SlideShare a Scribd company logo
‫תזונתיות‬ ‫וגנטיקה‬ ‫גנומיקה‬ ‫של‬ ‫הרצף‬:
‫התקדמות‬,‫רלוונטיות‬,‫ויישום‬
‫שרון‬ ‫אסנת‬,‫שני‬ ‫תואר‬,R.D.
Continuum of Nutritional Genomics and
Genetics: Progress, Relevance, Application
Ossie (Osnat) Sharon, M.Sc., R.D.
‫תקציר‬
‫של‬ ‫העתיד‬ ‫נחשבת‬ ‫היא‬ ‫אישית‬ ‫מותאמת‬ ‫תזונה‬
‫התזונה‬ ‫תורת‬,‫ידי‬ ‫על‬ ‫שנוצר‬ ‫צורך‬ ‫על‬ ‫שמענה‬
‫בין‬ ‫וסתירות‬ ‫להתערבויות‬ ‫עקביות‬ ‫לא‬ ‫תגובות‬
‫המלצות‬.‫אישית‬ ‫מותאמת‬ ‫לרפואה‬ ‫במקביל‬,
‫ונוטריגנומיקה‬ ‫נוטריגנטיקה‬ ‫כולל‬ ‫זה‬ ‫תחום‬–‫איך‬
‫האורגניזם‬ ‫ועל‬ ‫לזו‬ ‫זו‬ ‫על‬ ‫משפיעות‬ ‫ותזונת‬ ‫גנטיקה‬
‫כולו‬–‫ואפי‬‫ואפיגנומיקה‬ ‫גנטיקה‬–‫על‬ ‫ההשפעה‬
‫הבאים‬ ‫הדורות‬.‫גנים‬ ‫זוהו‬ ‫מחקרים‬,‫פולימורפיזם‬,
‫וסביבתיים‬ ‫הללו‬ ‫הגורמים‬ ‫בין‬ ‫גומלין‬ ‫ויחסי‬
‫חומרים‬ ‫חילוף‬ ‫את‬ ‫המניעים‬,‫שמושפעת‬ ‫למידה‬
‫פנוטיפית‬ ‫גמישות‬ ‫ידי‬ ‫על‬.‫הזוהו‬ ‫כה‬ ‫שעד‬ ‫אלה‬
‫ופעילות‬ ‫לתזונה‬ ‫הקשר‬ ‫מבחינת‬ ‫אמינים‬ ‫כהכי‬
‫לאסטרטג‬ ‫רתומים‬ ‫גופנית‬‫התערבותיות‬ ‫יות‬,‫ועוד‬
‫להתברר‬ ‫ממשיכים‬ ‫אחרים‬.
‫מונע‬ ‫יוקר‬ ‫רקע‬ ‫על‬,‫בדיקות‬
‫נוטריגנטקיה‬/‫נתן‬ ‫ויותר‬ ‫יותר‬ ‫הן‬ ‫נוטריגנומיקה‬
‫לצרכן‬ ‫להשיג‬,‫העולם‬ ‫ברחבי‬ ‫ופופולריות‬.‫אתגר‬
‫תוצאות‬ ‫קבלת‬ ‫לאחר‬ ‫הכוונה‬ ‫חוסר‬ ‫היה‬ ‫נוסף‬
‫הצרכן‬ ‫ידי‬ ‫על‬ ‫הבדיקות‬,‫בסיכונים‬ ‫שכרוכים‬.
‫לפנו‬ ‫מקודדים‬ ‫שגנים‬ ‫העובדה‬‫מרובים‬ ‫טיפים‬–
‫הרחב‬ ‫לקהל‬ ‫ידועים‬ ‫שפחות‬ ‫אלה‬ ‫כולל‬–‫מציג‬
‫רב‬ ‫קליני‬ ‫פרופיל‬-‫דורש‬ ‫קרובות‬ ‫שלעתים‬ ‫גורמי‬
‫לכאורה‬ ‫סותרות‬ ‫ואפילו‬ ‫מורכבות‬ ‫התערבויות‬,‫ולכן‬
‫לספק‬ ‫יכול‬ ‫מקצוען‬ ‫שרק‬ ‫מאוזנת‬ ‫אסטרטגיה‬.‫זה‬
‫האמריקאי‬ ‫הבריאות‬ ‫משרד‬ ‫את‬ ‫הניע‬,‫גם‬ ‫כמו‬
‫אמריקאיות‬ ‫שאינן‬ ‫חברות‬ ‫כמה‬,‫בדיק‬ ‫לדרוש‬‫ה‬
‫בלבד‬ ‫מומחים‬ ‫ידי‬ ‫על‬ ‫מכוונים‬ ‫וייעוץ‬–‫של‬ ‫במקרה‬
‫לתזונה‬ ‫הקשורות‬ ‫אינדיקציות‬,‫דיאטנים‬ ‫ידי‬ ‫על‬/‫ות‬.
‫מכך‬ ‫כתוצאה‬,‫דיאטנים‬/‫בחוד‬ ‫שומו‬ ‫נמצאים‬ ‫ות‬
‫ידי‬ ‫על‬ ‫אישית‬ ‫מותאמת‬ ‫תזונה‬ ‫של‬ ‫החנית‬
‫נלווים‬ ‫מקצועות‬,‫גם‬ ‫עבור‬ ‫חשובות‬ ‫השלכות‬ ‫עם‬
‫הצרכן‬ ‫בריאות‬ ‫וגם‬ ‫מקצועי‬ ‫קידום‬.‫להמשך‬ ‫בנוסף‬
‫ב‬ ‫של‬‫בישראל‬ ‫נוטריגנומיקה‬ ‫מחקרי‬ ‫יצוע‬,‫בדיקות‬
‫לדיאטנים‬ ‫המיועדות‬ ‫אישיות‬/‫לכאן‬ ‫הגיעו‬ ‫ות‬
‫לאחרונה‬,‫להזדמנויות‬ ‫דלתות‬ ‫פתיחת‬ ‫לטובת‬
‫דומות‬.
Abstract
Personalized nutrition is considered to be the
future of dietetics, answering a need created
by inconsistent responses to interventions and
resultant contradictions within/between
recommendations. Parallel to personalized
medicine, it comprises nutrigenetics and
nutrigenomics – how genetics and nutrition
impact one another and the organism as a
whole – and epigenetics and epigenomics –
how this impact affects future generations.
Research has identified genes,
polymorphisms, and interactions between
these and environmental factors that drive
metabolism, to an extent influenced by
phenotypic plasticity. Those most reliably
correlating to nutrition and physical activity
are currently being harnessed for
interventional strategies, while others
continue to be clarified.
Once prohibitively expensive, consumer
nutrigenetic/nutrigenomic testing is becoming
increasingly affordable and popular globally.
An additional challenge has been inadequate
direction once results are received by the
consumer, with associated risks. The fact that
genes code for multiple phenotypes –
including those less publicized – presents a
multifactorial clinical profile that often
requires composite and even seemingly
contradictory interventions, and therefore a
balanced strategy that only a professional can
provide. This has prompted the United States
Food and Drug Administration, as well as
some non-American companies, to require
expert-directed testing and counseling – in the
case of nutrition-related indications, by
dietitians.
As a result, dietitians are being put on the
forefront of personalized nutrition by
ancillary professions, with important
implications for both professional
advancement and consumer health. In
addition to Israel continuing to host
nutrigenomics research, personal testing
technologies have recently become available
to practitioners here, opening the doors to
similar opportunities.
Summary
Nutrigenomics is a blanket term for a new scientific discipline that uses modern genomics
technology to study the relationship between genes, nutrition and health. It has long been
apparent that some people respond differently from others to certain foods. Nutrigenomics
allows us to understand how our genes affect the way we respond to the foods, beverages and
supplements we consume [1-3]. It is the position of the Academy of Nutrition and Dietetics that
“nutritional genomics provides insight into how diet and genotype interactions affect
phenotype.”
Key Definitions
Nutritional genomics The relationship between the human genome, nutrition, and health
Nutrigenetics How the genotype drives the phenotypic response to dietary intake
• Monogenic Result of alteration to one particular gene
• Polygenic: Result of alterations to more than one gene
Nutrigenomics How dietary intake drives genotypic activity
Phenotypic Plasticity How the phenotype adapts to environmental changes, including diet
SNP Single/simple nucleotide polymorphism: common variation of a
single nucleotide (A, T, C, G) in a common DNA sequence
Metabolomics The study of chemical processes involving metabolites
Nutritional epigenetics How phenotypic response to diet drives the genotype over generations
Nutritional epigenomics How nutritional epigenetic modifications affect the entire genome
DoHAD Developmental origins of health and disease
• Impact of experiences during initial phases of somatic
development on lifelong health, including chronic disease risk
• Models proposed: thrifty phenotype, programming, predictive
adaptive response theories, concept of match or mismatch
• Possible mechanisms: environmental effects on gene
expression, hormonal signals transmitted to fetus/infant via
placenta/lactation
Key Tenets
• Common dietary chemicals act on the human genome, either directly or indirectly, to
alter gene expression or structure.
• Under certain circumstances and in some individuals, diet can be a serious risk factor for
a number of diseases.
• Some diet-regulated genes (and their common variants, i.e. “SNP”s) are likely to play a
role in the onset, incidence, progression, and/or severity of chronic diseases of
individuals and their descendants.
• The degree to which diet influences the balance between healthy and disease states may
depend on an individual’s genetic makeup.
• Dietary intervention based on knowledge of nutritional requirement, nutritional status,
and genotype (i.e., “individualized nutrition”) can be used to prevent, mitigate or cure
chronic disease.
Introduction
Since completion of the human genome project, understanding of complex interactions between
environmental factors such as diet and genes has progressed considerably. The knowledge that
metabolic pathways may be altered in individuals with genetic variants in the presence of certain
dietary exposures offers great potential for personalized nutrition advice, and epigenetics and
nutrigenetics have been used to assess the need and status of specific nutrients. This has
dramatically increased the potential to individualize diets using dietary, phenotypic and
genotypic data, with improved outcomes [1-6].
Recent genetic interest and research focus surrounds the direct connections between metabolism
and developmental dynamics, which now represents an important conceptual challenge to
explain many aspects of metabolic dysfunction. Several components of the epigenetic machinery
require intermediates of cellular metabolism for enzymatic function. For example, specific
epigenetic influences of dietary glucose and lipid consumption, as well as undernutrition, are
observed across numerous organs and pathways associated with metabolism. Studies have started
to define the chromatin-dependent mechanisms underlying persistent and pathophysiological
changes induced by altered metabolism. Importantly, numerous recent studies demonstrate that
gene regulation underlying phenotypic determinants of adult metabolic health is influenced by
maternal and early postnatal diet. These emerging concepts open new perspectives to combat the
rising global epidemic of metabolic disorders [7-9].
Despite widespread promotion of population-based healthy eating guidelines over the half-
century, preventable diseases remain the leading cause of mortality in the developed world.
Moreover, the greatest numbers of disease events occur in individuals with a lower conventional
risk of disease [10-14].
Inconsistent research outcomes and real-world responses limit utility of recommendations for
health attainment and maintenance, particularly in the areas of weight and chronic disease such
as type 2 diabetes mellitus, cardiovascular disease, and cancer [15, 16]. Examples include the
following:
• Saturated fat risks
• Omega-3 fatty acid needs
• Low-fat vs. low-carbohydrate diet for weight loss
• Increased fiber needs
• Intolerances, e.g. to lactose, gluten
• Sodium restriction risk vs. advantage
• Increased need for specific micronutrients
• Animal vs. plant-based
• Relative risk from high-risk cooking methods, such as charring and frying
• Harmful vs. beneficial effects of diet-related habits, e.g. caffeine, alcohol
• Physical activity needs and injury risk
• Efforts required for healthy weight loss and maintenance
• Impact of the above on perinatal outcomes, including long-term in offspring
Personalized nutrition interventions may have greater potential for reducing the global burden of
preventable diseases and for promoting better health across the lifespan than the conventional
“one size fits all” approach [4-6, 10]. Further, such measures during the prenatal period –
including prior to conception – have implications not only for improved maternal outcomes and
future health, but also for the long-term health of the offspring and future generations [17-22].
Genetic testing is increasingly being embraced for the purpose of personalized nutrition
planning. Once available only to physicians through medical testing, provision of genetic
polymorphism (SNP) information is now widely available direct-to-consumer.
Method
Background
Over the past decade, several companies have formed partnerships with laboratories to provide
an increasing variety of genetic and immunologic tests that indicate predispositions for
physiologic imbalances that, left unchecked, could ultimately lead to serious disorders [23].
Most of these tests are provided by having the consumer send a saliva sample to a central
laboratory, which then performs the analysis for a predetermined group of genes,
polymorphisms, and alleles. The results are then provided either to the consumer or to a
healthcare practitioner.
Practitioners benefit because they have a new set of tools with potential to improve their ability
to prevent disease and help restore health, and patients benefit from new opportunities to avert
disorders before they cause significant disruptions, doing away with the hit-or-miss, trial-and-
error that characterizes the pursuit of the ideal health-oriented lifestyle [20].
However, challenges and questions remain regarding use and acceptance of nutrigenomic testing,
including the following:
• Accuracy of tests provided
Research into the many polymorphisms involved in nutrigenomics has yielded many
promising leads, but only some have been consistently linked to health conditions in a
manner that can benefit from specific interventions.
• Professional acceptance
It is not clear to some health care providers how nutrigenetic testing and related
interventions will yield benefits over current standards of care. However, nutrigenetic
screening has been associated with greater improvements towards and maintenance of
health goals when compared to conventional intervention [24-26].
• Government acceptance
Government bodies are concerned that consumers must be protected from unrealistic
claims and misinterpretations of complex genomic information, and that direct-to-
consumer test results are often not adequately understood by patients and thus raise
concerns when used to self-manage [27].
• Fraudulent practices of service providers
Impartial evaluations have found that many internet-based direct-to-consumer genetic
tests were fraudulent in some way [28].
• Quality of associated care
Many companies are linked to nutritional supplement companies, encouraging pills over
food, where research suggests a food-based approach to be superior [29-31].
• Public acceptance
Many consumers remain unaware of the potentially controllable nature of genetic
influence on health. When informed, research has identified health and clear consumer
benefits as key motivators in the uptake of genetic testing, with individuals
reporting personal experience of disease/symptoms being more willing to
undergo genetic testing for the purpose of personalized nutrition [20], particularly when
information is delivered by a qualified healthcare professional such as a dietitian [32]. A
perceived susceptibility to disease has also been suggested to improve motivation to
change behavior, which is a key barrier in the success of any nutrition intervention [19].
• Compliance with recommendations
Where widespread lack of compliance with conventional nutrition recommendations has
cast doubt on the efficacy of methods, the nutrigenetic approach has resulted in increased
compliance with lifestyle interventions [33].
Sources of Testing
Some companies that offer nutrigenetic testing products are designing programs that bypass
principal concerns of multiple authorities.
• Testing only of SNPs that have consistently been linked to health conditions in a manner
that can benefit from specific interventions.
• Provision of nutrigenetic testing through qualified professionals, specifically dietitians,
packaged with nutritional counseling.
Genes, SNPs, and Implications
The following are examples of SNPs meeting the above criteria, pointing to a strong indication
for professional guidance by a dietitian/nutritionist:
• MTHFR 677 CT/TT
Methylenetetrahydrofolate reductase (MTHFR) catalyzes the metabolism of folate and
nucleotides needed for DNA synthesis and repair, and as such gene plays an important
role in the genomic integrity and genetic stability. The most common SNP, MTHFR
C677T is known primarily for increasing the need for dietary folate and most frequently
linked to homocysteinemia and its impact on cardiovascular risk [34].
MTHFR SNPs have also been found in research to be associated with cellular functions
related to various cancer linked to key nutritional factors beyond folate (including breast,
gastric, colon, thyroid) [35-40], ovarian [41, 42] and sperm integrity [43], gestational
outcomes beyond neural tube defects [44, 45], thyroid function [46-48], and neurological
disorders, including Parkinson’s [49] and autism [50, 51], and metabolic risks related to
premature menopause, bone health [52], and development of type 2 diabetes, as well as
increased need for riboflavin, pyridoxine, and other nutritional interventions related to the
above issues [53].
While over-the-counter folic acid supplements have been recommended as a solution,
