundantly with in- Transcription factors (TFs) are one of the key
creasing PUFA intake (32). molecules through with nutrients can alter the
gene expression. One of the most important groups
Nutrigenomics of nutrient sensors is PPARs TFs with 48
Nutrigenomics aims to identify the effects of members in the human genome. The majority of
several nutrients, including macronutrients and receptors in this superfamily bind nutrients, their
micronutrients on the genome (13) and explores metabolites, and influences expression of spe-
the interaction between genes and nutrients or cific genes involved in numerous metabolic proc-
food bioactives and their effects on human health ess in the liver, including fatty acid oxidation,
(33). The influence of nutrients on the transcription ketogenesis, gluconeogenesis, amino acid metabo-
activity, gene expression, and heterogeneous re- lism, cellular proliferation, and acute-phase re-
sponse of gene variants is also referred to as "Nutri- sponse (41). For example, the fatty acids palmitic
genomics". (16:0), oleic (18:1n9), linoleic (18:2n6), and ara-
Nutrigenomics also describes the use of func- chidonic acid (20:4n6) (42-45), and the eico-
tional genomic tools to study a biological system sanoids, 15deoxy-δ12, 14prostaglandinJ2 and 8-
to understanding of how nutritional molecules (S) hydroxyeicosatraenoic acid, are ligands for
affect metabolic pathways and homeostatic con- PPAR-δ (46-48). These nuclear receptors act as
trol. This branch of science will reveal the opti- sensors for fatty acids. Lipid sensors usually het-
mal diet form within a series of nutritional changes, erodimerize with retinoid receptor, whose ligand
whereas Nutrigenetics will yield critically important is derived from another dietary chemical, vitamin
information that assist clinicians in identifying A, and hyperforin, bind directly to nuclear re-
the optimal diet for a given individual, i.e. per- ceptors and influence gene expression (Table 1).
sonalized nutrition (13). Transcriptomics, proteo- The liver X receptor-α (binding cholesterol me-
mics, and metabolomics are also technologies tabolites), bind as a heteromers to specific nu-
that apply in Nutrigenomics research (33). cleotide sequence (response elements) in the pro-
According to numerous studies, nutrients can al- moter regions of a large member of genes. During
ter the expression of genes at the level of gene ligand binding, nuclear receptors undergo a con-
regulation, signal transduction, chromatin struc- formational change that results in coordinated dis-
ture and protein function (33). sociation of corepressors and recruitment of coac-
Epidemiological studies show association between tivator proteins to prepare transcriptional activation
food intake and the incidence and severity of (41). Thereby, a number of genes are induced
chronic diseases (34, 35). A large number of nu- such as those involved in fatty acid oxidation or
trition related pathologies (obesity, metabolic syn- fatty-acid storage, depending on the cellular meta-
dromes, type 2 diabetes, CVD, and some types bolic state (31). In metabolically active organs,
of cancers) are polygenic and multifactorial and such as the liver, intestine, and adipose tissue,
their onset and progression are related to multiple these TFs act as nutrient sensors by changing the
genes and their variants as well as several en- level of DNA transcription of specific genes in
vironmental factors, especially the diet (30). response to nutrients changes (41).
Dietary chemicals can affect gene expression di-
rectly or indirectly. At the cellular level nutrients

6
Iranian J Publ Health, Vol. 39, No.4, 2010, pp.1-14

Table 1: Nuclear receptors and dietary ligands. Umbers in parentheses indicate percent activity after ligand binding rela-
tive to estradiol (36, 37, 50, 52, 53)

Regulation Receptor Type Endogenous ligand Dietary ligand


ERα
17β-Esteradiol (100)
Endocrine: hormonal lipids: ERβ
Estrogen 17β-Esteradiol (100) Genisteine (4)
feedback paradigm
Progestrone

Progestrone Testoterone Genisteine (87)


Androgen 5α -dihydrotestosterone
Endogenous metabolism
Androgen Aldosterone cholestrol precursor
Glucocorticoid Cortisol

