ApoE
- NIH-funded research provides new clues on how ApoE4 affects Alzheimer’s risk. National Institutes of Health website.
https://www.nih.gov/news-events/news-releases/nih-funded-research-provides-new-clues-how-apoe4-affects-alzheimers-risk. Published May 16, 2012. Accessed December 4, 2019. - Liu CC, Liu CC, Kanekiyo T, Xu H, Bu G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol. 2013;9(2):106-18.
- Shankar GM, Li S, Mehta TH, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837-42.
- Prelli F, Castaño E, Glenner GG, Frangione B. Differences between vascular and plaque core amyloid in Alzheimer’s disease. J Neurochem. 1988;51(2):648-51.
- Strittmatter WJ, Saunders AM, Schmecheld et al. Apolipoprotein E: High-avidity binding to B-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. PNAS. 1993:90;1977-1981.
- Aleshkov S, Abraham CR, and Zannis VI. Interaction of nascent ApoE2, ApoE3, and ApoE4 isoforms expressed in mammalian cells with amyloid peptide beta (1-40). Relevance to Alzheimer’s disease. Biochemistry. 1997:36(34);10571-80.
- Liu Y, Yu JT, Wang HF, et al. APOE genotype and neuroimaging markers of Alzheimer’s disease: systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2015;86(2):127-34.
- Huang W, Qiu C, Von strauss E, Winblad B, Fratiglioni L. APOE genotype, family history of dementia, and Alzheimer disease risk: a 6-year follow-up study. Arch Neurol. 2004;61(12):1930-4.
- Weisgraber KH, Innerarity TL, Mahley RW. Abnormal lipoprotein receptor-binding activity of the human E apoprotein due to cysteine-arginine interchange at a single site. J Biol Chem. 1982;257(5):2518-21.
- Mahley RW, Huang Y, Rall SC. Pathogenesis of type III hyperlipoproteinemia (dysbetalipoproteinemia). Questions, quandaries, and paradoxes. J Lipid Res. 1999;40(11):1933-49.
- Mahley RW, Rall SC. Apolipoprotein E: far more than a lipid transport protein. Annu Rev Genomics Hum Genet. 2000;1:507-37.
- Mediterranean diet: A heart-healthy eating plan. Mayo Clinic website. https://www.mayoclinic.org/healthy-lifestyle/nutrition-and-healthyeating/in-depth/mediterranean-diet/art-20047801. Accessed December 4, 2019.
- The Secret Behind the Mediterranean Diet. Life Extension website. https://www.lifeextension.com/magazine/2010/1/the-secret-behind-themediterranean- diet. Published January 2010. Accessed December 4, 2019.
- Nutrition and Healthy Eating. Mayo Clinic website. https://www.mayoclinic.org/healthy-lifestyle/nutrition-and-healthy-eating/indepth/nutrition-basics/hlv-20049477. Accessed December 4, 2019.
- Robinson JG, Stone NJ. Antiatherosclerotic and antithrombotic effects of omega-3 fatty acids. Am J Cardiol. 2006;98(4A):39i-49i.
- Thomas SR, Neuzil J, Mohr D, Stocker R. Coantioxidants make alpha-tocopherol an efficient antioxidant for low-density lipoprotein. Am J Clin Nutr. 1995;62(6 Suppl):1357S-1364S.
- Jie KG, Bots ML, Vermeer C, Witteman JC, Grobbee DE. Vitamin K status and bone mass in women with and without aortic atherosclerosis: a population-based study. Calcif Tissue Int. 1996;59(5):352-6.
- Geleijnse JM, Vermeer C, Grobbee DE, et al. Dietary intake of menaquinone is associated with a reduced risk of coronary heart disease: the Rotterdam Study. J Nutr. 2004;134(11):3100-519.
- Aviram M, Dornfeld L. Pomegranate juice consumption inhibits serum angiotensin converting enzyme activity and reduces systolic blood pressure. Atherosclerosis. 2001;158(1):195-8.
- Aviram M, Rosenblat M, Gaitini D, et al. Pomegranate juice consumption for 3 years by patients with carotid artery stenosis reduces common carotid intima-media thickness, blood pressure and LDL oxidation. Clin Nutr. 2004;23(3):423-33.
- Enhance Endothelial Health: How Pomegranate Protects Against Atherosclerosis. Life Extension website.
https://www.lifeextension.com/magazine/2014/11/enhance-endothelial-health-how-pomegranate-protects-against-atherosclerosis. Published November 2014. Accessed December 4, 2019. - Hu S, Belcaro G, Cornelli U, et al. Effects of Pycnogenol® on endothelial dysfunction in borderline hypertensive, hyperlipidemic, and hyperglycemic individuals: the borderline study. Int Angiol. 2015;34(1):43-52.
- Belcaro G, Dugall M, Hosoi M, et al. Pycnogenol® and Centella Asiatica for asymptomatic atherosclerosis progression. Int Angiol. 2014;33(1):20-6.
- Cesarone MR, Belcaro G, Nicolaides AN, et al. Increase in echogenicity of echolucent carotid plaques after treatment with total triterpenic fraction of Centella asiatica: a prospective, placebo-controlled, randomized trial. Angiology. 2001;52 Suppl 2:S19-25.
- Akhtar MS, Ramzan A, Ali A, Ahmad M. Effect of Amla fruit (Emblica officinalis Gaertn.) on blood glucose and lipid profile of normal subjects and type 2 diabetic patients. Int J Food Sci Nutr. 2011;62(6):609-16.
- Cai F, Li C, Wu J, et al. Modulation of the oxidative stress and nuclear factor kappaB activation by theaflavin 3,3′-gallate in the rats exposed to cerebral ischemia-reperfusion. Folia Biol (Praha). 2007;53(5):164-72.
- Englisch W, Beckers C, Unkauf M, Ruepp M, Zinserling V. Efficacy of Artichoke dry extract in patients with hyperlipoproteinemia. Arzneimittelforschung. 2000;50(3):260-5.
- Rondanelli M, Giacosa A, Opizzi A, et al. Beneficial effects of artichoke leaf extract supplementation on increasing HDL-cholesterol in subjects with primary mild hypercholesterolaemia: a double-blind, randomized, placebo-controlled trial. Int J Food Sci Nutr. 2013;64(1):7-15.
- Rondanelli M, Castellazzi AM, Riva A, et al. Natural Killer Response and Lipo-Metabolic Profile in Adults with Low HDL-Cholesterol and Mild Hypercholesterolemia: Beneficial Effects of Artichoke Leaf Extract Supplementation. Evid Based Complement Alternat Med. 2019;2019:2069701.
- Evans M, Rumberger JA, Azumano I, Napolitano JJ, Citrolo D, Kamiya T. Pantethine, a derivative of vitamin B5, favorably alters total, LDL and non-HDL cholesterol in low to moderate cardiovascular risk subjects eligible for statin therapy: a triple-blinded placebo and diet-controlled investigation. Vasc Health Risk Manag. 2014;10:89-100.
- Scarmeas N, Luchsinger JA, Schupf N, et al. Physical activity, diet, and risk of Alzheimer disease. JAMA. 2009;302(6):627-37.
- Larson EB, Wang L. Exercise, aging, and Alzheimer disease. Alzheimer Dis Assoc Disord. 2004;18(2):54-6.
- Hoffmann K, Sobol NA, Frederiksen KS, et al. Moderate-to-High Intensity Physical Exercise in Patients with Alzheimer’s Disease: A Randomized Controlled Trial. J Alzheimers Dis. 2016;50(2):443-53.
- How to Delay Brain Aging by 11 Years. Life Extension website. https://www.lifeextension.com/magazine/2016/4/how-to-delay-brain-aging-by-11-years. Published April 2016. Accessed December 5, 2019.
- Scarmeas N, Stern Y, Tang MX, Mayeux R, Luchsinger JA. Mediterranean diet and risk for Alzheimer’s disease. Ann Neurol. 2006;59(6):912-21.
- Yurko-mauro K, Mccarthy D, Rom D, et al. Beneficial effects of docosahexaenoic acid on cognition in age-related cognitive decline. Alzheimers Dement. 2010;6(6):456-64.
- Yassine HN, Feng Q, Azizkhanian I, et al. Association of Serum Docosahexaenoic Acid With Cerebral Amyloidosis. JAMA Neurol. 2016;73(10):1208-1216.
- Yassine HN, Braskie MN, Mack WJ, et al. Association of Docosahexaenoic Acid Supplementation With Alzheimer Disease Stage in Apolipoprotein E ε4 Carriers: A Review. JAMA Neurol. 2017;74(3):339-347.
- Kato-kataoka A, Sakai M, Ebina R, Nonaka C, Asano T, Miyamori T. Soybean-derived phosphatidylserine improves memory function of the elderly Japanese subjects with memory complaints. J Clin Biochem Nutr. 2010;47(3):246-55.
- Schreiber S, Kampf-sherf O, Gorfine M, Kelly D, Oppenheim Y, Lerer B. An open trial of plant-source derived phosphatydilserine for treatment of age-related cognitive decline. Isr J Psychiatry Relat Sci. 2000;37(4):302-7.
- Szilágyi G, Nagy Z, Balkay L, et al. Effects of vinpocetine on the redistribution of cerebral blood flow and glucose metabolism in chronic ischemic stroke patients: a PET study. J Neurol Sci. 2005;229-230:275-84.
- Dézsi L, Kis-varga I, Nagy J, Komlódi Z, Kárpáti E. [Neuroprotective effects of vinpocetine in vivo and in vitro. Apovincaminic acid derivatives as potential therapeutic tools in ischemic stroke]. Acta Pharm Hung. 2002;72(2):84-91.
- Pereira C, Agostinho P, Oliveira CR. Vinpocetine attenuates the metabolic dysfunction induced by amyloid beta-peptides in PC12 cells. Free Radic Res. 2000;33(5):497-506.
- Mashayekh A, Pham DL, Yousem DM, Dizon M, Barker PB, Lin DD. Effects of Ginkgo biloba on cerebral blood flow assessed by quantitative MR perfusion imaging: a pilot study. Neuroradiology. 2011;53(3):185-91.
- Motoi Y, Shimada K, Ishiguro K, Hattori N. Lithium and autophagy. ACS Chem Neurosci. 2014;5(6):434-42.
- Nunes MA, Schöwe NM, Monteiro-silva KC, et al. Chronic Microdose Lithium Treatment Prevented Memory Loss and Neurohistopathological Changes in a Transgenic Mouse Model of Alzheimer’s Disease. PLoS ONE. 2015;10(11):e0142267.
- Nunes MA, Viel TA, Buck HS. Microdose lithium treatment stabilized cognitive impairment in patients with Alzheimer’s disease. Curr Alzheimer Res. 2013;10(1):104-7.
- Szaniszlo P, German P, Hajas G, Saenz DN, Kruzel M, Boldogh I. New insights into clinical trial for Colostrinin in Alzheimer’s disease. J Nutr Health Aging. 2009;13(3):235-41.
- Leszek J, Inglot AD, Janusz M, et al. Colostrinin proline-rich polypeptide complex from ovine colostrum–a long-term study of its efficacy in Alzheimer’s disease. Med Sci Monit. 2002;8(10):PI93-6.
- Slutsky I, Abumaria N, Wu LJ, et al. Enhancement of learning and memory by elevating brain magnesium. Neuron. 2010;65(2):165-77.
- Li W, Yu J, Liu Y, et al. Elevation of brain magnesium prevents synaptic loss and reverses cognitive deficits in Alzheimer’s disease mouse model. Mol Brain. 2014;7:65.
- Wang J, Liu Y, Zhou LJ, et al. Magnesium L-threonate prevents and restores memory deficits associated with neuropathic pain by inhibition of TNF-α. Pain Physician. 2013;16(5):E563-75.
- Zhang HY, Tang XC. Neuroprotective effects of huperzine A: new therapeutic targets for neurodegenerative disease. Trends Pharmacol Sci. 2006;27(12):619-25.
- Liang YQ, Tang XC. Comparative effects of huperzine A, donepezil and rivastigmine on cortical acetylcholine level and acetylcholinesterase activity in rats. Neurosci Lett. 2004;361(1-3):56-9.
- Li WM, Kan KK, Carlier PR, Pang YP, Han YF. East meets West in the search for Alzheimer’s therapeutics – novel dimeric inhibitors from tacrine and huperzine A. Curr Alzheimer Res. 2007;4(4):386-96.
- Kennedy DO, Pace S, et al. Effects of cholinesterase inhibiting sage (Salvia officinalis) on mood, anxiety and performance on a psychological stressor battery. Neuropsychopharmacology. 2006;31(4):845-52.
- Lopresti A et al. Salvia (Sage): A Review of its Potential Cognitive-Enhancing and Protective Effects. Drugs in R&D. 2017;17(1):53-64.
- Scholey AB, et al. An extract of Salvia (sage) with anticholinesterase properties improves memory and attention in healthy older volunteers. Psychopharmacology. 2008;198(1):127-39.
Cardiac Health
- Roberts R and Stewart A. 9p21 and the genetic Revolution for Coronary Artery Disease. Clinical Chemistry. 2012; 58(1):104-112.
- Catt KJ et al. Angiotensin II blood levels in human hypertension. The Lancet. 1971; 297:459-464.
- Wang WZ. Association between T174M polymorphism in the angiotensinogen gene and risk of coronary artery disease: a meta-analysis. J Geriatr Cardiol. 2013: 10:59-65.
- Cosentino F and Luscher TF. Maintenance of vascular integrity: role of nitric oxide and other bradykinin mediators. Eur Heart J. 1995; 16 Suppl K:4-12.
- Tesauro M et. Al. Intracellular processing of endothelial nitric oxide synthase isoforms associated with differences in severity of cardiopulmonary diseases: cleavage of proteins with aspartate vs. glutamate at position 298. Proc Natl Acad Sci USA. 2006:2832-2835.
- Wang M et al. Association of G894T polymorphism in endothelial nitric oxide synthase gene with the risk of ischemic stroke: A meta-analysis.Biomed Rep. 2013: (1)1:144-150.
- Refsum H et al. The Hordaland Homocysteine Study: A community-Based Study of Homocysteine, Its Determinants, and Associations with Disease. J Nutr. 2006; 136:1731S-174OS.
- Frost P et. Al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet.1995; 10-111-113.
- Cotlarciuc I et al. Effect of genetic variants associated with plasma homocysteine levels on stroke risk. Stroke. 2014; 45(7):1920-4.
- Weisberg I et al. A second genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR) associated with decreased enzyme activity. Mol Genet Metab. 1998; 64:169-72
- Poort SR et.al. A common genetic variation in the 3’-untranslated region of the prothrombin gene is associated with elevated plasma prothrombin levels and an increase in venous thrombosis. Blood. Nov 15, 1996:88(10):3698-703.
- Simone B et al. Risk of venous thromboembolism associated with single and combined effects of Factor V Leiden, Prothrombin 20210A and Methylenetethraydrofolate reductase C677T: a meta-analysis involving over 11,000 cases and 21,000 controls. Eur J Epidemiol. 2013; 28(8):621-47.
- Gohil, R et al. The Genetics of Venous Thromboembolism: A meta-analysis involving 120,000 cases and 180,000 controls. Journal of Thrombosis and Haemostasis. 2009; 102: 360-370.
- Kujovich J et. al GeneReviews; 1999 “Factor V Leiden Thrombophilia”.
- Bertina RM et al. Mutation in blood coagulation factor V associated with resistance to activated protein C. Nature. 1994; 369(6475):64-7.
- Mahley RW et al. Apolipoprotein E: Far More Than a Lipid Transport Protein. Annu Rev Genomics Hum Genet. 2000; 1:507-537.
- Howard BV et al. Association of Apolipoprotein E Phenotype with Plasma lipoproteins in African-American and White Young Adults. Am J Epidemiol. 1998: 148(9):859-868.
- Niemi M et al. Organic Anion Transporting Protein 181:a Genetically Polymorphic Transporter of Major Importance for Hepatic Drug Uptake. Pharm Review. 2011; 63(1): 157-181.
- Saita E et al. Anti-inflammatory Diet for Atherosclerosis and Coronary Artery Disease: Antioxidant Foods. Clinical Medicine Insights: Cardiology. 2014:8(S3) 61-65.
- Fan E, Zhang L, Jiang S, Bai Y. Beneficial effects of resveratrol on atherosclerosis. J Med Food. 2008;11(4):610-4.
- Hosogoe N et al. Add-on Antiplatelet Effects of Eicosapentaenoic Acid with Tailored Dose Setting in Patients on Dual Antiplatelet Therapy. Int Heart J. 2017 Ag 3; 58 (4): 481-485.
- Ho, H. T. et al. A systematic review and meta-analysis of randomized controlled trials of the effect of konjac glucomannan, a viscous soluble fiber, on LDL cholesterol and the new lipid targets non-HDL cholesterol and apolipoprotein B. Am J Clin Nutr. 2017; 105(5):1239-1247.
