Hostname: page-component-848d4c4894-nr4z6 Total loading time: 0 Render date: 2024-04-30T16:43:42.928Z Has data issue: false hasContentIssue false

Utilising the precision nutrition toolkit in the path towards precision medicine

Published online by Cambridge University Press:  07 June 2023

Caleigh Sawicki
Affiliation:
Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
Danielle Haslam
Affiliation:
Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
Shilpa Bhupathiraju*
Affiliation:
Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
*
*Corresponding author: Shilpa Bhupathiraju, Email nhsnb@channing.harvard.edu

Abstract

The overall aim of precision nutrition is to replace the ‘one size fits all’ approach to dietary advice with recommendations that are more specific to the individual in order to improve the prevention or management of chronic disease. Interest in precision nutrition has grown with advancements in technologies such as genomics, proteomics, metabolomics and measurement of the gut microbiome. Precision nutrition initiatives have three major applications in precision medicine. First, they aim to provide more ‘precision’ dietary assessments through artificial intelligence, wearable devices or by employing omic technologies to characterise diet more precisely. Secondly, precision nutrition allows us to understand the underlying mechanisms of how diet influences disease risk and identify individuals who are more susceptible to disease due to gene–diet or microbiota–diet interactions. Third, precision nutrition can be used for ‘personalised nutrition’ advice where machine-learning algorithms can integrate data from omic profiles with other personal and clinical measures to improve disease risk. Proteomics and metabolomics especially provide the ability to discover new biomarkers of food or nutrient intake, proteomic or metabolomic signatures of diet and disease, and discover potential mechanisms of diet–disease interactions. Although there are several challenges that must be overcome to improve the reproducibility, cost-effectiveness and efficacy of these approaches, precision nutrition methodologies have great potential for nutrition research and clinical application.

Type
Conference on ‘Food and nutrition: Pathways to a sustainable future’
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Willett, W (2012) Nutritional Epidemiology. New York, NY: Oxford University Press.CrossRefGoogle Scholar
Tebani, A & Bekri, S (2019) Paving the way to precision nutrition through metabolomics. Front Nutr 6, 41.CrossRefGoogle ScholarPubMed
Trujillo, E, Davis, C & Milner, J (2006) Nutrigenomics, proteomics, metabolomics, and the practice of dietetics. J Am Diet Assoc 106, 403413.CrossRefGoogle ScholarPubMed
Wang, DD & Hu, FB (2018) Precision nutrition for prevention and management of type 2 diabetes. Lancet Diabetes Endocrinol 6, 416426.CrossRefGoogle ScholarPubMed
Rodgers, GP & Collins, FS (2020) Precision nutrition – the answer to ‘what to eat to stay healthy’. JAMA 324, 735736.CrossRefGoogle ScholarPubMed
National Institutes of Health (2020) Nutrition for precision health, powered by the all of us research program. https://commonfund.nih.gov/nutritionforprecisionhealthGoogle Scholar
Ferguson, LR (2010) Genome-wide association studies and diet. Personalized Nutr 101, 814.CrossRefGoogle ScholarPubMed
Maruvada, P, Lampe, JW, Wishart, DS et al. (2020) Perspective: dietary biomarkers of intake and exposure – exploration with omics approaches. Adv Nutr 11, 200215.CrossRefGoogle ScholarPubMed
Vockley, J, Andersson, HC, Antshel, KM et al. (2014) Phenylalanine hydroxylase deficiency: diagnosis and management guideline. Genet Med 16, 188200.CrossRefGoogle ScholarPubMed
