Article Data

  • Views 2362
  • Dowloads 249

Mini-Reviews

Open Access

The role of metabolomics in myocardial infarction: a recent mini-review

  • Xuguang Miao1
  • Cui Song1,*,
  • Pu Zhen2,*,

1Department of Cardiology, Beijing Anzhen Hospital, Affiliated with Capital Medical University, 100029 Beijing, China

2Department of Cardiology, People's Hospital of Lhasa, 850000 Lhasa, Tibet Autonomous Region, China

DOI: 10.22514/sv.2021.252 Vol.19,Issue 1,January 2023 pp.34-42

Submitted: 15 August 2021 Accepted: 10 November 2021

Published: 08 January 2023

*Corresponding Author(s): Cui Song E-mail: cuisongdoctor@163.com
*Corresponding Author(s): Pu Zhen E-mail: 18008919066@163.com

Abstract

Myocardial infarction (MI) is one of the most common and important causes of heart failure in critical care and emergency medicine. Incidence of MI and MI-related mortality have been on the rise in the recent past. Metabolomics is a new field that entails analysis of profiles of metabolites (<1250 Da) in living organisms. Currently, several studies have extensively explored the application of metabolomics in medical field. In the current article, the emerging applications of next-generation metabolomics on MI in the previous 10 years were reviewed. The present article thus provides references for further use of metabolomics to guide clinical treatment and provides a basis for prevention of cardiovascular events.


Keywords

Myocardial infarction; Metabolomics; Metabolic biomarkers; Occurrence prediction; Molecular mechanisms


Cite and Share

Xuguang Miao,Cui Song,Pu Zhen. The role of metabolomics in myocardial infarction: a recent mini-review. Signa Vitae. 2023. 19(1);34-42.

References

[1] Gill R, Kuriakose R, Gertz ZM, Salloum FN, Xi L, Kukreja RC. Remote ischemic preconditioning for myocardial protection: update on mechanisms and clinical relevance. Molecular and Cellular Biochemistry. 2015; 402: 41–49.

[2] Keller T, Zeller T, Peetz D, Tzikas S, Roth A, Czyz E, et al. Sensitive troponin i assay in early diagnosis of acute myocardial infarction. The New England Journal of Medicine. 2009; 361: 868–877.

[3] Fiehn O. Metabolomics by Gas Chromatography-Mass Spectrometry: Combined Targeted and Untargeted Profiling. Current Protocols in Molecular Biology. 2016; 114: 30.4.1–30.4.32.

[4] Lewis GD, Asnani A, Gerszten RE. Application of metabolomics to cardiovascular biomarker and pathway discovery. Journal of the American College of Cardiology. 2008; 52: 117–123.

[5] Wang J, Sun Y, Teng S, Li K. Prediction of sepsis mortality using metabolite biomarkers in the blood: a meta-analysis of death-related pathways and prospective validation. BMC Medicine. 2020; 18: 83.

[6] Li K, Naviaux JC, Bright AT, Wang L, Naviaux RK. A robust, single-injection method for targeted, broad-spectrum plasma metabolomics. Metabolomics. 2017; 13: 122.

[7] Li K, Naviaux JC, Monk JM, Wang L, Naviaux RK. Improved Dried Blood Spot-Based Metabolomics: A Targeted, Broad-Spectrum, Single-Injection Method. Metabolites. 2020; 10: 82.

[8] Cheng S, Shah SH, Corwin EJ, Fiehn O, Fitzgerald RL, Gerszten RE, et al. Potential Impact and Study Considerations of Metabolomics in Cardiovascular Health and Disease: A Scientific Statement from the American Heart Association. Circulation. Cardiovascular Genetics. 2017; 10: e000032.

[9] Barba I, Andrés M, Garcia-Dorado D. Metabolomics and Heart Diseases: From Basic to Clinical Approach. Current Medicinal Chemistry. 2019; 26: 46–59.

[10] McGarrah RW, Crown SB, Zhang G, Shah SH, Newgard CB. Cardiovas-cular Metabolomics. Circulation Research. 2018; 122: 1238–1258.

