Proteomics Research in Cardiovascular Medicine and Biomarker Discovery

Maggie P Y Lam, Peipei Ping, Elizabeth Murphy, Maggie P Y Lam, Peipei Ping, Elizabeth Murphy

Abstract

Proteomics is a systems physiology discipline to address the large-scale characterization of protein species within a biological system, be it a cell, a tissue, a body biofluid, an organism, or a cohort population. Building on advances from chemical analytical platforms (e.g., mass spectrometry and other technologies), proteomics approaches have contributed powerful applications in cardiovascular biomedicine, most notably in: 1) the discovery of circulating protein biomarkers of heart diseases from plasma samples; and 2) the identification of disease mechanisms and potential therapeutic targets in cardiovascular tissues, in both preclinical models and translational studies. Contemporary proteomics investigations offer powerful means to simultaneously examine tens of thousands of proteins in various samples, and understand their molecular phenotypes in health and disease. This concise review introduces study design considerations, example applications and use cases, as well as interpretation and analysis of proteomics data in cardiovascular biomedicine.

Keywords: mass spectrometry; molecular phenotyping; post-translational modifications; protein arrays; protein signatures; protein dynamics.

Published by Elsevier Inc.

Figures

Figure 1. Sensitivity and Coverage of Various…
Figure 1. Sensitivity and Coverage of Various Proteomics Study Designs
Properties of 3 common proteomics study designs are listed. Discovery (shotgun), targeted, and targeted discovery proteomics approaches take different strategies to address proteome coverage, number of samples, and sensitivity limits. Discovery proteomics experiments have high coverage (up to 10,000 proteins in a single sample), but have limitations in throughput; fewer samples/subjects can be analyzed. Targeted discovery approaches focus on analyzing a panel of high-potential targets in sufficient numbers of samples. Targeted proteomics is able to achieve the highest sensitivity, which allows the detection of low-abundance plasma markers, such as TNF and IL-6, but at the expense of scope and throughput. Graph (center) shows the typical sensitivity range of discovery, targeted discovery, and targeted approaches, juxtaposed with the concentrations of selected plasma proteins and disease markers. Example technological platforms and additional notes are shown on the right. CK-MB = creatine kinase-myocardial band; CRP = C-reactive protein; ELISA = enzyme-linked immunosorbent assay; IL-6 = interleukin 6; LDL = low-density lipoprotein; MS = mass spectrometry; NT-proBNP = N-terminal B-type natriuretic peptide; TNF = tumor necrosis factor.
Central Illustration. Overview of Proteomics Analyses for…
Central Illustration. Overview of Proteomics Analyses for Cardiovascular Diseases
This figure lists major components in common proteomics workflows, as well as their associated experimental considerations in biomarker discovery and disease studies. From top to bottom, protein samples are collected from the plasma of human cohorts or cardiac tissues in animal models according to study goals (either biomarker discovery or mechanistic studies). Three major study approaches (discovery, targeted, and targeted discovery) take different strategies between proteome coverage and analytical throughput, and utilize different technological platforms, including mass spectrometry and protein arrays. Following the acquisition of large-scale datasets, data are processed to identify the protein species present and to deduce their relative quantities across samples. Subsequently, a number of statistical and bioinformatics workflows are used in data interpretation to extract insights from datasets. Network analysis casts proteins in the context of interaction networks and/or altered cellular pathways. Statistical learning and modeling methods connect the identified molecular features to orthogonal phenotypes, identify signatures, and offer information on subject classification or predictive analysis. The identified protein signatures will require validation, which can be achieved by complementary translational studies, including in vitro biochemical analysis and large cohorts. PTM = post-translational modifications.