Hegele, RA, Jugenberg, M, Connelly, PW et al. (1997) Evidence for gene-diet interaction in the response of blood pressure to dietary fibre. Nutr Res 17, 12291238.CrossRefGoogle Scholar
Cornelis, MC, El-Sohemy, A & Campos, H (2007) Genetic polymorphism of the adenosine A2A receptor is associated with habitual caffeine consumption. Am J Clin Nutr 86, 240244.CrossRefGoogle ScholarPubMed
Tremblay, BL, Cormier, H, Rudkowska, I et al. (2015) Association between polymorphisms in phospholipase A2 genes and the plasma triglyceride response to an n-3 PUFA supplementation: a clinical trial. Lipids Health Dis 14, 12.CrossRefGoogle Scholar
Ling, C & Groop, L (2009) Epigenetics: a molecular link between environmental factors and type 2 diabetes. Diabetes 58, 27182725.CrossRefGoogle ScholarPubMed
Li, X & Qi, L (2022) Epigenetics in precision nutrition. J Pers Med 12, 533.CrossRefGoogle ScholarPubMed
Lillycrop, KA, Hoile, SP, Grenfell, L et al. (2014) DNA methylation, ageing and the influence of early life nutrition. Proc Nutr Soc 73, 413421.CrossRefGoogle ScholarPubMed
Ma, J, Rebholz, CM, Braun, KVE et al. (2020) Whole blood DNA methylation signatures of diet are associated with cardiovascular disease risk factors and all-cause mortality. Circ Genom Precis Med 13, e002766.CrossRefGoogle ScholarPubMed
Keller, M, Yaskolka Meir, A, Bernhart, SH et al. (2020) DNA methylation signature in blood mirrors successful weight-loss during lifestyle interventions: the CENTRAL trial. Genome Med 12, 97.CrossRefGoogle ScholarPubMed
Li, X, Shao, X, Bazzano, LA et al. (2022) Blood DNA methylation at TXNIP and glycemic changes in response to weight-loss diet interventions: the POUNDS lost trial. Int J Obes 46, 11221127.CrossRefGoogle ScholarPubMed
tom Dieck, H, Döring, F, Fuchs, D et al. (2005) Transcriptome and proteome analysis identifies the pathways that increase hepatic lipid accumulation in zinc-deficient rats. J Nutr 135, 199205.CrossRefGoogle ScholarPubMed
Zhao, Y, Barrere-Cain, RE & Yang, X (2015) Nutritional systems biology of type 2 diabetes. Genes Nutr 10, 31.CrossRefGoogle ScholarPubMed
Cox, J & Mann, M (2011) Quantitative, high-resolution proteomics for data-driven systems biology. Annu Rev Biochem 80, 273299.CrossRefGoogle ScholarPubMed
Cox, J & Mann, M (2007) Is proteomics the new genomics? Cell 130, 395398.CrossRefGoogle ScholarPubMed
Arbones-Mainar, JM, Ross, K, Rucklidge, GJ et al. (2007) Extra virgin olive oils increase hepatic fat accumulation and hepatic antioxidant protein levels in APOE-/- mice. J Proteome Res 6, 40414054.CrossRefGoogle ScholarPubMed
Kim, CH, Tworoger, SS, Stampfer, MJ et al. (2018) Stability and reproducibility of proteomic profiles measured with an aptamer-based platform. Sci Rep 8, 8382.CrossRefGoogle ScholarPubMed
Haslam, DE, Li, J, Dillon, ST et al. (2022) Stability and reproducibility of proteomic profiles in epidemiological studies: comparing the Olink and SOMAscan platforms. Proteomics 22, e2100170.CrossRefGoogle ScholarPubMed
Walker, ME, Song, RJ, Xu, X et al. (2020) Proteomic and metabolomic correlates of healthy dietary patterns: the Framingham heart study. Nutrients 12, E1476.CrossRefGoogle ScholarPubMed
Warensjö Lemming, E, Byberg, L, Stattin, K et al. (2019) Dietary pattern specific protein biomarkers for cardiovascular disease: a cross-sectional study in 2 independent cohorts. J Am Heart Assoc 8, e011860.CrossRefGoogle ScholarPubMed
David, LA, Maurice, CF, Carmody, RN et al. (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559563.CrossRefGoogle ScholarPubMed
Gill, SK, Rossi, M, Bajka, B et al. (2021) Dietary fibre in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol 18, 101116.CrossRefGoogle ScholarPubMed
Sonnenburg, JL & Bäckhed, F (2016) Diet–microbiota interactions as moderators of human metabolism. Nature 535, 5664.CrossRefGoogle ScholarPubMed