RARα All-trans retinoic acid Vitamin A


Mixed paradigm Retinoic acid RARβ All-trans retinoic acid Vitamin A
RARγ All-trans retinoic acid Vitamin A
TRα Iodine
Thyroid
TRβ Iodine
Vitamin D 1,25-dihydroxyvitamin D Vitamin D/Sunshine
Ecdisone Cholesterol derivatives Cholesterol
Lipid sensors: dietary lipids:
Retinoid X C is-9-retinoic acid Docsahexaenoic acid
feed-forward paradigm
PPARα FA
Pristinic/phytanic
PPAR PPARβ FA/eicosanoids
Pristinic/phytanic
PPARδ ?
Hyperforin
Estrogen
Pregnan X Coumesterol
Progesterone Progesterone
Genisteine
Liver X Oxysterols Cholesterol metabolites
Famosoid X Bile acids

Dietary chemicals indirectly regulate some of gallate (EGCG) that EGCG inhibits tyrosine
TFs. The sterol regulatory element binding pro- phosphorylation of Her-2/neu receptor and epi-
teins (SREBPs), for example, are activated by dermal growth factor receptor that reduces sig-
protease cleavage, an event regulated by low naling via the phosphatidyl inositol 3-kinase (PI-
levels of oxy sterols and changes in insulin/ 3)-AKt kinase-NF-kB pathway. Activation of the
glucose and PUFAS (49). The carbohydrate-re- NF-kB pathway is associated with some types of
sponsive element-binding protein (chREBP) is a breast cancer (50, 52, 53).
large TF, activated in response to high glucose PUFAs such as n-3 and n-6 are other micronutri-
levels, and is regulated by reversible phosphoryla- ents, which are also referred to as omega-3 and
tion events (50). This DNA binding protein serves omega-6 fatty acids, may influence gene expres-
as an effector of lipogenic gene expression (51). sion. Animal studies have demonstrated that PUFA
Moreover, dietary chemicals can directly affect intake can modulate the gene expression of sev-
signal transduction pathways. For example, green eral enzymes involved in lipid and carbohydrate
tea contains the polyphenol, 11-epigallocatechin-3- metabolism. A significant interaction has also been

7
DD Farhud et al: Nutrigenomics and Nutrigenetics…

shown for the PPARA Lue162Val polymorphism (Vit B, E, and carotenoids), cancer (folate,
n-6 PUFA intake. Individuals with the less com- carotenoids), neural tube defects (folate), and
mon V162 allele, increased n-6 PUFA intake is bone mass (Vit D) (54). B6, B12 and folate defi-
associated with a marked reduction in triacylglyc- ciencies, for example, are associated with in-
erol concentration, whereas this association is creased serum homocysteine levels. Hyperho-
not observed in L162 carriers. Conversely, in mocysteinemia is a risk factor and marker for
L162 and V162 carriers n-3 PUFA intakes results coronary artery disease. Deficiency of Vit B12,
in triacylglycerol concentrations reduction (32). folic acid, B6, niacin, C or E, iron or zinc
Approximately 40 micronutrients are needed in appears to imitate radiation in damaging DNA
the human diet. Suboptimal intakes of specific by causing single and double-strand breaks,
micronutrients have been associated with CVD oxidative lesions, or both (55), (Table 2).
Table 2: Examples of the role and effect of specific micronutrients deficiencies on genomic stability (56, 57)
Micronutrients Role in genomic stability Consequence of deficiency
Vits C and E Prevention of DNA and lipid oxidation. Increased baseline level of DNA strand
breaks, chromosome breaks, oxidative
Antioxidant activity by increasing glutathione level in DNA lesions and lipid peroxide adducts
Vit D on DNA.
normalm cell, inducuction apoptosis in cancer cells.
uracil misincorporation in DNA, increased
Folate and Maintenance methylation of DNA, synthesis of dTMP
chromosome breaks and DNA
Vits B2, B6, B12 from dUMP and efficient recycling of folate.
hypomethylation.
Required as substrate for poly (ADP- ribose)
Increased level of unrepaired nicks in DNA,
polymerase which is involved in cleavage and
Niacin, Nicotinic acid increased chromosome breaks and
rejoining of DNA and telomere length
rearrangement, sensitivity of mutagens.
Maintenance and DNA repair.
Zn, required as a cofactor for Cu/Zn supperoxid
dismutase, endonuclease IV, P53 function, DNA
replication and Zinc finger proteins such as poly (ADP-
Zinc, Manganese and ribose) polymerase. Increased DNA breaks and oxidation,
Selenium Mn, required as a component of elevated chromosomal damage rate.
mitochondrial Mn superoxide dismutase.
Se, required as a component of peroxidases e.g.
glutathione peroxidase.
Reduced DNA repair capacity, increased
Required as a component of ribonucleotide reductase
Iron propensity for oxidative damage to
and mitochondrial cytochromes.
mitochondrial DNA.
Mg, required as a cofactor for a variety of DNA
Reduced fidelity of DNA replication,
polymerases, in nucleotide excision Repaire, base
reduced DNA repaire capacity,
excision repair and mismatch Repair, essencial for
chromosome segregation Errors,survival of
Magnesium, Calcium microtubule Polymerization and chromosome
genomically aberrant cells.
segregation. Ca, plays an important role in
chromosome Segregation and is required for
apoptosis.

