Nutritional genomics: the next frontier in the postgenomic era
Jim Kaput and
Raymond L. Rodriguez
Laboratory for High Performance Computing and Informatics, Section of Molecular and Cellular Biology, University of California at Davis, Davis, California 95616
 |
ABSTRACT
|
---|
The interface between the nutritional environment and cellular/genetic processes is being referred to as "nutrigenomics." Nutrigenomics seeks to provide a molecular genetic understanding for how common dietary chemicals (i.e., nutrition) affect health by altering the expression and/or structure of an individuals genetic makeup. The fundamental concepts of the field are that the progression from a healthy phenotype to a chronic disease phenotype must occur by changes in gene expression or by differences in activities of proteins and enzymes and that dietary chemicals directly or indirectly regulate the expression of genomic information. We present a conceptual basis and specific examples for this new branch of genomic research that focuses on the tenets of nutritional genomics: 1) common dietary chemicals act on the human genome, either directly or indirectly, to alter gene expression or structure; 2) under certain circumstances and in some individuals, diet can be a serious risk factor for a number of diseases; 3) some diet-regulated genes (and their normal, common variants) are likely to play a role in the onset, incidence, progression, and/or severity of chronic diseases; 4) the degree to which diet influences the balance between healthy and disease states may depend on an individuals genetic makeup; and 5) 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.
nutrition; diet; diet-regulated genes; gene expression
 |
INTRODUCTION
|
---|
PROGRESS IN THE BATTLE against human disease and suffering is being accelerated by the availability of genomic information for humans, mice, and other organisms. The techniques and knowledge emerging from these genome projects have revolutionized the process of localizing and identifying genes involved in disease. To date, almost 1,000 human disease genes have been identified and partially characterized, 97% of which are now known to cause monogenic diseases (75). However, most cases of obesity, cardiovascular disease (CVD), diabetes, cancer, and other chronic diseases are due to complex interactions between several genes and environmental factors. It is not surprising, therefore, that the strategies for characterizing and identifying monogenic diseases have been unsuccessful when applied to chronic diseases. Despite the more than 600 association studies published as of 2002 (reviewed in Ref. 65), the molecular basis of chronic diseases remains elusive. Such results led to the development of the "common disease/common variant hypothesis" (i.e., CDCV hypothesis; Refs. 24 and 91), which states that chronic diseases are caused by sets of gene variants that collectively contribute to disease initiation and development. The complexity of genetic interactions and the number and spacing of mapping markers explain why it has been difficult for molecular epidemiological studies to localize genes associated with chronic diseases. More complete single-nucleotide polymorphism (SNP) and haplotype maps (31, 58, 76, 77, 173) will create additional resources for identifying genes involved in diseases. These genome-centric approaches, however, usually fail to take into account the most important variable in expression of genetic information and a major contributor to disease development, namely, dietary chemicals.
The interface between the nutritional environment and cellular/genetic processes is being referred to as nutritional genomics or "nutrigenomics." Nutrigenomics seeks to provide a genetic understanding for how common dietary chemicals (i.e., nutrition) affects the balance between health and disease by altering the expression and/or structure of an individuals genetic makeup. The conceptual basis for this new branch of genomic research can best be summarized with the following five tenets.
1) Common dietary chemicals act on the human genome, either directly or indirectly, to alter gene expression or structure.
2) Under certain circumstances and in some individuals, diet can be a serious risk factor for a number of diseases.
3) Some diet-regulated genes (and their normal, common variants) are likely to play a role in the onset, incidence, progression, and/or severity of chronic diseases.
4) The degree to which diet influences the balance between healthy and disease states may depend on an individuals genetic makeup.
5) 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.
Common Dietary Chemicals Can Alter Gene Expression Or Structure
Epidemiological studies repeatedly show associations between food intake and the incidence and severity of chronic diseases (reviewed in Refs. 74 and 165), but the concept that food contains bioactive chemicals is not apparent from the design of many molecular and genetic association studies or laboratory animal or cell culture experiments. As an example of the complexity of a "simple" food, the constituents of corn oil are shown in Table 1. The variety and concentrations of fatty acids, triglycerides, sterols, sterol esters, and tocopherols are likely to have many and diverse effects on physiology since dietary chemicals have several fates upon entering a cell.
Dietary chemicals can affect gene expression directly or indirectly. At the cellular level, nutrients may: 1) act as ligands for transcription factor receptors (32, 70); 2) be metabolized by primary or secondary metabolic pathways, thereby altering concentrations of substrates or intermediates; or 3) positively or negatively affect signal pathways (22, 42). This is shown schematically in Fig. 1. Fatty acids, for example, are metabolized via the ß-oxidation pathways to produce cellular energy (Fig. 1B). Altering intracellular energy balance may indirectly alter gene expression through changes in cellular NAD homeostasis (reviewed in Ref. 97). NAD reoxidation is associated with mitochondrial electron transport activity and is a cofactor for proteins involved in chromatin remodeling (59, 104). Chromatin remodeling processes have short- and long-term consequences for gene regulation due to reactions such as histone acetylation or DNA methylation that alter access to, and therefore regulation of, eukaryotic genes (reviewed in Ref. 43).

View larger version (21K):
[in this window]
[in a new window]
|
Fig. 1. Fate and activities of nutrients in the cell. Nutrients may act directly as ligands for transcription factor receptors (pathway A); may be metabolized by primary or secondary metabolic pathways, thereby altering concentrations of substrates or intermediates (pathway B) involved in gene regulation or cell signaling; or alter signal transduction pathways and signaling (pathway C). See text for details.
|
|
Some dietary chemicals also are ligands for nuclear receptors (Fig. 1A). Many, but not all genes involved in fatty acid metabolism are regulated by one of the three members of the peroxisome proliferator-activated receptor family (PPAR
, PPAR
, PPAR
) family (reviewed in Ref. 9). The surprising finding (at the time) was that the fatty acids, palmitic (16:0), oleic (18:1 n9), linoleic (18:2 n6), and arachidonic (20:4 n6) acid (41, 63, 133), and the eicosanoids, 15-deoxy-
12,14prostaglandin J2 and 8-(S)hydroxyeicosatraenoic acid (54, 80), are ligands for PPARs (reviewed in Ref. 81). That is, these nuclear receptors act as sensors for fatty acids. Lipid sensors usually heterodimerize with retinoid X receptor (RXR), whose ligand is derived from another dietary chemical, retinol (vitamin A) (32). Some dietary chemicals, such as genistein, vitamin A, and hyperforin, bind directly to nuclear receptors and influence gene expression (Table 2). Other transcription factors are indirectly regulated by dietary chemicals. The sterol regulatory element binding proteins (SREBPs) are activated by protease cleavage, an event regulated by low levels of oxysterols and changes in insulin/glucose and polyunsaturated fatty acids (PUFA; reviewed in Ref. 44). The carbohydrate-responsive element-binding protein (ChREBP) is activated in response to high glucose levels and is regulated by reversible phosphorylation events (reviewed in Ref. 152).
Metabolic conversion of dietary chemicals also serves as a control mechanism for gene expression (109). The level of steroid hormones, which are ultimately derived from cholesterol, is regulated by the activities of the combined 10 steps in the steroid biosynthetic pathway. In addition, various intermediates branch into other metabolic pathways. Degradative pathways will also influence the overall intracellular concentrations of intermediates and end products (Fig. 1B, and Metabolism, below). Hence, the concentration of any given ligand (109) will be greatly influenced by specific combinations of alleles for the enzymatic steps in these assorted pathways. That a specific pair of alleles may be heterozygous and vary in frequency from one subpopulation to another is a fundamental precept of nutrigenomics.
Dietary chemicals also can directly affect signal transduction pathways (Fig. 1C). Green tea contains the polyphenol, 11-epigallocatechin-3-gallate (EGCG). EGCG inhibits tyrosine phosphorylation of Her-2/neu receptor and epidermal growth factor receptor, which, in turn, reduces signaling via the phosphatidylinositol 3-kinase (PI-3)
Akt kinase
NF-
B pathway (101, 119). Activation of the NF-
B pathway is associated with some virulent forms of breast cancer. Platelet-derived growth factor receptor phosphorylation is also inhibited by EGCG and derivatives (129). Grains such as rice contain inositol hexaphosphate (InsP6), which inhibits TPA- or EGF-induced cell transformation through its effects on PI-3 kinase (37). Resveratrol, phenethyl isothiocyanate (PEITC), genistein, and retinoids (vitamin A and metabolites) also affect signal transduction pathways (reviewed in Ref. 38).
The fact that dietary chemicals play such key roles in regulating gene expression beyond their well-known roles of producing energy and affecting insulin levels is consistent with evolutionary theory. Given that the human genome is so exquisitely responsive to its nutritional environment, it is reasonable to conclude that many human genes evolved in response to the plant- and animal-derived dietary chemicals we consume.
Diet Can Be a Risk Factor for Disease
The idea that adverse diet/genome interactions can cause disease is not new. The first example was the discovery of galactosemia by F. Goppart in 1917 (http://www.ncbi.nlm.nih.gov/htbin-post/Omim/dispmim?230400). Galactosemia is a rare recessive defect in galactose-1-phosphate uridyltransferase (GALT). The lack of GALT results in the accumulation of galactose in the blood, causing a number of health problems including mental retardation. Phenylketonuria (PKU), another recessive trait, was discovered in 1934 by Asbjørn Følling (http://www.ncbi.nlm.nih.gov/htbin-post/Omim/dispmim?261600). PKU is a defect in the enzyme phenylalanine hydroxylase that results in an accumulation of phenylalanine in the blood. The accumulation of high levels of phenylalanine can cause neurological damage. Both PKU and galactosemia can be screened in infants shortly after birth, and these diseases can be managed with diets low in phenylalanine and lactose, respectively.