[11] Cui S, Li K, Ang L, Liu J, Cui L, Song X, et al. Plasma Phospholipids and Sphingolipids Identify Stent Restenosis after Percutaneous Coronary Intervention. JACC: Cardiovascular Interventions. 2017; 10: 1307–1316.

[12] Cui S, Li L, Zhang Y, Lu J, Wang X, Song X, et al. Machine Learning Identifies Metabolic Signatures that Predict the Risk of Recurrent Angina in Remitted Patients after Percutaneous Coronary Intervention: A Multicenter Prospective Cohort Study. Advanced Science. 2021; 8: 2003893.

[13] Al-Obaidi MK, Stubbs PJ, Collinson P, Conroy R, Graham I, Noble MI. Elevated homocysteine levels are associated with increased ischemic myocardial injury in acute coronary syndromes. Journal of the American College of Cardiology. 2000; 36: 1217–1222.

[14] Page JH, Ma J, Chiuve SE, Stampfer MJ, Selhub J, Manson JE, et al. Plasma total cysteine and total homocysteine and risk of myocardial infarction in women: a prospective study. American Heart Journal. 2010; 159: 599–604.

[15] Khan A, Choi Y, Back JH, Lee S, Jee SH, Park YH. High-resolution metabolomics study revealing l-homocysteine sulfinic acid, cysteic acid, and carnitine as novel biomarkers for high acute myocardial infarction risk. Metabolism. 2020; 104: 154051.

[16] Tayama J, Munakata M, Yoshinaga K, Toyota T. Higher plasma homocysteine concentration is associated with more advanced systemic arterial stiffness and greater blood pressure response to stress in hypertensive patients. Hypertension Research. 2006; 29: 403–409.

[17] Saibeni S, Lecchi A, Meucci G, Cattaneo M, Tagliabue L, Rondonotti E, et al. Prevalence of hyperhomocysteinemia in adult gluten-sensitive enteropathy at diagnosis: role of B12, folate, and genetics. Clinical Gastroenterology and Hepatology. 2005; 3: 574–580.

[18] Yoon H, Jeong Yang J, Rivera ES, Shu X, Xiang Y, Calcutt MW, et al. Urinary metabolites and risk of coronary heart disease: a prospective investigation among urban Chinese adults. Nutrition, Metabolism and Cardiovascular Diseases. 2020; 30: 467–473.

[19] Wang Y, Liu H, McKenzie G, Witting PK, Stasch J, Hahn M, et al. Kynurenine is an endothelium-derived relaxing factor produced during inflammation. Nature Medicine. 2010; 16: 279–285.

[20] Tong Q, Song J, Yang G, Fan L, Xiong W, Fang J. Simultaneous determination of tryptophan, kynurenine, kynurenic acid, xanthurenic acid and 5-hydroxytryptamine in human plasma by LC-MS/MS and its application to acute myocardial infarction monitoring. Biomedical Chromatography. 2018; 32.

[21] Ban Y, Watanabe T, Miyazaki A, Nakano Y, Tobe T, Idei T, et al. Impact of increased plasma serotonin levels and carotid atherosclerosis on vascular dementia. Atherosclerosis. 2007; 195: 153–159.

[22] Floegel A, Kühn T, Sookthai D, Johnson T, Prehn C, Rolle-Kampczyk U, et al. Serum metabolites and risk of myocardial infarction and ischemic stroke: a targeted metabolomic approach in two German prospective cohorts. European Journal of Epidemiology. 2018; 33: 55–66.

[23] Zhu M, Han Y, Zhang Y, Zhang S, Wei C, Cong Z, et al. Metabolomics Study of the Biochemical Changes in the Plasma of Myocardial Infarction Patients. Frontiers in Physiology. 2018; 9: 1017.

[24] Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011; 472: 57–63.

[25] Law SH, Chan ML, Marathe GK, Parveen F, Chen CH, Ke LY. An Updated Review of Lysophosphatidylcholine Metabolism in Human Diseases. International Journal of Molecular Sciences. 2019; 20: 1149.

[26] Kogot-Levin A, Saada A. Ceramide and the mitochondrial respiratory chain. Biochimie. 2014; 100: 88–94.