Nutrient deficiencies are more important than Amino acids can play the role of nutritional sig-
radiation because of constancy of exposure to nals in the modulation of expression of particu-
milieu promoting DNA damage (58-60). For lar genes. Studies have shown that cells can de-
example, folate deficiency breaks chromosomes tect variants in amino acid levels and respond by
due to substantial incorporation of uracil in hu- mechanism as control of transcription, mRNA
man DNA (4 million uracil/cell) (61). stabilization ,as well as by up or down regulation

8
of translation initiation (62). For example, in cleotide binding proteins, histon-modifing enzymes,
human cells amino acid L-tryptophan in supra- chromatin-remodeling factors, and their multi-
physiologic concentrations is a powerful inducer molecular complexes are involved in the overall
of collagenase gene expression at a transcrip- epigenetic process (8). The best studied epige-
tional level. The increase in collagenase mRNA netic modification is DNA methylation and in the
levels was reversible, time and L-tryptophan dose- mammals genome occurs at many of cytosine
dependent (63). residues that are followed by guanine residue
Simple and complex carbohydrates have differen- (CpG islands) and in most cases methylation in
tial effects on blood glucose concentrations. Foods these regions induces gene repression. However,
with a high glycemic index (GI) would increase this phenomenon can lead to the expression of
insulin production and, decrease synthesis of insu- neighboring genes (67). Studies identify that DNA
lin receptors. High glucose concentration also methylation is dependent on bioactive food com-
induces the transcription of several genes of the ponents ranging from alcohol to zinc (8) (Table 4).
glycolytic and lipogenic pathways (64).
Therefore, dietary chemicals are regularly ingested Table 4: Nutrients and chemicals involved in DNA
methylation (8, 68)
and are involved indirectly and directly in regu-
Micronutrients
lation gene expression, it follows that a subset of Alcohol Genistein
genes regulated by diet must be involved in disease Arsenic Methionine
initiation, progression, and severity (65, 66). Betaine Nickel
Cadmium Polyphenol
Choline Selenium
Nutritional epigenetics Coumestrol Vitamin A
The term "epigenetics" is used to gene expres- Equol Vitamin B6
sion that occurs without changes in the DNA se- Fiber VitaminB12
Folate Zinc
quence. Epigenetic regulation plays an impor-
tant role in development and is needed to gain Several dietary factors may influence the provi-
stable expression or repression of genes in spe- sion of methyl groups available for the formation
cific cell types or at defined developmental stages of S-adenosylmethinine. Moreover, dietary factors
(67). Epigenetic changes may influence cell cycle may modulate the use of methyl group by proc-
control, DNA damage, apoptosis, invasion, im- esses including change in DNA methyltransferase
printing, and aging (8). activity. The methyl groups’ status depends on
Epigenetic events can be modified by bioactive B vitamins as cofactors including folate, Vit B12,
food components (Table 3). and Vit B6 (29). The folate-dependent biosyn-
Table 3: Some nonessential nutrients and bioactive food thesis of nucleotide precursors for DNA synthe-
components that can alter genetic and epigenetic events (8) sis and of SAM for genome methylation is de-
pendent on the availability of many vitamins in-
Nutrient group Example cluding B12, B6, niacin, riboflavin and minerals
Isothiocyanates, allyl sulfur
Phytochemicals
Carotenoids, flavonoids, indoles,
(zinc, cobalt). Therefore, folate-mediated one-
carbon metabolism mediates communication be-
Zoochemicals Conjugated linoleic acid, n-3 fatty acids
tween the cellular nutrient environment and regula-
β-glucans, lentinan, schizophylan, and
Fungochemicals
other compounds in mushrooms.
tion of the genome. Impairments in one-carbon
metabolism and the SAM cycle induced by nu-
Equol, butyrate, and other compounds
Bacteriochemicals formed from gastrointestinal flora tritional deficiencies and/or SNPs in genes that
fermentation encode folate-dependent enzymes, alter genome
methylation patterns and gene expression levels.
A majority of regulatory proteins including DNA Disruptions in folate metabolism are common and
methyltransferases, methyl-cytosine guanine dinu- increase risk cancers, cardiovascular disease, neu-