PKU and galactosemia are single gene traits and thus easy to identify and treat by changes in diet. Many chronic diseases are polygenic in nature and result from interactions of a subset of genes with environmental factors. The association between specific food intake and chronic disease was first recorded in 1908 and was based upon observations of Ignatovski that rabbits fed meat, milk, and eggs developed arterial lesions resembling atherosclerosis in humans (reviewed in Ref. 26). The associations of cholesterol with hypercholesterolemia and hypercholesterolemia with atherosclerosis focused much attention on the link between amount of calories (reviewed in Ref. 165) and/or the levels and types of vitamins (50), fat (e.g., 84; and reviewed in Refs. 84 and 167), and carbohydrates (reviewed in Ref. 73) with atherosclerosis, diabetes, obesity, cancer, and other chronic diseases. Although some associations have been confirmed by subsequent genetic or biochemical studies in laboratory animals or humans, others remain controversial. The much-debated association between level and type of dietary fat with breast cancer incidence (reviewed in Refs. 139 and 166) illustrates the difficulty of epidemiological studies to prove causation.
The limitations of epidemiological studies may be in design, dietary assessment tools, measurement errors, statistical methods, sample size, and nutritionally insignificant differences in fat intake among study populations (96). In addition, although epidemiological methods often include family histories, individual genotypes are not usually analyzed. Nevertheless, the underlying assumption of nonmolecular epidemiology studies is that individual genetic variation is insignificant and that each individual responds similarly to their environment. As demonstrated by the human genome (92, 156) and SNP projects (58, 93, 128, 134), there are potentially millions of base pair differences between individuals, and some of these differences could affect the way one individual responds to their nutritional environment relative to another individual. Assumptions, therefore, that fail to take into account these genetic differences are unlikely to reveal meaningful connections between specific nutrients and chronic diseases. With the advent of genomic sequence information and high-throughput technologies and reagents, analyzing SNPs or other polymorphisms in multiple genes is now possible. Analyzing patterns of SNPs with patterns of disease subphenotypes will require sophisticated statistical tools and large populations or many family studies.
A related confounding factor is the differences in allele frequencies among human subpopulations. Following the migrations from Africa, humans became geographically isolated, limiting genetic exchanges and fixing distinct alleles and haplotypes (148). As one example, the gene encoding arylamine N-acetyltransferase, NAT2 (64, 125), is polymorphic. Variants encode a fast acetylator allele and several subtypes of slow acetylators. These allele types are represented differently among populations in different geographic regions: slow allele subtypes are found in 72% of the Caucasians in the United States but only in 31% of Japanese. Individuals with slow acetylator NAT2 allele are more susceptible to bladder cancer when exposed to procarcinogens (125). The allele frequencies of NAT2 illustrate the importance of knowing allele distributions in populations and exposure to environmental influences, since molecular epidemiology studies rely upon statistical associations of certain haplotypes or SNPs (alleles) with disease phenotypes, incidence, and/or severity. False-positive associations of SNPs (or haplotype) and disease may be found because of the distribution of alleles within the population studied rather than a true association between an allele and a phenotype or disease. Determining allele frequencies requires resequencing genes in ethnically different individuals (greater than or equal to
90), a costly enterprise that will ultimately be done for all genes. In the meantime, ethnic difference markers (25, 103, 114, 135) can determine the origin of chromosomal regions, which may encode ethnic-specific alleles and may be used to assess population substructures in epidemiological studies. Although population substructures confound statistical association studies, analyzing diverse admixtures may allow for the identification of genes contributing to certain specific chronic diseases (25, 103, 114, 135, 148), since certain ethnic populations are at increased risk for chronic diseases.
Analyzing genotype as a variable in association studies of disease phenotypes or subphenotypes will eliminate confounding due to population heterogeneity (e.g., 96, 116, 117, 134, 167). Monitoring or measuring dietary intakes must also be done, since epidemiological and laboratory animal studies have consistently demonstrated an effect of diet on disease initiation and progression. The literature linking diet to disease is too voluminous to review herein but certain recent advances are summarized below.
Micronutrients.
Approximately 40 micronutrients are required in the human diet. Suboptimal intakes of specific micronutrients have been associated with CVD (B vitamins, vitamin E, carotenoids), cancer (folate, carotenoids), neural tube defects (folate), and bone mass (vitamin D) (50). B6, B12, and folate deficiencies, for example, are associated with increased serum homocysteine levels. Hyperhomocysteinemia is a risk factor and marker for coronary artery disease, but the mechanism(s) is not understood at the molecular level (51) although several theories have been proposed to explain its action (e.g., 132). Many of these conclusions are based upon cohort, randomized trials, and meta analyses wherein the cause of the disease cannot be conclusively determined.
Deficiency of vitamins B12, folic acid, B6, niacin, C, or E, or iron or zinc appears to mimic radiation in damaging DNA by causing single- and double-strand breaks, oxidative lesions, or both (5) (Table 3). Nutrient deficiencies are orders of magnitude more important than radiation because of constancy of exposure to milieu promoting DNA damage (4, 5, 7). Folate deficiency breaks chromosomes due to substantial incorporation of uracil in human DNA (4 million uracil/cell) (14). Single-strand breaks in DNA are subsequently formed during base excision repair, with two nearby single-strand breaks on opposite DNA strands leading to chromosome fragmentation. Micronutrient deficiency may explain why the quarter of the US population that consume less than the recommended five portions a day of vegetables and fruits has approximately twice the rate for most types of cancer compared with the quarter with the highest intake (5). A number of other degenerative diseases of aging are also associated with low fruit and vegetable intake. Progress is also being made in determining specific mechanisms for the role of certain minerals (calcium, magnesium, manganese, copper, and selenium) and vitamins in heart disease from work done in humans and in cell culture systems (reviewed in Ref. 168).
Macronutrients: Fats.
Unbalanced intake of any of the three major macronutrients, fat, carbohydrates, or protein, contributes to the initiation, development, progression, and/or severity of chronic diseases. Intake of saturated fatty acids (SFA) is correlated with increased levels of low-density lipoprotein (LDL) cholesterol, the principal target of intervention for coronary disease risk reduction (86). In addition to CVDs, SFA may contribute to obesity and diabetes (87) because these diseases are also characterized by dyslipidemias (35, 39, 71, 140, 153, 172). Large numbers of human and laboratory animal experiments support the associations predicted from epidemiological studies.
As discussed above, human studies showing associations between amount and type of fat and prostate (10, 120, 154), colorectal (reviewed in Ref. 60), and breast (26, 96) cancers are inconsistent (139, 165). Laboratory animal studies in which genotype and environment can be more rigorously controlled consistently show that the type and level of dietary fat are associated with incidence and strongly associated with promotion (reviewed in Refs. 26 and 96) of certain cancers. Molecular studies that rely upon candidate genes identified from diet- and genotype-controlled laboratory animal studies may provide better candidate genes for human molecular epidemiology studies examining the role of dietary fats in human cancers.
Macronutrients: Carbohydrates.
Dietary guidelines continue to emphasize diets low in saturated and total fat for reducing in the risk of obesity and its related comorbidities: diabetes and CVD (86). Spurred by the increase in obesity and diabetes while fat intake barely decreased (36.4 to 34.1% of total calories) between 1970 and 1990 (48), a new focus of a number of epidemiological studies is carbohydrates. Simple and complex carbohydrates are metabolized at different rates and therefore have differential effects on blood glucose concentrations. The glycemic index (GI) is a quantitative measure of foods based upon postprandial blood glucose response (72). GI is expressed as a percentage of the response to an equivalent carbohydrate portion of white bread or glucose (169). Glycemic load (GL), the product of the average dietary GI and total carbohydrate intake, is a measure of total insulin demand (131). For example, if glucose has a GI of 100, potatoes have a GI of 87 and tomatoes, 9. Refined simple sugars or some polysaccharides that are cleaved rapidly to glucose produce higher blood glucose levels and a greater demand for insulin (8). High GI would increase insulin production and, at the same time, decrease synthesis of insulin receptors.
All but one cohort study of type 2 diabetes and GI (8) showed an association between GI and type 2 diabetes or coronary artery disease and colon or breast cancer and GI in case control studies. These associations were observed after multiple adjustments (e.g., for fiber or other dietary variables) and usually for the 5th quintile of GI (8). Molecular epidemiological analyses of the associations of candidate genes and nutritional variables are needed to determine the biochemical effects of GI on physiology.
Macronutrients: Protein.
Analyzing the effect of protein intake on health is difficult because fat and micronutrients are significant and variable components of meat. Broiling, frying, and baking differentially alters the chemical composition of meat and other foods (1) and in some cases creates nitrosamines and other carcinogens (e.g., 130). Different ways of preparing foods may produce different amounts and types of natural or heat-generated dietary chemicals, thereby introducing confounding into epidemiological analyses. With these caveats, meat consumption appears to be associated with increased chronic disease risk (reviewed in Refs. 3 and 82) including bowel (e.g., 12) and colorectal (e.g., 57) cancers and type 2 diabetes (e.g., 153). Molecular epidemiology suggests that certain genes, for example, epoxide hydrolase (151), glutathione-S-transferase (28), and other detoxifying enzymes (130), may modify the effect of meat on disease risk.
Increased metabolism of protein also will increase the production of urea, with the corresponding increase in membrane-permeable ammonia (NH3) and its ionized form NH4+. Ammonia released in the alimentary tract of animals by microbial enzymes can disrupt metabolic pathways (158), alter the gastrointestinal mucosa (53), inhibit rates of growth in animals (e.g., 160), alter brain function (52), and promote cancer (e.g., 23).
Caloric restriction.