[27] Pan W, Li L, Sun M, Wang C, Fang S, Yu B. Plasma ceramides are associated with coronary atherosclerotic burden in patients with ST-segment elevation myocardial infarction. International Journal of Cardiology. 2020; 320: 155–160.

[28] Jozefczuk E, Guzik TJ, Siedlinski M. Significance of sphingosine-1-phosphate in cardiovascular physiology and pathology. Pharmacological Research. 2020; 156: 104793.

[29] Nielsen MS, Schmidt EB, Stegger J, Gorst-Rasmussen A, Tjonneland A, Overvad K. Adipose tissue arachidonic acid content is associated with the risk of myocardial infarction: a Danish case-cohort study. Atherosclerosis. 2013; 227: 386–390.

[30] Sun Y, Koh HWL, Choi H, Koh W, Yuan J, Newman JW, et al. Plasma fatty acids, oxylipins, and risk of myocardial infarction: the Singapore Chinese Health Study. Journal of Lipid Research. 2016; 57: 1300–1307.

[31] Sun CW, Falck JR, Harder DR, Roman RJ. Role of tyrosine kinase and PKC in the vasoconstrictor response to 20-HETE in renal arterioles. Hypertension. 1999; 33: 414–418.

[32] Fan F, Sun CW, Maier KG, Williams JM, Pabbidi MR, Didion SP, et al. 20-Hydroxyeicosatetraenoic acid contributes to the inhibition of K+ channel activity and vasoconstrictor response to angiotensin II in rat renal microvessels. PLoS ONE. 2013; 8: e82482.

[33] Cheng J, Wu C, Gotlinger KH, Zhang F, Falck JR, Narsimhaswamy D, et al. 20-hydroxy-5,8,11,14-eicosatetraenoic acid mediates endothe-lial dysfunction via IkappaB kinase-dependent endothelial nitric-oxide synthase uncoupling. The Journal of Pharmacology and Experimental Therapeutics. 2010; 332: 57–65.

[34] Yuhki K, Kojima F, Kashiwagi H, Kawabe J, Fujino T, Narumiya S, et al. Roles of prostanoids in the pathogenesis of cardiovascular diseases: Novel insights from knockout mouse studies. Pharmacology and Therapeutics. 2011; 129: 195–205.

[35] Wolf G. Adipose-specific phospholipase as regulator of adiposity. Nutrition Reviews. 2009; 67: 551–554.

[36] Zordoky BNM, El-Kadi AOS. Effect of cytochrome P450 polymorphism on arachidonic acid metabolism and their impact on cardiovascular diseases. Pharmacology and Therapeutics. 2010; 125: 446–463.

[37] Kim M, Kim M, Yoo HJ, Lee A, Jeong S, Lee JH. Associations among FADS1 rs174547, eicosapentaenoic acid/arachidonic acid ratio, and arterial stiffness in overweight subjects. Prostaglandins, Leukotrienes, and Essential Fatty Acids. 2018; 130: 11–18.

[38] Ali SE, Farag MA, Holvoet P, Hanafi RS, Gad MZ. A Comparative Metabolomics Approach Reveals Early Biomarkers for Metabolic Response to Acute Myocardial Infarction. Scientific Reports. 2016; 6: 36359.

[39] Sansbury BE, DeMartino AM, Xie Z, Brooks AC, Brainard RE, Watson LJ, et al. Metabolomic analysis of pressure-overloaded and infarcted mouse hearts. Circulation. Heart Failure. 2014; 7: 634–642.

[40] Zhang Y, Yuan S, Pu J, Yang L, Zhou X, Liu L, et al. Integrated Metabolomics and Proteomics Analysis of Hippocampus in a Rat Model of Depression. Neuroscience. 2018; 371: 207–220.

[41] Gray LR, Tompkins SC, Taylor EB. Regulation of pyruvate metabolism and human disease. Cellular and Molecular Life Sciences. 2014; 71: 2577–2604.