Kovatcheva-Datchary, P, Nilsson, A, Akrami, R et al. (2015) Dietary fiber-induced improvement in glucose metabolism Is associated with increased abundance of Prevotella. Cell Metab 22, 971982.CrossRefGoogle ScholarPubMed
den Besten, G, van Eunen, K, Groen, AK et al. (2013) The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 54, 23252340.CrossRefGoogle ScholarPubMed
Samuel, BS, Shaito, A, Motoike, T et al. (2008) Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci USA 105, 1676716772.CrossRefGoogle ScholarPubMed
Rafiq, T, Azab, SM, Teo, KK et al. (2021) Nutritional metabolomics and the classification of dietary biomarker candidates: a critical review. Adv Nutr 12, 23332357.CrossRefGoogle ScholarPubMed
LeVatte, M, Keshteli, AH, Zarei, P et al. (2022) Applications of metabolomics to precision nutrition. Lifestyle Genom 15, 19.CrossRefGoogle ScholarPubMed
Heinzmann, SS, Brown, IJ, Chan, Q et al. (2010) Metabolic profiling strategy for discovery of nutritional biomarkers: proline betaine as a marker of citrus consumption. Am J Clin Nutr 92, 436443.CrossRefGoogle ScholarPubMed
Nokhoijav, E, Guba, A, Kumar, A et al. (2022) Metabolomic analysis of serum and tear samples from patients with obesity and type 2 diabetes mellitus. Int J Mol Sci 23, 4534.CrossRefGoogle ScholarPubMed
Yan, W, Apweiler, R, Balgley, BM et al. (2009) Systematic comparison of the human saliva and plasma proteomes. Proteomics Clin Appl 3, 116134.CrossRefGoogle ScholarPubMed
Sakanaka, A, Kuboniwa, M, Katakami, N et al. (2021) Saliva and plasma reflect metabolism altered by diabetes and periodontitis. Front Mol Biosci 8, 742002.CrossRefGoogle ScholarPubMed
Regueiro, J, Vallverdú-Queralt, A, Simal-Gándara, J et al. (2014) Urinary tartaric acid as a potential biomarker for the dietary assessment of moderate wine consumption: a randomised controlled trial. Br J Nutr 111, 16801685.CrossRefGoogle ScholarPubMed
Scalbert, A, Brennan, L, Manach, C et al. (2014) The food metabolome: a window over dietary exposure. Am J Clin Nutr 99, 12861308.CrossRefGoogle ScholarPubMed
Lépine, G, Fouillet, H, Rémond, D et al. (2021) A scoping review: metabolomics signatures associated with animal and plant protein intake and their potential relation with cardiometabolic risk. Adv Nutr 12, 21122131.CrossRefGoogle ScholarPubMed
Huang, NK, Matthan, NR, Matuszek, G et al. (2022) Plasma metabolite response to simple, refined and unrefined carbohydrate-enriched diets in older adults-randomized controlled crossover trial. Metabolites 12, 547.CrossRefGoogle ScholarPubMed
Bulló, M, Papandreou, C, Ruiz-Canela, M et al. (2021) Plasma metabolomic profiles of glycemic index, glycemic load, and carbohydrate quality Index in the PREDIMED study. J Nutr 151, 5058.CrossRefGoogle ScholarPubMed
Zheng, C, Gowda, GAN, Raftery, D et al. (2021) Evaluation of potential metabolomic-based biomarkers of protein, carbohydrate and fat intakes using a controlled feeding study. Eur J Nutr 60, 42074218.CrossRefGoogle ScholarPubMed
Garcia-Perez, I, Posma, JM, Chambers, ES et al. (2016) An analytical pipeline for quantitative characterization of dietary intake: application to assess grape intake. J Agric Food Chem 64, 24232431.CrossRefGoogle ScholarPubMed
Yin, X, Gibbons, H, Rundle, M et al. (2017) Estimation of chicken intake by adults using metabolomics-derived markers. J Nutr 147, 18501857.CrossRefGoogle ScholarPubMed
McNamara, AE, Collins, C, Harsha, PSCS et al. (2020) Metabolomic-based approach to identify biomarkers of apple intake. Mol Nutr Food Res 64, e1901158.CrossRefGoogle ScholarPubMed
Linko, A-M, Juntunen, KS, Mykkänen, HM et al. (2005) Whole-grain rye bread consumption by women correlates with plasma alkylresorcinols and increases their concentration compared with low-fiber wheat bread. J Nutr 135, 580583.CrossRefGoogle ScholarPubMed