- Zelman, K. April 07, 2016. Fiber: How Much Do I Need? WebMD. https://www.webmd.com/diet/guide/fiber-how-much-do-you-need#1. [2017,October 2]
- Nordmann A et al. Effects of Low-Carbohydrate vs Low-fat Diets on Weight loss and Cardiovascular Risk Factors. Arch Intern Med. 2006; 166:285-293.
Celiac – DQ2/DQ8
- Megiorni F et al. HLA-DQ and risk gradient for celiac disease. Hum Imm. 2009; 70:55-59.
- Megiorni F et al. HLA-DQA1 and HLA-DQB1 in celiac disease predisposition: practical implications of the HLA molecular typing. J of Biomed Sci. 2012; 19:88.
- Abadie et al. Integration of genetic and immunological insights into a model of celiac disease pathogenesis. Annu. Rev. Immunol. 2011; 29:493-525.
- Kagnoff MF. Celiac disease: pathogenesis of a model immunogenetic disease. J Clin Invest. 2007; 117:41-49.
- Megiorni F et al. HLA-DQ and susceptibility to celiac disease: evidence for gender differences and parent-of-origin effects. Am J Gastroenterol. 2008; 103:997-1003.
- Karell et al. HLA types in celiac disease patients not carrying the DQA1*05-DQB1*02(DQ2) heterodimer: results from the European genetics cluster of celiac disease. Hum Immunol. 2003; 64:469-477
Comprehensive Genetic
- Jalba MS, Rhoads GG, Demissie K. Association of codon 16 and codon 27 beta 2-adrenergic receptor gene polymorphisms with obesity: a meta-analysis. Obesity (Silver Spring). 2008;16(9):2096-106.
- Lange LA, Norris JM, Langefeld CD, et al. Association of adipose tissue deposition and beta-2 adrenergic receptor variants: the IRAS family study. Int J Obes (Lond). 2005;29(5):449-57.
- Martínez JA et al. Obesity risk is associated with carbohydrate intake in women with the Gln27Glu β2-adrenoreceptor polymorphism. J Nutr. 2003; 133:2549-2554.
- Large V et al. Human Beta-2 adrenoreceptor gene polymorphisms are highly frequent in obesity and associate with altered adipocyte Beta-2 adrenoceptor function. J Clin Invest. 1997; 100:3005-3013.
- Macho-Azcarate et al. Gln27Glu polymorphism in the beta2 adrenergic receptor gene and lipid metabolism during exercise in obese women. Int J Obesity. 2002; 26:1434-1441.
- HGNC. http://www.genenames.org/cgibin/gene_symbol_report? hgnc_id=HGNC:286.
- org. Nutrition and Healthy Eating: Carbohydrates: How carbs fit into a healthy diet. http://www.mayoclinic.org/healthy-living/nutrition-and-healthy-eating/in-depth/carbohydrates/art-20045705?pg=1.
- Layman DK. Dietary Guidelines should reflect new understandings about adult protein needs. Nutr Metab (Lond). 2009; 6:12.
- Phares DA et al. Association Between Body Fat Response to Exercise Training and Multilocus ADR Genotypes. Obes Res. 2004; 12(5):807-815.
- Corbalan MS et al. The 27Glu polymorphism of the Beta2-adrenergic receptor gene interacts with physical activity influencing obesity risk among female subjects. Clin Genet. 2002; 61:305-307.
- Zhang et al. Association of Gln27Glu and Arg16Gly Polymorphisms in Beta2-Adrenergic Receptor Gene with Obesity Susceptibility: A Meta-Analysis. PLoS ONE. 2014; 9(6): e100489.
- Martinez-lopez E, Garcia-garcia MR, Gonzalez-avalos JM, et al. Effect of Ala54Thr polymorphism of FABP2 on anthropometric and biochemical variables in response to a moderate-fat diet. Nutrition. 2013;29(1):46-51.
- Pratley RE, Baier L, Pan DA, et al. Effects of an Ala54Thr polymorphism in the intestinal fatty acid-binding protein on responses to dietary fat in humans. J Lipid Res. 2000;41(12):2002-8.
- Levy E et al. The polymorphism at codon 54 of the FABP2 gene increases fat absorption in human intestinal explants. J Biol Chem. 2001; 276:39679-39684.
- Marin C et al. The Ala54Thr polymorphism of the fatty acid-binding protein 2 gene is associated with a change in insulin sensitivity after a change in the type of dietary fat. Am J Clin Nutr. 2005; 82:196-200.
- Albala C et al. FABP2 Ala54Thr polymorphism and diabetes in Chilean elders. Diab Res Clin Pract. 2007; 77:245-250.
- Paglialunga S et al. Regulation of postprandial lipemia: an update on current trends. Appl Physiol Nutr Metab. 2007; 32:61-75.
- Takakura Y et al. Thr54 allele of the FABP2 gene affects resting metabolic rate and visceral obesity. Diabetes Res Clin Pract. 2005; 67:36-42.
- Hegele RA. A Review of Intestinal Fatty Acid Binding Protein Gene Variation and the Plasma Lipoprotein Response to Dietary Components. Clin Biochem. 1998; 31:609-612.
- Gaggini M et al. Non-Alcoholic Fatty Liver Disease (NAFLD) and Its Connection with Insulin Resistance, Dyslipidemia, Atherosclerosis and Coronary Heart Disease. Nutrients. 2013; 5:1544-1560.
- Almeida JC et al. The Ala54Thr polymorphism of the FABP2 gene influences the postprandial fatty acids in patients with type 2 diabetes. J Clin Endocrin Met. 2010; 95:3909-3917.
- Dworatzek PD et al. Postprandial lipemia in subjects with the threonine 54 variant of the fatty acid-binding protein 2 gene is dependent on the type of fat ingested. Am J Clin Nutr. 2004; 79:1110-1117.
- Weiss EP et al. FABP2 Ala54Thr genotype is associated with glucoregulatory function and lipid oxidation after a high-fat meal in sedentary nondiabetic men and women. Am J Clin Nutr. 2007; 85:102-108.
- Weickert MO et al. Metabolic Effects of Dietary Fiber consumption and Prevention of Diabetes. J Nutr. 2008; 138(3):439-442.
- org. Nutrition and Healthy Eating: Carbohydrates: How carbs fit into a healthy diet. http://www.mayoclinic.org/ healthy-living/nutrition-and-healthy-eating/in-depth/carbohydrates/ art-20045705?pg=1
- Pfeiffer M et al. The influence of walking performed immediately before meals with moderate fat content on postprandial lipemia. Lipids Health Dis. 2005; 4:24.
- Brandou F et al. Impact of high- and low-intensity targeted exercise training on the type of substrate utilization in obese boys submitted to a hypocaloric diet. Diabetes Metab. 2005; 31(4 Pt 1):327-325.
- Corella D et al. A High Intake of Saturated Fatty Acids Strengthens the Association between the Fat Mass and Obesity-Associated Gene and BMI. J of Nutr. 2011; 141:2219-2225.
- Kilpelainen TO et al. Physical Activity Attenuates the Influence of FTO Variants on Obesity Risk: A Meta-Analysis of 218,166 Adults and 19.268 Children. PLOS Medicine. 2011; 8(11):31001116.
- Frayling TM et al. A Common Variant in the FTO Gene is Associated with Body Mass Index and Predisposition to Childhood and Adult Obesity. Science. 2007; 316(58):889-894.
- Berulava T and Horsthemke B. The obesity-associated SNPs in intron 1 of the FTO gene affect primary transcript levels. Eur J Hum Genet. 2010; 18(9):1054-1058.
- Karra E et al. A link between FTO, ghrelin, and impaired brain food-cue responsivity. J Clin Invest. 2013; 123(8):3539-3551.
- Lu Y et al. Obesity genomics: assessing the transferability of susceptibility loci across diverse populations. Genome Med. 2013; 5:55.
- Cho YS et al. A large-scale genome-wide association study of Asian populations uncovers genetic factors influencing eight quantitative traits. Nat Genet. 2009; 41:527-534.
- Jaaskelainen A et al. Meal Frequencies Modify the Effect of Common Genetic Variants on Body Mass Index in Adolescents of the Northern Finland Birth Cohort 1986. PLOS One. 2013; 8(9):e73802.
- Zhang X et al. FTO Genotype and 2-Year Change in Body Composition and Fat Distribution in Response to Weight-Loss Diets: The POUNDS Lost Trial. Diabetes. 2012; 61:3005-3011.
- Ortega-Azorin C et al. Associations of the FTO rs9939609 and the MC4R rs17782313 polymorphisms with type 2 diabetes are modulated by diet, being higher when adherence to the Mediterranean diet pattern is low. Cardiovasc Diabetol. 2012; 11:137.
- Mitchell JA et al. FTO Genotype and the Weight Loss Benefits of Moderate Intensity Exercise. Obesity. 2010; 18(3):641-643.
- Lauria F et al. Prospective Analysis of the Association of a Common Variant of FTO (rs9939609) with Adiposity in Children: Results of the IDEFICS Study. PLoS One. 2012; 7: e48876.
- Velders FP et al. FTO at rs9939609, food responsiveness, emotional control and symptoms of ADHD in preschool children. PLoS One. 2012; 7:e49131.
- Wardle J et al. Obesity associated genetic variation in FTO is associated with diminished satiety. J Clin Endocrinol Metab. 2008; 93:3640–3643.
- den Hoed M et al. Postprandial responses in hunger and satiety are associated with the rs9939609 single nucleotide polymorphism in FTO. Am J Clin Nutr. 2009; 90:1426–1432.
- Loos RJF et al. Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat Genet. 2008; 40(6):768-775.
- Qi L et al. The common obesity variant near MC4R gene is associated with higher intakes of total energy and dietary fat, weight change and diabetes risk in women. Human Mol Genetics 2008; 17:3502-3508.
- Xi B et al. Common polymorphism near the MC4R gene is associated with type 2 diabetes: data from a meta-analysis of 123,373 individuals. Diabetologia 2012; 55:2660–2666.
- Stutzmann F et al. Common genetic variation near MC4R is associated with eating behaviour patterns in European populations. Int J Obes. 2009; 33:373-378.
- Xi B et al. Influence of physical inactivity on associations between single nucleotide polymorphisms and genetic predisposition to childhood obesity. Am J Epidemiology 2011; 173:1256-1262.
- Acosta A et al. Association of melanocortin 4 receptor gene variation with satiation and gastric emptying in overweight and obese adults. Genes Nutr. 2014; 9(2):384.
- Zlatohlavek L et al. FTO and MC4R gene variants determine BMI changes in children after intensive lifestyle intervention. Clin Biochem. 2013; 46:313-31.
- Raynor HA et al. Dietary energy density and successful weight loss maintenance. Eat Behav. 2011; 12(2):119-125.
- Otten JJ et al. DRI: Dietary Reference Intakes: The Essential Guide to Nutrient Requirements. Washington, DC. National Academies Press. c2006.
- OSU: Linus Pauling Institute. Glycemic Index and Glycemic Load. http://lpi.oregonstate.edu/infocenter/foods/grains/gigl.html
- Jaaskelainen A et al. Meal Frequencies Modify the Effect of Common Genetic Variants on Body Mass Index in Adolescents of the Northern Finland Birth Cohort 1986. PLOS One. 2013; 8(9):e73802.
- Little JP et al. A practical model of low-volume high-intensity interval training induces mitochondrial biogenesis in human skeletal muscle: potential mechanisms. J Physiol. 2010; 588 (Pt 6):1011-1022.
- Bauer F et al. Obesity genes identified in genome-wide association studies are associated with adiposity measures and potentially with nutrient-specific food preference. Am J Clin Nutr. 2009; 90:951–959.
- Volckmar AL et al. Mutation screen in the GWAS derived obesity gene SH2B1 including functional analyses of detected variants. BMC Med Genomics. 2012; 5:65.
- Jamshidi Y et al. The SH2B gene is associated with serum leptin and body fat in normal female twins. Obesity. 2007; 15:5-9.
- Ren D et al. Neuronal SH2B1 is essential for controlling energy and glucose homeostasis. J Clin Invest. 2007; 117:397–406.
- Morris DL et al. SH2B1 enhances insulin sensitivity by both stimulating the insulin receptor and inhibiting tyrosine dephosphorylation of insulin receptor substrate proteins. Diabetes. 2009; 58:2039-2047.
- McCaffery JM et al. Obesity susceptibility loci and dietary intake in the Look AHEAD Trial. Am J Clin Nutr. 2012; 95:1477-1486.
- Duan C et al. Disruption of the SH2-B gene causes age-dependent insulin resistance and glucose intolerance. Mol Cell Biol 2004; 24:7435–7443.
- Ren D et al. Identification of SH2-B as a key regulator of leptin sensitivity, energy balance, and body weight in mice. Cell Metabolism. 2005; 2:95–104.
- Wolpert HA et al. Dietary Fat Acutely Increases Glucose Concentrations and Insulin Requirements in Patients With Type 1 Diabetes: Implications for carbohydrate-based bolus dose calculation and intensive diabetes management. Diabetes Care. 2012; 36(4):810-816.
- Diabetes.org American Diabetes Association: Insulin Basics. http://www.diabetes.org/living-with-diabetes/treatment-and-care/ medication/insulin/insulin-basics.html.
- Klok MD et al. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: a review. Obes Rev. 2007; 8(1):21-34.
- Bachman JL et al. Eating frequency is higher in weight loss maintainers and normal-weight individuals than in overweight individuals. J Am Diet Assoc. 2011; 111(11):1730-4.
- Lichtenstein MB et al. Exercise Addiction in Men Is Associated With Lower Fat-Adjusted Leptin Levels. Clin J Sport Med. 2015; 25(2):138-43.
- OSU: Linus Pauling Institute. Glycemic Index and Glycemic Load. http://lpi.oregonstate.edu/infocenter/foods/grains/gigl.html
- Zhang Z et al. A high-legume low-glycemic index diet reduces fasting plasma leptin in middle-aged insulin-resistant and –sensitive men. Eur J Clin Nutr. 2011; 65(3):415-418.
- Fall T et al. The role of obesity-related genetic loci in insulin sensitivity. Diabet Med. 2012; 29(7):e62-66.
- Robiou-du-Pont S et al. Contribution of 24 obesity-associated genetic variants to insulin resistance, pancreatic beta-cell function and type 2 diabetes. Int J Obesity. 2013; 37:980-985.
- Weickert MO et al. Metabolic Effects of Dietary Fiber Consumption and Prevention of Diabetes. J Nutr. 2008; 138(3):439-442.
- Willett WC. Eat, Drink, and be Healthy: The Harvard Medical School Guide to Healthy Eating. New York: Simon & Schuster; 2001.
- Zaccaria M et al. Plasma leptin and energy expenditure during prolonged, moderate intensity, treadmill exercise. J Endocrinol Invest. 2013; 36(6):396-401.
- Li S et al. Physical Activity Attenuates the Genetic Predisposition to Obesity in 20,000 Men and Women from EPIC-Norfolk Prospective Population Study. PLOS Medicine. 2010; 7(8):e1000332.
- Lee S et al. Aerobic exercise but not resistance exercise reduces intrahepatic lipid content and visceral fat and improves insulin sensitivity in obese adolescent girls: a randomized controlled trial. Am J Physiol Endocrinol Metab. 2013; 305(10):E1222-1229.
- Karvanen J, Silander K, Kee F, et al. The impact of newly identified loci on coronary heart disease, stroke and total mortality in the MORGAM prospective cohorts. Genet Epidemiol. 2009;33(3):237-46.
- Ye S et al. Association of Genetic Variation on Chromosome 9p21 with Susceptibility and Progression of Atherosclerosis. JACC 2008; 52(5): 378-384.
- Roberts R. Genetics of Coronary Artery Disease. Circ Res. 2014; 114:1890-1903.
- The Wellcome Trust Case Consortium, Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007; 447(7145): 661-678.
- Genetics Home Reference. Genes: CDKN2A, http://ghr.nlm.nih.gov/gene/CDKN2A 4.
- Roberts R and Stewart A. 9p21 and the Genetic Revolution for Coronary Artery Disease. Clinical Chemistry. 2012; 58(1):104-112.
- Munir M et al. The association of 9p21-3 locus with coronary atherosclerosis: a systematic review and meta-analysis. BMC Medical Genetics. 2014; 15:66.
- Dandonna S et al. Gene Dosage of the Common Variant 9p21 Predicts Severity of Coronary Artery Disease. J Am Coll Cardiol. 2010; 56(6):479-488.
- Zakrzewski-jakubiak M, De denus S, Dubé MP, Bélanger F, White M, Turgeon J. Ten renin-angiotensin system-related gene polymorphisms in maximally treated Canadian Caucasian patients with heart failure. Br J Clin Pharmacol. 2008;65(5):742-51.
- Wang WZ. Association between T174M polymorphism in the angiotensinogen gene and risk of coronary artery disease: a meta-analysis. J Geriatr Cardiol. 2013; 10:59-65.
- Touyz RM and EL Schiffrin. Signal Transduction Mechanisms Mediating the Physiological and Pathophysiological Actions of Angiotensin II in Vascular Smooth Muscle Cells. Pharmacol Rev. 2000; 52(4):639-672.