[42] Bodi V, Sanchis J, Morales JM, Marrachelli VG, Nunez J, Forteza MJ, et al. Metabolomic profile of human myocardial ischemia by nuclear magnetic resonance spectroscopy of peripheral blood serum: a translational study based on transient coronary occlusion models. Journal of the American College of Cardiology. 2012; 59: 1629–1641.

[43] Eckle T, Köhler D, Lehmann R, El Kasmi K, Eltzschig HK. Hypoxia-inducible factor-1 is central to cardioprotection: a new paradigm for ischemic preconditioning. Circulation. 2008; 118: 166–175.

[44] Kido M, Du L, Sullivan CC, Li X, Deutsch R, Jamieson SW, et al. Hypoxia-inducible factor 1-alpha reduces infarction and attenuates progression of cardiac dysfunction after myocardial infarction in the mouse. Journal of the American College of Cardiology. 2005; 46: 2116–2124.

[45] Guth BD, Wisneski JA, Neese RA, White FC, Heusch G, Mazer CD, et al. Myocardial lactate release during ischemia in swine. Relation to regional blood flow. Circulation. 1990; 81: 1948–1958.

[46] Moselhy SS, Demerdash SH. Serum free L-carnitine in association with myoglobin as a diagnostic marker of acute myocardial infarction. Clinical Biochemistry. 2009; 42: 78–82.

[47] Subramaniam S, Fletcher C. Trimethylamine N-oxide: breathe new life. British Journal of Pharmacology. 2018; 175: 1344–1353.

[48] Wang Z, Liu Y, Liu G, Lu H, Mao C. L -Carnitine and heart disease. Life Sciences. 2018; 194: 88–97.

[49] Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nature Medicine. 2013; 19: 576–585.

[50] DiNicolantonio JJ, Lavie CJ, Fares H, Menezes AR, O’Keefe JH. L- carnitine in the secondary prevention of cardiovascular disease: systematic review and meta-analysis. Mayo Clinic Proceedings. 2013; 88: 544–551.

[51] Nam M, Jung Y, Ryu DH, Hwang G. A metabolomics-driven approach reveals metabolic responses and mechanisms in the rat heart following myocardial infarction. International Journal of Cardiology. 2017; 227: 239–246.

[52] Graham I. Homocysteine in health and disease. Annals of Internal Medicine. 1999; 131: 387–388.

[53] Jing L, He M, Chang Y, Mehta SL, He Q, Zhang J, et al. Coenzyme Q10 protects astrocytes from ROS-induced damage through inhibition of mitochondria-mediated cell death pathway. International Journal of Biological Sciences. 2015; 11: 59–66.

[54] Westman PC, Lipinski MJ, Luger D, Waksman R, Bonow RO, Wu E, et al. Inflammation as a Driver of Adverse Left Ventricular Remodeling after Acute Myocardial Infarction. Journal of the American College of Cardiology. 2016; 67: 2050–2060.

[55] Tousoulis D, Psaltopoulou T, Androulakis E, Papageorgiou N, Papaioan-nou S, Oikonomou E, et al. Oxidative stress and early atherosclerosis: novel antioxidant treatment. Cardiovascular Drugs and Therapy. 2015; 29: 75–88.

[56] Witting PK, Pettersson K, Letters J, Stocker R. Anti-atherogenic effect of coenzyme Q10 in apolipoprotein E gene knockout mice. Free Radical Biology and Medicine. 2000; 29: 295–305.

[57] Kuettner A, Pieper A, Koch J, Enzmann F, Schroeder S. Influence of coenzyme Q(10) and cerivastatin on the flow-mediated vasodilation of the brachial artery: results of the ENDOTACT study. International Journal of Cardiology. 2005; 98: 413–419.

[58] Kohlhauer M, Dawkins S, Costa ASH, Lee R, Young T, Pell VR, et al. Metabolomic Profiling in Acute ST‐Segment–Elevation Myocardial Infarction Identifies Succinate as an Early Marker of Human Ischemia–Reperfusion Injury. Journal of the American Heart Association. 2018; 7: e007546.

[59] Chouchani ET, Pell VR, Gaude E, Aksentijević D, Sundier SY, Robb EL, et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature. 2014; 515: 431–435.