there are concerns that standard options are not adequate to meet the needs of SNP
carriers; research has even suggested that common supplements may actually increase
risk of methylation-related disorders [54-56]. In carriers of the polymorphism, either a
special (more expensive) supplemental form of folate and/or food-based intervention is
preferred – particularly relevant when considering the additional advantages of foods
high in folate, which tend also to be naturally high in fiber. When frequently concurrent
conditions of hypothyroid and coagulation disorders occur, this demands balancing intake
of the above foods with medications, i.e. thyroxine and warfarin, as well as other B-
vitamins with which folate performs its activities, also warranting intervention by a
qualified dietitian/nutritionist.
• GSTT1 and GSMT1
The GSTT1 and GSMT1 genes produce proteins for the phase II metabolizing
glutathione S-transferase enzyme family, responsible detoxifying numerous potentially
cytotoxic/genotoxic compounds and attenuating inflammatory cytokines such as
interleukin-6 (IL6). GSTT1 is also known for its key role in the utilization of vitamin C,
and a dysfunctional version of the gene results in a reduced ability to process vitamin C
and need to ensure the minimum recommended intake (Dietary Reference Intake, DRI) to
maintain normal levels, where the functional version enables individuals to thrive with
less [18].
In clinical research, IL-6 was inversely associated with total isothiocyanate excretion in
GSTM1-null/GSTT1-null individuals. Inadequate upregulation of these protective
enzymes has been linked to cancer, suggesting a need not only to decrease intake of
offending compounds, but also to increase intake of foods rich in indole antioxidants,,
such as those in cruciferous vegetables [57].
When occurring in combination with the above-noted MTHFR polymorphism, there may
be increased risk of both acute and chronic myelogenous leukemias and autism spectrum
disorders (ASD) [51, 58-60]. Given the implications not only for individuals but also
their offspring, there is a clear advantage to the increased compliance with a preventive
diet that can be attained with personalized dietetic counseling.
• TCF7L2
Studies have consistently shown that common variants in the TCF7L2 gene significantly
predict type 2 diabetes risk [61-63], due to a deleterious effect related to β-cell function
[64-66] and downregulation of gastric inhibitory polypeptide (GIP) and glucagon-like
peptide-1 (GLP-1) receptors [65]. This has been demonstrated to be relevant to
gestational diabetes as well [67-70].
The TCF7L2 has been closely linked to the role of fiber in maintenance of normal
glucose metabolism [71-73], including prevention of progression to the prediabetic state
in high-risk individuals [74].
In addition, compelling evidence has suggested important roles of TCF7L2 in the
regulation of body weight and adiposity including 1) taking part in the Wnt signaling
cascade, which inhibits adipogenesis and stimulates leptin production in mature
adipocytes in response to nutritional status [75], 2) gene expression in adipose tissue
differing by genotype after caloric restriction, 3) the association with hunger-satiety
hormones that influence weight loss, and 4) the promotion of the transcription of
proglucagon, which induces the synthesis of glucagon-like peptide, which is a regulator
of insulin and glucagon secretion, appetite, and food intake [76-78].
Despite this evidence, reports on the association of TCF7L2 variants with body weight
and composition as well as diabetes risk have been contradictory. One possible
explanation for the discrepancies might be interactions between TCF7L2 genotypes and
environmental factors such as dietary intake [67, 73, 79]. Such interactions have been
reported in observational studies for dietary fat, carbohydrate intake, and animal protein,
and are largely dependent on specific SNPs. For example, a recent randomized trial
reported that obese individuals with the rs7903146 or rs12255372 risk allele have better
weight/adiposity loss responses in weight loss with a low- rather than high-fat dietary
intervention [79] and additional genes have been observed to interact with prenatal
nutrition in relation to T2D risk and glucose levels in later life [73].
Given that a possible predisposition to type 2 diabetes generally indicates a diet low in
carbohydrates and attainment and maintenance of a healthy weight, and that efforts to
reduce fat intake have historically resulted in high carbohydrate intake, guidance by a
dietitian is indicated to assist in constructing a balanced diet that can be sustained long-
term.
• NOS3
The NOS3 gene upregulates nitric oxide synthase and production of nitric oxide, which
plays an important role in the function of the vascular epithelium. New research has
shown that variations in the NOS3 gene interact with omega-3 fatty acids in different
ways to impact how the body processes triglycerides, and the degree to which elevated
triglycerides impair circulation. Previous studies have produced mixed results relating to
the effects of omega-3 fat on triglyceride levels between individuals. Some people
experience a significant reduction in triglyceride levels in response to increasing omega-3
fat intake, whereas others experience little benefit. The reasons for these differences have
been unclear until a recent breakthrough study* showed that the effect of omega-3 fat on
triglyceride levels depends on variations in a gene called NOS3. Those who have the GT
or TT variant of the gene are at greater risk of elevated triglyceride levels when
consuming a diet low in omega-3 fats, compared to those who have the GG variant.
Beyond this function, NOS3 SNPs such as rs1799983 have been and continue to be
investigated for influence on cancers, obesity, and perinatal disorders such as recurrent
miscarriage and pre-eclampsia. Pre-eclampsia is a well-known example of a potentially
preventable/manageable condition that in addition to omega-3 intake [80], nutrition-
related prevention and management of pre-eclampsia is multi-factorial [81, 82], and is an
accepted indication for dietitian involvement. Pre-eclampsia in turn increases
• APOA2
Saturated fats have been associated with health conditions such as diabetes,
cardiovascular disease, and obesity. However, the connection between saturated fats and
obesity, until recently, has been inconsistent. A number of studies have now shown that
the effect of saturated fat on obesity can be influenced by variations in APOA2 [83-87].
The APOA2 gene upregulates production of apolipoprotein A-II, which impacts dietary
fat utilization. Scientists now understand that there are different variations in the APOA2
gene present in the human population and that these different versions of the gene interact
with saturated fat in unique ways to influence energy balance and ultimately the risk of
obesity [83, 86]. Those people who have the CC variant of the gene are at a higher risk of
developing obesity when consuming a diet high in saturated fats than those possessing
the TT or TC variant of the gene [85].
Additionally, the CC polymorphism has been associated with elevated levels of visceral
adipose tissue and development of insulin resistance, independent of gender, age and
body mass index [87]. Related concerns point to the influence of APOA2 SNPs on
inflammation and blood lipids [88].
The above issues require guidance regarding a diet that balances low saturated fat with
low simple carbohydrate intake, while protecting against the oxidation risk [84].
• HLA DQ2/8
The human leukocyte antigen (HLA) gene is the most important genetic predictor of
gluten intolerance. Nearly all individuals with celiac disease [89-91] and more than half
of those with non-celiac gluten sensitivity (NCGS) [92, 93] have the DQ2 or DQ8 (“high
risk”) version of the HLA gene, compared to only 25% of the general population.
Celiac disease represents the most severe form of gluten intolerance and affects about 1%
of the population. Another 5-10% of the population have non-celiac gluten sensitivity
NCGS, which results in various gastrointestinal problems, and still others have wheat
sensitivities related to its carbohydrate fractions rather than gluten, which in turn may be
indicative of multiple sensitivities, e.g. to FODMAPs [94, 95], requiring guidance by a
dietitian [96-98].
Recent research has suggested that in predisposed individuals, celiac may represent the
final step in a continuum of severity of gluten intolerance following cumulative exposure
[99, 100]. Testing for the relevant HLA forms may enable preventive dietary measures
and/or redirect focus to other disorders with similar symptoms but differing interventions.
It can also rule-out a need to undergo an invasive biopsy procedure, currently standard of
care to rule out celiac in individuals experiencing characteristic symptom.
Additional genes with SNPs indicating a need for nutritional management and currently being
offered for direct-to-dietitian testing include APOE, ACE, CYP1A2, ADH1C, VDR, IL6, TNF,
SOD2, CAT, GPX1, LCT, and FOT, each with complexities requiring intervention by a qualified
professional.
The above polymorphisms can occur in combination, potentially magnifying risk not only of
characteristic disorders, but also to diseases specifically associated with concurrence of alleles,
again pointing to the need for qualified dietetic guidance for both prevention and management,
for the individual and future generations.
Conclusions
Personalized interventions that take into account genetic variations in response to nutritional
factors may have greater potential for disease prevention and management than a “one size fits
all” approach, and may be better accepted by patients.
The fact that genes code for multiple phenotypic effects presents a unique, multifactorial picture
for individuals that require the type of balancing between lifestyle factors that only a professional
can provide. This is highly likely to improve compliance and quality of results and in a much
shorter timeframe than are associated with conventional interventions.
As a result, dietitians are being put on the forefront of personalized nutrition by ancillary
professions, with important implications for both professional advancement and consumer
health. In addition to Israel continuing to host nutrigenomics research, personal testing
technologies have recently become available to practitioners here, opening the doors to similar
opportunities.
References
1. Isaak CK, Siow YL. The evolution of nutrition research. Can J Physiol Pharmacol 2013;
91 (4):257-267.
2. Mocchegiani E, Costarelli L, Giacconi R, Malavolta M, Basso A, Piacenza F, Ostan R,
Cevenini E, Gonos ES, Monti D. Micronutrient-gene interactions related to
inflammatory/immune response and antioxidant activity in ageing and inflammation. A
systematic review. Mech Ageing Dev 2014; 136-137:29-49.
3. Perez-Martinez P, Phillips CM, Delgado-Lista J, Garcia-Rios A, Lopez-Miranda J, Perez-
Jimenez F. Nutrigenetics, metabolic syndrome risk and personalized nutrition. Curr Vasc
Pharmacol 2013; 11 (6):946-953.
4. Bordoni A, Capozzi F. Foodomics for healthy nutrition. Curr Opin Clin Nutr Metab Care
2014; 17 (5):418-424.
5. Gibney MJ, McNulty BA, Ryan MF, Walsh MC. Nutritional phenotype databases and
integrated nutrition: from molecules to populations. Adv Nutr 2014; 5 (3):352S-357S.
6. Jones ML, Ganopolsky JG, Martoni CJ, Labbe A, Prakash S. Emerging science of the
human microbiome. Gut Microbes 2014; 5 (4):446-457.
7. Desai M, Jellyman JK, Ross MG. Epigenomics, gestational programming and risk of
metabolic syndrome. Int J Obes (Lond) 2015; 39 (4):633-641.
8. Keating ST, El-Osta A. Epigenetics and metabolism. Circ Res 2015; 116 (4):715-736.
9. Tarrade A, Panchenko P, Junien C, Gabory A. Placental contribution to nutritional
programming of health and diseases: epigenetics and sexual dimorphism. J Exp Biol
2015; 218 (Pt 1):50-58.
10. Bauer UE, Briss PA, Goodman RA, Bowman BA. Prevention of chronic disease in the
21st century: elimination of the leading preventable causes of premature death and
disability in the USA. Lancet 2014; 384 (9937):45-52.
11. Dwyer JT, Melanson KJ, Ms US, Cross P, Wilson M. Dietary Treatment of Obesity.
2000.
12. Kwon BJ, Kim DW, Her SH, Kim DB, Jang SW, Cho EJ, Ihm SH, Kim HY, Youn HJ,
Seung KB, Kim JH, Rho TH. Metabolically obese status with normal weight is associated
with both the prevalence and severity of angiographic coronary artery disease.
Metabolism 2013; 62 (7):952-960.
13. Karelis AD, St-Pierre DH, Conus F, Rabasa-Lhoret R, Poehlman ET. Metabolic and body
composition factors in subgroups of obesity: what do we know? J Clin Endocrinol Metab
2004; 89 (6):2569-2575.
14. Nowak M, Grzywa M. Metabolic obesity in normal weight individuals and
cardiovascular disease. Pol Merkur Lekarski 2011; 31 (182):118-121.
15. Alexander DD, Weed DL, Miller PE, Mohamed MA. Red Meat and Colorectal Cancer: A
Quantitative Update on the State of the Epidemiologic Science. J Am Coll Nutr 2010:1-
23.
16. Jenkins DJ, Josse AR, Dorian P, Burr ML, LaBelle Trangmar R, Kendall CW, Cunnane
SC. Heterogeneity in randomized controlled trials of long chain (fish) omega-3 fatty
acids in restenosis, secondary prevention and ventricular arrhythmias. J Am Coll Nutr
2008; 27 (3):367-378.
17. Bouchard C, Ordovas JM. Fundamentals of nutrigenetics and nutrigenomics. Prog Mol
Biol Transl Sci 2012; 108:1-15.
18. Cahill LE, Fontaine-Bisson B, El-Sohemy A. Functional genetic variants of glutathione
S-transferase protect against serum ascorbic acid deficiency. Am J Clin Nutr 2009; 90
(5):1411-1417.
19. Nielsen DE, El-Sohemy A. A randomized trial of genetic information for personalized
nutrition. Genes Nutr 2012; 7 (4):559-566.
20. Ronteltap A, van Trijp JC, Renes RJ. Consumer acceptance of nutrigenomics-based
personalised nutrition. Br J Nutr 2009; 101 (1):132-144.
21. Subbiah MT. Nutrigenetics and nutraceuticals: the next wave riding on personalized
medicine. Transl Res 2007; 149 (2):55-61.
22. Millis RM. Epigenetics and hypertension. Curr Hypertens Rep 2011; 13 (1):21-28.
23. Preuss C, Das MK, Pathak YV. Genomics and natural products: role of bioinformatics
and recent patents. Recent Pat Biotechnol; 8 (2):144-151.
24. Rizkalla SW, Prifti E, Cotillard A, Pelloux V, Rouault C, Allouche R, Laromiguiere M,
Kong L, Darakhshan F, Massiera F, Clement K. Differential effects of macronutrient
content in 2 energy-restricted diets on cardiovascular risk factors and adipose tissue cell
size in moderately obese individuals: a randomized controlled trial. Am J Clin Nutr 2012;
95 (1):49-63.
25. Roy N, Barnett M, Knoch B, Dommels Y, McNabb W. Nutrigenomics applied to an
animal model of Inflammatory Bowel Diseases: transcriptomic analysis of the effects of
eicosapentaenoic acid- and arachidonic acid-enriched diets. Mutat Res 2007; 622 (1-
2):103-116.
26. Sanchez-Muniz FJ, Canales A, Nus M, Bastida S, Guillen M, Corella D, Olmedilla-
Alonso B, Granado-Lorencio F, Benedi J. The antioxidant status response to low-fat and
walnut paste-enriched meat differs in volunteers at high cardiovascular Risk carrying