References

    1. McDonough JL, Labugger R, Pickett W, et al. Cardiac troponin I is modified in the myocardium of bypass patients. Circulation. 2001;103:58–64.
    1. Labugger R, Organ L, Collier C, et al. Extensive troponin I and T modification detected in serum from patients with acute myocardial infarction. Circulation. 2000;102:1221–1226.
    1. Weekes J, Wheeler CH, Yan JX, et al. Bovine dilated cardiomyopathy: proteomic analysis of an animal model of human dilated cardiomyopathy. Electrophoresis. 1999;20:898–906.
    1. Ping P, Zhang J, Pierce WM, et al. Functional proteomic analysis of protein kinase C ε signaling complexes in the normal heart and during cardioprotection. Circ Res. 2001;88:59–62.
    1. Edmondson RD, Vondriska TM, Biederman KJ, et al. Protein kinase C ε signaling complexes include metabolism- and transcription/translation-related proteins: complimentary separation techniques with LC/MS/MS. Mol Cell Proteomics. 2002;1:421–433.
    1. Lindsey ML, Goshorn DK, Comte-Walters S, et al. A multidimensional proteomic approach to identify hypertrophy-associated proteins. Proteomics. 2006;6:2225–2235.
    1. Sun J, Picht E, Ginsburg KS, et al. Hypercontractile female hearts exhibit increased S-nitrosylation of the L-type Ca2+ channel α1 subunit and reduced ischemia/reperfusion injury. Circ Res. 2006;98:403–411.
    1. Ge Y, Rybakova IN, Xu Q, et al. Top-down high-resolution mass spectrometry of cardiac myosin binding protein C revealed that truncation alters protein phosphorylation state. Proc Natl Acad Sci U S A. 2009;106:12658–12663.
    1. Larance M, Lamond AI. Multidimensional proteomics for cell biology. Nat Rev Mol Cell Biol. 2015;16:269–280.
    1. Liu Y, Beyer A, Aebersold R. On the dependency of cellular protein levels on mRNA abundance. Cell. 2016;165:535–550.
    1. Vogel C, Abreu Rde S, Ko D, et al. Sequence signatures and mRNA concentration can explain two-thirds of protein abundance variation in a human cell line. Mol Syst Biol. 2010;6:400.
    1. Tian Q, Stepaniants SB, Mao M, et al. Integrated genomic and proteomic analyses of gene expression in mammalian cells. Mol Cell Proteomics. 2004;3:960–969.
    1. Schwanhäusser B, Busse D, Li N, et al. Global quantification of mammalian gene expression control. Nature. 2011;473:337–342.
    1. Lundberg E, Fagerberg L, Klevebring D, et al. Defining the transcriptome and proteome in three functionally different human cell lines. Mol Syst Biol. 2010;6:450.
    1. Marshall KD, Edwards MA, Krenz M, et al. Proteomic mapping of proteins released during necrosis and apoptosis from cultured neonatal cardiac myocytes. Am J Physiol Cell Physiol. 2014;306:C639–C647.
    1. Van Eyk JE. Proteomics: unraveling the complexity of heart disease and striving to change cardiology. Curr Opin Mol Ther. 2001;3:546–553.
    1. Nanjappa V, Thomas JK, Marimuthu A, et al. Plasma Proteome Database as a resource for proteomics research: 2014 update. Nucleic Acids Res. 2014;42:D959–D965.
    1. Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics. 2002;1:845–867.
    1. Pundir S, Magrane M, Martin MJ, et al. Searching and navigating UniProt databases. Curr Protoc Bioinformatics. 2015;50:1.27.1–1.27.10.
    1. Kusebauch U, Deutsch EW, Campbell DS, et al. Using PeptideAtlas, SRMAtlas, and PASSEL: comprehensive resources for discovery and targeted proteomics. Curr Protoc Bioinformatics. 2014;46:13.25.1–13.25.28.
    1. Zong N, Li H, Li H, et al. Integration of cardiac proteome biology and medicine by a specialized knowledgebase. Circ Res. 2013;113:1043–1053.
    1. Riley NM, Hebert AS, Coon JJ. Proteomics moves into the fast lane. Cell Syst. 2016;2:142–143.
    1. Bensimon A, Heck AJ, Aebersold R. Mass spectrometry-based proteomics and network biology. Annu Rev Biochem. 2012;81:379–405.
    1. Zhang Y, Fonslow BR, Shan B, et al. Protein analysis by shotgun/bottom-up proteomics. Chem Rev. 2013;113:2343–2394.