9
DD Farhud et al: Nutrigenomics and Nutrigenetics…

rological disorders and developmental anomalies and biotin) affect genome stability in human in
such as spina bifida, cleft palate, and spontaneous vivo (4). Folate and vitamin B12 are need for
abortion. Therefore, folate supplementation can re- DNA replication, repair and maintenance of
duce the risk of these disorders developing (69). DNA methylation patterns. Both in vivo and in
DNA methylation patterns may effect on the re- vitro studies with human cells clearly show that
sponse to bioactive food components and thereby folate and vitamin B12 deficiencies and elevated
account for differences in response in normal and plasma homocysteine are associated with the
neoplastic cells (70). expression of chromosomal fragile sites, chromo-
somal breaks, excessive uracile in DNA and DNA
Genome health and disease prevention hypomethylation. Nicotinic acid (niacin) also plays
It is clear that even the small damages in the ge- a fundamental role in chromosome integrity and
nome can cause crucial effects in whole human reduction of cancer risk (Table 2) (70).
life. DNA metabolism and repair is depending Reactive oxygen species (ROS) such as highly
on a variety of dietary factors that act as cofac- reactive hydroxyl radical and superoxide radical
tors or substrates. Nutritional requirements is contributes to DNA damage. Antioxidants (Vit
important for the prevention of DNA oxidation C and E) and enzymes such as superoxid dismu-
(i.e. antioxidants such as carotenoids, Vit E and tase, catalase and glutathionperoxidase may con-
C), prevention of uracil incorporation into DNA trol lipid and protein oxidation induced by ROS (3).
(i.e. folate), maintenance methylation of CpG in Since developmental, degenerative diseases and
DNA (methionine, cholin, folate and vitamin aging are partly caused by DNA damage, defin-
B12), as cofactors or as components of DNA re- ing optimal requirements of key minerals and
pair enzymes (Zn, Mg), maintenance of telomere vitamins for preventing nuclear and mitochon-
length (niacin, folate) (56, 69, 70). drial DNA damage is important (70).
Many chronic diseases are polygenic and result Infertility is another consequence of genome dam-
from interaction between genes and environ- age on human health. Genome damage results
mental factor. Dietary intervention based on nutri- from specific micronutrient deficiencies may cause
tional requirement, nutritional status, and genotype developmental defects in the fetus or increased
(i.e., “individualized nutrition"), can be used to pre- risk of cancer in the child. For example, inadequate
vent, control or treatment of chronic disease such Vit C intake results in increased oxidation of
as cardiovascular diseases (CVD), metabolic syn- sperm DNA; folate deficiency increases risk of
dromes, and cancer (41). These disorders are NTDs and genome damage. Increased risk of
partly mediated by chronic exposure to certain childhood leukemia in children with mothers who
food components. Fore example, the association did not intake enough folic acid supplementation
between amount of calories (35), the levels and during pregnancy has demonstrated. Additionally,
types of vitamins (71), fat (72), and carbohydrates zinc deficiency induces oxidative damage to DNA
with atherosclerosis, diabetes, obesity, cancer, and impairs DNA repair, which has a terato-
hypertension, and other chronic diseases is dem- genic effects (70, 74).
onstrated (73).
Telomere and nutritional status
Genome damage and nutritional deficiency Telomeres are nucleoprotein structures that cap
As mentioned earlier, nutritional status influences the ends of chromosomes, and maintain chromo-
genome stability and deficiency of certain mic- some stability. Degeneration of telomeres leads
ronutrients can result in critical damages in the to whole chromosomal instability, and chromo-
genome. Studies have shown that at least nine somal fusion and therefore gene amplification,
micronutrients (Vit E, Ca, folate, retinol, nico- an important risk factor for cancer (3). Folate
tinic acid, β-caroten, riboflavin, pantothenic acid, and nicotinic acid deficiency increased oxidative