- Catt KJ et al. Angiotensin II blood-levels in human hypertension. The Lancet. 1971; 297:459-464.
- Million Hearts: strategies to reduce the prevalence of leading cardiovascular disease risk factors. United States, 2011. MMWR 2011; 60(36):1248–51.
- Li X et al. AGT gene polymorphisms (M235T, T174M) are associated with coronary heart disease in a Chinese population. J Renin Angiotensin Aldosterone Syst. 2013; 14(4):354-9.
- Gardemann A et al. Angiotensinogen T174M and M235T gene polymorphisms are associated with the extent of coronary atherosclerosis. Atherosclerosis. 1999; 145(2):309-14.
- Irvin MR, Kabagambe EK, Tiwari HK, et al. Apolipoprotein E polymorphisms and postprandial triglyceridemia before and after fenofibrate treatment in the Genetics of Lipid Lowering and Diet Network (GOLDN) Study. Circ Cardiovasc Genet. 2010;3(5):462-7.
- Zende PD, Bankar MP, Kamble PS, Momin AA. Apolipoprotein e gene polymorphism and its effect on plasma lipids in arteriosclerosis. J Clin Diagn Res. 2013;7(10):2149-52.
- Mahley RW et al. Apolipoprotein E: Far More Than a Lipid Transport Protein. Annu Rev Genomics Hum Genet. 2000; 1:507-537.
- Eichner JE et al. Apolipoprotein E Polymorphism and Cardiovascular Disease: A HuGE Review. Am J Epidemiol. 2002; 155(6):487-495.
- Villeneuve S et al. The potential applications of Apolipoprotein E in personalized medicine. Front Aging Neurosci. 2014; 6:154.
- Howard BV et al. Association of Apolipoprotein E Phenotype with Plasma Lipoproteins in African-American and White Young Adults. Am J Epidemiol. 1998; 148(9):859-868.
- Yassine H. et al. DHA brain uptake and APOE4 status: a PET study with (1-11C)-DHA. Alzheimer’s Research & Therapy 2017; 9:23.
- Casas JP et al. Endothelial Nitric Oxide Synthase Genotype and Ischemic Heart Disease: Meta-Analysis of 26 Studies Involving 23,028 Subjects. Circulation. 2004; 109:1359-1365.
- Luo JQ, Wen JG, Zhou HH, Chen XP, Zhang W. Endothelial nitric oxide synthase gene G894T polymorphism and myocardial infarction: a meta-analysis of 34 studies involving 21,068 subjects. PLoS ONE. 2014;9(1):e87196.
- Alkaitis M et al. Recoupling the Cardiac Nitric Oxide Synthases: Tetrahydrobiopterin Synthesis and Recycling. Curr Heart Fail Rep. 2012; 9:200–210.
- Xie X et al. Endothelial nitric oxide synthase gene single nucleotide polymorphisms and the risk of hypertension: A meta-analysis involving 63,258 subjects. Clinical and Experimental Hypertension. 2017; 39(2):175-182.
- Shengyuan Liu et al. The nitric oxide synthase 3 G894T polymorphism associated with Alzheimer’s disease risk: a meta-analysis. Sci Rep. 2015; 5:13598.
- Marsden PA et al. Structure and chromosomal localization of the human constitutive endothelial nitric oxide synthase gene. J Biol Chem. 1992; 68:17478-17488.
- Cosentino F and Luscher TF. Maintenance of vascular integrity: role of nitric oxide and other bradykinin mediators. Eur Heart J. 1995; 16 Suppl K:4-12.
- Huang PL. eNOS, metabolic syndrome and cardiovascular disease. Trends Endocrinol Metab. 2009; 20(6):295-302.
- Hogg N et al. Inhibition of low-density lipoprotein oxidation by nitric oxide. Potential role in atherogenesis. FEBS Lett. 1993; 334(2):170-174.
- Radomski MW et al. Endogenous nitric oxide inhibits human platelet adhesion to vascular endothelium. Lancet. 1987; 2:1057-1058.
- Tesauro M et al. Intracellular processing of endothelial nitric oxide synthase isoforms associated with differences in severity of cardiopulmonary diseases: cleavage of proteins with aspartate vs. glutamate at position 298. Proc Natl Acad Sci U S A. 2000; 6:2832-2835.
- Niu W and Y Qi. An Updated Meta-Analysis of Endothelial Nitric Oxide Synthase Gene: Three Well-Characterized Polymorphisms with Hypertension. Plos One. 2011; 6(9):e24266.
- Wang M et al. Association of G894T polymorphism in endothelial nitric oxide synthase gene with the risk of ischemic stroke: A meta-analysis. Biomed Rep. 2013; 1(1):144-150.
- Zigra AM et al. eNOS gene variants and the risk of premature myocardial infarction. Dis Markers. 2013; 34(6):431-436.
- Gilbody S et al. Methylenetetrahydrofolate Reductase (MTHFR) Genetic Polymorphisms and Psychiatric Disorders: A HuGE Review. Am J Epidemiol. 2007; 165:1-13.
- Ho V et al. Effects of methionine synthase and methylenetetrahydrofolate reductase gene polymorphisms on markers of one-carbon metabolism. Genes Nutr. 2013; 8(6):571-80.
- Agata R. et al. The MAOA, COMT, MTHFR and ESR1 gene polymorphisms are associated with the risk of depression in menopausal women. Maturitas 84. 2016; 84:42–54.
- Van der Put NM et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998; 62(5):1044–51.
- Wagner C. Biochemical role of folate in cellular metabolism. In: Bailey LB, editor. Folate in health and disease. New York, NY: Marcel Dekker Inc.; 1995. p. 23–42.
- Bailey LB and JF Gregory III. Polymorphisms of Methylenetetrahydrofolate Reductase and Other Enzymes: Metabolic Significance, Risks and Impact on Folate Requirement. J Nutr. 1999; 129(5):919-22.
- Stover PJ. Polymorphisms in 1-Carbon Metabolism, Epigenetics and Folate-Related Pathologies. J. Nutrigenet Nutrigenomics. 2012; 4(5):293-305.
- Refsum H et al. The Hordaland Homocysteine Study: A Community-Based Study of Homocysteine, Its Determinants, and Associations with Disease. J Nutr. 2006; 136:1731S-1740S.
- Frosst P et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet. 1995; 10:111–113.
- Weisberg I et al. A second genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR) associated with decreased enzyme activity. Mol Genet Metab. 1998; 64:169–72.
- Teng Z. The 677C>T (rs1801133) polymorphism in the MTHFR gene contributes to colorectal cancer risk: a meta-analysis based on 71 research studies. PLoS One. 2013; 8(2):e55332.
- Yang L, et al. Impact of methylenetetrahydrofolate reductase (MTHFR) polymorphisms on methotrexate-induced toxicities in acute lymphoblastic leukemia: a meta-analysis. Tumor Biol. 2012; 33(5):1445–54.
- Bjelland I et al. Folate, Vitamin B12, Homocysteine, and the MTHFR 677CT Polymorphism in Anxiety and Depression: The Hordaland Homocysteine Study. Arch Gen Psychiatry. 2003; 60(6):618-626.
- Beydoun MA et al. Serum folate, vitamin B-12 and homocysteine and their association with depressive symptoms among US adults. Psychosom Med. 2010; 72(9):862-873.
- Nurk E et al. Plasma Total Homocysteine and Memory in the Elderly: The Hordaland Homocysteine Study. Ann Neurol. 2005; 58:847-857.
- Rajagopalan P et al. Common folate gene variant, MTHFR C677T, is associated with brain structure in two independent cohorts of people with mild cognitive impairment. Neuroimage Clin. 2012; 1(1):179-187.
- Lewis SJ et al. The thermolabile variant of MTHFR is associated with depression in the British Women’s Heart and Health Study and a meta-analysis. Molecular Psychiatry. 2006; 11:352-360.
- Steluti J et al. Genetic Variants Involved in One-Carbon Metabolism: Polymorphism Frequencies and Differences in Homocysteine Concentrations in the Folic Acid Fortification Era. Nutrients 2017; 9:539.
- Chen D, et al. Folate metabolism genetic polymorphisms and meningioma and glioma susceptibility in adults. Oncotarget 2017; 8(34): 57265-57277.
- Lim U et al. Gene-nutrient interactions among determinants of folate and one-carbon metabolism on the risk of non-Hodgkin lymphoma: NCI-SEER Case-Control Study. 2007; 109:3050-3059.
- Han X et al. Genetic variants and increased risk of meningioma: an updated meta-analysis. OncoTargets and Therapy. 2017; 10:1875–1888.
- Carr DF, O’meara H, Jorgensen AL, et al. SLCO1B1 genetic variant associated with statin-induced myopathy: a proof-of-concept study using the clinical practice research datalink. Clin Pharmacol Ther. 2013;94(6):695-701.
- Donnelly LA, Doney AS, Tavendale R, et al. Common nonsynonymous substitutions in SLCO1B1 predispose to statin intolerance in routinely treated individuals with type 2 diabetes: a go-DARTS study. Clin Pharmacol Ther. 2011;89(2):210-6.
- Niemi M et al. Organic Anionic Transporting Protein 1B1: a Genetically Polymorphic Transporter of Major Importance for Hepatic Drug Uptake. Pharm Review. 2011; 63(1):157-181.
- Ramsey LB et al. The Clinical Pharmacogenetics Implementation Consortium Guideline for SLCO1B1 and Simvastatin-Induced Myopathy: 2014 Update. Clin Pharmacol Ther. 2014; 96(4):423-428.
- Huang Y et al. Association between prediabetes and risk of cardiovascular disease and all-cause mortality: systematic review and meta-analysis. 2016; 355:i5953.
- Ma C. et al. Effects of weight loss interventions for adults who are obese on mortality, cardiovascular disease, and cancer: systematic review and meta-analysis. BMJ 2017; 359:j4849.
- Meikle J et al. Exercise in a Healthy Heart Program: A Cohort Study. Cardiology 2013; 7:145–151.
- Alessandro A et al. Mediterranean Diet and Cardiovascular Disease: A critical Evaluation of A Priori Dietary Indexes. 2015; 7:7863-7888.
- Akarolo-Anthony S et al. Plasma Magnesium and the Risk of Ischemic Stroke among Women. Stroke 2014; 45(10): 2881–2886.
- Lee S et el. The relationship between Magnesium and Endothelial Function in End-Stage Renal Disease Patients on Hemodialysis. Yonsei Med J 2016; 57(6):1446-1453.
- Ashor A et al. Effect of vitamin C and vitamin E supplementation on endothelial function: a systematic review and meta-analysis of randomized controlled trials. British Journal of Nutrition 2015; 113:1182–1194.
- Wilson A et al. Functionally Null Mutations in Patients with the cblG-Variant Form of Methionine Synthase Deficiency. Am. J. Hum. Genet. 1998; 63:409–414.
- Saita E et al. Anti-inflammatory Diet for Atherosclerosis and Coronary Artery Disease: Antioxidant Foods. Clinical Medicine Insights: Cardiology 2014; 8(S3) 61-65.
- Hosogoe N et al. Add-on Antiplatelet Effects of Eicosapentaenoic Acid with Tailored Dose Setting in Patients on Dual Antiplatelet Therapy. Int Heart J. 2017; 58(4): 481-485.
- Ho, H. T. et al. A systematic review and meta-analysis of randomized controlled trials of the effect of konjac glucomannan, a viscous soluble fiber, on LDL cholesterol and the new lipid targets non-HDL cholesterol and apolipoprotein B. Am J Clin Nutr. 2017; 105(5):1239-1247.
- Zelman, K. Fiber: How Much Do I Need? WebMD. 2011 https://www.webmd.com/diet/guide/fiber-how-much-do-you-need#1.
- Nordmann A et al. Effects of Low-Carbohydrate vs Low-fat Diets on Weight loss and Cardiovascular Risk Factors. Arch Intern Med. 2006; 166:285-293.
- Huang Y et al. Association between prediabetes and risk of cardiovascular disease and all-cause mortality: systematic review and meta-analysis. 2016; 355:i5953.
- Ferrucci L et al. Common Variation in the B-Carotene 15, 15’-Monooxygenase 1 Gene Affects Circulating Levels of Carotenoids: A Genome-wide Association Study. The American Journal of Human Genetics. 2009; 84, 123-133.
- Lietz G et al. Single Nucleotide Polymorphisms Upstream from the B-Carotene 15, 15’-Monoxygenase Gene Influence Provitamin A Conversion Efficiency in Female Volunteers. J. Nutr. 2012; 142(1):161S-165S
- Borel P et al. Genetic Variations Involved in Interindividual Variability in Carotenoid Status. Mol Nutr Food Res. 2012; 56(2): 228-40.
- Wyss A et al. Expression pattern and localization of β,β-carotene 15,15′-dioxygenase in different tissues. Biochem. J. 2001; 354:521–529.
- Li LH, Yin XY, Wu XH, et al. Serum 25(OH)D and vitamin D status in relation to VDR, GC and CYP2R1 variants in Chinese. Endocr J. 2014;61(2):133-41.
- Nissen J et al. Common Variants in CYP2R1 and GC Genes Predict Vitamin D Concentrations in Healthy Danish Children and Adults. PLoS One. 2014; 9(2):e89907.
- Cheng JB et al. Genetic evidence that the human CYP2R1 enzyme is a key vitamin D 25-hydroxylase. Proc Natl Acad Sci U S A. 2004; 101:7711–7715.
- Ahn J et al. Genome-wide association study of circulating vitamin D levels. Human Molecular Genetics. 2010; 19(13): 2739-2745.
- Harris HW et al. Supplementation might help patients with depression, seasonal mood disturbances. Current Psych. 2013; 12(4):19-25.
- Houssein-Nezhad A et al. Vitamin D for Health: A Global Perspective. Mayo Clin. Proc. 2013; 88(7):720-755.
- Holick MF and M Garabedian. Vitamin D: photobiology, metabolism, mechanism of action, and clinical applications. In: Favus MJ, ed. Primer on the metabolic bone diseases and disorders of mineral metabolism. 6th ed. Washington, DC: American Society for Bone and Mineral Research. 2006; 129-137.
- Lips P and NM van Schoor. The effect of vitamin D on bone and osteoporosis. Best Pract Res Clin Endocrinol Metab. 2011; 25:585–591.
- Wang T et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010; 376(9736):180-188.
- Bischoff-Ferrari HA et al. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012; 367:40–9.
- Grober U et al. Vitamin D: Update 2013: From rickets prophylaxis to general preventive healthcare. Dermato-Endocrinology. 2013; 5(3):331-47.
- Seppälä et al. Association between vitamin b12 levels and melancholic depressive symptoms: a Finnish population-based study. BMC Psychiatry 2013; 13:145.
- Tanaka T et al. Genome-wide Association Study of Vitamin B6, Vitamin B12, Folate, and Homocysteine Blood Concentrations. The American Journal of Human Genetics. 2009; 84:477-482.
- Hazra A et al. Common variants of FUT2 are associated with plasma vitamin B12 levels. Nat. Genet. 2008; 40:1160–1162.
- Kelly RJ et al. Sequence and expression of a candidate for the human Secretor blood group alpha (1,2)fucosyltransferase gene (FUT2). Homozygosity for an enzyme-inactivating nonsense mutation commonly correlates with the non-secretor phenotype. J Biol Chem. 1995; 270:4640–4649.
- Hazra A et al. Genome-wide significant predictors of metabolites in the one-carbon metabolism pathway. Hum Mol Gen. 2009; 18(23):4677-4687.
- Semmes BJ. Depression: a role for omega-3 fish oils and B vitamins? Evid. Based Integr. Med. 2005; 2:229–237.
- Tiemeier H et al. Vitamin B12, Folate, and Homocysteine in Depression: The Rotterdam Study. Am J Psychiatry. 2002; 159:2099-2101.
- Frankenburg, FR. The role of one-carbon metabolism in schizophrenia and depression. Harv. Rev. Psychiatry. 2007; 15:146–160.
- Wang W, Ingles SA, Torres-mejía G, et al. Genetic variants and non-genetic factors predict circulating vitamin D levels in Hispanic and non-Hispanic White women: the Breast Cancer Health Disparities Study. Int J Mol Epidemiol Genet. 2014;5(1):31-46.
- Nissen J et al. Common Variants in CYP2R1 and GC Genes Predict Vitamin D Concentrations in Healthy Danish Children and Adults. PLoS One. 2014; 9(2):e89907.
- Gozdzik A et al. Association of vitamin D binding protein (VDBP) polymorphisms and serum 25(OH)D concentrations in a sample of young Canadian adults of different ancestry. J Steroid Biochem Mol Biol. 2011; 127:405–412.
- Ahn J et al. Genome-wide association study of circulating vitamin D levels. Human Molecular Genetics. 2010; 19(13) 2739-2745.
- Foucan L et al. Polymorphisms in GC and NADSYN1 Genes are associated with vitamin D status and metabolic profile in non-diabetic adults. BMC Endocrine Disorders. 2013; 13:36.
- Wang T et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010; 376(9736):180-188.
- Grober U et al. Vitamin D: Update 2013: From rickets prophylaxis to general preventive healthcare. Dermato-Endocrinology. 2013; 5(3):331-47.