different PON-1 polymorphisms. J Am Coll Nutr 2012; 31 (3):194-205.
27. Food and Drug Administration (FDA). Warning Letter: 23andMe, Inc. 11/22/13. 2013.
28. Saukko P. State of play in direct-to-consumer genetic testing for lifestyle-related
diseases: market, marketing content, user experiences and regulation. Proc Nutr Soc
2013; 72 (1):53-60.
29. Fortmann SP, Burda BU, Senger CA, Lin JS, Whitlock EP. Vitamin and mineral
supplements in the primary prevention of cardiovascular disease and cancer: An updated
systematic evidence review for the U.S. Preventive Services Task Force. Ann Intern Med
2013; 159 (12):824-834.
30. Jacobs DR, Jr., Gross MD, Tapsell LC. Food synergy: an operational concept for
understanding nutrition. Am J Clin Nutr 2009; 89 (5):1543S-1548S.
31. Ward E. Addressing nutritional gaps with multivitamin and mineral supplements. Nutr J
2014; 13:72.
32. Nielsen DE, Shih S, El-Sohemy A. Perceptions of genetic testing for personalized
nutrition: a randomized trial of DNA-based dietary advice. J Nutrigenet Nutrigenomics
2014; 7 (2):94-104.
33. Nielsen DE, El-Sohemy A. Disclosure of genetic information and change in dietary
intake: a randomized controlled trial. PLoS One 2014; 9 (11):e112665.
34. Trimmer EE. Methylenetetrahydrofolate reductase: biochemical characterization and
medical significance. Curr Pharm Des 2013; 19 (14):2574-2593.
35. Slaoui M, Razine R, Ibrahimi A, Attaleb M, Mzibri ME, Amrani M. Breast cancer in
Morocco: a literature review. Asian Pac J Cancer Prev 2014; 15 (3):1067-1074.
36. Xia LZ, Liu Y, Xu XZ, Jiang PC, Ma G, Bu XF, Zhang YJ, Yu F, Xu KS, Li H.
Methylenetetrahydrofolate reductase C677T and A1298C polymorphisms and gastric
cancer susceptibility. World J Gastroenterol 2014; 20 (32):11429-11438.
37. Nassiri M, Kooshyar MM, Roudbar Z, Mahdavi M, Doosti M. Genes and SNPs
associated with non-hereditary and hereditary colorectal cancer. Asian Pac J Cancer Prev
2013; 14 (10):5609-5614.
38. Chen Y, Wang B, Yan S, Wang YG. Significant association between MTHFR C677T
polymorphism and thyroid cancer risk: evidence from a meta-analysis. Genet Test Mol
Biomarkers 2014; 18 (10):695-702.
39. Yan Y, Han F, Fu H, Xia W, Qin X. Association between MTHFR C677T polymorphism
and thyroid cancer risk: a meta-analysis. Tumour Biol 2014; 35 (8):7707-7712.
40. Yang YM, Zhang TT, Yuan L, Ren Y. The association between the C677T
polymorphism in MTHFR gene and the risk of thyroid cancer: a meta-analysis. Eur Rev
Med Pharmacol Sci 2014; 18 (15):2097-2101.
41. Casarini L, Simoni M. Gene polymorphisms in female reproduction. Methods Mol Biol
2014; 1154:75-90.
42. Rah H, Jeon YJ, Choi Y, Shim SH, Yoon TK, Choi DH, Cha SH, Kim NK. Association
of methylenetetrahydrofolate reductase (MTHFR 677C>T) and thymidylate synthase
(TSER and TS 1494del6) polymorphisms with premature ovarian failure in Korean
women. Menopause 2012; 19 (11):1260-1266.
43. Singh K, Jaiswal D. One-carbon metabolism, spermatogenesis, and male infertility.
Reprod Sci 2012; 20 (6):622-630.
44. Lewis SJ. Commentary: One-carbon metabolism has major implications for fetal growth
and development beyond neural tube defects. Int J Epidemiol 2014; 43 (5):1498-1499.
45. Yajnik CS, Chandak GR, Joglekar C, Katre P, Bhat DS, Singh SN, Janipalli CS, Refsum
H, Krishnaveni G, Veena S, Osmond C, Fall CH. Maternal homocysteine in pregnancy
and offspring birthweight: epidemiological associations and Mendelian randomization
analysis. Int J Epidemiol 2014; 43 (5):1487-1497.
46. Arakawa Y, Watanabe M, Inoue N, Sarumaru M, Hidaka Y, Iwatani Y. Association of
polymorphisms in DNMT1, DNMT3A, DNMT3B, MTHFR and MTRR genes with
global DNA methylation levels and prognosis of autoimmune thyroid disease. Clin Exp
Immunol 2012; 170 (2):194-201.
47. Kakkoura MG, Demetriou CA, Loizidou MA, Loucaides G, Neophytou I, Marcou Y,
Hadjisavvas A, Kyriacou K. Single-nucleotide polymorphisms in one-carbon metabolism
genes, Mediterranean diet and breast cancer risk: a case-control study in the Greek-
Cypriot female population. Genes Nutr 2015; 10 (2):453.
48. Lambrinoudaki I, Kaparos G, Rizos D, Galapi F, Alexandrou A, Sergentanis TN, Creatsa
M, Christodoulakos G, Kouskouni E, Botsis D. Apolipoprotein E and paraoxonase 1
polymorphisms are associated with lower serum thyroid hormones in postmenopausal
women. Clin Endocrinol (Oxf) 2009; 71 (2):284-290.
49. Wu YL, Ding XX, Sun YH, Yang HY, Sun L. Methylenetetrahydrofolate reductase
(MTHFR) C677T/A1298C polymorphisms and susceptibility to Parkinson's disease: a
meta-analysis. J Neurol Sci 2013; 335 (1-2):14-21.
50. Frustaci A, Neri M, Cesario A, Adams JB, Domenici E, Dalla Bernardina B, Bonassi S.
Oxidative stress-related biomarkers in autism: systematic review and meta-analyses. Free
Radic Biol Med 2012; 52 (10):2128-2141.
51. James SJ, Melnyk S, Jernigan S, Cleves MA, Halsted CH, Wong DH, Cutler P, Bock K,
Boris M, Bradstreet JJ, Baker SM, Gaylor DW. Metabolic endophenotype and related
genotypes are associated with oxidative stress in children with autism. Am J Med Genet
B Neuropsychiatr Genet 2006; 141B (8):947-956.
52. Clarke M, Ward M, Strain JJ, Hoey L, Dickey W, McNulty H. B-vitamins and bone in
health and disease: the current evidence. Proc Nutr Soc 2014; 73 (2):330-339.
53. Reilly R, McNulty H, Pentieva K, Strain JJ, Ward M. MTHFR 677TT genotype and
disease risk: is there a modulating role for B-vitamins? Proc Nutr Soc 2014; 73 (1):47-56.
54. Christensen KE, Mikael LG, Leung KY, Levesque N, Deng L, Wu Q, Malysheva OV,
Best A, Caudill MA, Greene ND, Rozen R. High folic acid consumption leads to pseudo-
MTHFR deficiency, altered lipid metabolism, and liver injury in mice. Am J Clin Nutr
2015; 101 (3):646-658.
55. Scaglione F, Panzavolta G. Folate, folic acid and 5-methyltetrahydrofolate are not the
same thing. Xenobiotica 2014; 44 (5):480-488.
56. Shorter KR, Felder MR, Vrana PB. Consequences of dietary methyl donor supplements:
Is more always better? Prog Biophys Mol Biol 2015.
57. Navarro SL, Schwarz Y, Song X, Wang CY, Chen C, Trudo SP, Kristal AR, Kratz M,
Eaton DL, Lampe JW. Cruciferous vegetables have variable effects on biomarkers of
systemic inflammation in a randomized controlled trial in healthy young adults. J Nutr
2014; 144 (11):1850-1857.
58. Cummings AM, Kavlock RJ. Gene-environment interactions: a review of effects on
reproduction and development. Crit Rev Toxicol 2004; 34 (6):461-485.
59. Ding H, Liu W, Yu X, Wang L, Shao L, Yi W. Risk association of meningiomas with
MTHFR C677T and GSTs polymorphisms: a meta-analysis. Int J Clin Exp Med 2014; 7
(11):3904-3914.
60. Lordelo GS, Miranda-Vilela AL, Akimoto AK, Alves PC, Hiragi CO, Nonino A,
Daldegan MB, Klautau-Guimaraes MN, Grisolia CK. Association between methylene
tetrahydrofolate reductase and glutathione S-transferase M1 gene polymorphisms and
chronic myeloid leukemia in a Brazilian population. Genet Mol Res 2012; 11 (2):1013-
1026.
61. Grant SF, Thorleifsson G, Reynisdottir I, Benediktsson R, Manolescu A, Sainz J,
Helgason A, Stefansson H, Emilsson V, Helgadottir A, Styrkarsdottir U, Magnusson KP,
Walters GB, Palsdottir E, Jonsdottir T, Gudmundsdottir T, Gylfason A, Saemundsdottir J,
Wilensky RL, Reilly MP, Rader DJ, Bagger Y, Christiansen C, Gudnason V, Sigurdsson
G, Thorsteinsdottir U, Gulcher JR, Kong A, Stefansson K. Variant of transcription factor
7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 2006; 38 (3):320-323.
62. Meigs JB, Shrader P, Sullivan LM, McAteer JB, Fox CS, Dupuis J, Manning AK, Florez
JC, Wilson PW, D'Agostino RB, Sr., Cupples LA. Genotype score in addition to common
risk factors for prediction of type 2 diabetes. N Engl J Med 2008; 359 (21):2208-2219.
63. Tong Y, Lin Y, Zhang Y, Yang J, Liu H, Zhang B. Association between TCF7L2 gene
polymorphisms and susceptibility to type 2 diabetes mellitus: a large Human Genome
Epidemiology (HuGE) review and meta-analysis. BMC Med Genet 2009; 10:15.
64. Migliorini A, Lickert H. Beyond association: A functional role for Tcf7l2 in beta-cell
development. Mol Metab 2015; 4 (5):365-366.
65. Shu L, Matveyenko AV, Kerr-Conte J, Cho JH, McIntosh CH, Maedler K. Decreased
TCF7L2 protein levels in type 2 diabetes mellitus correlate with downregulation of GIP-
and GLP-1 receptors and impaired beta-cell function. Hum Mol Genet 2015; 24
(10):3004.
66. Yao DD, Yang L, Wang Y, Liu C, Wei YJ, Jia XB, Yin W, Shu L. Geniposide promotes
beta-cell regeneration and survival through regulating beta-catenin/TCF7L2 pathway.
Cell Death Dis 2015; 6:e1746.
67. Pagan A, Sabater-Molina M, Olza J, Prieto-Sanchez MT, Blanco-Carnero JE, Parrilla JJ,
Gil A, Larque E. A gene variant in the transcription factor 7-like 2 (TCF7L2) is
associated with an increased risk of gestational diabetes mellitus. Eur J Obstet Gynecol
Reprod Biol 2014; 180:77-82.
68. Reyes-Lopez R, Perez-Luque E, Malacara JM. Metabolic, hormonal characteristics and
genetic variants of TCF7L2 associated with development of gestational diabetes mellitus
in Mexican women. Diabetes Metab Res Rev 2014; 30 (8):701-706.
69. Shi X, Cai Q, Zou M, Shen Y. [Correlation between TCF7L2 gene polymorphism and
genetic susceptibility in women with gestational diabetes mellitus]. Zhonghua Fu Chan
Ke Za Zhi 2014; 49 (8):588-593.
70. Zhang C, Bao W, Rong Y, Yang H, Bowers K, Yeung E, Kiely M. Genetic variants and
the risk of gestational diabetes mellitus: a systematic review. Hum Reprod Update 2013;
19 (4):376-390.
71. Heni M, Herzberg-Schafer S, Machicao F, Haring HU, Fritsche A. Dietary fiber intake
modulates the association between variants in TCF7L2 and weight loss during a lifestyle
intervention. Diabetes Care 2012; 35 (3):e24.
72. Hindy G, Sonestedt E, Ericson U, Jing XJ, Zhou Y, Hansson O, Renstrom E, Wirfalt E,
Orho-Melander M. Role of TCF7L2 risk variant and dietary fibre intake on incident type
2 diabetes. Diabetologia 2012; 55 (10):2646-2654.
73. Qi L, Liang J. Interactions between genetic factors that predict diabetes and dietary
factors that ultimately impact on risk of diabetes. Curr Opin Lipidol 2010; 21 (1):31-37.
74. Wirstrom T, Hilding A, Gu HF, Ostenson CG, Bjorklund A. Consumption of whole grain
reduces risk of deteriorating glucose tolerance, including progression to prediabetes. Am
J Clin Nutr 2012; 97 (1):179-187.
75. Chen ZL, Shao WJ, Xu F, Liu L, Lin BS, Wei XH, Song ZL, Lu HG, Fantus IG, Weng
JP, Jin TR. Acute Wnt pathway activation positively regulates leptin gene expression in
mature adipocytes. Cell Signal 2015; 27 (3):587-597.
76. Helgason A, Palsson S, Thorleifsson G, Grant SF, Emilsson V, Gunnarsdottir S,
Adeyemo A, Chen Y, Chen G, Reynisdottir I, Benediktsson R, Hinney A, Hansen T,
Andersen G, Borch-Johnsen K, Jorgensen T, Schafer H, Faruque M, Doumatey A, Zhou
J, Wilensky RL, Reilly MP, Rader DJ, Bagger Y, Christiansen C, Sigurdsson G,
Hebebrand J, Pedersen O, Thorsteinsdottir U, Gulcher JR, Kong A, Rotimi C, Stefansson
K. Refining the impact of TCF7L2 gene variants on type 2 diabetes and adaptive
evolution. Nat Genet 2007; 39 (2):218-225.
77. Lee YS, Jun HS. Anti-diabetic actions of glucagon-like peptide-1 on pancreatic beta-
cells. Metabolism 2014; 63 (1):9-19.
78. Xiong X, Shao W, Jin T. New insight into the mechanisms underlying the function of the
incretin hormone glucagon-like peptide-1 in pancreatic beta-cells: the involvement of the
Wnt signaling pathway effector beta-catenin. Islets 2012; 4 (6):359-365.
79. Mattei J, Qi Q, Hu FB, Sacks FM, Qi L. TCF7L2 genetic variants modulate the effect of
dietary fat intake on changes in body composition during a weight-loss intervention. Am
J Clin Nutr 2012; 96 (5):1129-1136.
80. Fereidooni B, Jenabi E. The use of omega 3 on pregnancy outcomes: a single-center
study. J Pak Med Assoc 2014; 64 (12):1363-1365.
81. Dodd JM, O'Brien C, Grivell RM. Preventing pre-eclampsia - are dietary factors the key?
BMC Med 2014; 12:176.
82. Endeshaw M, Abebe F, Bedimo M, Asart A. Diet and Pre-eclampsia: A Prospective
Multicentre Case-Control Study in Ethiopia. Midwifery 2015; 31 (6):617-624.
83. Corella D, Tai ES, Sorli JV, Chew SK, Coltell O, Sotos-Prieto M, Garcia-Rios A, Estruch
R, Ordovas JM. Association between the APOA2 promoter polymorphism and body
weight in Mediterranean and Asian populations: replication of a gene-saturated fat
interaction. Int J Obes (Lond) 2011; 35 (5):666-675.
84. Martinez-Gonzalez MA, Salas-Salvado J, Estruch R, Corella DD, Fito M, Ros E. Benefits
of the Mediterranean Diet: Insights from the PREDIMED Study. Prog Cardiovasc Dis
2015.
85. Smith CE, Tucker KL, Arnett DK, Noel SE, Corella D, Borecki IB, Feitosa MF,
Aslibekyan S, Parnell LD, Lai CQ, Lee YC, Ordovas JM. Apolipoprotein A2
polymorphism interacts with intakes of dairy foods to influence body weight in 2 U.S.
populations. J Nutr 2013; 143 (12):1865-1871.
86. Zaki ME, Amr KS, Abdel-Hamid M. APOA2 Polymorphism in Relation to Obesity and
Lipid Metabolism. Cholesterol 2011; 2013:289481.
87. Zaki ME, Amr KS, Abdel-Hamid M. Evaluating the association of APOA2
polymorphism with insulin resistance in adolescents. Meta Gene 2014; 2:366-373.
88. Peloso GM, Demissie S, Collins D, Mirel DB, Gabriel SB, Cupples LA, Robins SJ,
Schaefer EJ, Brousseau ME. Common genetic variation in multiple metabolic pathways
influences susceptibility to low HDL-cholesterol and coronary heart disease. J Lipid Res
2010; 51 (12):3524-3532.
89. Koning F. The molecular basis of celiac disease. J Mol Recognit 2003; 16 (5):333-336.
90. Meresse B, Ripoche J, Heyman M, Cerf-Bensussan N. Celiac disease: from oral tolerance
to intestinal inflammation, autoimmunity and lymphomagenesis. Mucosal Immunol 2009;
2 (1):8-23.
91. Schuppan D, Zimmer KP. The diagnosis and treatment of celiac disease. Dtsch Arztebl
Int 2013; 110 (49):835-846.
92. Brottveit M, Beitnes AC, Tollefsen S, Bratlie JE, Jahnsen FL, Johansen FE, Sollid LM,
Lundin KE. Mucosal cytokine response after short-term gluten challenge in celiac disease
and non-celiac gluten sensitivity. Am J Gastroenterol 2013; 108 (5):842-850.
93. Kabbani TA, Vanga RR, Leffler DA, Villafuerte-Galvez J, Pallav K, Hansen J,
Mukherjee R, Dennis M, Kelly CP. Celiac disease or non-celiac gluten sensitivity? An
approach to clinical differential diagnosis. Am J Gastroenterol 2014; 109 (5):741-746;
quiz 747.
94. Fasano A, Sapone A, Zevallos V, Schuppan D. Nonceliac gluten sensitivity.
Gastroenterology 2014; 148 (6):1195-1204.
95. Shepherd SJ, Halmos E, Glance S. The role of FODMAPs in irritable bowel syndrome.
Curr Opin Clin Nutr Metab Care 2014; 17 (6):605-609.
96. Ashat M, Kochhar R. Non-celiac gluten hypersensitivity. Trop Gastroenterol 2014; 35
(2):71-78.
97. Mansueto P, Seidita A, D'Alcamo A, Carroccio A. Non-celiac gluten sensitivity:
literature review. J Am Coll Nutr 2014; 33 (1):39-54.
98. Nijeboer P, Bontkes HJ, Mulder CJ, Bouma G. Non-celiac gluten sensitivity. Is it in the
gluten or the grain? J Gastrointestin Liver Dis 2013; 22 (4):435-440.
99. Meresse B, Malamut G, Cerf-Bensussan N. Celiac disease: an immunological jigsaw.
Immunity 2012; 36 (6):907-919.
100. Uibo R, Tian Z, Gershwin ME. Celiac disease: a model disease for gene-environment
interaction. Cell Mol Immunol 2011; 8 (2):93-95.