Early epidemiological studies neglected to account for the differences in energy content between carbohydrates and proteins (each at
4 kcal/g) and lipids (
9 kcal/g). Virtually all association studies show an increased risk for common diseases with increased energy intake (78). Laboratory animal studies have consistently shown that reducing caloric intake is the most effective means to reduce the incidence and severity of chronic diseases, retard the effects of aging, and increase genetic fidelity (reviewed in Refs. 149 and 163). Experiments in Saccharomyces cerevisiae suggest that caloric restriction may produce its largest effects by increasing respiration with the concomitant increase in the NAD:NADH (reviewed in Ref. 97). Energy balance may be monitored through changes in reducing equivalents. NAD also is a cofactor for Sir2, a histone deacetylase involved in chromatin silencing of nucleolar rDNA, telomere, and mating type locus (59, 104). In mammals, other cellular targets, such as uncoupling proteins, and neuroendocrine peptides (e.g., leptin) of the central nervous system (CNS), are potential targets of regulation by caloric restriction.
Summary.
The hunt for a single macronutrient or micronutrient that will prevent chronic diseases is destined to fail. It is more likely that dietary imbalances, from micronutrient deficiencies to overconsumption of macronutrients or dietary supplements, are the modifiers of metabolism and potentiators of chronic disease. Although the complexity of food and genotypic variations appears daunting, molecular and genetic technologies may provide the means for identifying causative genes (or their variants) and the nutrients that regulate them.
Some Diet-Regulated Genes Can Play a Role in Chronic Diseases
The progression from a healthy phenotype to a chronic disease phenotype must occur by changes in gene expression or by differences in activities of proteins and enzymes. Since dietary chemicals are regularly ingested and participate indirectly and directly in regulating gene expression, it follows that a subset of genes regulated by diet must be involved in disease initiation, progression, and severity (79, 113). The clearest example of genotype-diet interactions in chronic disease is type 2 diabetes, a condition that frequently occurs in sedentary, obese individuals and certain minority groups (13, 15). Once diagnosed with type 2 diabetes, some individuals can control symptoms by increasing physical activities and by reducing caloric (and specific fat) intake (108), i.e., expression of genomic information is changed by changing environmental (i.e., dietary) variables. Other individuals are refractory to such environmental interventions and require drug treatments. Many chronic diseases do not show the phenotypic plasticity seen in some type 2 diabetics; that is, symptoms are not reversible after some initiating event. Chromatin remodeling and changes in DNA methylation induced by unbalanced diets are possible mechanisms that contribute to irreversible gene expression changes. Nevertheless, genotype x diet interactions contribute to the incidence and severity of obesity, atherosclerosis, many cancers, asthma, and other chronic conditions (106, 124, 145, 167).
Molecular approach.
One approach to understanding the molecular mechanisms whereby diet alters health is to identify diet-regulated genes that cause or contribute to disease process. This can be done by examining the expression of a candidate gene or groups of genes (e.g., 142) in response to diets, an approach pioneered by Goodridge and coworkers (105; reviewed in Ref. 62). Many laboratories characterize the expression of candidate genes in a variety of tissues in laboratory animals in response to dietary variables (49, 61; reviewed in Refs. 21, 29, 33) and caloric restriction (17, 94, 95, 121). DNA and oligo-array technologies have extended this approach to multiple genes within a pathway (36) or all genes on an array (17, 94, 162; reviewed in Refs. 66, 141, 145, 155). Changes in gene expression are then associated with phenotype and can be explained by genetic variants in nuclear receptors, cis-acting elements in promoters, or differences in metabolism that produce altered concentrations of transcriptional ligands.
The limitations of assessing regulation of individual or multiple genes by diet are 1) determining cause from effect for each gene; that is, what are the subset of causative genes for a given phenotype?; and 2) gene expression patterns in one strain (or genotype) may be unique to that genotype. The results from inbred mouse strains (Kaput J, Klein KG, Reyes EJ, Visek WJ, Kibbe WA, Jovanovic B, Cooney CA, and Wolff G, unpublished observations) would suggest that individual humans (170) may have unique patterns of gene expression depending upon their genotype and diet. Such individual qualitative and quantitative differences will complicate attempts to find patterns in gene expression results for dietary intake. Since diet recall is imprecise and controlling diets difficult in large population studies, identifying these complex interactions will be challenging.
A separate confounding influence on analyses of diet-induced changes in gene expression patterns is the health of the subject (laboratory animal or human). The presence of a disease can be considered an additional environmental influence that could affect gene expression patterns. For example, the presence of obesity unmasks additional type 2 diabetes loci in C57BL/6 and BTBR mice (143). Specifically, phenotypic expression of two interacting loci that affected fasting glucose and insulin levels was observed only in obese mice, and the alleles from the two parental strains (C57BL/6 and BTBR) had different effects on the diabetic subphenotypes. Hence, one would predict changes in gene expression based upon the presence or absence of disease processes and changes caused by dietary differences. Separating these variables will be an important component of future experimental designs for determining the effect of diet on susceptibility and disease progression.
Inbred strains of laboratory animals have proven useful for examining diet-disease interactions at the molecular level because 1) each individual member of a strain is genetically identical; 2) their environment can be rigorously controlled; 3) statistics can be applied to molecular, physiological, and genetic measurements; and 4) experiments can be repeated (e.g., 56, 98). The limitation of a unique genotype can be overcome by examining multiple strains of mice (159). One of our laboratories (79, 113) introduced a comparative method for laboratory animals that identifies genes regulated differently by dietary variables between two or more genotypes. The genotypes (or inbred strains) of mice are selected based upon their susceptibility to disease caused by diet. We found that certain genes were differentially regulated based upon genotype (in this case, Avy/A obese yellow vs. A/a agouti mice) and/or on caloric intake (100% vs. 70% calories) or by the interaction between diet and genotype (Kaput et al., unpublished observations). The criteria for identifying a candidate disease gene are 1) genes must be differentially regulated by diet and/or 2) differentially regulated by genotype and 3) must map to chromosomal regions [e.g., quantitative trait loci (QTL)] associated with the disease (79, 113). This approach identifies candidate genes in an unbiased manner, and additional testing in humans or animal models is necessary to validate these.
Genetic approach.
Strategies for identifying genes that cause chronic diseases in humans have been greatly influenced by the successes in identifying genes that cause monogenic diseases (75). The difficulty in identifying chronic disease genes (65) has been attributed to factors such as small sample size, poorly matched control groups, population stratification, and overinterpreting data (among others, see: 18, 90, 127, 147). These methods and approaches are being improved to eliminate such errors and to reliably identify genes involved in chronic diseases (25, 34, 103, 114, 118, 122).
However, noticeably missing from discussions of the limitations of these genetic mapping techniques and gene association studies are the effects of environmental variables such as diet. Tanksley and coworkers (115) observed the importance of environment on expression of phenotypic traits in F2 and F3 generation tomato plants grown in Davis or Gilroy, CA, compared with plants grown near Rehovot, Israel. Only 4 of a total of 29 QTL were found at all three sites, and 10 QTL were found in only two sites. QTL identify multiple regions within all chromosomes that collectively contribute to a complex phenotype (126). Since an individual QTL encode many genes, identifying the causative genes within the QTL is challenging (147). So far, the QTL mapping that accounts for differences in diet has not been done, largely because controlling for diet in large population association studies is often not possible.
The complexities of gene-environment interactions are compounded by the same factors that affect molecular (gene expression) analyses: epigenetic interactions between genes (the obesity example in mice, Ref. 143), in utero effects, diet-gene interactions, and the "environmental history"; that is, the life-long exposure to changing diets may alter expression of genetic information later in life (136). Maternal nutrition during pregnancy also has been linked to altered phenotypes in laboratory and farm animals (e.g., 27, 30, 69, 164), and such epigenetic phenomena will likely affect gene-disease association studies. Maternal exposure to different nutrients and an organisms exposure during its lifetime to changing diets is likely to also produce different health outcomes because the exposure to food and xenobiotics may act upon the genome to alter gene expression. An excellent discussion of the complexity of genotype x environmental history is presented by Sing and colleagues (136).
The Balance Between Health and Disease States May Depend on an Individuals Genetic Makeup
All humans are 99.9% identical at the gene sequence level. The 0.1% variations in sequence, however, produce the differences in phenotypes (hair and skin color, height, weight, etc.) and an individuals susceptibility to disease or health. Alterations in phenotype result from differences in gene expression or altered macromolecular activities.
1) A striking and simple example of how SNPs alter gene expression is a polymorphism that alters tolerance to dietary lactose (milk). Adult mammals are typically lactose intolerant. A mutation occurred
9,000 years ago in Northern Europeans that allowed expression of the lactase-phlorizin hydrolase gene (LCH locus) to continue into adulthood. Although there are 11 polymorphisms in this gene clustered into 4 (A, B, C, U) prevalent haplotypes (>0.05%), a C13910T SNP located 14 kb upstream of the LCH (47) is highly associated with lactase persistence (lactose tolerance). This polymorphism is thought to alter regulatory protein-DNA interactions controlling expression of the gene (67). The A haplotype conferring lactose tolerance has an 86% frequency in the Northern European population, but only 36% in Southern European populations. The persistence of this variant in populations may confer selective advantages that include improved nutrition, prevention of dehydration, and improved calcium absorption. Regulatory SNPs (rSNPs) in other promoters are likely to play a role in regulating gene expression (e.g., 11, 16, 89)
2) SNPs may also alter splicing. Two insulin receptor splice variants differ in the presence (type B) or absence (type A) of exon 11. The type A isoform is associated with hyperinsulinemia (68, 144). Over 30,000 alternative splice sites have been identified in a genome-wide analysis of humans (93).