Landberg, R, Kamal-Eldin, A, Andersson, A et al. (2008) Alkylresorcinols as biomarkers of whole-grain wheat and rye intake: plasma concentration and intake estimated from dietary records. Am J Clin Nutr 87, 832838.CrossRefGoogle ScholarPubMed
McKeown, NM, Marklund, M, Ma, J et al. (2016) Comparison of plasma alkylresorcinols (AR) and urinary AR metabolites as biomarkers of compliance in a short-term, whole-grain intervention study. Eur J Nutr 55, 12351244.CrossRefGoogle Scholar
Ross, AB, Bourgeois, A, Macharia, H et al. (2012) Plasma alkylresorcinols as a biomarker of whole-grain food consumption in a large population: results from the WHOLEheart intervention study. Am J Clin Nutr 95, 204211.CrossRefGoogle Scholar
Muli, S, Goerdten, J, Oluwagbemigun, K et al. (2021) A systematic review of metabolomic biomarkers for the intake of sugar-sweetened and low-calorie sweetened beverages. Metabolites 11, 546.CrossRefGoogle ScholarPubMed
Vázquez-Manjarrez, N, Ulaszewska, M, Garcia-Aloy, M et al. (2020) Biomarkers of intake for tropical fruits. Genes Nutr 15, 11.CrossRefGoogle ScholarPubMed
McNamara, AE, Walton, J, Flynn, A et al. (2020) The potential of multi-biomarker panels in nutrition research: total fruit intake as an example. Front Nutr 7, 577720.CrossRefGoogle ScholarPubMed
Li, C, Imamura, F, Wedekind, R et al. (2022) Development and validation of a metabolite score for red meat intake: an observational cohort study and randomized controlled dietary intervention. Am J Clin Nutr 116, 511522.CrossRefGoogle ScholarPubMed
Wedekind, R, Keski-Rahkonen, P, Robinot, N et al. (2021) Pepper alkaloids and processed meat intake: results from a randomized trial and the European prospective investigation into cancer and nutrition (EPIC) cohort. Mol Nutr Food Res 65, e2001141.CrossRefGoogle ScholarPubMed
Garcia-Perez, I, Posma, JM, Gibson, R et al. (2017) Objective assessment of dietary patterns by use of metabolic phenotyping: a randomised, controlled, crossover trial. Lancet Diabetes Endocrinol 5, 184195.CrossRefGoogle ScholarPubMed
Bhupathiraju, SN & Hu, FB (2017) One (small) step towards precision nutrition by use of metabolomics. Lancet Diabetes Endocrinol 5, 154155.CrossRefGoogle ScholarPubMed
Lindqvist, HM, Rådjursöga, M, Malmodin, D et al. (2019) Serum metabolite profiles of habitual diet: evaluation by 1H-nuclear magnetic resonance analysis. Am J Clin Nutr 110, 5362.CrossRefGoogle ScholarPubMed
Schmidt, JA, Rinaldi, S, Ferrari, P et al. (2015) Metabolic profiles of male meat eaters, fish eaters, vegetarians, and vegans from the EPIC-Oxford cohort. Am J Clin Nutr 102, 15181526.CrossRefGoogle ScholarPubMed
Rossitch, E & Bullard, DE (1988) The autonomic dysfunction syndrome: aetiology and treatment. Br J Neurosurg 2, 471478.CrossRefGoogle ScholarPubMed
Kim, H, Hu, EA, Wong K, E et al. (2021) Serum metabolites associated with healthy diets in African Americans and European Americans. J Nutr 151, 4049.CrossRefGoogle ScholarPubMed
Rebholz, CM, Lichtenstein, AH, Zheng, Z et al. (2018) Serum untargeted metabolomic profile of the dietary approaches to stop hypertension (DASH) dietary pattern. Am J Clin Nutr 108, 243255.CrossRefGoogle ScholarPubMed
Willis, ND, Lloyd, AJ, Xie, L et al. (2020) Design and characterisation of a randomized food intervention that mimics exposure to a typical UK diet to provide urine samples for identification and validation of metabolite biomarkers of food intake. Front Nutr 7, 561010.CrossRefGoogle ScholarPubMed
Rothwell, JA, Fillâtre, Y, Martin, J-F et al. (2014) New biomarkers of coffee consumption identified by the non-targeted metabolomic profiling of cohort study subjects. PLoS ONE 9, e93474.CrossRefGoogle ScholarPubMed
Drouin-Chartier, J-P (2022) Plasma lipidomic profiles of dairy consumption: a new window on their cardiometabolic effects. Hypertension 79, 16291632.CrossRefGoogle ScholarPubMed