More Related Content

What's hot

Will the real vegetarian please stand up? An investigation of dietary restrai...
Will the real vegetarian please stand up? An investigation of dietary restrai...Will the real vegetarian please stand up? An investigation of dietary restrai...
Will the real vegetarian please stand up? An investigation of dietary restrai...kurutemanko
 
Nxy292
Nxy292Nxy292
Lec 2 community dietary assessment
Lec 2 community dietary assessmentLec 2 community dietary assessment
Lec 2 community dietary assessmentSiham Gritly
 
Dietary evaluation in the community
Dietary evaluation in the communityDietary evaluation in the community
Dietary evaluation in the community
Chamath Fernando
 
Rao 6a varieties of measurement for food security
Rao 6a  varieties of measurement for food securityRao 6a  varieties of measurement for food security
Rao 6a varieties of measurement for food securitySizwan Ahammed
 
Flechtner
FlechtnerFlechtner
Flechtner
silver1111
 
How Major Restaurant Chains Plan Their Menus
How Major Restaurant Chains Plan Their MenusHow Major Restaurant Chains Plan Their Menus
How Major Restaurant Chains Plan Their MenusMark Lyons
 
Nutritional support in surgical patients
Nutritional support in surgical patientsNutritional support in surgical patients
Nutritional support in surgical patients
OmarAlaidaroos3
 
Bmjopen 2017-020574
Bmjopen 2017-020574Bmjopen 2017-020574
Bmjopen 2017-020574
Universidade Federal do Piauí
 
Nutritiona status frederici Bozzeti Nutrition
Nutritiona status frederici Bozzeti NutritionNutritiona status frederici Bozzeti Nutrition
Nutritiona status frederici Bozzeti NutritionVladislava DJurasinovic
 
Perspectives of Nursing in the Care of the Patient with Diabetes Mellitus-Cr...
Perspectives of Nursing in the Care of the  Patient with Diabetes Mellitus-Cr...Perspectives of Nursing in the Care of the  Patient with Diabetes Mellitus-Cr...
Perspectives of Nursing in the Care of the Patient with Diabetes Mellitus-Cr...
CrimsonPublishersIOD
 
Gene and Environment Interactions
Gene and Environment InteractionsGene and Environment Interactions
Gene and Environment Interactions
Nadine Taylor
 
Levels of Anxiety and Quality of Life in Overweight Individuals Under Nutriti...
Levels of Anxiety and Quality of Life in Overweight Individuals Under Nutriti...Levels of Anxiety and Quality of Life in Overweight Individuals Under Nutriti...
Levels of Anxiety and Quality of Life in Overweight Individuals Under Nutriti...
CrimsonPublishersIOD
 
FOODOMICS:A NEW APPROACH TO FOOD SAFETY AND PRODUCTION
FOODOMICS:A NEW APPROACH TO FOOD SAFETY AND PRODUCTIONFOODOMICS:A NEW APPROACH TO FOOD SAFETY AND PRODUCTION
FOODOMICS:A NEW APPROACH TO FOOD SAFETY AND PRODUCTION
maryamarif31
 
Validation mcn
Validation mcnValidation mcn
Validation mcngisa_legal
 

What's hot (19)

Will the real vegetarian please stand up? An investigation of dietary restrai...
Will the real vegetarian please stand up? An investigation of dietary restrai...Will the real vegetarian please stand up? An investigation of dietary restrai...
Will the real vegetarian please stand up? An investigation of dietary restrai...
 
Nxy292
Nxy292Nxy292
Nxy292
 
Lec 2 community dietary assessment
Lec 2 community dietary assessmentLec 2 community dietary assessment
Lec 2 community dietary assessment
 
Dietary evaluation in the community
Dietary evaluation in the communityDietary evaluation in the community
Dietary evaluation in the community
 
Rao 6a varieties of measurement for food security
Rao 6a  varieties of measurement for food securityRao 6a  varieties of measurement for food security
Rao 6a varieties of measurement for food security
 
Flechtner
FlechtnerFlechtner
Flechtner
 
How Major Restaurant Chains Plan Their Menus
How Major Restaurant Chains Plan Their MenusHow Major Restaurant Chains Plan Their Menus
How Major Restaurant Chains Plan Their Menus
 
Rehabilitation moringa
Rehabilitation moringaRehabilitation moringa
Rehabilitation moringa
 
Nutrition Ref
Nutrition RefNutrition Ref
Nutrition Ref
 
Nutritional support in surgical patients
Nutritional support in surgical patientsNutritional support in surgical patients
Nutritional support in surgical patients
 
Bmjopen 2017-020574
Bmjopen 2017-020574Bmjopen 2017-020574
Bmjopen 2017-020574
 
Nutritiona status frederici Bozzeti Nutrition
Nutritiona status frederici Bozzeti NutritionNutritiona status frederici Bozzeti Nutrition
Nutritiona status frederici Bozzeti Nutrition
 
Perspectives of Nursing in the Care of the Patient with Diabetes Mellitus-Cr...
Perspectives of Nursing in the Care of the  Patient with Diabetes Mellitus-Cr...Perspectives of Nursing in the Care of the  Patient with Diabetes Mellitus-Cr...
Perspectives of Nursing in the Care of the Patient with Diabetes Mellitus-Cr...
 
Gene and Environment Interactions
Gene and Environment InteractionsGene and Environment Interactions
Gene and Environment Interactions
 
Levels of Anxiety and Quality of Life in Overweight Individuals Under Nutriti...
Levels of Anxiety and Quality of Life in Overweight Individuals Under Nutriti...Levels of Anxiety and Quality of Life in Overweight Individuals Under Nutriti...
Levels of Anxiety and Quality of Life in Overweight Individuals Under Nutriti...
 