    1. Keshishian H, Addona T, Burgess M, et al. Quantification of cardiovascular biomarkers in patient plasma by targeted mass spectrometry and stable isotope dilution. Mol Cell Proteomics. 2009;8:2339–2349.
    1. Lam MP, Lau E, Scruggs SB, et al. Site-specific quantitative analysis of cardiac mitochondrial protein phosphorylation. J Proteomics. 2013;81:15–23.
    1. Zhang P, Kirk JA, Ji W, et al. Multiple reaction monitoring to identify site-specific troponin I phosphorylated residues in the failing human heart. Circulation. 2012;126:1828–1837.
    1. Percy AJ, Yang J, Hardie DB, et al. Precise quantitation of 136 urinary proteins by LC/MRM-MS using stable isotope labeled peptides as internal standards for biomarker discovery and/or verification studies. Methods. 2015;81:24–33.
    1. Fu Q, Chen Z, Zhang S, et al. Multiple and selective reaction monitoring using triple quadrupole mass spectrometer: preclinical large cohort analysis. Methods Mol Biol. 2016;1410:249–264.
    1. Keshishian H, Addona T, Burgess M, et al. Quantification of cardiovascular biomarkers in patient plasma by targeted mass spectrometry and stable isotope dilution. Mol Cell Proteomics. 2009;8:2339–2349.
    1. Gerstein HC, Paré G, McQueen MJ, et al. Outcome Reduction With Initial Glargine Intervention Trial Investigators. Identifying novel biomarkers for cardiovascular events or death in people with dysglycemia. Circulation. 2015;132:2297–2304.
    1. Sajic T, Liu Y, Aebersold R. Using data-independent, high-resolution mass spectrometry in protein biomarker research: perspectives and clinical applications. Proteomics Clin Appl. 2015;9:307–321.
    1. Ngo D, Sinha S, Shen D, et al. Aptamer-based proteomic profiling reveals novel candidate biomarkers and pathways in cardiovascular disease. Circulation. 2016;134:270–285.
    1. Ganz P, Heidecker B, Hveem K, et al. Development and validation of a protein-based risk score for cardiovascular outcomes among patients with stable coronary heart disease. JAMA. 2016;315:2532–2541.
    1. Lind L, Siegbahn A, Lindahl B, et al. Discovery of new risk markers for ischemic stroke using a novel targeted proteomics chip. Stroke. 2015;46:3340–3347.
    1. Sabatine MS. Using aptamer-based technology to probe the plasma proteome for cardiovascular disease prediction. JAMA. 2016;315:2525–2526.
    1. Gramolini A, Lau E, Liu PP. Identifying low-abundance biomarkers: aptamer-based proteomics potentially enables more sensitive detection in cardiovascular diseases. Circulation. 2016;134:286–289.
    1. Keshishian H, Burgess MW, Gillette MA, et al. Multiplexed, quantitative workflow for sensitive biomarker discovery in plasma yields novel candidates for early myocardial injury. Mol Cell Proteomics. 2015;14:2375–2393.
    1. Cominetti O, Nuñez Galindo A, Corthésy J, et al. Proteomic biomarker discovery in 1000 human plasma samples with mass spectrometry. J Proteome Res. 2016;15:389–399.
    1. Olsen JV, Mann M. Status of large-scale analysis of post-translational modifications by mass spectrometry. Mol Cell Proteomics. 2013;12:3444–3452.
    1. Huang KY, Su MG, Kao HJ, et al. dbPTM 2016: 10-year anniversary of a resource for post-translational modification of proteins. Nucleic Acids Res. 2016;44:D435–D446.
    1. Thingholm TE, Jørgensen TJ, Jensen ON, et al. Highly selective enrichment of phosphorylated peptides using titanium dioxide. Nat Protoc. 2006;1:1929–1935.
    1. Lundby A, Andersen MN, Steffensen AB, et al. In vivo phosphoproteomics analysis reveals the cardiac targets of β-adrenergic receptor signaling. Sci Signal. 2013;6:rs11.
    1. Schechter MA, Hsieh MK, Njoroge LW, et al. Phosphoproteomic profiling of human myocardial tissues distinguishes ischemic from non-ischemic end stage heart failure. PLoS ONE. 2014;9:e104157.
    1. Karamanlidis G, Lee CF, Garcia-Menendez L, et al. Mitochondrial complex I deficiency increases protein acetylation and accelerates heart failure. Cell Metab. 2013;18:239–250.
