Challenges and Opportunities of Metabolic Profiling

Last Updated: May 16, 2023


Disclosure: None
Pub Date: Thursday, Mar 30, 2017
Author: Catriona Syme, PhD and Zdenka Pausova, MD, FAHA
Affiliation: The Hospital for Sick Children, University of Toronto, Toronto, Canada , Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada

Metabolomics is a comprehensive study of metabolites and metabolism – it measures simultaneously the levels of a large number of metabolites in a biosample, providing a snapshot of the metabolic activity in the sample. The metabolite levels are determined largely by genetics1, but are also influenced by environmental exposures, including the gut microbiome, diet, and physical activity. With the characterization of metabolites in relation to cardiovascular health, disease, and interventions, metabolomics has the potential to identify novel biomarkers for diagnostics and drug targets for therapy, uncover pathobiology of disease, and monitor treatment response.

Metabolomics technologies are continually evolving. As they become more sensitive, they are able to distinguish more and more metabolites2. A recent study using ultra-high resolution metabolomics yielded close to 10,000 distinct signals between the mass-to-charge ratio (m/z) range of 85 to 12753. Beyond detecting these unique spectral signals, however, identification of the chemical structure remains a significant challenge. Typically, only a fraction can be identified and the process can take weeks or months of analysis2. Several spectral databases exist to aid in the identification4. The Human Metabolome Database lists over 40,000 metabolites that have been detected, or that are expected to be present, in biosamples (hmdb.ca). In this AHA statement on metabolomics in cardiovascular disease (CVD), the authors provide an in-depth overview of the many challenges regarding technology, processing, standardization and analytics that need to be addressed for metabolomics to have the expected considerable impact on diagnostics and therapeutics.

Despite the existing challenges, metabolomics is already providing new insight into etiology, biomarkers, and interventions for CVD. Trimethylamine N-oxide (TMAO) is an example of one identified metabolite that is providing insight into all of the above. Animal studies have shown it to be an atherotoxin5 that disrupts cholesterol balance6 (etiology). It correlates strongly with subsequent adverse myocardial events7 (biomarker), and it is a byproduct of trimethylamine, which gut microorganisms produce in response to meat and phospholipids in the diet5. As such, it represents a mechanism of atherosclerosis that could be treated or prevented by management of diet and/or the microbiome (intervention). Indeed two novel protein targets to reduce its levels have been identified8.

Similarly, metabolomics studies have shown that branched-chain amino acids (BCAA), which are insulin analogues, associate with insulin resistance9-11, and predict development of diabetes12. Obesity is not always associated with metabolic complications, and in metabolically healthy individuals with overweight/obesity, BCAA are lower than in those with complications13. Their levels can be affected by both diet and the gut microbiome14. Animal studies have shown that a high-fat diet with BCAA caused insulin resistance, while high-fat diet alone or standard chow with BCAA did not10. BCAA are also associated with coronary artery disease, even after adjusting for other CVD-risk factors15, 16.

Metabolomics studies may unveil common metabolites key to other health complications that are associated with cardiometabolic disturbances. For example, systemic inflammation is proposed to be related to both atherosclerosis and dementia17-20. Recently, small subsets of metabolites were identified that predicted both incident coronary artery disease21 and Alzheimer’s disease22 in large-scale prospective studies. One circulating lysophosphatidylcholine, PC 18:2/0:0, was common to both subsets. Lysophosphatidylcholines are glycerophosphocholine metabolites. They are lipid species that are involved in the modulation of systemic inflammation; they influence signaling and phagocytic functions of immune cells, as well as their migratory and adhesive behaviour and thus their capacity to cross endothelial barriers23-25.

Metabolomics is also redefining dyslipidemia. Traditionally assessed by ‘high-abundance’ lipids, such as cholesterols and triacylglycerols, which circulate at millimolar blood levels, recent research in older individuals suggests that ‘low-abundance’ lipids, including glycerophosphocholine metabolites, may improve the prediction of CVD outcomes21, 26-28. These lipids circulate at micro- or nano-molar blood levels29-32, and advances in mass spectrometry enable their quantification. An untargeted lipidomics study identified several lipid species that predicted CVD outcomes; a few were glycerophosphocholine metabolites21. Recently, one of the profiled species, that was associated with pre-clinical CVD-risk factors (adiposity and triacylglycerols)21 in older adults, was also reported to be associated with higher CVD-risk factors in a much younger population-based cohort of adolescents33. Further, using targeted lipidomics in the adolescent cohort, novel glycerophosphocholine metabolites were identified and quantified that showed even stronger associations with these risk factors33. Further research is needed to assess the clinical relevance of these novel metabolites to CVD outcomes.