Feeding Strategies paper
Feeding Strategies paperFeeding Strategies paper
Feeding Strategies paper
 
Paper nutyicion de carne
Paper nutyicion de carnePaper nutyicion de carne
Paper nutyicion de carne
 
FOODOMICS:A NEW APPROACH TO FOOD SAFETY AND PRODUCTION
FOODOMICS:A NEW APPROACH TO FOOD SAFETY AND PRODUCTIONFOODOMICS:A NEW APPROACH TO FOOD SAFETY AND PRODUCTION
FOODOMICS:A NEW APPROACH TO FOOD SAFETY AND PRODUCTION
 
Validation mcn
Validation mcnValidation mcn
Validation mcn
 

Viewers also liked

Mi primer slideshare
Mi primer slideshareMi primer slideshare
Mi primer slideshare
amaliapatrnina
 
Guide To Hire An Event Photographer
Guide To Hire An Event PhotographerGuide To Hire An Event Photographer
Guide To Hire An Event Photographer
Absolute Amusements
 
About form
About formAbout form
About form
sri rahayu ningsih
 
Why Event Professionals Should Care About Games
Why Event Professionals Should Care About GamesWhy Event Professionals Should Care About Games
Why Event Professionals Should Care About Games
Absolute Amusements
 
Expert Tips For A Perfect Christmas Party
Expert Tips For A Perfect Christmas PartyExpert Tips For A Perfect Christmas Party
Expert Tips For A Perfect Christmas Party
Absolute Amusements
 

Viewers also liked (8)

DELIVER_Circ 2004
DELIVER_Circ 2004DELIVER_Circ 2004
DELIVER_Circ 2004
 
Mi primer slideshare
Mi primer slideshareMi primer slideshare
Mi primer slideshare
 
Guide To Hire An Event Photographer
Guide To Hire An Event PhotographerGuide To Hire An Event Photographer
Guide To Hire An Event Photographer
 
About form
About formAbout form
About form
 
Why Event Professionals Should Care About Games
Why Event Professionals Should Care About GamesWhy Event Professionals Should Care About Games
Why Event Professionals Should Care About Games
 
resume
resumeresume
resume
 
Expert Tips For A Perfect Christmas Party
Expert Tips For A Perfect Christmas PartyExpert Tips For A Perfect Christmas Party
Expert Tips For A Perfect Christmas Party
 
STATE BANK INDIA.PDF
STATE BANK INDIA.PDFSTATE BANK INDIA.PDF
STATE BANK INDIA.PDF
 

Similar to Outline_2015_עתיד

Genomics and proteomics
Genomics and proteomicsGenomics and proteomics
Genomics and proteomicsAmshumala S
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
Razib Datta Shubhra
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
Raffia Siddique
 
Nutrigenomics
NutrigenomicsNutrigenomics
nutrigenomics-150422112851-conversion-gate01.pptx
nutrigenomics-150422112851-conversion-gate01.pptxnutrigenomics-150422112851-conversion-gate01.pptx
nutrigenomics-150422112851-conversion-gate01.pptx
Dr-Faisal Al-Shormany
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
Krushna Yadav D K
 
Individualization of dosage regime
Individualization of dosage regimeIndividualization of dosage regime
Individualization of dosage regime
SwarnaPriyaBasker
 
Sajjad.moradi
Sajjad.moradiSajjad.moradi
Nutrigenomics: Perio-nutrition interrelationship
Nutrigenomics: Perio-nutrition interrelationshipNutrigenomics: Perio-nutrition interrelationship
Nutrigenomics: Perio-nutrition interrelationship
Anushri Gupta
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
Cholleti Vennela
 
Personalized Nutrition
Personalized NutritionPersonalized Nutrition
Personalized Nutrition
redx9
 
Copyright © 2016 John Wiley & Sons, Inc. All rights reserved..docx
Copyright © 2016 John Wiley & Sons, Inc. All rights reserved..docxCopyright © 2016 John Wiley & Sons, Inc. All rights reserved..docx
Copyright © 2016 John Wiley & Sons, Inc. All rights reserved..docx
dickonsondorris
 
PPT nutrigenomics obesity in nutrition.ppt
PPT nutrigenomics obesity in nutrition.pptPPT nutrigenomics obesity in nutrition.ppt
PPT nutrigenomics obesity in nutrition.ppt
InnaMutmainnahMusa
 
Nutritional regulation of gene expression
Nutritional regulation of gene expressionNutritional regulation of gene expression
Nutritional regulation of gene expression
emmanueltendekayi
 
Nutragenomics 2
Nutragenomics 2Nutragenomics 2
Nutragenomics 2
maria khatoon
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
Shubhra RD
 
Bisphenol A and obesity, the estrogenic endocrine disrupting chemical
Bisphenol A and obesity, the estrogenic endocrine disrupting chemicalBisphenol A and obesity, the estrogenic endocrine disrupting chemical
Bisphenol A and obesity, the estrogenic endocrine disrupting chemical
ricguer
 

Similar to Outline_2015_עתיד (20)

Genomics and proteomics
Genomics and proteomicsGenomics and proteomics
Genomics and proteomics
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
 
nutrigenomics-150422112851-conversion-gate01.pptx
nutrigenomics-150422112851-conversion-gate01.pptxnutrigenomics-150422112851-conversion-gate01.pptx
nutrigenomics-150422112851-conversion-gate01.pptx
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
 
Neutrigenomics
Neutrigenomics Neutrigenomics
Neutrigenomics
 
Individualization of dosage regime
Individualization of dosage regimeIndividualization of dosage regime
Individualization of dosage regime
 
Sajjad.moradi
Sajjad.moradiSajjad.moradi
Sajjad.moradi
 
Nutrigenomics: Perio-nutrition interrelationship
Nutrigenomics: Perio-nutrition interrelationshipNutrigenomics: Perio-nutrition interrelationship
Nutrigenomics: Perio-nutrition interrelationship
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
 
Personalized Nutrition
Personalized NutritionPersonalized Nutrition
Personalized Nutrition
 
Copyright © 2016 John Wiley & Sons, Inc. All rights reserved..docx
Copyright © 2016 John Wiley & Sons, Inc. All rights reserved..docxCopyright © 2016 John Wiley & Sons, Inc. All rights reserved..docx
Copyright © 2016 John Wiley & Sons, Inc. All rights reserved..docx
 
PPT nutrigenomics obesity in nutrition.ppt
PPT nutrigenomics obesity in nutrition.pptPPT nutrigenomics obesity in nutrition.ppt
PPT nutrigenomics obesity in nutrition.ppt
 
Nutritional regulation of gene expression
Nutritional regulation of gene expressionNutritional regulation of gene expression
Nutritional regulation of gene expression
 
Nutragenomics 2
Nutragenomics 2Nutragenomics 2
Nutragenomics 2
 
Nutrigenomics
NutrigenomicsNutrigenomics
Nutrigenomics
 
Bisphenol A and obesity, the estrogenic endocrine disrupting chemical
Bisphenol A and obesity, the estrogenic endocrine disrupting chemicalBisphenol A and obesity, the estrogenic endocrine disrupting chemical
Bisphenol A and obesity, the estrogenic endocrine disrupting chemical
 