3) A subset of coding SNPs (cSNPS) will alter biochemical activities of enzymes, proteins, and cellular processes. As one example, steroid 5
-reductase (designated SRD5A2), a key enzyme in androgen metabolism in the prostate, has 13 naturally occurring variants in the human population. Nine of these variants reduce SRD5A2 activity by 20% or more, and three increase activity by more than 15% (99). Since SRD5A produces dihydrotestosterone (DHT) and DHT regulates genes in the prostate, variants in steroid 5
-reductase may affect incidence or severity of prostate cancer (123). Although these studies focused on coding SNPs, polymorphisms in 5' or 3' regulatory regions or splice sites may also alter the level of expression of a gene or the variant produced (e.g., 88, 170).
Dietary Intervention Based on Individualized Nutrition
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. This assertion is obvious for nutritional deficiencies such as scurvy and beriberi or the potential harm from dietary phenylalanine for phenylketonurics. Less obvious are treatments for
50 genetic diseases in humans caused by variants in enzymes (6). As many as one-third of enzyme variants are due to increased Km for a coenzyme, resulting in a lower rate of reaction (6). The Michaelis-Menten constant, Km, is a measure of binding affinity of an enzyme for its ligand (substrate or coenzyme) and is defined as the concentration of ligand required to fill half of the ligand-binding sites. Ames et al. (6) proposed the "Km hypothesis" to describe the effects of polymorphisms on enzymatic activity. Intracellular concentrations of coenzyme may be increased by high doses of the corresponding vitamin, which would partially restore enzymatic activity and potentially ameliorate the phenotype. Changing substrate concentrations may be a general approach to circumvent decreased coenzyme binding or decreased enzymatic activities caused by a given cSNP. Some examples include the following (6).
1) Glucose-6-phosphate dehydrogenase A44G (DNA:C131GP) with NADP as cofactor. Defects in this gene are involved in favism and hemolytic anemia. Increased dietary intake of nicotinic acid or nicotinamide might increase NADPH coenzyme concentrations enough to alter the equilibrium of GPDH 
GPDH + NADPH.
2) NAD is a cofactor for aldehyde dehydrogenase (ALDH), an enzyme involved in alcohol intolerance and linked to Alzheimers and cancer. A cSNP causes E487K, which increases the Km 150-fold. This variant could not be treated with diet because the NAD substrate concentration could not be increased sufficiently to overcome the increased Km.
Ames and coworkers (46) have established a web site entitled "Km Mutants" (http://www.kmmutants.org/), which summarizes nutritional information for a large number of enzymes requiring coenzymes.
Directed dietary intervention for prevention or treatment of chronic diseases in an individual is inherently more challenging because multiple genes interacting with each other and with environmental variables contribute to disease etiology. Identifying genes that contribute the most to chronic disease initiation or progression and understanding their regulation by dietary variables is a likely first step in this process. A number of candidate gene-disease-diet association studies (reviewed in Refs. 100, 111, 157, 171) have shown the promise of this approach, as follows.
Hypertension.
The amount of circulating angiotensinogen (ANG) is associated with increased blood pressure. A SNP, designated AA, at nucleotide position -6 of the ANG gene is linked with the level of circulating ANG protein. Individuals with the AA genotype who eat the Dietary Approaches to Stop Hypertension (DASH) diet show reduced blood pressure, but the same diet was less effective in reducing blood pressure in individuals with a GG genotype. A large percentage (
60%) of African Americans have the AA variant, and the remainder are heterozygotic (AG) at this position (146).
Cardiovascular health.
Apo-A1 plays a central role in lipid metabolism and coronary heart disease. The G-to-A transition in the promoter of APOA1 gene is associated with increased HDL cholesterol concentrations. The A allele (or variant) was associated with decreased serum HDL levels (112). For example, women who eat more PUFA relative to saturated fats (SF) and monounsaturated fats (MUFA) have increased serum HDL levels. This type-of-fat effect is significant in men when alcohol consumption and tobacco smoking were considered in the analyses.
Individuals with small, dense LDL particles (phenotype B) have an increased risk of coronary artery disease relative to those individuals exhibiting large, less dense LDL particles (phenotype A) (reviewed in Ref. 86). In a classic crossover experiment, Krauss and coworkers (40) showed that the LDL patterns are influenced by low-fat diets. Thirty-eight men exhibiting phenotype A LDL were switched from a 32% fat diet to a diet containing 10% fat. Twelve of these 38 exhibited phenotype B LDL after 10 days on the low-fat diet (40), suggesting that for these 12, low-fat diets were not beneficial. Although not directly analyzed, these results suggest three distinct genotypes. Two genotypes produce either the A or B phenotype. A third genotype produces the A phenotype when these individuals eat a diet containing 32% fat, but a B phenotype when fed 10% fat, a result that can be explained by a genotype x environment interactions.
Cancer.
Methylenetetrahydrofolate reductase (MTHFR) is a key gene in one-carbon metabolism and, indirectly, in all methylation reactions. Several laboratories have noted that the C667T polymorphism (Ala to Val), which reduces enzymatic activity, is inversely associated with occurrence of colorectal cancer (e.g., 20, 138, 150) and acute lymphocyte leukemia (137). Low intake of folate, vitamin B12, vitamin B6, or methionine was associated with increased risk for cancer among those with the MTHFR TT genotype. MTHFR variants are also implicated in CVD (83).
The effects of dietary chemicals on these polymorphisms raise the possibility that candidate gene or SNP association studies may be more accurate if diets and nutritional status are included as variables in the analyses. Dietary histories are notoriously inaccurate, and nutritional status is difficult to assess. Nevertheless these environmental factors are likely to alter results of association studies.
Summary
Recent advances in the field of pharmacogenomics underscores the importance of genotype x environment interactions by showing how individual genetic variation in human populations can affect a drugs efficacy and the severity of undesirable side effects (102, 110). For this reason, pharmaceutical companies are incorporating genotyping as part of their clinical trails to predict drug safety, toxicity, and efficacy. By relating phenotype to genotype, drug companies are designing and developing better drugs with fewer adverse side effects. By identifying the nonresponding subpopulations, pharmacogenomics can also develop new drugs from compounds previously thought too toxic for human use.
The concept of "personalized" medicine is now being extended to the field of nutrition (85, 107, 161). It is now accepted that nutrients (i.e., macronutrients, micronutrients and antinutrients) alter molecular processes such as DNA structure, gene expression, and metabolism, and these in turn may alter disease initiation, development, or progression. Individual genetic variation can influence how nutrients are assimilated, metabolized, stored, and excreted by the body. The same tools and methods used in pharmacogenomics (SNP analysis, gene expression profiling, proteomics, metabolomics, and bioinformatics) are being used to examine an individuals response to his or her nutritional environment. In the near future, quick, low-cost, point-of-care tests will be available to assist patients and physicians to achieve, manage, and prolong health through dietary invention. The desired outcome of nutrigenomics is the use of personalized diets to delay the onset of disease and optimize and maintain human health.
The studies and examples cited here and by others (107) provide a conceptual basis for the emerging field of nutritional genomics. Building on this foundation will be challenging and will likely focus on the following broadly defined questions.
1) What are the quantitative nutritional requirements to produce optimal metabolism, particularly for the macronutrients?
2) How can we optimize nutrient intake for each individual, given the genetic diversity and complexity of common dietary chemicals?
3) How can we link dietary chemicals to subtle, long-term regulation of metabolism?
4) How can we assess the changing nutritional needs of an individual from birth through death, given the available molecular and genomic technologies?
5) How do we ensure that nutritional genomic information is used in a socially responsible manner, particularly as it relates to health disparities in subpopulations, such as ethnic racial minorities, the poor, and the uninsured?
 |
NOTE ADDED IN PROOF
|
---|
The following articles address issues related to nutritional genomics:
Orzechowski A, Ostaszewski P, Jank M, and Berwid SJ. Bioactive substances of plant origin in food: impact on genomics. Reprod Nutr Dev 42: 461477, 2002.
Swanson KS, Schook LB, and Fahey GC Jr. Nutritional genomics: implications for companion animals. J Nutr 133: 30333040, 2003.
 |
ACKNOWLEDGMENTS
|
---|
We thank George Wolff, Willard Visek, Steven Watkins, Ted Powers, and Su Ju Lin for helpful discussion and critical review of the manuscript. We also thank Willard Visek, for providing information and references for the discussion on the role of protein in chronic diseases, and Steven Watkins, who contributed the questions that end the article.
GRANTS
This work was supported by National Center for Minority Health and Health Disparities Center of Excellence in Nutritional Genomics Grant MD-00222.
DISCLOSURES
J. Kaput is the Chief Scientific Officer of NutraGenomics, Inc., a new biotechnology company that is investigating the science of nutritional genomics.
 |
FOOTNOTES
|
---|
Article published online before print. See web site for date of publication (http://physiolgenomics.physiology.org).
Address for reprint requests and other correspondence: J. Kaput, Laboratory for High Performance Computing and Informatics, Section of Molecular and Cellular Biology, Univ. of California at Davis, One Shields Ave., Davis, CA 95616 (E-mail: jkaput{at}ucdavis.edu).
10.1152/physiolgenomics.00107.2003.
 |
REFERENCES
|
---|
- Ames BN and Gold LS. The causes and prevention of cancer: gaining perspective. Environ Health Perspect 105, Suppl 4: 865873, 1997.
- Ames BN. Micronutrients prevent cancer and delay aging. Toxicol Lett 102103: 518, 1998.