10
stress and telomere dysfunction. Under folate other chronic diseases, and perhaps increase hu-
deficiency uracil is incorporated into DNA in- man longevity.
stead of thymidine, leading to chromosome break-
age. Similarly, oxidative stress causes telomere
Ethical Considerations
shortening. Folate and other methyl donors such
All ethical issues including plagiarism, Informed
as Vit B12, cholin, and methionin have an im-
Consent, misconduct, data fabrication and/or
portant role in maintenance methylation of cyto-
falsification, double publication and/or submis-
sine. Defects in the DNA methylation can cause
sion, redundancy, etc have been completely ob-
excessive telomere elongation and homologous
served by the author.
recombination between telomeres and telomeres
fusion. Hypomethylation or hypermethylation of
the CpG islands is in the promoter of telomerase Acknowledgments
may cause excessive expression of telomerase or The authors are thankful to Prof. A. Jazayery, Dept.
silence the gene respectively (3). of Nutrition, school of Public Health and Dr. H.
Telomere shortening has been observed in num- Sadighi, Genetic Clinic, for their critical comments
ber of conditions including obesity, psychologi- and corrections. The authors declare that they
cal stress, immune dysfunction, cancer, and have no conflicts of interest.
CVD. In vitro studies antioxidant treatment has
been found to prevent telomere damage (3).
References
1. Shils ME, Shike M, Ross A.C, Caballero B,
Conclusions J-Cousins R (2006). Modern Nutrition in
Nutritional genomics elucidate the interaction among Health and Disease. 10th ed. lippincott Wil-
nutrients, metabolic intermediates, and the mam- liams A wolters Kluwer Company. Phila-
malian genome. The response to bioactive food delphia.chapter:39.
components is dependent on genetic background 2. Subbiah MT (2007). Nutrigenetics and nutra-
(Nutrigenetics effects) that can influence absorp- ceuticals: the next wave riding on person-
tion and metabolism targets or sites of action. alized medicine. Transl Res, 149(2): 55–61.
Likewise, the response to food components de- 3. Bull C, Fenech M (2008). Genome health
pends on DNA methylation and other epigenetic nutrigenomics and nutrigenetics: nutritional
events. The ability of bioactive food components requirements for chromosomal stability and
to influence gene expression patterns (nutrigenom- telomere maintenance at the individual level.
ics effects) is also a factor in determining the Proc Nutr Soc, 67(2):146-56.
overall response. Finally, bioactive food compo- 4. Fenech M (2008). Genome health nutrigenomics
nents may influence protein synthesis, degrada- and nutrigenetics-diagnosis and nutritional
tion, and posttranslational modification. Under- treatment of genome damage on an individual
standing the interrelationships among human ge- basis. Food Chem Toxicol, 46(4): 1365-70.
netics diversity, genome function, and dietary com- 5. El-Sohemy A (2007). Nutrigenetics. Forum
ponents will enable precise manipulation of genome Nutr, 60: 25-30.
function and stability throughout the life cycle for 6. Dolinoy DC, Jirtle RL (2008). Environmental
optimal human health and disease prevention. epigenetics in human health. Environ Mol
With greater insight in to the gene-nutrient inter- Mutagen, 49(1): 4-8.
action, alterations in diet and single nutrient inter- 7. Paoloni-Giacobino A, Grimble R, Pichard C
ventions may help us to better protect against can- (2003). Genetics and nutrition .Clin Nutr,
cer, decrease the occurrence of cardiovascular and 22(5): 429-35.