Outline_2015_עתיד

  • 1. ‫תזונתיות‬ ‫וגנטיקה‬ ‫גנומיקה‬ ‫של‬ ‫הרצף‬: ‫התקדמות‬,‫רלוונטיות‬,‫ויישום‬ ‫שרון‬ ‫אסנת‬,‫שני‬ ‫תואר‬,R.D. Continuum of Nutritional Genomics and Genetics: Progress, Relevance, Application Ossie (Osnat) Sharon, M.Sc., R.D. ‫תקציר‬ ‫של‬ ‫העתיד‬ ‫נחשבת‬ ‫היא‬ ‫אישית‬ ‫מותאמת‬ ‫תזונה‬ ‫התזונה‬ ‫תורת‬,‫ידי‬ ‫על‬ ‫שנוצר‬ ‫צורך‬ ‫על‬ ‫שמענה‬ ‫בין‬ ‫וסתירות‬ ‫להתערבויות‬ ‫עקביות‬ ‫לא‬ ‫תגובות‬ ‫המלצות‬.‫אישית‬ ‫מותאמת‬ ‫לרפואה‬ ‫במקביל‬, ‫ונוטריגנומיקה‬ ‫נוטריגנטיקה‬ ‫כולל‬ ‫זה‬ ‫תחום‬–‫איך‬ ‫האורגניזם‬ ‫ועל‬ ‫לזו‬ ‫זו‬ ‫על‬ ‫משפיעות‬ ‫ותזונת‬ ‫גנטיקה‬ ‫כולו‬–‫ואפי‬‫ואפיגנומיקה‬ ‫גנטיקה‬–‫על‬ ‫ההשפעה‬ ‫הבאים‬ ‫הדורות‬.‫גנים‬ ‫זוהו‬ ‫מחקרים‬,‫פולימורפיזם‬, ‫וסביבתיים‬ ‫הללו‬ ‫הגורמים‬ ‫בין‬ ‫גומלין‬ ‫ויחסי‬ ‫חומרים‬ ‫חילוף‬ ‫את‬ ‫המניעים‬,‫שמושפעת‬ ‫למידה‬ ‫פנוטיפית‬ ‫גמישות‬ ‫ידי‬ ‫על‬.‫הזוהו‬ ‫כה‬ ‫שעד‬ ‫אלה‬ ‫ופעילות‬ ‫לתזונה‬ ‫הקשר‬ ‫מבחינת‬ ‫אמינים‬ ‫כהכי‬ ‫לאסטרטג‬ ‫רתומים‬ ‫גופנית‬‫התערבותיות‬ ‫יות‬,‫ועוד‬ ‫להתברר‬ ‫ממשיכים‬ ‫אחרים‬. ‫מונע‬ ‫יוקר‬ ‫רקע‬ ‫על‬,‫בדיקות‬ ‫נוטריגנטקיה‬/‫נתן‬ ‫ויותר‬ ‫יותר‬ ‫הן‬ ‫נוטריגנומיקה‬ ‫לצרכן‬ ‫להשיג‬,‫העולם‬ ‫ברחבי‬ ‫ופופולריות‬.‫אתגר‬ ‫תוצאות‬ ‫קבלת‬ ‫לאחר‬ ‫הכוונה‬ ‫חוסר‬ ‫היה‬ ‫נוסף‬ ‫הצרכן‬ ‫ידי‬ ‫על‬ ‫הבדיקות‬,‫בסיכונים‬ ‫שכרוכים‬. ‫לפנו‬ ‫מקודדים‬ ‫שגנים‬ ‫העובדה‬‫מרובים‬ ‫טיפים‬– ‫הרחב‬ ‫לקהל‬ ‫ידועים‬ ‫שפחות‬ ‫אלה‬ ‫כולל‬–‫מציג‬ ‫רב‬ ‫קליני‬ ‫פרופיל‬-‫דורש‬ ‫קרובות‬ ‫שלעתים‬ ‫גורמי‬ ‫לכאורה‬ ‫סותרות‬ ‫ואפילו‬ ‫מורכבות‬ ‫התערבויות‬,‫ולכן‬ ‫לספק‬ ‫יכול‬ ‫מקצוען‬ ‫שרק‬ ‫מאוזנת‬ ‫אסטרטגיה‬.‫זה‬ ‫האמריקאי‬ ‫הבריאות‬ ‫משרד‬ ‫את‬ ‫הניע‬,‫גם‬ ‫כמו‬ ‫אמריקאיות‬ ‫שאינן‬ ‫חברות‬ ‫כמה‬,‫בדיק‬ ‫לדרוש‬‫ה‬ ‫בלבד‬ ‫מומחים‬ ‫ידי‬ ‫על‬ ‫מכוונים‬ ‫וייעוץ‬–‫של‬ ‫במקרה‬ ‫לתזונה‬ ‫הקשורות‬ ‫אינדיקציות‬,‫דיאטנים‬ ‫ידי‬ ‫על‬/‫ות‬. ‫מכך‬ ‫כתוצאה‬,‫דיאטנים‬/‫בחוד‬ ‫שומו‬ ‫נמצאים‬ ‫ות‬ ‫ידי‬ ‫על‬ ‫אישית‬ ‫מותאמת‬ ‫תזונה‬ ‫של‬ ‫החנית‬ ‫נלווים‬ ‫מקצועות‬,‫גם‬ ‫עבור‬ ‫חשובות‬ ‫השלכות‬ ‫עם‬ ‫הצרכן‬ ‫בריאות‬ ‫וגם‬ ‫מקצועי‬ ‫קידום‬.‫להמשך‬ ‫בנוסף‬ ‫ב‬ ‫של‬‫בישראל‬ ‫נוטריגנומיקה‬ ‫מחקרי‬ ‫יצוע‬,‫בדיקות‬ ‫לדיאטנים‬ ‫המיועדות‬ ‫אישיות‬/‫לכאן‬ ‫הגיעו‬ ‫ות‬ ‫לאחרונה‬,‫להזדמנויות‬ ‫דלתות‬ ‫פתיחת‬ ‫לטובת‬ ‫דומות‬. Abstract Personalized nutrition is considered to be the future of dietetics, answering a need created by inconsistent responses to interventions and resultant contradictions within/between recommendations. Parallel to personalized medicine, it comprises nutrigenetics and nutrigenomics – how genetics and nutrition impact one another and the organism as a whole – and epigenetics and epigenomics – how this impact affects future generations. Research has identified genes, polymorphisms, and interactions between these and environmental factors that drive metabolism, to an extent influenced by phenotypic plasticity. Those most reliably correlating to nutrition and physical activity are currently being harnessed for interventional strategies, while others continue to be clarified. Once prohibitively expensive, consumer nutrigenetic/nutrigenomic testing is becoming increasingly affordable and popular globally. An additional challenge has been inadequate direction once results are received by the consumer, with associated risks. The fact that genes code for multiple phenotypes – including those less publicized – presents a multifactorial clinical profile that often requires composite and even seemingly contradictory interventions, and therefore a balanced strategy that only a professional can provide. This has prompted the United States Food and Drug Administration, as well as some non-American companies, to require expert-directed testing and counseling – in the case of nutrition-related indications, by dietitians. As a result, dietitians are being put on the forefront of personalized nutrition by ancillary professions, with important implications for both professional advancement and consumer health. In addition to Israel continuing to host nutrigenomics research, personal testing technologies have recently become available to practitioners here, opening the doors to similar opportunities.
  • 2. Summary Nutrigenomics is a blanket term for a new scientific discipline that uses modern genomics technology to study the relationship between genes, nutrition and health. It has long been apparent that some people respond differently from others to certain foods. Nutrigenomics allows us to understand how our genes affect the way we respond to the foods, beverages and supplements we consume [1-3]. It is the position of the Academy of Nutrition and Dietetics that “nutritional genomics provides insight into how diet and genotype interactions affect phenotype.” Key Definitions Nutritional genomics The relationship between the human genome, nutrition, and health Nutrigenetics How the genotype drives the phenotypic response to dietary intake • Monogenic Result of alteration to one particular gene • Polygenic: Result of alterations to more than one gene Nutrigenomics How dietary intake drives genotypic activity Phenotypic Plasticity How the phenotype adapts to environmental changes, including diet SNP Single/simple nucleotide polymorphism: common variation of a single nucleotide (A, T, C, G) in a common DNA sequence Metabolomics The study of chemical processes involving metabolites Nutritional epigenetics How phenotypic response to diet drives the genotype over generations Nutritional epigenomics How nutritional epigenetic modifications affect the entire genome DoHAD Developmental origins of health and disease • Impact of experiences during initial phases of somatic development on lifelong health, including chronic disease risk • Models proposed: thrifty phenotype, programming, predictive adaptive response theories, concept of match or mismatch • Possible mechanisms: environmental effects on gene expression, hormonal signals transmitted to fetus/infant via placenta/lactation Key Tenets • Common dietary chemicals act on the human genome, either directly or indirectly, to alter gene expression or structure. • Under certain circumstances and in some individuals, diet can be a serious risk factor for a number of diseases. • Some diet-regulated genes (and their common variants, i.e. “SNP”s) are likely to play a role in the onset, incidence, progression, and/or severity of chronic diseases of individuals and their descendants. • The degree to which diet influences the balance between healthy and disease states may depend on an individual’s genetic makeup. • Dietary intervention based on knowledge of nutritional requirement, nutritional status, and genotype (i.e., “individualized nutrition”) can be used to prevent, mitigate or cure chronic disease.
  • 3. Introduction Since completion of the human genome project, understanding of complex interactions between environmental factors such as diet and genes has progressed considerably. The knowledge that metabolic pathways may be altered in individuals with genetic variants in the presence of certain dietary exposures offers great potential for personalized nutrition advice, and epigenetics and nutrigenetics have been used to assess the need and status of specific nutrients. This has dramatically increased the potential to individualize diets using dietary, phenotypic and genotypic data, with improved outcomes [1-6]. Recent genetic interest and research focus surrounds the direct connections between metabolism and developmental dynamics, which now represents an important conceptual challenge to explain many aspects of metabolic dysfunction. Several components of the epigenetic machinery require intermediates of cellular metabolism for enzymatic function. For example, specific epigenetic influences of dietary glucose and lipid consumption, as well as undernutrition, are observed across numerous organs and pathways associated with metabolism. Studies have started to define the chromatin-dependent mechanisms underlying persistent and pathophysiological changes induced by altered metabolism. Importantly, numerous recent studies demonstrate that gene regulation underlying phenotypic determinants of adult metabolic health is influenced by maternal and early postnatal diet. These emerging concepts open new perspectives to combat the rising global epidemic of metabolic disorders [7-9]. Despite widespread promotion of population-based healthy eating guidelines over the half- century, preventable diseases remain the leading cause of mortality in the developed world. Moreover, the greatest numbers of disease events occur in individuals with a lower conventional risk of disease [10-14]. Inconsistent research outcomes and real-world responses limit utility of recommendations for health attainment and maintenance, particularly in the areas of weight and chronic disease such as type 2 diabetes mellitus, cardiovascular disease, and cancer [15, 16]. Examples include the following: • Saturated fat risks • Omega-3 fatty acid needs • Low-fat vs. low-carbohydrate diet for weight loss • Increased fiber needs • Intolerances, e.g. to lactose, gluten • Sodium restriction risk vs. advantage • Increased need for specific micronutrients • Animal vs. plant-based • Relative risk from high-risk cooking methods, such as charring and frying • Harmful vs. beneficial effects of diet-related habits, e.g. caffeine, alcohol • Physical activity needs and injury risk • Efforts required for healthy weight loss and maintenance • Impact of the above on perinatal outcomes, including long-term in offspring Personalized nutrition interventions may have greater potential for reducing the global burden of preventable diseases and for promoting better health across the lifespan than the conventional “one size fits all” approach [4-6, 10]. Further, such measures during the prenatal period – including prior to conception – have implications not only for improved maternal outcomes and future health, but also for the long-term health of the offspring and future generations [17-22]. Genetic testing is increasingly being embraced for the purpose of personalized nutrition planning. Once available only to physicians through medical testing, provision of genetic polymorphism (SNP) information is now widely available direct-to-consumer. Method Background Over the past decade, several companies have formed partnerships with laboratories to provide an increasing variety of genetic and immunologic tests that indicate predispositions for physiologic imbalances that, left unchecked, could ultimately lead to serious disorders [23]. Most of these tests are provided by having the consumer send a saliva sample to a central laboratory, which then performs the analysis for a predetermined group of genes,
  • 4. polymorphisms, and alleles. The results are then provided either to the consumer or to a healthcare practitioner. Practitioners benefit because they have a new set of tools with potential to improve their ability to prevent disease and help restore health, and patients benefit from new opportunities to avert disorders before they cause significant disruptions, doing away with the hit-or-miss, trial-and- error that characterizes the pursuit of the ideal health-oriented lifestyle [20]. However, challenges and questions remain regarding use and acceptance of nutrigenomic testing, including the following: • Accuracy of tests provided Research into the many polymorphisms involved in nutrigenomics has yielded many promising leads, but only some have been consistently linked to health conditions in a manner that can benefit from specific interventions. • Professional acceptance It is not clear to some health care providers how nutrigenetic testing and related interventions will yield benefits over current standards of care. However, nutrigenetic screening has been associated with greater improvements towards and maintenance of health goals when compared to conventional intervention [24-26]. • Government acceptance Government bodies are concerned that consumers must be protected from unrealistic claims and misinterpretations of complex genomic information, and that direct-to- consumer test results are often not adequately understood by patients and thus raise concerns when used to self-manage [27]. • Fraudulent practices of service providers Impartial evaluations have found that many internet-based direct-to-consumer genetic tests were fraudulent in some way [28]. • Quality of associated care Many companies are linked to nutritional supplement companies, encouraging pills over food, where research suggests a food-based approach to be superior [29-31]. • Public acceptance Many consumers remain unaware of the potentially controllable nature of genetic influence on health. When informed, research has identified health and clear consumer benefits as key motivators in the uptake of genetic testing, with individuals reporting personal experience of disease/symptoms being more willing to undergo genetic testing for the purpose of personalized nutrition [20], particularly when information is delivered by a qualified healthcare professional such as a dietitian [32]. A perceived susceptibility to disease has also been suggested to improve motivation to change behavior, which is a key barrier in the success of any nutrition intervention [19]. • Compliance with recommendations Where widespread lack of compliance with conventional nutrition recommendations has cast doubt on the efficacy of methods, the nutrigenetic approach has resulted in increased compliance with lifestyle interventions [33]. Sources of Testing Some companies that offer nutrigenetic testing products are designing programs that bypass principal concerns of multiple authorities. • Testing only of SNPs that have consistently been linked to health conditions in a manner that can benefit from specific interventions. • Provision of nutrigenetic testing through qualified professionals, specifically dietitians, packaged with nutritional counseling. Genes, SNPs, and Implications The following are examples of SNPs meeting the above criteria, pointing to a strong indication for professional guidance by a dietitian/nutritionist: • MTHFR 677 CT/TT