Further, metabolomics will be influential in drug development and precision medicine. For example, individual patient responses to drugs can be highly variable. Metabolomics can be used to assess drug metabolism, toxicity, and patient compliance, and to identify responders/non-responders (reviewed in34). Combined with the potential of metabolomics to provide biomarkers of disease risk, this ability to find effective drugs with optimal dosing will enable personalized plans for prevention and treatment, respectively.

Moving forward, as clearly stated in this AHA statement on metabolomics in CVD, methodological and analytic standards must be implemented, and high throughput technologies advanced. Metabolomic profiling of large, well phenotyped cohorts is needed to characterize the influence of intrinsic (e.g., genotype, sex, age, etc.) and extrinsic (e.g., diet, exercise, smoking, etc.) factors. Prospective studies are required to characterize metabolomic variations associated with lifestyle changes (i.e., in diet, physical activity, smoking, and medications) and comorbidities, and to distinguish metabolomics signatures that are causes vs. consequences of disease. Replication/validation studies are key to the generalizability of findings. While metabolomics will clearly aid in identifying biological pathways and risk prediction, it may also enable disease prevention through identification of ‘protective’ and ‘harmful’ metabolites and their sources (endogenous vs. exogenous). Determining the functional roles of novel metabolites in health and disease are needed to identify their relevance to clinical outcomes. It is well established that CVD has a long pre-clinical phase, which may start as early as during childhood and adolescence35. Conceivably, longitudinal studies could ascertain whether certain metabolomic profiles in youth, prior to disease onset, could identify those at increased risk of developing CVD, opening a window for prevention. Each of these considerations are relevant not only for ongoing research, but also must be addressed before translation to clinical practice will be an option.

Citation


Cheng S, Shah SH, Corwin EJ, Fiehn O, Fitzgerald RL, Gerszten RE, Illig T, Rhee EP, Srinivas PR, Wang TJ, Jain M; on behalf of the American Heart Association Council on Genomic and Precision Medicine; Council on Cardiovascular and Stroke Nursing; Council on Clinical Cardiology; and Stroke Council. Potential impact and study considerations of metabolomics in cardiovascular health and disease: a scientific statement from the American Heart Association [published online ahead of print Thursday, March 30, 2017]. Circ Cardiovasc Genet. doi: 10.1161/HCG.0000000000000032.