11
DD Farhud et al: Nutrigenomics and Nutrigenetics…

8. Trujillo E, Davis C, Milner J (2006). Nutri- 20. Brigeli S, Flohe R, Joost HG (2006). Nurti-
genomics, Proteomics, Metabolomics, and genomics (Translated in press, by Farhud
the Practice of Dietetics. J Am Diet Assoc, DD and Athari S).
106(3): 403-13. 21. Farhud DD, Sadighi H, Amirshahi P, Tavak-
9. Ferguson LR. (2006). Nutrigenomics: integrat- koli F SH (1993). Serum level measure-
ing genomics approaches into nutrition re- ments of Gc, Cp, IgG, IgA and IgM in pa-
search. Mol Diag Ther, 10(2):101-8. tients with Favism in Iran. Iranian J Public
10. Kaptur J, Raymond LR (2004). Nutritional Health, 22:1-4.
genomics: the next frontier in the Post ge- 22.Farhud DD, Yazdanpanah L (2008). G6PD
nomic era. Physiol Genomics, 16: 167-77. deficiency. Iranian J Public Health, 37(4):1-18.
11. Milner J. A (2004). Molecular Targets for 23. Farhud DD, Amirshahi P (1985). Ahaptoglo-
Bioactive Food Components. J Nutr, 134(9): binemia in Favism patients from Iran. Acta
2492S-2498S. Anthropogenet, 9(4): 206-13.
12. Simopoulos AP (2010). Genetic variants in 24. Hedayat SH, Farhud DD, Montazami K,
the metabolism of omega-6 and omega-3 Ghadiryan P (1981). The pattern of bean
fatty acids: their role in the determination consumption, laboratory findings in patients
of nutritional requirements and chronic with Favism, G6PD deficiency, and a con-
disease risk Experi Biol Med, 235:785-95. trol group. J Trop Pediatr, 27(2):110-13.
13. David M (2005). Mutch, Walter Wahli, Gary 25. Mark Lucock (2007). Molecular Nutrition and
Williamson. Nutrigenomics and nutrigenetics: Genomics (Translated in press, by Farhud
the emerging faces of Nutrition. FASEB J, DD and Zarif Yeganeh M).
19(12): 1602-16. 26. Enattah NS, Sahi T, Savilahti E, Terwilliger
14. Ames BN, Elson-Schwab I, Silver EA (2002). JD, Peltonen L, Järvelä I (2002). Identifi-
High-dose vitamin therapy stimulates variant cation of a variant associated with adult-
enzymes with decreased coenzyme binding type hypolactasia .Nat Genet, 30(2):233-7.
affinity (increased Km): relevance to genetic 27. Patrick J Stove (2006). Influence of human
disease and polymorphisms. Am J clin Nutr, genetic variation on nutritional requirements.
75(4): 616-58. Am J Clin Nutr, 83(2): 436S- 42S.
15. Palou A (2007). From nutrigenomics to per- 28. Virgili F, Perozzi G (2008). How Does
sonalized nutrition. Genes Nutr, 2(1): 5–7. Nutrigenomics Impact Human Health?
16. Farhud DD, Shalileh M (In press). Guidline IUBMB Life, 60(5): 341–44.
of nutritional regimens for Galactosemia, 29. Patrick J. Stover (2004). Nutritional genomics.
Dept. of Experimental Education. Physiol Genomics, 15; 16(2):161-5.
17. Krauss RM (2001). Dietary and genetic 30. Iacoviello L, Santimone I, Latella MC, de
effects on LDL heterogeneity. World Rev Gaetano G, Donati MB (2008). Nutrigenom-
Nutr Diet, 89:12-22. ics: a case for the common soil between car-
18. Ko DH, Chang HE, Song SH, Park KU, Kim diovascular disease and cancer. Genes Nutr,
JQ, Kim MC, Song YH, Hong YH, Lee 3(1):19-24.
DH, Song J (2010). Molecular and bio- 31. Ordovas JM (2006). Nutrigenetics, Plasma
chemical characterization of the GALT gene Lipids, and Cardiovascular Risk. J Am Diet
in Korean patients with galactose-1-phos- Assoc, 106(7):1074-81.
phate uridyltransferase deficiency. Clin Chim 32. Ordovas JM (2006). Genetic interactions
Acta, (in press). with diet influence the risk of cardiovascular
19. Farhud DD, Shalileh M (2008). Phenylke- disease. Am J Clin Nutr, 83(2):443S-446S.
tonuria and its dietary therapy in children. 33. El-Sohemy A (2008). The Science of Nutri-
Iranian J Pediatric, 18(suppl 1): 88-98. genomics. Health Law Review, 16(3): 5-8.