- Ames BN and Gold LS. The causes and prevention of cancer: the role of environment. Biotherapy 11: 205220, 1998.[CrossRef][ISI][Medline]
- Ames BN and Gold LS. Paracelsus to parascience: the environmental cancer distraction. Mutat Res 447: 313, 2000.[ISI][Medline]
- Ames BN. DNA damage from micronutrient deficiencies is likely to be a major cause of cancer. Mutat Res 475: 720, 2001.[ISI][Medline]
- Ames BN, Elson-Schwab I, and Silver EA. High-dose vitamin therapy stimulates variant enzymes with decreased coenzyme binding affinity (increased Km): relevance to genetic disease and polymorphisms. Am J Clin Nutr 75: 616658, 2002.[Abstract/Free Full Text]
- Ames BN and Wakimoto P. Are vitamin and mineral deficiencies a major cancer risk? Nat Rev Cancer 2: 694704, 2002.[CrossRef][ISI][Medline]
- Augustin LS, Franceschi S, Jenkins DJ, Kendall CW, and La Vecchia C. Glycemic index in chronic disease: a review. Eur J Clin Nutr 56: 10491071, 2002.[CrossRef][ISI][Medline]
- Auwerx J. Regulation of gene expression by fatty acids and fibric acid derivatives: an integrative role for peroxisome proliferator activated receptors. The Belgian Endocrine Society Lecture 1992. Horm Res 38: 269277, 1992.[ISI][Medline]
- Bairati I, Meyer F, Fradet Y, and Moore L. Dietary fat and advanced prostate cancer. J Urol 159: 12711275, 1998.[CrossRef][ISI][Medline]
- Benbow U, Tower GB, Wyatt CA, Buttice G, and Brinckerhoff CE. High levels of MMP-1 expression in the absence of the 2G single nucleotide polymorphism is mediated by p38 and ERK1/2 mitogen-activated protein kinases in VMM5 melanoma cells. J Cell Biochem 86: 307319, 2002.[CrossRef][ISI][Medline]
- Bingham S. Meat, starch and non-starch polysaccharides, are epidemiological and experimental findings consistent with acquired genetic alterations in sporadic colorectal cancer? Cancer Lett 114: 2534, 1997.[CrossRef][ISI][Medline]
- Black SA. Diabetes, diversity, and disparity: what do we do with the evidence? Am J Public Health 92: 543548, 2002.[Abstract/Free Full Text]
- Blount BC, Mack MM, Wehr CM, Macgregor JT, Hiatt RA, Wang G, Wickramasinghe SN, Everson RB, and Ames BN. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci USA 94: 32903295, 1997.[Abstract/Free Full Text]
- Boden G. Pathogenesis of type 2 diabetes. Insulin resistance. Endocrinol Metab Clin North Am 30: 801815, 2001.[ISI][Medline]
- Bream JH, Ping A, Zhang X, Winkler C, and Young HA. A single nucleotide polymorphism in the proximal IFN-gamma promoter alters control of gene transcription. Genes Immun 3: 165169, 2002.[CrossRef][ISI][Medline]
- Cao SX, Dhahbi JM, Mote PL, and Spindler SR. Genomic profiling of short- and long-term caloric restriction effects in the liver of aging mice. Proc Natl Acad Sci USA 98: 1063010635, 2001.[Abstract/Free Full Text]
- Cardon LR and Bell JI. Association study designs for complex diseases. Nat Rev Genet 2: 9199, 2001.[CrossRef][ISI][Medline]
- Chawla A, Repa JJ, Evans RM, and Mangelsdorf DJ. Nuclear receptors and lipid physiology: opening the X-files. Science 294: 18661870, 2001.[Abstract/Free Full Text]
- Chen J, Giovannucci EL, and Hunter DJ. Mthfr polymorphism, methyl-replete diets and the risk of colorectal carcinoma and adenoma among US men and women: an example of gene-environment interactions in colorectal tumorigenesis. J Nutr 129: 560S564S, 1999.[ISI][Medline]
- Clarke SD and Abraham S. Gene expression: nutrient control of pre- and posttranscriptional events. FASEB J 6: 31463152, 1992.[Abstract/Free Full Text]
- Clarke SD. Nutrient regulation of gene and protein expression. Curr Opin Clin Nutr Metab Care 2: 287289, 1999.[CrossRef][Medline]
- Clinton SK, Bostwick DG, Olson LM, Mangian HJ, and Visek WJ. Effects of ammonium acetate and sodium cholate on N-methyl-N'-nitro-N-nitrosoguanidine-induced colon carcinogenesis of rats. Cancer Res 48: 30353039, 1988.[Abstract]
- Collins FS, Guyer MS, and Charkravarti A. Variations on a theme: cataloging human DNA sequence variation. Science 278: 15801581, 1997.[Free Full Text]
- Collins-Schramm HE, Phillips CM, Operario DJ, Lee JS, Weber JL, Hanson RL, Knowler WC, Cooper R, Li H, and Seldin MF. Ethnic-difference markers for use in mapping by admixture linkage disequilibrium. Am J Hum Genet 70: 737750, 2002.[CrossRef][ISI][Medline]
- Committee on Diet and Health Food and Nutrition Board, Commission on Life Sciences, National Research Council, Diet and Health. Implications for Reducing Chronic Disease Risk. Washington, DC: National Academy Press, 1989.
- Cooney CA, Dave AA, and Wolff GL. Maternal methyl supplements in mice affect epigenetic variation and DNA methylation of offspring. J Nutr 132: 2393S2400S, 2002.[Abstract/Free Full Text]
- Cortessis V, Siegmund K, Chen Q, Zhou N, Diep A, Frankl H, Lee E, Zhu QS, Haile R, and Levy D. A case-control study of microsomal epoxide hydrolase, smoking, meat consumption, glutathione S-transferase M3, and risk of colorectal adenomas. Cancer Res 61: 23812385, 2001.[Abstract/Free Full Text]
- Cousins RJ. Nutritional regulation of gene expression. Am J Med 106: 20S23S, 50S51S, 1999.[Medline]
- Da Silva P, Aitken RP, Rhind SM, Racey PA, and Wallace JM. Impact of maternal nutrition during pregnancy on pituitary gonadotrophin gene expression and ovarian development in growth-restricted and normally grown late gestation sheep fetuses. Reproduction 123: 769777, 2002.[Abstract/Free Full Text]
- Daly MJ, Rioux JD, Schaffner SF, Hudson TJ, and Lander ES. High-resolution haplotype structure in the human genome. Nat Genet 29: 229232, 2001.[CrossRef][ISI][Medline]
- Dauncey MJ, White P, Burton KA, and Katsumata M. Nutrition-hormone receptor-gene interactions: implications for development and disease. Proc Nutr Soc 60: 6372, 2001.[ISI][Medline]
- De Caterina R, Madonna R, Hassan J, and Procopio AD. Nutrients and gene expression. World Rev Nutr Diet 89: 2352, 2001.[Medline]
- Deng HW, Chen WM, and Recker RR. Population admixture: detection by Hardy-Weinberg test and its quantitative effects on linkage-disequilibrium methods for localizing genes underlying complex traits. Genetics 157: 885897, 2001.[Abstract/Free Full Text]
- Despres JP. Health consequences of visceral obesity. Ann Med 33: 534541, 2001.[ISI][Medline]
- Dhahbi JM, Mote PL, Wingo J, Tillman JB, Walford RL, and Spindler SR. Calories and aging alter gene expression for gluconeogenic, glycolytic, and nitrogen-metabolizing enzymes. Am J Physiol Endocrinol Metab 277: E352E360, 1999.[Abstract/Free Full Text]
- Dong Z, Huang C, and Ma WY. PI-3 kinase in signal transduction, cell transformation, and as a target for chemoprevention of cancer. Anticancer Res 19: 37433747, 1999.[ISI][Medline]
- Dong Z. Effects of food factors on signal transduction pathways. Biofactors 12: 1728, 2000.[ISI][Medline]
- Douglas JA, Erdos MR, Watanabe RM, Braun A, Johnston CL, Oeth P, Mohlke KL, Valle TT, Ehnholm C, Buchanan TA, Bergman RN, Collins FS, Boehnke M, and Tuomilehto J. The peroxisome proliferator-activated receptor-gamma2 Pro12A1a variant: association with type 2 diabetes and trait differences. Diabetes 50: 886890, 2001.[Abstract/Free Full Text]
- Dreon DM, Fernstrom HA, Williams PT, and Krauss RM. A very-low-fat diet is not associated with improved lipoprotein profiles in men with a predominance of large, low-density lipoproteins. Am J Clin Nutr 69: 411418, 1999.[Abstract/Free Full Text]
- Dreyer C, Krey G, Keller H, Givel F, Helftenbein G, and Wahli W. Control of the peroxisomal beta-oxidation pathway by a novel family of nuclear hormone receptors. Cell 68: 879887, 1992.[ISI][Medline]
- Eastwood MA. A molecular biological basis for the nutritional and pharmacological benefits of dietary plants. QJM 94: 4548, 2001.[Abstract/Free Full Text]
- Eberharter A and Becker PB. Histone acetylation: a switch between repressive and permissive chromatin. Second in review series on chromatin dynamics. EMBO Rep 3: 224229, 2002.[Abstract/Free Full Text]
- Edwards PA, Tabor D, Kast HR, and Venkateswaran A. Regulation of gene expression by SREBP and SCAP. Biochim Biophys Acta 1529: 103113, 2000.[ISI][Medline]
- Ekins S, Mirny L, and Schuetz EG. A ligand-based approach to understanding selectivity of nuclear hormone receptors PXR, CAR, FXR, LXRalpha, and LXRbeta. Pharm Res 19: 17881800, 2002.[CrossRef][ISI][Medline]
- Elson-Schwab I, Poedjosoedarmo K, and Ames BN. KmMutants.org [Online]. Childrens Hospital Oakland Research Institute. http://www.kmmutants.org (updated 19 August 2002).