- Bischoff-Ferrari HA et al. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012; 367:40–9.
- Foucan L, Vélayoudom-céphise FL, Larifla L, et al. Polymorphisms in GC and NADSYN1 Genes are associated with vitamin D status and metabolic profile in Non-diabetic adults. BMC Endocr Disord. 2013;13:36.
- Wang T et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010; 376(9736):180-188.
- Hara N et al. Molecular identification of human glutamine- and ammonia-dependent NAD synthetases. Carbon-nitrogen hydrolase domain confers glutamine dependency. J. Biol. Chem. 2003; 278(13):10914-10921.
- Wassif CA et al. Mutations in the human sterol Δ7-reductase gene at 11q12–13 cause Smith–Lemli–Opitz syndrome. Am. J. Hum. Genet. 1998; 63:55–62.
- Harris HW et al. Supplementation might help patients with depression, seasonal mood disturbances. Current Psych. 2013; 12(4):19-25.
- Houssein-Nezhad A et al. Vitamin D for Health: A Global Perspective. Mayo Clinic. Proc. 2013; 88(7):720-755.
- Bischoff-Ferrari HA et al. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012; 367:40–9.
- Grober U et al. Vitamin D: Update 2013: From rickets prophylaxis to general preventive healthcare. Dermato-Endocrinology. 2013; 5(3):331-47.
- Benyamin B et al. Common variants in TMPRSS6 are associated with iron status and erythrocyte volume. Nature Genetics 2009; 41:1173-1175.
- Chambers JC et al. Genome-wide association study identifies variants in TMPRSS6 associated with hemoglobin levels. Nature Genetics 2009; 41:1170-1172.
- Stoltzfus RJ. Iron deficiency: global prevalence and consequences. Food Nutrition Bulletin. 2003; 24:S99–S103.
- Patel KV. Variability and heritability of hemoglobin concentration: an opportunity to improve understanding of anemia in older adults. Haematologica 2008; 93:1281-1283.
- Mccullough ML, Stevens VL, Diver WR, et al. Vitamin D pathway gene polymorphisms, diet, and risk of postmenopausal breast cancer: a nested case-control study. Breast Cancer Res. 2007;9(1):R9. Morris HA. Vitamin D Activities for Health Outcomes. Ann Lab Med 2014; 34:181-186.
- Palomba S et al. BsmI vitamin D receptor genotypes influence the efficacy of antiresorptive treatments in postmenopausal osteoporotic women. A 1-year multicenter, randomized and controlled trial. Osteoporosis Int. 2005; 16(8):943-52.
- Jia F, Sun RF, Li QH, et al. Vitamin D receptor BsmI polymorphism and osteoporosis risk: a meta-analysis from 26 studies. Genet Test Mol Biomarkers. 2013;17(1):30-4.
- Turner AG. Vitamin D and bone health. Scand J Clin Lab Invest Suppl. 2012; 243:65-72.
- Jia F et al. Vitamin D receptor BsmI polymorphism and osteoporosis risk: a meta-analysis from 26 studies. Genet Test Mol Biomarkers. 2013; 17(1):30-4.
- Ji GR. BsmI, TaqI, ApaI and FokI polymorphisms in the vitamin D receptor (VDR) gene and risk of fracture in Caucasians: a meta-analysis. Bone. 2010; 47(3):681-6.
- Vaughan-Shaw PG et al. The impact of vitamin D pathway genetic variation and circulating 25-hydroxyvitamin D on cancer outcome: Systemic review and meta-analysis. Br. J. Cancer 2017; 116:1092-1110.
- Deuster E et al. Vitamin D and VDR in Gynecological Cancers – A Systematic Review. Intl J Mol Sci 2017; 18:2328-2340.
- Gnagnarella P et al. Vitamin D receptor polymorphism FokI and cancer risk. A comprehensive meta-analysis. Carcinogenesis. 2014; 35:1913-1919.
- Laczmanski L et al. Association of the vitamin D receptor FokI gene polymorphism with sex- and non-sex-associated cancers: A meta-analysis. Tumor Biol 2017; 39(10):1-8.
- Mun M, Kim TH, Hwang JY, Jang WC. Vitamin D receptor gene polymorphisms and the risk for female reproductive cancers. Maturitas. 2015; 81(2):256-65.
- Muscogiuri G et al. Mechanisms in Endocrinology: Vitamin D as a potential contributor in endocrine health and disease. Eur J Endocrinol 2014; 171(3):R101-R110.
- Bagheri M et al. Vitamin D Receptor TaqI Gene Variant in Exon 9 and Polycystic Ovary Syndrome Risk. Int J Fertil Steril. 2013; 7(2):116-121.
- Reis G et al. Vitamin D receptor polymorphisms and the polycystic ovary syndrome: A systematic review. J. Obstet. Gynaecol Res 2017; 43(3):436-446.
- El-Shal AS et al. Genetic variation in the vitamin D receptor gene and vitamin D serum levels in Egyptian women with polycystic ovary syndrome. Mol Biol Rep 2013; 40(11):6063.
- Pal L et al. Vitamin D Status Relates to Reproductive Outcome in Women with Polycystic Ovary Syndrome: Secondary Analysis of a Multicenter Randomized Controlled Trial. J Clin Endocrinol Metab. 2016; 101(8):3027–3035.
- Mahley RW et al. Apolipoprotein E: Far More Than a Lipid Transport Protein. Annu Rev Genomics Hum Genet. 2000; 1:507-537.
- Eichner JE et al. Apolipoprotein E Polymorphism and Cardiovascular Disease: A HuGE Review. Am J Epidemiol. 2002; 155(6):487-495.
- Villeneuve S et al. The potential applications of Apolipoprotein E in personalized medicine. Front Aging Neurosci. 2014; 6:154.
- Howard BV et al. Association of Apolipoprotein E Phenotype with Plasma Lipoproteins in African-American and White Young Adults. Am J Epidemiol. 1998; 148(9):859-868.
- Yassine H. et al. DHA brain uptake and APOE4 status: a PET study with (1-11C)-DHA. Alzheimer’s Research & Therapy 2017; 9:23.
- Yassine H et al. Association of Docosahexaenoic Acid Supplementation With Alzheimer Disease Stage in Apolipoprotein E e4 Carriers: A Review. JAMA Neurol. 2017 March 01; 74(3): 339–347.
- Enoch MA, Xu K, Ferro E, Harris CR, Goldman D. Psychiatr Genet. 2003;13(1):33-41.
- Smolka MN, Schumann G, Wrase J, et al. Catechol-O-methyltransferase val158met genotype affects processing of emotional stimuli in the amygdala and prefrontal cortex. J Neurosci. 2005;25(4):836-42.
- Lacerda-pinheiro SF, Pinheiro junior RF, Pereira de lima MA, et al. Are there depression and anxiety genetic markers and mutations? A systematic review. J Affect Disord. 2014;168:387-98.
- Mier D et al. Neural substrates of pleiotropic action of genetic variation in COMT: a meta-analysis. Molecular Psychiatry 2010; 15:918-927.
- Agata R. et al. The MAOA, COMT, MTHFR and ESR1 gene polymorphisms are associated with the risk of depression in menopausal women. Maturitas 2016; 84:42–54.
- Crooke P et al. Estrogens, Enzyme Variants, and Breast Cancer: A Risk Model. Cancer Epidemiol Biomarkers Prev 2006; 15(9):1620-9.
- Lachman H et al. Human catechol-O-methyltransferase pharmacogenetics: description of a functional polymorphism and its potential application to neuropsychiatric disorders. Pharmacogenetics 1996; 6:243-250.
- Weinshilboum R et al. Methylation Pharmacogenetics: Catechol-O methyltransferase, Thiopurine Methyltransferase, and Histamine N-Methyltransferase. Annu. Rev. Pharmacol. Toxicol. 1999; 39:19-52.
- Mannisto P and S Kaakkola. Catechol-O-methyltransferase (COMT): Biochemistry, Molecular Biology, Pharmacology, and Clinical Efficacy of the New Selective COMT Inhibitors. Pharm Rev. 1999; 51(4):594-622.
- Dawling S et al. Catechol-O-Methyltransferase (COMT)-mediated Metabolism of Catechol Estrogens: Comparison of Wild-Type and Variant COMT Isoforms. Cancer Res. 2001; 61:6716-6722.
- Goldman D et al. The Genetics of Addictions: Uncovering the Genes. Nat Rev Genet. 2005; 6(7):521-532.
- Yuferov V et al. Search for Genetic Markers and Functional Variants Involved in the Development of Opiate and Cocaine Addiction, and Treatment. Ann N Y Acad Sci. 2010; 1187:184-207.
- Schellekens AF et al. COMT Val158Met modulates the effect of childhood adverse experiences on the risk of alcohol dependence. Addict Biol. 2013; 18(2):344-356.
- Bhakta SG et al. The COMT Met158 allele and violence in schizophrenia: a meta-analysis. Schizophr Res. 2012; 140(1-3):192-197.
- Godar SC and M Bortolato. Gene-sex interactions in schizophrenia: focus on dopamine neurotransmission. Front Behav Neurosci. 2014; 8:71.
- Pooley EC et al. The met158 allele of catechol-o-methyltransferase (COMT) is associated with obsessive-compulsive disorder in men: case-control study and meta-analysis. Mol Psych. 2007; 12:556-551.
- Konishi Y et al. Genexgenexgender interaction of BDNF and COMT genotypes associated with panic disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2014; 51:119-125.
- Kolassa IT et al. The risk of posttraumatic stress disorder after trauma depends on traumatic load and the catechol-o-methyltransferase Val(158)Met polymorphism. Biol Psychiatry. 2010; 67(4):304-308.
- Lee SY et al. COMT and BDNF interacted in bipolar II disorder not comorbid with anxiety disorder. Behav Brain Res. 2013; 237:243-248.
- Zhang Z. The Val/Met functional polymorphism in COMT confers susceptibility to bipolar disorder: evidence from an association study and a meta-analysis. J Neural Transm. 2009; 116(10):1193-200.
- Janicki PK. Pharmacogenetics of Pain Management. Comprehensive Treatment of Chronic Pain by Medical, Interventional, and Integrative Approaches. Edited by TR Deers. American Academy of Pain Medicine. 2013.
- Zubieta JK et al. COMT val158met Genotype Affects mu-opioid Neurotransmitter Responses to a Pain Stressor. Science. 2003; 299(5610):1240-1243.
- Klepstad P et al. Genetic variability and clinical efficacy of morphine. Acta Anasthesiol Scand. 2005; 49:902-908.
- Mannisto PT and S Kaakkola. Catechol-O-methyltransferase (COMT): Biochemistry, Molecular Biology, Pharmacology, and Clinical Efficacy of the New Selective COMT Inhibitors. Pharm Rev. 1999; 51(4):594-622.
- Corvol JC et al. The COMT Val158Met polymorphism affects the response to entacapone in Parkinson’s disease: a randomized crossover clinical trial. Ann Neurol. 2011; 69(1):111-118.
- Ball P and R Knuppen. Catecholoestrogens (2-and 4-hydroxyoestrogens): chemistry, biogenesis, metabolism, occurrence and physiological significance. Acta Endocrinol. Suppl. 1980; 232:1-127.
- Lakhani NJ et al. 2-Methoxyestradiol, a Promising Anticancer Agent. Pharmacotherapy. 2003; 23:165-172.
- Lavigne JA et al. The Effects of Catechol-O-Methyltransferase Inhibition on Estrogen Metabolite and Oxidative DNBA Damage Levels in Estradiol-treated MCF-7 Cells. Cancer Research. 2001; 61:7488-7494.
- Worda C et al. Influence of the catechol-O-methyltransferase (COMT) codon 158 polymorphism on estrogen levels in women. Human Reproduction. 2003; 18(2):262-266.
- Eriksson AL et al. The COMT val158met polymorphism Is Associated with Early Pubertal Development, Height and Cortical Bone Mass in Girls. Pediatr Res. 2005; 58(1):71-77.
- Masurier M et al. Effect of Acute Tyrosine Depletion in Using a Branched Chain Amino-Acid Mixture on Dopamine Neurotransmission in the Rat Brain. Neuropsychopharmacology. 2006; 31(2):310-317.
- Sarris J et al. S-adenosyl methionine (SAMe) versus escitalopram and placebo in major depression RCT: Efficacy and effects of histamine and carnitine as moderators of response. J Affect Disord. 2014; 164:76-81.
- Kennedy D et al. Effects of high-dose B vitamin complex with vitamin C and minerals on subjective mood and performance in healthy males. Psychopharmacology (Berl). 2010; 211(1):55-68.
- Li Y et al. Functional and structural comparisons of cysteine residues in the Val108 wild type and Met108 variant of human soluble catechol O-methyltransferase. Chem Biol Interact. 2005; 152(2-3):151-163.
- Fava M et al. Rapidity of onset of the antidepressant effect of parenteral S-adenosyl-l-methionine. Psychiatry Res. 1995; 56(3):295-297.
- Jeffery DR and JA Roth. Kinetic reaction mechanism for magnesium binding to membrane-bound and soluble catechol O-methyltransferase. Biochem. 1987; 26(10):2955-2958.
- Sowa-Kucma M et al. Zinc, magnesium and NMDA receptor alterations in the hippocampus of suicide victims. J Affect Disord. 2013; 151(3):924-931.
- Basheer MP et al. A study of serum magnesium, calcium and phosphorus level, and cognition in the elderly population of South India. Alexandria J Med. 2016; 52(4):303-308.
- Yary T et al. Dietary magnesium intake and the incidence of depression: A 20 year follow-up study. J Affect Disord. 2016; 193:94-98.
- Haan MN et al. Homocysteine, B vitamins, and the incidence of dementia and cognitive impairment: Results from the Sacramento Area Latino Study on Aging. Am J Clin Nutr. 2007; 85(2):511-517.
- Smith AD et al. Homocysteine-lowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: A randomized controlled trial. PLoS One. 2010; 5(9):1-10.
- Mitchell ES et al. B vitamin polymorphisms and behavior: Evidence of associations with neurodevelopment, depression, schizophrenia, bipolar disorder and cognitive decline. Neurosci Biobehav Rev. 2014; 47:307-320.
- Hiroi T et al. Dopamine formation from tyramine by CYP2D6. Biochem Biophys Res Commun. 1998; 249(3):838-843.
- Zhu ZT et al. Tyramine excites rat subthalamic neurons in vitro by a dopamine-dependent mechanism. Neuropharmacology. 2007; 52(4):1169-1178.
- Burchett SA et al. The mysterious trace amines: Protean neuromodulators of synaptic transmission in mammalian brain. Prog Neurobiol. 2006; 79:223-246.
- Papaleo F et al. Sex-dichotomous effects of functional COMT genetic variations on cognitive functions disappear after menopause in both health and schizophrenia. Eur Neuropsychopharmacol. 2015; 25(12):2349-2363.
- Almey A et al. Estrogen receptors in the central nervous system and their implication for dopamine-dependent cognition in females. Horm Behav. 2015; 74:125-138.
- McCann S et al. Changes in 2-hydroxyestrone and 16α-hydroxyestrone metabolism with flaxseed consumption: Modification by COMT and CYP1B1 genotype. Cancer Epidemiol Biomarkers Prev. 2007; 16(2):256-262.
- Rižner TL. Estrogen biosynthesis, phase I and phase II metabolism, and action in endometrial cancer. Mol Cell Endocrinol. 2013; 381(1-2):124-139.
- Masurier M et al. Effect of Acute Tyrosine Depletion in Using a Branched Chain Amino-Acid Mixture on Dopamine Neurotransmission in the Rat Brain. 2006; 31(2):310-317.
- Fernstrom HD and MH Fernstrom. Tyrosine, Phenylalanine, and Catecholamine Synthesis and Function in the Brain. J. Nutr. 2007; 137(6):1539S-1547S.
- Reus G et al. Kynurenine pathway dysfunction in the pathophysiology and treatment of depression: Evidences from animal and human studies. J Psychiatr Res. 2015; 68:316-328.
- Jangid P et al. Comparative study of efficacy of l-5-hydroxytryptophan and fluoxetine in patients presenting with first depressive episode. Asian J Psychiatr. 2013; 6(1):29-34.
- Lowe S et al. L-5-Hydroxytryptophan augments the neuroendocrine response to a SSRI. Psychoneuroendocrinology. 2006; 31(4):473-484.
- Lardner A et al. Neurobiological effects of the green tea constituent theanine and its potential role in the treatment of psychiatric and neurodegenerative disorders. Nutritional Neuroscience. 2014; 17(4):145-155.
- Mu W et al. An overview of biological production of L-theanine. Biotechnol Adv. 2015; 33(3-4):335-342.
- Kakuda T. Neuroprotective effects of theanine and its preventive effects on cognitive dysfunction. Pharmacol Res. 2011; 64(2):162-168.
- Tian X et al. Protective effect of l-theanine on chronic restraint stress-induced cognitive impairments in mice. Brain Res. 2013; 1503:24-32.