References


  1. Shin SY, Fauman EB, Petersen AK, Krumsiek J, Santos R, Huang J, Arnold M, Erte I, Forgetta V, Yang TP, Walter K, Menni C, Chen L, Vasquez L, Valdes AM, Hyde CL, Wang V, Ziemek D, Roberts P, Xi L, Grundberg E, Multiple Tissue Human Expression Resource C, Waldenberger M, Richards JB, Mohney RP, Milburn MV, John SL, Trimmer J, Theis FJ, Overington JP, Suhre K, Brosnan MJ, Gieger C, Kastenmuller G, Spector TD and Soranzo N. An atlas of genetic influences on human blood metabolites. Nature genetics. 2014;46:543-50.
  2. Zamboni N, Saghatelian A and Patti GJ. Defining the metabolome: size, flux, and regulation. Mol Cell. 2015;58:699-706.
  3. Jin R, Banton S, Tran VT, Konomi JV, Li S, Jones DP and Vos MB. Amino Acid Metabolism is Altered in Adolescents with Nonalcoholic Fatty Liver Disease-An Untargeted, High Resolution Metabolomics Study. J Pediatr. 2016;172:14-19 e5.
  4. Alonso A, Marsal S and Julia A. Analytical methods in untargeted metabolomics: state of the art in 2015. Front Bioeng Biotechnol. 2015;3:23.
  5. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH, Bushman FD, Lusis AJ and Hazen SL. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nature medicine. 2013;19:576-85.
  6. Warrier M, Shih DM, Burrows AC, Ferguson D, Gromovsky AD, Brown AL, Marshall S, McDaniel A, Schugar RC, Wang Z, Sacks J, Rong X, Vallim TA, Chou J, Ivanova PT, Myers DS, Brown HA, Lee RG, Crooke RM, Graham MJ, Liu X, Parini P, Tontonoz P, Lusis AJ, Hazen SL, Temel RE and Brown JM. The TMAO-Generating Enzyme Flavin Monooxygenase 3 Is a Central Regulator of Cholesterol Balance. Cell Rep. 2015.
  7. Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y and Hazen SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575-84.
  8. Brown JM and Hazen SL. The gut microbial endocrine organ: bacterially derived signals driving cardiometabolic diseases. Annu Rev Med. 2015;66:343-59.
  9. Felig P, Marliss E and Cahill GF, Jr. Plasma amino acid levels and insulin secretion in obesity. N Engl J Med. 1969;281:811-6.
  10. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, Haqq AM, Shah SH, Arlotto M, Slentz CA, Rochon J, Gallup D, Ilkayeva O, Wenner BR, Yancy WS, Jr., Eisenson H, Musante G, Surwit RS, Millington DS, Butler MD and Svetkey LP. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9:311-26.
  11. Shaham O, Wei R, Wang TJ, Ricciardi C, Lewis GD, Vasan RS, Carr SA, Thadhani R, Gerszten RE and Mootha VK. Metabolic profiling of the human response to a glucose challenge reveals distinct axes of insulin sensitivity. Mol Syst Biol. 2008;4:214.
  12. Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, Lewis GD, Fox CS, Jacques PF, Fernandez C, O'Donnell CJ, Carr SA, Mootha VK, Florez JC, Souza A, Melander O, Clish CB and Gerszten RE. Metabolite profiles and the risk of developing diabetes. Nature medicine. 2011;17:448-53.
  13. Batch BC, Shah SH, Newgard CB, Turer CB, Haynes C, Bain JR, Muehlbauer M, Patel MJ, Stevens RD, Appel LJ, Newby LK and Svetkey LP. Branched chain amino acids are novel biomarkers for discrimination of metabolic wellness. Metabolism. 2013;62:961-9.
  14. Neis EP, Dejong CH and Rensen SS. The role of microbial amino acid metabolism in host metabolism. Nutrients. 2015;7:2930-46.
  15. Bhattacharya S, Granger CB, Craig D, Haynes C, Bain J, Stevens RD, Hauser ER, Newgard CB, Kraus WE, Newby LK and Shah SH. Validation of the association between a branched chain amino acid metabolite profile and extremes of coronary artery disease in patients referred for cardiac catheterization. Atherosclerosis. 2014;232:191-6.
  16. Shah SH, Bain JR, Muehlbauer MJ, Stevens RD, Crosslin DR, Haynes C, Dungan J, Newby LK, Hauser ER, Ginsburg GS, Newgard CB and Kraus WE. Association of a peripheral blood metabolic profile with coronary artery disease and risk of subsequent cardiovascular events. Circ Cardiovasc Genet. 2010;3:207-14.
  17. Libby P. Inflammation in atherosclerosis. Arterioscler Thromb Vasc Biol. 2012;32:2045-51.
  18. Engelhart MJ, Geerlings MI, Meijer J, Kiliaan A, Ruitenberg A, van Swieten JC, Stijnen T, Hofman A, Witteman JC and Breteler MM. Inflammatory proteins in plasma and the risk of dementia: the rotterdam study. Arch Neurol. 2004;61:668-72.
  19. Schmidt R, Schmidt H, Curb JD, Masaki K, White LR and Launer LJ. Early inflammation and dementia: a 25-year follow-up of the Honolulu-Asia Aging Study. Annals of neurology. 2002;52:168-74.