12
34. Willett W (2002). Isocaloric diets are of pri- receptor superfamily that is activated by a
mary interest in experimental and epide- peroxisome proliferators and fatty acids.
miological Studies. Int J EPidemiol, 31(3): Mol Endocrinol, 6(10):1634-41.
694-5. 46. Forman BM, Tontonoz P, Chen J, Brun RP,
35. Jenkins D JA, Kendall ccw, Ransom TPP Spiegelman BM, Evans RM (1995). 15-
(1998). Dietary fiber, the evolution of the Deoxy-delta 12, 14- Prostaglandin J2 is a
human diet and Coronary heart disease. Ligand for the adipocyte determination factor
Nutr Res, 18: 633-52. PPAR gamma .Cell, 83(5): 803- 12.
36. Dauncey MJ, White P, Burton KA, Katsumata 47. Kliewer SA, lenhard Jm, willsan TM, Patel I,
M (2001). Nutrition hormone receptor- gene Morris DC, Lehmann JM. (1995). A Pros-
interactions: implications for development taglandin J2 metabolite binds Peroxisome Pro-
and disease. Proc Nutr Soc, 60(1): 63 -72. liferator-activated receptor gamma and Pro-
37.Jacobs MN, Lewis DF (2002). Steroid hormone motes adipocyte differentiation. Cell, 83(5):
receptors and dietary ligands: a selected 813-19.
review. Proc Nutr Soc, 61(1): 105-22. 48. Kliewer SA, Xu HE, lambert MH, Willson
38. Clarke SD (1999). Nutrient regulation of gene TM (2001). Peroxisome Proliferator-activated
and protein expression. Curr opin Clin receptors: from genes to Physiology. Re-
Nutr Metab care, 2(4): 287-9. cent Prog Horm Res, 56: 239-63.
39.Eastwood MA (2001). A molecular biological 49. Masuda M, Suzui M, Weinstein IB (2001).
basis for the nutritional and Pharmacological Effects of epigallocatechin - 3 -gallate on
benefits of dietary plants. QJM, 94(1): 45-8. growth, epidermal growth factor receptor
40. Lin SJ, Guarentel (2003). Nicotinamide ade- signaling pathways, gene expression, and
nine dinucleotid, a metabolic regulator of chemosensitivity in human head and neck
transcription, longevity and disease. Curr squamous cell carcinoma cell lines. Clin
opin, cell Biol 15(2): 241-6. cancer Res, 7(12): 4220 -29.
41. Afman L, Müller M (2006). Nutrigenomics: 50. Uyeda K, Yamashita H, Kawaguchi T
From Molecular Nutrition to Prevention of (2002). Carbohydrate responsive element-
Disease. J Am Diet Assoc, 106(4): 569-76. binding protein (chREBP): a key regulator
42. Auwerx J (1992). Regulation of gene expres- of glucose metabolism and fat storage.
sion by fatty acids and fibric acid derivatives: Biochem Pharmacol, 63(12): 2075-80.
an integrative role for Peroxisome Proliferator 51. Kosaku Uyeda1 and Joyce J. Repa (2006).
activated receptors. The Belgian Endocrine Carbohydrate response element binding pro-
Society lecture. Horm Res, 38(5-6): 269-77. tein, ChREBP, a transcription factor cou-
43. Dreyer C, Krey G, Keller H, Givel F, pling hepatic glucose utilization and lipid
Helftenbein G, Wahli W (1992). Control synthesis. Cell Metabolism, 4: 107-10.
of the peroxisomal beta- oxidation pathway 52. Edwards PA, Tabor D, Kast HR, Ven-
by a novel family of nuclear hormone re- kateswaran A (2000). Regulation of gene
ceptors. Cell, 68(5): 879-87. expression by SREBP and SCAP. Biochim
44. Göttlicher M, Widmark E, Li Q, Gustafsson Biophys Acta, 1529(1-3): 103 -13.
JA (1992). Fatty acids activate a chimera 53. Nobel S, Abrahmsen L, Oppermann U (2001).
of the clofibric acid -activated receptor and Metabolic conversion as a Pre-receptor
the glucocorticoid receptor. Proc Natl Acad control Mechanism for lipophilic hormones.
Sci USA, 89(10): 4653-57. Eur J Biochem, 268(15): 4113-25.
45. Schmidt A, Endo N, Rutledge SJ, Vogel R, 54. Fairfield KM, Fletcher RH (2002). Vitamins
Shinar D, Rodan GA (1992). Identification For chronic disease Prevention in adults:
of a new member of the steroid hormone Scientific review. JAMA, 287(23): 3116-26.