    1. Nguyen TT, Wong R, Menazza S, et al. Cyclophilin D modulates mitochondrial acetylome. Circ Res. 2013;113:1308–1319.
    1. Yang W, Paschen W. SUMO proteomics to decipher the SUMO-modified proteome regulated by various diseases. Proteomics. 2015;15:1181–1191.
    1. Barallobre-Barreiro J, Gupta SK, Zoccarato A, et al. Glycoproteomics reveals decorin peptides with anti-myostatin activity in human atrial fibrillation. Circulation. 2016;134:817–832.
    1. Parker BL, Palmisano G, Edwards AV, et al. Quantitative N-linked glycoproteomics of myocardial ischemia and reperfusion injury reveals early remodeling in the extracellular environment. Mol Cell Proteomics. 2011;10:M110.006833.
    1. Wang SB, Foster DB, Rucker J, et al. Redox regulation of mitochondrial ATP synthase: implications for cardiac resynchronization therapy. Circ Res. 2011;109:750–757.
    1. Kohr MJ, Aponte A, Sun J, et al. Measurement of S-nitrosylation occupancy in the myocardium with cysteine-reactive tandem mass tags: short communication. Circ Res. 2012;111:1308–1312.
    1. Zong N, Ping P, Lau E, et al. Lysine ubiquitination and acetylation of human cardiac 20S proteasomes. Proteomics Clin Appl. 2014;8:590–594.
    1. Fert-Bober J, Giles JT, Holewinski RJ, et al. Citrullination of myofilament proteins in heart failure. Cardiovasc Res. 2015;108:232–242.
    1. Lindsey ML, Iyer RP, Zamilpa R, et al. A novel collagen matricryptin reduces left ventricular dilation post-myocardial infarction by promoting scar formation and angiogenesis. J Am Coll Cardiol. 2015;66:1364–1374.
    1. Kerbey AL, Randle PJ, Cooper RH, et al. Regulation of pyruvate dehydrogenase in rat heart. Mechanism of regulation of proportions of dephosphorylated and phosphorylated enzyme by oxidation of fatty acids and ketone bodies and of effects of diabetes: role of coenzyme A, acetyl-coenzyme A and reduced and oxidized nicotinamideadenine dinucleotide. Biochem J. 1976;154:327–348.
    1. Sharma K, D'Souza RC, Tyanova S, et al. Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thr-based signaling. Cell Rep. 2014;8:1583–1594.
    1. Lundby A, Secher A, Lage K, et al. Quantitative maps of protein phosphorylation sites across 14 different rat organs and tissues. Nat Commun. 2012;3:876.
    1. Deng N, Zhang J, Zong C, et al. Phosphoproteome analysis reveals regulatory sites in major pathways of cardiac mitochondria. Mol Cell Proteomics. 2011;10:M110.000117.
    1. Lee DI, Zhu G, Sasaki T, et al. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature. 2015;519:472–476.
    1. Scholten A, Preisinger C, Corradini E, et al. Phosphoproteomics study based on in vivo inhibition reveals sites of calmodulin-dependent protein kinase II regulation in the heart. J Am Heart Assoc. 2013;2:e000318.
    1. Kooij V, Holewinski RJ, Murphy AM, et al. Characterization of the cardiac myosin binding protein-C phosphoproteome in healthy and failing human hearts. J Mol Cell Cardiol. 2013;60:116–120.
    1. Svinkina T, Gu H, Silva JC, et al. Deep, quantitative coverage of the lysine acetylome using novel anti-acetyl-lysine antibodies and an optimized proteomic workflow. Mol Cell Proteomics. 2015;14:2429–2440.
    1. Horton JL, Martin OJ, Lai L, et al. Mitochondrial protein hyperacetylation in the failing heart. JCI Insight. 2016;2:e84897.
    1. Sun J, Aponte AM, Menazza S, et al. Additive cardioprotection by pharmacological postconditioning with hydrogen sulfide and nitric oxide donors in mouse heart: S-sulfhydration vs. S-nitrosylation. Cardiovasc Res. 2016;110:96–106.
    1. Sun J, Nguyen T, Aponte AM, et al. Ischaemic preconditioning preferentially increases protein S-nitrosylation in subsarcolemmal mitochondria. Cardiovasc Res. 2015;106:227–236.
    1. Murray CI, Chung HS, Uhrigshardt H, et al. Quantification of mitochondrial S-nitrosylation by CysTMT6 switch assay. Methods Mol Biol. 2013;1005:169–179.