  • 5. Methylenetetrahydrofolate reductase (MTHFR) catalyzes the metabolism of folate and nucleotides needed for DNA synthesis and repair, and as such gene plays an important role in the genomic integrity and genetic stability. The most common SNP, MTHFR C677T is known primarily for increasing the need for dietary folate and most frequently linked to homocysteinemia and its impact on cardiovascular risk [34]. MTHFR SNPs have also been found in research to be associated with cellular functions related to various cancer linked to key nutritional factors beyond folate (including breast, gastric, colon, thyroid) [35-40], ovarian [41, 42] and sperm integrity [43], gestational outcomes beyond neural tube defects [44, 45], thyroid function [46-48], and neurological disorders, including Parkinson’s [49] and autism [50, 51], and metabolic risks related to premature menopause, bone health [52], and development of type 2 diabetes, as well as increased need for riboflavin, pyridoxine, and other nutritional interventions related to the above issues [53]. While over-the-counter folic acid supplements have been recommended as a solution, there are concerns that standard options are not adequate to meet the needs of SNP carriers; research has even suggested that common supplements may actually increase risk of methylation-related disorders [54-56]. In carriers of the polymorphism, either a special (more expensive) supplemental form of folate and/or food-based intervention is preferred – particularly relevant when considering the additional advantages of foods high in folate, which tend also to be naturally high in fiber. When frequently concurrent conditions of hypothyroid and coagulation disorders occur, this demands balancing intake of the above foods with medications, i.e. thyroxine and warfarin, as well as other B- vitamins with which folate performs its activities, also warranting intervention by a qualified dietitian/nutritionist. • GSTT1 and GSMT1 The GSTT1 and GSMT1 genes produce proteins for the phase II metabolizing glutathione S-transferase enzyme family, responsible detoxifying numerous potentially cytotoxic/genotoxic compounds and attenuating inflammatory cytokines such as interleukin-6 (IL6). GSTT1 is also known for its key role in the utilization of vitamin C, and a dysfunctional version of the gene results in a reduced ability to process vitamin C and need to ensure the minimum recommended intake (Dietary Reference Intake, DRI) to maintain normal levels, where the functional version enables individuals to thrive with less [18]. In clinical research, IL-6 was inversely associated with total isothiocyanate excretion in GSTM1-null/GSTT1-null individuals. Inadequate upregulation of these protective enzymes has been linked to cancer, suggesting a need not only to decrease intake of offending compounds, but also to increase intake of foods rich in indole antioxidants,, such as those in cruciferous vegetables [57]. When occurring in combination with the above-noted MTHFR polymorphism, there may be increased risk of both acute and chronic myelogenous leukemias and autism spectrum disorders (ASD) [51, 58-60]. Given the implications not only for individuals but also their offspring, there is a clear advantage to the increased compliance with a preventive diet that can be attained with personalized dietetic counseling. • TCF7L2 Studies have consistently shown that common variants in the TCF7L2 gene significantly predict type 2 diabetes risk [61-63], due to a deleterious effect related to β-cell function [64-66] and downregulation of gastric inhibitory polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) receptors [65]. This has been demonstrated to be relevant to gestational diabetes as well [67-70]. The TCF7L2 has been closely linked to the role of fiber in maintenance of normal glucose metabolism [71-73], including prevention of progression to the prediabetic state in high-risk individuals [74]. In addition, compelling evidence has suggested important roles of TCF7L2 in the regulation of body weight and adiposity including 1) taking part in the Wnt signaling cascade, which inhibits adipogenesis and stimulates leptin production in mature adipocytes in response to nutritional status [75], 2) gene expression in adipose tissue differing by genotype after caloric restriction, 3) the association with hunger-satiety hormones that influence weight loss, and 4) the promotion of the transcription of
  • 6. proglucagon, which induces the synthesis of glucagon-like peptide, which is a regulator of insulin and glucagon secretion, appetite, and food intake [76-78]. Despite this evidence, reports on the association of TCF7L2 variants with body weight and composition as well as diabetes risk have been contradictory. One possible explanation for the discrepancies might be interactions between TCF7L2 genotypes and environmental factors such as dietary intake [67, 73, 79]. Such interactions have been reported in observational studies for dietary fat, carbohydrate intake, and animal protein, and are largely dependent on specific SNPs. For example, a recent randomized trial reported that obese individuals with the rs7903146 or rs12255372 risk allele have better weight/adiposity loss responses in weight loss with a low- rather than high-fat dietary intervention [79] and additional genes have been observed to interact with prenatal nutrition in relation to T2D risk and glucose levels in later life [73]. Given that a possible predisposition to type 2 diabetes generally indicates a diet low in carbohydrates and attainment and maintenance of a healthy weight, and that efforts to reduce fat intake have historically resulted in high carbohydrate intake, guidance by a dietitian is indicated to assist in constructing a balanced diet that can be sustained long- term. • NOS3 The NOS3 gene upregulates nitric oxide synthase and production of nitric oxide, which plays an important role in the function of the vascular epithelium. New research has shown that variations in the NOS3 gene interact with omega-3 fatty acids in different ways to impact how the body processes triglycerides, and the degree to which elevated triglycerides impair circulation. Previous studies have produced mixed results relating to the effects of omega-3 fat on triglyceride levels between individuals. Some people experience a significant reduction in triglyceride levels in response to increasing omega-3 fat intake, whereas others experience little benefit. The reasons for these differences have been unclear until a recent breakthrough study* showed that the effect of omega-3 fat on triglyceride levels depends on variations in a gene called NOS3. Those who have the GT or TT variant of the gene are at greater risk of elevated triglyceride levels when consuming a diet low in omega-3 fats, compared to those who have the GG variant. Beyond this function, NOS3 SNPs such as rs1799983 have been and continue to be investigated for influence on cancers, obesity, and perinatal disorders such as recurrent miscarriage and pre-eclampsia. Pre-eclampsia is a well-known example of a potentially preventable/manageable condition that in addition to omega-3 intake [80], nutrition- related prevention and management of pre-eclampsia is multi-factorial [81, 82], and is an accepted indication for dietitian involvement. Pre-eclampsia in turn increases • APOA2 Saturated fats have been associated with health conditions such as diabetes, cardiovascular disease, and obesity. However, the connection between saturated fats and obesity, until recently, has been inconsistent. A number of studies have now shown that the effect of saturated fat on obesity can be influenced by variations in APOA2 [83-87]. The APOA2 gene upregulates production of apolipoprotein A-II, which impacts dietary fat utilization. Scientists now understand that there are different variations in the APOA2 gene present in the human population and that these different versions of the gene interact with saturated fat in unique ways to influence energy balance and ultimately the risk of obesity [83, 86]. Those people who have the CC variant of the gene are at a higher risk of developing obesity when consuming a diet high in saturated fats than those possessing the TT or TC variant of the gene [85]. Additionally, the CC polymorphism has been associated with elevated levels of visceral adipose tissue and development of insulin resistance, independent of gender, age and body mass index [87]. Related concerns point to the influence of APOA2 SNPs on inflammation and blood lipids [88]. The above issues require guidance regarding a diet that balances low saturated fat with low simple carbohydrate intake, while protecting against the oxidation risk [84]. • HLA DQ2/8 The human leukocyte antigen (HLA) gene is the most important genetic predictor of gluten intolerance. Nearly all individuals with celiac disease [89-91] and more than half
  • 7. of those with non-celiac gluten sensitivity (NCGS) [92, 93] have the DQ2 or DQ8 (“high risk”) version of the HLA gene, compared to only 25% of the general population. Celiac disease represents the most severe form of gluten intolerance and affects about 1% of the population. Another 5-10% of the population have non-celiac gluten sensitivity NCGS, which results in various gastrointestinal problems, and still others have wheat sensitivities related to its carbohydrate fractions rather than gluten, which in turn may be indicative of multiple sensitivities, e.g. to FODMAPs [94, 95], requiring guidance by a dietitian [96-98]. Recent research has suggested that in predisposed individuals, celiac may represent the final step in a continuum of severity of gluten intolerance following cumulative exposure [99, 100]. Testing for the relevant HLA forms may enable preventive dietary measures and/or redirect focus to other disorders with similar symptoms but differing interventions. It can also rule-out a need to undergo an invasive biopsy procedure, currently standard of care to rule out celiac in individuals experiencing characteristic symptom. Additional genes with SNPs indicating a need for nutritional management and currently being offered for direct-to-dietitian testing include APOE, ACE, CYP1A2, ADH1C, VDR, IL6, TNF, SOD2, CAT, GPX1, LCT, and FOT, each with complexities requiring intervention by a qualified professional. The above polymorphisms can occur in combination, potentially magnifying risk not only of characteristic disorders, but also to diseases specifically associated with concurrence of alleles, again pointing to the need for qualified dietetic guidance for both prevention and management, for the individual and future generations. Conclusions Personalized interventions that take into account genetic variations in response to nutritional factors may have greater potential for disease prevention and management than a “one size fits all” approach, and may be better accepted by patients. The fact that genes code for multiple phenotypic effects presents a unique, multifactorial picture for individuals that require the type of balancing between lifestyle factors that only a professional can provide. This is highly likely to improve compliance and quality of results and in a much shorter timeframe than are associated with conventional interventions. As a result, dietitians are being put on the forefront of personalized nutrition by ancillary professions, with important implications for both professional advancement and consumer health. In addition to Israel continuing to host nutrigenomics research, personal testing technologies have recently become available to practitioners here, opening the doors to similar opportunities.
  • 8. References 1. Isaak CK, Siow YL. The evolution of nutrition research. Can J Physiol Pharmacol 2013; 91 (4):257-267. 2. Mocchegiani E, Costarelli L, Giacconi R, Malavolta M, Basso A, Piacenza F, Ostan R, Cevenini E, Gonos ES, Monti D. Micronutrient-gene interactions related to inflammatory/immune response and antioxidant activity in ageing and inflammation. A systematic review. Mech Ageing Dev 2014; 136-137:29-49. 3. Perez-Martinez P, Phillips CM, Delgado-Lista J, Garcia-Rios A, Lopez-Miranda J, Perez- Jimenez F. Nutrigenetics, metabolic syndrome risk and personalized nutrition. Curr Vasc Pharmacol 2013; 11 (6):946-953. 4. Bordoni A, Capozzi F. Foodomics for healthy nutrition. Curr Opin Clin Nutr Metab Care 2014; 17 (5):418-424. 5. Gibney MJ, McNulty BA, Ryan MF, Walsh MC. Nutritional phenotype databases and integrated nutrition: from molecules to populations. Adv Nutr 2014; 5 (3):352S-357S. 6. Jones ML, Ganopolsky JG, Martoni CJ, Labbe A, Prakash S. Emerging science of the human microbiome. Gut Microbes 2014; 5 (4):446-457. 7. Desai M, Jellyman JK, Ross MG. Epigenomics, gestational programming and risk of metabolic syndrome. Int J Obes (Lond) 2015; 39 (4):633-641. 8. Keating ST, El-Osta A. Epigenetics and metabolism. Circ Res 2015; 116 (4):715-736. 9. Tarrade A, Panchenko P, Junien C, Gabory A. Placental contribution to nutritional programming of health and diseases: epigenetics and sexual dimorphism. J Exp Biol 2015; 218 (Pt 1):50-58. 10. Bauer UE, Briss PA, Goodman RA, Bowman BA. Prevention of chronic disease in the 21st century: elimination of the leading preventable causes of premature death and disability in the USA. Lancet 2014; 384 (9937):45-52. 11. Dwyer JT, Melanson KJ, Ms US, Cross P, Wilson M. Dietary Treatment of Obesity. 2000. 12. Kwon BJ, Kim DW, Her SH, Kim DB, Jang SW, Cho EJ, Ihm SH, Kim HY, Youn HJ, Seung KB, Kim JH, Rho TH. Metabolically obese status with normal weight is associated with both the prevalence and severity of angiographic coronary artery disease. Metabolism 2013; 62 (7):952-960. 13. Karelis AD, St-Pierre DH, Conus F, Rabasa-Lhoret R, Poehlman ET. Metabolic and body composition factors in subgroups of obesity: what do we know? J Clin Endocrinol Metab 2004; 89 (6):2569-2575. 14. Nowak M, Grzywa M. Metabolic obesity in normal weight individuals and cardiovascular disease. Pol Merkur Lekarski 2011; 31 (182):118-121. 15. Alexander DD, Weed DL, Miller PE, Mohamed MA. Red Meat and Colorectal Cancer: A Quantitative Update on the State of the Epidemiologic Science. J Am Coll Nutr 2010:1- 23. 16. Jenkins DJ, Josse AR, Dorian P, Burr ML, LaBelle Trangmar R, Kendall CW, Cunnane SC. Heterogeneity in randomized controlled trials of long chain (fish) omega-3 fatty acids in restenosis, secondary prevention and ventricular arrhythmias. J Am Coll Nutr 2008; 27 (3):367-378. 17. Bouchard C, Ordovas JM. Fundamentals of nutrigenetics and nutrigenomics. Prog Mol Biol Transl Sci 2012; 108:1-15.
  • 9. 18. Cahill LE, Fontaine-Bisson B, El-Sohemy A. Functional genetic variants of glutathione S-transferase protect against serum ascorbic acid deficiency. Am J Clin Nutr 2009; 90 (5):1411-1417. 19. Nielsen DE, El-Sohemy A. A randomized trial of genetic information for personalized nutrition. Genes Nutr 2012; 7 (4):559-566. 20. Ronteltap A, van Trijp JC, Renes RJ. Consumer acceptance of nutrigenomics-based personalised nutrition. Br J Nutr 2009; 101 (1):132-144. 21. Subbiah MT. Nutrigenetics and nutraceuticals: the next wave riding on personalized medicine. Transl Res 2007; 149 (2):55-61. 22. Millis RM. Epigenetics and hypertension. Curr Hypertens Rep 2011; 13 (1):21-28. 23. Preuss C, Das MK, Pathak YV. Genomics and natural products: role of bioinformatics and recent patents. Recent Pat Biotechnol; 8 (2):144-151. 24. Rizkalla SW, Prifti E, Cotillard A, Pelloux V, Rouault C, Allouche R, Laromiguiere M, Kong L, Darakhshan F, Massiera F, Clement K. Differential effects of macronutrient content in 2 energy-restricted diets on cardiovascular risk factors and adipose tissue cell size in moderately obese individuals: a randomized controlled trial. Am J Clin Nutr 2012; 95 (1):49-63. 25. Roy N, Barnett M, Knoch B, Dommels Y, McNabb W. Nutrigenomics applied to an animal model of Inflammatory Bowel Diseases: transcriptomic analysis of the effects of eicosapentaenoic acid- and arachidonic acid-enriched diets. Mutat Res 2007; 622 (1- 2):103-116. 26. Sanchez-Muniz FJ, Canales A, Nus M, Bastida S, Guillen M, Corella D, Olmedilla- Alonso B, Granado-Lorencio F, Benedi J. The antioxidant status response to low-fat and walnut paste-enriched meat differs in volunteers at high cardiovascular Risk carrying different PON-1 polymorphisms. J Am Coll Nutr 2012; 31 (3):194-205. 27. Food and Drug Administration (FDA). Warning Letter: 23andMe, Inc. 11/22/13. 2013. 28. Saukko P. State of play in direct-to-consumer genetic testing for lifestyle-related diseases: market, marketing content, user experiences and regulation. Proc Nutr Soc 2013; 72 (1):53-60. 29. Fortmann SP, Burda BU, Senger CA, Lin JS, Whitlock EP. Vitamin and mineral supplements in the primary prevention of cardiovascular disease and cancer: An updated systematic evidence review for the U.S. Preventive Services Task Force. Ann Intern Med 2013; 159 (12):824-834. 30. Jacobs DR, Jr., Gross MD, Tapsell LC. Food synergy: an operational concept for understanding nutrition. Am J Clin Nutr 2009; 89 (5):1543S-1548S. 31. Ward E. Addressing nutritional gaps with multivitamin and mineral supplements. Nutr J 2014; 13:72. 32. Nielsen DE, Shih S, El-Sohemy A. Perceptions of genetic testing for personalized nutrition: a randomized trial of DNA-based dietary advice. J Nutrigenet Nutrigenomics 2014; 7 (2):94-104. 33. Nielsen DE, El-Sohemy A. Disclosure of genetic information and change in dietary intake: a randomized controlled trial. PLoS One 2014; 9 (11):e112665. 34. Trimmer EE. Methylenetetrahydrofolate reductase: biochemical characterization and medical significance. Curr Pharm Des 2013; 19 (14):2574-2593.