- Enattah NS, Sahi T, Savilahti E, Terwilliger JD, Peltonen L, and Jarvela I. Identification of a variant associated with adult-type hypolactasia. Nat Genet 30: 233237, 2002.[CrossRef][ISI][Medline]
- Ernst ND, Sempos CT, Briefel RR, and Clark MB. Consistency between US dietary fat intake and serum total cholesterol concentrations: the National Health and Nutrition Examination Surveys. Am J Clin Nutr 66: 965S972S, 1997.[Abstract]
- Fafournoux P, Bruhat A, and Jousse C. Amino acid regulation of gene expression. Biochem J 351: 112, 2000.[CrossRef][ISI][Medline]
- Fairfield KM and Fletcher RH. Vitamins for chronic disease prevention in adults: scientific review. JAMA 287: 31163126, 2002.[Abstract/Free Full Text]
- Falk E, Zhou J, and Moller J. Homocysteine and atherothrombosis. Lipids 36, Suppl: S3S11, 2001.
- Felipo V and Butterworth RF. Neurobiology of ammonia. Prog Neurobiol 67: 259279, 2002.[CrossRef][ISI][Medline]
- Figura N. Helicobacter pylori factors involved in the development of gastroduodenal mucosal damage and ulceration. J Clin Gastroenterol 25, Suppl 1: S149S163, 1997.[CrossRef]
- Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, and Evans RM. 15-Deoxy-delta 12,14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell 83: 803812, 1995.[ISI][Medline]
- Francis GA, Fayard E, Picard F, and Auwerx J. Nuclear receptors and the control of metabolism. Annu Rev Physiol 65: 261311, 2003.[CrossRef][ISI][Medline]
- Frankel WN. Taking stock of complex trait genetics in mice. Trends Genet 11: 471477, 1995.[CrossRef][ISI][Medline]
- Freedman AN, Michalek AM, Marshall JR, Mettlin CJ, Petrelli NJ, Black JD, Zhang ZF, Satchidanand S, and Asirwatham JE. Familial and nutritional risk factors for p53 overexpression in colorectal cancer. Cancer Epidemiol Biomarkers Prev 5: 285291, 1996.[Abstract]
- Gabriel SB, Schaffner SF, Nguyen H, Moore JM, Roy J, Blumenstiel B, Higgins J, DeFelice M, Lochner A, Faggart M, Liu-Cordero SN, Rotimi C, Adeyemo A, Cooper R, Ward R, Lander ES, Daly MJ, and Altshuler D. The structure of haplotype blocks in the human genome. Science 296: 22252229, 2002.[Abstract/Free Full Text]
- Gasser SM and Cockell MM. The molecular biology of the SIR proteins. Gene 279: 116, 2001.[CrossRef][ISI][Medline]
- Gatof D and Ahnen D. Primary prevention of colorectal cancer: diet and drugs. Gastroenterol Clin North Am 31: 587623, xi, 2002.[ISI][Medline]
- Goodridge AG. Dietary regulation of gene expression: enzymes involved in carbohydrate and lipid metabolism. Annu Rev Nutr 7: 157185, 1987.[CrossRef][ISI][Medline]
- Goodridge AG. The role of nutrients in gene expression. World Rev Nutr Diet 63: 183193, 1990.[Medline]
- Gottlicher M, Widmark E, Li Q, and Gustafsson JA. Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor. Proc Natl Acad Sci USA 89: 46534657, 1992.[Abstract]
- Hietanen E, Husgafvel-Pursiainen K, and Vainio H. Interaction between dose and susceptibility to environmental cancer: a short review. Environ Health Perspect 105, Suppl 4: 749754, 1997.
- Hirschhorn JN, Lohmueller K, Byrne E, and Hirschhorn K. A comprehensive review of genetic association studies. Genet Med 4: 4561, 2002.[ISI][Medline]
- Hirschi KD, Kreps JA, and Hirschi KK. Molecular approaches to studying nutrient metabolism and function: an array of possibilities. J Nutr 131: 1605S1609S, 2001.[Abstract/Free Full Text]
- Hollox EJ, Poulter M, Wang Y, Krause A, and Swallow DM. Common polymorphism in a highly variable region upstream of the human lactase gene affects DNA-protein interactions. Eur J Hum Genet 7: 791800, 1999.[ISI][Medline]
- Huang Z, Bodkin NL, Ortmeyer HK, Zenilman ME, Webster NJ, Hansen BC, and Shuldiner AR. Altered insulin receptor messenger ribonucleic acid splicing in liver is associated with deterioration of glucose tolerance in the spontaneously obese and diabetic rhesus monkey: analysis of controversy between monkey and human studies. J Clin Endocrinol Metab 81: 15521556, 1996.[Abstract]
- Jackson AA. Nutrients, growth, and the development of programmed metabolic function. Adv Exp Med Biol 478: 4155, 2000.[ISI][Medline]
- Jacobs MN and Lewis DF. Steroid hormone receptors and dietary ligands: a selected review. Proc Nutr Soc 61: 105122, 2002.[ISI][Medline]
- James RW. Diabetes and other coronary heart disease risk equivalents. Curr Opin Lipidol 12: 425431, 2001.[CrossRef][ISI][Medline]
- Jenkins D, Wolever TMS, Taylor RH, Barker H, Fielden H, Baldwin JM, Bowling AC, Newman HC, Jenkins AL, and Goff DV. Glycemic index of foods: a physiological basis for carbohydrate exchange. Am J Clin Nutr 34: 1981.
- Jenkins DJ, Kendall CW, Augustin LS, Franceschi S, Hamidi M, Marchie A, Jenkins AL, and Axelsen M. Glycemic index: overview of implications in health and disease. Am J Clin Nutr 76: 266S273S, 2002.[Abstract/Free Full Text]
- Jenkins DJA, Kendall CCW, and Ransom TPP. Dietary fiber, the evolution of the human diet and coronary heart disease. Nutr Res 18: 633652, 1998.[CrossRef][ISI]
- Jimenez-Sanchez G, Childs B, and Valle D. Human disease genes. Nature 409: 853855, 2001.[CrossRef][ISI][Medline]
- Johnson GC, Esposito L, Barratt BJ, Smith AN, Heward J, Di Genova G, Ueda H, Cordell HJ, Eaves IA, Dudbridge F, Twells RC, Payne F, Hughes W, Nutland S, Stevens H, Carr P, Tuomilehto-Wolf E, Tuomilehto J, Gough SC, Clayton DG, and Todd JA. Haplotype tagging for the identification of common disease genes. Nat Genet 29: 233237, 2001.[CrossRef][ISI][Medline]
- Judson R, Salisbury B, Schneider J, Windemuth A, and Stephens JC. How many SNPs does a genome-wide haplotype map require? Pharmacogenomics 3: 379391, 2002.[ISI][Medline]
- Kant AK. Consumption of energy-dense, nutrient-poor foods by adult Americans: nutritional and health implications. The Third National Health and Nutrition Examination Survey, 19881994. Am J Clin Nutr 72: 929936, 2000.[Abstract/Free Full Text]
- Kaput J, Swartz D, Paisley E, Mangian H, Daniel WL, and Visek WJ. Diet-disease interactions at the molecular level: an experimental paradigm. J Nutr 124: 1296S1305S, 1994.[Medline]
- Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, and Lehmann JM. A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor gamma and promotes adipocyte differentiation. Cell 83: 813819, 1995.[ISI][Medline]
- Kliewer SA, Xu HE, Lambert MH, and Willson TM. Peroxisome proliferator-activated receptors: from genes to physiology. Recent Prog Horm Res 56: 239263, 2001.[Abstract/Free Full Text]
- Kolonel LN. Fat, meat, and prostate cancer. Epidemiol Rev 23: 7281, 2001.[ISI][Medline]
- Kostulas K, Crisby M, Huang WX, Lannfelt L, Hagenfeldt L, Eggertsen G, Kostulas V, and Hillert J. A methylenetetrahydrofolate reductase gene polymorphism in ischaemic stroke and in carotid artery stenosis. Eur J Clin Invest 28: 285289, 1998.[CrossRef][ISI][Medline]
- Krauss RM. Heterogeneity of plasma low-density lipoproteins and atherosclerosis risk. Curr Opin Lipidol 5: 339349, 1994.[Medline]
- Krauss RM and Dreon DM. Low-density-lipoprotein subclasses and response to a low-fat diet in healthy men. Am J Clin Nutr 62: 478S487S, 1995.[Abstract]
- Krauss RM. Dietary and genetic effects on LDL heterogeneity. World Rev Nutr Diet 89: 1222, 2001.[Medline]
- Krauss RM et al. (Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults). Executive summary of the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 285: 24862497, 2001.[Free Full Text]
- Kuokkanen M, Enattah NS, Oksanen A, Savilahti E, Orpana A, and Jarvela I. Transcriptional regulation of the lactase-phlorizin hydrolase gene by polymorphisms associated with adult-type hypolactasia. Gut 52: 647652, 2003.[Abstract/Free Full Text]
- Lamba JK, Lin YS, Schuetz EG, and Thummel KE. Genetic contribution to variable human CYP3A-mediated metabolism. Adv Drug Deliv Rev 54: 12711294, 2002.[CrossRef][ISI][Medline]
- Lander E and Kruglyak L. Genetic dissection of complex traits: guidelines for interpreting and reporting linkage results. Nat Genet 11: 241247, 1995.[ISI][Medline]
- Lander ES. The new genomics: global views of biology. Science 274: 536539, 1996.[Free Full Text]
- Lander ES et al. (International Human Genome Sequencing Consortium). Initial sequencing and analysis of the human genome. Nature 409: 860921, 2001.[CrossRef][ISI][Medline]
- Lee C, Atanelov L, Modrek B, and Xing Y. ASAP: the Alternative Splicing Annotation Project. Nucleic Acids Res 31: 101105, 2003.[Abstract/Free Full Text]
- Lee CK, Klopp RG, Weindruch R, and Prolla TA. Gene expression profile of aging and its retardation by caloric restriction. Science 285: 13901393, 1999.[Abstract/Free Full Text]
- Lee CK, Allison DB, Brand J, Weindruch R, and Prolla TA. Transcriptional profiles associated with aging and middle age-onset caloric restriction in mouse hearts. Proc Natl Acad Sci USA 99: 1498814993, 2002.[Abstract/Free Full Text]
- Lee MM and Lin SS. Dietary fat and breast cancer. Annu Rev Nutr 20: 221248, 2000.[CrossRef][ISI][Medline]
- Lin SJ and Guarente L. Nicotinamide adenine dinucleotide, a metabolic regulator of transcription, longevity and disease. Curr Opin Cell Biol 15: 241246, 2003.[CrossRef][ISI][Medline]
- Linder CC. The influence of genetic background on spontaneous and genetically engineered mouse models of complex diseases. Lab Anim (NY) 30: 3439, 2001.