    1. Kim TY, Wang D, Kim AK, et al. Metabolic labeling reveals proteome dynamics of mouse mitochondria. Mol Cell Proteomics. 2012;11:1586–1594.
    1. Price JC, Holmes WE, Li KW, et al. Measurement of human plasma proteome dynamics with 2H2O and liquid chromatography tandem mass spectrometry. Anal Biochem. 2012;420:73–83.
    1. Lam MP, Wang D, Lau E, et al. Protein kinetic signatures of the remodeling heart following isoproterenol stimulation. J Clin Invest. 2014;124:1734–1744.
    1. Lau E, Cao Q, Ng DC, et al. A large dataset of protein dynamics in the mammalian heart proteome. Sci Data. 2016;3:160015.
    1. Kolwicz SC, Jr, Tian R. Glucose metabolism and cardiac hypertrophy. Cardiovasc Res. 2011;90:194–201.
    1. Hsu YR, Chang WC, Mendiaz EA, et al. Selective deamidation of recombinant human stem cell factor during in vitro aging: isolation and characterization of the aspartyl and isoaspartyl homodimers and heterodimers. Biochemistry. 1998;37:2251–2262.
    1. Burgers PP, van der Heyden MA, Kok B, et al. A systematic evaluation of protein kinase A-A-kinase anchoring protein interaction motifs. Biochemistry. 2015;54:11–21.
    1. Aye TT, Soni S, van Veen TA, et al. Reorganized PKA-AKAP associations in the failing human heart. J Mol Cell Cardiol. 2012;52:511–518.
    1. Pankow S, Bamberger C, Calzolari D, et al. ΔF508 CFTR interactome remodelling promotes rescue of cystic fibrosis. Nature. 2015;528:510–516.
    1. Morris JH, Knudsen GM, Verschueren E, et al. Affinity purification-mass spectrometry and network analysis to understand protein-protein interactions. Nat Protoc. 2014;9:2539–2554.
    1. Huttlin EL, Ting L, Bruckner RJ, et al. The BioPlex network: a systematic exploration of the human interactome. Cell. 2015;162:425–440.
    1. Waldron L, Steimle JD, Greco TM, et al. The cardiac TBX5 interactome reveals a chromatin remodeling network essential for cardiac septation. Dev Cell. 2016;36:262–275.
    1. Rizzetto S, Priami C, Csikász-Nagy A. Qualitative and quantitative protein complex prediction through proteome-wide simulations. PLoS Comput Biol. 2015;11:e1004424.
    1. Goldfarb D, Hast BE, Wang W, et al. Spotlite: web application and augmented algorithms for predicting co-complexed proteins from affinity purification--mass spectrometry data. J Proteome Res. 2014;13:5944–5955.
    1. Gingras AC, Gstaiger M, Raught B, et al. Analysis of protein complexes using mass spectrometry. Nat Rev Mol Cell Biol. 2007;8:645–654.
    1. Leitner A, Faini M, Stengel F, et al. Crosslinking and mass spectrometry: an integrated technology to understand the structure and function of molecular machines. Trends Biochem Sci. 2016;41:20–32.
    1. Rifai N, Gillette MA, Carr SA. Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat Biotechnol. 2006;24:971–983.
    1. Percy AJ, Chambers AG, Yang J, et al. Multiplexed MRM-based quantitation of candidate cancer biomarker proteins in undepleted and non-enriched human plasma. Proteomics. 2013;13:2202–2215.
    1. Abbatiello SE, Schilling B, Mani DR, et al. Large-scale interlaboratory study to develop, analytically validate and apply highly multiplexed, quantitative peptide assays to measure cancer-relevant proteins in plasma. Mol Cell Proteomics. 2015;14:2357–2374.
    1. Goodman SN. Aligning statistical and scientific reasoning. Science. 2016;352:1180–1181.
    1. de Lemos JA, Rohatgi A, Ayers CR. Applying a big data approach to biomarker discovery: running before we walk? Circulation. 2015;132:2289–2292.
    1. Keshishian H, Burgess MW, Gillette MA, et al. Multiplexed, quantitative workflow for sensitive biomarker discovery in plasma yields novel candidates for early myocardial injury. Mol Cell Proteomics. 2015;14:1–45.