[60] Bai H, Sun K, Wu JH, Zhong ZH, Xu SL, Zhang HR, et al. Proteomic and metabolomic characterization of cardiac tissue in acute myocardial ischemia injury rats. PLoS ONE. 2020; 15: e0231797.

[61] Du X, Li Y, Wang Y, You H, Hui P, Zheng Y, et al. Increased branched-chain amino acid levels are associated with long-term adverse cardiovascular events in patients with STEMI and acute heart failure. Life Sciences. 2018; 209: 167–172.

[62] Cheng S, Rhee EP, Larson MG, Lewis GD, McCabe EL, Shen D, et al. Metabolite profiling identifies pathways associated with metabolic risk in humans. Circulation. 2012; 125: 2222–2231.

[63] Lu G, Sun H, She P, Youn J, Warburton S, Ping P, et al. Protein phosphatase 2Cm is a critical regulator of branched-chain amino acid catabolism in mice and cultured cells. Journal of Clinical Investigation. 2009; 119: 1678–1687.

[64] Sun H, Olson KC, Gao C, Prosdocimo DA, Zhou M, Wang Z, et al. Catabolic Defect of Branched-Chain Amino Acids Promotes Heart Failure. Circulation. 2016; 133: 2038–2049.

[65] Dong W, Zhou M, Dong M, Pan B, Liu Y, Shao J, et al. Keto acid metabolites of branched-chain amino acids inhibit oxidative stress-induced necrosis and attenuate myocardial ischemia-reperfusion injury. Journal of Molecular and Cellular Cardiology. 2016; 101: 90–98.

[66] McKirnan MD, Ichikawa Y, Zhang Z, Zemljic-Harpf AE, Fan S, Barupal DK, et al. Metabolomic analysis of serum and myocardium in compensated heart failure after myocardial infarction. Life Sciences. 2019; 221: 212–223.

[67] Zornoff LAM, Paiva SAR, Duarte DR, Spadaro J. Ventricular remodeling after myocardial infarction: concepts and clinical implications. Arquivos Brasileiros De Cardiologia. 2009; 92: 150–164.

[68] Murakami S, Sakurai T, Tomoike H, Sakono M, Nasu T, Fukuda N. Prevention of hypercholesterolemia and atherosclerosis in the hyperlipidemia- and atherosclerosis-prone Japanese (LAP) quail by taurine supplementation. Amino Acids. 2010; 38: 271–278.

[69] Ardisson LP, Rafacho BPM, Santos PP, Assalin H, Gonçalves AF, Azevedo PS, et al. Taurine attenuates cardiac remodeling after myocardial infarction. International Journal of Cardiology. 2013; 168: 4925–4926.

[70] Ibanez B, James S, Agewall S, Antunes MJ, Bucciarelli-Ducci C, Bueno H, et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC). European Heart Journal. 2018; 39: 119–177.

[71] Ueeda M, Doumei T, Takaya Y, Ohnishi N, Takaishi A, Hirohata S, et al. Association of serum levels of arachidonic acid and eicosapentaenoic acid with prevalence of major adverse cardiac events after acute myocardial infarction. Heart and Vessels. 2011; 26: 145–152.

[72] Lázaro I, Rueda F, Cediel G, Ortega E, García-García C, Sala-Vila A, et al. Circulating Omega-3 Fatty Acids and Incident Adverse Events in Patients with Acute Myocardial Infarction. Journal of the American College of Cardiology. 2020; 76: 2089–2097.

[73] Endres S. n-3 polyunsaturated fatty acids and human cytokine synthesis. Lipids. 1996; 31: S239–S242.

[74] Vignoli A, Tenori L, Giusti B, Valente S, Carrabba N, Balzi D, et al. Differential Network Analysis Reveals Metabolic Determinants Associated with Mortality in Acute Myocardial Infarction Patients and Suggests Potential Mechanisms Underlying Different Clinical Scores used to Predict Death. Journal of Proteome Research. 2020; 19: 949–961.