13
DD Farhud et al: Nutrigenomics and Nutrigenetics…

55. Ames BN (2001). DNA damage from micro- abundance differently in mice with distinct
nutrient deficiencies is likely to be a major susceptibilities todiet–influenced diseases.
cause of cancer .Mutat Re, 475(1-2): 7-20. J Nutr, 127(4): 566-73.
56. Fenech M (2007). Nutrition and genome 66. Ross SA (2007). Nutritional genomic ap-
health. Forum Nutr, 60:49-65. proaches to cancer prevention research. Ex-
57. Pianetti S, Guo S, Kavanagh KT, So-nen- perimental oncology, 29(4): 250-6.
shein GE (2002). Green tea polyphenol 67. Stover PJ, Caudill MA (2008). Genetic and
epigallocatechin -3 gallate inhibits Her-2 Epigenetic Contributions to Human Nutrition
/neu signaling, Proliferation, and transformed and Health: Managing Genome-Diet Interac-
Phenotype of breast cancer cell. Cancer Res, tions.
62(3): 652-55. 68. Davis CD, Uthus EO (2004). DNA methyla-
58. Ames BN, Gold LS (2000). Paracelsus to tion, cancer susceptibility, and nutrient in-
parascience: the environmental cancer dis- teractions. Exp Biol Med 229: 988–995.
traction. Mutat Res, 447(1): 3-13. 69. Fenech M (2005). The Genome Health Clinic
59. Ames BN, Wakimoto P (2002). Are vitamin and Genome Health Nutrigenomics concepts:
and mineral deficiencies a major cancer diagnosis and nutritional treatment of ge-
risk? Nat Rev Cancer, 2(9): 694-704. nome and epigenome damage on an indi-
60. Blount BC, mack MM, Wehr CM, Mac vidual basis. Mutagenesis, 20(4): 255-69.
Gregor JT, Hiatt RA, Wang G, Wickrama- 70. Fairfield KM, Fletcher RH (2002). Vitamins
singhe SN, Everson RB, Ames BN (1997). For chronic disease Prevention in adults:
Folate deficiency causes uracil misincor- Scientific review. JAMA, 287(23): 3116-26.
poration in to human DNA and chromo- 71. Willet WC (2002) .Balancing life-style and
some breakage: implications for cancer genomics research for disease Prevention.
and neuronal damage. Proc Natl Acod Sci, Science, 296(5568): 695-8.
USA 94(7): 3290-95. 72. Genkins DJ, Kendall cw, Augustin LS, etal
61. Jefferson LS, Kimball SR (2001). Amino (2002). Glycemic index: overview of impli-
acid regulation of gene expression. J Nutr, cation in health and Disease. Am J clin
131(9 Suppl):2460S-6S. Nutr, 76:266S-273S.
62. Varga J, LiL, Mauviel A, Jimenez SA 73. Kumar D (2007). From evidence-based me-
(1994). L-Tryptophan in supraphysiologic dicine to genomic medicine. Genomic Med,
concentrations Stimulates collagenase gene 1(3-4):95-104.
expression in human skin fibroblasts. Lab 74. Fenech M, R.Ferguson L (2001). Vitamins/
Invest, 70(2): 183-91. minerals and genomic stability in humans.
63. Augustin LS, Franceschi S, Jenkins DJ, Mut Res, 475: 1-6.
Kendall CW, La Vecchia C (2002). Glycemic
index in choronic disease: a review. Eur J
clin Nutr, 56(11): 1049-71.
64. Kaput J, Swartz D, paisley E, Mangian H,
Daniel WL, Visek WJ (1994). Diet- disease
interactions at the molecular level: an ex-
perimental paradigm. J Nutr, 124: (8 Suppl):
1296 S-305 S.
65. Park EI, paisley EA, Mangian HJ, Swartz
DA, Wu MX, O'Morchoe PJ, Behr SR,
Visek WJ, Kaput J (1997). Lipid level and
type alter stearoyl coA desaturase mRNA

14

Das könnte Ihnen auch gefallen