- Makridakis NM, di Salle E, and Reichardt JK. Biochemical and pharmacogenetic dissection of human steroid 5 alpha-reductase type II. Pharmacogenetics 10: 407413, 2000.[CrossRef][ISI][Medline]
- Masson LF, McNeill G, and Avenell A. Genetic variation and the lipid response to dietary intervention: a systematic review. Am J Clin Nutr 77: 10981111, 2003.[Abstract/Free Full Text]
- Masuda M, Suzui M, and Weinstein IB. Effects of epigallocatechin-3-gallate on growth, epidermal growth factor receptor signaling pathways, gene expression, and chemosensitivity in human head and neck squamous cell carcinoma cell lines. Clin Cancer Res 7: 42204229, 2001.[Abstract/Free Full Text]
- McCarthy JJ and Hilfiker R. The use of single-nucleotide polymorphism maps in pharmacogenomics. Nat Biotechnol 18: 505508, 2000.[CrossRef][ISI][Medline]
- McKeigue PM, Carpenter JR, Parra EJ, and Shriver MD. Estimation of admixture and detection of linkage in admixed populations by a Bayesian approach: application to African-American populations. Ann Hum Genet 64: 171186, 2000.[CrossRef][ISI][Medline]
- Moazed D. Enzymatic activities of Sir2 and chromatin silencing. Curr Opin Cell Biol 13: 232238, 2001.[CrossRef][ISI][Medline]
- Morris SM Jr, Nilson JH, Jenik RA, Winberry LK, McDevitt MA, and Goodridge AG. Molecular cloning of gene sequences for avian fatty acid synthase and evidence for nutritional regulation of fatty acid synthase mRNA concentration. J Biol Chem 257: 32253229, 1982.[Abstract/Free Full Text]
- Mucci LA, Wedren S, Tamimi RM, Trichopoulos D, and Adami HO. The role of gene-environment interaction in the aetiology of human cancer: examples from cancers of the large bowel, lung and breast. J Intern Med 249: 477493, 2001.[CrossRef][ISI][Medline]
- Muller M and Kersten S. Nutrigenomics: goals and strategies. Nat Rev Genet 4: 315322, 2003.[CrossRef][ISI][Medline]
- Nathan DM. Clinical practice. Initial management of glycemia in type 2 diabetes mellitus. N Engl J Med 347: 13421349, 2002.[Free Full Text]
- Nobel S, Abrahmsen L, and Oppermann U. Metabolic conversion as a pre-receptor control mechanism for lipophilic hormones. Eur J Biochem 268: 41134125, 2001.[Abstract/Free Full Text]
- Novelli G, Margiotti K, Sangiuolo F, and Reichardt JK. Pharmacogenetics of human androgens and prostatic diseases. Pharmacogenomics 2: 6572, 2001.[ISI][Medline]
- Ordovas JM. HDL genetics: candidate genes, genome wide scans and gene-environment interactions. Cardiovasc Drugs Ther 16: 273281, 2002.[CrossRef][ISI][Medline]
- Ordovas JM, Corella D, Cupples LA, Demissie S, Kelleher A, Coltell O, Wilson PW, Schaefer EJ, and Tucker K. Polyunsaturated fatty acids modulate the effects of the APOA1 G-A polymorphism on HDL-cholesterol concentrations in a sex-specific manner: the Framingham Study. Am J Clin Nutr 75: 3846, 2002.[Abstract/Free Full Text]
- Park EI, Paisley EA, Mangian HJ, Swartz DA, Wu MX, OMorchoe PJ, Behr SR, Visek WJ, and Kaput J. Lipid level and type alter stearoyl CoA desaturase mRNA abundance differently in mice with distinct susceptibilities to diet-influenced diseases. J Nutr 127: 566573, 1997.[Abstract/Free Full Text]
- Parra EJ, Marcini A, Akey J, Martinson J, Batzer MA, Cooper R, Forrester T, Allison DB, Deka R, Ferrell RE, and Shriver MD. Estimating African American admixture proportions by use of population-specific alleles. Am J Hum Genet 63: 18391851, 1998.[CrossRef][ISI][Medline]
- Patterson AH, Damon Hewitt S, JD, Zamir D, Rabinowitch HD, Lincoln SE, Lander ES, and Tanksley SD. Mendelian factors underlying quantitative traits in tomato: comparison across species, generations, and environments. Genetics 127: 181197, 1991.[Abstract/Free Full Text]
- Perera FP and Weinstein IB. Molecular epidemiology: recent advances and future directions. Carcinogenesis 21: 517524, 2000.[Abstract/Free Full Text]
- Perusse L and Bouchard C. Gene-diet interactions in obesity. Am J Clin Nutr 72: 1285S1290S, 2000.[Abstract/Free Full Text]
- Pfaff CL, Parra EJ, Bonilla C, Hiester K, McKeigue PM, Kamboh MI, Hutchinson RG, Ferrell RE, Boerwinkle E, and Shriver MD. Population structure in admixed populations: effect of admixture dynamics on the pattern of linkage disequilibrium. Am J Hum Genet 68: 198207, 2001.[CrossRef][ISI][Medline]
- Pianetti S, Guo S, Kavanagh KT, and Sonenshein GE. Green tea polyphenol epigallocatechin-3 gallate inhibits Her-2/neu signaling, proliferation, and transformed phenotype of breast cancer cells. Cancer Res 62: 652655, 2002.[Abstract/Free Full Text]
- Powell IJ and Meyskens FL Jr. African American men and hereditary/familial prostate cancer: intermediate-risk populations for chemoprevention trials. Urology 57: 178181, 2001.[CrossRef][ISI][Medline]
- Prolla TA. DNA microarray analysis of the aging brain. Chem Senses 27: 299306, 2002.[Abstract/Free Full Text]
- Reich DE and Goldstein DB. Detecting association in a case-control study while correcting for population stratification. Genet Epidemiol 20: 416, 2001.[CrossRef][ISI][Medline]
- Reichardt JK. GEN GEN: the genomic genetic analysis of androgen-metabolic genes and prostate cancer as a paradigm for the dissection of complex phenotypes. Front Biosci 4: D596D600, 1999.[Medline]
- Reifsnyder PC, Churchill G, and Leiter EH. Maternal environment and genotype interact to establish diabesity in mice. Genome Res 10: 15681578, 2000.[Abstract/Free Full Text]
- Risch A, Wallace DM, Bathers S, and Sim E. Slow n-acetylation genotype is a susceptibility factor in occupational and smoking related bladder cancer. Hum Mol Genet 4: 231236, 1995.[Abstract]
- Risch N, Ghosh S, and Todd JA. Statistical evaluation of multiple-locus linkage data in experimental species and its relevance to human studies: application to nonobese diabetic (NOD) mouse and human insulin-dependent diabetes mellitus (IDDM). Am J Hum Genet 53: 702714, 1993.[ISI][Medline]
- Risch N. Evolving methods in genetic epidemiology. II. Genetic linkage from an epidemiologic perspective. Epidemiol Rev 19: 2432, 1997.[ISI][Medline]
- Sachidanandam R, Weissman D, Schmidt SC, Kakol JM, Stein LD, Marth G, Sherry S, Mullikin JC, Mortimore BJ, Willey DL, Hunt SE, Cole CG, Coggill PC, Rice CM, Ning Z, Rogers J, Bentley DR, Kwok PY, Mardis ER, Yeh RT, Schultz B, Cook L, Davenport R, Dante M, Fulton L, Hillier L, Waterston RH, McPherson JD, Gilman B, Schaffner S, Van Etten WJ, Reich D, Higgins J, Daly MJ, Blumenstiel B, Baldwin J, Stange-Thomann N, Zody MC, Linton L, Lander ES, and Altshuler D. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 409: 928933, 2001.[CrossRef][ISI][Medline]
- Sachinidis A, Skach RA, Seul C, Ko Y, Hescheler J, Ahn HY, and Fingerle J. Inhibition of the PDGF beta-receptor tyrosine phosphorylation and its downstream intracellular signal transduction pathway in rat and human vascular smooth muscle cells by different catechins. FASEB J 16: 893895, 2002.[Abstract/Free Full Text]
- Sachse C, Smith G, Wilkie MJ, Barrett JH, Waxman R, Sullivan F, Forman D, Bishop DT, and Wolf CR. A pharmacogenetic study to investigate the role of dietary carcinogens in the etiology of colorectal cancer. Carcinogenesis 23: 18391849, 2002.[Abstract/Free Full Text]
- Salmeron JM, JE, Stampfer MJ, Colditz GA, Wing AL, and Willett WC. Dietary fiber, glycemic load, and risk of NIDDM in men. Diabetes Care 20: 545550, 1997.[Abstract]
- Schaefer EJ. Lipoproteins, nutrition, and heart disease. Am J Clin Nutr 75: 191212, 2002.[Abstract/Free Full Text]
- Schmidt A, Endo N, Rutledge SJ, Vogel R, Shinar D, and Rodan GA. Identification of a new member of the steroid hormone receptor superfamily that is activated by a peroxisome proliferator and fatty acids. Mol Endocrinol 6: 16341641, 1992.[Abstract]
- Schork NJ, Fallin D, and Lanchbury JS. Single nucleotide polymorphisms and the future of genetic epidemiology. Clin Genet 58: 250264, 2000.[CrossRef][ISI][Medline]