[75] Sibilitz KL, Benn M, Nordestgaard BG. Creatinine, eGFR and association with myocardial infarction, ischemic heart disease and early death in the general population. Atherosclerosis. 2014; 237: 67–75.

[76] Naito K, Anzai T, Yoshikawa T, Anzai A, Kaneko H, Kohno T, et al. Impact of chronic kidney disease on postinfarction inflammation, oxidative stress, and left ventricular remodeling. Journal of Cardiac Failure. 2008; 14: 831–838.

[77] Vignoli A, Tenori L, Giusti B, Takis PG, Valente S, Carrabba N, et al. NMR-based metabolomics identifies patients at high risk of death within two years after acute myocardial infarction in the AMI-Florence II cohort. BMC Medicine. 2019; 17: 3.

[78] Mardinoglu A, Stančáková A, Lotta LA, Kuusisto J, Boren J, Blüher M, et al. Plasma Mannose Levels are Associated with Incident Type 2 Diabetes and Cardiovascular Disease. Cell Metabolism. 2017; 26: 281–283.

[79] DeFilippis AP, Trainor PJ, Hill BG, Amraotkar AR, Rai SN, Hirsch GA, et al. Identification of a plasma metabolomic signature of thrombotic myocardial infarction that is distinct from non-thrombotic myocardial infarction and stable coronary artery disease. PLoS ONE. 2017; 12: e0175591.

[80] Chen J, Hong T, Ding S, Deng L, Abudupataer M, Zhang W, et al. Aggravated myocardial infarction-induced cardiac remodeling and heart failure in histamine-deficient mice. Scientific Reports. 2017; 7: 44007.

[81] Moradi-Arzeloo M, Farshid AA, Tamaddonfard E, Asri-Rezaei S. Effects of histidine and vitamin C on isoproterenol-induced acute myocardial infarction in rats. Veterinary Research Forum. 2016; 7: 47–54.

[82] Han J, Xia Y, Lin L, Zhang Z, Tian H, Li K. Next-generation Metabolomics in the Development of New Antidepressants: Using Albiflorin as an Example. Current Pharmaceutical Design. 2018; 24: 2530–2540.


Abstracted / indexed in

Science Citation Index Expanded (SciSearch) Created as SCI in 1964, Science Citation Index Expanded now indexes over 9,200 of the world’s most impactful journals across 178 scientific disciplines. More than 53 million records and 1.18 billion cited references date back from 1900 to present.

Journal Citation Reports/Science Edition Journal Citation Reports/Science Edition aims to evaluate a journal’s value from multiple perspectives including the journal impact factor, descriptive data about a journal’s open access content as well as contributing authors, and provide readers a transparent and publisher-neutral data & statistics information about the journal.

Chemical Abstracts Service Source Index The CAS Source Index (CASSI) Search Tool is an online resource that can quickly identify or confirm journal titles and abbreviations for publications indexed by CAS since 1907, including serial and non-serial scientific and technical publications.

Index Copernicus The Index Copernicus International (ICI) Journals database’s is an international indexation database of scientific journals. It covered international scientific journals which divided into general information, contents of individual issues, detailed bibliography (references) sections for every publication, as well as full texts of publications in the form of attached files (optional). For now, there are more than 58,000 scientific journals registered at ICI.

Geneva Foundation for Medical Education and Research The Geneva Foundation for Medical Education and Research (GFMER) is a non-profit organization established in 2002 and it works in close collaboration with the World Health Organization (WHO). The overall objectives of the Foundation are to promote and develop health education and research programs.

Scopus: CiteScore 1.0 (2022) Scopus is Elsevier's abstract and citation database launched in 2004. Scopus covers nearly 36,377 titles (22,794 active titles and 13,583 Inactive titles) from approximately 11,678 publishers, of which 34,346 are peer-reviewed journals in top-level subject fields: life sciences, social sciences, physical sciences and health sciences.

Embase Embase (often styled EMBASE for Excerpta Medica dataBASE), produced by Elsevier, is a biomedical and pharmacological database of published literature designed to support information managers and pharmacovigilance in complying with the regulatory requirements of a licensed drug.

Submission Turnaround Time

Conferences

Top