- Martínez-Banaclocha M et al. N-acetyl-cysteine in the treatment of Parkinson’s disease. What are we waiting for? Med Hypotheses. 2012; 79(1):8-12.
- Dean O et al. N-acetyl cysteine restores brain glutathione loss in combined 2-cyclohexene-1-one and d-amphetamine-treated rats: Relevance to schizophrenia and bipolar disorder. Neurosci Lett. 2011; 499(3):149-153.
- Botsakis K et al. 17β-Estradiol/N-acetylcysteine interaction enhances the neuroprotective effect on dopaminergic neurons in the weaver model of dopamine deficiency. Neuroscience. 2016; 320:221-229.
- Tunbridge E et al. Polymorphisms in the catechol‐O‐methyltransferase (COMT) gene influence plasma total homocysteine levels. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics 147.6 (2008):996-999.
- Paul R et al. The potential physiological crosstalk and interrelationship between two sovereign endogenous amines, melatonin and homocysteine. Life Sci. 2015; 139:97-107.
- Hursel R et al. The Role of Catechol-o-Methyl Transferase Val (108/158) MET Polymorphism (rs4680) in the effect of Green Tea on Resting Energy Expenditure and Fat Oxidation: A Pilot Study. 2014; 9(9): e106220.
- Lorenz M et al. The activity of catechol-O-methyltransferase (COMT) is not impaired by high doses of epigallocatechin-3-gallate (EGCG) in vivo. Eur J Pharmacol. 2014; 740: 645-651.
- Kang K et al. Beneficial effects of natural phenolics on levodopa methylation and oxidative neurodegeneration. Brain Res. 2013; 1497:1-14.
- Kang K et al. Dual beneficial effects of (-)-epigallocatechin-3-gallate on levodopa methylation and hippocampal neurodegeneration: In vitro and in vivo studies. PLoS One. 2010; 5(8): e11951.
- Xie X et al. Adenosine and dopamine receptor interactions in striatum and caffeine-induced behavioral activation. Comp Med. 2007; 57(6):538-545.
- Witte A et al. COMT Val158Met polymorphism modulates cognitive effects of dietary intervention. Front Aging Neurosci. 2010; 2:146.
- Voelcker-Rehage C et al. COMT gene polymorphisms, cognitive performance, and physical fitness in older adults. Psychol Sport Exerc. 2015; 20:20-28.
- Zhu BT et al. Effects of tea polyphenols and flavonoids on liver microsomal glucuronidation of estradiol and estrone. J Steroid Biochem Mol Biol. 1998; 64(3-4):207-215.
- Moon YJ et al. Dietary flavonoids: Effects on xenobiotic and carcinogen metabolism. Toxicol Vitr. 2006; 20 (2): 187-210.
- Ullah N et al. Green tea phytocompounds as anticancer: A review. Asian Pacific J Trop Dis. 2016; 6(4):330-336.
- Wang L et al. CYP1A1 lle462Val Polymorphism Is Associated with Cervical Cancer Risk in Caucasians Not Asians: A Meta-Analysis. Front. Physiol. 2017; 8:1081.
- Wu B et al. Mspl and lle462Val Polymorphisms in CYP1A1 and Overall Cancer Risk: A Meta-Analysis. PLOS ONE 2013; 8(12): e85166.
- Crooke P et al. Estrogens, Enzyme Variants, and Breast Cancer: A Risk Model. Cancer Epidemiol Biomarkers Prev. 2006; 15(9).
- Wang Y et al. The association of the CYP1A1 lle462Val polymorphism with head and neck cancer risk: evidence based on a cumulative meta-analysis. OncoTargets and Therapy 2016; 9 2927–2934.
- Zeng W et al. CYP1A1 rs1048943 and rs4646903 polymorphisms associated with laryngeal cancer susceptibility among Asian populations: a meta-analysis. J. Cell. Mol. Med. 2016; 20 (2): 287-293.
- Zhu X et al. Associations between CYP1A1 rs1048943 A>G and rs4646903. T>C genetic variations and colorectal cancer risk: Proof from 26 case-control studies. Oncotarget 2016; 7 (32):51365-51374.
- Lu J et al. Genetic polymorphisms of CYP1A1 and risk of leukemia: a met-analysis. OncoTargets and Therapy 2015; 8: 2883–2902.
- Ji YN et al. CYP1A1 Ile462Val Polymorphism Contributes to Lung Cancer Susceptibility among Lung Squamous Carcinoma and Smokers: A Meta-Analysis. PLoS ONE 2012; 7(8): e43397.
- Wright C et al. Genetic association study of CYP1A1 polymorphisms identifies risk haplotypes in non-small cell lung cancer. Eur Respir J 2010; 35: 152–159.
- Koller DL et al. Meta-analysis of genome-wide studies identifies WNT16 and ESR1 snps associated with bone mineral density in premenopausal women. J Bone Miner Res. 2013; 28(3):547–558.
- Styrkarsdottir U et al. Multiple Genetic Loci for Bone Mineral Density and Fractures. N Engl J Med 2008; 358:2355-65.
- Loannidis JP et al. Differential genetic effects of ESR1 gene polymorphisms on osteoporosis outcomes. JAMA. 2004; 292(17):2105–2114.
- Agata R et al. The MAOA, COMT, MTHFR and ESR1 gene polymorphisms are associated with the risk of depression in menopausal women. Maturitas 2016; 84:42–54.
- Li WF et al. Genetics of Osteoporosis: Perspectives for Personalized Medicine. Personalized Medicine 2010; 7(6):655-668.
- Raisz LG. Pathogenesis of osteoporosis: concepts, conflicts, and prospects J. Clin. Invest. 2005; 115:3318–3325.
- Falahati-Nini A et al. Relative contributions of testosterone and estrogen in regulating bone resorption and formation in normal elderly men. J. Clin. Invest. 2000; 106:1553–1560.
- Luo L et al. Association of ESR1 and C6orf97 gene polymorphism with osteoporosis in postmenopausal women. Mol Biol Rep. 2014; 41(5):3235-43.
- Wang K et al. Association of estrogen receptor alpha gene polymorphisms with bone mineral density: a meta-analysis. Chinese Med. Journal 2012; 125(14):2589-2597.
- Wedren S et al. Estrogen receptor alpha gene polymorphism and endometrial cancer risk- a case-control study. BMC Cancer 2008; 8:322.
- Martinaityte I et al. Bone mineral density is associated with vitamin D related rs6013897 and estrogen receptor polymorphism rs4870044: The Tromsø study. PLoS ONE 2017; 12(3):e0173045.
- Sheng et al. Prognostic role of methylated GSTP1, p16, ESR1 and PITX2 in patients with breast cancer. Medicine 2017; 96:28.
- Lee Y et al. Estrogen receptor 1 PvuII and XbaI polymorphisms and susceptibility to Alzheimer’s disease: a meta-analysis. Genet. Mol. Res. 2015; 14 (3):9361-9369.
- Zhou X et al. Eight Functional Polymorphisms in the Estrogen Receptor 1 Gene and Endometrial Cancer Risk: A Meta-Analysis. PLoS ONE 2013; 8(4):e60851.
- Zhang J, A cis-phase interaction study of genetic variants within the MAOA gene in major depressive disorder. Biol Psychiatry 2010; 68(9):795-800.
- Pinsonneault JK, Papp AC, Sadee W. Allelic mRNA expression of X-linked monoamine oxidase a (MAOA) in human brain: dissection of epigenetic and genetic factors. Human Molecular Genetics. 2006; 15(17):2636-2649.
- Leuchter AF, McCracken JT, et al. Monoamine oxidase a and catechol-o-methyltransferase functional polymorphisms and the placebo response in major depressive disorder. J Clin Psychopharmacol. 2009; 29(4):372-7.
- Estrada K et al. Genome-wide meta-analysis identifies 56 bone mineral density loci and reveals 14 loci associated with risk of fracture. Nat Genet. 2012; 44(5):491–501.
- Koller et al. Meta-analysis of genome-wide studies identifies WNT16 and ESR1 snps associated with bone mineral density in premenopausal women. J Bone Miner Res. 2013; 28(3):547–558.
- Kim et al. Wnt signaling in bone formation and its therapeutic potential for bone diseases. Ther Adv Musculoskel Dis. 2013; 5(1):13–31.
- Monroe D et al. Update on Wnt signaling in bone cell biology and bone disease. Gene 2012; 492:1–18.
- Medina-Gomez et al. Meta-Analysis of Genome-Wide Scans for Total Body BMD in Children and Adults Reveals Allelic Heterogeneity and Age-Specific Effects at the WNT16 Locus. PLOS Genet. 2012; 8(7):e1002718.
- Yasko, A. 2005. Genetic ByPass, Using Nutrition to Bypass Genetic Mutations. Matrix Development Publishing.
- London, M. CBS Upregulation, Myth or Reality? MIT- Massachusetts Institute of Technology. http://web.mit.edu/london/www/cbs.html.
- McEvoy M. March 27 2013. Metabolic Gateways: CBS Mutations & Glutathione. Metabolic Healing Empowering Your Health. https://metabolichealing.com/metabolic-gateways-cbs-gene-mutations-glutathione.
- Wang L et al. Modulation of Cystathionine β-Synthase Levels Regulates Total Serum Homocysteine in Mice. American Heart Association Journal. 2004; 94:1318-1324.
- Aras O et al. Influence of 699C-T and 1080C-T Polymorphisms of the Cystathionine β-synthase gene on Plasma Homocysteine Levels. Clin Gen. 2000; 58:455-459.
- Yasko, A. 2009. Autism: Pathways To Recovery. Neurological Research Institute, LLC.
- Stipanuk, M. and I. Ueki. Dealing with methionine/homocysteine sulfur: cysteine metabolism to taurine and inorganic sulfur. J Inherit Metab Dis. 2011; 34(1):17-32.
- Rumbeiha, W. et al. Acute hydrogen sulfide-induced neuropathology and neurological sequelae: Challenges for translational neuroprotective research. Ann. N.Y. Acad. Sci. 2016; 1378:5-16.
- Ingenbleek, Y. and H. Kimura. Nutritional essentiality of sulfur in health and disease. Nutrition Reviews. 2013; 71(7):413-432.
- Alkaitis M et al. Recoupling the Cardiac Nitric Oxide Synthases: Tetrahydrobiopterin Synthesis and Recycling. Curr Heart Fail Rep. 2012; 9:200–210.
- Xie X et al. Endothelial nitric oxide synthase gene single nucleotide polymorphisms and the risk of hypertension: A meta-analysis involving 63,258 subjects. Clinical and Experimental Hypertension. 2017; 39(2):175-182.
- Shengyuan Liu et al. The nitric oxide synthase 3 G894T polymorphism associated with Alzheimer’s disease risk: a meta-analysis. Sci Rep. 2015; 5:13598.
- Marsden PA et al. Structure and chromosomal localization of the human constitutive endothelial nitric oxide synthase gene. J Biol Chem. 1992; 68:17478-17488.
- Cosentino F and Luscher TF. Maintenance of vascular integrity: role of nitric oxide and other bradykinin mediators. Eur Heart J. 1995; 16 Suppl K:4-12.
- Huang PL. eNOS, metabolic syndrome and cardiovascular disease. Trends Endocrinol Metab. 2009; 20(6):295-302.
- Hogg N et al. Inhibition of low-density lipoprotein oxidation by nitric oxide. Potential role in atherogenesis. FEBS Lett. 1993; 334(2):170-174.
- Radomski MW et al. Endogenous nitric oxide inhibits human platelet adhesion to vascular endothelium. Lancet. 1987; 2:1057-1058.
- Tesauro M et al. Intracellular processing of endothelial nitric oxide synthase isoforms associated with differences in severity of cardiopulmonary diseases: cleavage of proteins with aspartate vs. glutamate at position 298. Proc Natl Acad Sci U S A. 2000; 6:2832-2835.
- Casas JP et al. Endothelial Nitric Oxide Synthase Genotype and Ischemic Heart Disease: Meta-Analysis of 26 Studies Involving 23,028 Subjects. Circulation. 2004; 109:1359-1365.
- Niu W and Y Qi. An Updated Meta-Analysis of Endothelial Nitric Oxide Synthase Gene: Three Well-Characterized Polymorphisms with Hypertension. Plos One. 2011; 6(9):e24266.
- Wang M et al. Association of G894T polymorphism in endothelial nitric oxide synthase gene with the risk of ischemic stroke: A meta-analysis. Biomed Rep. 2013; 1(1):144-150.
- Zigra AM et al. eNOS gene variants and the risk of premature myocardial infarction. Dis Markers. 2013; 34(6):431-436.
- Agata R. et al. The MAOA, COMT, MTHFR and ESR1 gene polymorphisms are associated with the risk of depression in menopausal women. Maturitas 84. 2016; 84:42–54.
- Van der Put NM et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998; 62(5):1044–51.
- Wagner C. Biochemical role of folate in cellular metabolism. In: Bailey LB, editor. Folate in health and disease. New York, NY: Marcel Dekker Inc.; 1995. p. 23–42.
- Bailey LB and JF Gregory III. Polymorphisms of Methylenetetrahydrofolate Reductase and Other Enzymes: Metabolic Significance, Risks and Impact on Folate Requirement. J Nutr. 1999; 129(5):919-22.
- Stover PJ. Polymorphisms in 1-Carbon Metabolism, Epigenetics and Folate-Related Pathologies. J. Nutrigenet Nutrigenomics. 2012; 4(5):293-305.
- Refsum H et al. The Hordaland Homocysteine Study: A Community-Based Study of Homocysteine, Its Determinants, and Associations with Disease. J Nutr. 2006; 136:1731S-1740S.
- Frosst P et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet. 1995; 10:111–113.
- Weisberg I et al. A second genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR) associated with decreased enzyme activity. Mol Genet Metab. 1998; 64:169–72.
- Teng Z. The 677C>T (rs1801133) polymorphism in the MTHFR gene contributes to colorectal cancer risk: a meta-analysis based on 71 research studies. PLoS One. 2013; 8(2):e55332.
- Yang L, et al. Impact of methylenetetrahydrofolate reductase (MTHFR) polymorphisms on methotrexate-induced toxicities in acute lymphoblastic leukemia: a meta-analysis. Tumor Biol. 2012; 33(5):1445–54.
- Bjelland I et al. Folate, Vitamin B12, Homocysteine, and the MTHFR 677CT Polymorphism in Anxiety and Depression: The Hordaland Homocysteine Study. Arch Gen Psychiatry. 2003; 60(6):618-626.
- Beydoun MA et al. Serum folate, vitamin B-12 and homocysteine and their association with depressive symptoms among US adults. Psychosom Med. 2010; 72(9):862-873.
- Nurk E et al. Plasma Total Homocysteine and Memory in the Elderly: The Hordaland Homocysteine Study. Ann Neurol. 2005; 58:847-857.
- Rajagopalan P et al. Common folate gene variant, MTHFR C677T, is associated with brain structure in two independent cohorts of people with mild cognitive impairment. Neuroimage Clin. 2012; 1(1):179-187.
- Gilbody S et al. Methylenetetrahydrofolate Reductase (MTHFR) Genetic Polymorphisms and Psychiatric Disorders: A HuGE Review. Am J Epidemiol. 2007; 165:1-13.
- Lewis SJ et al. The thermolabile variant of MTHFR is associated with depression in the British Women’s Heart and Health Study and a meta-analysis. Molecular Psychiatry. 2006; 11:352-360.
- Steluti J et al. Genetic Variants Involved in One-Carbon Metabolism: Polymorphism Frequencies and Differences in Homocysteine Concentrations in the Folic Acid Fortification Era. Nutrients 2017; 9:539.
- Chen D, et al. Folate metabolism genetic polymorphisms and meningioma and glioma susceptibility in adults. Oncotarget 2017; 8(34): 57265-57277.
- Lim U et al. Gene-nutrient interactions among determinants of folate and one-carbon metabolism on the risk of non-Hodgkin lymphoma: NCI-SEER Case-Control Study. 2007; 109:3050-3059.
- Han X et al. Genetic variants and increased risk of meningioma: an updated meta-analysis. OncoTargets and Therapy. 2017; 10:1875–1888.
- Steluti J et al. Genetic Variants Involved in One-Carbon Metabolism: Polymorphism Frequencies and Differences in Homocysteine Concentrations in the Folic Acid Fortification Era. Nutrients 2017; 9(6):539.
- Li WX et al. Homocysteine Metabolism Gene Polymorphisms (MTHFR C677T, MTHFR A1298C, MTR A2756G and MTRR A66G) Jointly Elevate the Risk of Folate Deficiency. Nutrients 2015; 7(8):6670-6687.
- Ho V et al. Effects of methionine synthase and methylenetetrahydrofolate reductase gene polymorphisms on markers of one-carbon metabolism. Genes Nutr. 2013; 8(6):571-80.
- Wang P et al. Association of MTRR A66G polymorphism with cancer susceptibility: Evidence from 85 studies. J Cancer. 2017; 8(2):266-277.
- Han D et al. Methionine synthase reductase A66G polymorphism contributes to tumor susceptibility: evidence from 35 case-control studies. Mol Biol Rep. 2012; 39(2):805-816.