- Shibata A and Whittemore AS. Genetic predisposition to prostate cancer: possible explanations for ethnic differences in risk. Prostate 32: 6572, 1997.[CrossRef][ISI][Medline]
- Sing CF, Stengard JH, and Kardia SL. Genes, environment, and cardiovascular disease. Arterioscler Thromb Vasc Biol 23: 11901196, 2003.[Abstract/Free Full Text]
- Skibola CF, Smith MT, Kane E, Roman E, Rollinson S, Cartwright RA, and Morgan G. Polymorphisms in the methylenetetrahydrofolate reductase gene are associated with susceptibility to acute leukemia in adults. Proc Natl Acad Sci USA 96: 1281012815, 1999.[Abstract/Free Full Text]
- Slattery ML, Potter JD, Samowitz W, Schaffer D, and Leppert M. Methylenetetrahydrofolate reductase, diet, and risk of colon cancer. Cancer Epidemiol Biomarkers Prev 8: 513518, 1999.[Abstract/Free Full Text]
- Smith-Warner SA, Spiegelman D, Adami HO, Beeson WL, van den Brandt PA, Folsom AR, Fraser GE, Freudenheim JL, Goldbohm RA, Graham S, Kushi LH, Miller AB, Rohan TE, Speizer FE, Toniolo P, Willett WC, Wolk A, Zeleniuch-Jacquotte A, and Hunter DJ. Types of dietary fat and breast cancer: a pooled analysis of cohort studies. Int J Cancer 92: 767774, 2001.[CrossRef][ISI][Medline]
- Sniderman AD, Scantlebury T, and Cianflone K. Hypertriglyceridemic hyperapob: the unappreciated atherogenic dyslipoproteinemia in type 2 diabetes mellitus. Ann Intern Med 135: 447459, 2001.[Abstract/Free Full Text]
- Song F, Srinivasan M, Aalinkeel R, and Patel MS. Use of a cDNA array for the identification of genes induced in islets of suckling rats by a high carbohydrate nutritional intervention. Diabetes 50: 20532060, 2001.[Abstract/Free Full Text]
- Spindler SR, Crew MD, Mote PL, Grizzle JM, and Walford RL. Dietary energy restriction in mice reduces hepatic expression of glucose-regulated protein 78 (BiP) and 94 mRNA. J Nutr 120: 14121417, 1990.[ISI][Medline]
- Stoehr JP, Nadler ST, Schueler KL, Rabaglia ME, Yandell BS, Metz SA, and Attie AD. Genetic obesity unmasks nonlinear interactions between murine type 2 diabetes susceptibility loci. Diabetes 49: 19461954, 2000.[Abstract]
- Sugimoto K, Murakawa Y, Zhang W, Xu G, and Sima AA. Insulin receptor in rat peripheral nerve: its localization and alternatively spliced isoforms. Diabetes Metab Res Rev 16: 354363, 2000.[CrossRef][ISI][Medline]
- Sunde RA. Research needs for human nutrition in the post-genome-sequencing era. J Nutr 131: 33193323, 2001.[Abstract/Free Full Text]
- Svetkey LP, Moore TJ, Simons-Morton DG, Appel LJ, Bray GA, Sacks FM, Ard JD, Mortensen RM, Mitchell SR, Conlin PR, and Kesari M. Angiotensinogen genotype and blood pressure response in the Dietary Approaches to Stop Hypertension (DASH) study. J Hypertens 19: 19491956, 2001.[CrossRef][ISI][Medline]
- Tabor HK, Risch NJ, and Myers RM. Candidate-gene approaches for studying complex genetic traits: practical considerations. Nat Rev Genet 3: 391397, 2002.[CrossRef][ISI][Medline]
- Tishkoff SA and Williams SM. Genetic analysis of African populations: human evolution and complex disease. Nat Rev Genet 3: 611621, 2002.[ISI][Medline]
- Turturro A, Blank K, Murasko D, and Hart R. Mechanisms of caloric restriction affecting aging and disease. Ann NY Acad Sci 719: 159170, 1994.[Abstract]
- Ulrich CM, Kampman E, Bigler J, Schwartz SM, Chen C, Bostick R, Fosdick L, Beresford SA, Yasui Y, and Potter JD. Colorectal adenomas and the C677T MTHFR polymorphism: evidence for gene-environment interaction? Cancer Epidemiol Biomarkers Prev 8: 659668, 1999.[Abstract/Free Full Text]
- Ulrich CM, Bigler J, Whitton JA, Bostick R, Fosdick L, and Potter JD. Epoxide hydrolase Tyr113His polymorphism is associated with elevated risk of colorectal polyps in the presence of smoking and high meat intake. Cancer Epidemiol Biomarkers Prev 10: 875882, 2001.[Abstract/Free Full Text]
- Uyeda K, Yamashita H, and Kawaguchi T. Carbohydrate responsive element-binding protein (ChREBP): a key regulator of glucose metabolism and fat storage. Biochem Pharmacol 63: 20752080, 2002.[CrossRef][ISI][Medline]
- van Dam RM, Willett WC, Rimm EB, Stampfer MJ, and Hu FB. Dietary fat and meat intake in relation to risk of type 2 diabetes in men. Diabetes Care 25: 417424, 2002.[Abstract/Free Full Text]
- Veierod MB, Laake P, and Thelle DS. Dietary fat intake and risk of prostate cancer: a prospective study of 25,708 Norwegian men. Int J Cancer 73: 634638, 1997.[CrossRef][ISI][Medline]
- Velazquez A. Interactions between the genome and the environment, with special reference to nutrient variation. New concepts, emerging methodologies and challenges. World Rev Nutr Diet 89: 93107, 2001.[Medline]
- Venter JC et al. (Celera Genomics). The sequence of the human genome. Science 291: 13041351, 2001.[Abstract/Free Full Text]
- Vincent S, Planells R, Defoort C, Bernard MC, Gerber M, Prudhomme J, Vague P, and Lairon D. Genetic polymorphisms and lipoprotein responses to diets. Proc Nutr Soc 61: 427434, 2002.[CrossRef][ISI][Medline]
- Visek WJ. Effects of urea hydrolysis on cell life-span and metabolism. Fed Proc 31: 11781193, 1972.[ISI][Medline]
- Wade CM, Kulbokas EJ III, Kirby AW, Zody MC, Mullikin JC, Lander ES, Lindblad-Toh K, and Daly MJ. The mosaic structure of variation in the laboratory mouse genome. Nature 420: 574578, 2002.[CrossRef][ISI][Medline]
- Wamberg S, Engel K, and Kildeberg P. Balance of net base in the rat. V. Effects of oral ammonium chloride loading. Biol Neonate 36: 99108, 1979.[ISI][Medline]
- Watkins SM, Hammock BD, Newman JW, and German JB. Individual metabolism should guide agriculture toward foods for improved health and nutrition. Am J Clin Nutr 74: 283286, 2001.[Abstract/Free Full Text]
- Weindruch R, Kayo T, Lee CK, and Prolla TA. Microarray profiling of gene expression in aging and its alteration by caloric restriction in mice. J Nutr 131: 918S923S, 2001.[Abstract/Free Full Text]
- Weindruch R, Keenan KP, Carney JM, Fernandes G, Feuers RJ, Floyd RA, Halter JB, Ramsey JJ, Richardson A, Roth GS, and Spindler SR. Caloric restriction mimetics: metabolic interventions. J Gerontol A Biol Sci Med Sci 56: 2033, 2001. (Spec No. 1)[Abstract/Free Full Text]
- Whorwood CB, Firth KM, Budge H, and Symonds ME. Maternal undernutrition during early to midgestation programs tissue-specific alterations in the expression of the glucocorticoid receptor, 11beta-hydroxysteroid dehydrogenase isoforms, and type 1 angiotensin II receptor in neonatal sheep. Endocrinology 142: 28542864, 2001.[Abstract/Free Full Text]
- Willett W. Isocaloric diets are of primary interest in experimental and epidemiological studies. Int J Epidemiol 31: 694695, 2002.[ISI][Medline]
- Willett WC. Diet and breast cancer. J Intern Med 249: 395411, 2001.[CrossRef][ISI][Medline]
- Willett WC. Balancing life-style and genomics research for disease prevention. Science 296: 695698, 2002.[Abstract/Free Full Text]
- Witte KK, Clark AL, and Cleland JG. Chronic heart failure and micronutrients. J Am Coll Cardiol 37: 17651774, 2001.[CrossRef][ISI][Medline]
- Wolever TMS, Jenkins DJA, Jenkins AL, and Josse RG. The glycemic index: methodology and clinical implications. Am J Clin Nutr 54: 846854, 1991.[Abstract]
- Yan H, Yuan W, Velculescu VE, Vogelstein B, and Kinzler KW. Allelic variation in human gene expression. Science 297: 1143, 2002.[Free Full Text]
- Ye SQ and Kwiterovich PO Jr. Influence of genetic polymorphisms on responsiveness to dietary fat and cholesterol. Am J Clin Nutr 72: 1275S1284S, 2000.[Abstract/Free Full Text]
- Yoshino G, Hirano T, and Kazumi T. Atherogenic lipoproteins and diabetes mellitus. J Diabetes Complications 16: 2934, 2002.[CrossRef][ISI][Medline]
- Zaykin DV, Westfall PH, Young SS, Karnoub MA, Wagner MJ, and Ehm MG. Testing association of statistically inferred haplotypes with discrete and continuous traits in samples of unrelated individuals. Hum Hered 53: 7991, 2002.[ISI][Medline]