Yun, H, Sun, L, Wu, Q et al. (2022) Lipidomic signatures of dairy consumption and associated changes in blood pressure and other cardiovascular risk factors among Chinese adults. Hypertension 79, 16171628.CrossRefGoogle ScholarPubMed
Li, J, Guasch-Ferré, M, Chung, W et al. (2020) The Mediterranean diet, plasma metabolome, and cardiovascular disease risk. Eur Heart J 41, 26452656.CrossRefGoogle ScholarPubMed
Kim, H, Yu, B, Li, X et al. (2022) Serum metabolomic signatures of plant-based diets and incident chronic kidney disease. Am J Clin Nutr 116, 151164.CrossRefGoogle ScholarPubMed
Wang, F, Baden, MY, Guasch-Ferré, M et al. (2022) Plasma metabolite profiles related to plant-based diets and the risk of type 2 diabetes. Diabetologia 65, 11191132.CrossRefGoogle ScholarPubMed
Smith, E, Ericson, U, Hellstrand, S et al. (2022) A healthy dietary metabolic signature is associated with a lower risk for type 2 diabetes and coronary artery disease. BMC Med 20, 122.CrossRefGoogle ScholarPubMed
Floegel, A, Stefan, N, Yu, Z et al. (2013) Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes 62, 639648.CrossRefGoogle ScholarPubMed
Alshehry, ZH, Mundra, PA, Barlow, CK et al. (2016) Plasma lipidomic profiles improve on traditional risk factors for the prediction of cardiovascular events in type 2 diabetes mellitus. Circulation 134, 16371650.CrossRefGoogle ScholarPubMed
Hang, D, Zeleznik, OA, He, X et al. (2020) Metabolomic signatures of long-term coffee consumption and risk of type 2 diabetes in women. Diabetes Care 43, 25882596.CrossRefGoogle ScholarPubMed
Kim, Y, Lu, S, Ho, JE et al. (2021) Proteins as mediators of the association between diet quality and incident cardiovascular disease and all-cause mortality: the Framingham heart study. J Am Heart Assoc 10, e021245.CrossRefGoogle ScholarPubMed
Koeth, RA, Wang, Z, Levison, BS et al. (2013) Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 19, 576585.CrossRefGoogle ScholarPubMed
Tang, WHW, Wang, Z, Levison, BS et al. (2013) Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 368, 15751584.CrossRefGoogle ScholarPubMed
Li, J, Li, Y, Ivey, KL et al. (2022) Interplay between diet and gut microbiome, and circulating concentrations of trimethylamine N-oxide: findings from a longitudinal cohort of US men. Gut 71, 724733.CrossRefGoogle ScholarPubMed
Qi, Q, Li, J, Yu, B et al. (2022) Host and gut microbial tryptophan metabolism and type 2 diabetes: an integrative analysis of host genetics, diet, gut microbiome and circulating metabolites in cohort studies. Gut 71, 10951105.CrossRefGoogle ScholarPubMed
Zeevi, D, Korem, T, Zmora, N et al. (2015) Personalized nutrition by prediction of glycemic responses. Cell 163, 10791094.CrossRefGoogle ScholarPubMed
Price, ND, Magis, AT, Earls, JC et al. (2017) A wellness study of 108 individuals using personal, dense, dynamic data clouds. Nat Biotechnol 35, 747756.CrossRefGoogle ScholarPubMed
Celis-Morales, C, Livingstone, KM, Marsaux, CF et al. (2017) Effect of personalized nutrition on health-related behaviour change: evidence from the Food4Me European randomized controlled trial. Int J Epidemiol 46, 578588.Google ScholarPubMed
Merino, J (2022) Precision nutrition in diabetes: when population-based dietary advice gets personal. Diabetologia 65, 18391848.CrossRefGoogle ScholarPubMed
Adams, SH, Anthony, JC, Carvajal, R et al. (2020) Perspective: guiding principles for the implementation of personalized nutrition approaches that benefit health and function. Adv Nutr 11, 2534.CrossRefGoogle ScholarPubMed
Ramos-Lopez, O, Milagro, FI, Riezu-Boj, JI et al. (2021) Epigenetic signatures underlying inflammation: an interplay of nutrition, physical activity, metabolic diseases, and environmental factors for personalized nutrition. Inflamm Res 70, 2949.CrossRefGoogle ScholarPubMed