- Clark S and J Melki. DNA methylation and gene silencing in cancer: which is the guilty party? Oncogene. 2002; 21(35):5380-5387.
- Zeng XT et al. Association of methionine synthase rs1801394 and methionine synthase reductase rs1805087 polymorphisms with meningioma in adults: A meta-analysis. BioMed Rep. 2014; 2(3):432-436.
COMT
- Agata R. et al. The MAOA, COMT, MTHFR and ESR1 gene polymorphisms are associated with the risk of depression in menopausal women. Maturitas 2016; 84:42–54.
- Crooke P et al. Estrogens, Enzyme Variants, and Breast Cancer: A Risk Model. Cancer Epidemiol Biomarkers Prev 2006; 15(9):1620-9.
- Lachman H et al. Human catechol-O-methyltransferase pharmacogenetics: description of a functional polymorphism and its potential application to neuropsychiatric disorders. Pharmacogenetics 1996; 6:243-250.
- Weinshilboum R et al. Methylation Pharmacogenetics: Catechol-O methyltransferase, Thiopurine Methyltransferase, and Histamine N-Methyltransferase. Annu. Rev. Pharmacol. Toxicol. 1999; 39:19-52.
- Mannisto P and S Kaakkola. Catechol-O-methyltransferase (COMT): Biochemistry, Molecular Biology, Pharmacology, and Clinical Efficacy of the New Selective COMT Inhibitors. Pharm Rev. 1999; 51(4):594-622.
- Dawling S et al. Catechol-O-Methyltransferase (COMT)-mediated Metabolism of Catechol Estrogens: Comparison of Wild-Type and Variant COMT Isoforms. Cancer Res. 2001; 61:6716-6722.
- Mier D et al. Neural substrates of pleiotropic action of genetic variation in COMT: a meta-analysis. Molecular Psychiatry 2010; 15:918-927.
- Goldman D et al. The Genetics of Addictions: Uncovering the Genes. Nat Rev Genet. 2005; 6(7):521-532.
- Yuferov V et al. Search for Genetic Markers and Functional Variants Involved in the Development of Opiate and Cocaine Addiction, and Treatment. Ann N Y Acad Sci. 2010; 1187:184-207.
- Schellekens AF et al. COMT Val158Met modulates the effect of childhood adverse experiences on the risk of alcohol dependence. Addict Biol. 2013; 18(2):344-356.
- Bhakta SG et al. The COMT Met158 allele and violence in schizophrenia: a meta-analysis. Schizophr Res. 2012; 140(1-3):192-197.
- Godar SC and M Bortolato. Gene-sex interactions in schizophrenia: focus on dopamine neurotransmission. Front Behav Neurosci. 2014; 8:71.
- Pooley EC et al. The met158 allele of catechol-o-methyltransferase (COMT) is associated with obsessive-compulsive disorder in men: case-control study and meta-analysis. Mol Psych. 2007; 12:556-551.
- Konishi Y et al. Genexgenexgender interaction of BDNF and COMT genotypes associated with panic disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2014; 51:119-125.
- Kolassa IT et al. The risk of posttraumatic stress disorder after trauma depends on traumatic load and the catechol-o-methyltransferase Val(158)Met polymorphism. Biol Psychiatry. 2010; 67(4):304-308.
- Lee SY et al. COMT and BDNF interacted in bipolar II disorder not comorbid with anxiety disorder. Behav Brain Res. 2013; 237:243-248.
- Zhang Z. The Val/Met functional polymorphism in COMT confers susceptibility to bipolar disorder: evidence from an association study and a meta-analysis. J Neural Transm. 2009; 116(10):1193-200.
- Janicki PK. Pharmacogenetics of Pain Management. Comprehensive Treatment of Chronic Pain by Medical, Interventional, and Integrative Approaches. Edited by TR Deers. American Academy of Pain Medicine. 2013.
- Zubieta JK et al. COMT val158met Genotype Affects mu-opioid Neurotransmitter Responses to a Pain Stressor. Science. 2003; 299(5610):1240-1243.
- Klepstad P et al. Genetic variability and clinical efficacy of morphine. Acta Anasthesiol Scand. 2005; 49:902-908.
- Mannisto PT and S Kaakkola. Catechol-O-methyltransferase (COMT): Biochemistry, Molecular Biology, Pharmacology, and Clinical Efficacy of the New Selective COMT Inhibitors. Pharm Rev. 1999; 51(4):594-622.
- Corvol JC et al. The COMT Val158Met polymorphism affects the response to entacapone in Parkinson’s disease: a randomized crossover clinical trial. Ann Neurol. 2011; 69(1):111-118.
- Ball P and R Knuppen. Catecholoestrogens (2-and 4-hydroxyoestrogens): chemistry, biogenesis, metabolism, occurrence and physiological significance. Acta Endocrinol. Suppl. 1980; 232:1-127.
- Lakhani NJ et al. 2-Methoxyestradiol, a Promising Anticancer Agent. Pharmacotherapy. 2003; 23:165-172.
- Lavigne JA et al. The Effects of Catechol-O-Methyltransferase Inhibition on Estrogen Metabolite and Oxidative DNBA Damage Levels in Estradiol-treated MCF-7 Cells. Cancer Research. 2001; 61:7488-7494.
- Worda C et al. Influence of the catechol-O-methyltransferase (COMT) codon 158 polymorphism on estrogen levels in women. Human Reproduction. 2003; 18(2):262-266.
- Eriksson AL et al. The COMT val158met polymorphism Is Associated with Early Pubertal Development, Height and Cortical Bone Mass in Girls. Pediatr Res. 2005; 58(1):71-77.
- Masurier M et al. Effect of Acute Tyrosine Depletion in Using a Branched Chain Amino-Acid Mixture on Dopamine Neurotransmission in the Rat Brain. Neuropsychopharmacology. 2006; 31(2):310-317.
- Sarris J et al. S-adenosyl methionine (SAMe) versus escitalopram and placebo in major depression RCT: Efficacy and effects of histamine and carnitine as moderators of response. J Affect Disord. 2014; 164:76-81.
- Kennedy D et al. Effects of high-dose B vitamin complex with vitamin C and minerals on subjective mood and performance in healthy males. Psychopharmacology (Berl). 2010; 211(1):55-68.
- Li Y et al. Functional and structural comparisons of cysteine residues in the Val108 wild type and Met108 variant of human soluble catechol O-methyltransferase. Chem Biol Interact. 2005; 152(2-3):151-163.
- Fava M et al. Rapidity of onset of the antidepressant effect of parenteral S-adenosyl-l-methionine. Psychiatry Res. 1995; 56(3):295-297.
- Jeffery DR and JA Roth. Kinetic reaction mechanism for magnesium binding to membrane-bound and soluble catechol O-methyltransferase. Biochem. 1987; 26(10):2955-2958.
- Sowa-Kucma M et al. Zinc, magnesium and NMDA receptor alterations in the hippocampus of suicide victims. J Affect Disord. 2013; 151(3):924-931.
- Basheer MP et al. A study of serum magnesium, calcium and phosphorus level, and cognition in the elderly population of South India. Alexandria J Med. 2016; 52(4):303-308.
- Yary T et al. Dietary magnesium intake and the incidence of depression: A 20 year follow-up study. J Affect Disord. 2016; 193:94-98.
- Haan MN et al. Homocysteine, B vitamins, and the incidence of dementia and cognitive impairment: Results from the Sacramento Area Latino Study on Aging. Am J Clin Nutr. 2007; 85(2):511-517.
- Smith AD et al. Homocysteine-lowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: A randomized controlled trial. PLoS One. 2010; 5(9):1-10.
- Mitchell ES et al. B vitamin polymorphisms and behavior: Evidence of associations with neurodevelopment, depression, schizophrenia, bipolar disorder and cognitive decline. Neurosci Biobehav Rev. 2014; 47:307-320.
- Hiroi T et al. Dopamine formation from tyramine by CYP2D6. Biochem Biophys Res Commun. 1998; 249(3):838-843.
- Zhu ZT et al. Tyramine excites rat subthalamic neurons in vitro by a dopamine-dependent mechanism. Neuropharmacology. 2007; 52(4):1169-1178.
- Burchett SA et al. The mysterious trace amines: Protean neuromodulators of synaptic transmission in mammalian brain. Prog Neurobiol. 2006; 79:223-246.
- Papaleo F et al. Sex-dichotomous effects of functional COMT genetic variations on cognitive functions disappear after menopause in both health and schizophrenia. Eur Neuropsychopharmacol. 2015; 25(12):2349-2363.
- Almey A et al. Estrogen receptors in the central nervous system and their implication for dopamine-dependent cognition in females. Horm Behav. 2015; 74:125-138.
- McCann S et al. Changes in 2-hydroxyestrone and 16α-hydroxyestrone metabolism with flaxseed consumption: Modification by COMT and CYP1B1 genotype. Cancer Epidemiol Biomarkers Prev. 2007; 16(2):256-262.
- Rižner TL. Estrogen biosynthesis, phase I and phase II metabolism, and action in endometrial cancer. Mol Cell Endocrinol. 2013; 381(1-2):124-139.
- Masurier M et al. Effect of Acute Tyrosine Depletion in Using a Branched Chain Amino-Acid Mixture on Dopamine Neurotransmission in the Rat Brain. 2006; 31(2):310-317.
- Fernstrom HD and MH Fernstrom. Tyrosine, Phenylalanine, and Catecholamine Synthesis and Function in the Brain. J. Nutr. 2007; 137(6):1539S-1547S.
- Reus G et al. Kynurenine pathway dysfunction in the pathophysiology and treatment of depression: Evidences from animal and human studies. J Psychiatr Res. 2015; 68:316-328.
- Jangid P et al. Comparative study of efficacy of l-5-hydroxytryptophan and fluoxetine in patients presenting with first depressive episode. Asian J Psychiatr. 2013; 6(1):29-34.
- Lowe S et al. L-5-Hydroxytryptophan augments the neuroendocrine response to a SSRI. Psychoneuroendocrinology. 2006; 31(4):473-484.
- Lardner A et al. Neurobiological effects of the green tea constituent theanine and its potential role in the treatment of psychiatric and neurodegenerative disorders. Nutritional Neuroscience. 2014; 17(4):145-155.
- Mu W et al. An overview of biological production of L-theanine. Biotechnol Adv. 2015; 33(3-4):335-342.
- Kakuda T. Neuroprotective effects of theanine and its preventive effects on cognitive dysfunction. Pharmacol Res. 2011; 64(2):162-168.
- Tian X et al. Protective effect of l-theanine on chronic restraint stress-induced cognitive impairments in mice. Brain Res. 2013; 1503:24-32.
- Martínez-Banaclocha M et al. N-acetyl-cysteine in the treatment of Parkinson’s disease. What are we waiting for? Med Hypotheses. 2012; 79(1):8-12.
- Dean O. et al. N-acetyl cysteine restores brain glutathione loss in combined 2-cyclohexene-1-one and d-amphetamine-treated rats: Relevance to schizophrenia and bipolar disorder. Neurosci Lett. 2011; 499(3):149-153.
- Botsakis K et al. 17β-Estradiol/N-acetylcysteine interaction enhances the neuroprotective effect on dopaminergic neurons in the weaver model of dopamine deficiency. Neuroscience. 2016; 320:221-229.
- Tunbridge E et al. Polymorphisms in the catechol‐O‐methyltransferase (COMT) gene influence plasma total homocysteine levels. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics 147.6 (2008):996-999.
- Paul R and A. Borah. The potential physiological crosstalk and interrelationship between two sovereign endogenous amines, melatonin and homocysteine. Life Sci. 2015; 139:97-107.
- Hursel R et al. The Role of Catechol-o-Methyl Transferase Val (108/158) MET Polymorphism (rs4680) in the effect of Green Tea on Resting Energy Expenditure and Fat Oxidation: A Pilot Study. 2014; 9(9): e106220.
- Lorenz M et al. The activity of catechol-O-methyltransferase (COMT) is not impaired by high doses of epigallocatechin-3-gallate (EGCG) in vivo. Eur J Pharmacol. 2014; 740: 645-651.
- Kang K et al. Beneficial effects of natural phenolics on levodopa methylation and oxidative neurodegeneration. Brain Res. 2013; 1497:1-14.
- Kang K et al. Dual beneficial effects of (-)-epigallocatechin-3-gallate on levodopa methylation and hippocampal neurodegeneration: In vitro and in vivo studies. PLoS One. 2010; 5(8): e11951.
- Xie X et al. Adenosine and dopamine receptor interactions in striatum and caffeine-induced behavioral activation. Comp Med. 2007; 57(6):538-545.
- Witte A et al. COMT Val158Met polymorphism modulates cognitive effects of dietary intervention. Front Aging Neurosci. 2010; 2:146.
- Voelcker-Rehage C et al. COMT gene polymorphisms, cognitive performance, and physical fitness in older adults. Psychol Sport Exerc. 2015; 20:20-28.
- Zhu BT et al. Effects of tea polyphenols and flavonoids on liver microsomal glucuronidation of estradiol and estrone. J Steroid Biochem Mol Biol. 1998; 64(3-4):207-215.
- Moon YJ et al. Dietary flavonoids: Effects on xenobiotic and carcinogen metabolism. Toxicol Vitr. 2006; 20 (2): 187-210.
- Ullah N et al. Green tea phytocompounds as anticancer: A review. Asian Pacific J Trop Dis. 2016; 6(4):330-336.
Mood Profile
- Lachman H et al. Human catechol-O-methyltransferase pharmacogenetics: description of a functional polymorphism and its potential application to neuropsychiatric disorders. Pharmacogenetics 1996; 6:243-250.
- Weinshilboum R et al. Methylation Pharmacogenetics: Catechol-O methyltransferase, Thiopurine Methyltransferase, and Histamine N-Methyltransferase. Annu. Rev. Pharmacol. Toxicol. 1999; 39:19-52.
- Genetics Home Reference. Genes: COMT. http://ghr.nlm.nih.gov/gene/COMT.
- Mier D et al. Neural substrates of pleiotropic action of genetic variation in COMT: a meta-analysis. Molecular Psychiatry. 2010; 15:918-927.
- Lester K et al. Therapygenetics: Using genetic markers to predict response to psychological treatment for mood and anxiety disorders. Biology of Mood & Anxiety Disorders. 2013; 3:4.
- Cheng JB et al. Genetic evidence that the human CYP2R1 enzyme is a key vitamin D 25-hydroxylase. Proc Natl Acad Sci U S A. 2004; 101:7711–7715.
- Ahn J et al. Genome-wide association study of circulating vitamin D levels. Human Molecular Genetics. 2010; 19(13): 2739-2745.
- Nissen J et al. Vitamin D Concentrations in Healthy Danish Children and Adults. PLOS One. 2014; 9(2):e89907.
- Harris HW et al. Supplementation might help patients with depression, seasonal mood disturbances. Current Psych. 2013; 12(4):19-25.
- Houssein-Nezhad A et al. Vitamin D for Health: A Global Perspective. Mayo Clin. Proc. 2013; 88(7):720-755.
- Kelly RJ et al. Sequence and expression of a candidate for the human Secretor blood group alpha (1,2)fucosyltransferase gene (FUT2). Homozygosity for an enzyme-inactivating nonsense mutation commonly correlates with the non-secretor phenotype. J Biol Chem. 1995; 270:4640–4649.
- Hazra A et al. Genome-wide significant predictors of metabolites in the one-carbon metabolism pathway. Hum Mol Gen. 2009; 18(23):4677-4687.
- Semmes BJ. Depression: a role for omega-3 fish oils and B vitamins? Evid. Based Integr. Med. 2005; 2:229–237.
- Tiemeier H et al. Vitamin B12, Folate, and Homocysteine in Depression: The Rotterdam Study. Am J Psychiatry. 2002; 159:2099-2101.
- Frankenburg, FR. The role of one-carbon metabolism in schizophrenia and depression. Harv. Rev. Psychiatry. 2007; 15:146–160.
- Seppälä et al. Association between vitamin b12 levels and melancholic depressive symptoms: a Finnish population-based study. BMC Psychiatry 2013;13:145.
- Tanaka T et al. Genome-wide Association Study of Vitamin B6, Vitamin B12, Folate, and Homocysteine Blood Concentrations. The American Journal of Human Genetics. 2009; 84:477-482.
- Gozdzik A et al. Association of vitamin D binding protein (VDBP) polymorphisms and serum 25(OH)D concentrations in a sample of young Canadian adults of different ancestry. J Steroid Biochem Mol Biol. 2011; 127:405–412.
- Ahn J et al. Genome-wide association study of circulating vitamin D levels. Human Molecular Genetics. 2010; 19(13) 2739-2745.
- Nissen J et al. Vitamin D Concentrations in Healthy Danish Children and Adults. PLOS One. 2014; 9(2):e89907.
- Foucan L et al. Polymorphisms in GC and NADSYN1 Genes are associated with vitamin D status and metabolic profile in non-diabetic adults. BMC Endocrine Disorders. 2013; 13:36.
- Wang T et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010; 376(9736):180-188.