  • 10. 35. Slaoui M, Razine R, Ibrahimi A, Attaleb M, Mzibri ME, Amrani M. Breast cancer in Morocco: a literature review. Asian Pac J Cancer Prev 2014; 15 (3):1067-1074. 36. Xia LZ, Liu Y, Xu XZ, Jiang PC, Ma G, Bu XF, Zhang YJ, Yu F, Xu KS, Li H. Methylenetetrahydrofolate reductase C677T and A1298C polymorphisms and gastric cancer susceptibility. World J Gastroenterol 2014; 20 (32):11429-11438. 37. Nassiri M, Kooshyar MM, Roudbar Z, Mahdavi M, Doosti M. Genes and SNPs associated with non-hereditary and hereditary colorectal cancer. Asian Pac J Cancer Prev 2013; 14 (10):5609-5614. 38. Chen Y, Wang B, Yan S, Wang YG. Significant association between MTHFR C677T polymorphism and thyroid cancer risk: evidence from a meta-analysis. Genet Test Mol Biomarkers 2014; 18 (10):695-702. 39. Yan Y, Han F, Fu H, Xia W, Qin X. Association between MTHFR C677T polymorphism and thyroid cancer risk: a meta-analysis. Tumour Biol 2014; 35 (8):7707-7712. 40. Yang YM, Zhang TT, Yuan L, Ren Y. The association between the C677T polymorphism in MTHFR gene and the risk of thyroid cancer: a meta-analysis. Eur Rev Med Pharmacol Sci 2014; 18 (15):2097-2101. 41. Casarini L, Simoni M. Gene polymorphisms in female reproduction. Methods Mol Biol 2014; 1154:75-90. 42. Rah H, Jeon YJ, Choi Y, Shim SH, Yoon TK, Choi DH, Cha SH, Kim NK. Association of methylenetetrahydrofolate reductase (MTHFR 677C>T) and thymidylate synthase (TSER and TS 1494del6) polymorphisms with premature ovarian failure in Korean women. Menopause 2012; 19 (11):1260-1266. 43. Singh K, Jaiswal D. One-carbon metabolism, spermatogenesis, and male infertility. Reprod Sci 2012; 20 (6):622-630. 44. Lewis SJ. Commentary: One-carbon metabolism has major implications for fetal growth and development beyond neural tube defects. Int J Epidemiol 2014; 43 (5):1498-1499. 45. Yajnik CS, Chandak GR, Joglekar C, Katre P, Bhat DS, Singh SN, Janipalli CS, Refsum H, Krishnaveni G, Veena S, Osmond C, Fall CH. Maternal homocysteine in pregnancy and offspring birthweight: epidemiological associations and Mendelian randomization analysis. Int J Epidemiol 2014; 43 (5):1487-1497. 46. Arakawa Y, Watanabe M, Inoue N, Sarumaru M, Hidaka Y, Iwatani Y. Association of polymorphisms in DNMT1, DNMT3A, DNMT3B, MTHFR and MTRR genes with global DNA methylation levels and prognosis of autoimmune thyroid disease. Clin Exp Immunol 2012; 170 (2):194-201. 47. Kakkoura MG, Demetriou CA, Loizidou MA, Loucaides G, Neophytou I, Marcou Y, Hadjisavvas A, Kyriacou K. Single-nucleotide polymorphisms in one-carbon metabolism genes, Mediterranean diet and breast cancer risk: a case-control study in the Greek- Cypriot female population. Genes Nutr 2015; 10 (2):453. 48. Lambrinoudaki I, Kaparos G, Rizos D, Galapi F, Alexandrou A, Sergentanis TN, Creatsa M, Christodoulakos G, Kouskouni E, Botsis D. Apolipoprotein E and paraoxonase 1 polymorphisms are associated with lower serum thyroid hormones in postmenopausal women. Clin Endocrinol (Oxf) 2009; 71 (2):284-290. 49. Wu YL, Ding XX, Sun YH, Yang HY, Sun L. Methylenetetrahydrofolate reductase (MTHFR) C677T/A1298C polymorphisms and susceptibility to Parkinson's disease: a meta-analysis. J Neurol Sci 2013; 335 (1-2):14-21.
  • 11. 50. Frustaci A, Neri M, Cesario A, Adams JB, Domenici E, Dalla Bernardina B, Bonassi S. Oxidative stress-related biomarkers in autism: systematic review and meta-analyses. Free Radic Biol Med 2012; 52 (10):2128-2141. 51. James SJ, Melnyk S, Jernigan S, Cleves MA, Halsted CH, Wong DH, Cutler P, Bock K, Boris M, Bradstreet JJ, Baker SM, Gaylor DW. Metabolic endophenotype and related genotypes are associated with oxidative stress in children with autism. Am J Med Genet B Neuropsychiatr Genet 2006; 141B (8):947-956. 52. Clarke M, Ward M, Strain JJ, Hoey L, Dickey W, McNulty H. B-vitamins and bone in health and disease: the current evidence. Proc Nutr Soc 2014; 73 (2):330-339. 53. Reilly R, McNulty H, Pentieva K, Strain JJ, Ward M. MTHFR 677TT genotype and disease risk: is there a modulating role for B-vitamins? Proc Nutr Soc 2014; 73 (1):47-56. 54. Christensen KE, Mikael LG, Leung KY, Levesque N, Deng L, Wu Q, Malysheva OV, Best A, Caudill MA, Greene ND, Rozen R. High folic acid consumption leads to pseudo- MTHFR deficiency, altered lipid metabolism, and liver injury in mice. Am J Clin Nutr 2015; 101 (3):646-658. 55. Scaglione F, Panzavolta G. Folate, folic acid and 5-methyltetrahydrofolate are not the same thing. Xenobiotica 2014; 44 (5):480-488. 56. Shorter KR, Felder MR, Vrana PB. Consequences of dietary methyl donor supplements: Is more always better? Prog Biophys Mol Biol 2015. 57. Navarro SL, Schwarz Y, Song X, Wang CY, Chen C, Trudo SP, Kristal AR, Kratz M, Eaton DL, Lampe JW. Cruciferous vegetables have variable effects on biomarkers of systemic inflammation in a randomized controlled trial in healthy young adults. J Nutr 2014; 144 (11):1850-1857. 58. Cummings AM, Kavlock RJ. Gene-environment interactions: a review of effects on reproduction and development. Crit Rev Toxicol 2004; 34 (6):461-485. 59. Ding H, Liu W, Yu X, Wang L, Shao L, Yi W. Risk association of meningiomas with MTHFR C677T and GSTs polymorphisms: a meta-analysis. Int J Clin Exp Med 2014; 7 (11):3904-3914. 60. Lordelo GS, Miranda-Vilela AL, Akimoto AK, Alves PC, Hiragi CO, Nonino A, Daldegan MB, Klautau-Guimaraes MN, Grisolia CK. Association between methylene tetrahydrofolate reductase and glutathione S-transferase M1 gene polymorphisms and chronic myeloid leukemia in a Brazilian population. Genet Mol Res 2012; 11 (2):1013- 1026. 61. Grant SF, Thorleifsson G, Reynisdottir I, Benediktsson R, Manolescu A, Sainz J, Helgason A, Stefansson H, Emilsson V, Helgadottir A, Styrkarsdottir U, Magnusson KP, Walters GB, Palsdottir E, Jonsdottir T, Gudmundsdottir T, Gylfason A, Saemundsdottir J, Wilensky RL, Reilly MP, Rader DJ, Bagger Y, Christiansen C, Gudnason V, Sigurdsson G, Thorsteinsdottir U, Gulcher JR, Kong A, Stefansson K. Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 2006; 38 (3):320-323. 62. Meigs JB, Shrader P, Sullivan LM, McAteer JB, Fox CS, Dupuis J, Manning AK, Florez JC, Wilson PW, D'Agostino RB, Sr., Cupples LA. Genotype score in addition to common risk factors for prediction of type 2 diabetes. N Engl J Med 2008; 359 (21):2208-2219. 63. Tong Y, Lin Y, Zhang Y, Yang J, Liu H, Zhang B. Association between TCF7L2 gene polymorphisms and susceptibility to type 2 diabetes mellitus: a large Human Genome Epidemiology (HuGE) review and meta-analysis. BMC Med Genet 2009; 10:15. 64. Migliorini A, Lickert H. Beyond association: A functional role for Tcf7l2 in beta-cell development. Mol Metab 2015; 4 (5):365-366.
  • 12. 65. Shu L, Matveyenko AV, Kerr-Conte J, Cho JH, McIntosh CH, Maedler K. Decreased TCF7L2 protein levels in type 2 diabetes mellitus correlate with downregulation of GIP- and GLP-1 receptors and impaired beta-cell function. Hum Mol Genet 2015; 24 (10):3004. 66. Yao DD, Yang L, Wang Y, Liu C, Wei YJ, Jia XB, Yin W, Shu L. Geniposide promotes beta-cell regeneration and survival through regulating beta-catenin/TCF7L2 pathway. Cell Death Dis 2015; 6:e1746. 67. Pagan A, Sabater-Molina M, Olza J, Prieto-Sanchez MT, Blanco-Carnero JE, Parrilla JJ, Gil A, Larque E. A gene variant in the transcription factor 7-like 2 (TCF7L2) is associated with an increased risk of gestational diabetes mellitus. Eur J Obstet Gynecol Reprod Biol 2014; 180:77-82. 68. Reyes-Lopez R, Perez-Luque E, Malacara JM. Metabolic, hormonal characteristics and genetic variants of TCF7L2 associated with development of gestational diabetes mellitus in Mexican women. Diabetes Metab Res Rev 2014; 30 (8):701-706. 69. Shi X, Cai Q, Zou M, Shen Y. [Correlation between TCF7L2 gene polymorphism and genetic susceptibility in women with gestational diabetes mellitus]. Zhonghua Fu Chan Ke Za Zhi 2014; 49 (8):588-593. 70. Zhang C, Bao W, Rong Y, Yang H, Bowers K, Yeung E, Kiely M. Genetic variants and the risk of gestational diabetes mellitus: a systematic review. Hum Reprod Update 2013; 19 (4):376-390. 71. Heni M, Herzberg-Schafer S, Machicao F, Haring HU, Fritsche A. Dietary fiber intake modulates the association between variants in TCF7L2 and weight loss during a lifestyle intervention. Diabetes Care 2012; 35 (3):e24. 72. Hindy G, Sonestedt E, Ericson U, Jing XJ, Zhou Y, Hansson O, Renstrom E, Wirfalt E, Orho-Melander M. Role of TCF7L2 risk variant and dietary fibre intake on incident type 2 diabetes. Diabetologia 2012; 55 (10):2646-2654. 73. Qi L, Liang J. Interactions between genetic factors that predict diabetes and dietary factors that ultimately impact on risk of diabetes. Curr Opin Lipidol 2010; 21 (1):31-37. 74. Wirstrom T, Hilding A, Gu HF, Ostenson CG, Bjorklund A. Consumption of whole grain reduces risk of deteriorating glucose tolerance, including progression to prediabetes. Am J Clin Nutr 2012; 97 (1):179-187. 75. Chen ZL, Shao WJ, Xu F, Liu L, Lin BS, Wei XH, Song ZL, Lu HG, Fantus IG, Weng JP, Jin TR. Acute Wnt pathway activation positively regulates leptin gene expression in mature adipocytes. Cell Signal 2015; 27 (3):587-597. 76. Helgason A, Palsson S, Thorleifsson G, Grant SF, Emilsson V, Gunnarsdottir S, Adeyemo A, Chen Y, Chen G, Reynisdottir I, Benediktsson R, Hinney A, Hansen T, Andersen G, Borch-Johnsen K, Jorgensen T, Schafer H, Faruque M, Doumatey A, Zhou J, Wilensky RL, Reilly MP, Rader DJ, Bagger Y, Christiansen C, Sigurdsson G, Hebebrand J, Pedersen O, Thorsteinsdottir U, Gulcher JR, Kong A, Rotimi C, Stefansson K. Refining the impact of TCF7L2 gene variants on type 2 diabetes and adaptive evolution. Nat Genet 2007; 39 (2):218-225. 77. Lee YS, Jun HS. Anti-diabetic actions of glucagon-like peptide-1 on pancreatic beta- cells. Metabolism 2014; 63 (1):9-19. 78. Xiong X, Shao W, Jin T. New insight into the mechanisms underlying the function of the incretin hormone glucagon-like peptide-1 in pancreatic beta-cells: the involvement of the Wnt signaling pathway effector beta-catenin. Islets 2012; 4 (6):359-365.
  • 13. 79. Mattei J, Qi Q, Hu FB, Sacks FM, Qi L. TCF7L2 genetic variants modulate the effect of dietary fat intake on changes in body composition during a weight-loss intervention. Am J Clin Nutr 2012; 96 (5):1129-1136. 80. Fereidooni B, Jenabi E. The use of omega 3 on pregnancy outcomes: a single-center study. J Pak Med Assoc 2014; 64 (12):1363-1365. 81. Dodd JM, O'Brien C, Grivell RM. Preventing pre-eclampsia - are dietary factors the key? BMC Med 2014; 12:176. 82. Endeshaw M, Abebe F, Bedimo M, Asart A. Diet and Pre-eclampsia: A Prospective Multicentre Case-Control Study in Ethiopia. Midwifery 2015; 31 (6):617-624. 83. Corella D, Tai ES, Sorli JV, Chew SK, Coltell O, Sotos-Prieto M, Garcia-Rios A, Estruch R, Ordovas JM. Association between the APOA2 promoter polymorphism and body weight in Mediterranean and Asian populations: replication of a gene-saturated fat interaction. Int J Obes (Lond) 2011; 35 (5):666-675. 84. Martinez-Gonzalez MA, Salas-Salvado J, Estruch R, Corella DD, Fito M, Ros E. Benefits of the Mediterranean Diet: Insights from the PREDIMED Study. Prog Cardiovasc Dis 2015. 85. Smith CE, Tucker KL, Arnett DK, Noel SE, Corella D, Borecki IB, Feitosa MF, Aslibekyan S, Parnell LD, Lai CQ, Lee YC, Ordovas JM. Apolipoprotein A2 polymorphism interacts with intakes of dairy foods to influence body weight in 2 U.S. populations. J Nutr 2013; 143 (12):1865-1871. 86. Zaki ME, Amr KS, Abdel-Hamid M. APOA2 Polymorphism in Relation to Obesity and Lipid Metabolism. Cholesterol 2011; 2013:289481. 87. Zaki ME, Amr KS, Abdel-Hamid M. Evaluating the association of APOA2 polymorphism with insulin resistance in adolescents. Meta Gene 2014; 2:366-373. 88. Peloso GM, Demissie S, Collins D, Mirel DB, Gabriel SB, Cupples LA, Robins SJ, Schaefer EJ, Brousseau ME. Common genetic variation in multiple metabolic pathways influences susceptibility to low HDL-cholesterol and coronary heart disease. J Lipid Res 2010; 51 (12):3524-3532. 89. Koning F. The molecular basis of celiac disease. J Mol Recognit 2003; 16 (5):333-336. 90. Meresse B, Ripoche J, Heyman M, Cerf-Bensussan N. Celiac disease: from oral tolerance to intestinal inflammation, autoimmunity and lymphomagenesis. Mucosal Immunol 2009; 2 (1):8-23. 91. Schuppan D, Zimmer KP. The diagnosis and treatment of celiac disease. Dtsch Arztebl Int 2013; 110 (49):835-846. 92. Brottveit M, Beitnes AC, Tollefsen S, Bratlie JE, Jahnsen FL, Johansen FE, Sollid LM, Lundin KE. Mucosal cytokine response after short-term gluten challenge in celiac disease and non-celiac gluten sensitivity. Am J Gastroenterol 2013; 108 (5):842-850. 93. Kabbani TA, Vanga RR, Leffler DA, Villafuerte-Galvez J, Pallav K, Hansen J, Mukherjee R, Dennis M, Kelly CP. Celiac disease or non-celiac gluten sensitivity? An approach to clinical differential diagnosis. Am J Gastroenterol 2014; 109 (5):741-746; quiz 747. 94. Fasano A, Sapone A, Zevallos V, Schuppan D. Nonceliac gluten sensitivity. Gastroenterology 2014; 148 (6):1195-1204. 95. Shepherd SJ, Halmos E, Glance S. The role of FODMAPs in irritable bowel syndrome. Curr Opin Clin Nutr Metab Care 2014; 17 (6):605-609.
  • 14. 96. Ashat M, Kochhar R. Non-celiac gluten hypersensitivity. Trop Gastroenterol 2014; 35 (2):71-78. 97. Mansueto P, Seidita A, D'Alcamo A, Carroccio A. Non-celiac gluten sensitivity: literature review. J Am Coll Nutr 2014; 33 (1):39-54. 98. Nijeboer P, Bontkes HJ, Mulder CJ, Bouma G. Non-celiac gluten sensitivity. Is it in the gluten or the grain? J Gastrointestin Liver Dis 2013; 22 (4):435-440. 99. Meresse B, Malamut G, Cerf-Bensussan N. Celiac disease: an immunological jigsaw. Immunity 2012; 36 (6):907-919. 100. Uibo R, Tian Z, Gershwin ME. Celiac disease: a model disease for gene-environment interaction. Cell Mol Immunol 2011; 8 (2):93-95.