  20. Desikan RS, Schork AJ, Wang Y, Thompson WK, Dehghan A, Ridker PM, Chasman DI, McEvoy LK, Holland D, Chen CH, Karow DS, Brewer JB, Hess CP, Williams J, Sims R, O'Donovan MC, Choi SH, Bis JC, Ikram MA, Gudnason V, DeStefano AL, van der Lee SJ, Psaty BM, van Duijn CM, Launer L, Seshadri S, Pericak-Vance MA, Mayeux R, Haines JL, Farrer LA, Hardy J, Ulstein ID, Aarsland D, Fladby T, White LR, Sando SB, Rongve A, Witoelar A, Djurovic S, Hyman BT, Snaedal J, Steinberg S, Stefansson H, Stefansson K, Schellenberg GD, Andreassen OA, Dale AM, Inflammation Working G, International Genomics of Alzheimer's Disease P and DemGene I. Polygenic Overlap Between C-Reactive Protein, Plasma Lipids, and Alzheimer Disease. Circulation. 2015;131:2061-9.
  21. Ganna A, Salihovic S, Sundstrom J, Broeckling CD, Hedman AK, Magnusson PK, Pedersen NL, Larsson A, Siegbahn A, Zilmer M, Prenni J, Arnlov J, Lind L, Fall T and Ingelsson E. Large-scale metabolomic profiling identifies novel biomarkers for incident coronary heart disease. PLoS genetics. 2014;10:e1004801.
  22. Mapstone M, Cheema AK, Fiandaca MS, Zhong X, Mhyre TR, MacArthur LH, Hall WJ, Fisher SG, Peterson DR, Haley JM, Nazar MD, Rich SA, Berlau DJ, Peltz CB, Tan MT, Kawas CH and Federoff HJ. Plasma phospholipids identify antecedent memory impairment in older adults. Nature medicine. 2014;20:415-8.
  23. Yan JJ, Jung JS, Lee JE, Lee J, Huh SO, Kim HS, Jung KC, Cho JY, Nam JS, Suh HW, Kim YH and Song DK. Therapeutic effects of lysophosphatidylcholine in experimental sepsis. Nature medicine. 2004;10:161-7.
  24. Sevastou I, Kaffe E, Mouratis MA and Aidinis V. Lysoglycerophospholipids in chronic inflammatory disorders: the PLA(2)/LPC and ATX/LPA axes. Biochimica et biophysica acta. 2013;1831:42-60.
  25. Marathe GK, Pandit C, Lakshmikanth CL, Chaithra VH, Jacob SP and D'Souza CJ. To hydrolyze or not to hydrolyze: the dilemma of platelet-activating factor acetylhydrolase. Journal of lipid research. 2014;55:1847-54.
  26. Stegemann C, Pechlaner R, Willeit P, Langley SR, Mangino M, Mayr U, Menni C, Moayyeri A, Santer P, Rungger G, Spector TD, Willeit J, Kiechl S and Mayr M. Lipidomics profiling and risk of cardiovascular disease in the prospective population-based Bruneck study. Circulation. 2014;129:1821-31.
  27. Meikle PJ, Wong G, Tsorotes D, Barlow CK, Weir JM, Christopher MJ, MacIntosh GL, Goudey B, Stern L, Kowalczyk A, Haviv I, White AJ, Dart AM, Duffy SJ, Jennings GL and Kingwell BA. Plasma lipidomic analysis of stable and unstable coronary artery disease. Arteriosclerosis, thrombosis, and vascular biology. 2011;31:2723-32.
  28. Jove M, Mauri-Capdevila G, Suarez I, Cambray S, Sanahuja J, Quilez A, Farre J, Benabdelhak I, Pamplona R, Portero-Otin M and Purroy F. Metabolomics predicts stroke recurrence after transient ischemic attack. Neurology. 2015;84:36-45.
  29. Hinterwirth H, Stegemann C and Mayr M. Lipidomics: quest for molecular lipid biomarkers in cardiovascular disease. Circulation Cardiovascular genetics. 2014;7:941-54.
  30. Xu H, Valenzuela N, Fai S, Figeys D and Bennett SA. Targeted lipidomics - advances in profiling lysophosphocholine and platelet-activating factor second messengers. The FEBS journal. 2013;280:5652-67.
  31. Shevchenko A and Simons K. Lipidomics: coming to grips with lipid diversity. Nat Rev Mol Cell Biol. 2010;11:593-8.
  32. Quehenberger O, Armando AM, Brown AH, Milne SB, Myers DS, Merrill AH, Bandyopadhyay S, Jones KN, Kelly S, Shaner RL, Sullards CM, Wang E, Murphy RC, Barkley RM, Leiker TJ, Raetz CR, Guan Z, Laird GM, Six DA, Russell DW, McDonald JG, Subramaniam S, Fahy E and Dennis EA. Lipidomics reveals a remarkable diversity of lipids in human plasma. J Lipid Res. 2010;51:3299-305.
  33. Syme C, Czajkowski S, Shin J, Abrahamowicz M, Leonard G, Perron M, Richer L, Veillette S, Gaudet D, Strug L, Wang Y, Xu H, Taylor G, Paus T, Bennett S and Pausova Z. Glycerophosphocholine Metabolites and Cardiovascular Disease Risk Factors in Adolescents: A Cohort Study. Circulation. 2016;134:1629-1636.
  34. Wishart DS. Emerging applications of metabolomics in drug discovery and precision medicine. Nature reviews Drug discovery. 2016;15:473-84.
  35. Berenson GS, Srinivasan SR, Bao W, Newman WP, 3rd, Tracy RE and Wattigney WA. Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. The Bogalusa Heart Study. The New England journal of medicine. 1998;338:1650-6.

Science News Commentaries

View All Science News Commentaries

-- The opinions expressed in this commentary are not necessarily those of the editors or of the American Heart Association --