- Grober U et al. Vitamin D: Update 2013: From rickets prophylaxis to general preventive healthcare. Dermato-Endocrinology. 2013; 5(3):331-47.
- Bischoff-Ferrari HA et al. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012; 367:40–9.
- Bjelland I et al. Folate, Vitamin B12, Homocysteine, and the MTHFR 677CT Polymorphism in Anxiety and Depression: The Hordaland Homocysteine Study. Arch Gen Psychiatry. 2003; 60(6):618-626.
- Beydoun MA et al. Serum folate, vitamin B-12 and homocysteine and their association with depressive symptoms among US adults. Psychosom Med. 2010;72(9):862-873.
- Refsum H et al. The Hordaland Homocysteine Study: A Community-Based Study of Homocysteine, Its Determinants, and Associations with Disease. J Nutr. 2006; 136:1731S-1740S.
- Frosst P et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet. 1995; 10:111–113.
- Lewis SJ et al. The thermolabile variant of MTHFR is associated with depression in the British Women’s Heart and Health Study and a meta-analysis. Molecular Psychiatry. 2006; 11:352-360.
- Van der Put NM et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998; 62(5):1044–51.
- Weisberg I et al. A second genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR) associated with decreased enzyme activity. Mol Genet Metab. 1998; 64:169–72.
- Nurk E et al. Plasma Total Homocysteine and Memory in the Elderly: The Hordaland Homocysteine Study. Ann Neurol. 2005; 58:847-857.
- Rajagopalan P et al. Common folate gene variant, MTHFR C677T, is associated with brain structure in two independent cohorts of people with mild cognitive impairment. Neuroimage Clin. 2012; 1(1):179-187. 179-187.
- Lewis SJ et al. The thermolabile variant of MTHFR is associated with depression in the British Women’s Heart and Health Study and a meta-analysis. Molecular Psychiatry. 2006; 11:352-360.
- Gilbody S et al. Methylenetetrahydrofolate Reductase (MTHFR) Genetic Polymorphisms and Psychiatric Disorders: A HuGE Review. Am J Epidemiol. 2007;165:1-13.
MTHFR
- Van der Put NM et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998; 62(5):1044–51.
- Wagner C. Biochemical role of folate in cellular metabolism. In: Bailey LB, editor. Folate in health and disease. New York, NY: Marcel Dekker Inc.; 1995. p. 23–42.
- Bailey LB and JF Gregory III. Polymorphisms of Methylenetetrahydrofolate Reductase and Other Enzymes: Metabolic Significance, Risks and Impact on Folate Requirement. J Nutr. 1999; 129(5):919-22.
- Stover PJ. Polymorphisms in 1-Carbon Metabolism, Epigenetics and Folate-Related Pathologies. J. Nutrigenet Nutrigenomics. 2012; 4(5):293-305.
- Refsum H et al. The Hordaland Homocysteine Study: A Community-Based Study of Homocysteine, Its Determinants, and Associations with Disease. J Nutr. 2006; 136:1731S-1740S.
- Frosst P et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet. 1995; 10:111–113.
- Weisberg I et al. A second genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR) associated with decreased enzyme activity. Mol Genet Metab. 1998; 64:169–72.
- Teng Z. The 677C>T (rs1801133) polymorphism in the MTHFR gene contributes to colorectal cancer risk: a meta-analysis based on 71 research studies. PLoS One. 2013; 8(2):e55332.
- Yang L, et al. Impact of methylenetetrahydrofolate reductase (MTHFR) polymorphisms on methotrexate-induced toxicities in acute lymphoblastic leukemia: a meta-analysis. Tumor Biol. 2012; 33(5):1445–54.
- Bjelland I et al. Folate, Vitamin B12, Homocysteine, and the MTHFR 677CT Polymorphism in Anxiety and Depression: The Hordaland Homocysteine Study. Arch Gen Psychiatry. 2003; 60(6):618-626.
- Beydoun MA et al. Serum folate, vitamin B-12 and homocysteine and their association with depressive symptoms among US adults. Psychosom Med. 2010; 72(9):862-873.
- Nurk E et al. Plasma Total Homocysteine and Memory in the Elderly: The Hordaland Homocysteine Study. Ann Neurol. 2005; 58:847-857.
- Rajagopalan P et al. Common folate gene variant, MTHFR C677T, is associated with brain structure in two independent cohorts of people with mild cognitive impairment. Neuroimage Clin. 2012; 1(1):179-187. 179-187.
- Cotlarciuc I et al. Effect of genetic variants associated with plasma homocysteine levels on stroke risk. Stroke. 2014; 45(7):1920-4.
- Refsum H et al. The Hordaland Homocysteine Study: A community- based study of homocysteine, its determinants, and associations with disease. The Journal of Nutrition. 2006; 136:1731S-1740S.
- Simpson J et al. Micronutrients and women of reproductive potential: required dietary intake and consequences of dietary deficiency or excess. Part I – Folate, Vitamin B12, Vitamin B6. J Matern Fetal Neonatal Med. 2011; 24(1):1-24.
- NIH: Office of Dietary Supplements. Folate. http://ods.od.nih.gov/factsheets/Folate-HealthProfessional/
- Pietrzrik K. Folic acid and L-5-Methyltetrahydrofolate: Comparison of Clinical Pharmacokinetics and Pharmacodynamics. Cln Pharmacokinet. 2010; 49(8):535-548.
- Jain R et al. Beyond the resistance: how novel neurobiological understandings of depression may lead to advanced treatment strategies. J Clin Psychiatry. 2012; 73(11):e30.
- Soleimani A et al. Comparison of oral folic acid and folinic acid on blood homocysteine level of patients on hemodialysis. Iran J Kidney Dis. 2011: 5(1):45-9.
- Chen Z et al. Purification and Kinetic Mechanism of a Mammalian Methionine Synthase from Pig Liver. J Biol Chem. 1994; 269(44):27193-27197.
- Ho G et al. Methylenetetrahydrofolate Reductase Polymorphisms and Homocysteine-Lowering Effect of Vitamin Therapy in Singaporean Stroke Patients. Stroke. 2006; 37:456-460.
- Chen X et al. Contrasting behaviors of mutant cystathionine beta-synthase enzymes associated with pyridoxine response. Hum Mutat. 2006; 275(5):474-482.
- Dell’Edera D et al. Effect of multivitamins on plasma homocysteine in patients with the 5,10 methylenetetrahydrofolate reductase C677T homozygous state. Molecular Medicine Reports. 2013; 8:609-612.
- Betaine: Drug Information. http://www.uptodate.com/contents/betaine-drug-information?source=search_result&search=betaine&selectedTitle=1~9
- Lawson-Yuen A eta l. The use of betaine in treatment of elevated homocysteine. Mol Genet Metab. 2006; 88(3):201-207.
- Hultberg B et al. Plasma homocysteine and thiol compound fractions after oral administration of N-acetylcysteine. Scand J Clin Lab Invest. 1994; 54:417–422.
Nutritional Health
- Ferrucci L et al. “Common Variation in the B-Carotene 15, 15’-Monooxygenase 1 Gene Affects Circulating Levels of Carotenoids: A Genome-wide Association Study.” The American Journal of Human Genetics. 2009; 84, 123-133.
- Borel P et al. “Genetic Variations Involved in Interindividual Variability in Carotenoid Status.” Mol Nutr Food Res. 2012; 56(2): 228-40.
- Wyss A et al. Expression pattern and localization of β,β-carotene 15,15′-dioxygenase in different tissues. Biochem. J. 2001; 354:521–529.
- Lietz G et al. “Single Nucleotide Polymorphisms Upstream from the B-Carotene 15, 15’-Monoxygenase Gene Influence Provitamin A Conversion Efficiency in Female Volunteers.” J. Nutr. 2012; 142(1):161S-165S.
- Cheng JB et al. Genetic evidence that the human CYP2R1 enzyme is a key vitamin D 25-hydroxylase. Proc Natl Acad Sci U S A. 2004; 101:7711–7715.
- Ahn J et al. Genome-wide association study of circulating vitamin D levels. Human Molecular Genetics. 2010; 19(13): 2739-2745.
- Nissen J et al. Vitamin D Concentrations in Healthy Danish Children and Adults. PLOS One. 2014; 9(2):e89907.
- Harris HW et al. Supplementation might help patients with depression, seasonal mood disturbances. Current Psych. 2013; 12(4):19-25.
- Houssein-Nezhad A et al. Vitamin D for Health: A Global Perspective. Mayo Clin. Proc. 2013; 88(7):720-755.
- Holick MF and M Garabedian. Vitamin D: photobiology, metabolism, mechanism of action, and clinical applications. In: Favus MJ, ed. Primer on the metabolic bone diseases and disorders of mineral metabolism. 6th ed. Washington, DC: American Society for Bone and Mineral Research. 2006; 129-137.
- Lips P and NM van Schoor. The effect of vitamin D on bone and osteoporosis. Best Pract Res Clin Endocrinol Metab. 2011; 25:585–591.
- Wang T et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010; 376(9736):180-188.
- Bischoff-Ferrari HA et al. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012; 367:40–9.
- Grober U et al. Vitamin D: Update 2013: From rickets prophylaxis to general preventive healthcare. Dermato-Endocrinology. 2013; 5(3):331-47.
- Kelly RJ et al. Sequence and expression of a candidate for the human Secretor blood group alpha (1,2)fucosyltransferase gene (FUT2). Homozygosity for an enzyme-inactivating nonsense mutation commonly correlates with the non-secretor phenotype. J Biol Chem. 1995; 270:4640–4649.
- Hazra A et al. Genome-wide significant predictors of metabolites in the one-carbon metabolism pathway. Hum Mol Gen. 2009; 18(23):4677-4687.
- Semmes BJ. Depression: a role for omega-3 fish oils and B vitamins? Evid. Based Integr. Med. 2005; 2:229–237.
- Tiemeier H et al. Vitamin B12, Folate, and Homocysteine in Depression: The Rotterdam Study. Am J Psychiatry. 2002; 159:2099-2101.
- Frankenburg, FR. The role of one-carbon metabolism in schizophrenia and depression. Harv. Rev. Psychiatry. 2007; 15:146–160.
- Seppälä et al. Association between vitamin b12 levels and melancholic depressive symptoms: a Finnish population-based study. BMC Psychiatry 2013; 13:145.
- Tanaka T et al. Genome-wide Association Study of Vitamin B6, Vitamin B12, Folate, and Homocysteine Blood Concentrations. The American Journal of Human Genetics. 2009; 84:477-482.
- Hazra A et al. Common variants of FUT2 are associated with plasma vitamin B12 levels. Nat. Genet. 2008; 40:1160–1162.
- http://lpi.oregonstate.edu/infocenter/vitamins/vitaminB12/.
- Gozdzik A et al. Association of vitamin D binding protein (VDBP) polymorphisms and serum 25(OH)D concentrations in a sample of young Canadian adults of different ancestry. J Steroid Biochem Mol Biol. 2011; 127:405–412.
- Ahn J et al. Genome-wide association study of circulating vitamin D levels. Human Molecular Genetics. 2010; 19(13) 2739-2745.
- Nissen J et al. Vitamin D Concentrations in Healthy Danish Children and Adults. PLoS One. 2014; 9(2):e89907.
- Foucan L et al. Polymorphisms in GC and NADSYN1 Genes are associated with vitamin D status and metabolic profile in non-diabetic adults. BMC Endocrine Disorders. 2013; 13:36.
- Wang T et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010; 376(9736):180-188.
- Grober U et al. Vitamin D: Update 2013: From rickets prophylaxis to general preventive healthcare. Dermato-Endocrinology. 2013; 5(3):331-47.
- Bischoff-Ferrari HA et al. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012; 367:40–9.
- Wagner C. Biochemical role of folate in cellular metabolism. In: Bailey LB, editor. Folate in health and disease. New York, NY: Marcel Dekker Inc.; 1995. p. 23–42.
- Bailey LB and JF Gregory III. Polymorphisms of Methylenetetrahydrofolate Reductase and Other Enzymes: Metabolic Significance, Risks and Impact on Folate Requirement. J Nutr. 1999; 129(5):919-22.
- Stover PJ. Polymorphisms in 1-Carbon Metabolism, Epigenetics and Folate-Related Pathologies. J. Nutrigenet Nutrigenomics. 2012; 4(5):293-305.
- Refsum H et al. The Hordaland Homocysteine Study: A Community-Based Study of Homocysteine, Its Determinants, and Associations with Disease. J Nutr. 2006; 136:1731S-1740S.
- Frosst P et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet. 1995; 10:111–113.
- Van der Put NM et al. A second common mutation in the methylenetetrahydrofolate reductase gene: an additional risk factor for neural-tube defects? Am J Hum Genet. 1998; 62(5):1044–51.
- Weisberg I et al. A second genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR) associated with decreased enzyme activity. Mol Genet Metab. 1998; 64:169–72.
- Teng Z. The 677C>T (rs1801133) polymorphism in the MTHFR gene contributes to colorectal cancer risk: a meta-analysis based on 71 research studies. PLoS One. 2013; 8(2):e55332.
- Hara N et al. Molecular identification of human glutamine- and ammonia-dependent NAD synthetases. Carbon-nitrogen hydrolase domain confers glutamine dependency. J. Biol. Chem. 2003; 278(13):10914-10921.
- Wassif CA et al. Mutations in the human sterol Δ7-reductase gene at 11q12–13 cause Smith–Lemli–Opitz syndrome. Am. J. Hum. Genet. 1998; 63:55–62.
- Wang T et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010; 376(9736):180-188.
- Harris HW et al. Supplementation might help patients with depression, seasonal mood disturbances. Current Psych. 2013; 12(4):19-25.
- Houssein-Nezhad A et al. Vitamin D for Health: A Global Perspective. Mayo Clinic. Proc. 2013; 88(7):720-755.
- Bischoff-Ferrari HA et al. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012; 367:40–9.
- Grober U et al. Vitamin D: Update 2013: From rickets prophylaxis to general preventive healthcare. Dermato-Endocrinology. 2013; 5(3):331-47.
- Stoltzfus RJ. Iron deficiency: global prevalence and consequences. Food Nutrition Bulletin. 2003; 24:S99–S103.
- Patel KV. “Variability and heritability of hemoglobin concentration: an opportunity to improve understanding of anemia in older adults.” Haematologica 2008; 93:1281-1283.
- Benyamin B et al. Common variants in TMPRSS6 are associated with iron status and erythrocyte volume. Nature Genetics 2009; 41:1173-1175.
- Chambers JC et al. Genome-wide association study identifies variants in TMPRSS6 associated with hemoglobin levels. Nature Genetics 2009; 41:1170-1172.
- Morris HA. Vitamin D Activities for Health Outcomes. Ann Lab Med 2014; 34:181-186.
- Turner AG. Vitamin D and bone health. Scand J Clin Lab Invest Suppl. 2012; 243:65-72.
- Palomba S et al. BsmI vitamin D receptor genotypes influence the efficacy of antiresorptive treatments in postmenopausal osteoporotic women. A 1-year multicenter, randomized and controlled trial. Osteoporosis Int. 2005; 16(8):943-52.
- Jia F et al. Vitamin D receptor BsmI polymorphism and osteoporosis risk: a meta-analysis from 26 studies. Genet Test Mol Biomarkers. 2013; 17(1):30-4.
- Ji GR. BsmI, TaqI, ApaI and FokI polymorphisms in the vitamin D receptor (VDR) gene and risk of fracture in Caucasians: a meta-analysis. Bone. 2010; 47(3):681-6.
- Bischoff-Ferrari HA et al. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012; 367:40–9.
- Grober U et al. Vitamin D: Update 2013: From rickets prophylaxis to general preventive healthcare. Dermato-Endocrinology. 2013; 5(3):331-47.
PGx Comprehensive
- Bloom D et al. The Global Economic Burden of Non-Communicable Diseases. Geneva: World Economic Forum. 2011:1-48.
- Ganesh SK et al. Genetics and genomics for the prevention and treatment of cardiovascular disease: update: a scientific statement from the American Heart Association. Circulation. 2013; 128(25):2813-51.
- Haddad PM et al. Nonadherence with antipsychotic medication in schizophrenia: challenges and management strategies. Patient Relat Outcome Meas. 2014; 5:43-62.
- Hicks JK et al. Clinical Pharmacogenetics Implementation Consortium guideline for CYP2D6 and CYP2C19 genotypes and dosing of tricyclic antidepressants. Clin Pharmacol Ther. 2013; 93(5):402-8.
- Janicki PK. Comprehensive Treatment of Chronic Pain by Medical, Interventional, and Integrative Approaches. Deer TR et al. eds. 2013.
- Jannetto PJ and Bratanow NC. Pharmacogenomic considerations in the opioid management of pain. Genome Med. 2010; 2:66.
- Johnson J et al. Clinical Pharmacogenetics Implementation Consortium Guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin Pharmacol Ther. 2011; 90(4):625-9.
- Johnson JA et al. Institutional profile: University of Florida and Shands Hospital Personalized Medicine Program: clinical implementation of pharmacogenetics. Pharmacogenomics. 2013; 14(7):723-6.