    1. Yin X, Subramanian S, Hwang SJ, et al. Protein biomarkers of new-onset cardiovascular disease: prospective study from the systems approach to biomarker research in cardiovascular disease initiative. Arterioscler Thromb Vasc Biol. 2014;34:939–945.
    1. Addona TA, Shi X, Keshishian H, et al. A pipeline that integrates the discovery and verification of plasma protein biomarkers reveals candidate markers for cardiovascular disease. Nat Biotechnol. 2011;29:635–643.
    1. Mertins P, Mani DR, Ruggles KV, et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature. 2016;534:55–62.
    1. Williams EG, Wu Y, Jha P, et al. Systems proteomics of liver mitochondria function. Science. 2016;352:aad0189.
    1. Kong SW, Hu YW, Ho JW, et al. Heart failure-associated changes in RNA splicing of sarcomere genes. Circ Cardiovasc Genet. 2010;3:138–146.
    1. Song HK, Hong SE, Kim T, et al. Deep RNA sequencing reveals novel cardiac transcriptomic signatures for physiological and pathological hypertrophy. PLoS ONE. 2012;7:e35552.
    1. Hebert AS, Merrill AE, Bailey DJ, et al. Neutron-encoded mass signatures for multiplexed proteome quantification. Nat Methods. 2013;10:332–334.
    1. Stubbs P, Seed M, Lane D, et al. Lipoprotein(a) as a risk predictor for cardiac mortality in patients with acute coronary syndromes. Eur Heart J. 1998;19:1355–1364.
    1. McQueen MJ, Hawken S, Wang X, et al. INTERHEART study investigators. Lipids, lipoproteins, and apolipoproteins as risk markers of myocardial infarction in 52 countries (the INTERHEART study): a case-control study. Lancet. 2008;372:224–233.
    1. de Lemos JA, McGuire DK, Drazner MH. B-type natriuretic peptide in cardiovascular disease. Lancet. 2003;362:316–322.
    1. Emerging Risk Factors Collaboration. C-reactive protein, fibrinogen, and cardiovascular disease prediction. N Engl J Med. 2012;367:1310–1320.
    1. Puleo PR, Guadagno PA, Roberts R, et al. Early diagnosis of acute myocardial infarction based on assay for subforms of creatine kinase-MB. Circulation. 1990;82:759–764.
    1. Shlipak MG, Sarnak MJ, Katz R, et al. Cystatin C and the risk of death and cardiovascular events among elderly persons. N Engl J Med. 2005;352:2049–2060.
    1. Daniels LB, Laughlin GA, Sarno MJ, et al. Lipoprotein-associated phospholipase A2 is an independent predictor of incident coronary heart disease in an apparently healthy older population: the Rancho Bernardo Study. J Am Coll Cardiol. 2008;51:913–919.
    1. Brennan ML, Penn MS, Van Lente F, et al. Prognostic value of myeloperoxidase in patients with chest pain. N Engl J Med. 2003;349:1595–1604.
    1. Kavsak PA, MacRae AR, Newman AM, et al. Effects of contemporary troponin assay sensitivity on the utility of the early markers myoglobin and CKMB isoforms in evaluating patients with possible acute myocardial infarction. Clin Chim Acta. 2007;380:213–216.
    1. Johnson BD, Kip KE, Marroquin OC, et al. Serum amyloid A as a predictor of coronary artery disease and cardiovascular outcome in women: the National Heart, Lung, and Blood Institute-Sponsored Women's Ischemia Syndrome Evaluation (WISE) Circulation. 2004;109:726–732.
    1. Adams JE, III, Bodor GS, Dávila-Román VG, et al. Cardiac troponin I. A marker with high specificity for cardiac injury. Circulation. 1993;88:101–106.
    1. Reichlin T, Hochholzer W, Bassetti S, et al. Early diagnosis of myocardial infarction with sensitive cardiac troponin assays. N Engl J Med. 2009;361:858–867.
    1. Danese E, Montagnana M. An historical approach to the diagnostic biomarkers of acute coronary syndrome. Ann Transl Med. 2016;4:194.
    1. Ladenson JH. A personal history of markers of myocyte injury [myocardial infarction] Clin Chim Acta. 2007;381:3–8.
    1. Eng JK, McCormack AL, Yates JR. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J Am Soc Mass Spectrom. 1994;5:976–989.
    1. O'Leary NA, Wright MW, Brister JR, et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res. 2016;44:D733–D745.

Source: PubMed

3
Abonnieren