- Kelly K et al. Toward achieving optimal response: understanding and managing antidepressant side effects. Dialogues Clin Neurosci. 2008;10(4):409-18.
- Kitzmiller JP et al. Pharmacogenomic testing: relevance in medical practice: why drugs work in some patients but not in others. Cleve Clin J Med. 2011; 78(4):243-57.
- Kitzmiller JP et al. Statin pharmacogenomics: pursuing biomarkers for predicting clinical outcomes. Discov Med. 2013; 16(86):45-51.
- Lamba J et al. PharmGKB summary: very important pharmacogene information for CYP3A5. Pharmacogenet Genomics 2012; 22(7):555-8.
- Link E et al. SLCO1B1 variants and statin-induced myopathy–a genomewide study. N Engl J Med. 2008; 359(8):789-99.
- McNulty H et al. Homocysteine, B-vitamins and CVD. Proc Nutr Soc. 2008; 67(2):232-7.
- Miranda-Massari JR et al. Metabolic Correction in the Management of Diabetic Peripheral Neuropathy: Improving Clinical Results Beyond Symptom Control. Curr Clin Pharmacol. 2011; 6(4):260-273.
- Phillips KA et al. Potential Role of Pharmacogenomics in Reducing Adverse Drug Reactions. JAMA. 2001; 286(18):2270.
- Ramsey LB et al. The clinical pharmacogenetics implementation consortium guideline for SLCO1B1 and simvastatin-induced myopathy: 2014 update. Clin Pharmacol Ther. 2014; 96(4):423-8.
- Refsum H et al. The Hordaland Homocysteine Study: A Community-Based Study of Homocysteine, Its Determinants, and Associations with disease. J Nutr. 2006; 136:1731S-1740S.
- Rush AJ et al. Acute and Longer-Term Outcomes in Depressed Outpatients Requiring One or Several Treatment Steps: A STAR*D Report. 2006; 163:1905-1917.
- Sadhasivam S and Chidambaran V. Pharmacogenomics of opioids and perioperative pain management. Pharmacogenomics. 2012; 13(15):1719-40.
- Samer CF et al. Applications of CYP450 testing in the clinical setting. Mol Diagn Ther. 2013; 17(3):165-84.
- Scott S et al. Clinical Pharmacogenetics Implementation Consortium guidelines for cytochrome P450-2C19 (CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther. 2011; 90(2):328-332.
- Shaheen K et al. Factor V Leiden: how great is the risk of venous thromboembolism? Cleve Clin J Med. 2012; 79(4):265-72.
- Sim SC et al. Pharmacogenomics of drug-metabolizing enzymes: a recent update on clinical implications and endogenous effects. Pharmacogenomics J. 2013; 13(1):1-11.
- Tomaszewski M et al. Statin-induced myopathies. Pharmacol Rep. 2011; 63(4):859-866.
- Trescot AM and Faynboym S. A Review of the Role of Genetic Testing in Pain Medicine. Pain Physician. 2014; 17:425-445.
- U.S. Food and Drug Administration. Table of Pharmacogenomic Biomarkers in Drug Labeling. 08/18/2014. Available at: http://www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ucm083378.htm.
- Van Driest SL et al. Clinically actionable genotypes among 10,000 patients with preemptive pharmacogenomic testing. Clin Pharmacol Ther. 2014; 95(4):423-31.
- Varga E and Moll S. Prothrombin 20210 mutation (factor II mutation). Circulation. 2004; 110:e15-8.
- Velligan DI et al. The expert consensus guideline series: adherence problems in patients with serious and persistent mental illness. J Clin Psychiatry. 2009; 70 Suppl 4:1-46; quiz 47-8.
- Voora D and Ginsburg GS. Clinical application of cardiovascular pharmacogenetics. J Am Coll Cardiol. 2012; 60(1):9-20.
- Xu Y and Johnson A. Opioid therapy pharmacogenomics for noncancer pain: efficacy, adverse events, and costs. Pain Res Treat. 2013; 2013:943014.
- Zandi PP and Judy JT. The Promise and Reality of Pharmacogenetics in Psychiatry. Psychiatr Clin North Am. 2010;33(1):181-224.
Weight Management
- Martínez JA et al. Obesity risk is associated with carbohydrate intake in women with the Gln27Glu β2-adrenoreceptor polymorphism. J Nutr. 2003; 133:2549-2554.
- Large V et al. Human Beta-2 adrenoreceptor gene polymorphisms are highly frequent in obesity and associate with altered adipocyte Beta-2 adrenoceptor function. J Clin Invest. 1997; 100:3005-3013.
- Macho-Azcarate et al. Gln27Glu polymorphism in the beta2 adrenergic receptor gene and lipid metabolism during exercise in obese women. Int J Obesity. 2002; 26:1434-1441.
- HGNC. http://www.genenames.org/cgibin/gene_symbol_report? hgnc_id=HGNC:286.
- org. Nutrition and Healthy Eating: Carbohydrates: How carbs fit into a healthy diet. http://www.mayoclinic.org/healthy-living/nutrition-and-healthy-eating/in-depth/carbohydrates/art-20045705?pg=1.
- Layman DK. Dietary Guidelines should reflect new understandings about adult protein needs. Nutr Metab (Lond). 2009; 6:12.
- Phares DA et al. Association Between Body Fat Response to Exercise Training and Multilocus ADR Genotypes. Obes Res. 2004; 12(5):807-815.
- Corbalan MS et al. The 27Glu polymorphism of the Beta2-adrenergic receptor gene interacts with physical activity influencing obesity risk among female subjects. Clin Genet. 2002; 61:305-307.
- Zhang et al. Association of Gln27Glu and Arg16Gly Polymorphisms in Beta2-Adrenergic Receptor Gene with Obesity Susceptibility: A Meta-Analysis. PLoS ONE. 2014; 9(6): e100489.
- Levy E et al. The polymorphism at codon 54 of the FABP2 gene increases fat absorption in human intestinal explants. J Biol Chem. 2001; 276:39679-39684.
- Marin C et al. The Ala54Thr polymorphism of the fatty acid-binding protein 2 gene is associated with a change in insulin sensitivity after a change in the type of dietary fat. Am J Clin Nutr. 2005; 82:196-200.
- Albala C et al. FABP2 Ala54Thr polymorphism and diabetes in Chilean elders. Diab Res Clin Pract. 2007; 77:245-250.
- Paglialunga S et al. Regulation of postprandial lipemia: an update on current trends. Appl Physiol Nutr Metab. 2007; 32:61-75.
- Takakura Y et al. Thr54 allele of the FABP2 gene affects resting metabolic rate and visceral obesity. Diabetes Res Clin Pract. 2005; 67:36-42.
- Hegele RA. A Review of Intestinal Fatty Acid Binding Protein Gene Variation and the Plasma Lipoprotein Response to Dietary Components. Clin Biochem. 1998; 31:609-612.
- Gaggini M et al. Non-Alcoholic Fatty Liver Disease (NAFLD) and Its Connection with Insulin Resistance, Dyslipidemia, Atherosclerosis and Coronary Heart Disease. Nutrients. 2013; 5:1544-1560.
- Almeida JC et al. The Ala54Thr polymorphism of the FABP2 gene influences the postprandial fatty acids in patients with type 2 diabetes. J Clin Endocrin Met. 2010; 95:3909-3917.
- Dworatzek PD et al. Postprandial lipemia in subjects with the threonine 54 variant of the fatty acid-binding protein 2 gene is dependent on the type of fat ingested. Am J Clin Nutr. 2004; 79:1110-1117.
- Weiss EP et al. FABP2 Ala54Thr genotype is associated with glucoregulatory function and lipid oxidation after a high-fat meal in sedentary nondiabetic men and women. Am J Clin Nutr. 2007; 85:102-108.
- Weickert MO et al. Metabolic Effects of Dietary Fiber consumption and Prevention of Diabetes. J Nutr. 2008; 138(3):439-442.
- org. Nutrition and Healthy Eating: Carbohydrates: How carbs fit into a healthy diet. http://www.mayoclinic.org/ healthy-living/nutrition-and-healthy-eating/in-depth/carbohydrates/ art-20045705?pg=1
- Pfeiffer M et al. The influence of walking performed immediately before meals with moderate fat content on postprandial lipemia. Lipids Health Dis. 2005; 4:24.
- Brandou F et al. Impact of high- and low-intensity targeted exercise training on the type of substrate utilization in obese boys submitted to a hypocaloric diet. Diabetes Metab. 2005; 31(4 Pt 1):327-325.
- Corella D et al. A High Intake of Saturated Fatty Acids Strengthens the Association between the Fat Mass and Obesity-Associated Gene and BMI. J of Nutr. 2011; 141:2219-2225.
- Kilpelainen TO et al. Physical Activity Attenuates the Influence of FTO Variants on Obesity Risk: A Meta-Analysis of 218,166 Adults and 19.268 Children. PLOS Medicine. 2011; 8(11):31001116.
- Frayling TM et al. A Common Variant in the FTO Gene is Associated with Body Mass Index and Predisposition to Childhood and Adult Obesity. Science. 2007; 316(58):889-894.
- Berulava T and Horsthemke B. The obesity-associated SNPs in intron 1 of the FTO gene affect primary transcript levels. Eur J Hum Genet. 2010; 18(9):1054-1058.
- Karra E et al. A link between FTO, ghrelin, and impaired brain food-cue responsivity. J Clin Invest. 2013; 123(8):3539-3551.
- Lu Y et al. Obesity genomics: assessing the transferability of susceptibility loci across diverse populations. Genome Med. 2013; 5:55.
- Cho YS et al. A large-scale genome-wide association study of Asian populations uncovers genetic factors influencing eight quantitative traits. Nat Genet. 2009; 41:527-534.
- Jaaskelainen A et al. Meal Frequencies Modify the Effect of Common Genetic Variants on Body Mass Index in Adolescents of the Northern Finland Birth Cohort 1986. PLOS One. 2013; 8(9):e73802.
- Zhang X et al. FTO Genotype and 2-Year Change in Body Composition and Fat Distribution in Response to Weight-Loss Diets: The POUNDS Lost Trial. Diabetes. 2012; 61:3005-3011.
- Ortega-Azorin C et al. Associations of the FTO rs9939609 and the MC4R rs17782313 polymorphisms with type 2 diabetes are modulated by diet, being higher when adherence to the Mediterranean diet pattern is low. Cardiovasc Diabetol. 2012; 11:137.
- Mitchell JA et al. FTO Genotype and the Weight Loss Benefits of Moderate Intensity Exercise. Obesity. 2010; 18(3):641-643.
- Lauria F et al. Prospective Analysis of the Association of a Common Variant of FTO (rs9939609) with Adiposity in Children: Results of the IDEFICS Study. PLoS One. 2012; 7: e48876.
- Velders FP et al. FTO at rs9939609, food responsiveness, emotional control and symptoms of ADHD in preschool children. PLoS One. 2012; 7:e49131.
- Wardle J et al. Obesity associated genetic variation in FTO is associated with diminished satiety. J Clin Endocrinol Metab. 2008; 93:3640–3643.
- den Hoed M et al. Postprandial responses in hunger and satiety are associated with the rs9939609 single nucleotide polymorphism in FTO. Am J Clin Nutr. 2009; 90:1426–1432.
- Loos RJF et al. Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat Genet. 2008; 40(6):768-775.
- Qi L et al. The common obesity variant near MC4R gene is associated with higher intakes of total energy and dietary fat, weight change and diabetes risk in women. Human Mol Genetics 2008; 17:3502-3508.
- Xi B et al. Common polymorphism near the MC4R gene is associated with type 2 diabetes: data from a meta-analysis of 123,373 individuals. Diabetologia 2012; 55:2660–2666.
- Stutzmann F et al. Common genetic variation near MC4R is associated with eating behaviour patterns in European populations. Int J Obes. 2009; 33:373-378.
- Xi B et al. Influence of physical inactivity on associations between single nucleotide polymorphisms and genetic predisposition to childhood obesity. Am J Epidemiology 2011; 173:1256-1262.
- Acosta A et al. Association of melanocortin 4 receptor gene variation with satiation and gastric emptying in overweight and obese adults. Genes Nutr. 2014; 9(2):384.
- Zlatohlavek L et al. FTO and MC4R gene variants determine BMI changes in children after intensive lifestyle intervention. Clin Biochem. 2013; 46:313-31.
- Raynor HA et al. Dietary energy density and successful weight loss maintenance. Eat Behav. 2011; 12(2):119-125.
- Otten JJ et al. DRI: Dietary Reference Intakes: The Essential Guide to Nutrient Requirements. Washington, DC. National Academies Press. c2006.
- OSU: Linus Pauling Institute. Glycemic Index and Glycemic Load. http://lpi.oregonstate.edu/infocenter/foods/grains/gigl.html
- Jaaskelainen A et al. Meal Frequencies Modify the Effect of Common Genetic Variants on Body Mass Index in Adolescents of the Northern Finland Birth Cohort 1986. PLOS One. 2013; 8(9):e73802.
- Little JP et al. A practical model of low-volume high-intensity interval training induces mitochondrial biogenesis in human skeletal muscle: potential mechanisms. J Physiol. 2010; 588 (Pt 6):1011-1022.
- Bauer F et al. Obesity genes identified in genome-wide association studies are associated with adiposity measures and potentially with nutrient-specific food preference. Am J Clin Nutr. 2009; 90:951–959.
- Volckmar AL et al. Mutation screen in the GWAS derived obesity gene SH2B1 including functional analyses of detected variants. BMC Med Genomics. 2012; 5:65.
- Jamshidi Y et al. The SH2B gene is associated with serum leptin and body fat in normal female twins. Obesity. 2007; 15:5-9.
- Ren D et al. Neuronal SH2B1 is essential for controlling energy and glucose homeostasis. J Clin Invest. 2007; 117:397–406.
- Morris DL et al. SH2B1 enhances insulin sensitivity by both stimulating the insulin receptor and inhibiting tyrosine dephosphorylation of insulin receptor substrate proteins. Diabetes. 2009; 58:2039-2047.
- McCaffery JM et al. Obesity susceptibility loci and dietary intake in the Look AHEAD Trial. Am J Clin Nutr. 2012; 95:1477-1486.
- Duan C et al. Disruption of the SH2-B gene causes age-dependent insulin resistance and glucose intolerance. Mol Cell Biol 2004; 24:7435–7443.
- Ren D et al. Identification of SH2-B as a key regulator of leptin sensitivity, energy balance, and body weight in mice. Cell Metabolism. 2005; 2:95–104.
- Wolpert HA et al. Dietary Fat Acutely Increases Glucose Concentrations and Insulin Requirements in Patients With Type 1 Diabetes: Implications for carbohydrate-based bolus dose calculation and intensive diabetes management. Diabetes Care. 2012; 36(4):810-816.
- Diabetes.org American Diabetes Association: Insulin Basics. http://www.diabetes.org/living-with-diabetes/treatment-and-care/ medication/insulin/insulin-basics.html.
- Klok MD et al. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: a review. Obes Rev. 2007; 8(1):21-34.
- Bachman JL et al. Eating frequency is higher in weight loss maintainers and normal-weight individuals than in overweight individuals. J Am Diet Assoc. 2011; 111(11):1730-4.
- Lichtenstein MB et al. Exercise Addiction in Men Is Associated With Lower Fat-Adjusted Leptin Levels. Clin J Sport Med. 2015; 25(2):138-43.
- OSU: Linus Pauling Institute. Glycemic Index and Glycemic Load. http://lpi.oregonstate.edu/infocenter/foods/grains/gigl.html
- Zhang Z et al. A high-legume low-glycemic index diet reduces fasting plasma leptin in middle-aged insulin-resistant and –sensitive men. Eur J Clin Nutr. 2011; 65(3):415-418.
- Fall T et al. The role of obesity-related genetic loci in insulin sensitivity. Diabet Med. 2012; 29(7):e62-66.
- Robiou-du-Pont S et al. Contribution of 24 obesity-associated genetic variants to insulin resistance, pancreatic beta-cell function and type 2 diabetes. Int J Obesity. 2013; 37:980-985.
- Weickert MO et al. Metabolic Effects of Dietary Fiber Consumption and Prevention of Diabetes. J Nutr. 2008; 138(3):439-442.
- Willett WC. Eat, Drink, and be Healthy: The Harvard Medical School Guide to Healthy Eating. New York: Simon & Schuster; 2001.
- Zaccaria M et al. Plasma leptin and energy expenditure during prolonged, moderate intensity, treadmill exercise. J Endocrinol Invest. 2013; 36(6):396-401.
- Li S et al. Physical Activity Attenuates the Genetic Predisposition to Obesity in 20,000 Men and Women from EPIC-Norfolk Prospective Population Study. PLOS Medicine. 2010; 7(8):e1000332.
- Lee S et al. Aerobic exercise but not resistance exercise reduces intrahepatic lipid content and visceral fat and improves insulin sensitivity in obese adolescent girls: a randomized controlled trial. Am J Physiol Endocrinol Metab. 2013; 305(10):E1222-1229.
Please contact Kashi Health for further scientific reference papers.